Calnuc Binds to LRP9 and Affects its

Transcription

Calnuc Binds to LRP9 and Affects its
Université de Sherbrooke
Rôle de Calnuc dans le triage endosomial des récepteurs lysosomiaux et
implication potentielle dans les maladies du lysosome
Par
Heidi Larkin
Programme de pharmacologie
Thèse présentée à la Faculté de médecine et des sciences de la santé
en vue de l’obtention du grade de philosophiae doctor (Ph.D.) en pharmacologie
Sherbrooke, Québec, Canada
21 janvier 2016
Membres du jury d’évaluation
Dre. LAVOIE Christine
Directrice de recherche, Pre. Département de pharmacologie, Sherbrooke
Dr. PSHEZHETSKY Alexey
Juge externe, Pr. Université de Montréal, Montréal
Dr. ROUCOU Xavier
Juge interne, Pr. Département de biochimie, Sherbrooke
Dr. GRANDBOIS Michel
Directeur de programme, Pr. Département de pharmacologie, Sherbrooke
© Heidi Larkin, 2015
RÉSUMÉ FRANÇAIS
Rôle de Calnuc dans le triage endosomial des récepteurs lysosomiaux et implication
potentielle dans les maladies du lysosome
Par
Heidi Larkin
Programme de doctorat en pharmacologie
Thèse présentée à la Faculté de médecine et des sciences de la santé en vue de l’obtention
du diplôme de philosophiae doctor (Ph.D.) en pharmacologie à la Faculté de médecine et
des sciences de la santé, Université de Sherbrooke, Sherbrooke, Québec, Canada, J1H 5N4
Calnuc est une protéine ubiquitaire qui lie le calcium et qui est présente au réseau transgolgien (TGN) ainsi qu'aux endosomes. Notre groupe a précédemment mis en évidence le rôle
de Calnuc dans le transport de Low density lipoprotein receptor-related protein 9 (LRP9), un
récepteur aux lipoprotéines de faible densité qui cycle entre le TGN et les endosomes. Les
récepteurs lysosomiaux au mannose-6-phosphate (MPR) et Sortiline sont bien caractérisés et
empruntent également cette voie. À l'image de LRP9, nous avons montré que Calnuc prévient
leur dégradation aux lysosomes en participant à leur recyclage à partir des endosomes vers le
TGN. En fait, Calnuc est importante pour l'activation et l'association membranaire de Rab7, une
petite protéine G qui recrute ensuite le complexe Rétromère responsable du transport rétrograde
des récepteurs. La glycoprotéine lysosomiale Ceroid lipofuscinosis neuronal 5 (CLN5) est
également impliquée dans ce processus. La structure et la fonction de cette dernière n'étant pas
clairement définies, nous avons établi qu'elle est synthétisée sous forme d’une glycoprotéine
transmembranaire de type II, mais son domaine N-terminal cytoplasmique et son segment
transmembranaire sont rapidement éliminés suivant le clivage du peptide signal de manière à
former une protéine CLN5 mature fortement associée à la membrane par une hélice
amphipathique (AH). La compréhension des propriétés de base de CLN5 est particulièrement
pertinente puisque la protéine est impliquées dans certaines variantes de céroïdeslipofuscinoses neuronales (NCL), une maladie neurodégénérative rare causée par une surcharge
des lysosomes. D'ailleurs, nos données indiquent que les mutants pathologiques de CLN5
dépourvus de cette AH perdent leur association membranaire, sont retenus au réticulum
endoplasmique et sont rapidement dégradés. En raison de la similitude des fonctions de Calnuc
et de CLN5 au niveau du triage endosomial, nous avons exploré le lien entre les deux protéines.
Calnuc cytosolique et CLN5 luminale semblent former un complexe, par l'intermédiaire de la
protéine transmembranaire CLN3, de façon à influencer l'activité de Rab7. CLN3 étant aussi
associée aux NCL, nous avons finalement exploré la potentielle implication de Calnuc dans la
maladie. L'absence de Calnuc entraîne des phénotypes cellulaires typiques des NCL comme un
engorgement des lysosomes, une accumulation de matériel autofluorescent et une augmentation
de l'autophagie. Les niveaux protéiques de Calnuc sont diminués dans toutes les lignées de
fibroblastes de patients atteints de NCL disponibles ce qui indique que Calnuc pourrait être
impliquée dans certains types de NCL. La présente thèse couvre donc la découverte de la
fonction de Calnuc dans le transport intracellulaire, jusqu'à son implication potentielle dans les
NCL, de même qu'une étude topologique de CLN5.
Mots clés : Calnuc, CLN3, CLN5, MPR, Sortiline, Rab, Rétromère, lipofuscinose
RÉSUMÉ ANGLAIS
Calnuc fonction in endosomal sorting of lysosomal receptors and potential implication
in lysosomal diseases
Par
Heidi Larkin
Programme de doctorat en pharmacologie
Thèse présentée à la Faculté de médecine et des sciences de la santé en vue de l’obtention
du diplôme de philosophiae doctor (Ph.D.) en pharmacologie à la Faculté de médecine et
des sciences de la santé, Université de Sherbrooke, Sherbrooke, Québec, Canada, J1H 5N4
Calnuc is a ubiquitous Ca2+-binding protein present on the trans-Golgi network (TGN)
and endosomes. We previously highlighted the role of Calnuc in the transport of Low density
lipoprotein receptor-related protein 9 (LRP9), a low density lipoprotein (LDL) receptor that
cycles between the TGN and endosomes. Lysosomal receptors mannose-6-phosphate receptor
(MPR) and Sortilin are well-characterized and also use the TGN-to-endosome trafficking
pathway. Similarly to LPR9, we showed that Calnuc prevent their degradation in lysosomes by
acting in their recycling from endosomes to the TGN. In fact, Calnuc is a important for the
activation and the membrane association of Rab7, a small G protein which then recruit the
Retromer complex known to be responsible for the retrograde transport of receptors. Lysosomal
glycoprotein Ceroid lipofuscinosis neuronal 5 (CLN5) is also involved in this process. Because
its structure and function have not yet been clearly defined, we established that it is synthesized
as a type II transmembrane (TM) glycoprotein, but its cytoplasmic N-terminus and TM
segment are rapidly removed following signal-peptide cleavage to generate mature CLN5
which is tightly associated to membrane through an amphipathic helix (AH). The understanding
of the basic properties of CLN5 is particularly important given that CLN5 is involved in some
variants of neuronal ceroid lipofuscinosis (NCL), a rare neurodegenerative disease caused by
lysosomal overload. Moreover, our data indicate that CLN5 pathological mutants deprived of
AH lose their membrane association, are retained in the endoplasmic reticulum, and are rapidly
degraded. Based on the similarity featured by Calnuc and CLN5 in endosomal sorting, we
explored the link between these two proteins. Cytosolic Calnuc and luminal CLN5 seem to
form a complex, through the transmembrane protein CLN3, in order to influence the activity of
Rab7. As CLN3 is also associated with NCL, we finally explored the potential involvement of
Calnuc in this disease. Canuc depletion leads to typical NCL phenotypes such as lysosome
enlargement, accumulation of autofluorescent material and of an increased of autophagy
induction. Canuc's levels are decreased in all fibroblasts cell lines of NCL patients available
indicating that Calnuc could be involved in some types of NCL. This thesis thus covers the
discovery of the function of Calnuc in intracellular transport up to its potential involvement in
the NCL, as well as a topological study CLN5.
Mots clés : Calnuc, CLN3, CLN5, MPR, Sortiline, Rab, rétromère, lipofuscinosis
iii
À tous les étudiants
qui passeront à travers ce tumultueux périple
Aux Débrouillards
qui influencèrent une génération de scientifiques
Mais surtout,
En mémoire de mon oncle Paul
que je n'ai que trop brièvement connu
La science ne sert guère qu'à nous donner une
idée de l'étendue de notre ignorance.
Félicité Robert de Lamennais
TABLE DES MATIÈRES
RÉSUMÉ FRANÇAIS .........................................................................................................II
RÉSUMÉ ANGLAIS ......................................................................................................... III
LISTE DES FIGURES ................................................................................................... VIII
LISTE DES TABLEAUX .................................................................................................. IX
LISTE DES ABRÉVIATIONS ........................................................................................... X
INTRODUCTION ................................................................................................................ 1
1
CALNUC ....................................................................................................................... 1
1.1 Une nouvelle protéine d’intérêt .......................................................................... 1
1.2 Structure et localisation cellulaire ..................................................................... 2
1.3 Homologues ........................................................................................................ 6
1.3.1 Cab45 .............................................................................................................. 7
1.3.2 Calréticuline .................................................................................................... 7
1.3.3 NUCB2 ........................................................................................................... 8
1.4 Partenaires d’interaction, fonctions et maladies associées.............................. 12
1.4.1 Calcium et minéralisation ............................................................................. 14
1.4.2 Protéines G et sécrétion régulée.................................................................... 16
1.4.3 ADN et lupus ................................................................................................ 19
1.4.4 Cancer, apoptose et Caspases ....................................................................... 20
1.4.5 COX et inflammation.................................................................................... 21
1.4.6 Activité de protéase à sérine ......................................................................... 23
1.4.7 ATF6 et réponse au stress réticulaire ............................................................ 23
1.4.8 Necdin ........................................................................................................... 24
1.4.9 APP et Alzheimer ......................................................................................... 25
1.4.10 LRP10 et transport intracellulaire ................................................................. 26
2 TRANSPORT INTRACELLULAIRE ................................................................................. 27
2.1 Transport antérograde ...................................................................................... 27
2.1.1 Transport dépendant de M6P - MPR ............................................................ 28
2.1.2 Transport indépendant de M6P - Sortiline.................................................... 29
2.1.3 Transport du Golgi aux endosomes .............................................................. 31
2.1.4 Système endolysosomial ............................................................................... 32
2.2 Transport rétrograde ........................................................................................ 35
2.2.1 Transport des endosomes précoces au Golgi ................................................ 36
2.2.2 Transport des endosomes tardifs au Golgi .................................................... 41
2.3 Rab comme principaux régulateurs du transport intracellulaire ..................... 42
2.3.1 Cycle d'activation des Rab ............................................................................ 42
2.3.2 Localisation des Rab ..................................................................................... 44
2.3.3 Fonctions des Rab dans le transport entre le Golgi et les endosomes .......... 45
3 MALADIES ASSOCIÉES AUX LYSOSOMES .................................................................... 49
3.1 Lysosomes ......................................................................................................... 49
3.2 Maladies de surcharge lysosomiale .................................................................. 51
3.3 Céroïdes-lipofuscinoses neuronales ................................................................. 52
3.3.1 Causes génétiques et protéines associées...................................................... 55
3.3.2 CLN3 ............................................................................................................ 56
3.3.3 CLN5 ............................................................................................................ 61
3.3.4 Interactosome des CLN ................................................................................ 65
3.3.5 Aspects cliniques .......................................................................................... 66
4 PROBLÉMATIQUE, HYPOTHÈSE ET OBJECTIFS ............................................................. 69
4.1 Problématique ................................................................................................... 69
4.2 Hypothèse.......................................................................................................... 69
4.3 Objectifs ............................................................................................................ 70
ARTICLE 1 ......................................................................................................................... 71
RÉSUMÉ – ARTICLE 1 .................................................................................................... 72
ARTICLE 2 ....................................................................................................................... 111
RÉSUMÉ – ARTICLE 2 .................................................................................................. 112
ARTICLE 3 ....................................................................................................................... 146
RÉSUMÉ – ARTICLE 3 .................................................................................................. 147
DISCUSSION .................................................................................................................... 181
CONCLUSION ................................................................................................................. 196
REMERCIEMENTS ........................................................................................................ 197
LISTE DES PUBLICATIONS ........................................................................................ 200
ANNEXE 1......................................................................................................................... 231
ANNEXE 2......................................................................................................................... 254
ANNEXE 3......................................................................................................................... 284
vii
LISTE DES FIGURES
FIGURE 1 - STRUCTURE DE CALNUC. ....................................................................................... 3
FIGURE 2 - COMPARAISON ENTRE CALNUC (NUCB1) ET NUCB2. ........................................ 11
FIGURE 3 - DISTRIBUTION DE CALNUC ET DE SES DIVERS PARTENAIRES D'INTERACTION. ...... 13
FIGURE 4 - PRINCIPAUX MÉCANISMES DE TRANSPORT ANTÉROGRADE ET RÉTROGRADE. ....... 33
FIGURE 5 - LES RAB COMME PRINCIPAUX RÉGULATEURS DU TRANSPORT INTRACELLULAIRE.
...................................................................................................................................... 43
FIGURE 6 - FORMATION ET ULTRASTRUCTURES DE LA LIPOFUSCINE. ..................................... 54
FIGURE 7 - SCHÉMA DE LA STRUCTURE DE CLN3. ................................................................. 58
FIGURE 8 - SCHÉMA DE LA STRUCTURE DE CLN5. ................................................................. 62
FIGURE 9 - CALNUC INTERAGIT AVEC FURINE, TRKA ET CD-MPR, MAIS PAS AVEC P75/NTR
OU EGFR. .................................................................................................................... 183
FIGURE 10 - LA DÉPLÉTION DE CALNUC INFLUENCE LES NIVEAUX DE CI-MPR, FURINE, CDMPR ET EGFR. ........................................................................................................... 184
FIGURE 11 - RÔLES POTENTIELS DE CALNUC DANS LE CYCLE D'ACTIVATION DES RAB. ....... 185
FIGURE 12 - MODÈLE DU COMPLEXE CALNUC-CLN3-CLN5 INFLUENÇANT LE RECRUTEMENT
DE RAB7 ET DU COMPLEXE RÉTROMÈRE. ..................................................................... 192
LISTE DES TABLEAUX
TABLEAU 1 - PATHOLOGIE DES NCL. .................................................................................... 56
TABLEAU 2 - PRINCIPALES MUTATIONS DANS CLN5. ............................................................ 65
LISTE DES ABRÉVIATIONS
A
aa, Acide aminé
AA, Acide arachidonique
AAK1, Adaptor-associated protein kinase 1
ACTH, Hormone adrénocorticotropine
ADN, Acide désoxyribonucléique
AICD, Domaine intracellulaire d’APP
AP, Adaptator protein
ApoE, Apolipoprotéine E
APP, Protéine précurseur de la β-amyloïde
AR, Région acide (Acidic region)
Arf-1, ADP-ribosylation factor-binding protein 1
ARN, Acide ribonucléique
ARNm, ARN messager
ARTS-1, Aminopeptidase regulator of TNRF1 shedding
ASM, Sphingomyélinase
ATF6, Activating transcription factor 6
ATP13A2, Probable cation-transporting ATPase 13A2
Aβ, Peptide β-amyloïde
B
BACE1, ß-sécrétase
BAR, Bin-Amphiphysine-Rvs
Btn, Battenine
BCMP84, Breast cancer membrane protein
BDNF, Brain derived neurotrophic factor
BR, Région basique (Basic region)
C
Ca2+, Calcium
Cab45, 45 kDa calcium-binding protein
Casp, Caspase
CCDC53, Coiled-coil domain-containing protein 53
CD-MPR, Cation-dependant MPR
CerS, Ceramide synthase
CI-MPR, Cation-independant MPR
CL, Curviligne
CLC-7, Chloride channel 7
CLEAR, Coordinated Lysosomal Expression And Regulation
CLN, Ceroid lipofuscinosis neuronal
CMH, Complexe majeur d'histocompatibilité
COG, Conserved oligomeric Golgi
CORVET, Class C core vacuole/endosome tethering
COX, Cyclooxygénase
CPY, Carboxypeptidase Y
Creg1, E1A-stimulated genes 1
CRH, Corticolibérine
CRT, Caréticuline
CSP, Cysteine string protein
CTF, Fragment C-terminal
CTSB, Cathepsine B
CTSD, Cathepsine D
CTSF, Cathepsine F
CUB, Complement C1r/C1s, Uegf, Bmp1
D
DIC, Chaîne intermédiaire de la Dynéine
DMT1, Divalent metal transporter 1
DNAJC5, DnaJ homolog subfamily C member 5
E
E1A, Adenovirus early region 1A protein
EEA1, Early endosome antigen 1
EF, Main EF (EF-hand)
EHD1, EH domain-containing protein 1
ENT1, Equilibrative nucleoside transporter 1
ERC55, Endoplasmic reticulum calcium-binding protein of 55 kDa
ERSE, Élément de réponse au stress réticulaire
EUR, Européenne
F
FAAH, Amidohydrolase des acides gras
Fe65, Amyloid beta A4 precursor protein-binding family B member 1
FinM, Finlandaise majeure
Finm, Finlandaise mineure
FP, Empreinte digitale (fingerprint)
FYCO1, FYVE and coiled-coil domain–containing protein 1
G
GAE, Gamma-adaptin ear
GAP, Protéine d’accélération de la GTPase
GAPvD1, GTPase activating protein and VPS9 domains 1
GARP, Golgi-associated retrograde protein
GAT, GGA and TOM
GDF, Facteur de déplacement de GDI
xi
GDI, Inhibiteur de la séparation des nucléotides de guanine
GDP, Guanosine diphosphate
GEF, Facteur d’échange des nucléotides de guanine
GGA, Golgi-localized, gamma-ear containing Arf binding protein
GGT, Geranylgeranyl transférase
GM, Granulocyte/monocyte colony stimulation factor
GM130, Golgi matrix protein 130
GPCR, Récepteur couplé aux protéines G
GPI, Glycosylphosphatidyl inositol
GROD, Granular osmiophilic deposit
GRN, Granulins precursor
GTP, Guanosine triphosphate
H
HOPS, Homotypic fusion and vacuole protein sorting
Hpse, Héparanase
Hsc70, Heat shock cognate protein 70
I
IGF-2, Insulin-like growth factor-2
IgG, Immunoglobuline G
J
JNK, Jun N-terminal kinase
K
KCTD7, Potassium channel tetramerisation domain containing 7
Kd, Constante de dissociation
KIF3A, Kinesin Family Member 3A
L
LAMP, Lysosome-associated membrane protein
LC3, Microtubule-associated protein 1A/1B-light chain 3
LD, Gouttelette lipidique (Lipid droplet)
LDL-A, Répétition de classe A
LDLR, Récepteur aux lipoprotéines de faible densité
LIMP, Lysosomal integral membrane protein
Lpl, Lipoprotein lipase
LRP, Low density lipoprotein receptor-related protein
LSD, Maladie de surcharge lysosomiale (Lysosomal storage disease)
LYNUS, Lysosome nutrient sensing
LysoNaATP, canal sodique ATP-sensible
LZ, Fermeture/Glissière en leucine (Leucine zipper)
M
M6P, Mannose-6-phosphate
Man2b1, α-Mannosidase B1
xii
MFSD8, Major facilitator superfamily domain-containing protein 8
Mg2+, Magnésium
MPR, Récepteur au mannose-6-phosphate (Mannose-6-phosphate receptor)
mTORC1, Mammalian target of rapamycin complex 1
N
NCL, Céroïde-lipofuscinose neuronale (Neuronal ceroid lipofuscinose)
Necdin, Neurally differentiated embryonal carcinoma-derived protein
NEFA, DNA binding, EF-hand, acidic region
Nesfatin, NEFA/NUCB2-encoded satiety- and fat-influencing protein
NFL, Terre-Neuve
NGF, Nerve growth factor
NLP, Nesfatin-1-like peptide
NLS, Signal de localisation nucléaire (Nuclear localisation signal)
NPC1, Niemann-Pick C1 protein
NSF, N-éthylmaleimide-sensitive-factor
NT, Neurotensine
NTR3, Neurotensin receptor 3
NUC, Nucleobindin
NUCB1, Calnuc
O
OCRL1, Lowe oculocerebrorenal syndrome protein ou Inositol polyphosphate 5phosphatase
ORP1L, Oxysterol‐binding protein
P
PAI-2, Inhibiteur 2 de l’activateur du Plasminogène (Plasminogen activator inhibitor-2)
PAT, Perilipine, Adipophiline, TIP47
PC, Prohormone convertase
PDZ, Postsynaptic density 95/discs large/zonus occludens-1
PG, Prostaglandine
PI(3)P, Phosphatidylinositol-3-phosphate
PI(4,5)P2, Phosphatidylinositol-4,5-bisphosphate
PI, Phosphatidylinositol
PI3K, Phosphatidylinositol-3-kinase
PPAR-g, Peroxisome proliferator-activated receptor gamma
pRB, Retinoblastoma protein
Protéine G, Guanosine nucleotide-binding protein
Psap, Prosaposine
PX, Phox homology
R
RAB11FIP2, RAB11 family-interacting protein 2
REP, Protéine escorte de Rab
xiii
RhoBTB3, Rho-related BR-C, ttk and bab (BTB) domain-containing protein 3
RL, Rectiligne
RILP, Rab7-interacting lysosomal protein
RT-PCR, Transcription inverse combinées à des réactions en chaîne par polymérase
S
S1P, Site-1-protease
SAP, Saposine ou Sphingolipid activator protein
SBDS, Shwachmann-Bodian-Diamond syndrome protein
SCMAS, Sous-unité C de l'ATP synthase mitochondriale
SDF4, Stromal cell derived factor
SERCA, Sarco/endoplasmic reticulum Ca2+-ATPase
shRNA, Petits ARN en épingle à cheveux
siRNA, Petits ARN interférents
SLC29, Solute carrier family 29
SNAP, NSF attachment protein
SNARE, Soluble N-éthylmaleimide-sensitive-factor (NSF) attachment protein (SNAP) receptor
SNX, Sorting Nexin
SorLA, Sorting protein-related receptor containing LDLR class A repeats
SP, Peptide signal (Signal peptide)
sp1, Specificity protein 1
SRP, Particule de reconnaissance du signal (Signal recognition particule)
SWE, Suédoise
SV40-TAg, Simian Vacuolating Virus 40 large T antigen
SWIP, Strumpellin and WASH-interacting protein
T
TAA, Antigène associé aux tumeurs
TBC1D5, Tre2-Bub2-Cdc16-domain-containing protein
TFEB, Facteur de transcription EB
TfR, Transferrin receptor
TGN, Réseau trans-golgien (trans Golgi network)
TIP47, Tail interacting protein of 47 kDa
Tip60, 60 kDa Tat-interactive protein
TNF, Tumor necrosis factor
TNFR1, Tumor necrosis factor receptor 1
TPP1, Tripeptidyl peptidase I
TRAPP, Transport protein particle
TRPML1, Transient receptor potential cation channel, mucolipin subfamily, member 1
U
uPAR, Récepteur de l'Urokinase
UPR, Unfold protein response
UPRE-like, UPR element-like
xiv
V
VAMP, Vesicule-associated membrane protein
VHS, Vps27, Hrs, Stam
Vps, Protéine de triage vacuolaire (Vacuolar protein sorting)
W
WASH, Wiskott–Aldrich Syndrome protein (WASP) and Suppressor of cAR (cAMP
receptor) (SCAR) homologue
WASP, Wiskott–Aldrich Syndrome protein
Z
ZBD, Domaine de liaison au zinc (Zinc-binding domain)
Zn2+, Zinc
Autres
β2-AR, Récepteur bêta-adrénergique
Ø, Résidu hydrophobe encombrant
Φ, Résidu aromatique
Ψ, Résidu aliphatique
xv
INTRODUCTION
Le transport des hydrolases acides jusqu’aux lysosomes nécessite leur triage, dans le
réseau trans-golgien (TGN), par des récepteurs tels que le Récepteur au mannose-6phosphate (MPR) et Sortiline. Ces derniers permettent l’empaquetage des enzymes dans
des vésicules de clathrine puis leur transport antérograde du TGN vers les endosomes. Dans
l’environnement acide des endosomes, les hydrolases se dissocient des récepteurs et sont
acheminées aux lysosomes pour assurer la dégradation des macromolécules dispensables à
la cellule tandis que les récepteurs sont redirigés vers le TGN pour un nouveau cycle de
transport. Ce transport rétrograde requiert de nombreuses protéines dont le complexe
Rétromère et la protéine Ceroid lipofuscinosis neuronal 5 (CLN5). L’ensemble des
mécanismes de régulation du triage endosomial est essentiel pour maintenir la fonction
lysosomiale. En effet, un défaut à ce niveau influence la livraison des hydrolases aux
lysosomes, causant une accumulation intracellulaire de métabolites qui sont caractéristiques
des maladies de désordres lysosomiaux, comme les céroïdes-lipofuscinoses neuronales
(NCL).
Nous avons récemment démontré l’implication de Calnuc dans le triage rétrograde de
Low density lipoprotein receptor-related protein 9 (LRP9), un récepteur peu caractérisé qui
cycle entre le TGN et les endosomes. Le but de la présente étude est, dans un premier
temps, de démontrer que Calnuc agit comme régulateur général du transport rétrograde des
récepteurs des enzymes lysosomiales, CI-MPR et Sortiline. D’autre part, nous examinerons
le rôle de Calnuc dans les désordres lysosomiaux de type NCL.
1
Calnuc
1.1
Une nouvelle protéine d’intérêt
Calnuc a été isolée en 1986 à partir d’un surnageant de lymphocytes B issu d’un
modèle murin (MRL/lpr) de lupus érythémateux disséminé, une maladie systémique autoimmune chronique (Kanai et al., 1986). Originalement nommée Nucleobindin (NUC) en
raison de sa capacité à lier l’acide désoxyribonucléique (ADN) in vitro (Miura et al., 1992),
Calnuc a alors été identifiée alors comme une protéine soluble de 55 kDa promouvant la
formation d’anticorps dirigés contre l’ADN (Kanai et al., 1990; Kanai et al., 1986).
Les études subséquentes ont permis de démontrer que Calnuc est hautement répandue
dans le règne animal étant présente de Ciona intestinalis à Homo sapiens (Kanuru et al.,
2009). L’analyse de sa structure primaire révèle que les différents domaines sont bien
conservés à l’intérieur d’espèces rapprochées d’un point de vue évolutif indiquant que la
protéine doit maintenir ses fonctions. Aussi, sa séquence est davantage conservée chez les
organismes supérieurs qui présentent deux isoformes. Ces derniers, codés par des loci
géniques indépendants, proviendraient de la duplication d’un gène ancestral, modifié au
travers de l’évolution.
Par contre, malgré une structure protéique conservée, le gène codant Calnuc est
considérablement variable, suivant la complexité de l’organisme, en ce qui concerne son
organisation ainsi que sa quantité d’exons et d’introns (Aradhyam et al., 2010). Chez
l’humain, le gène qui code NUCB1 est localisé au niveau du chromosome 19q13.2-q13.4 et
est composé de 13 exons dont 12 codants (Miura et al., 1996). La région en amont contient
des promoteurs communs aux gènes ubiquitaires nécessaires pour le maintien de la fonction
cellulaire de base comme des sites de liaison pour le facteur de transcription Specificity
protein 1 (sp1), une boîte CCAAT, une boîte TATA et plusieurs sites d’initiation (Miura et
al., 1996). D’ailleurs, Calnuc est exprimée dans tous les tissus et cellules humains
investigués jusqu’à présent (Lin et al., 1998; Miura et al., 1996). Le caractère ubiquitaire de
Calnuc laisse croire qu’elle aurait un rôle important dans la maintenance de la machinerie
cellulaire.
1.2
Structure et localisation cellulaire
La forme humaine de Calnuc compte 461 acides aminés arrangés en structure
principalement hélicoïdale (Kanuru et al., 2009; Miura et al., 1994). La partie N-terminale
est compacte et globulaire alors que la partie C-terminale est allongée (Kapoor et al., 2010).
2
Calnuc contient, dans sa séquence, plusieurs motifs et domaines (figure 1) (Ren et al.,
2009), dont certains qui influencent sa localisation cellulaire faisant d’elle une protéine
modulaire multicompartimentale.
Figure 1 - Structure de Calnuc.
Représentation des différents éléments structuraux de Calnuc à l'échelle. SP (peptide
signal), BR (région basique), AR (région acide), LZ (fermeture en leucine), ZBD
(domaine de liaison au zinc), NLS (signal de localisation nucléaire), EF (main EF). Figure
crée avec DOG 2.0, DOG 1.0 : Illustrator of Protein Domain Structures. Ren et al. Cell
Research (2009) 19:271-273.
Entre autres, elle possède un signal bipartite de localisation nucléaire (NLS) (figure
1) (Dingwall and Laskey, 1991) suggérant sa présence au noyau (figure 3A) (Miura et al.,
1994; Wang et al., 1994). En effet, ce motif basique est reconnu par des Importines et
permet le transport à travers les pores du noyau. Il se trouve d’ailleurs dans 50% des
protéines nucléaires contre seulement 5% des protéines non nucléaires (Petersson et al.,
2004). Quoiqu’allant de pair avec la capacité de Calnuc à lier l’ADN, cette localisation est
encore aujourd’hui controversée.
Aussi, la séquence de Calnuc débute par un peptide signal (SP) de 26 acides aminés
(figure 1). Cette structure tripartite au cœur hydrophobe se lie à la Particule de
reconnaissance du signal (SRP) ce qui permet l’adressage au réticulum endoplasmique lors
de la traduction (figure 3A). Calnuc est donc synthétisée au niveau de la lumière du
réticulum endoplasmique, à travers le Translocon, et subit le clivage de son SP par une
peptidase signal. Rapidement, la protéine est transportée au Golgi (figure 3A),
probablement via un récepteur encore inconnu et par un mécanisme dépendant de sa proline
3
en position 28 (figure 1) (Tsukumo et al., 2009). À cet endroit, Calnuc va subir des
modifications post-traductionnelles, dont des O-glycosylations, des sialylations et des
sulfatations, la faisant passer de 60 à 63 kDa (Lavoie et al., 2002; Tsukumo et al., 2007).
La particularité principale de Calnuc, est de posséder deux mains EF (figure 1) qui lui
permettent de lier le calcium (Ca2+). Ces motifs sont constitués d’une hélice alpha (notée
‘E’), d’une boucle et d’une seconde hélice α (notée ‘F’). L’ion est maintenu par la boucle
qui forme une pochette de liaison composée d’atomes d’oxygène de la chaîne latérale
(positions 1, 3, 5, 12), la chaîne principale (position 7) et une molécule d’eau (position 9).
Un résidu hydrophobe (position 8) et une glycine (position 6) sont également nécessaires
(Kretsinger, 1987). La première main EF (EF1) de Calnuc présente ce profil idéal et lui
confère une forte affinité pour le calcium avec une constante de dissociation (Kd1) de 6,3
µM. Quant à elle, la seconde main (EF2) possède une arginine en position 6 nuisant
considérablement à la liaison du calcium (Kd2 = 73,5 µM) (Lin et al., 1999). Globalement,
Calnuc lie donc le calcium avec une faible capacité (~1,1 µmol Ca2+/µmol de protéines) et
une haute affinité (Kd = 6,6 μM) dans un compartiment comme le Golgi qui est riche en cet
ion (0,3 mM). La liaison du calcium entraîne une augmentation du contenu hélicoïdal de
Calnuc de 3,3% (Miura et al., 1994) (pouvant potentiellement représenter une hélice allant
jusqu'à 15 acides aminés) de même que l’exposition de sites hydrophobes ayant le potentiel
d'influencer des interactions avec d’autres protéines. Il est à noter que le magnésium (Mg2+)
peut également se lier faiblement au niveau des mains EF (Kd en millimolaire) (Kanuru et
al., 2009). En revanche, les changements structuraux engendrés par ce dernier sont deux
fois moindres que ceux observés avec le calcium et les deux ions semblent avoir un effet
synergique (Kanuru et al., 2009). Il a été proposé que le magnésium aurait pour rôle de
maintenir une conformation intermédiaire facilitant la liaison du calcium (Gifford et al.,
2007). En 2012, une étude a révélé la présence de deux sites putatifs conservés pour la
liaison de zinc (Zn2+) en N-terminal de Calnuc (figure 1) (Kanuru et al., 2013). Ces motifs
HXXEXnH (où n correspond à 108-135 résidus) sont similaires à ceux retrouvés chez les
protéases de type carboxypeptidase A. Par contre, Calnuc est dénuée d’une telle activité
catalytique qui dépend de plus amples résidus. La liaison du zinc est de haute affinité (Kd =
21 nM), permettant une interaction dans les compartiments riches comme certains organites
4
et le milieu extracellulaire (concentration de l'ordre du micromolaire), mais insuffisante
pour la faible concentration du cytosol (concentration de l'ordre du picomolaire). Il en
résulte un changement de conformation qui expose la surface hydrophobe de Calnuc, de
façon indépendante du calcium (Kanuru et al., 2013). Les variations intracellulaires de zinc,
mais aussi de calcium et de magnésium, pourraient donc s’avérer primordiales pour
modifier la conformation structurale de Calnuc de manière à moduler ses fonctions.
En C-terminal, Calnuc possède également une glissière en leucines (LZ) (figure 1)
(Kanai and Tanuma, 1992; Miura et al., 1992) caractérisée par une hélice alpha qui présente
des leucines à intervalle de sept résidus. Ces dernières sont orientées du même côté de
manière à former, avec les résidus non polaires adjacents, une région hydrophobe
longitudinale qui peut s’enrouler autour d’une autre hélice de même nature. Cette structure
tridimensionnelle permet normalement de lier l’ADN et, par conséquent, est principalement
retrouvée au niveau des facteurs de transcription comme Fos et Jun. Quoique Calnuc
possède également un signal de localisation nucléaire et une région basique de liaison
potentielle à l’ADN (figure 1), aucune évidence n’a démontré son rôle comme régulateur de
l’expression génique. La glissière en leucines de Calnuc lui permet de dimériser (Kanuru et
al., 2009; Kapoor et al., 2010) tout en constituant un site potentiel pour lier diverses
protéines. D'ailleurs, elle a été proposée comme élément permettant à Calnuc de se fixer à
la membrane du Golgi via des interactions protéiques.
D’autres mécanismes, on été proposés comme étant responsables de la rétention de
Calnuc au Golgi. Entre autres, Calnuc possède une région de 15 résidus hydrophobes en Cterminal (figure 1) (Lin et al., 1998) qui pourrait, comme c’est le cas pour Synaptobrevin,
suffire à son ancrage (Kutay et al., 1993; Whitley et al., 1996). Le point isoélectrique de 4,9
de la protéine suggère qu’elle est fortement liée à la membrane de l’organite empêchant son
extraction par des traitements alcalins. Finalement, en 2001, un groupe a démontré qu’une
région riche en leucines et en isoleucines située en N-terminal (acides aminés (aa) 67-120)
(figure 1) est essentielle pour le maintien de la protéine au Golgi (Nesselhut et al., 2001).
Calnuc est donc retenue au Golgi faisant d’elle une protéine résidente (Lin et al., 1998).
C’est d’ailleurs à cet endroit qu’on la retrouve principalement et, de manière plus marquée,
5
au niveau du réseau cis-golgien et du cis-Golgi (Lin et al., 1998). Elle représente environ
0,4% (3,8 g/mg) des protéines golgiennes chez les cellules de foie de rat (Lin et al., 1999)
faisant d'elle la protéine soluble la plus abondante du compartiment (Gilchrist et al., 2006).
Calnuc va demeurer dans l'organite jusqu’à 24h avant d’être sécrétée par les voies
constitutive et constitutive-like (Lavoie et al., 2002) pour se retrouver dans le milieu
extracellulaire (figure 3A).
Outre sa localisation au niveau de la voie sécrétoire et au niveau extracellulaire,
Calnuc se retrouve aussi dans le cytosol, plus particulièrement à la surface du Golgi, de la
membrane plasmique, des endosomes et des granules de sécrétion (figure 3A) (Brodeur et
al., 2009; Lin et al., 2009). Cette localisation au cytoplasme est quelque peu inhabituelle
pour une protéine possédant un peptide signal, mais peut s’expliquer par deux hypothèses.
D’abord, Calnuc pourrait subir une rétrotranslocation vers le cytoplasme via le Translocon,
comme c’est le cas pour Calréticuline (Afshar et al., 2005). D’autres protéines liant le Ca2+,
dont la Phospholipase A2, peuvent transloquer au Golgi en réponse à des variations du Ca2+
cytosolique (Evans et al., 2001). Aussi, il est possible que la reconnaissance du peptide
signal de Calnuc fasse parfois défaut ce qui préviendrait l’insertion de Calnuc dans la
lumière du RE et mènerait à sa synthèse au niveau du cytosol. C’est d’ailleurs le cas pour
l’Inhibiteur 2 de l’activateur du Plasminogène (PAI-2) qui subit une translocation variable
(Belin et al., 1996). La différence de mobilité de Calnuc lors de fractionnements suggère
l’ajout de modifications post-traductionnelles distinctes en fonction de la localisation ce qui
suggère une régulation du peptide signal (Lin et al., 2000).
1.3
Homologues
L'homologie entre protéines est d'une grande utilité afin d'identifier des domaines et
motifs d'intérêt qui apportent des indices relativement aux fonctions et interactions
protéiques. Calnuc possède peu de similarité avec d'autres protéines si ce n'est avec son
paralogue NUCB2 ainsi qu'avec deux autres protéines liant le calcium, soit Cab45 et
Calréticuline.
6
1.3.1 Cab45
Tout comme Calnuc, 45 kDa calcium-binding protein (Cab45), aussi connu sous le
nom de Stromal cell derived factor (SDF4), est une protéine soluble résidente du Golgi qui
lie le calcium (Scherer et al., 1996). L'ion est nécessaire à sa rétention au Golgi et ses 362
acides aminés contiennent six mains EF (Koivu et al., 1997). Cab45 lie des cargos de façon
dépendante du calcium et permet leur triage au TGN vers la voie de sécrétion (von Blume
et al., 2012). Elle pourrait aussi être nécessaire pour l'homéostasie calcique, d'autant plus
qu'elle présente une forte homologie avec les protéines Réticulocalbine et Endoplasmic
reticulum calcium-binding protein of 55 kDa (ERC55) qui lient le calcium au RE (Scherer
et al., 1996). Aussi, Cab45 possède environ 30% de similarité, mais aucune homologie
marquée, avec Calnuc à l'exception d'une courte séquence AANXE (E/D) (Lin et al., 1998).
1.3.2 Calréticuline
Caréticuline (CRT) est une protéine chaperonne soluble qui interagit avec les
protéines mal repliées de manière à prévenir leur export du RE. Malgré l'absence de mains
EF dans sa séquence, elle lie également le calcium avec une faible affinité et une haute
capacité pour en faire une réserve mobilisable au niveau du même organite. Calnuc a une
homologie de séquence considérable de 30% avec CRT de rat (Lin et al., 1998). La
similarité la plus importante (43%) se trouve dans la région C-terminale qui présente
beaucoup de résidus acides et deux motifs conservés AY(I/A)EE et QRLX(Q/E)E(I/E)E de
fonction inconnue (Lin et al., 1998). Caractéristique de la famille des lectines, le domaine P
(riche en prolines) de CRT contient trois motifs hélice-boucle-hélice (Michalak et al., 1996)
et est semblable (36%) aux mains EF de Calnuc (Lin et al., 1998) alors que la région Nterminale présente moins de similarités (26%) (Lin et al., 1998). Par contre, plusieurs
motifs d'importance de CRT sont absents chez Calnuc comme les trois répétitions NPD/E,
le motif KPEDWD typique de la famille Calnexine/Calréticuline (Nash et al., 1994) et le
motif KDEL de rétention au RE expliquant leur divergence de localisation (Lin et al.,
1998). Il est à noter que CRT peut également être sécrétée par les odontoblastes (Somogyi
7
et al., 2003) et inhiberait la minéralisation (St-Arnaud et al., 1995). Comme nous le vérrons
ultérieurement, Calnuc aurrait également une fonction au niveau des os.
1.3.3 NUCB2
En 1994, le groupe de Hilschmann a identifié une protéine de 55 kDa hautement
homologue à Calnuc et possédant 61,56% d'identité (Barnikol-Watanabe et al., 1994;
Karabinos et al., 1996). Nommée alors DNA binding, EF-hand, acidic region (NEFA),
cette dernière fut rebaptisée NUCB2 étant probablement un isoforme de Calnuc (NUCB1).
Une analyse rapide par le programme 'Lalign' confirme 61,4% d'identité et 81,9% de
similarité sur 415 acides aminés des séquences humaines (figure 2A). Les deux protéines
coexistent uniquement chez les organismes supérieurs et sont issues de loci géniques
différents (19q13.2-q13.4 et 11p15.1-p14) suggérant qu'elles ont évolué à partir d'un gène
ancestral commun modifié au travers l’évolution. En effet, les protéines à mains EF sont
formées à partir d'un précurseur eucaryotique à un domaine nommé CTER (Nakayama et
al., 1992). Ce dernier s'est ensuite transformé en un dérivé à quatre domaines duquel, selon
l'analyse phylogénétique, originent Calnuc et NUCB2 (Karabinos et al., 1996). Il serait
responsable de la présence de domaines fonctionnels communs dont le SP, le NLS, le LZ,
la région acide et évidemment les mains EF (figure 2A). Chez NUCB2, ces dernières lient
le calcium avec des kd de 0,08 μM et 0,2 μM permettant une capacité deux fois supérieure
à Calnuc (2 μmol Ca2+/μmol protéine) et une augmentation quatre fois supérieure (13%) du
contenu hélicoïdal (Kroll et al., 1999). Les deux protéines sont divergentes au niveau des
régions N-terminale et C-terminal ainsi qu'au niveau du LZ, mais la principale différence
réside dans le fait que, comparée à NUCB2, Calnuc a une extension d'environ 40 résidus
(figure 2A).
Les variations entre Calnuc et NUCB2 ne sont pas sans rappeler que les deux
protéines ont acquis, au travers leurs 830 millions d'années d'évolution indépendante (Gu,
1998), des différences induisant l'apparition de fonctions distinctes. D'ailleurs, jusqu'à
présent les études portant sur NUCB2 mettent en lumière des rôles très divergents de ceux
attribués à Calnuc. Entre autres, il a été montré que l'association, dépendante du calcium, de
8
NUCB2 avec le Aminopeptidase regulator of TNRF1 shedding (ARTS-1), au niveau des
vésicules exosome-like, est nécessaire pour promouvoir la relâche extracellulaire du Tumor
necrosis factor (TNF) receptor 1 (TNFR1) (Islam et al., 2006). Ce dernier peut alors
moduler l'effet de la cytokine TNF dans une foule de processus tels que l'inflammation, les
fonctions immunitaires et l'apoptose, ainsi que dans les pathologies associées comme
l'arthrite, le cancer et la sclérose en plaque.
Pratiquement au même moment, NUCB2 a été identifiée comme protéine régulée par
le récepteur nucléaire Peroxisome proliferator-activated receptor gamma (PPAR-g) qui
contrôle l'homéostasie du glucose et des lipides de même que la prise de nourriture (Oh et
al., 2006). NUCB2 agit d'ailleurs comme suppresseur de l'appétit. La présence de plusieurs
fragments de NUCB2 ainsi que de sites de clivages conservés pour les prohormones
convertases (PC) a indiqué aux auteurs de l'étude que NUCB2 pourrait être à l'origine de
plusieurs peptides bioactifs (Oh et al., 2006). Basé sur le motif consensus R/K–Xn–R/K↓
(où X est un acide aminé, n est le nombre de résidus espaceurs (soit 0, 2, 4 ou 6) et ↓ est le
site de clivage) (Seidah and Chretien, 1999), NUCB2 serait divisé en trois fragments.
Suivant la nouvelle fonction de NUCB2, ils ont été nommés NEFA/NUCB2-encoded
satiety- and fat-influencing protein (Nesfatin)-1 (résidus 1-82), Nesfatin-2 (résidus 85-163)
et Nesfatin-3 (résidus 166-396) (figure 2B) (Oh et al., 2006). Nesfatin-1 a été montré
comme étant responsable de l'effet anorexigénique et son clivage, vraisemblablement par
PC1/3 et PC2, est nécessaire à cette fonction (Oh et al., 2006). En effet, l'injection centrale
(intracérébroventriculaire) ou périphérique de Nesfatin-1 réduit de façon significative la
prise de nourriture chez les rongeurs (Goebel et al., 2011; Oh et al., 2006; Stengel et al.,
2010) alors que le jeûne diminue l'expression de l'acide ribonucléique messager (ARNm)
de NUCB2 dans les glandes endocrines gastriques (Stengel et al., 2009). Le mécanisme qui
sous-tend l'effet anorexigénique demeure nébuleux et serait indépendant de la voie de la
Leptine (Oh et al., 2006; Shimizu et al., 2009), dépendant de celle de la Mélanocortine (Oh
et al., 2006) et potentiellement médié par celles de l'Oxytocine (Maejima et al., 2009) et de
la Corticolibérine (CRH). Actuellement, Nesfatin-1 est l'objet d'un engouement pour son
potentiel dans le traitement de l'obésité. Son fragment central de 30 résidus (M30) (figure
9
2B), responsable de l'effet anorectique (Shimizu et al., 2009), constitue un modèle pour la
conception rationnelle d'analogues thérapeutiques. Nesfatin-1 aurait également un effet
anti-hyperglycémique insulino-dépendant chez les souris obèses hyperglycémiques (Su et
al., 2010) indiquant un potentiel rôle dans la pathogenèse du diabète. L'expression de
Nesfatin-1 dans les tissus adipeux est augmentée par une diète induisant l'obésité alors que
la privation de nourriture a l'effet contraire (Ramanjaneya et al., 2010). On observe aussi
une diminution des niveaux plasmatique Nesfatin-1 chez les patients à jeun atteints de
diabète de type 2 (Li et al., 2010). De plus amples études seront nécessaires pour
comprendre la fonction de Nesfatin-1 dans cette maladie. Les voies responsables du
contrôle de la masse corporelle sont généralement impliquées dans la puberté et la fertilité.
Ces phénomènes ont rapidement été associés à Nesfatin-1 qui est exprimée dans les
gonades (Garcia-Galiano et al., 2012). Dans ce sens, l'expression hypothalamique de
NUCB2/nesfatin-1 augmente pendant la puberté et sa diminution retarde les signes de
puberté et diminue les niveaux circulant de LH chez les rats femelles (Garcia-Galiano et al.,
2010; Garcia-Galiano and Tena-Sempere, 2013).
Nesfatin-1 fait également partie des neuropeptides qui participent à la création de la
réponse au stress (Emmerzaal and Kozicz, ; Merali et al., 2008). L'injection centrale de
Nesfatin-1 entraîne une élévation des niveaux circulant de composantes majeures de l'axe
hypothalamo-hypophyso-surrénalien,
soit
l'axe
du
stress,
comme
l'hormone
Adrénocorticotropine (ACTH) et la Corticostérone (Konczol et al., 2010; Yoshida et al.,
2010). On observe également une modulation de la réponse cardiovasculaire, dont de
l'hypertension, via la voie Oxytocine-Mélanocortine (Yosten and Samson, 2009; Yosten
and Samson, 2010; Yosten and Samson, 2013; Yosten and Samson, 2014).
Tout comme Calnuc, NUCB2 est localisée au niveau extracellulaire et intracellulaire,
dans le cytosol et au Golgi (Lavoie et al., 2002; Morel-Huaux et al., 2002). Il a été proposé
que, tout comme Calnuc (voir section 'Calcium et minéralisation'), NUCB2 formerait une
réserve de calcium mobilisable par agoniste (Kalnina et al., 2009). De son côté, Nesfatin-1
est dépourvu de mains EF, mais serait tout de même impliqué dans l'homéostasie calcique
en induisant une augmentation intracellulaire de l'ion via les canaux de type L, N ou P/Q
10
Figure 2 - Comparaison entre Calnuc (NUCB1) et NUCB2.
(A) Alignement des séquences humaines de NUCB1 et NUCB2 marquant le degré de
conservation des acides aminés selon l'échelle allant du rouge au bleu, où rouge (10)
11
représente une forte conservation et bleu (0) une faible conservation. Figure crée avec
PRALINE multiple sequence alignment (http://www.ibi.vu.nl/programs/pralinewww). (B)
Représentations à l'échelle de NUCB1, tel que présenté en figure 1, et de NUCB2 avec
ses différents fragments (incluant le SP). Les domaines de Nesfatin-1 sont également
représentés. Figure crée avec DOG 2.0, DOG 1.0 : Illustrator of Protein Domain
Structures. Ren et al. Cell Research (2009) 19:271-273.
(Brailoiu et al., 2007). Cet effet est bloqué par la toxine pertussique indiquant qu'un
récepteur couplé aux protéines Gi/o serait également impliqué. GPCR12, GPCR3 et
GPCR6 sont les principaux candidats (Osei-Hyiaman, 2011), mais jusqu'à présent Nesfatin1 demeure considérée comme un peptide bioactif orphelin.
Récemment, le groupe d'Unniappan, a démontré que Calnuc contient potentiellement
un peptide de 77 acides aminés hautement similaire à Nesfatin-1 et surtout, à la région
biologiquement active M30 (identité 76,6%) (Ramesh et al., 2015). Ce dernier, nommé
Nesfatin-1-like peptide (NLP), est localisé après le SP et est flanqué de sites potentiels de
clivage par des convertases de prohormones (sites Lys-Arg) (Ramesh et al., 2015). Des
expériences de transcription inverse combinées à des réactions en chaîne par polymérase
(RT-PCR) et des expériences d'immunoréactivités ont révélé que le pancréas, et plus
particulièrement les cellules bêta productrices d'insuline dans les îlots de Langerhans, sont
des sources de Calnuc et possiblement de NLP (Ramesh et al., 2015; Williams et al., 2014).
De plus, l'incubation de ces cellules avec du NLP synthétique (10 et 100 nM) démontre une
activité insulinotropique à forte concentration de glucose (Ramesh et al., 2015). Cette étude
laisse croire que les deux Nucléobindin sont des précurseurs de peptides biologiquement
actifs.
1.4
Partenaires d’interaction, fonctions et maladies associées
Calnuc est une protéine multicompartimentale ubiquitaire hautement conservée
indiquant qu’elle doit être d’une grande importance pour le maintien des fonctions
cellulaires. De plus, elle présente une structure complexe composée de plusieurs domaines
modulaires qui constituent des sites d’interactions potentiels pour d’autres protéines. Au
travers les années, l’étude de ses différentes composantes et la découverte de partenaires
12
Figure 3 - Distribution de Calnuc et de ses divers partenaires d'interaction.
(A) Calnuc, représentée par les points noirs, se localise dans les différents compartiments
de la voie de biosynthèse de même qu'au niveau du cytosol et du milieu extracellulaire. À
chaque endroit, (B) Calnuc interagit avec différents partenaires. Modifié de Calnuc :
Emerging Roles in Calcium Signaling and Human Diseases. Aradhyam et al. IUBMB Life,
62(6): 436–446, June 2010 avec la permission de John Wiley and Sons. (C) Représentation
à l'échelle de Calnuc (NUCB1), tel que présenté en figure 1, et des sites d'interactions de
13
ses partenaires. Casp (Caspase), DGLDP (site de clivage par les caspases), D328-H339S378 (triade catalytique attribuée à l'activité de protéase à sérine), GBA (G alpha binding
and activating), GEF (facteur d’échange des nucléotides de guanine). Figure crée avec
DOG 2.0, DOG 1.0 : Illustrator of Protein Domain Structures. Ren et al. Cell Research
(2009) 19:271-273.
d’interactions variés ont permis d’associer Calnuc à plusieurs fonctions. Bien que ces rôles
restent à être approfondis, ils laissent supposer que la préservation de l’intégrité structurale
et fonctionnelle de la protéine est nécessaire pour nous prévaloir de multiples pathologies.
1.4.1 Calcium et minéralisation
Le calcium est un élément chimique omniprésent essentiel au bon fonctionnement
d'une variété de fonctions biologiques. Il contribue entre autres à la formation osseuse et au
maintien du potentiel d'action membranaire tout en agissant comme cofacteur enzymatique
et comme second messager dans la transduction de signaux. Au niveau cellulaire,
l'efficacité du calcium repose sur sa capacité de lier de nombreuses protéines dont la
principale famille est celle des protéines à mains EF. Ces dernières assurent sa
compartimentation et sa spécificité d'action agissant d'une part comme tamponneur dans
l'homéostasie calcique ou, d'autre part, comme senseur dans la traduction de signaux (da
Silva and Reinach, 1991). Les tamponneurs, comme Calbindin D9k et Parvalbumin,
chélatent les ions grâce à une haute affinité (Kd <10-7 M) et subissent peu de changements
conformationnels (Ikura, 1996). De leur côté, malgré une affinité relativement faible pour
le calcium (Kd >10-5 M), les senseurs, comme Calmodulin et Troponin C, subissent des
changements conformationnels marqués créant des domaines d’interaction pour une cible à
moduler (Ikura, 1996). À l'image de Calbindin D28k (Berggard et al., 2002), Calnuc a été
proposée comme étant à la fois un senseur et un tamponneur de calcium (de Alba and
Tjandra, 2004; Kanuru et al., 2009) allant de pair avec sa localisation au Golgi et au
cytoplasme (figure 3A et 3C).
Calnuc comme senseur
La fraction cytosolique de Calnuc joue probablement un rôle de senseur permettant la
régulation de nombreuses protéines en réponse à une relâche de calcique. Son affinité pour
14
le Ca2+, ainsi que sa sélectivité pour l'ion au détriment du Mg2+, jouent en cette faveur. Lors
d'une activation cellulaire, telle une contraction musculaire menant à l'ouverture des canaux
Ryanodine, la concentration de calcium dans le cytoplasme passe d'environ 0,1 uM à 1-10
uM (Berggard et al., 2002) entraînant la liaison de l'ion par Calnuc. Cette liaison provoque
des changements conformationnels importants dont le repliement des mains EF ainsi que
l'exposition des surfaces hydrophobes pouvant servir de sites d'interaction (de Alba and
Tjandra, 2004). De multiples interactions décrites plus bas dans cette section sont d'ailleurs
influencées par la liaison du calcium laissant croire que Calnuc pourrait agir comme
effecteur ou modulateur dans la signalisation.
Calnuc comme tamponneur
Le calcium est nécessaire pour la cellule mais devient toxique à haute concentration.
Il doit donc être emmagasiné dans des compartiments comme le réticulum endoplasmique
et le Golgi (Grohovaz et al., 1996) de manière à être rapidement mobilisé au cytosol suivant
un stimulus. Ces réserves sont maintenues par des protéines liant le calcium tel que
Calréticuline au RE. À l'intérieur du Golgi, Calnuc est la principale protéine liant le
calcium (3,8 mg Calnuc/mg protéines dans les cellules NRK, soit 0,4% des protéines) (Lin
et al., 1998). En effet, la surexpression de Calnuc au Golgi mène à une augmentation de
deux à trois fois de la quantité de calcium emmagasiné et pouvant être mobilisée par
l'action d'agonistes ciblant le récepteur IP3 de type 1, comme l'ATP et l'IP3 (Lin et al.,
1999). Calnuc agirait donc comme tamponneur à cet endroit grâce à sa main EF1 ainsi qu'à
l'action de la pompe Sarco/endoplasmic reticulum Ca2+-ATPase (SERCA) qui assure le
mouvement des ions vers la lumière du Golgi (Taylor et al., 1997).
Calnuc dans la minéralisation
Le tissu osseux forme une réserve calcique d'un tout autre type. En 1995, Calnuc a été
identifiée comme étant une constituante mineure de la matrice extracellulaire des os
(Wendel et al., 1995), puis de la dentine (Somogyi et al., 2004). En effet, Calnuc est
produite par de nombreuses cellules impliquées dans la formation osseuse, soit les
ostéoblastes, les ostéocytes (Wendel et al., 1995) et les odontoblastes (Somogyi et al.,
2004), mais est indétectable par hybridation in situ dans les cellules connexes comme les
15
chondrocytes (cellules du cartilage), les ostéoclastes et cellules de la moelle (Petersson et
al., 2004). Calnuc est sécrétée dans l'ostéoïde, c'est-à-dire la matrice osseuse non
minéralisée essentiellement constituée de collagène de type 1. Cette dernière doit donc
subir une étape de minéralisation pendant laquelle s'ajoute une matrice minérale composée
de phosphate tricalcique (hydroxyapatite), de carbonates de calcium et de phosphates de
magnésium. Suivant sa capacité à être sécrétée et à lier l'hydroxyapatite (Wendel et al.,
1995), l'implication de Calnuc dans ce procédé a été proposée. Des analyses
ultrastructurelles ont mis en évidence une forte présence de Calnuc au niveau des ostéoïdes
d'os nouvellement créés. De plus, durant la prolifération et la différenciation cellulaire,
Calnuc a été localisée au niveau du noyau alors que pendant la minéralisation Calnuc se
retrouve principalement dans le cytoplasme (Petersson et al., 2004). Ce déplacement
intracellulaire est accompagné d'une faible expression durant la phase de prolifération qui
augmente considérablement durant la différentiation et la maturation de la matrice pour
ensuite diminuer après l'initialisation de la minéralisation (Petersson et al., 2004). Ces
données suggèrent que Calnuc contribuerait à l'accumulation et au transport des ions
calciques et pourrait agir comme modulateur de la maturation avant le dépôt de minéraux.
Il est à noter que Calnuc pourrait aussi agir indirectement dans la destruction de l'os via son
interaction avec COX-2 qui a pour effet de stimuler la synthèse de PGE2 et la formation
d'ostéoclastes.
1.4.2 Protéines G et sécrétion régulée
Les Guanosine nucleotide-binding proteins (protéines G), sont d’importants
messagers qui transmettent des signaux provenant de récepteurs couplés aux protéines G
(GPCR) de la membrane plasmique vers l’intérieur de la cellule. Pour ce faire, le récepteur
activé par un ligand catalyse l’échange de guanosine diphosphate (GDP) pour du guanosine
triphosphate (GTP) sur la sous-unité Gce qui l'active à son tour. À ce moment, les sousunités Get G associées au récepteur se dissocient pour aller moduler indépendemment
une variété d’effecteurs différents. Éventuellement, l'activité GTPase intrinsèque de
Gprovoque son inactivation, via l’hydrolyse du GTP, et conséquemment sa réassociation
16
avec le récepteur. Récemment, il a été mis en évidence que les protéines G, ainsi que le
dimère G, sont présentes à la membrane de compartiments intracellulaires, comme le
Golgi et les endosomes (Wilson et al., 1994), mais leur rôle à ce niveau demeure encore
inconnu.
En 1995, une expérience de double hybride a permis au groupe d’Insel d’identifier
Calnuc comme partenaire d’interaction direct des protéines Gde type Gi2 (Mochizuki et
al., 1995). Ultérieurement, Calnuc a été montrée comme étant capable d’interagir, de façon
dépendante du Ca2+ et Mg2+ (Lin et al., 2000), avec les types Gi1 et Gi3, ainsi qu’avec
les Gi2, Go1, Gz et Gs, mais pas avec les Gq, G12 et G13 (figure 3B) (Lin et al.,
1998). L’étude plus approfondie de la liaison avec Gi3 a permis d’établir que les mains
EF de Calnuc, ainsi que la région acide très conservée et flanquée entre ces structures, sont
essentielles pour l’interaction (figure 3C) (Lin et al., 1998). Les premières expériences
visant à établir la fonction de ces interactions ont montré que la surexpression de Calnuc
augmente les niveaux de Gi2 (Mochizuki et al., 1995). À ce moment, l’interaction a été
montrée comme étant indépendante de l’état d'activation de la protéine G (Lin et al., 1998).
Toutefois, une protéine G active augmente la liaison de Ca2+ sur Calnuc alors qu’une
protéine G inactive a pour effet d’entraîner une relâche de l’ion par Calnuc (Kanuru et al.,
2009). Ce phénomène laisse croire que Calnuc pourrait être agir comme effecteur dans la
signalisation des protéines G ou dans la modulation de leurs fonctions.
Outre leur localisation à la membrane plasmique, les protéines Gi3 sont retrouvées à
la surface du Golgi (Stow et al., 1991) et les Gi1/2 à la surface des vésicules de sécrétion
(Wilson et al., 1994). Calnuc a également été observée à ces différents endroits (Lavoie et
al., 2002) et est partiellement distribuée avec Gi1/2 et Gi3 à la surface des granules de
sécrétion (Lin et al., 2009). Il s’agit également de la seule protéine ayant la capacité de lier
à la fois les G et le calcium, deux éléments qui stimulent la sécrétion régulée. Son
implication dans ce processus physiologique a donc rapidement été confirmée, dans les
cellules neuroendocrines, par une augmentation de la sécrétion régulée d’ACTH
dépendante des G (Lin et al., 2009). La surexpression de Calnuc entraîne aussi une
17
redistribution de Gi3 sur les granules de sécrétion et de Gi1/2 à la membrane plasmique
sans affecter leur activité. Ces résultats indiquent que Calnuc influencerait la sécrétion
régulée en agissant comme modulateur, possiblement sensible au calcium, dans la
dynamique de distribution intracellulaire des protéines G.
La signalisation des protéines G est dépendante de leur cycle d’activation-inactivation
lui-même finement régulée par de nombreuses protéines accessoires. Les facteurs
d’échange des nucléotides de guanine (GEF) catalysent l’activation des protéines G via
l’échange du GDP pour du GTP. De leur côté, les protéines d’accélération de la GTPase
(GAP), dépourvues d’activité catalytique, stimulent l’activité GTPase intrinsèque des
protéines G pour mener à leur inactivation. Finalement, les inhibiteurs de la séparation des
nucléotides de guanine (GDI) maintiennent les protéines G dans un état d’inactivation.
Dernièrement, deux études ont suggéré l’implication de Calnuc dans cette mécanistique en
lui attribuant les rôles, quelque peu opposés, de GDI et de GEF. En 2010, le groupe de
Sakmar a montré que la forme cytosolique de Calnuc est un dimère physiologique qui peut
interagir avec Gαi1-GDP, suivant une affinité de 18,3 μM, mais pas avec Gαi1-GTP
(Kapoor et al., 2010). Calnuc a pour effet d’inhiber la relâche du GDP de Gαi1, prévenant
ainsi l’activation de la protéine G et l’inhibition de la voie de signalisation subjacente
(Kapoor et al., 2010). Ces résultats suggèrent que Calnuc agirait comme une GDI malgré
l’absence dans sa structure d’un motif GoLoco normalement associé à cette fonction. La
région N-terminale de Calnuc (26-333) serait plutôt en cause (Kapoor et al., 2010).
Pratiquement au même moment, le groupe de Farquhar identifiait un motif G alpha binding
and activating (GBA) (figure 3C) attribué à l’activité GEF dans plusieurs protéines, dont la
G alpha-interacting vesicle-associated protein (GIV) (Garcia-Marcos et al., 2011). Ce
motif Ψ-S/T-Φ/Ψ-X-D/E-F-Ψ (où Ψ est un résidu aliphatique et Φ est un résidu
aromatique) se trouve conservé dans Calnuc et NUCB2 et leur permet de lier
préférentiellement la forme inactive des Gi au niveau de la crevasse entre l’hélice alpha3
et la région Switch II (Garcia-Marcos et al., 2011). Ceci contredit une étude du même
groupe qui impliquait l’hélice alpha5 de Gi3 (Lin et al., 2000), une région d’importance
impliquée dans la liaison de GDP (Mixon et al., 1995; Sprang, 1997), de GPCR (Lambright
18
et al., 1996) et d’effecteurs (Sunahara et al., 1997). Néanmoins, Gi3 lie davantage de GTP
en présence de Calnuc ou NUCB2 indiquant que ces dernières agiraient comme GEF
(Garcia-Marcos et al., 2011). La divergence des fonctions apportées par ces articles peut
être imputable à l’utilisation de concentrations protéiques différentes (sous les Kd ou
affectant les réactions enzymatiques) ou à des lectures artéfactuelles. Par contre, les deux
groupes s’entendent pour dire que Calnuc lie uniquement les formes inactives des protéines
Get que le calcium abolit l’interaction. En effet, l’ion entraîne le repliement des mains EF
réquisitionnant et dissimulant des résidus nécessaires pour l’interaction avec G (GarciaMarcos et al., 2011). D’ailleurs, l’interaction entre Calnuc et Gi3 se produit dans un
milieu pauvre en calcium, soit dans le cytosol à la surface du Golgi (Weiss et al., 2001).
Calnuc est alors libre d’ions et lie Gseulement s’il est ancré aux membranes par ses
ancrages lipidiques (Weiss et al., 2001). De plus amples études seront nécessaires afin
d’éclaircir le rôle de Calnuc dans le cycle des protéines G, les régions impliquées ainsi que
l’effet de l’élévation physiologique du calcium.
1.4.3 ADN et lupus
Calnuc possède une région riche en acides aminés basiques qui lui permet de lier
l’ADN (figure 3B et 3C) (Miura et al., 1992) et une fermeture en leucines connue chez
plusieurs protéines pour faciliter cette liaison. Une hypothèse stipule que l'interaction entre
Calnuc et l'ADN serait importante pour l'induction d'anticorps contre l'ADN, un
phénomène en cause dans la physiopathologie du lupus érythémateux disséminé. Il s’agit
d’une maladie chronique qui attaque principalement les tissus conjonctifs. Le système
immunitaire produit alors des anticorps spécifiques contre des entités normalement
présentes dans l'organisme dans le but de les éliminer. À l’origine, Calnuc a été identifiée
comme étant un facteur promouvant la formation de tels auto-anticorps ciblant l’ADN
double brin, la Ribonucléoprotéine U1, la protéine Lupus La et le facteur rhumatoïde
(Kanai et al., 1995b; Kanai et al., 1993). D’ailleurs, l’injection intrapéritonéale de Calnuc
cause des symptômes associés au lupus dont une surproduction d’immunoglobuline G
(IgG) suivie par l'élévation d'auto-anticorps de même type ce qui accélère la réponse auto19
immune (Kanai et al., 1995b). Les souris injectées présentent aussi une fragmentation
d’ADN liée à de l’apoptose dans des tissus lymphoïdes comme le thymus et entraînant une
accumulation de segments d’ADN dans le sérum. Ce phénomène pourrait être à l’origine de
la production d’anticorps dirigés contre l’ADN et de l’auto-immunité (Kanai et al., 1995a).
Des études mécanistiques plus approfondies devront être faites pour comprendre les
relations de causalité et élucider le rôle nébuleux de Calnuc dans cette maladie.
1.4.4 Cancer, apoptose et Caspases
Le cancer est une maladie caractérisée par une prolifération cellulaire incontrôlée en
dépit
d'anomalies
génétiques.
Le
système
immunitaire
peut
détecter
certains
dysfonctionnements cellulaires associés aux tumeurs comme étant immunogéniques.
Plusieurs protéines impliquées dans la tumorigenèse servent alors d’antigènes pour la
production d’auto-anticorps ce qui n’est pas sans rappeler l’induction d’auto-anticorps par
Calnuc dans un contexte de lupus. Dans cette logique, plusieurs groupes ont étudié
l’expression antigénique de Calnuc. Dès 1994, une augmentation de Calnuc est décelée
dans 10% des adénocarcinomes gastriques et 56% de ceux présentant des métastases aux
ganglions lymphatiques (Wang et al., 1994). Également, le niveau d’expression de Calnuc
dans les lymphomes non hodgkiniens reflète le degré de prolifération des cellules et donc le
grade de la pathologie (Kubota et al., 1998). Aussi, Calnuc est hautement spécifique pour le
cancer du côlon puisqu’environ 60% des tissus affectés surexpriment cette protéine en
comparaison aux côlons sains qui ne semblent pas l'exprimer en immunohistochimie (Chen
et al., 2007). Dans ce même article de 2007, des auto-anticorps contre Calnuc ont été
retrouvés dans 11,5% des cancers du côlon contre seulement 1,2% des côlons sains. Calnuc
semble donc bel et bien agir comme un antigène associé aux tumeurs (TAA) et contribue
favorablement aux outils diagnostiques. De plus amples études seront nécessaires pour
établir la fonction exacte de Calnuc et afin de déterminer si la réaction immunitaire est en
cause dans le développement du caractère malin de la maladie. En effet, en raison de sa
capacité à lier le calcium et les protéines G, Calnuc perturberait l’adhésion et la motilité
cellulaire (Mermelstein et al., 2003) de même que la croissance, la prolifération et la
migration. Pour les mêmes raisons, Calnuc pourrait être impliquée dans le mécanisme
20
d'action d'autres protéines associées à des cancers. Entre autres, par double hybride, Calnuc
a été identifiée comme partenaire d'interaction de la protéine peu caractérisée nommée
Breast cancer membrane protein (BCMP84) (Adam et al., 2003). Cette dernière est
surexprimée et est déplacée à la membrane plasmique dans les carcinomes du sein et de la
vessie.
Le cancer est induit par le blocage de l'apoptose, le processus de mort physiologique
par lequel une cellule déclenche son auto-destruction. Ce mécanisme mène à l’activation de
multiples Caspases, des protéases à cystéine qui clivent nombre de substrats au niveau de
sites consensus spécifiques pour entraîner le démantèlement cellulaire. L’expression de
Calnuc dans certains cancers de même que l’induction de l’apoptose dans les tissus
lymphoïdes lors de son administration exogène (Kanai et al., 1995b) suggèrent
l’implication de la protéine à ce niveau. Dans le même ordre d'idées, in vitro, NUCB1 a été
identifiée comme substrat potentiel pour les Caspases 3, 6 et 8 alors que son homologue
NUCB2 serait clivé par Caspase 3, 6, 7, 8 et 9 (Valencia et al., 2008). Un site général de
clivage DXXD est hautement conservé à travers les espèces. Chez la forme humaine de
Calnuc, ce motif non optimal DGLDP (figure 3C) est localisé entre le NLS et la première
main EF suggérant que les Caspases pourraient découpler les domaines fonctionnels en
situation d’apoptose. En effet, le fragment C-terminal comprend les mains EF et la glissière
en leucines qui sont essentielles pour la liaison du calcium et la liaison à des partenaires,
dont l’ADN. Par contre, il est dépourvu de son domaine riche en leucines et isoleucines
ainsi que de son NLS, ce qui signifie qu’il n’est pas retenu au Golgi et qu’il ne transloque
pas au noyau. De son côté, le fragment N-terminal contenant les domaines de liaison au
zinc est rapidement dégradé.
1.4.5 COX et inflammation
Les Cyclooxygénases (COX) sont des enzymes qui permettent la synthèse de
prostanoïdes, à partir d’acide arachidonique (AA) de manière à moduler divers
effets physiologiques. Alors que la COX-1 est constitutive, la COX-2 est inductible par de
multiples facteurs pro-inflammatoires dont le Granulocyte/monocyte colony stimulation
21
factor (GM) et le TNF. En 1996, une étude a démontré que Calnuc peut interagir avec les
COX, de manière indépendante du calcium, via une région qui comprend ses 123 résidus en
N-terminal (figure 3B et 3C) (Ballif et al., 1996). Malgré une affinité plus forte pour la
COX-1 que la COX-2, les deux enzymes, fortement liées à la membrane luminale du RE,
diminuent de plus de 80% la sécrétion de Calnuc en condition de surexpression. Elles
participeraient ainsi à la rétention intracellulaire de Calnuc (Ballif et al., 1996). Pour faire
suite, un groupe québécois s’est concentré sur la COX-2, qui prédomine dans la production
de prostaglandines (PG) E2 dans les cellules inflammatoires telles que les neutrophiles. Ils
ont démontré que Calnuc interagit et colocalise avec la COX-2 au niveau du RE et du Golgi
dans des neutrophiles stimulés au GM/TNF (Leclerc et al., 2008). Calnuc se retrouve
également dans les mêmes fractions que les enzymes impliquées dans la génération de
prostanoïdes (Microsomal PGE2-synthase-1, Thromboxane-synthase, Calcium-dependent
phospholipase A2) (Leclerc et al., 2008). À ce sujet, des essais sur neutrophiles stimulés ont
démontré que Calnuc recombinante augmente de 5 fois la synthèse de PGE2 via la COX-2
et de manière dépendante de la dose (Leclerc et al., 2008). Calnuc présente donc des
propriétés régulatrices des COX durant l’inflammation alors que ces dernières jouent un
rôle dans la rétention intracellulaire de Calnuc.
Dans le même ordre d'idées, en 2015, Calnuc a été identifiée comme protéine liant les
lipides (Niphakis et al., 2015). Plus de 1000 protéines, dont plusieurs transporteurs,
enzymes et protéines peu caractérisées, ont été enrichies avec une sonde semblable à l'acide
arachidonique (arachidonoyl) (Niphakis et al., 2015). Il a été montré que le lipide se lie à
Calnuc au niveau du domaine d'interaction des COX et que le calcium augmente
significativement et sélectivement l'interaction (Niphakis et al., 2015). Calnuc perturbe le
métabolisme oxydatif et hydrolytique des endocannabinoïdes et des eicosanoïdes. En effet,
l'utilisation de deux ligands sélectifs de Calnuc (MJN228 et KML110) ou de petits acides
ribonucléiques (ARN) en épingle à cheveux (shRNA) dirigés contre Calnuc a pour effet
d'élever la concentration cellulaire d'acides gras (Niphakis et al., 2015). Ces résultats
tendent à démonter que Calnuc faciliterait le métabolisme des acides gras en les transférant
à des enzymes comme l'Amidohydrolase des acides gras (FAAH) et la COX2 (Niphakis et
al., 2015).
22
1.4.6 Activité de protéase à sérine
En 2012, un groupe indien a démontré, par essais enzymatiques, que Calnuc possède
une activité de protéase à sérine (Kanuru et al., 2013). Cette activité est attribuable à une
triade catalytique en C-terminal composée de l’aspartate328 du motif DTG (séquence
consensus de protéases aspartiques (Rawlings and Barrett, 1995)), de l’histidine339 et de la
sérine378 du motif GXSXG (séquence consensus de lipases (Wagner et al., 2002)) (figure
3C). En utilisant une variété de substrats différents, l’équipe a établi que Calnuc, à l’image
de la trypsine, clive en aval d’une arginine. Aussi, l’interaction avec la sous-unité alpha des
protéines G à proximité de la sérine catalytique a pour effet de réduire l’activité
protéolytique. La liaison allostérique du zinc produit le même effet inhibiteur possiblement
en nuisant à la géométrie du site actif. Cette étude suggère une grande importance des ions
dans la régulation des fonctions de Calnuc qui existerait, de manière dynamique, entre
autres comme protéase à sérine.
1.4.7 ATF6 et réponse au stress réticulaire
La réponse au stress lié à l'accumulation de protéines mal repliées dans la lumière du
réticulum endoplasmique (ou UPR, unfold protein response) est un mécanisme activé pour
restaurer les fonctions normales de la cellule. Elle agit de façon à mettre fin à la traduction
des protéines, à dégrader les protéines mal repliées et à augmenter la production de
chaperonnes moléculaires. L'Activating transcription factor 6 (ATF6) est alors transporté
du réticulum endoplasmique au Golgi pour être clivé par la Site-1-protease (S1P) et S2P
(Ye et al., 2000). Alors sous sa forme courte, p50ATF6 transloque au noyau pour lier et
activer des éléments de réponse au stress réticulaire (ERSE) au niveau de promoteur de
gènes cibles (Haze et al., 1999). En 2007, il a été démontré que le promoteur de Calnuc
contient des séquences telles que ERSEII et UPR element-like (UPRE-like) qui permettent
son induction par ATF6 (Tsukumo et al., 2007). Calnuc agit alors comme rétrorépresseur
en situation de stress réticulaire. En effet, elle inhibe le clivage d’ATF6 (figure 3B) en
réduisant indirectement son interaction avec S1P de façon à prévenir la translocation au
noyau.
23
1.4.8 Necdin
La Neurally differentiated embryonal carcinoma-derived protein (Necdin) est un
suppresseur de croissance exprimé de façon ubiquitaire dans les tissus humains (Jay et al.,
1997), mais principalement dans les neurones postmitotiques. Au niveau du noyau de ces
cellules (Maruyama et al., 1991), Necdin maintiendrait l'état postmitotique en interagissant
avec les facteurs des transcriptions p53 et E2F1 de manière à réprimer leur action (Taniura
et al., 1999). Necdin interagit aussi avec des oncoprotéines virales comme l'Adenovirus
early region 1A protein (E1A) et le Simian Vacuolating Virus 40 large T antigen (SV40TAg) (Taniura et al., 1999) lui permettant d'exercer un contrôle négatif sur le cycle
cellulaire via la Retinoblastoma protein (pRB). Outre sa localisation nucléaire, Necdin est
aussi présente au niveau du cytosol (Niinobe et al., 2000) et est sécrétée (BarnikolWatanabe et al., 1994) ce qui suggère d'autres fonctions. En l'an 2000, un groupe a identifié
Calnuc ainsi que NUCB2 comme partenaires d'interaction pour Necdin (figure 3B)
(Taniguchi et al., 2000). À cause de la forte homologie entre NUCB2 et Calnuc, il a été
proposé que les mêmes régions d'interaction et les mêmes fonctions soient attribuées à
Calnuc dans le contexte de Necdin (Taniguchi et al., 2000). Il a été démontré que NUCB2
lie la région centrale de Necdin (102-325), nécessaire aux interactions avec E2F1, p53 et
SV4-TAg laissant croire que NUCB2 pourrait séquestrer Necdin l'empêchant de lier les
facteurs de transcription. Du côté de NUCB2, l'interaction est attribuée à la région Cterminale qui englobe à la fois la région acide et les mains EF (souris 214–358) (figure 3C),
mais elle est indépendante du calcium (Taniguchi et al., 2000). Necdin augmente la
rétention intracellulaire de NUCB2 et bloque la sécrétion dans le milieu extracellulaire
(Taniguchi et al., 2000). Puisque NUCB2 lie le calcium et est principalement localisée au
RE, les auteurs ont investigué les relâches calciques de l'organite par stimulation des
récepteurs Ryanodine à la caféine. La sortie de calcium est augmentée par la surexpression
de NUCB2 et cet effet est exacerbé par la coexpression de Necdin indiquant un rôle dans
l'homéostasie calcique (Taniguchi et al., 2000). Necdin a donc un effet modulatoire sur la
distribution et la fonction de NUCB2 ce qui permet de spéculer que Calnuc, semblable à
NUCB2, pourrait être affecté de façon similaire.
24
1.4.9 APP et Alzheimer
L’Alzheimer est une maladie neurodégénérative qui entraîne des pertes de fonctions
mentales irréversibles. Au point de vue mécanistique, la pathologie est caractérisée par un
dysfonctionnement cellulaire résultant de l’accumulation de deux protéines. Au niveau
intracellulaire, la protéine Tau s’agglutine en neurofibrilles et, au niveau extracellulaire, le
peptide β-amyloïde (Aβ) forme des plaques amyloïdes. Ce dernier est issu du clivage
anormal du précurseur de la protéine précurseur de la β-amyloïde (APP). Plusieurs facteurs
impliqués dans ces processus participeraient également à un déséquilibre de l’homéostasie
calcique menant à la dysfonction synaptique et à l’apoptose (LaFerla, 2002; Mattson and
Chan, 2003; Yang and Cook, 2004). Quoique mécompris, le maintien des niveaux de zinc
serait aussi primordial pour se prévaloir de l’Alzheimer (Cuajungco and Lees, 1997a;
Cuajungco and Lees, 1997b). Calnuc, par sa capacité à lier à la fois le calcium et le zinc, a
logiquement été associée à la maladie. Il a été démontré que Calnuc interagit directement,
et de façon sensible au Ca2+, avec le domaine intracellulaire d’APP (AICD) (figure 3B)
(Lin et al., 2007). La population cytosolique de Calnuc serait donc impliquée lorsque
présente à la surface du Golgi où elle colocalise avec APP (Lin et al., 2007). D’ailleurs, la
surexpression de Calnuc réduit l’ARNm d’APP ce qui entraîne une diminution d’APP et de
ses produits protéolytiques (APPα, Aβ et le fragment C-terminal (CTF)) (Lin et al., 2007).
À l’opposé, la réduction de Calnuc par petits ARN interférents (siRNA) augmente la
biosynthèse d’APP (Lin et al., 2007). Calnuc affecterait donc la transcription d’APP
possiblement en séquestrant le fragment AICD l’empêchant d’agir en complexe avec
d’autres protéines (Amyloid beta A4 precursor protein-binding family B member 1 (Fe65)
et 60 kDa Tat-interactive protein (Tip60)) pour réguler l’expression de gènes incluant APP
(Cao and Sudhof, 2001; Kim et al., 2003a; von Rotz et al., 2004). Ces résultats sont
appuyés par une diminution de Calnuc dans les cerveaux de patients souffrant de la maladie
d’Alzheimer (Lin et al., 2007).
25
1.4.10 LRP10 et transport intracellulaire
En 2009, notre groupe a voulu identifier de nouveaux partenaires d'interaction pour
Calnuc dans le but d'en apprendre davantage sur ses fonctions intracellulaires. Des
expériences de double hybride chez la levure ont apporté les premières évidences d'une
interaction, qui s'est avérée directe, entre la région N-terminale de Calnuc et la queue
cytoplasmique de la Low density lipoprotein receptor (LDLR)-related protein 10 (LRP10)
(figure 3B et 3C) (Brodeur et al., 2009). Ce récepteur de type 1, aussi connu sous le nom de
LRP9, fait partie d'une sous-famille de récepteurs aux lipoprotéines de faible densité
(LDLR) qui inclue également LRP3 et LRP12. De façon générale, les LDLR sont
caractérisés par des répétitions de classe A (LDL-A) extracellulaires permettant la fixation
de lipoprotéines à internaliser dans un contexte de métabolisme des lipides. La sous-famille
de LRP10 se distingue par l'absence de répétitions EGF-like et de motifs YWTD ainsi que
par une grande queue cytoplasmique comportant un domaine riche en prolines, des
domaines Complement C1r/C1s, Uegf, Bmp1 (CUB) extracellulaires pour la liaison de
ligands, mais surtout au moins un motif acide-dileucine (DXXLL). Ce dernier motif lie des
adaptateurs de clathrine nommés Golgi-localized, gamma-ear containing Arf binding
protein (GGA) et Adaptator protein (AP) qui permettent l'entrée du récepteur dans des
vésicules de clathrine et son transport subséquent entre la membrane plasmique, le TGN et
les endosomes (Boucher et al., 2008; Doray et al., 2008). LRP10 a été proposée pour
participer à l'internalisation de l'Apolipoprotéine E (Sugiyama et al., 2000), mais sa
localisation préférentielle au TGN et aux endosomes (Boucher et al., 2008; Doray et al.,
2008) indique plutôt une fonction potentielle à ce niveau. À ce sujet, il été démontré que
LRP10 agit comme récepteur de transport, interagissant directement avec l'ectodomaine
d'APP pour la maintenir au Golgi et prévenir son clivage amyloïdogénique (Brodeur et al.,
2012). Au contraire, une diminution de LRP10 a pour effet de redistribuer APP aux
endosomes précoces et d'augmenter la production du fragment Aβ par la β-sécrétase qui s'y
trouve (Brodeur et al., 2012). Ceci n'est pas sans rappeler qu'une réduction de Calnuc
augmente la biosynthèse d’APP (Lin et al., 2007). D'ailleurs, comme c'est le cas pour
Calnuc (Lin et al., 2007), une diminution de LRP10 est observée chez les patients
souffrants de la maladie d'Alzheimer (Brodeur et al., 2012).
26
L'implication de Calnuc et de LRP10 dans l'Alzheimer supporte un rôle fonctionnel
pour leur interaction qui est indépendante du calcium (Brodeur et al., 2009). Dans la queue
cytosolique de LRP10, une région riche en arginines, qui forme un segment amphiphilique
propice à des interactions, s'est avérée importante, mais pas nécessaire, pour la liaison avec
Calnuc (Brodeur et al., 2009). Les deux protéines colocalisent au niveau du TGN et des
endosomes et la déplétion de Calnuc, par siRNA, a pour conséquence d'entraîner une
diminution des niveaux du récepteur (Brodeur et al., 2009). En effet, ce dernier n'est alors
plus recyclé vers le Golgi et est plutôt redistribué aux lysosomes où il est dégradé (Brodeur
et al., 2009). Ce phénomène peut être renversé par l'expression d'un mutant strictement
cytosolique de Calnuc, impliquant cette fraction protéique dans l'effet observé (Brodeur et
al., 2009). De la même manière, Calnuc interagit et affecte le triage de LRP3, un autre
LDLR de la même sous-famille que LRP10, qui possède aussi une région riche en arginines
(Brodeur et al., 2009). Ces travaux indiquent que Calnuc serait impliquée dans le transport
intracellulaire de LRP10 et LRP3 et laisse croire que Calnuc pourrait également affecter
d'autres récepteurs transitant entre le TGN et les endosomes.
2
Transport intracellulaire
Le transport bidirectionnel intracellulaire permet de recevoir, dissocier et trier les
cargos de la membrane plasmique ou de la voie de synthèse tout en maintenant l'identité de
chaque compartiment. La voie antérograde assure la distribution de nombreuses protéines
du RE, en passant par le Golgi, vers les lysosomes, la membrane plasmique et le milieu
extracellulaire. De son côté, la voie rétrograde permet l'internalisation de multiples
molécules ainsi que le recyclage de matériel réutilisable jusqu'au Golgi, et même jusqu'au
RE. Dans le cadre de la présente étude, seuls les mécanismes d'échanges et de régulation
entre le Golgi et le système endolysosomial seront abordés (figure 4A).
2.1
Transport antérograde
La voie de transport antérograde achemine des protéines nouvellement synthétisées
au RE vers leur lieu d'action. Leur passage à travers l'organite leur permet de se replier
27
adéquatement, d'oligomériser et d'acquérir les premières modifications telles que des
glycosylations,
des
ponts
disulfures
et
des
ancrages
lipidiques
de
type
Glycosylphosphatidyl inositol (GPI). Elles gagnent ensuite le Golgi, organite décrit en 1898
par l'histologiste italien du même nom, qui modifie davantage les protéines et qui agit
comme centre de trie et de distribution. En effet, en traversant les différentes citernes
contenant un assortiment d'enzymes variées, les protéines subissent de plus amples
altérations. Entre autres, la modification de l'arborescence de glycosylations, combinée à
une phosphorylation, engendre la formation d'un groupement mannose-6-phosphate (M6P)
(Rohrer and Kornfeld, 2001), agissant comme signal d'adressage vers le lysosome, pour
plus de 60 précurseurs d'hydrolases acides (Johannes and Wunder). Ces dernières sont
transportées, grâce aux récepteurs au M6P (MPR), vers le système endo-lysosomial pour y
subir une activation protéolytique. D'autres récepteurs, comme Sortiline et LIMP2 assurent
le transport indépendant des groupements M6P.
2.1.1 Transport dépendant de M6P - MPR
Il existe deux types de récepteurs MPR, soit le récepteur dépendant des cations
manganèse (CD-MPR) et le récepteur indépendant des cations (CI-MPR). Ces
glycoprotéines homodimériques (Dahms and Hancock, 2002) transmembranaires de type-1
sont homologues à seulement 20% et sont les seuls membres de la famille des récepteurs de
lectine de type P (Dahms and Hancock, 2002), c’est-à-dire qui ont la capacité de lier des
groupements M6P. Ils transportent des populations différentes, mais chevauchantes
d’hydrolases acides en utilisant des mécanismes similaires de reconnaissance des
carbohydrates (Roberts et al., 1998). Ainsi, CD-MPR a comme cargo la Tripeptidyl
peptidase I (TPP1), E1A-stimulated genes 1 (Creg1) et l'Héparanase (Hpse) alors que CIMPR lie préférentiellement l'α-Mannosidase B1 (Man2b1), la Cathepsine D (CTSD) et la
Prosaposine (Psap) (Qian et al., 2008). Le monomère de CD-MPR a une masse moléculaire
de 46 kDa et possède un peptide signal en N-terminal, suivit d’une région luminale de 159
acides aminés qui contient un motif liant M6P (Lobel et al., 1988). Son domaine
cytoplasmique inclut des signaux de triage, tels que des palmitoylations (Schweizer et al.,
1996) et des phosphorylations (Meresse et al., 1990; Rosorius et al., 1993), ainsi que des
28
sites d’interactions pour les protéines de transport (protéines adaptatrices et rétromères). De
son côté, CI-MPR est conservé chez les mammifères et invertébrés et il est de loin le
récepteur le mieux caractérisé de la voie endosomiale. Le monomère indépendant des
cations est beaucoup plus massif avec ses 300 kDa. Sa structure générale est la même que
celle de CD-MPR, mais sa région luminale compte 15 répétitions contigües de 147 acides
aminés séparées par des espaceurs (linkers) de 5-12 résidus (Lobel et al., 1988) qui
restreignent les arrangements moléculaires (Brown et al., 2002). On y retrouve quatre
motifs de liaison pour des groupements M6P (Olson et al., 2015), un site de liaison au
récepteur de l'Urokinase (uPAR), un site pour le Plasminogène et un site pour l'Insulin-like
growth factor-2 (IGF-2) (Garmroudi et al., 1996; Hancock et al., 2002; Marron-Terada et
al., 2000; Schmidt et al., 1995). En effet, les MPR sont retrouvés en faible quantité à la
surface cellulaire (3-10% de la quantité totale des MPR) (Braulke et al., 1987; Breuer et al.,
1997; Geuze et al., 1988), où seul CI-MPR internalise plusieurs ligands glycosylés ou non
glycosylés. CI-MPR possède donc de multiples fonctions qui dépassent la seule ségrégation
d'enzymes lysosomiales.
2.1.2 Transport indépendant de M6P - Sortiline
L'étude de la mucolipidose II (Inclusion-cell disease) a rapidement imposé un mode
de transport alternatif entre le TGN et les endosomes qui soit indépendant des M6P. En
effet, la localisation de nombreuses enzymes n'est pas affectée par l'inhibition de la
formation des groupements M6P qui caractérise la pathologie. C'est principalement
Sortiline (95kDa), aussi connu sous le nom de Neurotensin receptor 3 (NTR3), qui assure
alors le triage de ces cargos, dont la Psap (Lefrancois et al., 2003), la GM2-activator
protein (Lefrancois et al., 2003) et la Sphingomyélinase (ASM) (Ni and Morales, 2006). Il
appartient à la famille des protéines structurellement liées à la protéine de triage vacuolaire
(Vps) 10 de la levure (Vps10p) qui comprend également SorCS1, SorCS2, SorCS3 et
Sorting protein-related receptor containing LDLR class A repeats (SorLA) (Marcusson et
al., 1994). Au niveau de la région luminale, ils sont caractérisés par un domaine riche en
cystéines homologue à celui de Vps10p qui permet la liaison des ligands. Le motif reconnu
par ce domaine n'a toujours pas été identifié, mais tous les cargos lysosomiaux possèdent
29
des hélices alpha proéminentes qui pourraient être impliquées (Canuel et al., 2009).
Sortiline dispose aussi d'un site de clivage pour la Furine (Munck Petersen et al., 1999) et
d'une courte queue cytoplasmique homologue à celle de CI-MPR qui contient des motifs
d'interaction pour la machinerie de transport intracellulaire (Nielsen et al., 2001). Tout
comme les MPR, Sortiline se retrouve également en faible quantité (10% de la quantité
totale de Sortiline) à la surface cellulaire où il participe à l’endocytose d'une variété de
molécules, dont la Neurotensine (NT) (Quistgaard et al., 2009). D'ailleurs, Sortiline peut
agir en tant que corécepteur, entre autres avec NTR1, pour moduler la signalisation de ce
neuropeptide.
Sortiline est exprimé surtout au niveau des neurones du système nerveux central et
périphérique (Petersen et al., 1997; Sarret et al., 2003) où il agit comme récepteurs pour des
molécules proapoptotiques comme le précurseur du Nerve growth factor (NGF) (Nykjaer et
al., 2004) et du Brain derived neurotrophic factor (BDNF) (Teng et al., 2005). Sortiline
serait donc important pour l'induction de la mort neuronale et aurait aussi un rôle dans la
neurodégénération associée à la maladie d'Alzheimer. En effet, Sortiline médie le transport
rétrograde de la ß-sécrétase (BACE1), des endosomes précoces vers le TGN, ce qui
favorise le clivage amyloïdogénique menant à la formation du fragment Aß42 (Finan et al.,
2011). À son tour, ce fragment toxique hautement oligomérisable cause une augmentation
des niveaux de Sortiline (Saadipour et al., 2013). Ces données sont appuyées par une
surexpression de Sortiline dans les cerveaux de patients atteints d'Alzheimer (Saadipour et
al., 2013). D'un autre côté, Sortiline favorise également le clivage non amyloïdogénique
d'APP par l'α-sécrétase (Gustafsen et al., 2013). Il médie l'internalisation d'APP soluble et
facilite sa dégradation au lysosome (Gustafsen et al., 2013). De plus, Sortiline permet
l'internalisation d'Aß neurotoxique séquestré par l'Apolipoprotéine E (ApoE) au niveau
extracellulaire et mène à sa dégradation (Carlo et al., 2013). Les souris n'exprimant pas
Sortiline montrent d'ailleurs une augmentation d'ApoE due à un défaut de clairance, des
niveaux exacerbés d'Aß et la formation de plaques amyloïdes (Carlo et al., 2013). Sortiline
serait donc un récepteur neuronal ayant un rôle protecteur en contrant l'accumulation d'Aß
dans le cerveau grâce à l'ApoE et formerait un lien mécanistique entre le métabolisme des
lipoprotéines et la survie neuronale. Sortiline se retrouve également au niveau de cellules
30
spécialisées comme les hépatocytes et les adipocytes (Morris et al., 1998). Au niveau du
foie, il interagit avec les Apolipoprotéines A–V pour contrôler l'export de lipoprotéines vers
la circulation sanguine (Kjolby et al., 2010). Finalement, chez les adipocytes, il régulerait le
système de transport du glucose en réponse à l'insuline et médierait l'internalisation et la
dégradation de la Lipoprotein lipase (Lpl) (Nielsen et al., 1999), une enzyme qui hydrolyse
les triglycérides contenus dans les lipoprotéines.
Il est à noter qu'en plus de la voie de transport directe indépendante des groupements
M6P, certaines
enzymes peuvent emprunter une voie indirecte en fonction du type
cellulaire. Entre autres, au niveau des fibroblastes, de 5 à 20% des protéines lysosomiales
nouvellement synthétisées sont sécrétées. Dans ces cas, suite à la sécrétion il y a recapture
par endocytose grâce à des récepteurs (Dittmer et al., 1999). Deux membres de la famille
des LDLR, soit Megalin et LRP1 seraient ainsi capables de transporter CTSB (Nielsen et
al., 2007b) ainsi que Psap et CTSD (Markmann et al., 2015), respectivement.
2.1.3 Transport du Golgi aux endosomes
À la suite de leur passage au Golgi, les diverses protéines, telles les hydrolases
acides, atteignent le réseau trans-golgien où elles subissent un tri qui les orientera vers leur
destination. Pour ce faire, elles sont reconnues par des récepteurs de triage, comme MPR et
Sortiline. Le complexe récepteur-cargo est ensuite séquestré dans un microdomaine
correspondant à une vésicule de clathrine bourgeonnante qui s'enrichit de diverses protéines
utiles ultérieurement. Au TGN, la formation de ces structures est initiée par l'action de la
GTPase ADP-ribosylation factor-binding protein 1 (Arf-1) qui recrute à la membrane des
protéines adaptatrices de clathrine nommées GGA et AP (figure 4B) (Cooper and
Hausman, 2006; Doray et al., 2002). Chez l'humain, il existe trois types de GGA (1-3) qui
assemblent le manteau de clathrine grâce à leur région non structurée (Hinge) et leurs
domaines modulaires : Vps27, Hrs, Stam (VHS), GGA and TOM (GAT) et Gamma-adaptin
ear (GAE). Le domaine GAT est responsable de la liaison à Arf, le domaine VHS reconnaît
le motif dileucine (DXXLL) (Chen et al., 1993; Chen et al., 1997; Johnson and Kornfeld,
1992a; Johnson and Kornfeld, 1992b) de la queue cytoplasmique des récepteurs, la région
31
Hinge lie la clathrine alors que le domaine GAE recrute des protéines accessoires
(Bonifacino, 2004). Le modèle actuel veut que GGA interagisse avec AP (Doray et al.,
2002), de manière à lui transférer le complexe récepteur-cargo (figure 4B) (Doray et al.,
2002). Il existe cinq hétérotétramères AP (1-5) (Hirst et al., 2011) ayant une fonction
similaire aux GGA, mais AP-1 serait le principal acteur au niveau du TGN (Hirst et al.,
2011), où il reconnaît les motifs [D/E]XXXL[L/I] et YXXØ (où Ø est un résidu
hydrophobe encombrant) des récepteurs (Bonifacino and Traub, 2003). Les adaptatrices de
clathrine permettent le recrutement de plusieurs protéines à domaine de liaison aux
phospholipides ENTH/ANTH, dont Epsin, qui induit la polymérisation du triskèle de
clathrine (Rosenthal et al., 1999) et la formation d’une invagination (Ford et al., 2002)
(figure 4B). Alors que la vésicule croît, la Dynamine s’autoassemble autour du cou du
bourgeon se resserrant jusqu’au détachement de la vésicule (Praefcke and McMahon,
2004). Rapidement, le manteau se disloque suivant l’action de la chaperonne Heat shock
cognate protein 70 (Hsc70) (Schlossman et al., 1984) et de son partenaire Auxiline (Prasad
et al., 1993) qui fixent la chaîne lourde de clathrine. Dès lors, la vésicule nue expose la
machinerie nécessaire à l’attachement et la fusion spécifique de la vésicule avec les
endosomes précoces, la première composante du système endolysosomial. À cet endroit,
l’environnement
acide
entraîne
la
dissociation
des
complexes
récepteur-cargo.
Conséquemment, les cargos comme les hydrolases sont acheminés aux lysosomes pour
effectuer leur fonction dans la dégradation, tandis que les récepteurs sont redirigés vers le
TGN pour un nouveau cycle de transport.
2.1.4 Système endolysosomial
Le système endolysososomial est un réseau complexe de compartiments
intracellulaires qui assure le tri de molécules dans le but de les réutiliser (recycler) ou de les
détruire.
Il
se
compose
d'endosomes
précoces,
d'endosomes
tardifs
(corps
multivésiculaires) et de lysosomes (figure 4A) (Huotari and Helenius, 2011). Jadis perçu
comme des compartiments distincts stables échangeant du matériel par l'intermédiaire de
vésicules, on sait aujourd'hui qu'il s'agit plutôt de compartiments dynamiques dont la
composition hétérogène indique un phénomène de maturation en continuité (Cullen, 2011).
32
Figure 4 - Principaux mécanismes de transport antérograde et rétrograde.
(A) Schématisation du système endolysosomial. Des vésicules de clathrine contenant de
multiples cargos bourgeonnent du Golgi pour aller fusionner avec les endosomes
précoces issus des processus d'internalisation à la membrane plasmique. Ces derniers sont
caractérisés par des domaines de triage tubulaire qui permettent le recyclage sélectif de
cargos vers le Golgi ou la membrane plasmique. Alors que les endosomes se déplacent le
long des microtubules vers la région périnucléaire, ils maturent en endosomes tardifs qui
fusionnent avec les lysosomes. Figure modifiée de Endosome maturation, Huotari and
Helenius, The EMBO Journal (2011) 30, 3481–3500 avec la permission de John Wiley
and Sons. (B) Formation d'une vésicule de clathrine sur le TGN. L'activation d'Arf
permet le recrutement de protéines adaptatrices de clathrine qui concentre les cargos dans
des microdomaines formant des vésicules qui émergent du Golgi. Arf (ADP-ribosylation
factor-binding protein), GEF (facteur d’échange des nucléotides de guanine), GGA
(Golgi-localized, gamma-ear containing Arf binding protein) et AP (Adaptator protein).
Figure modifiée de The cell : A molecular approach 6e, Figure 10.36 Cooper and
Hausman avec la permission de Sinauer Associates. (C) Modèle général du recrutement
du complexe Rétromère. Le sous-complexe de Vps est recruté sur l'endosome par Rab7
de manière à ségréger les cargos dans des microdomaines tubulaires créés par l'action du
sous-complexe de SNX. L'association de protéines motrices et la polymérisation de
l'actine génèrent alors des forces qui entraînent le détachement du transporteur en
direction du Golgi. Vps (Vacuolar protein sorting), SNX (Sorting Nexin), WASH (WASP
and SCAR homologue). Figure inspirée de A global analysis of SNX27–retromer
assembly and cargo specificity reveals a function in glucose and metal ion transport.
Steinberg et al. Nat Cell Biol. 2013 May;15(5):461-71 avec la permission de Nature
Publishing Group (Macmillan Publishers Ltd).
33
Le système endolysosomial est généré par la fusion de vésicules d'endocytose formant des
endosomes précoces localisés en périphérie. Suivant l'action de la pompe à proton de
type vacuolaire (type V) (Yamashiro et al., 1983), ces derniers sont légèrement acides (pH
6,8–5,9) ce qui assure la dissociation des complexes récepteur-cargo en provenance de la
membrane plasmique ou du TGN. Ils sont constitués d'un domaine sphérique duquel
émergent de nombreux tubules (Chia et al., 2013) servant au triage des molécules en transit
(Cullen, 2011). Par exemple, les récepteurs permettant l'internalisation de nutriments,
comme Transferrin receptor (TfR), sont recyclés vers la membrane plasmique alors que les
récepteurs de transport, comme MPR et Sortiline, sont acheminés vers leur point d'origine,
soit le TGN. TfR constitue donc un marqueur caractéristique de l'organite de même que les
protéines cytosoliques associées comme EEA1, Rab4 et Rab5. L'endosome précoce va
graduellement maturer et sa région sphérique va former un endosome tardif. L'endosome
tardif est typiquement rond, sans tubules, et contient une grande quantité de vésicules
intraluminales, d'où son nom alternatif de corps multivésiculaire. Il se déplace vers le noyau
et peut devenir un lysosome par l'acidification graduelle de son contenu, passant de pH 6,0–
4,9 à 5,0-4,5 (Yamashiro and Maxfield, 1987), ou par la fusion avec des lysosomes
préexistants. En fusionnant avec les lysosomes, l'endosome tardif suit une voie
essentiellement unidirectionnelle puisque la plupart de ses composantes seront dégradées.
En effet, les lysosomes sont principalement des compartiments de dégradation où converge
le matériel de la voie d'endocytose, de phagocytose, d'autophagie et de synthèse. De façon
générale, le lysosome se distingue par une haute teneur en hydrolases, par la présence de
nombreuses glycoprotéines protégeant sa membrane, telles les Lysosome-associated
membrane proteins (LAMP) et les Lysosomal integral membrane proteins (LIMP), et par
l'accumulation de matériel résistant à la dégradation comme la lipofuscine. Par contre, dû à
la grande diversité de matériel à dégrader et le degré de cette dégradation, il s'agit de
compartiments très hétérogènes tant au niveau de leur composition, de leur morphologie
que de leur localisation. Les fonctions et composantes des lysosomes seront plus
amplement développées ultérieurement.
34
2.2
Transport rétrograde
Les premières évidences de l'existence d'un système de transport rétrograde des
endosomes vers le Golgi sont venues de l'étude de la toxine du ricin. Cette glycoprotéine
retrouvée dans les graines d'un arbrisseau produit son effet toxique en altérant l'ARN
ribosomique de manière à inhiber la synthèse protéique (Olsnes and Pihl, 1972). En 1975,
la ricine a été observée au niveau du Golgi indiquant la présence d'un système permettant à
la molécule exogène de pénétrer par endocytose pour remonter la voie de synthèse
(Gonatas et al., 1975). Cette découverte a ouvert la voie à l'identification de protéines
cellulaires, dont MPR, qui opère en cyclant entre le TGN et les endosomes (Duncan and
Kornfeld, 1988). On sait aujourd'hui que la voie de transport rétrograde peut remonter
jusqu'au RE et peut être empruntée par moult molécules classifiées selon cinq groupes, soit
les récepteurs de triage (MPR, Sortiline, LRP9 et Wntless), les protéases membranaires
intégrales (β-sécretase et Furine), les protéines médiant la fusion des vésicules, les
transporteurs de nutriments et les toxines (toxine Shiga (Sandvig et al., 1992), toxine du
choléra, toxine pertussique, toxine du ricin) (Burd, 2011). Elle permet le maintien des
fonctions du Golgi en le réapprovisionnant de ces molécules pour leur réutilisation.
Plusieurs processus cellulaires dépendent donc du bon fonctionnement de la voie de
transport rétrograde. Son dysfonctionnement entraîne, entre autres, une l'élévation du
peptide β-amyloïde associé à la maladie d'Alzheimer, un débalancement de l'homéostasie
des nutriments/éléments (glucose, fer, cuivre) et la perturbation du gradient de
morphogenèse de Wnt, une protéine multifonctionnelle d'importance (Burd, 2011).
Depuis l'étude des toxines qui a montré le lien à contresens entre la membrane
plasmique, les endosomes, le Golgi et le RE, les mécanismes permettant le transport
rétrograde ont été approfondis. On distingue désormais deux voies de recyclage
existant en parallèle et servant des cargos différents. Ainsi, les molécules peuvent se
rendre au TGN à partir des endosomes précoces ou des endosomes tardifs en utilisant
des mécanismes différents.
35
2.2.1 Transport des endosomes précoces au Golgi
Les endosomes précoces, ou plus particulièrement les tubules qui émergent de leur
domaine sphérique, constituent la principale station de triage de la voie endosomiale. À ce
niveau, le transport rétrograde vers le Golgi ou la membrane plasmique est orchestré par
l'assemblage d'un complexe qui, à l'image du manteau de clathrine, reconnaît et concentre
des cargos dans un transporteur. Ce complexe, nommé Rétromère, agit pendant la
maturation de l'endosome de manière à prévenir la dégradation au lysosome de nombreuses
protéines dont la fonction dépend de leur retour dans la voie de biosynthèse. Le cargo le
mieux caractérisé de cette voie est CI-MPR qui cycle entre les endosomes et le Golgi
(Seaman, 2004). Sortiline, le transporteur ionique Divalent metal transporter 1 (DMT1)-II
(Tabuchi et al., 2010), Wntless la protéine de transport de Wnt (Eaton, 2008), la protéine de
polarité cellulaire nommée Crumbs (Pocha et al., 2011) et SorLA un récepteur d'APP
(Nielsen et al., 2007a) empruntent également cette voie de transport rétrograde. Le
complexe Rétromère a été découvert en 1997 (Seaman et al., 1997), chez la levure
Saccharomyces cerevisiae. L'équipe du Dr. Emr a alors démontré son importance pour le
recyclage du récepteur à la Carboxypeptidase Y (CPY) des endosomes vers le Golgi
(Seaman et al., 1997; Seaman et al., 1998). Il s'agit d'un hétéropentamère formé par
l'association transitoire de deux sous-complexes distincts, soit un dimère de Sorting Nexin
(SNX) et un trimère de Vacuolar protein sorting (Vps).
Vps et capture du cargo
Chez les mammifères, le trimère de Vps est responsable de la reconnaissance et de la
sélection des cargos (figure 4C) (Seaman et al., 1998; Steinberg et al., 2013). Il comprend
les protéines Vps26, Vps29 et Vps35 qui ont été remarquablement conservées au cours de
l'évolution des eucaryotes (Koumandou et al., 2011). La composante centrale, Vps35, se
présente sous forme d'un solénoïde α-hélicoïdal (α-helical solenoid) en forme de fer à
cheval sur lequel Vps26 et Vps29 s'associent aux extrémités opposées, soit N-terminale et
C-terminale respectivement (Hierro et al., 2007; Norwood et al., 2010). Vps35 est
également le principal responsable de la reconnaissance des cargos (Nothwehr et al., 2000).
Il lie au moins un motif conservé de séquence consensus Trp/Phe-Leu-Met/Val (F/W-L36
M/V) dans la queue cytosolique de CI-MPR (motif WLM) et Sortiline (motif FLV)
(Seaman, 2007). Chez CI-MPR, ce dernier chevauche le motif de liaison d'AP-1
([D/E]XXXL[L/I]) démontrant une potentielle régulation structurelle entre le transport
antérograde et rétrograde. Des mécanismes de reconnaissance alternatifs, impliquant les
autres Vps, commencent à émerger. Vps26 lie directement un motif hydrophobe FANSHY
dans la portion cytoplasmique de SorLA (Fjorback et al., 2012) et servirait à la
reconnaissance des cargos. Chez l'humain, Vps26 présente deux paralogues (Vps26A et
Vps26B) (Kerr et al., 2005) qui pourraient agir à des endroits distincts et pour des cargos
différents, comme suggéré par la préférence de CI-MPR pour les complexes possédant
Vps26A (Bugarcic et al., 2011). De son côté, Vps29 serait un régulateur du transport
rétrograde. En plus d'un rôle structural (Collins et al., 2005a), il possède une activité
phosphatase, et ce malgré un repliement de type métallophosphoestérase (Damen et al.,
2006). In vitro, Vps29 a la capacité de déphosphoryler la sérine précédant le motif dileucine
antérograde (SDEDLL) de CI-MPR et Sortiline. Cette dernière agit comme signal pour
déclencher le transport antérograde et sa déphosphorylation pourrait être importante pour le
transport vers le TGN (Damen et al., 2006). De plus, la palmitoylation réversible de
certains récepteurs est nécessaire pour l'interaction avec les rétromères et, conséquemment,
pour la séquestration des cargos dans des microdomaines de transport vers le TGN
(McCormick et al., 2008). Il pourrait s'agir là d'un autre mécanisme de régulation agissant,
cette fois, en altérant la position et l'accessibilité du motif de triage.
SNX et tubulation des endosomes
Chez l'humain, le deuxième sous-complexe de rétromères, soit l'hétérodimère de
SNX (figure 4C), est généralement composé de SNX1 ou de SNX2 associée à SNX5 ou
SNX6 (Wassmer et al., 2007). Il est responsable de détecter la courbure membranaire et de
générer la tubulation de l'endosome (Carlton et al., 2004). Il a été proposé que la formation
de tubules permettrait de réduire la diffusion latérale des protéines afin de faciliter leur
ségrégation (Cullen and Korswagen). Aussi, cette région de haute courbure serait moins
épaisse et contiendrait peu de lipides saturés rigides (cholestérol et sphingomyéline) (Roux
et al., 2005) ce qui faciliterait la concentration préférentielle des cargos à courts domaines
transmembranaires (Sharpe et al., 2010), tels que SorLA et TfR (Chia et al., 2011). Pour
37
générer des microdomaines tubuleux, les SNX lient le PI3P et le PI(3,5)P2 enrichis dans les
endosomes, grâce à leur domaine Phox homology (PX) (Cozier et al., 2002) qui favorise
leur autoassemblage (Kurten et al., 2001). La dimérisation s'effectue par le domaine Cterminal Bin-Amphiphysine-Rvs (BAR) et résulte en une structure en arc qui épouse et lie
les membranes de courbure positive par des interactions électrostatiques (Peter et al., 2004).
Les hélices amphipathiques des SNX, à l'interface entre le cytosol et les lipides, génèrent
alors des tensions sur la membrane qui s'y accommode en se déformant (Peter et al., 2004).
Ainsi, les SNX agissent à la fois comme détecteur de courbure et comme responsables de la
tubulation des endosomes. Toutefois, la famille des SNX est grande et, au cours de
l'évolution, ses membres sont de plus en plus nombreux et divergents. Plusieurs formes de
complexes Rétromère peuvent donc être formées avec divers SNX de manière à agir à des
endroits distincts pour des cargos distincts. Par exemple, il existe un complexe composé des
Vps en association avec SNX3, qui ne possède pas de domaine de dimérisation BAR, et qui
permet le retrait de Wntless vers le TGN (Harterink et al., 2011). Les SNX-BAR et SNX3
semblent constituer des voies différentes agissant à la transition entre l'endosome précoce et
tardif (van Weering et al., 2012) et aux endosomes précoces (Harterink et al., 2011),
respectivement. Aussi, ils utilisent des transporteurs morphologiquement distincts, les
SNX-BAR se retrouvant au niveau de tubules (Mari et al., 2008) alors que les SNX3 sont
associés à des vésicules (Harterink et al., 2011) et à la clathrine (Skanland et al., 2009).
D'ailleurs, le rôle de la clathrine dans le transport rétrograde, ainsi que de multiples
protéines en relation, comme les adaptateurs EpsinR et AP-1 et l'enzyme de polymérisation
Lowe oculocerebrorenal syndrome protein ou Inositol polyphosphate 5-phosphatase
(OCRL1) est incertain. Ces dernières pourraient former une région de séquestration
précédant l'assemblage de certaines SNX (McGough and Cullen, 2011). Il existe également
un complexe composé de l'association entre SNX27 et les Vps qui, de son côté, permet le
recyclage à partir de l'endosome vers la membrane plasmique (Temkin et al., 2011). Plus de
70 cargos, dont le récepteur bêta-adrénergique (β2-AR), empruntent ce mécanisme
dépendant du motif de triage Postsynaptic density 95/discs large/zonus occludens-1 (PDZ)
(Lauffer et al., 2010) ou NPxY/NxxY (Ghai et al., 2013) reconnu directement par SNX27.
Bien d'autres SNX émergent comme étant impliquées dans la formation de complexes
38
Rétromère laissant présager une mécanistique complexe optimisée de manière à favoriser le
retrait d'un maximum de cargos.
Recrutement des rétromères
Pendant longtemps, le recrutement du complexe Rétromère aux endosomes a été
attribué à la seule association du sous-complexe SNX aux phospholipides membranaires
(Rojas et al., 2007). Ce dernier génère des domaines tubulaires puis, via sa région Nterminale non structurée, agirait comme plateforme pour le recrutement du sous-complexe
Vps avant de capturer le cargo. Depuis, ce modèle s'est avéré être une version simpliste ne
reflétant guère la complexité de la machinerie intracellulaire. En effet, les SNX sont
nécessaires, mais insuffisants pour le recrutement des Vps. Elles présentent seulement des
interactions faibles avec Vps35 et avec plusieurs sites de Vps29 (Swarbrick et al., 2011), ce
qui est peu propice à un phénomène de recrutement (Seaman, 2012). Il est maintenant établi
que Rab7-GTP participe aussi au recrutement et à la stabilisation du trimère de Vps (figure
4C) (Liu et al., 2012; Nakada-Tsukui et al., 2005; Rojas et al., 2008). Cette petite protéine
G lie Vps35 sur l'endosome tardif (Zelazny et al., 2013) de manière à coordonner l'export
du cargo avec la maturation de l'endosome. D'ailleurs, les transporteurs tubulaires
bourgeonnent majoritairement à partir des endosomes ayant acquis Rab7 (van Weering et
al., 2012). En retour, Vps29 interagit directement avec Tre2-Bub2-Cdc16-domaincontaining protein (TBC1D5), une GAP potentielle de Rab7 (Harbour et al., 2010; Seaman
et al., 2009). Ceci mènerait, une fois le transporteur tubulaire généré, à la dissociation de
Rab7 et conséquemment à la modification des phosphoinositides ainsi qu'au démantèlement
du manteau de rétromères (Seaman et al., 2009).
Implication du cytosquelette
Afin de permettre le détachement du transporteur tubulaire de l'endosome, les
rétromères recrutent une panoplie de protéines additionnelles, dont le complexe Wiskott–
Aldrich Syndrome protein (WASP) and Suppressor of cAR (cAMP receptor) (SCAR)
homologue (WASH) (figure 4C). L'activité de WASH a été montrée comme étant requise
pour le triage dans le recyclage de CI-MPR à partir des endosomes vers le Golgi (Gomez
and Billadeau, 2009) ainsi que pour le recyclage de TfR et de β2-AR des endosomes vers la
39
membrane plasmique (Derivery et al., 2009; Temkin et al., 2011). Comme les autres
membres de la famille Wiskott–Aldrich Syndrome protein (WASP), WASH est un facteur
qui promeut la formation de réseaux d'actine ramifiés (Campellone and Welch, 2010; Rotty
et al., 2013). Il est formé par l'assemblage de la protéine WASH avec les protéines
régulatrices Strumpelline, FAM21, Strumpellin and WASH-interacting protein (SWIP) et
Coiled-coil domain-containing protein 53 (CCDC53) (Derivery and Gautreau, 2010).
Quoique de nombreuses interactions stabilisantes soient répertoriées entre les composantes
du complexe Rétromère et celles du complexe WASH (Gomez and Billadeau, 2009), c'est
FAM21, via sa liaison à Vps35, qui est l'élément clé pour le recrutement à la membrane
endosomiale. FAM21 possède 21 copies du motif de liaison reconnu par Vps35 suggérant
que la protéine peut s'associer avec de multiples complexes Retromère de manière à les
enrichir dans des microdomaines tubulaires (Jia et al., 2012; Puthenveedu et al., 2010). À
cet endroit, l'activation locale de WASH entraîne une forte polymérisation de l'actine qui
génère une force de poussée longitudinale (figure 4C) (Derivery et al., 2009). De son côté,
les SNX5 et SNX6 s'associent avec une composante du complexe moteur de la Dynéine
(Hong et al., 2009). En s'éloignant sur le microtubule, cette dernière génère une force de
traction sur la région tubulaire de l'endosome (figure 4C) (Hong et al., 2009; Wassmer et
al., 2009). Les rétromères, en combinaison avec l'actine et les microtubules, sont donc
nécessaires à la génération de ces deux forces qui se combinent pour augmenter la tension
sur la membrane et entraîner le détachement du tubule. À ce sujet, la protéine EH domaincontaining protein 1 (EHD1) lie Vps26 et, par sa similitude avec la Dynamine (Daumke et
al., 2007), pourrait s'assembler en structures oligomériques pour permettre l’étranglement
du tubule en plus de stabiliser sa formation (Gokool et al., 2007).
Maladies associées aux rétromères
L'importance du transport rétrograde s'illustre par le nombre grandissant de maladies
qui lui sont associées. Brièvement, une réduction des niveaux protéiques des rétromères est
liée à la maladie d'Alzheimer (Muhammad et al., 2008) puisqu'il y a augmentation du
clivage amyloïdogénique d'APP possiblement aux endosomes (Choy et al., 2012; Wen et
al., 2011). Aussi, une mutation précise dans Vps35(Asp620Asn) est à l'origine d'une forme
de maladie de Parkinson (Zimprich et al., 2011). La perte de l'interaction entre le sous40
complexe de Vps et Rab7, causée par la mutation Rab7(K157N), provoque la maladie de
Charcot-Marie-Tooth (De Luca et al., 2008). Toutes ces pathologies sont caractérisées par
une atteinte du système nerveux. Il est fort à parier que la découverte de nouveaux joueurs
dans le transport rétrograde conduira à l'identification d'autres mutations aboutissant à
diverses maladies neurodégénératives.
2.2.2 Transport des endosomes tardifs au Golgi
En 1993, le groupe de Pfeffer a suggéré pour la première fois que des récepteurs
puissent être recyclés vers le TGN à partir des endosomes tardifs (Lombardi et al., 1993).
Rab9 et son effecteur Tail interacting protein of 47 kDa (TIP47) sont les deux principales
protéines impliquées dans ce processus. Rab9 colocalise avec MPR au niveau des
endosomes tardifs et stimule le transport rétrograde du récepteur (Lombardi et al., 1993) in
vitro (Goda and Pfeffer, 1988) alors que l'expression d'un dominant négatif a l'effet inverse
in vivo (Riederer et al., 1994). De son côté, TIP47 a été identifiée comme protéine
interagissant directement avec les queues cytosoliques des MPR, par l'intermédiaire d'un
motif Phe-Trp pour CD‐MPR (Diaz and Pfeffer, 1998; Schweizer et al., 1997) et d'un
domaine riche en prolines pour CI‐MPR (Orsel et al., 2000). Sa déplétion empêche le
recyclage des récepteurs vers le TGN (Diaz and Pfeffer, 1998) qui sont alors acheminés aux
lysosomes (Ganley et al., 2004). TIP47 interagit aussi directement avec Rab9 actif faisant
de lui un effecteur. Il augmente l'affinité de TIP47 pour MPR qui à son tour augmente la
liaison de TIP47 à Rab9 (Carroll et al., 2001; Hanna et al., 2002). Rab9 n'interagit pas avec
les récepteurs, mais facilite le recrutement de TIP47 spécifiquement au niveau des
endosomes tardifs et stimule la ségrégation des récepteurs par TIP47 dans la vésicule de
transport en formation. À ce moment, TIP47 formerait un hexamère grâce à son domaine
d'oligomérisation en N-terminal. Cet assemblage est dispensable pour la reconnaissance des
récepteurs, mais nécessaire pour la stimulation du transport rétrograde (Sincock et al.,
2003). Rho-related BR-C, ttk and bab (BTB) domain-containing protein 3 (RhoBTB3), une
Rho atypique hydrolysant de l'ATP plutôt que du GTP, a été suggéré comme étant
responsable de la libération de TIP47 de la vésicule (Espinosa et al., 2009). Elle lie
spécifiquement et directement Rab9 ainsi que TIP47 au niveau des membranes (Espinosa et
41
al., 2009). À ce jour, la Furine, une proprotéine convertase, est le seul autre récepteur
identifié pouvant utiliser la voie de recyclage dépendante de Rab9 (Chia et al., 2011). Par
contre, TIP47 ne lie pas la Furine ainsi que d'autres protéines cyclant entre les endosomes
et le TGN comme TGN38 et la Carboxypeptidase D (Hanna et al., 2002). De plus amples
études seront nécessaires afin de mettre en lumière tous les détails et les composantes de ce
mécanisme.
Alors que de nombreuses études soutiennent le rôle de Rab9 et TIP47 dans un
transport rétrograde à partir des endosomes tardifs, d'autres contestent cette conclusion. En
effet, TIP47 est le plus couramment décrite comme protéine des gouttelettes lipidiques
(LD, lipid droplets) (Ducharme and Bickel, 2008; Wolins et al., 2001) et serait recrutée à ce
niveau et non aux endosomes. D'ailleurs, l'interaction entre Rab9 et TIP47 ne semble pas
reproductible et l'affinité, rapportée comme étant deux fois supérieure (Ganley et al., 2004)
pour la forme active (96 nM) par rapport à l'inactive (159 nM), soulève un doute sérieux
relatif au rôle suggéré d'effecteur (Bulankina et al., 2009). La déplétion de TIP47 n'affecte
pas la distribution de MPR, ni le triage des enzymes lysosomiales, mais bloque plutôt la
maturation des LD et l'incorporation de triacylglycérol (Wolins et al., 2001). De surcroît,
par sa structure, TIP47 fait partie de la famille des protéines liant les LD PAT (Perilipine,
Adipophiline, TIP47) (Brasaemle, 2007) et montre beaucoup d'homologie avec l'ApoE au
niveau de la région de liaison des lipides (Bulankina et al., 2009). Il reste à savoir si TIP47
peut avoir des rôles différents dans le triage et le métabolisme des lipides.
2.3
Rab comme principaux régulateurs du transport intracellulaire
2.3.1 Cycle d'activation des Rab
Avec plus de 60 membres chez l'humain, les Rab constituent la plus grande sousfamille de petites protéines G Ras (Rojas et al., 2012). Elles sont conservées de la levure
aux mammifères (Pereira-Leal and Seabra, 2001) et sont d'expression ubiquitaire, tissu
spécifique ou régulée au cours du développement (Leung et al., 2006). Elles servent
principalement de régulateurs spatio-temporels lors de multiples processus liés au transport
42
Figure 5 - Les Rab comme principaux régulateurs du transport intracellulaire.
(A) Illustration des mécanismes du transport intracellulaire régulés par les Rab, soit le
bourgeonnement des vésicules, le désassemblage du manteau de clathrine, la motilité,
l'arrimage et la fusion de l'endosome. (B) Cycle d'activation des Rab. Les Rab
nouvellement synthétisées lient le GDP et sont escortées par une REP pour subir l'ajout de
groupements geranylgeranyl par la GGT. Elles peuvent alors s'ancrer aux membranes et
être activées par une GEF pour ensuite agir sur des effecteurs avant qu'une GAP ne
provoque leur retour à l'état inactif. Cet état est maintenu par une GDI jusqu'à ce qu'une
GDF dicte son recrutement à une membrane précise. GAP (protéine d’accélération de la
GTPase), GDF (facteur de déplacement de GDI), GDI (inhibiteur de la séparation des
nucléotides de guanine), GEF (facteur d’échange des nucléotides de guanine), GGT
(Geranylgeranyl transférase) et REP (protéine escorte de Rab). Figure modifiée de RAB11mediated trafficking in host–pathogen interactions. Guichard et al. Nat Rev
Microbiol. 2014 Sep;12(9):624-34. (C) Échange de Rab5 pour Rab7 lors de la maturation
de l'endosome précoce en endosome tardif. Rab5-GDP est activé par la GEF Rabex-5 ce
qui permet à son effecteur Rabaptin-5 de lier Rabex-5 pour promouvoir l'activation de
Rab5. Le complexe Mon1-Ccz1 (GEF de Rab7) lie alors Rab5 et Rabex-5 entraînant leur
dissociation et le recrutement de Rab7. Figure modifiée de Endosome maturation, Huotari
and Helenius, The EMBO Journal (2011) 30, 3481–3500 avec la permission de John Wiley
and Sons.
43
intracellulaire (figure 5A). L'efficacité des Rab repose sur leur cycle d'activation-inhibition
(figure 5B) (Guichard et al., 2014) qui leur permet d'agir comme interrupteurs moléculaires
auprès de divers effecteurs. Comme toutes les GTPases, les Rab possèdent deux
conformations structurelles en fonction de leur liaison au GTP ou au GDP. La première est
généralement reconnue par les effecteurs comme étant la forme active alors que la seconde
correspond à la forme inactive. Les facteurs d’échange des nucléotides de guanine (GEF)
catalysent l’activation des Rab en permettant l’échange du GDP pour du GTP. De leur côté,
les protéines d’accélération de la GTPase (GAP) catalysent l'hydrolyse du GTP en GDP de
manière à inactiver les Rab (Pan et al., 2006). Cet état est maintenu par des inhibiteurs de la
séparation des nucléotides de guanine (GDI) qui préviennent la relâche du GDP (Matsui et
al., 1990). La mécanistique des Rab est complexe en raison du nombre croissant d'acteurs
impliqués qui agissent en multiples combinaisons (Stenmark, 2009). Entre autres, certains
effecteurs sont soumis à plusieurs Rab distinctes. Aussi, l'effet d'une Rab peut être amplifié
par le recrutement d'un complexe d'effecteurs contenant une GEF pour la même Rab. Il
peut y avoir une activation couplée, si cette GEF active plutôt une autre Rab. Finalement,
l'activation couplée peut mener à une conversion de Rab s'il y a recrutement d'une GAP qui
inactive la Rab d'origine.
2.3.2 Localisation des Rab
La localisation intracellulaire distincte des différents Rab au niveau de compartiments
intracellulaires et de microdomaines contribue à la précision de leurs actions (Chavrier et
al., 1990). Les Rab nouvellement synthétisées doivent subir l'ajout d'un ancrage lipidique
pour permettre leur attachement réversible aux membranes (figure 5B). Pour ce faire, elles
sont reconnues par des protéines escortes de Rab (REP) qui les présentent à une
Geranylgeranyl transférase (GGT). Cette dernière ajoute des groupements du même nom à
un ou deux résidus cystéine de la région C-terminale des Rab suivant les motifs CC ou
CXC (Seabra et al., 1992). Les REP escortent alors les Rab jusqu'à une membrane tout en
agissant comme GDI (Alory and Balch, 2001). Par la suite, le recrutement d'une Rab à une
membrane précise sera contrôlé par un facteur de déplacement de GDI (GDF) associé à
cette dernière (figure 5B) (Sivars et al., 2003). Dans le système endolysosomial,
44
l'attachement spécifique des Rab en fait d'importants régulateurs spatio-temporels conférant
l'identité fonctionnelle des endosomes. Par exemple, Rab5 définit l'endosome précoce par
son action. Rab5 est recrutée à la membrane de l'endosome (figure 5C) (Huotari and
Helenius, 2011; Ullrich et al., 1994) à la suite de son activation par la GEF Rabex-5
(Yamashiro and Maxfield, 1987). Ce phénomène est amplifié par l'action de l'effecteur
Rabaptin-5 qui se lie à Rabex-5 (Horiuchi et al., 1997) pour favoriser l'activation de Rab5
(Lippe et al., 2001). Rab5 recrute également son effecteur la Phosphatidylinositol-3-kinases
(PI3K) de classe 3 (Christoforidis et al., 1999b). Cette enzyme convertit le
phosphatidylinositol (PI) membranaire en phosphatidylinositol-3-phosphate (PI(3)P) (Schu
et al., 1993) ce qui permet le recrutement de diverses protéines. L'endosome précoce va
graduellement maturer pour former un endosome tardif. Cette évolution est marquée par
l'échange de Rab5 pour Rab7 (figure 5C). Pour ce faire, les protéines Mon1 (Sand1 chez les
mammifères) et Ccz1 (GEF de Rab7) lient Rab5, le PI3P et Rabex-5 entraînant la
dissociation de ce dernier et, par le fait même, de Rab5 (Poteryaev et al., 2010). La GAP
responsable de mettre fin à l'activité de Rab5 est encore inconnue (Huotari and Helenius,
2011). Le complexe Mon1-Ccz1 permet également le recrutement de Rab7 (Poteryaev et
al., 2010) qui assure une production soutenue de PI(3)P en liant la PI3K (Stein et al., 2003).
2.3.3 Fonctions des Rab dans le transport entre le Golgi et les endosomes
Le recrutement spécifique des Rab confère l'identité fonctionnelle des membranes.
Elles agissent alors comme régulateurs dans plusieurs processus principalement liés au
transport vésiculaire, dont la formation, la motilité, l'arrimage et la fusion des vésicules
ainsi que le désassemblage de complexes de protéines (figure 5A).
Bourgeonnement et désassemblage du manteau protéique
La fonction des Rab au niveau du bourgeonnement de vésicules est de loin la plus
nébuleuse (figure 5A). Par exemple, Rab5 est une composante des vésicules de clathrine
qui serait essentielle pour la formation des puits. Pour ce faire, Rab5 permettrait la
séquestration de récepteurs par les protéines adaptatrices (Bucci et al., 1992).
L'internalisation de TfR est d'ailleurs directement liée à l'activité de Rab5 (Bucci et al.,
45
1992). D'autres Rab auraient un rôle semblable auprès d'autres compartiments dont Rab1
qui agirait au niveau de la formation des vésicules COP-I à partir de l'appareil de Golgi
(Alvarez et al., 2003; Peter et al., 1994) et Rab9 qui, comme mentionné antérieurement, agit
au niveau des endosomes tardifs. Par la suite, les Rab agissent comme régulateurs du
dépouillement de vésicules (figure 5A) de par l'exemple de Rab5. Suivant l'action de sa
GEF GTPase activating protein and VPS9 domains 1 (GAPvD1, aussi connue sous le nom
de RME-6, GAPex5 (Lodhi et al., 2007) ou RAP6 (Hunker et al., 2006)), Rab5 est activée
ce qui lui permet de recruter des effecteurs modifiant les éléments cruciaux à la stabilisation
de la protéine adaptatrice AP-2 au niveau des vésicules d'endocytose (Semerdjieva et al.,
2008). Dans un premier temps, Rab5 stimule la conversion du phosphatidylinositol-4,5bisphosphate (PI(4,5)P2) en PI(3)P via le recrutement de phosphatases et de kinases et,
dans un second temps, Rab5 provoque la déphosphorylation d'AP-2 via le déplacement de
la Adaptor-associated protein kinase 1 (AAK1) (Semerdjieva et al., 2008).
Motilité vésiculaire
Suite à leur dénudement, les vésicules peuvent voyager le long de filaments d'actine
et de microtubules afin d'atteindre leurs destinations. Les Rab participent également à ce
phénomène de motilité (figure 5A) en recrutant des effecteurs qui agissent comme
adaptateurs de protéines motrices. Par exemple, Rab11, par l'intermédiaire de RAB11
family-interacting protein 2 (RAB11FIP2) (Hales et al., 2002), lie la Myosine Vb qui
assure des déplacements courts et lents sur l'actine. Quand à eux, les déplacements longs et
rapides se produisent le long des microtubules et sont assurés par les protéines motrices
Kinésine, qui se dirige vers la périphérie, ou Dynéine, qui se déplace vers le centre
d'organisation des microtubules. L'attachement de l'endosome tardif à la Dynéine constitue
un exemple particulièrement pertinent dans le cadre de la présente étude. L'assemblage du
moteur protéique est initié par l'activation de Rab7 qui lie ses effecteurs Rab7-interacting
lysosomal protein (RILP) et Oxysterol‐binding protein (ORP1L) de manière à former un
dimère d'hétérotrimères (Johansson et al., 2007). RILP recrute alors le complexe DynéineDynactine qui est transféré au récepteur Spectrin betaIII par ORP1L activant le moteur
protéique rétrograde (Johansson et al., 2007). Un mécanisme semblable impliquant Rab7,
son effecteur FYVE and coiled-coil domain–containing protein 1 (FYCO1) et Microtubule46
associated protein 1A/1B-light chain 3 (LC3) permet le transport des vésicules
autophagiques par la Kinésine (Bento et al., 2013; Pankiv et al., 2010).
Arrimage
Une fois arrivée à proximité de leur destination, les vésicules doivent s'attacher
spécifiquement à la membrane du compartiment cible. Encore une fois, la localisation
intracellulaire distincte des Rab au niveau des compartiments intracellulaires en fait un
régulateur spatio-temporel de choix. Les différentes Rab facilitent le recrutement de
facteurs d'arrimage (figure 5A) précis qui servent en quelque sorte d'ancre. Il existe deux
groupes de facteurs d'arrimage (tethering factors), soit les protéines constituées de longues
superhélices (long coiled-coil proteins) et les complexes à multiples unités. Le premier
groupe englobe des protéines dimériques hydrophiles constituées de deux têtes globulaires
suivies de superhélices permettant un premier contact de faible affinité entre deux
membranes à grande distance (environ 200 nm) (Brocker et al., 2010). Les golgines qui
arriment les vésicules au Golgi, comme Golgi matrix protein 130 (GM130), en font partie
de même que Early endosome antigen 1 (EEA1). Ce dernier possède deux sites
d'interaction opposés pour Rab5 ce qui permet de lier ensemble deux endosomes précoces
arborant la GTPase (Christoforidis et al., 1999a). De son côté, le groupe des complexes à
multiples unités compte huit membres : Transport protein particle (TRAPP) I et II, Dsl1,
Conserved oligomeric Golgi (COG), Exocyst, Class C core vacuole/endosome tethering
(CORVET), Homotypic fusion and vacuole protein sorting (HOPS) et Golgi-associated
retrograde protein (GARP). Ils sont composés de trois à dix unités et ils agissent à
différents endroits dans la cellule vraisemblablement pour renforcer l'attachement des
vésicules lorsqu'elles parviennent à moins de 30 nm de distance. CORVET et HOPS sont
des hexamères ayant quatre unités en communs et deux unités spécifiques pour la liaison de
Rab distinctes (Plemel et al., 2011). CORVET est un complexe effecteur de Rab5 qui
permet l'arrimage entre endosomes précoces alors que HOPS agit plutôt sous l'influence de
Rab7, après la maturation de l'endosome, pour l'attachement impliquant les endosomes
tardifs et les lysosomes.
47
Fusion
Les Rab sont également d'importants régulateurs de la fusion des vésicules avec leur
compartiment de destination (figure 5A) à cause de leur effet sur les Soluble Néthylmaleimide-sensitive-factor (NSF) attachment protein (SNAP) receptor (SNARE). Les
SNARE sont des protéines membranaires majoritairement intégrales agissant comme
principales responsables de la fusion spécifique des membranes (McNew et al., 2000).
Chez l'humain, il existe 36 membres, nommés Vesicule-associated membrane protein
(VAMP), Syntaxine ou SNAP, qui sont différentiellement répartis sur les diverses
membranes intracellulaires (Jahn and Scheller, 2006). Elles sont caractérisées par la
présence d'une séquence homologue d'environ 60 acides aminés formant une superhélice
(Weimbs et al., 1997). Ce motif SNARE, permet l'assemblage complémentaire entre quatre
SNARE pour former un faisceau parallèle entremêlé nommé trans-SNARE (Sutton et al.,
1998). Le centre (zéro) de ce complexe est généralement composé d'une arginine (R) et de
trois glutamines (Q) entourés d'une fermeture en leucines étanche protégeant l'interaction
ionique (Antonin et al., 2002; Sutton et al., 1998). Il existe donc deux catégories de
SNARE, soit les R-SNARE et les Q-SNARE qui contribuent à la formation d'un complexe
spécifique via leur arginine (R) et leur glutamine (Q), respectivement (Fasshauer et al.,
1998). De façon générale, le R-SNARE est fourni par la vésicule (v-SNARE) alors que les
Q-SNARE proviennent du compartiment accepteur (target(t)-SNARE). Une fois
assemblées, les hélices du trans-SNARE s'enroulent et se resserrent de manière à
rapprocher deux membranes opposées. Les SNARE subissent alors un stress associé à leur
flexion qui les pousse à s'éloigner du lieu de fusion (Risselada et al., 2011). Il en résulte une
réduction de la répulsion entre les membranes et la génération d'une force nécessaire à leur
fusion (Risselada et al., 2011). Une fois sa fonction accomplie, le complexe SNARE
demeure sur la membrane fusionnée et est désigné cis-SNARE. Il est désassemblé par
l'ATPase NSF et son cofacteur SNAP pour permettre sa réutilisation (Marz et al., 2003;
Sollner et al., 1993). Les Rab agissent comme régulateurs indirects de la fusion puisque
plusieurs de leurs effecteurs sont des partenaires d'interaction des SNARE. Certains sont
des facteurs d'arrimage qui facilitent la fusion entre les membranes en régulant l'assemblage
des SNARE. Entre autres, EEA1, l'effecteur de Rab5, interagit avec le SNARE Syntaxin-13
pour permettre la fusion entre endosomes précoces (McBride et al., 1999). Le rôle de Rab5
48
est essentiel pour la fusion homotypique comme démontré par l'usage courant du mutant
constitutivement actif Rab5QL pour stimuler la fusion et ainsi faciliter l'observation
d'endosomes géants. Au niveau de l'endosome tardif, l'association d'un effecteur de Rab7 a
également été répertoriée en l'exemple de HOPS qui stimule la formation de complexes
impliquant VAMP3 et VAMP7 (Collins et al., 2005b).
3
Maladies associées aux lysosomes
3.1
Lysosomes
Le lysosome a été découvert en 1955 par Christian de Duve (De Duve et al., 1955),
prix Nobel de physiologie ou de médecine de 1974. Il a alors été décrit comme
compartiment cellulaire responsable de la digestion intracellulaire et du recyclage de
macromolécules. Il s'agit d'un organite juxtanucléaire et hétérogène morphologiquement
qui se définit aujourd'hui comme étant acide et riche en hydrolases, mais exempt de MPR
(Kornfeld and Mellman, 1989). La biogenèse du lysosome repose vraisemblablement sur la
maturation de l'endosome visant l'exclusion de certaines molécules et l'ajout graduel de
composantes caractéristiques (Mullins and Bonifacino, 2001). Ces dernières lui permettent
de remplir sa fonction de dégradation et comprennent plus de 50 hydrolases acides, telles
que des phosphatases, des nucléases, des glycosidases, des protéases, des sulphatases et des
lipases, ainsi que 120 protéines membranaires (Braulke and Bonifacino, 2009), dont au
moins 20 transporteurs (Sagne and Gasnier, 2008). La plupart des protéines membranaires
décorent la face luminale des lysosomes et sont hautement glycosylées (Fukuda, 1990)
formant une couche nommée glycocalyx qui protège l'organite de son pH acide. Les plus
abondantes sont LAMP1/2 et LIMP-1/2, les LAMP représentant 50% du protéome
membranaire. De son côté, l'acidité lysosomiale (pH 5) est principalement maintenue par la
pompe à proton de type vacuolaire (V-ATPase) qui hydrolyse de l'ATP pour permettre le
transport ionique au travers la membrane (Grabe and Oster, 2001). Cette activité est
cruciale pour la fonction de l'organite puisque de nombreuses hydrolases opèrent
préférentiellement en milieu acide. Entre autres, la protéase aspartique Cathepsine D subit
des changements de conformation dépendants d'une forte protonation qui libère son site
49
actif pour l'activer (Lee et al., 1998). De plus, cette enzyme doit préalablement être clivée le
long de la voie endolysosomiale par d'autres cathepsines dont l'efficacité est optimale à des
pH qui leurs sont propres (Laurent-Matha et al., 2006). Le matériel à dégrader aux
lysosomes provient de l'internalisation à la membrane plasmique, comme c'est le cas pour
certains récepteurs et leurs ligands, de la voie de synthèse ou de la séquestration par
autophagie. Certaines protéines membranaires, comme LAMP2A, permettraient également
la translocation de substrats cytosoliques précis vers la lumière de l'organite via des
chaperonnes (Bandyopadhyay and Cuervo, 2008; Bandyopadhyay et al., 2008), un
processus référé sous le nom d'autophagie médiée par des chaperonnes. Les produits
générés par la dégradation sont ensuite transportés vers le cytosol par des transporteurs
spécifiques. Parmi ces transporteurs, on compte Cystinosine et Sialine, des exportateurs de
cystéines et d'acides sialiques, respectivement. Transient receptor potential cation channel,
mucolipin subfamily, member 1 (TRPML1) transporte principalement le fer et le calcium
(Cheng et al., 2010) alors que, quoique non démontré, Niemann-Pick C1 protein (NPC1)
permetterait l'efflux de cholestérol. Par l'intermédiaires de nombreux canaux ioniques,
comme l'échangeur chlore/proton ClC-7 (Chloride channel 7), les lysosomes contrôlent
également la concentration ionique et le pH du cytosol. Les lysosomes possèdent aussi une
lysosome nutrient sensing (LYNUS) machinery qui détecte les variations en acides aminés
lysosomiaux via la V-ATPase (Zoncu et al., 2011) et le canal sodique ATP-sensible
(lysoNaATP) (Cang et al., 2013). Une forte concentration permet au Mammalian target of
rapamycin complex 1 (mTORC1) d'être activé et de s'associer à LYNUS à la surface des
lysosomes (Sancak et al., 2010; Zoncu et al., 2011). Alors, mTORC1 séquestre le facteur de
transcription EB (TFEB) (Cang et al., 2013) et induit la biosynthèse (Laplante and Sabatini,
2012). Au contraire, en condition de jeûne, mTORC1 est inactivé et relâché de LYNUS
libérant TFEB qui transloque au noyau pour favoriser l'autophagie ainsi que le catabolisme
de molécules. Ainsi, les lysosomes peuvent détecter la présence de nutriments et déclencher
une cascade de signalisation de réponse à leur absence.
Par le biais de sa capacité à dégrader, le lysosome est impliqué dans plusieurs autres
fonctions qui en font un compartiment central et dynamique. Les protéases qu'il contient
sont nécessaires dans l'immunité puisqu'elles traitent des antigènes pour la présentation par
50
le Complexe majeur d'histocompatibilité (CMH) de classe II. Les enzymes peuvent aussi
être libérées dans le milieu extracellulaire par exocytose des lysosomes pour inactiver des
organismes pathogènes ou participer à la dégradation de composantes de la matrice comme
dans le cas d'ostéoclastes. À la suite de la perméabilisation de l'organite, la relâche de
cathepsines peut aussi se faire dans le cytosol suivant un processus de mort cellulaire par le
lysosome, une forme d'apoptose peu connue (Saftig and Klumperman, 2009).
3.2
Maladies de surcharge lysosomiale
Les maladies de surcharge lysosomiale (LSD, Lysosomal storage diseases)
regroupent une cinquantaine de troubles héréditaires rares de type autosomal récessif, qui
ont en commun un dysfonctionnement des lysosomes. Les LSD affectent principalement
les enfants et présente une incidence combinée d'environ 1:5000 (Platt et al., 2012) et une
prévalence individuelle allant de 1:50000 à 1:4000000 (Meikle et al., 1999; Meikle et al.,
2004). Au point de vue cellulaire, les mutations génétiques responsables des LSD altèrent
des protéines membranaires intégrales des lysosomes, des protéines de transport
membranaire, des protéines impliquées dans les modifications post-traductionnelles ou,
plus fréquemment, l'activité d'enzymes particulières du lysosome (Schultz et al., 2011).
Chaque défaut entraîne un ralentissement ou une interruption de voies métaboliques
impliquées dans la dégradation de molécules. Il s'en suit une accumulation, aux lysosomes,
du substrat généralement directement en lien avec la fonction de l'enzyme fautive. La
classification conviviale des LSD repose d'ailleurs sur la nature de ce matériel laissant place
à des appellations telles que les lipidoses, les glycogénoses, les céroïdes-lipofuscinoses, les
pycnodysostoses,
les
mucopolysaccharidoses,
les
oligosaccharidoses
et
les
glycoprotéinoses. Il y a alors formation de lésions histologiques qui constituent la
principale caractéristique des maladies de surcharge lysosomiale (Heard et al., 2010). Les
inclusions cellulaires observées sont hautement hétérogènes au sein d'une même cellule et
selon le type cellulaire. Elles se présentent sous forme de larges structures d'aspect
vacuolaire contenant divers types de matériels, dont des débris, des produits clairs, des
vésicules, des fragments membranaires, des agrégats denses, des structures multilamellaires
et des corps zébrés (Heard et al., 2010). La toxicité, plutôt que la quantité, du matériel est
51
davantage critique pour les fonctions biologiques du lysosome (Heard et al., 2010). De
nombreux processus associés à l'organite peuvent être affectés comme la régulation du pH,
l'endocytose, l'exocytose, la maturation des vésicules, l'homéostasie du calcium et
l'autophagie (Ballabio and Gieselmann, 2009; Bellettato and Scarpa, 2010; Vitner et al.,
2010). À ce moment, la pathogenèse ne repose pas uniquement sur un problème de
contenu, mais aussi sur un problème de contenant (Heard et al., 2010). En effet, le
dysfonctionnement entraîne l'accumulation de produits secondaires qui participent à
l'hétérogénéité des inclusions cellulaires (Heard et al., 2010). Ces phénomènes entraînent à
la longue la mort cellulaire. Quoique l'ensemble des tissus soit affecté, dans la plupart des
cas, le système nerveux central demeure le plus sévèrement touché (Mitchison et al., 2004)
ce qui se traduit par une neurodégénération évolutive et polyhandicapante.
3.3
Céroïdes-lipofuscinoses neuronales
Parmi les LSD figurent les céroïdes-lipofuscinoses neuronales (NCL) dont le premier
cas fut décrit par Dr. Christian Stengel en 1826 (Brean, 2004; Stengel, 1826a). Il s'agissait
de quatre enfants norvégiens, issus de parents d'apparence saine, dont les symptômes sont
apparus vers l'âge de six ans (Brean, 2004; Stengel, 1826a). Au cours du début du siècle
suivant, de nombreux cas semblables ont été rapportés par Batten (Batten, 1903; Batten,
1914) et Spielmeyer (Spielmeyer, 1905a; Spielmeyer, 1905b). En l'honneur du pionnier, les
NCL sont d'ailleurs globalement désignées comme Batten disease. Suivant leurs
ressemblances cliniques et la présence d'accumulations cellulaires, elles ont originalement
été classées comme 'amaurotic family idiocy'. Dès 1969, l'appellation Neuronal ceroid
lipofuscinose a été proposée par Zeman et Dyken pour souligner le caractère distinct des
lipopigments accumulés (Zeman and Dyken, 1969). En effet, les NCL se distinguent des
autres LSD par l'accumulation anormalement forte d'un matériel périnucléaire et surtout
lysosomial, nommé lipofusine. Décrite pour la première fois en 1843 par Adolph Hannover
(Hannover, 1843), la lipofuscine, ou pigment d'âge, s'est rapidement révélée être une
caractéristique naturelle du vieillissement, inversement corrélée à l’espérance de vie d'une
cellule (Koneff, 1886). Le terme lipofuscine, proposé par Hueck (Hueck, 1912), provient de
l'union du mot grec lipo, soit gras et latin fuscus signifiant brun. De son côté, le mot ceroïde
52
est issu du latin cera et du grecque eidos signifiant 'de type cireux'. Ces termes décrivent
donc la lipofuscine qui est un produit brun jaunâtre, présentant une forte densité
électronique et une autofluorescence à spectre large (Brunk and Terman, 2002a; Porta,
2002; Terman and Brunk, 1998). Il est en effet possible d'observer la lipofucine par
excitation dans l'ultraviolet (330–380 nm), le bleu (450–490 nm) et le vert (510–560 nm)
(Porta, 2002). Cette importante propriété proviendrait de réactions entre les carbonyles
résultant de la peroxydation de lipides et les composés aminés qui produisent
principalement des bases de Schiff (Terman and Brunk, 1998). En fait, les lipofucines sont
formées de peptides réticulés (cross-link) et de composés variés fortement oxydés qui
résistent à la dégradation. Elles comprennent 30-70% de protéines, 20-50% de lipides
(triglycérides, acides gras libres, cholestérols et phospholipides) (Porta, 1991), 4-7% de
carbohydrates et 2% de métaux (fer, cuivre, aluminium, zinc, calcium et manganèse) (Jolly
et al., 1995; Terman and Brunk, 1998).
La respiration mitochondriale est à l'origine de la production de lipofuscine. Elle
entraîne la production de radicaux libres qui causent des modifications oxydatives au
niveau de macromolécules mitochondriales. Les mitochondries entrent ensuite dans les
lysosomes par autophagie et y déversent leur contenu résistant à la dégradation. Le faible
pH et la présence de substances réductrices favorisent également la production de radicaux
directement au niveau du lysosome causant de plus amples dommages. Par la suite, les
lysosomes engorgés se déchirent et libèrent la lipofuscine dans le cytosol. Ce modèle
(figure 6A) (Brunk and Terman, 2002b; Jung et al., 2007) explique pourquoi la sous-unité
C de l'ATP synthase mitochondriale (SCMAS) a été la première protéine identifiée comme
matériel emmagasiné dans les NCL (Palmer et al., 1989). Les protéines lysosomiales
Sphingolipid activator proteins (saposines ou SAP) A et D sont également bien
représentées (Tyynela et al., 1993). Les saposines sont associées à l'apparition
d'ultrastructures granulaires (GROD, granular osmiophilic deposits) (figure 6B) (Haltia,
2003) alors que la SCMAS forme des ultrastructures variées pouvant coexister et dites
curvilignes, rectilignes ou en empreintes digitales. Les GROD sont des corps membranaires
ronds finement granulaires mesurant 0,5 μm de diamètre et formant des agrégats atteignant
jusqu'à 5 μm (Mole et al., 2011). Le profil curviligne (CL) (figure 6B) (Haltia, 2003) est
53
Figure 6 - Formation et ultrastructures de la lipofuscine.
(A) Modèle de la formation de la lipofuscine selom l'hypothèse proposée par Brunk et
Terman en 2002. L'oxydation entraîne la réticulation des protéines et le dysfonctionnement
des mitochondries qui s'en trouve élargies. Ces protéines et ces mitochondries sont
acheminées aux lysosomes causant leur engorgement et, éventuellement, leur déchirement
et le déversement de leur contenu dans le cytosol. Figure tirée de Lipofuscin: formation,
distribution, and metabolic consequences, Jung et al., Annals of the New York Academy
of Sciences (2007) 1119, 97-111 avec la permission de John Wiley and Sons. (B) Les
ultrastructures associées à la lipofuscine se présentent sous quatres profils différents en
microscopie électronique, soit granulaire (GROD) ×10000, curviligne (CL) ×20000, en
empreintes digitales (FP) ×30000 et rectiligne (RL) ×15000. Figure tirée de The Neuronal
Ceroid-Lipofuscinoses, Haltia, Journal of Neuropathology & Experimental Neurology
(2003) 62(1), 1-13 avec la permission de Oxford University Press.
54
composé de citernes lamellaires minces et courtes (1,9-2,4 nm) uniformément courbées
alternant du noir au blanc (Mole et al., 2011). Il ne doit pas être confondu avec les
ultrastructures rectilignes (RL) (figure 6B) (Haltia, 2003), plus grosses et moins régulières,
qui présentent de courtes (2,8-3,8 nm) citernes oligolamellaires légèrement ondulées dont la
ligne centrale est proéminente (Mole et al., 2011). Finalement, le profil en empreintes
digitales (FP, fingerprint) (figure 6B) (Haltia, 2003) présente des lignes parallèles en paires
de 7,6-9,6 nm de large parfois fragmentées ou très denses (Mole et al., 2011).
3.3.1 Causes génétiques et protéines associées
Les NCL sont définies comme un groupe de « maladies génétiques dégénératives
progressives du cerveau et, dans la plupart des cas, de la rétine, en association avec une
accumulation intracellulaire d'un matériel qui est morphologiquement caractérisé comme
ou similaire à la céroïde-lipofuscine » (Mole et al., 2011). Leur classification originale était
principalement basée sur l'âge à l'apparition des symptômes, la progression de la maladie et
les caractéristiques des ultrastructures intracellulaires. On distinguerait alors les formes
infantile, infantile-tardive, juvénile et adulte qui ont rapidement présenté nombre de cas
atypiques et de variantes. En effet, les NCL se prêtent peu à ce système. Elles sont dites à la
fois génétiquement hétérogènes, puisque plusieurs mutations dans différents gènes peuvent
causer un phénotype clinique similaire, et hétérogènes au niveau allélique, puisque diverses
mutations dans le même gène peuvent engendrer des phénotypes cliniques différents (Mole
et al., 2011). Il existe également une hétérogénéité phénotypique qui se traduit par le fait
qu'une même mutation peut engendrer des symptômes cliniques et des progressions
différentes (Mole et al., 2011). Heureusement, l'évolution de la science a permis une
nouvelle classification incontestable et simple qui repose sur les déterminants génétiques.
Le premier gène impliqué dans la maladie a été identifié comme étant Palmitoyl-protein
thioesterase 1 (PPT1), par Vesa en 1995 (Vesa et al., 1995). Depuis 14 gènes, renommés
de façon uniforme Ceroid lipofuscinose neuronal 1-14 (CLN), figurent au tableau des NCL
(tableau 1) et des centaines de mutations sont répertoriées pour chacun. De plus amples
joueurs seront sans doute découverts puisqu'il existe encore des patients de phénotype
orphelin, c'est-à-dire que la cause génétique de leur phénotype est inconnue. De grands
55
efforts ont mené à l'identification de la majorité des protéines associées aux gènes CLN,
mais la fonction de chacun et leur rôle dans la pathogenèse restent encore obscurs. On
retrouve entre autres des enzymes directement impliquées dans la dégradation au lysosome
(PPT1 (ou CLN1), TPP1 (CLN2), CTSD (CLN10), CTSF (CLN13)) et des protéines
transmembranaires (CLN3, CLN6, Major facilitator superfamily domain-containing
protein 8 (MFSD8) ou CLN7), Probable cation-transporting ATPase 13A2 (ATP13A2) ou
CLN12)). La plupart de ces protéines se retrouvent au niveau de la voie endolysosomiale ou
du réticulum endoplasmique. Dans le contexte de la présente étude, l'accent sera mis sur les
protéines CLN3 et CLN5.
Tableau 1 - Pathologie des NCL.
Les gènes responsables des NCL, les protéines associées et leurs localisations, les protéines
accumulées dans la maladie, les principales ultrastructures observées et le nombre de
mutations répertoriées. CL (curviligne), FP (empreinte digitale), RL (rectiligne), GROD
(granular osmiophilic deposits), SAP (Sphingolipid activator proteins), SCMAS (Subunit
C of mitochondrial ATP synthase). Figure inspirée de Human NCL Neuropathology, Radke
et al., et mis à jour à partir de NCL Resource à l'adresse http://www.ucl.ac.uk/ncl/.
3.3.2 CLN3
Le gène CLN3 a été isolé en 1995 par The International Batten Disease
Consortium(The International Batten Disease Consortium, 1995). Il code la protéine
56
CLN3, aussi connue sous le nom de Battenine (Btn) en l'honneur de Dr. Frederick Batten,
un pionnier dans la découverte des NCL. Il s'agit d'une protéine hautement hydrophobe de
438 acides aminés comprenant six domaines transmembranaires, des extrémités
cytoplasmiques (Ezaki et al., 2003; Janes et al., 1996; Kaczmarski et al., 1999; Kyttala et
al., 2004) et une hélice amphipathique luminale entre le cinquième et le sixième segment
transmembranaire (figure 7) (Nugent et al., 2008). Elle est très conservée et présente, chez
l'humain, au moins sept isoformes produits par épissage alternatif. Son orthologue (Btn1)
chez la levure Saccharomyces cerevisiae lui est 30% identique et 59% similaire (Pearce and
Sherman, 1997). CLN3 compte de nombreux sites de potentielles modifications posttraductionnelles, dont trois pour des N-glycosylation de type hautement mannosylé (high
mannose) (Jarvela et al., 1998; Mao et al., 2003; Storch et al., 2007), six de
phosphorylation (Nugent et al., 2008), un de N-myristoylation en N-terminal (Kaczmarski
et al., 1999) et un motif CaaX de prénylation/farnésylation en C-terminal (Pullarkat and
Morris, 1997). CLN3 se retrouve en faible quantité, mais de façon ubiquitaire, et
étonnament son expression n'est pas particulièrement marquée dans le système nerveux
central (Mole et al., 2011). La présence de deux motifs de triage lysosomial assure sa
localisation, indépendante de MPR (Kyttala et al., 2004), dans l'organite ainsi que son
passage dans le RE, le Golgi et la voie endolysosomiale (Haskell et al., 2000; Jarvela et al.,
1998; Kida et al., 1999; Kyttala et al., 2004). Il s'agit en fait d'un motif dileucine dans une
boucle cytosolique (Kyttala et al., 2004; Storch et al., 2004) et d'un motif peu courant en Cterminal, composé d'une méthionine et d'une glycine séparées par neuf résidus (Kyttala et
al., 2004). CLN3 se retrouve également à la membrane plasmique dans une proportion
d'environ 10% (Mao et al., 2003; Persaud-Sawin et al., 2004; Storch et al., 2007) et aux
synapses (synaptosomes) des neurones primaires (Jarvela et al., 1999; Luiro et al., 2001).
Les NCL de type CLN3 sont les plus communes avec une prévalence de 0,6 cas par
100 000 naissances au Canada (MacLeod et al., 1976). Du point de vue histologique, la
maladie présente un profil en empreintes digitales, parfois accompagné de profils curviligne
et rectiligne (Elleder, 1999), avec une accumulation principale de la SCMAS (Elleder et al.,
1997). La grande majorité des patients, soit 85%, sont homo ou hétérozygotes pour une
même mutation qui consiste en une déplétion de 1 kb provoquant la perte des exons 7 et 8
57
(Cln3Δex7/8) (Munroe et al., 1997). Il en résulte deux transcripts qui, au niveau protéique, se
manifestent par la production de deux mutants. Le produit majeur provient d'un
changement de cadre de lecture au 153e acide aminé et possède seulement 28 nouveaux
résidus en aval de la mutation. Le produit secondaire est tronqué des résidus 154–263 car il
est issu de la perte d'un exon, de la création d'un nouveau site d'épissage et du retour au bon
cadre de lecture (Kitzmuller et al., 2008). Malgré le phénotype engendré et une forte
rétention au réticulum endoplasmique, ces peptides seraient suffisamment biologiquement
actifs pour maintenir la morphologie du lysosome (Kitzmuller et al., 2008). D'ailleurs,
l'incapacité de CLN3 à atteindre les lysosomes semble directement corrélée avec la sévérité
des phénotypes, suggérant un rôle important dans cet organite (Jarvela et al., 1999).
Figure 7 - Schéma de la structure de CLN3.
CLN3 est une protéine à six domaines transmembranaires comprenant des extrémités
cytoplasmiques ainsi qu'une hélice amphipathique entre les cinquième et sixième
segments transmembranaires. Figure créée avec Microsoft Office PowerPoint 2007 et
inspirée de The transmembrane topology of Batten disease protein CLN3 determined by
consensus computational prediction constrained by experimental data. Nugent et al.
FEBS Lett 582:1019–1024.
À ce jour, aucun homologue de CLN3 n'a été identifié et sa fonction demeure encore
inconnue. Il a été proposé que la protéine soit impliquée dans de nombreux processus
cellulaires, dont la balance ionique, l'acidification des lysosomes, les modifications de
protéolipides, l'apoptose et la signalisation. Brièvement, chez la drosophile, CLN3 interagit
avec Notch et Jun N-terminal kinase (JNK) et modulerait leurs voies de signalisation
58
(Tuxworth et al., 2009). L'absence d'activité palmitoyldésaturase chez les cellules déplétées
en CLN3 suggère que cette réaction enzymatique pourrait lui être associée (Narayan et al.,
2006; Narayan et al., 2008). Une augmentation de l'apoptose a aussi été observée dans ces
conditions (Persaud-Sawin and Boustany, 2005; Puranam et al., 1999). CLN3 interagit
d'ailleurs avec Calséniline, une protéine à mains EF qui contribue à la mort induite par le
calcium (Chang et al., 2007), et protègerait la cellule par la régulation des céramides
intracellulaires qui agissent comme second messager dans la cascade apoptotique (Puranam
et al., 1999). CLN3 possède un domaine conservé de liaison de galactosylceramide (GalCer
lipid raft-binding domain) et participerait à son transport du Golgi vers les radeaux
lipidiques influençant la composition membranaire (Rusyn et al., 2008). Dans un autre
ordre d'idées, l'absence de CLN3 ou sa mutation entraîne une diminution du pH lysosomial,
c'est-à-dire une acidification (Golabek et al., 2000; Holopainen et al., 2001; Padilla-Lopez
and Pearce, 2006; Pearce et al., 1999a; Pearce et al., 1999b). CLN3 participe donc au
maintien de l'homéostasie protonique et affecte même la maturation dépendante du pH de
protéines comme APP et CTSD (Golabek et al., 2000). Le mécanisme qui sous-tend cet
effet est encore inconnu, mais pourrait être associé à la régulation de CLN3 par
Shwachmann-Bodian-Diamond syndrome protein (SBDS), une protéine impliquée dans la
maturation des ribosomes et qui influencerait l'expression de la pompe ATPase vacuolaire
(Vitiello et al., 2010). CLN3 agirait aussi comme transporteur puisqu'elle présente une
distante, mais significative similarité de séquence avec Equilibrative nucleoside transporter
1 (ENT1), un membre de la famille des Solute carrier family 29 (SLC29), qui équilibre les
nucléosides et les nucléobases à la membrane plasmique (Baldwin et al., 2004). D'ailleurs,
CLN3 aurait un rôle dans l'osmorégulation rénale, soit le contrôle de l'équilibre en eau et en
potassium (Stein et al., 2010). Son expression est aussi plus élevée et sa localisation plus
lysosomiale et périphérique en condition d'hyperosmolarité (Getty et al., 2013). Dans le
même ordre d'idées, un transport défectueux d'arginines en l'absence de CLN3 ou en
présence d'un mutant pathologique tend à démontrer son implication dans l'import de
résidus basiques à travers la membrane lysosomiale (Chan et al., 2009; Kim et al., 2003b;
Ramirez-Montealegre and Pearce, 2005). Cet effet pourrait simplement être une
conséquence de la régulation à la hausse de Btn2 qui module le transporteur Arginine59
lysine-histidine-permease-Can1p, tel que montré chez
Saccharomyces cerevisiae
(Chattopadhyay and Pearce, 2002).
Toutefois, la plupart des évidences pointent vers un rôle dans le transport post-TGN
vraisemblablement en lien avec le cytosquelette. En conséquence à une perte de
Btn1/CLN3, plusieurs anomalies ont été rapportées relativement à des processus de
transport, notamment à l'endocytose (Fossale et al., 2004; Luiro et al., 2004; Uusi-Rauva et
al., 2008), au transport axonal du nerf optique (Weimer et al., 2006), à la maturation des
vacuoles autophagiques (Cao et al., 2006), à la morphologie golgienne (Codlin and Mole,
2009) et surtout au triage dans le Golgi (Codlin and Mole, 2009). Ce dernier altère la sortie
de MPR qui s'accumule au TGN (Metcalf et al., 2008) menant à une maturation aberrante
d'enzymes lysosomiales comme des cathepsines (Fossale et al., 2004; Golabek et al., 2000;
Metcalf et al., 2008). CLN3 contribue également à la localisation des endosomes et des
lysosomes. Plusieurs changements dans la distribution vésiculaire, induits par des mutants
de CLN3, ont été observés en fonction des cellules, dont une dispersion causée par
Cln3Δex7/8 (Fossale et al., 2004) et un regroupement périnucléaire faisant suite à l'expression
de CLN3E295K (Uusi-Rauva et al., 2012). D'ailleurs, CLN3 interagit avec des
composantes motrices impliquées à la fois dans le transport antérograde et rétrograde le
long des microtubules, soit beta-tubuline, p150Glued du complexe de Dynactine, la chaîne
intermédiaire de la Dynéine (DIC) et la sous-unité Kinesin Family Member 3A (KIF3A) du
complexe Kinésine-2 (Uusi-Rauva et al., 2012). L'association membranaire de ces
composantes est régulée par les protéines Rab ou plus précisément par Rab7 au niveau des
endosomes tardifs. Le domaine cytoplasmique N-terminal de CLN3 interagit directement
avec Rab7, préférentiellement actif, de façon à influencer son recrutement et possiblement
son cycle d'activation (Uusi-Rauva et al., 2012). Il a été montré que CLN3 peut alors
former un complexe en liant, avec une de ses boucles cytosoliques, RILP, un effecteur de
Rab7 (Uusi-Rauva et al., 2012). Le couple Rab7-RILP est bien connu pour faciliter le
recrutement du complexe Dynéine/Dynactine aux endosomes tardifs et pourrait constituer
le déterminant moléculaire par lequel CLN3 affecte le positionnement vésiculaire et
subséquemment la fonction lysosomiale. Le partenaire d'interaction Btn2, aussi connu sous
le nom de Hook1 chez les vertébrés, est aussi impliqué dans la liaison des microtubules
60
(Walenta et al., 2001) et pourrait compétitionner avec CLN3 pour Rab7 (Luiro et al., 2004).
Btn2/Hook1 lie également les SNARE (Kama 2007), laissant croire que CLN3
influencerait la fusion. CLN3 est associée à la migration cellulaire, la formation de plaques
d'actine (Codlin and Mole, 2009) et la distribution de la Myosine possiblement en lien avec
son interaction avec la Myosine-IIB (Getty et al., 2011). CLN3 interagit avec Fodrine et la
pompe Na+/K+ ATPase associée (Uusi-Rauva et al., 2008). La première est une protéine
associée à la membrane plasmique qui contrôle la distribution de surface de la pompe en la
fixant au cytosquelette d'actine. La déplétion de CLN3 cause une perturbation de la
distribution de Fodrine ainsi que de l'endocytose de la pompe Na+/K+ ATPase (Uusi-Rauva
et al., 2008) mettant en péril le gradient ionique qui fait partie intégrale de la fonction
neuronale. La fonction principale de CLN3 reste inconnue et sa multitude de partenaires
d'interaction lui confère une potentielle influence directe ou indirecte sur nombre de
processus biologiques.
3.3.3 CLN5
Le gène CLN5 a été isolé en 1998 (Savukoski et al., 1998), il est conservé chez les
vertébrés uniquement et code une protéine de 407 acides aminés du même nom qui n'a pas
d'homologue. À l'origine, les outils informatiques, comme PHDhtm et TMpred, prédisaient
que la protéine contiendrait jusqu'à deux segments transmembranaires, soit globalement des
résidus 74 à 91 et 352 à 375 (Savukoski et al., 1998). La région hydrophobe 76-91 a ensuite
été confirmée comme étant transmembranaire (Vesa et al., 2002) alors que la seconde
région pourrait assurer une association membranaire. Aujourd'hui, CLN5 mature est
considérée comme étant une protéine soluble (Holmberg et al., 2004; Schmiedt et al., 2010)
puisque la portion N-terminale contenant la région transmembranaire est incluse dans un
long peptide signal (figure 8). Lors de la synthèse, il y a donc clivage en position 96, par
une peptidase signal non identifiée et la région transmembranaire est retenue dans le RE
(Schmiedt et al., 2010). Alors que la plupart des espèces possèdent un seul codon initiateur
pour CLN5 (Holmberg et al., 2004), chez l'humain, il en existe quatre aux positions 1, 30,
50 et 62 (Savukoski et al., 1998). Les quatre transcrits produits ont des masses moléculaires
61
de 39 à 47 kDa (Isosomppi et al., 2002; Vesa et al., 2002), mais subissent le même clivage
de manière à produire une proprotéine identique.
Figure 8 - Schéma de la structure de CLN5.
(A) CLN5 immature est une protéine transmembranaire de type 2 qui est clivée en
position 96 par une peptidase signal suite à son insertion dans la membrane du RE. (B)
CLN5 mature est une protéine luminale. Le segment transmembranaire de la forme
immature est représentée en rouge, la forme mature correspond à la partie en bleu, les
ciseaux représentent le site de clivage par la peptidase signal. Figure créée avec Microsoft
Office PowerPoint 2007.
Après sa synthèse, CLN5 passe au travers de la voie de synthèse et subit des Nglycosylations hautement mannosylé (high mannose) et de sucres complexes sur jusqu'à
huit sites potentiels (Asn179, 192, 227, 252, 304, 320, 330 et 401) (Moharir et al., 2013;
Savukoski et al., 1998). Les sucres en position Asn179, 252, 304, 320 et 330 seraient
nécessaires à l'export du RE. L'ajout de mannose-6-phosophate au niveau des résidus
Asn320, Asn330 et Asn401 (Sleat et al., 2006) a été appuyé par des mesures en
spectroscopie de masse, mais seule la modification sur l'Asn401 permettrait de quitter le
Golgi (Moharir et al., 2013). CLN5 possède aussi des sites prédits pour d'autres
modifications postranductionnelles, dont sept de phosphorylation par des protéine-kinases
C, des caséine-kinases II et des tyrosine-kinases ainsi deux sites chevauchants de Nmyristoylation (Savukoski et al., 1998). Ces modifications n'ont pas été confirmées
62
expérimentalement mais expliqueraient la présence, après traitements de déglycosylation,
de deux formes de CLN5 (Schmiedt et al., 2010). CLN5 est donc synthétisée comme une
préproprotéine d'environ 70 kDa qui est clivée au RE en une proprotéine de 60 kDa avant
d'être glycosylée (60–80 kDa) et finalement taillée pour atteindre 50 kDa (Schmiedt et al.,
2010). La forme mature résultante est faiblement sécrétée, mais se rend principalement aux
lysosomes (Isosomppi et al., 2002; Vesa et al., 2002). Elle emprunte MPR (Kollmann et al.,
2005; Sleat et al., 2006) et peut vraisemblablement utiliser une voie alternative comme
indiqué par sa présence inaltérée au compartiment acide dans des fibroblastes déficients en
MPR (Schmiedt et al., 2010). Le récepteur LIMP-2 serait le principal candidat à cette
fonction puisque Sortiline a été montré comme interagissant avec CLN5 sans toutefois
influencer son transport (Mamo et al., 2012).
Les NCL de type CLN5 ont fait leur apparition en Finlande où l'incidence est de 2,6
cas par million d'habitants (Uvebrant and Hagberg, 1997). En histologie, la maladie partage
les mêmes caractéristiques que les types CLN3, 6, 7 et 8, soit l'accumulation majoritaire de la
SCMAS (Tyynela et al., 1997) et la présence de profils en empreintes digitales accompagnés de
profils curvilignes et rectilignes. La première mutation identifiée, et aussi la plus commune,
représente 94% des cas de NCL liés à CLN5 (Mole et al., 2011). Il s'agit d'une délétion de 2
paires de bases dans l'exon 4 (c.1175_1176delAT) produisant un codon de terminaison en
position 392 (p.Tyr392∗) (Holmberg et al., 2000) et formant une glycoprotéine de 47-52
kDa (Isosomppi et al., 2002; Vesa et al., 2002). Dans certaines sous-populations côtières de
Finlande, jusqu'à 1 personne sur 24 est porteuse de ce défaut génétique (Savukoski et al.,
1998), d'où son nom de mutation Finlandaise majeure (FinM) (tableau 2). Cette appellation
perdure bien que l'on retrouve maintenant de très rares cas à l'extérieur de ce pays. Parmi
les mutations les mieux connues, on compte également la mutation Finlandaise mineure
(Finm) (tableau 2) dans l’exon 1 (c.225G>A) qui produit une protéine de 74 résidus et une
de 12 kDa (p.Trp75∗) (Holmberg et al., 2000; Xin et al., 2010), la mutation Européenne
(EUR) (tableau 2) dans l'exon 4 (c.835G>A) qui provoque un changement de résidu en
position 279 (p.Asp279Asn) (Holmberg et al., 2000; Savukoski et al., 1998) et la mutation
Suédoise (SWE) (tableau 2) qui entraîne une terminaison prématurée (c.757G>T) et qui
63
code une protéine tronquée de 34 kDa (p.E253∗) ainsi que deux polypeptides de 37 et 40
kDa (Vesa et al., 2002). Ce dernier mutant possède seulement deux des sucres importants
pour l'export du RE, mais peut tout de même passer les contrôles de qualité du RE (Vesa et
al., 2002). Finalement, quoique peu répandu, le mutant de Terre-Neuve (NFL) (tableau 2)
est pertinant à la présente étude et consiste en une mutation dans l'exon 4 (c.1054G>T) qui
crée un codon de terminaison (p.Glu352∗) (Moore et al., 2008). Certains mutants seraient
retenus au RE cependant l'atteinte du lysosome ne semble pas être en corrélation avec la
sévérité du phénotype (Lebrun et al., 2009; Schmiedt et al., 2010). Cela indique que malgré
la faible présence de CLN5 au Golgi et au RE (Isosomppi et al., 2002), il pourrait s'agir là
de lieux d'interactions importantes.
CLN5 est exprimée dans tous les tissus humains (Savukoski et al., 1998), mais
surtout au cerveau notamment dans les cellules microgliales, les astrocytes, les
oligodendrocytes et les neurones (Heinonen et al., 2000; Holmberg et al., 2004; Schmiedt,
2012). La protéine semble particulièrement importante à ce niveau puisque son absence
entraîne un défaut de myélination, une altération du profil lipidique sérique et un
ralentissement du transport intracellulaire des sphingolipides. Ces observations tendent à
indiquer un rôle dans le métabolisme des lipides (Schmiedt et al., 2012). Tout comme les
cellules déficientes en CLN3, 2, 6 et 8 (Lane et al., 1996), les fibroblastes CLN5-/- arborent
une augmentation de l'apoptose en lien avec une diminution des niveaux de sphingolipides,
comme les céramides (Haddad et al., 2012). Il a été proposé que CLN5 agirait comme
activateurs de la Ceramide synthase (CerS), l'enzyme responsable de la synthèse de novo
(Haddad et al., 2012). L'absence de CLN5 entraîne également une inflammation avec
activation hâtive de la microglie et des astrocytes qui pourrait être à l'origine de la mort
neuronale (Schmiedt et al., 2012; von Schantz et al., 2008).
En ce qui concerne le transport intracellulaire, l'équipe du Dr. Lefrançois a montré
qu'en condition de surexpression CLN5 interagit avec le récepteur Sortiline sans toutefois
agir comme cargo (Mamo et al., 2012). La déplétion de CLN5 par ARN interférent entraîne
une dégradation de Sortiline, ainsi que de CI-MPR, aux lysosomes (Mamo et al., 2012). En
64
effet, ces derniers ne sont plus recyclés des endosomes vers le TGN ce qui limite la
maturation de l'enzyme CTSD (Mamo et al., 2012). CLN5 serait donc nécessaire au
fonctionnement du complexe Retromère. Plus particulièrement, la déplétion de CLN5
affecte le recrutement aux endosomes de deux protéines qui amorcent le transport
rétrograde et avec lesquelles CLN5 interagit indépendamment de leur activation, soit Rab5
et surtout Rab7 (Mamo et al., 2012). CLN5 serait impliquée dans l’activation de Rab7
puisque la capacité de cette dernière à interagir avec son effecteur RILP est réduite en
l'absence de CLN5 (Mamo et al., 2012). Il a été proposé que CLN5 activerait, directement
ou par l'intermédiaire d'une protéine transmembranaire, la GEF de Rab7, identifiée comme
étant Mon1-Ccz1 chez la levure (Mamo et al., 2012). Suivant la réduction de Rab7 aux
endosomes, les sous-unités VPS26 et VPS35 du complexe Rétromère sont également sousreprésentées sur les endosomes (Mamo et al., 2012).
Tableau 2 - Principales mutations dans CLN5.
Les impacts génétiques (localisations, changements, types) et protéiques (changements et
masses moléculaires) des mutations FinM (Finlandaise majeure) et Finm (Finlandaise
mineure), EUR (Européenne), SWE (Suédoise) et NFL (Terre-Neuvienne). Basé sur NCL
Resource à l'adresse http://www.ucl.ac.uk/ncl/.
3.3.4 Interactosome des CLN
De plus en plus d'évidences tendent à démontrer des relations et des effets
compensatoires entre les protéines CLN tant en ce qui a trait à leurs fonctions qu'à leurs
expressions. Des études in vitro ont montré les premières possibilités d'interactions. Entre
autres, CLN5 pourrait lier CLN1 (Lyly et al., 2009), CLN2 via son C-terminal (de CLN5)
(Vesa et al., 2002), CLN3 via son N-terminal (de CLN5) (Vesa et al., 2002), CLN6 (Lyly et
65
al., 2009), CLN8 (Lyly et al., 2009). Concernant l'expression, CLN1 et CLN5 partagent des
patrons d’expression similaires et, les souris déficientes en CLN5, présentent une
augmentation de l’ARNm de CLN1 (Lyly et al., 2009). Il pourrait s'agir là d'un effet
compensatoire en lien avec le fait que CLN1 semble influencer positivement le transport de
CLN5 (Schmiedt et al., 2010). Il a aussi été proposé que CLN1 et CLN5 formeraient un
complexe avec l'ATP synthase (Lyly et al., 2008; Lyly et al., 2009), un récepteur de
l’ApoA-I, appuyant davantage un rôle pour CLN5 dans l’homéostasie des lipides (Martinez
et al., 2003).
Dans le même ordre d'idées, une réduction des protéines CLN2 et CLN5 entraîne une
diminution de l’ARNm de CLN3 (Bessa et al., 2006). D'ailleurs, l’activité de CLN2 est
plus forte dans les fibroblastes portant une mutation dans CLN5 (Vesa et al., 2002) ou
CLN3 (Sleat et al., 1998). Dans ces mêmes cellules, le pH des lysosomes est augmenté
laissant croire que les deux protéines pourraient collaborer pour maintenir l’homéostasie du
pH (Holopainen et al., 2001; Pearce et al., 1999a). Ces observations suggèrent une relation
ou une redondance fonctionnelle entre les CLN2, 3 et 5. CLN5 partagerait aussi le rôle
d'activateurs de synthases de ceramides avec CLN8, car CLN8 à la capacité de corriger les
défauts apoptotiques engendrés par l'absence de CLN5 (Haddad et al., 2012).
De plus amples études seront nécessaires afin de clarifier cet interactosome. La
découverte, en 2011, du motif Coordinated Lysosomal Expression And Regulation (CLEAR)
pourrait expliquer la coordination d'expression de certaines protéines CLN. Entre autres,
CLN1, CLN2, CLN3, CLN5, CLN7, CLN10, CLN11 et CLN13 ont été clairement
identifiés comme protéines possédant le motif CLEAR et comme étant vraisemblalement
régulées par TFEB (Sardiello et al., 2009). Ce sujet sera plus amplement développé dans la
discussion.
3.3.5 Aspects cliniques
Les
céroïdes-lipofuscinoses neuronales constituent le groupe de
maladies
neurodégénératives progressives et récessives le plus commun chez les enfants avec une
66
incidence mondiale estimée de 1:100000 (Santavuori, 1988), mais fortement accentuée
dans les pays nordiques comme la Finlande à 1:12500 (Rider and Rider, 1988; Williams et
al., 1999). Elle est diagnostiquée par la combinaison de plusieurs examens. Les premiers
indices de la maladie peuvent être détectés lors d'examens visuels analysant la réponse
électrique du nerf optique ou de la rétine (Mole et al., 2011). Aussi, la tomodensitométrie et
l'imagerie par résonance magnétique sont utilisées pour révéler des changements
structuraux au niveau du cerveau (Williams et al., 2006). Pour parvenir à un diagnostic
définitif et précis, les patients sont rapidement soumis à des essais enzymatiques visant à
identifier une enzyme défectueuse (Williams et al., 2006). Toutefois, cette technique est
valable pour un nombre limité de sous-types de la maladie, n'étant pas tous causés par un
problème enzymatique. En microscopie électronique, l'observation de biopsies provenant
principalement de la peau, permet également de cibler les NCL grâce aux dépôts cellulaires
qui leurs sont caractéristiques (Williams et al., 2006). D'ailleurs, dans les familles affectées
par la maladie, l'étude prénatale des inclusions et des ultrastructures sur les prélèvements
des villosités choriales révèle la maladie dès le troisième mois de la grossesse (Mole et al.,
2011). Finalement, l'analyse de mutations établit un diagnostic incontestable (Williams et
al., 2006). À ce sujet, le criblage à haut débit (high-throughput screening) facilite les
diagnostiques moléculaires des LSD causées par des mutations dans des protéines autres
que des enzymes (Winchester, 2013). La nouvelle approche de séquençage ciblé Lysoplex
constitue également une voie prometteuse pour l'identification de mutations pathogéniques
dans 891 gènes en lien avec les lysosomes, l'endocytose et l'autophagie (Di Fruscio et al.,
2015).
Les NCL de même que leur évolution varie considérablement en fonction du soustype de la maladie, de la mutation en cause ainsi que de l'individu. Les premiers signes
cliniques de la maladie apparaissent généralement au bout de plusieurs années, mais parfois
dès la naissance ou au début de l'âge adulte. Des lésions s'accumulent au niveau des
organes entraînant des troubles graves et irréversibles qui provoquent une mort prématurée
après des années de symptômes divers. Parmi ces derniers, on compte un retard de
développement, de la surdité, de la cécité, des désordres moteurs, des problèmes de posture
et des crises d'épilepsie symptomatiques généralisées (Mole et al., 2011). Au quotidien, les
67
parents des enfants atteints doivent gérer de la constipation, des sécrétions orales excessives
accompagnées de difficultés à avaler et mastiquer, des troubles du sommeil et des
problèmes de communications comme l'emploi de thème récurent de conversation, un
manque de fluidité verbale et de la difficulté à initier des phrases (Mole et al., 2011). La
santé mentale et émotionnelle des patients est affectée par l'ensemble des symptômes qui
entraîne de la frustration, de la démence, de la dépression, de l'anxiété et des hallucinations
tant visuelles qu'auditives et sensitives (Mole et al., 2011). Des soins palliatifs sont donc
nécessaires pour assurer une certaine qualité de vie aux personnes atteintes. Une routine
structurée et variée, un environnement familier et une médication comprenant des
antiépileptiques, des antipsychotiques, des laxatifs, des relaxants musculaires, l'installation
de tubes nasogastriques ou une gastrotomie, de la physiothérapie et des activités plaisantes
aident à soulager plusieurs des symptômes associés aux NCL (Mole et al., 2011).
À l'heure actuelle, aucun traitement n'est disponible pour guérir les personnes
atteintes de céroïdes-lipofuscinoses neuronales ou autres maladies de surcharge
lysosomiale. Par contre, de nombreuses stratégies thérapeutiques sont en développement et
montrent des signes prometteurs. Dans plusieurs cas, une thérapie visant le remplacement
de l’enzyme défectueuse par infusion intraveineuse (Enzyme remplacement therapy) permet
de maintenir des niveaux adéquats (Eng et al., 2001). Lorsque la mutation affecte le
repliement plutôt que sa fonctionnalité protéique, il est possible de favoriser le repliement
des
enzymes
thermodynamiquement
instables
en
utilisant
des
chaperonnes
pharmacologiques (Enzyme enhancement therapy) (Desnick, 2004). La thérapie de
réduction du substrat (Substrate reduction therapy) constitue une autre approche qui a pour
but de réduire la synthèse du substrat de l’enzyme faisant défaut de manière à prévenir son
accumulation (Cox, 2005). De façon plus globale, la transplantation de cellules souches de
moelle épinière ou de sang de cordon ombilical s’avère efficace pour certaines maladies de
surcharge lysosomiale comme pour bien d’autres pathologies (Giugliani et al., 2016). Par
contre, l’ensemble de ces thérapies n’affecte pas l’origine génétique des maladies. En ce
sens, la thérapie génique, qui consiste à faire pénétrer des gènes dans les cellules pour
remplacer un gène défectueux, serait la stratégie expérimentale la plus prometteuse.
L’avancement des thérapies repose en grande partie sur l’évolution des connaissances
68
scientifiques relatives à la pathogenèse des maladies. L’identification des gènes impliqués
dans les NCL devrait constituer une priorité de même que l’élucidation de la fonction des
protéines codées par ces gènes, de leurs substrats et de leurs partenaires d'interaction. Une
meilleure compréhension des mécanismes menant aux accumulations cellulaires et à la
mort préférentielle des neurones aiderait grandement au développement pharmaceutique.
4
Problématique, hypothèse et objectifs
4.1
Problématique
Calnuc est une protéine peu caractérisée dont la fonction au niveau du TGN et des
endosomes demeure encore inconnue. Notre groupe a précédemment démontré que Calnuc
influence le recyclage du récepteur LRP9 ce qui laisse croire qu’elle pourrait avoir un rôle
général dans le transport d'autres récepteurs entre les endosomes et le Golgi. La voie
endosomiale est particulièrement importante pour le maintien des lysosomes puisqu’elle est
responsable du transit des divers constituants de l’organite. D’ailleurs, des défauts dans les
protéines responsables de sa structure et de sa fonction ont été associés à des désordres
lysosomiaux, nommés céroïdes-lipofuscinoses neuronales.
4.2
Hypothèse
Notre hypothèse de recherche est que Calnuc constituerait un régulateur important du
transport rétrograde des endosomes vers le Golgi. De ce fait, une altération de Calnuc,
comme une mutation ou une réduction de ses niveaux intracellulaires, causerait un mauvais
triage de récepteurs, dont des récepteurs lysosomiaux, menant à des désordres du lysosome,
comme les céroïdes-lipofuscinoses neuronales.
69
4.3
Objectifs
Article 1 - Impact et mécanisme d'action de Calnuc sur le transport rétrograde des
récepteurs lysosomiaux
Nos études précédentes ont démontré que Calnuc interagit, colocalise et influence le
transport rétrograde de LRP9, un récepteur peu caractérisé qui transite entre le TGN et les
endosomes. Nous étudierons donc l'existence d'une fonction plus générale de Calnuc à ce
niveau en examinant son impact sur CI-MPR et Sortiline. Nous identifierons ensuite le
mécanisme moléculaire responsable de cet effet.
Article 2 - Caractérisation topologique de CLN5 et de ses principaux mutants
pathologiques
Depuis sa découverte, la protéine CLN5 fait l'objet de données contradictoires quant à
sa localisation, sa topologie et sa solubilité. Nous caractériserons donc la protéine CLN5 de
même que quelques-uns de ses mutants les plus fréquents.
Article 3 - Calnuc, un nouveau partenaire d'interaction pour CLN3 et CLN5, est
potentiellement impliqué dans les céroïdes-lipofuscinoses neuronales
Calnuc affecte le transport rétrograde de récepteurs lysosomiaux de manière
semblable à CLN5, une protéine impliquée dans les céroïdes-lipofuscinoses neuronales.
Nous étudierons donc le lien entre ces deux protéines de même que l'implication de Calnuc
dans la pathologie.
70
ARTICLE 1
Calnuc function in endosomal sorting of lysosomal receptors
Auteurs de l’article: Heidi Larkin, Santiago Costantino and Christine Lavoie
Statut de l’article: Publié
Traffic. 2016 Jan 12. doi: 10.1111/tra.12374. [Epub ahead of print]
Avant-propos: J'ai écrit la première version des sections 'Introduction', 'Résultats',
'Matériel et méthodes' ainsi que des descriptions des figures. J'ai réalisé toutes les
expériences retrouvées dans l'article ainsi que tous les montages. Christine Lavoie a
effectué les corrections du texte et a rédigé la discussion. Elle a également contribué à la
prise de photos brutes pour les figures d'immunofluorescence et aux expériences présentées
en figures 1C et S1. La quantification de l'intensité des endosomes a été effectuée par
Santiago Costantino.
71
RÉSUMÉ – ARTICLE 1
Rôle de Calnuc dans le triage endosomial des récepteurs lysosomiaux
Calnuc est une protéine ubiquitaire qui lie le calcium et qui est présente au réseau
trans-golgien (TGN) ainsi qu'aux endosomes. Cependant, son rôle précis à ce niveau est
peu connu. Notre groupe a précédemment mis en évidence le rôle de Calnuc dans le
transport de LRP9, un membre des récepteurs aux lipoprotéines de faible densité (LDL) qui
cycle entre le TGN et les endosomes. L'objectif de la présente étude est d'explorer la
fonction de Calnuc dans le triage endosomial de deux récepteurs lysosomiaux bien
caractérisés, qui cycle aussi entre le TGN et les endosomes, soit le récepteur au mannose-6phosphate (MPR) et Sortiline. Par techniques de biochimie et de microscopie, nous avons
montré que Calnuc lie la queue cytoplasmique des deux récepteurs et colocalise avec eux
sur les endosomes. La déplétion de Calnuc par ARN interférent cause leur redistribution
vers les lysosomes où ils sont dégradés. Ce phénomène résulte d'un défaut dans le
mécanisme de recrutement du complexe Rétromère qui agit comme principal mécanisme de
transport rétrograde des endosomes vers le TGN. Finalement, nous avons montré que la
déplétion de Calnuc altère l'activation et l'association membranaire de Rab7, une petite
protéine G nécessaire pour le recrutement des rétromères aux endosomes. Dans l'ensemble,
nos données montrent que Calnuc est un important joueur du transport rétrograde des
récepteurs lysosomiaux des endosomes vers le TGN via la régulation et le recrutement du
complexe Rétromère.
72
Article 1
Calnuc function in endosomal sorting of lysosomal receptors
Heidi Larkin1, Santiago Costantino2 and Christine Lavoie1‡
Department of Pharmacology, Faculty of Medicine and Health Sciences, Université de
1
Sherbrooke, Sherbrooke, QC, Canada
Centre de recherche de l’Hôpital Maisonneuve-Rosemont, Université de Montréal,
2
Montréal, Canada H1T 2M
Running Title: Calnuc modulates lysosomal receptor trafficking
Key words: Calnuc, nucleobindin, mannose-6-phosphate receptor, Sortiline, lysosomal
receptor, retromers, endosomal sorting, lysosome, Rab7
‡
Correspondence should be addressed to:
Christine Lavoie
Department of Pharmacology
Faculty of Medicine
Université de Sherbrooke
3001—12e Avenue Nord
Sherbrooke, QC, Canada J1H 5N4
Telephone: (819) 820-6868, ext. 12732
Fax: (819) 564-5400
Email: [email protected]
73
Early view
Synopsis
Calnuc is a ubiquitous Ca2+-binding protein which function is poorly characterized. In this
study, we demonstrate that Calnuc plays a role in the endosome-to-TGN retrograde
transport of the lysosomal receptors CI-MPR and Sortilin through the activation and
membrane association of Rab7, a small G protein required for the endosomal recruitment of
retromers.
Abstract
Calnuc is a ubiquitous Ca2+-binding protein present on the trans-Golgi network
(TGN) and endosomes. However, the precise role of Calnuc in these organelles is poorly
characterized. We previously highlighted the role of Calnuc in the transport of LRP9, a new
member of a low density lipoprotein (LDL) receptor subfamily that cycles between the
TGN and endosomes. The objective of this study was to explore the role of Calnuc in the
endocytic sorting of mannose-6-phosphate receptor (MPR) and Sortilin, two wellcharacterized lysosomal receptors that transit between the TGN and endosomes. Using
biochemical and microscopy assays, we showed that Calnuc binds to the cytoplasmic tail of
these receptors through an indirect association and colocalizes with them on endosomes.
74
We found that Calnuc depletion (by siRNA) causes the misdelivery to and degradation in
lysosomes of CI-MPR and Sortilin due to a defect in the endosomal recruitment of
retromers, which are key components of the endosome-to-Golgi retrieval machinery.
Indeed, we demonstrated that Calnuc depletion impairs the activation and membrane
association of Rab7, a small G protein required for the endosomal recruitment of retromers.
Overall, our data indicate a novel role for Calnuc in the endosome-to-TGN retrograde
transport of lysosomal receptors through the regulation of Rab7 activity and the recruitment
of retromers to endosomes.
Introduction
Calnuc, also named NUCB1, is a ubiquitous, well-conserved protein; however, its
precise physiological and cellular functions are poorly understood. Calnuc is a
multifunctional protein comprised of a signal peptide, a putative DNA-binding domain, a
leucine zipper and 2 EF-hand motifs, which both possess the ability to bind Ca2+ (Lin, et
al., 1998). Calnuc is also a multicompartmental protein, with Golgi luminal, extracellular
and cytosolic pools. The predominant Golgi pool of Calnuc is located in the lumen of cisGolgi cisternae, where it acts as a major Ca2+-binding protein and a significant Ca2+ storage
pool (Lin, et al., 1998; Lin, et al., 1999). After a long period of retention in the Golgi,
Calnuc is secreted via constitutive and constitutive-like pathways (Lavoie, et al., 2002). In
addition, a significant cytoplasmic pool of Calnuc (Lin, et al., 1998; Lin, et al., 2000) is
either free or associated with the surface of the Golgi, endosomal membranes and secretory
granules (Brodeur, et al., 2009; Lin, et al., 2009). The multiple domains and localization of
Calnuc potentiates its interactions with various partners, and the growing list of Calnucinteracting partners, such as DNA (Miura, et al., 1992), heterotrimeric Gα proteins (Lin, et
al., 1998) or COX (Ballif, et al., 1996), suggests its importance in the regulation of many
cellular events. Recently, we have shown that Calnuc interacts with the cytosolic tail of a
novel LDLR-related protein known as LRP9 in mice (i.e., LRP10 in humans) (Brodeur, et
al., 2009). We have also demonstrated that LRP9 cycles between the trans-Golgi network
(TGN) and early endosomes (Boucher, et al., 2008) and that Calnuc is essential for
75
retrieving LRP9 from endosomes to the TGN (Brodeur, et al., 2009). These data
highlighted the potential role of Calnuc in endosomal sorting.
Similar to LRP9, other receptors such as Sortilin and cationic-dependent and independent mannose-6-phosphate receptor (CD-MPR and CI-MPR) transit between the
TGN and endosomes (Braulke and Bonifacino, 2009). These lysosomal receptors are
involved in the efficient delivery of soluble digestive enzymes to the lysosome. They bind
newly synthesized lysosomal cargos in the Golgi, and are then packaged into clathrincoated vesicles by the adaptors AP1 and GGA (Golgi-localized, γ-ear-containing, ADPribosylation factor-binding proteins), and anterogradely transported to endosomes
(Bonifacino and Lippincott-Schwartz, 2003). When the receptor/cargo complex reaches this
acidic endosomal environment, the cargo dissociates from the receptor and is transported to
lysosomes, while the unoccupied receptor is then recycled back to the Golgi for another
round of sorting (Bonifacino and Rojas, 2006). This retrograde transport depends on the
retromer complex (Arighi, et al., 2004) composed of two subcomplexes: the vacuolar
protein-sorting (Vps) trimer Vps26/Vps29/Vps35, which participates in cargo recognition,
and a membrane-targeting complex, which is composed of a heterodimeric sorting nexin
(SNX) that consists of an undefined combination of SNX1, SNX2, SNX5 and SNX6
(McGough and Cullen, 2011). The recruitment of the retromer complex is initiated by the
sequential action of the small G proteins Rab5 and Rab7 on endosomes. Through the
recruitment of class III PI3K (Christoforidis, et al., 1999), Rab5 increases endosomal PI3P
levels and consequently increases the membrane recruitment of the SNX subcomplex. The
mammalian SNX dimer is necessary but not sufficient for the recruitment of Vps
subcomplex (Rojas, et al., 2007). Another Rab5 effector is the Rab7 guanine nucleotide
exchange factor (GEF), which recruits and activates Rab7 on endosomal membranes.
Active Rab7 recruits the retromer Vps subcomplex on endosome via direct interactions
with Vps35/Vps29/Vps26 trimer (Rojas, et al., 2008; Seaman, et al., 2009). The
cooperation of Rab5 and Rab7 is thus necessary for retromer recruitment and for the sorting
of lysosomal receptors into retrograde tubules that are targeted to the Golgi. Identifying
novel players involved in the regulation of lysosomal receptor sorting and trafficking is
76
crucial because a loss of regulation can result in aberrant receptor localization and,
subsequently, lysosomal-associated diseases.
In the present study, we explored the role of Calnuc in the trafficking of CI-MPR and
Sortilin, two well-characterized lysosomal receptors that transit between the TGN and
endosomes. We demonstrate the function of Calnuc in the endosomal retrieval of these
receptors through modulating Rab7 activity and the retromer recruitment machinery,
suggesting a general role in the endosomal sorting of receptors.
Materials and Methods
Antibodies and reagents
Anti-actin monoclonal antibodies (mAbs) were purchased from Sigma-Aldrich (Saint
Louis, MO, USA), anti-EEA1 mAbs from BD Transduction Laboratories (Franklin Lakes,
NJ, USA), anti-Flag M2 mAbs from Sigma Aldrich (Saint Louis, MO, USA), anti-GFP
mAbs from Clontech (Mountain View, CA, USA), anti-HA mAbs from Covance
(Emeryville, CA, USA), anti-CI-MPR mAbs from Cedarlane Laboratories (Burlington,
ON, Canada), anti-CI-MPR mAbs from Abcam (Cambridge, MA, USA), anti-Myc mAbs
from Cell signalling Technology (Danvers, MA, USA), anti-Rab7 mAbs and anti-EGFR
pAbs from Cell signalling Technology (Danvers, MA, USA), anti-SNX1 mAbs from BD
Transduction Laboratories (Franklin Lakes, NJ, USA), anti-SNX2 mAbs from BD
Transduction Laboratories (Franklin Lakes, NJ, USA), anti-Sortilin (Neurotensin receptor
3) mAbs from BD Transduction Laboratories (Franklin Lakes, NJ, USA), anti-TfR mAbs
from Invitrogen (Carlsbad, CA, USA). Anti-Calnuc polyclonal antibodies (pAbs) were
purchased from Aviva Systems Biology (San Diego, CA, USA), anti-CatD pAbs from
Calbiochem (La Jolla, CA, USA), anti-CD8 pAbs from Santa Cruz Biotechnology (Santa
Cruz, CA, USA), anti-EEA1 pAbs from Thermo Scientific (Rockford, IL, USA), anti-Flag
pAbs from Sigma Aldrich (Saint Louis, MO, USA), anti-GFP pAbs from Molecular Probes
(Eugene, OR, USA), anti-hVps26 pAbs from Abcam (Cambridge, MA, USA).
77
DNA constructs
Mammalian expression vector pcDNA3.1 containing Calnuc-GFP fusion protein or
ΔSP-Calnuc (Calnuc without signal peptide) are described elsewhere (Weiss, et al., 2001).
These constructions were used to generate Calnuc-HA, Calnuc--Calnuc-Flag. 3xHA-Rab5
and HA-Rab7 were obtained by subcloning of human Rab5 and Rab7 sequences purchased
from Addgene.
Cell culture and transfection
HeLa cells were purchased from the American Type Culture Collection (Manassas,
VA, USA), COS7 cells from Dr Klaus Hahn (University of North Carolina, Chapel Hill,
NC, USA) and HEK293T cells were obtained from Dr. Alexandra Newton (University of
California, San Diego, CA, USA). HeLa cells stably expressing GFP-rab7 were obtained
from Matthew Seaman (University of Cambridge). The cells were grown in Dulbecco’s
modified Eagle’s high glucose medium (Invitrogen, Carlsbad, CA, USA) containing 10%
fetal bovine serum (FBS) (Hyclone Laboratories, Logan, UT, USA) and 1% penicillin,
Glutamine and streptomycin (Invitrogen, Carlsbad, CA, USA). The stable cell line was also
supplemented with 0.5 mg/ml G418 (Invitrogen, Carlsbad, CA, USA). HeLa cells and the
HEK cells were transfected with Lipofectamine 2000 transfection reagent (Invitrogen,
Carlsbad, CA, USA) according to the manufacturers’ instructions.
Coimmunoprecipitation
HEK293 cells were plated in 60-mm culture dishes and transfected with the various
constructs. After 48 h, the cells were lysed in 50 mM Tris buffer (pH 7.4) containing 100
mM NaCl, 1% NP-40, and protease inhibitors. Cells were incubated for 1 h at 4oC and then
centrifuged at 15,800xg for 20 min. The cleared supernatants were incubated with primary
antibodies overnight at 4oC and then with protein A-sepharose (GE Healthcare, Piscataway,
NJ, USA) or protein G-Sepharose (Zymed, San Francisco, CA, USA) for 1 h. The beads
were washed three times in lysis buffer and boiled in Laemmli sample buffer. Bound
immune complexes were analyzed by SDS-PAGE and immunoblotting.
78
Glutathione S-transferase (GST) pull-down assays
GST fusion proteins were expressed in E. coli BL21 and were purified on
glutathione-Sepharose 4B beads (Pharmacia, Piscataway, NJ, USA) according to the
manufacturer’s instructions. Lysate from HeLa cells obtained as described above was
incubated overnight at 4oC with 20-100 μg of GST fusion proteins immobilized beads. The
beads were washed three times in lysis buffer and boiled in Laemmli sample buffer. The
bound proteins were separated by SDS-PAGE and detected by autoradiography or
immunoblotting.
Immunoblotting
The protein samples were separated on 10% or 12% SDS-PAGE gels and transferred
to nitrocellulose membranes (Perkin Elmer, Woodbridge, ON, Canada). The membranes
were blocked in Tris-buffered saline (20 mM Tris-HCl, pH 7.5, 150 mM NaCl) containing
0.1% Tween 20 and 5% nonfat dry milk, bovine serum albumin (BSA), or foetal bovine
serum (FBS) and incubated with primary antibodies for 1 h at RT and then with horseradish
peroxidase-conjugated goat anti-rabbit or anti-mouse IgG (Bio-Rad, Richmond, ON,
Canada) and enhanced chemiluminescence detection reagent (Pierce Chemical, Rockford,
IL, USA).
RNA Interference and Rescue
Nonspecific (CTL) siRNA scrambled II duplex and Calnuc siRNA were purchased
from Dharmacon Research (Chicago, IL, USA). HeLa cells were transfected with a final
concentration of 100 nM siRNA duplex using Lipofectamine 2000 reagent (Invitrogen)
according to the manufacturer’s instructions. The cells were analyzed 72 h after the
transfection. Tagged proteins were transfected with Fugene 12 h before the
immunofluorescence experiments. Reversal of phenotype (rescue) was performed as
described in (Brodeur, et al., 2009). Briefly, HeLa cells were transfected with cDNA
encoding rat Calnuc-GFP, rat ΔSP-Calnuc-GFP or GFP alone using Fugene 8–10 h after
the initial transfection with human Calnuc siRNA, and the cells were analyzed after 38–40
h.
79
Cycloheximide chase
Mock- and siRNA-treated cells were incubated at 37ºC in complete DMEM
containing 25 mM Hepes buffer, pH 7.4, and 40 μg/ml of cycloheximide (Sigma-Aldrich,
Saint Louis, MO, USA). At the corresponding time points, the cells were lysed as described
above, and analyzed by SDS-PAGE and immunoblotting.
Treatment with lysosomal inhibitors
Mock- and siRNA-treated cells were incubated for 3h at 37ºC in complete DMEM
containing 40 μg/ml of cycloheximide (Sigma-Aldrich, Saint Louis, MO, USA), 1 mg/ml of
leupeptin (Roche Diagnostics, Indianapolis, IN, USA), 100 μM of pepstatin and 20 μg/ml
of E64 (Sigma-Aldrich, Saint Louis, MO, USA). The cells were collected, lysed, and
analyzed by SDS-PAGE and immunoblotting.
Immunofluorescence
HeLa cells were plated on coverslips. Twelve hours after the transfection, the cells
were fixed for 30 min in 3% paraformaldehyde (PFA) 100 mM phosphate buffer, pH 7.4,
permeabilized with 0.1% Triton X-100 for 10 min, blocked with 10% goat or fetal bovine
serum for 30 min, and incubated with primary antibodies for 1 h at RT, followed by Alexa
Fluor-488, 594 or 647-conjugated antibodies (Molecular Probes, Eugene, OR, USA) for 1 h
at RT. The specimens were visualized using an inverted confocal laser-scanning
microscope (FV1000, Olympus, Tokyo, Japan) equipped with a PlanApo 60x/1.42 oil
immersion objective (Olympus, Tokyo, Japan). Olympus Fluoview software version 1.6a
was used for image acquisition and analysis. The images were further processed using
Adobe Photoshop (Adobe Systems, San Jose, CA, USA). The quantification of the
immunofluorescence signal on endosome was previously described (Mamo, et al.).
Cell fractionation (S100/P100 centrifugation)
HeLa cells were washed in PBS, collected into homogenization buffer (50 mM Tris,
pH 7.4, 150 mM NaCl, 5 mM ethylenediaminetetraacetic acid (EDTA) and 200 mM
sucrose with protease inhibitors), and centrifuged for 5 min at 500×g. The pellet was
homogenized by 60 passages through a 25 G 5/8 needle and centrifuged at 1000×g for 10
80
min to remove nuclei and unbroken cells. The post-nuclear supernatant was collected and
centrifuged at 100,000×g for 1 h at 4ºC. The soluble (cytosolic fraction, S100) was
collected, and the pellet (membrane fraction, P100) was resuspended using a pestle in an
equal volume of buffer. The same volumes of membrane and cytosolic fractions were
analyzed by SDS-PAGE and immunoblotting.
Statistical analysis
Experiments were performed at least in triplicate and results are expressed as means
intensity of protein normalised with loading control ± SD. Statistical significance between
two groups was assessed using the Student t-test whereas ANOVA was used for
comparison with three groups or more. P value < 0.05 was considered significant (*) and p
< 0.001 very significant (**).
Results
Calnuc interacts and colocalizes with the lysosomal sorting receptors Sortilin and CIMPR.
Because Calnuc has been previously shown to interact with and alter the endosomal
sorting of LRP9 (Brodeur, et al., 2009), we first examined whether Calnuc could interact
with Sortilin and CI-MPR, two receptors that follow the same trafficking pathway as LRP9.
To verify the specificity of these interactions, we also examined whether Calnuc bound to
epidermal growth factor receptor (EGFR), which does not cycle between the TGN and
endosomes. We first performed immunoprecipitation experiments using HEK293 cells
expressing HA-tagged Calnuc alone or cells expressing GFP-tagged Calnuc or GFP
together with Myc-tagged Sortilin or EGFR (Figure 1A). We found a weak but highly
reproducible interaction between Calnuc-HA and endogenous CI-MPR as well as between
Calnuc-GFP and Myc-Sortilin using anti-CI-MPR or anti-GFP IgG immunoprecipitation,
respectively (Figure 1A). However, no interaction was detected between Calnuc-GFP and
EGFR using anti-GFP IgG immunoprecipitation (Figure 1A). Furthermore, endogenous
Calnuc interacts with endogenous CI-MPR (Figure 1B). Because the cytosolic tail of LRP9
was previously shown to interact with Calnuc (Brodeur, et al., 2009), we then performed
81
Figure 1 - A small proportion of Calnuc interacts with the cytoplasmic tails of CIMPR and Sortilin and colocalizes with CI-MPR and Sortilin on endosomes.
(A) Coimmunoprecipitation of Calnuc with full-length CI-MPR and Sortilin but not with
82
EGFR. Lysates from HEK cells transfected with Calnuc-HA alone or with GFP or Calnuc–
GFP together with Myc-Sortilin or EGFR were immunoprecipitated with control (lane 1),
anti-CI-MPR (lane 2), anti-GFP (lanes 3-6) antibodies and then immunoblotted with antiHA or anti-GFP to detect Calnuc, with anti-Myc to detect Sortilin, with anti-CI-MPR or
anti-EGFR. Calnuc-GFP and Calnuc-HA are observed as two bands, the lower of which
most likely represents an N-terminal cleavage product. (B) Coimmunoprecipitation of
endogenous Calnuc and CI-MPR. Lysates from HeLa cells were immunoprecipitated with
control (CTL) or anti-CI-MPR antibodies and then immunoblotted with anti-Calnuc and
anti-CI-MPR. (C) Coimmunoprecipitation of Calnuc-GFP with CD8-CI-MPR and CD8Sortilin reporter proteins containing the cytoplasmic domains (C-term) of these receptors.
Lysates from HEK cells transfected with CD8-CI-MPR or CD8-Sortilin and GFP or
Calnuc-GFP were immunoprecipitated with anti-GFP (lanes 1-4) and then immunoblotted
with anti-CD8 to detect CI-MPR C-term and Sortilin C-term or with anti-GFP to detect
GFP and Calnuc. (D) Comparison of the intracellular distribution of endogenous Calnuc
and CI-MPR. HeLa cells transfected with GFP-tagged Rab5Q79L (to create enlarged
endosomes) were fixed, permeabilized, immunostained with anti-Calnuc and anti-CI-MPR
and examined by confocal fluorescence microscopy. Calnuc was found mainly in the Golgi
cisternae (arrowheads), and a small amount was also detected in Rab5-labeled endosomes
(b, e). CI-MPR was localized in Rab5-labeled endosomes (c, d). The merged image (f)
shows a partial overlap between Calnuc and CI-MPR in specific membrane microdomains
of endosomes. Scale bar, 10 μm. (E) Comparison of the intracellular distribution of the
cytoplasmic pool of Calnuc and CI-MPR or Sortilin. COS7 cells were transfected with
Calnuc devoid of its signal sequence (ΔSP-Calnuc-GFP), which is only expressed in the
cytosol, in the absence (a, b) or presence of Myc-tagged Sortilin (c, d). (a, c) ΔSP-CalnucGFP was mainly detected at the plasma membrane and the cytosolic side of the Golgi
cisternae (left insets), and a small amount was found on vesicles corresponding to
endosomes (right insets). The merged image (yellow) shows a partial overlap between
ΔSP-Calnuc-GFP and CI-MPR (a, right inset) or Myc-Sortlin (c, right inset) on the
endosome surface. (b, d) ΔSP-Calnuc-GFP partially colocalized with endogenous CI-MPR
and Myc-Sortilin on enlarged early endosomes created by the expression of Rab5Q79L (right
insets). Twelve hours after transfection, the cells were fixed, permeabilized and
immunostained using anti-GFP (a-d) and anti-CI-MPR (a-b) or anti-Myc antibodies (c-d).
The labeled cells were examined by confocal fluorescence microscopy. Scale bar, 10 μm.
co-immunoprecipitation experiments using the cytosolic tails of both CI-MPR and Sortilin
fused to the reporter protein CD8 (Cluster of differentiation 8), a transmembrane
glycoprotein that allows adequate trafficking along the cell (Seaman, 2004) and acts as a
tag for detection. HEK293 cells were transfected with GFP-tagged Calnuc or GFP together
with CD8-MPR or CD8-Sortilin (Figure 1C), and the proteins were immunoprecipitated
using an antibody against GFP. The results showed that Calnuc-GFP, but not GFP alone,
interacts with the cytosolic tails of both CI-MPR and Sortilin (Figure 1C). However, using
in vitro translation pull-down assays, we were unable to detect a direct interaction between
83
Calnuc and the cytosolic tails of CI-MPR and Sortilin (data not shown), suggesting an
indirect interaction between Calnuc and the lysosomal receptors.
To determine whether Calnuc colocalizes with these lysosomal receptors, we next
compared the intracellular distribution of endogenous Calnuc and CI-MPR in HeLa cells
using confocal microscopy. In order to help visualize the colocalization of Calnuc and CIMPR, enlarged endosomes were created by expressing Rab5 GTPase-deficient mutant
(Rab5Q79L) that increases homo- and heterotypic fusion, leading to the formation of
enlarged early endosomes (Stenmark, et al., 1994). As previously reported (Lin, et al.,
1998; Brodeur, et al., 2009), endogenous Calnuc was predominantly localized in the Golgi
region (Figure 1D (arrowheads, g)). However, a small amount of Calnuc was also detected
in enlarged Rab5-labeled endosomes (Figure 1D(b,e)). Endogenous CI-MPR was also
localized in Rab5-labeled endosomes (Figure 1D(c,d)) and partially overlaps with Calnuc
in specific membrane microdomains (Figure 1D(f)). However, these images did not
indicate whether it is the luminal or cytoplasmic pool of Calnuc that colocalizes with CIMPR. Given that Calnuc interacts with the cytoplasmic tail of MPR, we next examined
whether the specific cytosolic pools of Calnuc and CI-MPR colocalize. COS7 cells were
transfected with Calnuc-GFP lacking its signal peptide (ΔSP-Calnuc-GFP), which prevents
its insertion into the lumen of the Golgi and limits its distribution to the cytoplasm. As
previously reported (Brodeur, et al., 2009), ΔSP-Calnuc-GFP accumulated mainly in the
cytoplasm and plasma membrane but was also detected at the surface of Golgi (Figure
1E(a), left inset) and of endosomes (Figure 1E(a), right inset). A small proportion of this
pool of Calnuc partially colocalized with CI-MPR at the surface of endosomes (Figure
1E(a), right insets). This endosomal colocalization was confirmed in cells co-expressing a
Rab5 GTPase-deficient mutant (Rab5Q79L) (Figure 1E(b)). The same confocal analyses
were performed in COS7 cells expressing Myc-Sortilin together with ΔSP-Calnuc-GFP in
the absence or presence of Rab5Q79L (Figure 1E(c, d)). As observed with CI-MPR, partial
colocalization between ΔSP-Calnuc-GFP and Myc-Sortilin was found at the surface of
endosomes (Figure 1E(c,d), right insets). Together, these immunofluorescence and
biochemical interaction assays suggest that a small proportion of the cytoplasmic pool of
Calnuc indirectly interacts with the C-terminal tails of MPR and Sortilin on endosomes.
84
Calnuc is implicated in the trafficking of the lysosomal sorting receptors Sortilin and
CI-MPR.
To investigate the functional role of Calnuc in CI-MPR and Sortilin trafficking, we
examined the steady-state distribution of these lysosomal receptors in Calnuc-depleted
cells. HeLa cells were transfected with control or Calnuc siRNA alone or together with
Myc-Sortilin and were then analyzed by confocal microscopy. Calnuc was strongly (≥
90%) depleted at 72 h after transfection with a Calnuc-specific small interfering RNA
(siRNA), as observed in immunofluorescence (Figure 2A) and immunoblotting
experiments (Figure 2B). In Calnuc knockdown cells, the intensities of the
immunofluorescence signals for endogenous CI-MPR and Myc-Sortilin (Figure 2A(d,e))
were reduced compared with control cells (Figure 2A(a,b)). These results were confirmed
by western blotting analysis. The steady-state levels of endogenous CI-MPR and MycSortilin were reduced in Calnuc knockdown cells compared with control cells (Figure 2B).
In contrast, the level of Transferrin receptor (TfR), a plasma membrane receptor that does
not traffic between the TGN and endosomes, was not affected by Calnuc depletion (Figure
2B). The specificity of the effect of Calnuc on the levels of these lysosomal receptors was
also validated by a rescue experiment with siRNA-resistant forms of wild-type and
cytosolic Calnuc (ΔSP-Calnuc). Rat Calnuc was used as a rescue construct because it
differs in many nucleotides from human siRNA target sequences, rendering it resistant to
the siRNA. The results showed that reintroducing rat Calnuc-GFP or ΔSP-Calnuc-GFP into
Calnuc-depleted cells restored the basal levels of CI-MPR to levels similar to those of
control cells (Figure 3A, B). These results suggest that the cytoplasmic pool of Calnuc
participates in the intracellular trafficking of lysosomal receptors.
To evaluate the impact of Calnuc depletion on CI-MPR and Sortilin stability, we
performed cycloheximide chase experiments in which the levels of CI-MPR and Sortilin in
control and Calnuc siRNA-treated cells were examined at different times after protein
synthesis inhibition (Figure 2C). CI-MPR and Sortilin were degraded more rapidly in the
Calnuc-depleted cells than in the control cells (Figure 2C). In mock cells, the half-lives of
CI-MPR and Sortilin were greater than 6 h, whereas the half-lives of these receptors were
85
Figure 2 - CI-MPR and Sortilin levels are reduced when Calnuc is depleted due to
missorting to lysosomes.
HeLa cells were transfected with control or Calnuc siRNA for 3 days and Myc-Sortilin
cDNA was transfected 12 h before the experiment. (A) Confocal microscopy analysis of
86
CI-MPR and Myc-Sortilin distribution in control or Calnuc-depleted cells. Cells were
fixed with PFA and immunostained with anti-CI-MPR (a, d), anti-Myc (b, e) and antiCalnuc (c, f) and confocal images were acquired using identical instrument settings. CIMPR and Myc-Sortilin localized mainly in the perinuclear region in control cells, and
Calnuc knockdown induced a decrease in the intensity level of both receptors. Bars, 10
μm. (B). Western blot analysis of control and Calnuc-depleted cell lysates. Proteins were
separated by SDS-PAGE and immunoblotted with anti-CI-MPR, anti-Myc, antiTransferrin receptor (TfR), anti-Early endosome antigen 1 (EEA1) and anti-Calnuc. The
steady-state levels of endogenous CI-MPR and Myc-Sortilin, but not TfR, were reduced
in Calnuc-depleted cells. (C) The half-lives of CI-MPR and Sortilin are shorter in Calnucdepleted cells than in control cells. Hela cells were incubated with 40 µg/ml of
cycloheximide for the time indicated, and total cell lysates were subjected to western blot
analysis, as described in (B). (D) Endogenous CI-MPR is degraded in lysosomes in
Calnuc-depleted cells. Control and Calnuc-depleted HeLa cells were treated or not with
the lysosomal inhibitors leupeptin (1 mg/ml), E64 (20 μg/ml) and pepstatin (100 μM) for
5 h. The cells were lysed, and the proteins were analyzed by western blotting, as
described in (B). EEA1 was used as loading control. (E) Bar graph showing the
quantification of CI-MPR degradation in cells treated as described in (D) and expressed
as a percentage of the CI-MPR present in control cells. The results are shown as the
means ± SD (n = 6). There is no significant difference between columns 1 and 3 (P>0.05);
**p < 0.01 (compared with control cells).
reduced to <4 h in Calnuc siRNA-treated cells (Figure 2C). To determine whether this
degradation occurred in lysosomes, we incubated the siRNA-treated cells with the
lysosomal inhibitors leupeptin, pepstatin and E64 for 5 h. As shown in Figures 2D and 2E,
treatment with lysosomal inhibitors significantly prevented the degradation of endogenous
CI-MPR. Forty-one percent of the CI-MPR pool was degraded in the Calnuc knockdown
cells in the absence of lysosomal inhibitors, whereas the CI-MPR level was similar to that
in the control cells in the presence of lysosomal inhibitors (Figure 2E). Lysosomal inhibitor
treatment thus significantly prevented a decrease in CI-MPR levels. Taken together, these
observations suggest that depleting Calnuc increases the delivery of CI-MPR and Sortilin to
lysosomes. The shorter half-lives of these lysosomal receptors should result in their
depletion from the TGN, leading to the missorting of newly synthesized lysosomal cargo
proteins, such as Cathepsin D or prosaposin. An immunoblot analysis indicated increased
amounts of the precursor and intermediate forms of Cathepsin D in the Calnuc-depleted
cells compared to the control cells (Supplementary Fig. S1), which was indicative of
impaired transport of the enzyme to lysosomes. Overall, these results are consistent with
87
the role of Calnuc in regulating the endosomal sorting of lysosomal receptors and in the
proper delivery of their cargo.
Figure 3 - Expression of the cytoplasmic form of Calnuc rescues the lysosomal
degradation of CI-MPR.
(A) Steady state levels of CI-MPR are increased in Calnuc-depleted cells rescued with
siRNA-resistant full-length Calnuc-GFP or cytoplasmic Calnuc-GFP (ΔSP-Calnuc-GFP)
unlike cells rescued with GFP. HeLa cells treated with human Calnuc siRNA were
transfected with GFP, siRNA-resistant Calnuc-GFP or ΔSP-Calnuc-GFP at 5 h after
knockdown. The cells were lysed 3 days later, and the proteins were analyzed by western
blotting. Endogenous human Calnuc was below the detection level in the samples treated
with the Calnuc-specific siRNA. (B) Bar graph showing the quantification of CI-MPR
degradation in cells treated as described in (A) and expressed as a percentage of CI-MPR
present in control siRNA cells. Results are shown as means ± SD (n=4). *p<0.05
(compared with control siRNA cells). There is no significant difference between columns
1 and 3 (P>0.05) and columns 1 and 4 (P>0.05).
Calnuc is required for the recruitment of retromers to endosomes.
Given that Calnuc depletion induces the misdelivery and degradation of CI-MPR and
Sortilin to lysosomes, we reasoned that Calnuc is specifically involved in the endosomal
sorting step that targets these receptors to retrograde carriers. The key component of the
machinery involved in endosomal cargo capture and retrieval is the heteropentameric
88
retromer complex. Accordingly, retromer depletion leads to an effect on CI-MPR and
Sortilin stability and localization that is similar to that observed upon Calnuc depletion
(Arighi et al. 2004; Rojas et al. 2007). Therefore, we examined whether Calnuc depletion
affects the intracellular distribution of retromer subunits in HeLa cells. Two HeLa cell
populations were transfected with either control or Calnuc-specific siRNA. After 48 hours,
the two populations of cells were mixed and subsequently fixed and stained with antiVps26, anti-SNX2 and anti-Calnuc antibodies (Figure 4A). This allows a direct side-byside comparison of control versus Calnuc-suppressed cells by confocal microscopy. A
decrease in the intensity of endogenous hVps26 and SNX2 immunofluorescence staining
was observed in Calnuc-depleted cells compared to control cells (Figure 4A). Z-stack
imaging was performed on these cells to confirm that the difference in Vps26 and SNX2
staining was observed in all the focal planes of the cells (Supplementary Fig. S2). This
observation was evaluated quantitatively using an ad hoc algorithm that was previously
developed to specifically detect and quantify the fluorescence intensity of endosomes
(Mamo, et al., 2012). This quantification revealed a 35% and 30% decrease in SNX2 and
hVps26 signals, respectively, on endosomes in the Calnuc-depleted cells compared to the
control
cells
(Figure
4B).
In
contrast,
no difference was
observed in
the
immunofluorescence staining intensity of the early endosomal marker EEA1 between the
control and Calnuc knockdown cells (Figure 4A, B), indicating that the observed effect on
retromers was not due to non-specific or general effects of Calnuc depletion on endosomes.
Furthermore, the observed decrease in endosomal retromer intensity was not due to
degradation because immunoblot analyses did not reveal differences in SNX1, SNX2,
hVps26 or hVps35 protein levels between the control and Calnuc knockdown cells (Figure
4C). Altogether, these results suggest a specific role for Calnuc in the trafficking of
lysosomal receptors by controlling the recruitment of retromers to endosomal membranes.
We next investigated whether the observed effect of Calnuc on retromer recruitment was
due to the direct action of Calnuc on the retromer machinery. We first examined whether
Calnuc colocalizes with SNX2 and hVps35 retromer components using confocal
microscopy. Because retromer subunits are cytosolic proteins, the immunofluorescence
experiments were performed using the cytosolic form of Calnuc,
89
Figure 4 - Calnuc depletion alters the recruitment of retromers to endosomes.
(A) Knocking down Calnuc affects the intracellular distribution of retromers but not of
EEA1. Mixed control and Calnuc siRNA-treated HeLa cells were fixed, permeabilized
and triple-labeled with anti-hVps26 or anti-SNX2 together with anti-EEA1 and antiCalnuc antibodies followed by Alexa Fluor-488, 594 and 647-conjugated antibodies. The
stained cells were then examined by confocal fluorescence microscopy. Scale bar, 10 μm.
90
(B) Quantification of the relative fluorescence intensities of retromers on endosomes in
control and Calnuc-depleted cells. The data represent the relative fluorescence intensities
of SNX2, Vps26 and EEA1 for more than 15,000 endosomes per condition. The results
are expressed as a percentage of arbitrary units and are shown as the means ± SD (n = 3).
* p < 0.05 compared with control siRNA cells. (C) The levels of endogenous retromers
are not affected by the depletion of Calnuc. HeLa cells were transfected as described in
(A) and lysed. The proteins were separated by SDS-PAGE and detected with specific
anti-SNX1, anti-SNX2, anti-hVps26, anti-hVps35 and anti-EEA1 and anti-Calnuc
antibodies. (D) Calnuc colocalizes with retromers on endosomes. COS7 cells transfected
with ΔSP-Calnuc-GFP (cytoplasmic form of Calnuc) in the absence (a) or presence of
Myc-hVps35 (b) were fixed, permeabilized and immunostained with anti-GFP (a, b), antiSNX2 (a) and anti-Myc (b). The stained cells were then examined by confocal
fluorescence microscopy. ΔSP-Calnuc-GFP was found at the plasma membrane, the
cytosolic side of the Golgi and small vesicular structures in the cytoplasm (insets).
Endogenous SNX2 (a) and Myc-hVps35 (b) were found on endosomes distributed
throughout the cell. The merged image shows partial overlap in the vesicular distribution
of ΔSP-Calnuc-GFP and SNX2 (a) or Myc-hVps35 (b). Scale bar, 10 μm.
ΔSP-Calnuc-GFP, transfected either alone (Figure 4D(a)) or with Myc-hVps35 (Figure
4D(b)) into COS7 cells. Both endogenous SNX2 (Figure 4D(a)) and Myc-hVps35 subunits
(Figure 4D(b)) were located on punctated structures corresponding to Golgi surrounding
vesicles and endosomes. A partial colocalization between ΔSP-Calnuc-GFP and
endogenous SNX2 (Figure 4D(a)) as well as with Myc-hVps35 (Figure 4D(b)) was
observed on endosomes. However, we were unable to detect an interaction between Calnuc
and any subunit of the retromer complex (data not shown). Although these results negate
the possibility that Calnuc is an adaptor that recruits retromers to endosomes, they do
suggest that Calnuc has an indirect role by possibly acting on the machinery that regulates
the recruitment of retromers.
Calnuc regulates the localization and activity of Rab7.
To identify the molecular mechanism by which Calnuc influences the recruitment of
retromers, we next investigated Calnuc’s function with regard to the machinery known to
recruit retromers to endosomal membranes. Recently, it was shown that the activation and
cooperation of Rab5 and Rab7 are required for the recruitment of Vps retromers to
endosomes as well as for lysosomal receptor retrieval (Rojas, et al., 2008; Seaman, et al.,
2009). Because we obtained similar effects with Calnuc depletion, we first examined the
role of Calnuc in Rab5 and Rab7 localization and activity.
91
Figure 5 - Calnuc partially interacts and colocalizes with Rab7 on endosomes.
(A) Comparison of the intracellular distribution of Calnuc with Rab5, Rab7 or Rab9.
COS7 cells transfected with ΔSP-Calnuc-Flag (cytoplasmic form of Calnuc) together
with HA-tagged Rab5, Rab7 or Rab9 were fixed, permeabilized and immunostained with
anti-Flag (a, d, g), and anti-HA (b, e, h). ΔSP-Calnuc-GFP was distributed at the plasma
membrane, the cytosolic side of the Golgi and small vesicular structures in the cytoplasm
92
(insets) (a, d, g). Rabs were found on endosomes distributed throughout the cell. The
merged images show partial overlap in the vesicular distribution of ΔSP-Calnuc-GFP and
Rab5 (c) or Rab7 (f) and also Rab9 (i). Scale bar, 10 μm. (B) Coimmunoprecipitation of
Flag-tagged Calnuc with HA-tagged Rab7 but not with HA-tagged Rab5 or HA-tagged
Rab9. Lysates from HEK cells transfected with Calnuc-Flag together with HA, 3xHARab5, HA-Rab7 or HA-Rab9 were immunoprecipitated with an anti-Flag antibody and
then immunoblotted with anti-HA or anti-Flag to detect Rab proteins or Calnuc,
respectively. The 3xHA tag added ≈3-4 kDa to the MW of Rab5 which explain its higher
molecular weight. (C) The interaction of Calnuc and Rab7 is independent of the
activation status of Rab7. Lysates from HEK cells transfected with Calnuc-Flag together
with HA, wild-type HA-Rab7, dominant-active HA-Rab7Q67L or dominant-inactive HARab7T22N were immunoprecipitated and immunoblotted as described in (B).
Using confocal microscopy, we examined the presence of Calnuc on Rab5- and
Rab7-labeled endosomes in COS7 cells (Figure 5A). Rab9 was also examined because it is
involved in late endosome-to-TGN trafficking (Lombardi, et al., 1993) but is not involved
in retromer recruitment (Seaman, et al. 2009). ΔSP-Calnuc-Flag was mainly localized on
the plasma membrane, Golgi region and cytoplasm (Figure 5A(a,d,g)). A minor pool of
ΔSP-Calnuc-Flag was also detected on some punctate structures distributed throughout the
cytoplasm that partially overlapped with HA-tagged Rab5 (Figure 5A(c)), HA-tagged Rab7
(Figure 5A(f)) as well as HA-tagged Rab9 (Figure 5A(i)). We next investigated whether
Calnuc could form a protein complex with Rab5, Rab7 or Rab9 (Figure 5B). HEK293 cells
were transfected with Calnuc-Flag together with HA-Rab5, HA-Rab7 or HA-Rab9.
Following immunoprecipitation with anti-Flag antibodies, an interaction between CalnucFlag and HA-Rab7 but not between Calnuc-Flag and HA-Rab5 or HA-Rab9 was observed
(Figure 5B), suggesting that Calnuc interacts with Rab7 on endosomes. However, using in
vitro translation pull-down assays, we were unable to detect a direct interaction between
Calnuc and Rab7 (data not shown). We next examined whether the Calnuc/Rab7 interaction
depends on the activation state of Rab7. We tested the interaction of Calnuc-Flag with the
dominant-active form of Rab7 (HA-Rab7Q67L) and the dominant-inactive form of Rab7
(HA-Rab7T22N) (Figure 5C). Coimmunoprecipitation experiments showed that wild-type,
active and inactive Rab7 bound Calnuc at similar levels (Figure 5C), suggesting that the
Rab7 activation state did not influence this interaction. Similarly, to determine whether the
interaction of Calnuc and Rab5 is GTP-dependent, we examined the interaction of Calnuc
93
Figure 6 - Rab7 localization and activity is altered in Calnuc-depleted cells.
(A) Calnuc knockdown alters the intracellular distribution of Rab7. HeLa cells stably
expressing GFP-Rab7 were transfected with control or Calnuc siRNA. After 48 hours, the
two populations of cells were mixed and subsequently fixed, permeabilized and triple94
labeled with anti-GFP, anti-EEA1 and anti-Calnuc. The stained cells were then examined
by confocal fluorescence microscopy. Scale bar, 10 μm. (B) The fluorescence intensity of
endosomal GFP-Rab7 and EEA1 was quantified as described in Figure 4B. The data
represent the relative fluorescence intensities of GFP-Rab7 and EEA1 for more than
15,000 endosomes per condition. The results are expressed as a percentage of arbitrary
units and are shown as the means ± SD (n = 3). ** p < 0.001 compared with control
siRNA cells. (C) Calnuc depletion does not affect the total levels of GFP-Rab7 or EEA1.
Lysates from cells treated with control or Calnuc-specific siRNA were analyzed by
western blotting with specific anti-GFP, anti-EEA1 and anti-Calnuc antibodies. (D)
Depletion of Calnuc increases the amount of Rab7, hVps26 and SNX2 in the cytosolic
fraction. HeLa cells treated with control or Calnuc siRNA were fractionated into
membrane (P) and cytosolic (S) fractions as described in Material and Methods, followed
by immunobloting with antibodies against Rab7, Vps26, SNX2, Calnuc, EEA1, and TfR.
(E) Quantification of the amount of Rab7 and Vps26 in the cytosolic and membrane
fractions of control and Calnuc-depleted cells. Data (n=3) are presented as percent of total
protein (S+P).
and dominant-active (Rab5Q79L) and inactive (Rab5S34N) forms of Rab5 (Supplementary
Fig. S3). The inability to detect an interaction negates the possibility that Calnuc is a Rab5
effector. These results suggest that Calnuc is not an effector of Rab5 and Rab7 but may be
part of the Rab7 activation machinery.
To investigate the function of the Calnuc/Rab7 interaction, we used confocal
microscopy to compare the intracellular distribution of Rab7 in HeLa cells stably
expressing GFP-tagged Rab7 and transfected with control or Calnuc siRNA (Figure 6A).
After mixing the two cell populations for a direct side-by-side staining comparison, a
decrease in the intensity of GFP-Rab7 immunofluorescence staining on punctate structures
was observed in the Calnuc-depleted cells compared to the control cells (Figure 6A). Zstack imaging was performed on these cells to confirm that the difference in Rab7 signal
intensity was observed in all focal planes of the cells (Supplementary Fig. S4).
Quantification of the endosomal fluorescence intensity revealed a 28% decrease in the
Rab7 signal on endosomes in the Calnuc-depleted cells compared to the control cells
(Figure 6B), whereas no difference was observed in the endosomal intensity of EEA1 in the
same cells (Figure 6A, B). Western blotting using whole-cell lysates revealed that the total
amounts of GFP-Rab7 in the control and Calnuc-depleted cells were similar (Figure 6C),
suggesting that the change in Rab7 endosomal intensity was not due to the deregulation of
95
Rab7 protein synthesis or degradation. To confirm that the redistribution of Rab7 observed
by immunofluorescence resulted from a change in the amount of these proteins that was
associated with membranes, HeLa cells treated with control or Calnuc siRNA were
fractionated into membrane (pellet, P) and cytosolic (supernatant, S) fractions and analyzed
by SDS-PAGE and immunoblotting (Figure 6D). The loss of Calnuc expression resulted in
a significant shift of Rab7 to the supernatant fraction, consistent with a redistribution of this
protein to the cytoplasm. In agreement and as shown in Figure 4, less membrane-bound and
more cytosolic hVps26 and SNX2 was found in the Calnuc-depleted cells (Figure 6D), and
this effect was found to be specific because Calnuc depletion did not affect the membrane
distribution of other endosomal markers such as EEA1 (Figure 6D). The membrane protein
TfR was detected in the pellet fractions, indicating that fractionation did not cause
membrane into the supernatant fraction. The data from three separate experiments were
quantified and are shown in Figure 6E. In control cells, 64% and 56% of Rab7 and Vps26,
respectively, were found in the membrane fraction, whereas, in Calnuc-depleted cells, the
membrane fractions of Rab7 and Vps26 were reduced to 45% and 43%, respectively. These
results suggest a specific role for Calnuc in the recruitment of Rab7 and consequently of
retromers to endosomal membranes.
The
recruitment
of
retromers
depends
on
proper
Rab5
and
Rab7
activation/inactivation in both time and space, which are governed by their cycling between
GTP- and GDP-bound forms (Rojas, et al., 2008). Because an interaction was detected
between Calnuc and Rab7 (Figure 5B, C) and Calnuc depletion induced the cytoplasmic
redistribution of Rab7 (Figure 6), we next investigated whether Calnuc influences Rab7
activation. To test this hypothesis, we used an effector pull-down assay in which the Rab7binding domain of its effector protein RILP (Rab-interacting lysosomal protein) was used
to selectively isolate GTP-loaded Rab7 (Romero Rosales, et al., 2009). Recombinant GSTRILP immobilized on glutathione beads was incubated with lysates from control and
Calnuc-depleted HeLa cells, and bound (active) Rab7 proteins were analyzed by
immunoblotting (Figure 7A). The data from three separate experiments were quantified and
indicated that GTP-loaded Rab7 was reduced by 66% in Calnuc-depleted cells compared to
control cells (Figure 7B). Taken together, these results suggest that Calnuc regulates the
96
recruitment and activation of Rab7, which is directly involved in the endosomal recruitment
of retromers.
Figure 7 - Knocking down Calnuc affects the activation of Rab7.
(A) Lysates from HeLa cells treated with control or Calnuc-specific siRNA were
incubated with GST or GST-RILP, which specifically interacts with the active (GTPbound) form of Rab7. Bound proteins were then analyzed by western blotting with antiRab7. The lower panel is Ponceau stained blot showing GST-tagged proteins. (B) Bar
graph showing the quantification of Rab7 associated with GST-RILP in cells treated as
described in (A) and expressed as a percentage of the Rab7 present in input lysates. The
results are shown as the means ± SD (n = 3). ** p < 0.001 (compared with control cells).
Discussion
In this study, we demonstrate that Calnuc plays a role in the endosomal sorting of the
lysosomal receptors CI-MPR and Sortilin through the modulation of Rab7 activity and the
recruitment of retromers to endosomes.
Our results show that a small proportion of Calnuc indirectly interacts with the
cytoplasmic tails of CI-MPR and Sortilin and partially colocalizes with these receptors on
endosomes. Furthermore, Calnuc depletion (by siRNA) causes the misdelivery and
degradation of these receptors in lysosomes, leading to a defect in the maturation and
secretion of the lysosomal enzyme cathepsin D. These data suggest a role for Calnuc in the
endosomal sorting of lysosomal receptors for proper retrieval to the TGN. Using siRNA
rescue experiments, we identified the cytoplasmic pool of Calnuc as the functional pool
97
involved in the endosomal sorting of lysosomal receptors. These results are reminiscent of
our previous work demonstrating that cytoplasmic Calnuc plays a role in the endosomal
retrieval of LRP9, an LDLR subtype that also cycles between the TGN and endosomes
(Brodeur, et al., 2009), and suggest that Calnuc is involved in the general mechanism of
this endosomal-sorting pathway. In accordance, our results suggest the implication of
Calnuc in the recruitment of retromers to endosomes, which is key for the retrograde
trafficking of various receptors, including lysosomal sorting receptors (Arighi, et al., 2004;
Rojas, et al., 2007; Rojas, et al., 2008). Indeed, as shown by immunofluorescence and
membrane fractionation assays, Calnuc depletion resulted in a partial cytosolic
redistribution of Vps26 and SNX2. The inability to detect an interaction between Calnuc
and retromer subunits negates the possibility that Calnuc is an adaptor that recruits
retromers to endosomal membrane. We thus investigated Calnuc’s role on the machinery
that recruits retromers onto endosomes, i.e., the small G-proteins Rab5 and Rab7. The
activation and cooperation of Rab5 and Rab7 are required for both the recruitment of the
SNX and Vps components of retromers to endosomes and for lysosomal receptor retrieval
(Rojas, et al., 2008; Seaman, et al., 2009). Our results indicate that Calnuc binds to Rab7
but not to Rab5 or Rab9. Although the interaction between Calnuc and Rab7 is not direct,
we observed that Calnuc plays a role in Rab7 endosomal recruitment and activity. The loss
of Calnuc led to a reduction in the amount of Rab7-GTP that was in concordance with a
decrease in the amount of Rab7 found on endosomal membranes. It has been reported that
the depletion of Rab7 or the expression of a constitutively inactive form of Rab7 decreases
the association of Vps26 and Vps35 on endosomes, but the association of SNX1 and SNX2
is either unaffected or less dramatically altered than that of the Vps proteins (Rojas, et al.,
2008; Seaman, et al., 2009; Vardarajan, et al., 2012). Calnuc depletion induced a partial
cytosolic redistribution of the retromer Vps26 and SNX2 subcomplexes, supporting its
action on Rab7 and the possibility that Calnuc could affect another effector acting on SNX
subcomplex recruitment or stability. The endosomal PI3P levels regulate the membrane
recruitment of the SNX subcomplex (Carlton, et al., 2004; Cozier, et al., 2002; Rojas, et al.,
2008). Interestingly, calcium/calmodulin have been suggested to impact Vps34/PI3K on
early endosomes (Vergne, et al., 2003). Therefore, we examined whether the loss of Calnuc
alters the endosomal PI3P levels. Using a FYVE-GFP probe that specifically recognizes
98
PI3P, we did not observe any clear difference between FYVE-GFP labeling in control and
Calnuc-depleted cells (data not shown). Furthermore, no difference was observed in the
immunofluorescence staining intensity of the early endosomal marker EEA1 (which
contains a FYVE domain) between control and Calnuc-knockdown cells (Figure 4A, B and
Figure 6A, B). These results suggest that the activity of PI3K is not altered in Calnucdepleted cells, and thus, that Calnuc does not alter SNX2 membrane recruitment by
modulating the level of endosomal PI3P.
An attractive hypothesis is that the fraction of Calnuc protein that binds to lysosomal
receptors at the endosome targets Rab7 activity and retromer recruitment to sites where the
receptors are localized. Using various truncation mutants of CI-MPR cytoplasmic tail, we
were unable to determine the domain/motif of interaction given that multiple domains
interacted with Calnuc (data not shown), preventing the generation of a CI-MPR mutant
that cannot bind Calnuc. This could be due to the fact that Calnuc does not bind directly to
the cytoplasmic tail of CI-MPR. Indeed, further work is required to elucidate how the
association between Calnuc and lysosomal receptors is mediated. Therefore, it is unclear
from our results, whether Calnuc actively modulates and recruits the retromer sorting
machinery to lysosomal receptors on endosomes.
In addition to its role in retromer-dependent endosome-to-TGN retrograde transport,
Rab7 has traditionally been involved in transport to late endosomes and lysosomes
(Mukhopadhyay et al. 1997; Press et al. 1998; Vonderheit and Helenius 2005; Ceresa and
Bahr 2006; Vanlandingham and Ceresa 2009; Zhang et al. 2009). In accordance with Rab7
involvement in protein transport pathways that lead both toward and away from lysosomes,
the interference with Rab7 function can lead to the accumulation of CI-MPR in endosomes
(Rojas, et al., 2008). In contrast, retromer depletion results in targeting of the CI-MPR to
lysosomes (Arighi, et al., 2004; Carlton, et al., 2004; Rojas, et al., 2007; Seaman, 2004),
which is caused by a lack of lysosomal receptor removal from maturing endosomes, thus
leading to their passive missorting to lysosomes. It is noteworthy that delivery to lysosomes
is the fate of the CI-MPR and Sortilin in the absence of Calnuc, although we showed that
Rab7 activity was altered. This observation could be explained by the strictly limited action
99
of Calnuc on Rab7 to sites where the retromer is present, thereby allowing Rab7 to regulate
fusion of endosomes with lysosomes at other sites within the endocytic pathway or by the
exposition of sorting determinants within these lysosomal receptors that would direct its
targeting to lysosomes.
Whereas there is considerable divergence of sorting nexin proteins between species,
the cargo-selective components (CSC) Vps26/Vps29/Vps35 heterotrimer is strikingly
conserved throughout eukaryotic evolution (Koumandou, et al., 2011) and is considered to
constitute the core functional component of retromer. Indeed, these components were first
identified in the budding yeast Saccharomyces cerevisiae and name “class A” vacuolar
protein sorting (Vps) proteins and shown as essential to select cargo for endosome-to-Golgi
retrieval of the CPY-sorting receptor, Vps10p. The “class B” Vps5p and Vps17p, the yeast
SNX1 and SNX2, were identified at the same time and shown to assemble onto the
membrane to promote vesicle/tubule formation (Seaman, et al., 1998). Conservation of the
yeast retromer complex components in higher eukaryotes suggests an important general
role for this complex in endosome protein sorting. However, since their first identification
in yeast, a variety of systems have identified multiple cargo proteins that require retromer
for their localization, and accessory proteins that function with CSC retromer in endosomal
protein sorting. For example, the small GTPase Rab7 associates directly with the CSC
complex to mediate its localization to endosomes in yeast and mammals (Balderhaar, et al.,
2010; Liu, et al., 2012; Priya, et al., 2015; Rojas, et al., 2008; Seaman, et al., 2009).
TBC1D5 regulates the membrane association of retromer through a direct interaction with
Vps29 (Harbour, et al., 2010) and via its GAP activity on Rab7 (Seaman, et al., 2009).
Recent studies also demonstrated the role of SNX3, a member of SNX family of proteins
that do not contain a BAR domain, in retromer recruitment via direct interactions with the
CSC subcomplex (Harrison, et al., 2014; Harterink, et al., 2011). However, SNX3 is part of
an alternative retromer pathway that mediates endosome sorting of specific cargo proteins
such as Wntless and transferrin receptor (Chen, et al., 2013 ; Harterink, et al., 2011 ; Zhang,
et al. 2011). Furthermore, unlike Rab7, SNX3 is not conserved across all eukaryotes being
absent in plants suggesting that it is dispensable in some eukaryotes (Vardarajan, et al.,
2012). Eps15-homology domain 1 (EHD1), as well as EHD-1 interacting protein
100
Rabankyrin-5, have also been shown to associate with the CSC, and have a regulatory role
in retromer-mediated endosome-to-Golgi retrieval (Gokool, et al., 2007; Zhang, et al.,
2012). In this study, we identify Calnuc as a novel accessory protein that regulates retromer
recruitment to endosomes, possibly by impacting Rab7 activity. It is noteworthy that yeast
does not seem to have a Calnuc homolog. Therefore, while Calnuc seems to be involved in
the recruitment of retromers in mammalian cells, it is dispensable in simpler organisms
(e.g. yeast). It suggests that Calnuc serves as a regulatory module in the endosome sorting
machinery in higher organisms, rather than an evolutionarily conserved core component.
Rab GTPases such as Rab7 cycle between GTP-bound, membrane-associated, and
GDP-bound cytosolic states and, in doing so, function to regulate the recruitment of
effector proteins, such as retromers, to membranes. Our results show a significant decrease
in the active GTP-bound form of Rab7 in Calnuc-depleted cells. However, the manner by
which Calnuc alters the amount of active Rab7 requires further clarification. Given that
Calnuc does not directly interact with Rab7, these results could be explained by the action
of Calnuc on a GDP Exchange Factor (GEF) of Rab7. An alteration of the activity of Rab7GEF would impair the exchange of GDP for GTP, leading to an increase in the amount of
inactive Rab7 and its dissociation from membranes. The only GEF identified for Rab7 is
Mon1-Ccz1 (Gerondopoulos, et al., 2012; Nordmann, et al., 2010), and it is possible that
Calnuc regulates the recruitment of this GEF to localize and activate Rab7. Alternatively,
Calnuc could regulate the activity and recruitment of TBC1D5, a Rab7-GAP that
negatively regulates retromer recruitment and causes Rab7 to dissociate from membranes
(Seaman, et al., 2009). A recent paper has also linked the activity of ceroid-lipofuscinosis
neuronal 5 (CLN5) to the regulation of Rab7 endosomal recruitment (Mamo, et al., 2012).
CLN5 is mutated in a rare type of lysosomal storage disease named neuronal ceroid
lipofuscinosis (NCL), and the loss of CLN5 expression results in reduced levels of active
Rab7 and compromised retromer recruitment, leading to degradation of the lysosomal
receptors Sortilin and CI-MPR (Mamo, et al., 2012). Because CLN5 depletion phenotypes
are reminiscent of that observed upon Calnuc depletion, it will be of interest to test whether
Calnuc can interact with and regulate the activity of CLN5. If true, these results would
imply a potential role of Calnuc in ceroid lipofuscinosis pathogenesis.
101
Recent studies have shown that the receptor Sortilin regulates the trafficking and
processing of amyloid-β (Aβ), a key neurotoxic peptide in Alzheimer’s disease (AD).
Sortilin binds to amyloid precursor protein (APP) and thereby promotes the production of a
nontoxic soluble product instead of Aβ (Gustafsen, et al., 2013). Moreover, Sortilindeficient mice have increased brain Aβ levels and plaque burden compared with wild-type
mice (Carlo, 2013). LRP10 (i.e., LRP9 in mice), another APP receptor that regulates APP
trafficking and cleavage into Aβ (Brodeur, et al., 2012), has previously been shown to
interact with Calnuc (Brodeur, et al., 2009). As reported for Sortilin and CI-MPR in the
present study, the loss of Calnuc leads to the missorting to lysosomes and degradation of
LRP10 (Brodeur, et al., 2009). Moreover, the expression of the LRP10 protein is
significantly reduced in the brains of AD patients (Brodeur, et al., 2012). Thus, Calnuc
expression affects the trafficking and levels of two APP receptors that protect against the
amyloidogenic processing of APP and accumulation of Aβ. Interestingly, Calnuc levels are
also significantly reduced in the brains of AD patients, and the modulation of Calnuc
expression in neuronal cell lines modulates endogenous APP levels (Lin et al. 2007). Our
results suggest that the action of Calnuc on Rab7 and retromer recruitment could be
involved in the molecular mechanisms by which Calnuc affects APP-sorting receptors and
amyloidogenic cleavage. In agreement with this, the loss of retromer function is known to
increase the amyloidogenic processing of APP, and studies of tissues from AD patients
indicate that retromer expression could be downregulated in neuronal tissue (Small, et al.
2005; Muhammad, et al. 2008; Lane, et al. 2010; Wen, et al. 2011). By altering retromer
recruitment, Calnuc is likely to play an important role in AD pathogenesis.
In conclusion, the results presented here identify Calnuc as a novel player in the
Rab7/retromer-mediated endosomal sorting machinery that determines whether lysosomal
receptors are recycled back to the Golgi or degraded in lysosomes. Consequently, an
alteration in Calnuc expression or activity may affect the proper sorting of CI-MPR and
Sortilin receptors and their cargo and may be potentially involved in the pathological
dysfunction that leads to lysosomal storage or AD.
102
Acknowledgments
We are grateful to Matthew Seaman (University of Cambridge) for the CD8-MPR
and CD8-Sortilin cDNA, the truncation mutants of CI-MPR cytoplasmic tail as well as the
HeLa cells stably expressing GFP-Rab7, to Claus M. Petersen (University of Aarhus) for
untagged Sortilin cDNA and to Francis S. Lee (Weill Medical College of Cornell
University) for Myc-Sortilin cDNA, to Jean-Luc Parent (Université de Sherbrooke) for the
HA-Rab9 cDNA, to Aimee Edinger (UC Irvine) for pGEX-RILP construct. We also thank
Juan S. Bonifacino (NIH) for Myc-hVps35 cDNA and anti-Vps35 pAbs and Thomas
Braulke (University of Hamburg) for the anti-MPR pAbs. We wish to thank Marilène
Paquette for her technical assistance. H.L. holds a fellowship from the National Sciences
and Engineering Research Council of Canada (NSERC). This work was supported by
grants from the Canadian Institutes for Health Research and a Canada Research Chair to
C.L.L. The authors declare that they have no conflict of interest.
103
References
Arighi CN, Hartnell LM, Aguilar RC, Haft CR, Bonifacino JS. 2004. Role of the
mammalian retromer in sorting of the cation-independent mannose 6-phosphate
receptor. J Cell Biol 165(1):123-33.
Balderhaar HJ, Arlt H, Ostrowicz C, Brocker C, Sundermann F, Brandt R, Babst M,
Ungermann C. 2010. The Rab GTPase Ypt7 is linked to retromer-mediated receptor
recycling and fusion at the yeast late endosome. J Cell Sci 123(Pt 23):4085-94.
Ballif BA, Mincek NV, Barratt JT, Wilson ML, Simmons DL. 1996. Interaction of
cyclooxygenases with an apoptosis- and autoimmunity-associated protein. Proc Natl
Acad Sci U S A 93(11):5544-9.
Bonifacino JS, Lippincott-Schwartz J. 2003. Coat proteins: shaping membrane transport.
Nat Rev Mol Cell Biol 4(5):409-14.
Bonifacino JS, Rojas R. 2006. Retrograde transport from endosomes to the trans-Golgi
network. Nat Rev Mol Cell Biol 7(8):568-79.
Boucher R, Larkin H, Brodeur J, Gagnon H, Theriault C, Lavoie C. 2008. Intracellular
trafficking of LRP9 is dependent on two acidic cluster/dileucine motifs. Histochem
Cell Biol 130(2):315-27.
Braulke T, Bonifacino JS. 2009. Sorting of lysosomal proteins. Biochim Biophys Acta
1793(4):605-14.
Brodeur J, Larkin H, Boucher R, Theriault C, St-Louis SC, Gagnon H, Lavoie C. 2009.
Calnuc binds to LRP9 and affects its endosomal sorting. Traffic 10(8):1098-114.
Brodeur J, Theriault C, Lessard-Beaudoin M, Marcil A, Dahan S, Lavoie C. 2012. LDLRrelated protein 10 (LRP10) regulates amyloid precursor protein (APP) trafficking and
processing: evidence for a role in Alzheimer's disease. Mol Neurodegener 7:31.
Bucci C, Thomsen P, Nicoziani P, McCarthy J, van Deurs B. 2000. Rab7: a key to
lysosome biogenesis. Mol Biol Cell 11(2):467-80.
Carlo AS. 2013. Sortilin, a novel APOE receptor implicated in Alzheimer disease. Prion
7(5):378-82.
Carlton J, Bujny M, Peter BJ, Oorschot VM, Rutherford A, Mellor H, Klumperman J,
McMahon HT, Cullen PJ. 2004. Sorting nexin-1 mediates tubular endosome-to-TGN
transport through coincidence sensing of high- curvature membranes and 3phosphoinositides. Curr Biol 14(20):1791-800.
Chen C, Garcia-Santos D, Ishikawa Y, Seguin A, Li L, Fegan KH, Hildick-Smith GJ, Shah
DI, Cooney JD, Chen W and others. 2013. Snx3 regulates recycling of the transferrin
receptor and iron assimilation. Cell Metab 17(3):343-52.
Christoforidis S, Miaczynska M, Ashman K, Wilm M, Zhao L, Yip SC, Waterfield MD,
Backer JM, Zerial M. 1999. Phosphatidylinositol-3-OH kinases are Rab5 effectors.
Nat Cell Biol 1(4):249-52.
Cozier GE, Carlton J, McGregor AH, Gleeson PA, Teasdale RD, Mellor H, Cullen PJ.
2002. The phox homology (PX) domain-dependent, 3-phosphoinositide-mediated
association of sorting nexin-1 with an early sorting endosomal compartment is
required for its ability to regulate epidermal growth factor receptor degradation. J
Biol Chem 277(50):48730-6.
Feng Y, Press B, Wandinger-Ness A. 1995. Rab 7: an important regulator of late endocytic
membrane traffic. J Cell Biol 131(6 Pt 1):1435-52.
104
Gerondopoulos A, Langemeyer L, Liang JR, Linford A, Barr FA. 2012. BLOC-3 mutated
in Hermansky-Pudlak syndrome is a Rab32/38 guanine nucleotide exchange factor.
Curr Biol 22(22):2135-9.
Gokool S, Tattersall D, Seaman MN. 2007. EHD1 interacts with retromer to stabilize
SNX1 tubules and facilitate endosome-to-Golgi retrieval. Traffic 8(12):1873-86.
Gustafsen C, Glerup S, Pallesen LT, Olsen D, Andersen OM, Nykjaer A, Madsen P,
Petersen CM. 2013. Sortilin and SorLA display distinct roles in processing and
trafficking of amyloid precursor protein. J Neurosci 33(1):64-71.
Harbour ME, Breusegem SY, Antrobus R, Freeman C, Reid E, Seaman MN. 2010. The
cargo-selective retromer complex is a recruiting hub for protein complexes that
regulate endosomal tubule dynamics. J Cell Sci 123(Pt 21):3703-17.
Harrison MS, Hung CS, Liu TT, Christiano R, Walther TC, Burd CG. 2014. A mechanism
for retromer endosomal coat complex assembly with cargo. Proc Natl Acad Sci U S
A 111(1):267-72.
Harterink M, Port F, Lorenowicz MJ, McGough IJ, Silhankova M, Betist MC, van Weering
JR, van Heesbeen RG, Middelkoop TC, Basler K and others. 2011. A SNX3dependent retromer pathway mediates retrograde transport of the Wnt sorting
receptor Wntless and is required for Wnt secretion. Nat Cell Biol 13(8):914-23.
Koumandou VL, Klute MJ, Herman EK, Nunez-Miguel R, Dacks JB, Field MC. 2011.
Evolutionary reconstruction of the retromer complex and its function in Trypanosoma
brucei. J Cell Sci 124(Pt 9):1496-509.
Lane RF, Raines SM, Steele JW, Ehrlich ME, Lah JA, Small SA, Tanzi RE, Attie AD,
Gandy S. 2010. Diabetes-associated SorCS1 regulates Alzheimer's amyloid-beta
metabolism: evidence for involvement of SorL1 and the retromer complex. J
Neurosci 30(39):13110-5.
Lavoie C, Meerloo T, Lin P, Farquhar MG. 2002. Calnuc, an EF-hand Ca(2+)-binding
protein, is stored and processed in the Golgi and secreted by the constitutive-like
pathway in AtT20 cells. Mol Endocrinol 16(11):2462-74.
Lin P, Fischer T, Lavoie C, Huang H, Farquhar MG. 2009. Calnuc plays a role in dynamic
distribution of Galphai but not Gbeta subunits and modulates ACTH secretion in
AtT-20 neuroendocrine secretory cells. Mol Neurodegener 4:15.
Lin P, Fischer T, Weiss T, Farquhar MG. 2000. Calnuc, an EF-hand Ca(2+) binding
protein, specifically interacts with the C-terminal alpha5-helix of G(alpha)i3. Proc
Natl Acad Sci U S A 97(2):674-9.
Lin P, Le-Niculescu H, Hofmeister R, McCaffery JM, Jin M, Hennemann H, McQuistan T,
De Vries L, Farquhar MG. 1998. The mammalian calcium-binding protein,
nucleobindin (CALNUC), is a Golgi resident protein. J Cell Biol 141(7):1515-27.
Lin P, Yao Y, Hofmeister R, Tsien RY, Farquhar MG. 1999. Overexpression of CALNUC
(nucleobindin) increases agonist and thapsigargin releasable Ca2+ storage in the
Golgi. J Cell Biol 145(2):279-89.
Liu TT, Gomez TS, Sackey BK, Billadeau DD, Burd CG. 2012. Rab GTPase regulation of
retromer-mediated cargo export during endosome maturation. Mol Biol Cell
23(13):2505-15.
Lombardi D, Soldati T, Riederer MA, Goda Y, Zerial M, Pfeffer SR. 1993. Rab9 functions
in transport between late endosomes and the trans Golgi network. Embo J 12(2):67782.
105
Mamo A, Jules F, Dumaresq-Doiron K, Costantino S, Lefrancois S. 2012. The role of
ceroid lipofuscinosis neuronal protein 5 (CLN5) in endosomal sorting. Mol Cell Biol
32(10):1855-66.
McGough IJ, Cullen PJ. 2011. Recent advances in retromer biology. Traffic 12(8):963-71.
Miura K, Titani K, Kurosawa Y, Kanai Y. 1992. Molecular cloning of nucleobindin, a
novel DNA-binding protein that contains both a signal peptide and a leucine zipper
structure. Biochem Biophys Res Commun 187(1):375-80.
Muhammad A, Flores I, Zhang H, Yu R, Staniszewski A, Planel E, Herman M, Ho L,
Kreber R, Honig LS and others. 2008. Retromer deficiency observed in Alzheimer's
disease causes hippocampal dysfunction, neurodegeneration, and Abeta
accumulation. Proc Natl Acad Sci U S A 105(20):7327-32.
Ng EL, Gan BQ, Ng F, Tang BL. 2012. Rab GTPases regulating receptor trafficking at the
late endosome-lysosome membranes. Cell Biochem Funct 30(6):515-23.
Nordmann M, Cabrera M, Perz A, Brocker C, Ostrowicz C, Engelbrecht-Vandre S,
Ungermann C. 2010. The Mon1-Ccz1 complex is the GEF of the late endosomal
Rab7 homolog Ypt7. Curr Biol 20(18):1654-9.
Priya A, Kalaidzidis IV, Kalaidzidis Y, Lambright D, Datta S. 2015. Molecular Insights
into Rab7-Mediated Endosomal Recruitment of Core Retromer: Deciphering the Role
of Vps26 and Vps35. Traffic.
Rojas R, Kametaka S, Haft CR, Bonifacino JS. 2007. Interchangeable but essential
functions of SNX1 and SNX2 in the association of retromer with endosomes and the
trafficking of mannose 6-phosphate receptors. Mol Cell Biol 27(3):1112-24.
Rojas R, van Vlijmen T, Mardones GA, Prabhu Y, Rojas AL, Mohammed S, Heck AJ,
Raposo G, van der Sluijs P, Bonifacino JS. 2008. Regulation of retromer recruitment
to endosomes by sequential action of Rab5 and Rab7. J Cell Biol 183(3):513-26.
Romero Rosales K, Peralta ER, Guenther GG, Wong SY, Edinger AL. 2009. Rab7
activation by growth factor withdrawal contributes to the induction of apoptosis. Mol
Biol Cell 20(12):2831-40.
Seaman MN. 2004. Cargo-selective endosomal sorting for retrieval to the Golgi requires
retromer. J Cell Biol 165(1):111-22.
Seaman MN, Harbour ME, Tattersall D, Read E, Bright N. 2009. Membrane recruitment of
the cargo-selective retromer subcomplex is catalysed by the small GTPase Rab7 and
inhibited by the Rab-GAP TBC1D5. J Cell Sci 122(Pt 14):2371-82.
Seaman MN, McCaffery JM, Emr SD. 1998. A membrane coat complex essential for
endosome-to-Golgi retrograde transport in yeast. J Cell Biol 142(3):665-81.
Small SA, Kent K, Pierce A, Leung C, Kang MS, Okada H, Honig L, Vonsattel JP, Kim
TW. 2005. Model-guided microarray implicates the retromer complex in Alzheimer's
disease. Ann Neurol 58(6):909-19.
Stenmark H, Parton RG, Steele-Mortimer O, Lutcke A, Gruenberg J, Zerial M. 1994.
Inhibition of rab5 GTPase activity stimulates membrane fusion in endocytosis. Embo
J 13(6):1287-96.
Vardarajan BN, Bruesegem SY, Harbour ME, Inzelberg R, Friedland R, St George-Hyslop
P, Seaman MN, Farrer LA. 2012. Identification of Alzheimer disease-associated
variants in genes that regulate retromer function. Neurobiol Aging 33(9):2231 e152231 e30.
106
Vergne I, Chua J, Deretic V. 2003. Tuberculosis toxin blocking phagosome maturation
inhibits a novel Ca2+/calmodulin-PI3K hVPS34 cascade. J Exp Med 198(4):653-9.
Wang T, Ming Z, Xiaochun W, Hong W. 2011. Rab7: role of its protein interaction
cascades in endo-lysosomal traffic. Cell Signal 23(3):516-21.
Weiss TS, Chamberlain CE, Takeda T, Lin P, Hahn KM, Farquhar MG. 2001. Galpha i3
binding to calnuc on Golgi membranes in living cells monitored by fluorescence
resonance energy transfer of green fluorescent protein fusion proteins. Proc Natl
Acad Sci U S A 98(26):14961-6.
Wen L, Tang FL, Hong Y, Luo SW, Wang CL, He W, Shen C, Jung JU, Xiong F, Lee DH
and others. 2011. VPS35 haploinsufficiency increases Alzheimer's disease
neuropathology. J Cell Biol 195(5):765-79.
Zhang J, Reiling C, Reinecke JB, Prislan I, Marky LA, Sorgen PL, Naslavsky N, Caplan S.
2012. Rabankyrin-5 interacts with EHD1 and Vps26 to regulate endocytic trafficking
and retromer function. Traffic 13(5):745-57.
Zhang P, Wu Y, Belenkaya TY, Lin X. 2011. SNX3 controls Wingless/Wnt secretion
through regulating retromer-dependent recycling of Wntless. Cell Res 21(12):167790.
107
Supporting Information
Supp. Figure S1 - Depletion of Calnuc causes missorting of the lysosomal enzyme
Cathepsin D.
Lysates from cells treated with control or Calnuc siRNA were analyzed by western
blotting with specific anti-Cathepsin D (CatD), anti-CI-MPR, anti-Calnuc and anti-Actin
antibodies. Precursor (p), intermediate (i) and mature (m) forms of Cathepsin D are
shown.
108
Supp. Figure S2 - Confocal Z-stack images of cells presented in Figure 4A.
Cells were prepared as described in Figure 4A. Series of confocal z-stack images were
acquired at 0.12 μm (A) or 0.2 μm (B) intervals. The last panels display merged z stacks
and signal intensities. The color bar ranges from low intensity (black) to high intensity
(white). Scale bar, 10 μm.
109
Supp. Figure S3 - The interaction of Calnuc and Rab5 is independent of the
activation status of Rab5.
Lysates from HEK cells transfected with Calnuc-Flag together with HA, wild-type HARab5, dominant-active HA-Rab5Q79L or dominant-inactive HA-Rab5S34N were
immunoprecipitated with an anti-Flag antibody and then immunoblotted with anti-HA or
anti-Flag to detect Rab5 proteins or Calnuc, respectively.
Supp. Figure S4 - Confocal Z-stack images of cells presented in Figure 6A.
Cells were prepared as described in Figure 6A. Series of confocal z-stack images were
acquired at 0.32 μm intervals. The last panels display merged z stacks and signal
intensities. The color bar ranges from low intensity (black) to high intensity (white).
Scale bar, 10 μm.
110
ARTICLE 2
Topology and membrane anchoring of the lysosomal storage disease-related protein
CLN5
Auteurs de l’article: Heidi Larkin, Maria Gil Ribeiro et Christine Lavoie
Statut de l’article: Publié
Human mutation, vol. 34, no 12 (déc. 2013), p. 1688-1697
Avant-propos: J'ai écrit la première version des sections 'Matériel et méthodes' ainsi que
des descriptions des figures. J'ai réalisé toutes les expériences retrouvées dans l'article ainsi
que tous les montages. Christine Lavoie a effectué les corrections du texte et a rédigé
l'introduction, les résultats et la discussion. Elle a également effectué l'expérience présentée
en figure 2C et les schémas 4A-C. Maria Gil Ribeiro a fourni l'anticorps CLN5-C/32.
111
RÉSUMÉ – ARTICLE 2
Topologie et ancrage membranaire de CLN5, une protéine associée à une maladie du
lysosome
Certaines variantes infantiles tardives de céroïdes-lipofuscinoses neuronales, une
maladie neurodégénérative, sont causées par des mutations dans le gène CLN5. CLN5 code
une glycoprotéine lysosomiale dont la structure et la fonction n’ont pas encore été
clairement définies. Dans la présente étude, nous utilisons diverses constructions de CLN5
comprenant des étiquettes afin de déterminer la topologie et la solubilité de la protéine.
Notre étude indique que CLN5 est synthétisée sous forme d’une glycoprotéine
transmembranaire de type II, comprenant un domaine N-terminal cytoplasmique, un
segment transmembranaire et un grand domaine C-terminal luminal contenant une hélice
amphipathique. Les domaines cytoplasmique et transmembranaire sont rapidement éliminés
suivant le clivage du peptide signal de manière à former la protéine CLN5 mature. Cette
dernière demeure fortement associée du côté luminal de la membrane par une hélice
amphipathique. Les mutants pathologiques de CLN5 dépourvus de cette hélice
amphipathique perdent leur association membranaire, sont retenus au réticulum
endoplasmique et sont rapidement dégradés par la machinerie du protéasome. Nous avons
défini, de façon expérimentale, la topologie de CLN5 et nous avons démontré l’existence
d’une hélice amphipathique qui permet l’ancrage de la protéine à la membrane. Notre
travail met en lumière les propriétés de base de CLN5 permettant une meilleure
compréhension de ses fonctions biologiques et de son implication dans la pathogenèse des
céroïdes-lipofuscinoses neuronales.
112
Article 2
Topology and Membrane Anchoring of the Lysosomal Storage Disease-Related
Protein CLN5
Heidi Larkin,1 Maria Gil Ribeiro,2,3 and Christine Lavoie1∗
1
Department of Pharmacology, Faculty of Medicine and Health Sciences, Universite de
Sherbrooke, Sherbrooke, Quebec, Canada
2
Research and Development Unit, Department of Genetics, National Health Institute Dr.
Ricardo Jorge, Porto, Portugal
3
Faculty of Health Sciences, Fernando Pessoa University, Porto, Portugal
Communicated by Elizabeth F. Neufeld
∗Correspondence
to: Christine Lavoie,
Department of Pharmacology, Faculty of Medicine, Universite de Sherbrooke,
3001-12e Avenue Nord, Sherbrooke, QC J1H 5N4, Canada
E-mail: [email protected]
Received 28 May 2013
Accepted revised manuscript 6 September 2013.
Published online 13 September 2013 in Wiley Online Library
(www.wiley.com/humanmutation). DOI: 10.1002/humu.22443
Keywords: neuronal ceroid lipofuscinoses (NCL);CLN5;topology;amphipathic
helix;transmembrane domain
113
Early view
Elucidation of the topology of human CLN5, a lysosomal protein involved in neuronal
ceroid lipofuscinosis pathogenesis.
CLN5 is synthesized as a single pass type II transmembrane precursor glycoprotein. The
signal peptide (red) is rapidly cleaved, and the resulting mature protein (blue) is tightly
associated with the lumen of the membrane through an amphipathic anchor region (AH).
Pathological mutants deprived of AH (pGlu253* and pGlu352*) lose their membrane
association and are rapidly degraded.
Abstract
One late infantile variant of the neurodegenerative disease neuronal ceroid
lipofuscinosis (NCL) is caused by a mutation in the CLN5 gene. CLN5 encodes a
lysosomal glycoprotein whose structure and function have not yet been clearly defined. In
the present study, we used epitope-tagged CLN5 to determine the topology and solubility
of the CLN5 protein. Our results indicated that CLN5 is synthesized as a type II
transmembrane (TM) glycoprotein with a cytoplasmic N-terminus, one TM segment, and a
large luminal C-terminal domain containing an amphipathic helix (AH). The cytoplasmic
and TM domains were rapidly removed following signal-peptide cleavage, and the
resulting mature CLN5 was tightly associated with the lumen of the membrane through the
AH. CLN5 pathological mutants deprived of AH lose their membrane association, are
114
retained in the endoplasmic reticulum, and are rapidly degraded by the proteasomal
machinery. We experimentally define the topology of CLN5 and demonstrate the existence
of an AH that anchors the protein to the membrane. Our work sheds light on the basic
properties of CLN5 required to better understand its biological functions and involvement
in NCL pathogenesis.
Introduction
Neuronal ceroid lipofuscinoses (NCLs) are severe neurodegenerative lysosomal
storage diseases that manifest mostly in childhood and are characterized by developmental
regression, seizures, visual failure, and mental retardation, all of which culminate in
premature death [Mole et al., 2005]. NCLs are relatively rare worldwide (1:100,000 live
births), but are more common (1:12,500 live births) in certain North American and
Northern European populations [Haltia, 2006; Moore et al., 2008; Santavuori, 1988]. These
disorders result from mutations in at least 13 ceroid lipofuscinosis neuronal genes (CLN1–
CLN14) [Kollmann et al., 2013]. Mutations in the human CLN5 gene (MIM #608102)
result in late infantile, juvenile, and adult forms of NCL [Mole et al., 2011]. To date, over
20 disease-causing mutations have been identified in this gene (http://www.ucl.ac.uk/ncl).
CLN5 encodes a 407 amino acid (aa) polypeptide with a predicted molecular mass of
46 kDa [Savukoski et al., 1998]. However, CLN5 expressed in cells has been reported to
have a higher molecular weight ranging from 50 to 75 kDa due to eight potential Nglycosylation sites [Isosomppi et al., 2002; Schmiedt et al., 2010; Vesa et al., 2002]. CLN5
also contains a signal peptide (SP) that is cleaved in the endoplasmic reticulum (ER) and
the mature polypeptide is mainly localized in lysosomes [Holmberg et al., 2004; Isosomppi
et al., 2002; Schmiedt et al., 2010; Vesa et al., 2002]. However, the full topology of CLN5
is not known, and its solubility versus membrane-spanning property has been a
controversial issue. CLN5 was initially described as having two transmembrane (TM)
domains and has been detected in a Triton X-114 (TX-114) TM protein fraction [Bessa et
al., 2006; Savukoski et al., 1998; Vesa et al., 2002]. However, it has also been described as
a soluble protein and has been detected in culture media [Holmberg et al., 2004; Sleat et al.,
115
2006b]. To date, none of the CLN5 disease-causing mutations studied have been shown to
have an effect on CLN5 synthesis and maturation, but they are known to affect the
lysosomal targeting of CLN5 [Isosomppi et al., 2002; Lebrun et al., 2009; Schmiedt et al.,
2010; Vesa et al., 2002].
While CLN5 was identified 15 years ago [Savukoski et al., 1998], its cellular function
remains poorly understood. Previous studies have shown that CLN5 interacts with other
NCL proteins [Lyly et al., 2009; Vesa et al., 2002], but the purpose of these interactions
remains elusive. Moreover, recent studies have shown that CLN5 interacts with the
lysosomal-sorting receptor sortilin and affects its endosome-to-TGN (trans-Golgi Network)
retrieval by regulating retromer membrane recruitment [Mamo et al., 2012]. This provided
the first link between CLN5 and lysosomal function. To better understand the impact of
CLN5 mutations and the role and mechanism of the action of CLN5, some of its basic
properties, including its topology and solubility, need to be elucidated. By introducing
epitope tags on the N- or C-terminus or an internal portion of CLN5, we determined that
CLN5 is synthesized as a single pass type II TM protein that is cleaved after its SP to
produce a mature luminal form of CLN5 that remains tightly associated with the membrane
through an amphipathic anchor region.
Materials and Methods
Antibodies
Anti-Calnexin polyclonal antibody (pAb) was from Millipore (Billerica, MA), antiCalnuc pAb was from Aviva Systems Biology (San Diego, CA), anti-EEA1 pAb was from
Thermo Scientific (Rockford, IL), anti-Flag monoclonal antibody (mAb) was from Sigma–
Aldrich (Saint Louis, MO), anti-GFP mAb was from Clontech (Mountain View, CA), antiGM130 mAb was from BD Transduction Laboratories (Franklin Lakes, NJ), anti-LAMP1
(H4A3) mAb was from Developmental Studies Hybridoma Bank (University of Iowa, IA),
anti-HA mAb and pAb were from Covance (Emeryville, CA), and anti-transferrin receptor
(TfR) mAb was from Invitrogen (Carlsbad, CA). CLN5-C/32 rabbit pAb directed against
116
the C-terminus extremity of human CLN5 has been previously described [Schmiedt et al.,
2010].
DNA Constructs
Human 3xHA-CLN5 in the pReceiver-M06 vector was a generous gift from Dr.
Stéphane Lefrançois (Université de Montréal, QC, Canada). Untagged CLN5, CLN5-HA,
and SP-Flag-CLN5 were generated by subcloning in the pcDNA3.1 vector. The SP-FlagCLN5 construct was obtained by the insertion of the Flag epitope at amino acid position 97.
The pathological mutants EUR (European) (c.835G>A, p.Asp279Asn), the FinM (Finnish
major) (c.1175_1176delAT, p.Tyr392*), the SWE (Swedish) (c.757G>T, p.Glu253*), and
the NFL (Newfoundland) (c.1054G>T, p.Glu352*) were generated from SP-Flag-CLN5 by
QuickChange site-directed mutagenesis as previously described [Schmiedt et al., 2010].
Nucleotide numbering reflects cDNA numbering, with +1 corresponding to the A of the
ATG translation initiation codon in the reference sequence (GenBank NM_006493.2)
according to journal guidelines (www.hgvs.org/mutnomen). The initiation codon is codon
1.
Cell Culture and Transfection
HeLa cells were from ATCC (Manassas, VA) and HEK293T cells were from Dr.
Alexandra Newton (UCSD, CA). The cells were grown in DMEM high glucose medium
(Invitrogen) containing 10% fetal bovine serum (FBS) (Hyclone Laboratories, Logan, UT),
penicillin, and streptomycin. HEK cells were transfected with Lipofectamine 2000
transfection reagent (Invitrogen), and HeLa cells with Fugene6 transfection reagent (Roche
Diagnostic, Indianapolis, IN), both according to the manufacturers’ instructions.
In Vitro Translation Assay
In vitro translation of human CLN5 pcDNA3.1 was performed using the TNT T7
Quick Coupled Transcription/Translation system (Promega, Madison, WI) in the presence
or absence of canine pancreatic microsomes (Promega) according to the manufacturer's
protocol.
117
Pulse-Chase Analysis
HEK293 cells transiently expressing the various tagged CLN5 proteins were
incubated for 30 min in methionine-free DMEM. The cells were pulsed for 5 min with 150
μCi/ml of [35S]methionine/cysteine (>1000 Ci/mmol; EasyTagTM Expression Protein
Labeling Mix, Perkin Elmer, Waltham, MA), washed in PBS, and chased for 0, 0.5, 1, 2, or
4 hr in complete culture medium. Cells and medium were collected after each chase. The
medium was cleared by centrifugation (15,800g for 30 min). Tagged CLN5 proteins were
immunoprecipitated from the cell lysates or medium, separated by SDS-PAGE, and
detected by autoradiography.
Subcellular Fractionation and Extraction Assays
HEK293 cells were washed in cold PBS, delicately scraped into TNES buffer (50
mM tris, pH 7.4, 150 mM NaCl, 5 mM EDTA, 250 mM sucrose, and protease inhibitors),
and centrifuged for 3 min at 500g. The pellet was homogenized in TNES buffer by 30
passages through a 25 G 5/8 needle. Nuclei and unbroken cells were removed by
centrifugation at 1,000g for 10 min. The postnuclear supernatant (PNS) was collected and
was centrifuged at 100,000g for 1 hr at 4°C. The soluble fraction (S100) was collected, and
the pellet or membrane fraction (P100) was resuspended using a pestle in an equal volume
of TNES buffer. Approximately 200 μg of P100 was then incubated with 500 μL of 1 M
NaCl, or 150 mM Na2CO3 (pH 11.5), or 5 M urea for 30 min on ice. The solution was
centrifuged at 100,000g for 1 hr at 4°C. The soluble fraction (SP100) was precipitated with
10% trichloroacetic acid (TCA) overnight at −80°C. The precipitate (PP100) was directly
resuspended in loading buffer. The same volumes of S100 and P100 and the entire SP100
and PP100 fractions were analyzed by SDS-PAGE and immunoblotting.
TX-114 Fractionation
TX-114 fractionation was performed on the P100 membrane fraction using MemPER Eukaryotic Membrane Protein Extraction Reagent kits from Thermo Scientific
(Waltham, MA) using the manufacturer's protocol [Qoronfleh et al., 2003]. The aqueous
phase was precipitated with 10% TCA overnight at −80°C, whereas the detergent phase
was precipitated using 1 ml of ethanol/ether (4:1) overnight at −20°C. The fractions were
118
centrifuged at 15,800g for 20 min and the pellets were resuspended in SDS-PAGE-loading
buffer.
Treatment with Glycosidase and Proteinase K
Deglycosylation was carried out according to the manufacturer's instructions. Briefly,
50 μg of cell lysate were incubated with 1 unit of PNGase F (NEB, Ipswich, MA) overnight
at 37°C. For the topology assays, 50 μg of P100 membrane fraction were incubated with
100 μg/ml of proteinase K (Invitrogen) in the absence or presence of 0.5% TX-100. The
protease reactions were stopped by adding 1 mM phenylmethylsulfonyl fluoride (Sigma–
Aldrich) and incubating for 10 min on ice.
Immunoblotting
The protein samples were separated on SDS-PAGE gels and were transferred to
nitrocellulose membranes. The membranes were blocked in tris-buffered saline containing
0.1% Tween 20 and 5% nonfat dry milk and were incubated with primary antibodies for 1
hr at room temperature (RT) followed by horseradish peroxidase-conjugated goat antirabbit or anti-mouse immunoglobulin G (Bio-Rad, Richmond, ON) for 1 hr at RT and then
with enhanced chemiluminescence (ECL) detection reagent (Pierce Chemical, Rockford,
IL).
Immunofluorescence
HeLa cells were plated on coverslips. Twelve hours after transfection, the cells were
fixed and permeabilized in cold methanol for 10 min. The cells were blocked with 10%
goat or bovine fetal serum (FBS) for 30 min and were incubated with primary antibodies
for 1 hr at RT followed by Alexa Fluor-conjugated secondary antibodies (Molecular
Probes, Eugene, OR). The specimens were visualized using an inverted confocal laserscanning microscope FV1000 (Olympus, Tokyo, Japan) equipped with a PlanApo 60x/1.42
oil immersion objective. Olympus Fluoview software version 1.6a was used to acquire and
analyze the images, which were further processed using Adobe Photoshop (Adobe Systems,
San Jose, CA).
119
Results
Tagging the CLN5 Protein Does not Affect Its Maturation, Secretion, or Localization
To clarify the topology of CLN5, three different epitope-tagged human CLN5
constructs were generated. The first had three HA tags at the N-terminus of the first
initiation methionine of CLN5 (3xHA-CLN5), the second had an HA tag at the C-terminus
(CLN5-HA), and the third had a Flag tag after the putative SP cleavage site (SP-FlagCLN5) (Fig. 1A). To validate the reliability of these tools, we first confirmed that the
epitope-tagged forms of CLN5 were expressed and processed appropriately. To that end,
wild-type (WT; untagged) and epitope-tagged CLN5 were transiently expressed in HeLa
cells, and their molecular weights (MW) were analyzed by Western blotting. Since CLN5
contains an SP cleavage site and eight potential N-glycosylation sites (Fig. 1A), the size of
the protein detected depends on the fragment recognized by the antibody and the level of
glycosylation. In Figure 1B, proteins were detected using either antibodies directed against
the Flag or HA tags or a specific CLN5 peptide antibody (α-C/32) directed against the Cterminal region of CLN5 (aa 393–407) that has already been used to detect the mature form
of overexpressed untagged CLN5 [Schmiedt et al., 2010]. In addition, portions of cell
lysates were treated with PNGase F to remove all N-linked oligosaccharide side chains
from the CLN5 proteins. As previously reported [Schmiedt et al., 2010], untagged CLN5 is
detected as a 60-kDa polypeptide in the absence of PNGase F and a 35-kDa polypeptide in
the presence of PNGase F, which correspond to the glycosylated and unglycosylated
cleaved (mature) forms of CLN5 (96–407 aa fragment), respectively. CLN5-HA and SPFlag-CLN5 detected with antibodies directed against their tags had the same expression
pattern as untagged CLN5 in the absence and presence of PNGase F, suggesting that the
insertion of these tags did not affect the cleavage or glycosylation of CLN5. N-terminustagged 3xHA-CLN5 was detected as three bands of ∼73, 50, and 15 kDa corresponding to
the predicted MW of the glycosylated and unglycosylated full-length (uncleaved or
precursor) forms of CLN5 and the SP fragment (aa 1–96), respectively. This is in
agreement with the results of Vesa et al. (2002), who detected a 75-kDa glycosylated
polypeptide and a 47-kDa unglycosylated polypeptide using antibodies raised against aa 1–
75 of CLN5. Using the α-C/32 antibody, we also detected the mature 60 kDa fragment in
120
Figure 1 - Epitope-tagged CLN5 are appropriately expressed, processed, and secreted.
A: Schematic representation of the different CLN5 cDNA constructs. Putative TM domains
are represented by hatched boxes, the SP cleavage site as a dotted line, and the Nglycosylation sites by stars. HA or Flag epitope tags (represented by black boxes) were
inserted at the N- or C-terminus as well as after the putative SP (aa 96) of CLN5. B:
Analysis of expression patterns of untagged and epitope-tagged CLN5 by Western blotting.
Lysates from HeLa cells transiently transfected with the untagged or tagged CLN5 were
121
incubated in the absence or presence of PNGase F. The proteins were separated by SDSPAGE and were immunoblotted with anti-HA, anti-Flag, or anti-CLN5 C/32 antibody.
Schematic representations of the corresponding proteins are indicated on the left or right
side of the bands. The 3xHA tag added ∼3–4 kDa to the MW of CLN5, whereas the HA
and Flag tags added ∼1 kDa. C: Analysis of the maturation and secretion of untagged and
tagged-CLN5. HEK293 cells transiently expressing CLN5-HA, SP-Flag-CLN5, or
untagged CLN5 were pulsed for 5 min with 35S-methionine and were chased for the
indicated periods of time. Tagged and untagged CLN5 proteins were immunoprecipitated
from the cell lysates, and culture media was collected after each chase period using antiHA, anti-Flag, or anti-C/32 antibodies. The proteins were separated by SDS-PAGE and
analyzed by autoradiography. *, Nonspecific band; black arrowhead, precursor polypeptide;
white arrowhead, mature polypeptide; boxed lane, longer exposure of the 0 min chase of
SP-Flag-CLN5 to show the presence of precursor and mature polypeptides.
the 3xHA-CLN5 cell lysate (data not shown), confirming that the appropriate cleavage and
glycosylation had occurred. These results indicated that the different epitope-tagged CLN5
proteins were correctly expressed, cleaved, and glycosylated.
To confirm that the epitope-tagged forms of CLN5 were appropriately localized, the
intracellular distributions of 3xHA-CLN5, CLN5-HA, SP-Flag-CLN5, and untagged CLN5
expressed in HeLa cells were next analyzed by confocal microscopy (Supp. Fig. S1).
Schmiedt et al. (2010) reported that the precursor form or N-terminal fragment of GFPCLN5 is retained in the ER, whereas the mature form of CLN5 exits the ER and is mainly
targeted to lysosomes. We observed that the intracellular distribution of CLN5-HA and SPFlag-CLN5 was very similar to the untagged CLN5 stained with anti-C/32 antibody and
partially colocalized with the lysosomal marker LAMP1, which is in agreement with the
results of Schmiedt et al. (2010). These three CLN5 proteins were also detected in a
juxtanuclear compartment corresponding to TGN since they partially colocalized with
TGN46 (data not shown). On the other hand, 3xHA-CLN5 colocalized exclusively with the
ER marker Calnexin. These results indicated that the different epitope-tagged CLN5 were
appropriately distributed in the cells.
WT CLN5 has also previously been reported to be secreted [Isosomppi et al., 2002].
To confirm that the maturation and secretion of epitope-tagged CLN5 were not altered, we
next compared the kinetics of synthesis and secretion of SP-Flag-CLN5 and CLN5-HA to
122
that of untagged CLN5 using a pulse-chase assay. HEK293 cells expressing SP-FlagCLN5, CLN5-HA, or untagged CLN5 were labeled for 5 min with [35S]methionine, chased
for up to 2 hr, immunoprecipitated from the cell lysate and the medium, separated by SDSPAGE, and detected by autoradiography (Fig. 1C). Similar kinetics was obtained with
tagged and untagged CLN5 proteins. After a 0-min chase, untagged and tagged CLN5 were
detected as a ∼70-kDa glycosylated precursor and a ∼60-kDa processed/mature form of
CLN5 in the cell lysates (Fig. 1C; intracellular, black and white arrowheads, respectively).
The weaker signal obtained with SP-Flag-CLN5 may be due to less efficient detection of
the internal epitope tag. With longer chases, the 70-kDa precursor of tagged and untagged
CLN5 was rapidly processed, giving rise to the 60-kDa mature form, which was still
detected in the cell lysate after a 2-hr chase (Fig. 1C, intracellular). The fact that the 60-kDa
mature form of CLN5 was observed after a 5-min pulse indicated that the N-glycosylation
and SP cleavage of CLN5 are cotranslational events, as previously suggested by Schmiedt
et al. (2010). Moreover, beginning with a 1-hr chase, small amounts of CLN5-HA, SPFlag-CLN5, and untagged CLN5 were detected in the extracellular medium (Fig. 1C,
extracellular), indicating that mature CLN5 is partially secreted. This was confirmed with a
chase at 20°C, which is known to arrest protein transport in the TGN [Matlin and Simons,
1983], that completely blocked the secretion of tagged and untagged CLN5 (Supp. Fig. S2).
In summary, the presence of tags did not markedly alter the kinetics of maturation and
secretion of CLN5. The small difference observed may be due to slight intracellular
retention or slower degradation of the tagged CLN5 proteins compared with their WT
counterpart or to differences in antibody affinity or native protein epitope recognition.
Overall, these results confirmed that tagging CLN5 does not alter its intracellular
maturation, secretion, or localization since the tagged CLN5 behaved mostly as untagged
CLN5. Therefore, these tagged CLN5 proteins represent suitable and valuable tools for
addressing key issues regarding the basic properties of CLN5.
123
Determination of the Topology of CLN5
A bioinformatic analysis of the CLN5 sequence using online SACS and ExPASy TM
prediction tools identified up to two hydrophobic regions predicted to function as
membrane-spanning regions (Fig. 1A). One TM domain was unanimously predicted at
residues ranging from 73 to 93, whereas a second potential TM domain (aa 353–373) was
only predicted by a few tools. Two models can thus be proposed based on these predictions
and on the presence of an SP cleavage site at position 96 (Fig. 2A). In one model, CLN5
encodes two TM regions with the N- and C-termini in the cytoplasm and an intraluminal
loop (Fig. 2A, Model 1). In the second model, CLN5 contains one TM, a cytoplasmic Nterminus, and an intraluminal C-terminus (Fig. 2A, Model 2). To determine the topological
orientation of the amino- and carboxyl-termini of CLN5, we performed a protease
protection assay using the previously characterized N-terminally, C-terminally, and
internally tagged CLN5 proteins (Fig. 1). Since the protease cannot cross the membrane,
regions or tags in the lumen of intact compartments would be protected from digestion,
whereas the cytoplasmic regions or tags would be digested. Crude membrane compartment
fractions isolated from HeLa cells expressing the different tagged CLN5 were incubated
with proteinase K in the presence or absence of TX-100 and were analyzed by Western
blotting (Fig. 2B). In the absence of TX-100, the N-terminus HA tags were digested by the
protease, resulting in the complete loss of immunoreactivity, whereas the C-terminus HAtag and the Flag-tag inserted after the SP were not digested. However, when TX-100 was
added to the proteinase K digestion mixture, these two tags were also digested (Fig. 2B).
These results indicated that the N-terminus tail of CLN5 is located in the cytoplasm,
whereas its C-terminus tail is located in the lumen, as depicted in Model 2 (Fig. 2A).
The topology of CLN5 was confirmed using a cell-free protein expression system
containing canine pancreatic microsomes. This in vitro system, which functionally and
topologically mimics the ER, accurately reproduces the translation, membrane insertion
and translocation, SP cleavage, and N-linked glycosylation of membrane and secreted
proteins that occurs in vivo. Protease protection assays can thus be used with this system to
determine the topology of membrane proteins. Untagged CLN5 was translated in vitro in
the absence or presence of microsomal membranes and was analyzed by SDS-PAGE and
124
Figure 2 - Topology of CLN5.
A: Schematic representation of two models representing a possible topology of CLN5.
Model 1 depicts two TM regions with cytoplasmic N- and C-termini and an intraluminal
loop. Model 2 depicts one TM region, a cytoplasmic N-terminus, and an intraluminal Cterminus. B: Membrane fractions isolated from HeLa cells transiently expressing the tagged
CLN5 proteins were incubated in the absence or presence of proteinase K, with or without
125
TX-100. The proteins were separated by SDS-PAGE and were immunoblotted with antiHA or anti-Flag antibody. Endogenous luminal calnuc and cytoplasmic EEA1 were used as
controls. C: Untagged CLN5 cDNA subcloned into the pcDNA3.1 vector was translated in
vitro with [35S]methionine using reticulocytes in the absence or presence of microsomes.
The translated proteins were incubated in the absence or presence of PNGase F or
proteinase K, with or without TX-100. The proteins were separated by SDS-PAGE and
were analyzed by autoradiography. Schematic representations of the corresponding bands
are indicated on the right side of the gel.
autoradiography. In the absence of microsomes, a major 48-kDa band was observed (Fig.
2C, lane I, band 1), which corresponded to the MW of the unglycosylated precursor form of
CLN5. The lower MW bands most likely corresponded to CLN5 translated from other
potential AUG codons (aa 30, 50, and 62), as previously reported [Isosomppi et al., 2002;
Vesa et al., 2002], or to degradation products. In the presence of microsomes, additional
bands with apparent MW of 70–72 kDa and 60 kDa were observed (Fig. 2C, lane II, bands
2 and 3, respectively). These bands corresponded to the expected MW of the glycosylated
precursor and the mature forms (in which the SP has been cleaved) of CLN5, respectively.
Indeed, the PNGase F treatment induced the deglycosylation and a ∼25-kDa downshift of
the precursor and mature forms of CLN5, resulting in 48 and 34 kDa bands (Fig. 2C, lane
III, bands 1 and 4, respectively). These results indicated that CLN5 is glycosylated and
cleaved in the presence of microsomes. When the microsomes were treated with proteinase
K, the 60-kDa mature form of CLN5 was protected (Fig. 2C, lane IV, band 3), whereas the
70–72-kDa precursor was shifted to an apparent MW of 65 kDa (Fig. 2C lane IV, band 5).
This 8–9 kDa shift corresponded to the digestion of the cytoplasmic N-terminus of CLN5
(76 amino acids). This is in agreement with topology Model 2 (Fig. 2A). Furthermore, the
unprocessed precursors and the degradation products observed in lane I were also degraded
in the presence of proteinase K, indicating that they are located on the cytoplasmic side of
the microsomes (Fig. 2C, lane IV). When TX-100 was added to the proteinase K treatment,
the bands observed in lane IV became sensitive to digestion (Fig. 2C, lane V), showing that
these proteins are located in the lumen. Taken together, these results indicated that the Nterminus of CLN5 is located in the cytoplasm, that its first hydrophobic region is a TM
domain, that the second hydrophobic region does not cross the membrane, and that the
protein C-terminus is located in the lumen of the microsomes. CLN5 is thus synthesized as
a type II TM precursor protein that is rapidly glycosylated and cleaved at the SP site
126
located after the TM domain, with the resulting mature form of CLN5 being entirely
located in the lumen of the intracellular compartments.
CLN5 is Tightly Bound to the Membrane
CLN5 has been previously reported to be a soluble protein [Holmberg et al., 2004;
Isosomppi et al., 2002]. However, even though the mature form of CLN5 is devoid of a TM
domain, we observed that only a small amount is secreted into the medium (Fig. 1C),
suggesting that it is directly or indirectly attached to the membrane. We then investigated
whether CLN5 was associated with the membrane using subcellular fractionation assays on
HEK293 cells expressing the various tagged CLN5 proteins. Cell homogenates (PNS) were
centrifuged at high speed, and the resulting pelleted membrane fractions (P) and
supernatants containing soluble proteins (S) were analyzed by Western blotting using
antibodies directed against the tagged CLN5 proteins. 3xHA-CLN5 was mainly found in
the membrane fraction, as expected, since this precursor form contains a TM domain.
However, the CLN5-HA and SP-Flag-CLN5 proteins, which do not have a TM region,
were also mainly found in the membrane fraction (Fig. 3A).
To determine the biochemical nature of the association of CLN5 with the membrane,
we conducted a series of extraction experiments to discriminate between peripheral and
integral membrane associations. The isolated membrane fraction described in Figure 3A
was incubated with 1 M NaCl or 150 mM Na2CO3 (pH 11.5), two agents known to release
loosely attached peripheral membrane proteins, or with 5 M urea, one chaotropic agent that
can extract strongly attached peripheral membrane proteins (Fig. 3B). The membranes were
centrifuged, and the pellets (P) and supernatants (S) were analyzed for the partitioning of
tagged CLN5 proteins. As shown in Figure 3B, both the precursor and the mature forms of
CLN5 remained associated with the membrane under all the extraction conditions. These
results strongly suggested that mature CLN5 is tightly associated with membrane and
behaves as an integral membrane protein.
To further analyze the association of mature CLN5 with the membrane, TX-114
partitioning was performed to segregate soluble and membrane-bound proteins into the
127
Figure 3 - Mature CLN5 is tightly associated with the membranes.
A: Membrane fractionation of tagged CLN5 proteins. HEK293 cells transiently expressing
3×HA-CLN5, CLN5-HA, or SP-Flag-CLN5 were lysed in TNES buffer and were
centrifuged at low speed. The PNS was centrifuged at 100,000g to separate the membrane
fraction (P) and the soluble proteins (S). The same volumes of S and P were separated by
SDS-PAGE and were immunoblotted using anti-HA or anti-Flag antibody. B: Membrane
subfractionation of tagged CLN5 proteins. Membranes (P) isolated as described in (A) were
resuspended in TNES buffer containing 1 M NaCl, 150 mM sodium carbonate (pH 11.5),
or 5 M urea, and were incubated for 30 min at 4°C. The membranes were sedimented at
100,000g. The supernatant (S) and pellet (P) fractions were separated by SDS-PAGE and
were immunoblotted using anti-HA or anti-Flag antibody. C: TX-114 partitioning of tagged
CLN5 proteins. Membranes (P) isolated as described in (A) were solubilized and were
extracted using TX-114 membrane protein extraction reagent Mem-PER protein extraction
kits. The supernatant (aqueous [AQ]) and pellet (detergent [Det]) fractions were separated
by SDS-PAGE and were immunoblotted using anti-HA or anti-Flag antibody. Endogenous
cytosolic protein EEA1, peripheral protein GM130, and transmembrane protein TfR were
used as fractionation controls.
aqueous phase and integral membrane proteins into the detergent phase [Bordier, 1981].
The membrane pellet fraction described in Figure 3A was dissolved in TX-114 buffer,
incubated, and centrifuged. The two phases were then analyzed by Western blotting (Fig.
3C). The 3xHA-CLN5 precursor form containing a TM domain remained mainly
membrane bound, whereas CLN5-HA and SP-Flag-CLN5 were distributed almost evenly
between the aqueous and detergent fractions. These results indicated that the mature form
of CLN5 is tightly bound to the membrane but that it does not behave like a typical integral
membrane protein.
128
CLN5 is Anchored to the Membrane Through an Amphipathic Helix
The aa 353–373 region of human CLN5, which was predicted to be a potential TM
domain, contains a pattern of hydrophobic and nonpolar residues that is well conserved
among CLN5 orthologues (Fig. 4A). We showed that this region is not a TM region. A
helical wheel projection using the amino acid interfacial hydrophobicity scale described by
Wimley and White (1996) revealed that the aa 353–392 fragment of CLN5 can adopt an
amphipathic helix (AH) conformation, with two hydrophobic patches at each end (Fig. 4B).
As depicted in the helical net diagram (Fig. 4C), these two patches lie on one face of the
helix and can directly adsorb at the interface of the lipidic membrane. To test the hypothesis
that the predicted AH mediates the association with the membrane, several deletion mutants
of the mature form of CLN5 with or without the AH were generated and fused to the Cterminus of GFP (Fig. 4D). The membrane association of these GFP-tagged proteins
expressed in HEK293 cells was examined by subcellular fractionation and urea extraction
followed by Western blotting (Fig. 4E). All the GFP fusion proteins containing the AH
domain (GFP-CLN5 353–392, GFP-CLN5 353–407, and GFP-CLN5 97–407) were mainly
present in the membrane-enriched fraction, even after urea extraction, indicating that the
AH is sufficient for membrane association. On the other hand, the GFP-fusion protein
without the AH (GFP-CLN5 97–352) was found exclusively in the soluble fraction after the
urea treatment, as was GFP alone. These results suggested that the AH is a key factor in
membrane anchoring of the mature form of CLN5.
Effect of Genetic Pathological Mutations on the Membrane Anchoring and Secretion
of CLN5
To improve our understanding of the impact of NCL pathological mutations on the
function of CLN5, we analyzed the membrane association of SP-Flag-CLN5 carrying four
variant late infantile CLN5 disease-causing mutations (Fig. 5A): the EUR p.Asp279Asn
mutation [Savukoski et al., 1998], the FinM p.Tyr392* mutation [Savukoski et al., 1998],
the SWE p.Glu253* mutation [Holmberg et al., 2000], and the NFL p.Glu352* mutation
[Moore et al., 2008]. The SWE and NFL mutations both resulted in truncated proteins
without the AH and the C-terminal region, whereas the FinM mutant retained the AH but
not the C-terminal region. The EUR mutant was included because it retained the AH and
129
Figure 4 - An AH in CLN5 mediates tight membrane association.
A: Sequence comparison of the potential AH from different species. The numbers refer to
amino acid positions in human CLN5. *, Fully conserved residues., similar residues. B:
Helical wheel projection of CLN5 aa 353–392. The amino acids are color-coded based on
the hydrophobic character of the residue: green for hydrophobic (W, F, Y, L, I, M, C),
white for neutral (A, T, S, V, G), and red for hydrophilic and charged (E, D, K, R, H, Q, P,
N). Two hydrophobic patches can be seen on the two sides of the helix. C: Helical net
130
projection of CLN5 aa 353–392. In these representations, the cylindrical helix region is cut
longitudinally and flattened into the plane of the page. The amino acids are color-coded as
described in (B). Two hydrophobic patches can also be seen in this diagram. D: Schematic
representation of the different GFP-tagged CLN5 cDNA constructs. GFP-CLN5 97–407,
the mature form of CLN5; GFP-CLN5 97–352, which does not contain the putative AH;
GFP-CLN5 353–407, which contains the AH and the C-terminus of CLN5; and GFP-CLN5
353–392, which contains only the putative AH. E: Membrane association analysis of the
different GFP-tagged CLN5 proteins. HEK293 cells transiently expressing the GFP or the
GFP-CLN5 proteins described in (D) were subfractionated and treated with 5 M urea as
described in Figure 3. The PNS, supernatant (S), and pellet (P) fractions were separated by
SDS-PAGE and were immunoblotted using anti-GFP antibody. Cytosolic EEA1, peripheral
GM130, and TM protein TfR were used as fractionation controls. Representative blots are
shown.
the C-terminus. The membrane associations of these pathological CLN5 mutants expressed
in HEK293 cells were examined by subcellular fractionation and urea extraction followed
by Western blotting (Fig. 5B). The EUR mutation did not affect the membrane association
since it behaves like WT SP-Flag-CLN5. The FinM mutant protein was more sensitive to
urea since it was partially found in the soluble fraction following urea extraction. In
contrast, the SWE and NFL mutant proteins were found exclusively in the soluble fraction
following urea extraction. These results suggested that CLN5 pathological mutants
deprived of their AH are not tightly associated with the membrane.
Pulse-chase assays were then performed to determine whether the mutations altered
the secretion of CLN5. HEK293 cells expressing WT SP-Flag-CLN5 or carrying the EUR,
FinM, SWE, or NFL mutations were pulsed for 5 min and were chased for different periods
of time, after which CLN5 was immunoprecipitated from the medium and cell extracts
(Fig. 5C). The EUR and FinM mutations did not affect the kinetics of maturation and
secretion of CLN5 since these mutants behave like WT SP-Flag-CLN5. A deglycosylation
analysis confirmed that the glycosylation of the EUR and FinM mutant proteins are not
altered (Supp. Fig. S3). However, the SWE and NFL mutations affected the maturation and
stability of CLN5 (Fig. 5C; intracellular). After a 0-min chase, bands were detected at the
predicted MW of the SWE and NFL glycosylated precursors (37–39 and 60 kDa bands,
respectively; Fig. 5C, black arrowheads) and glycosylated mature forms (∼29 and 50 kDa
bands, respectively; Fig. 5C, white arrowheads), suggesting cotranslational glycosylation
131
Figure 5 - Membrane association and secretion of Flag-tagged CLN5 carrying the
NCL pathological mutations.
A: Schematic representation of the relative localization and amino acid changes of four
pathological CLN5 mutations causing variant late infantile CLN5 disease: EUR
(c.835G>A, p.Asp279Asn), FinM (c.1175_1176delAT, p.Tyr392*) SWE (c.757G>T,
p.Glu253*), and NFL (c.1054G>T, p.Glu352*) mutations. Numbering is based on the
132
cDNA sequence (GenBank: NM_006493.2). The A of ATG of the initiator methionine
codon is denoted as nucleotide 1. TM, transmembrane region; black box, Flag tag. B:
Membrane association analysis of Flag-tagged CLN5 mutant proteins. HEK293 cells
transiently expressing the WT SP-Flag-CLN5 or carrying the EUR, FinM, SWE, or NFL
pathological mutation were subfractionated and treated with 5 M urea as described in
Figure 3. The supernatant (S) and pellet (P) fractions were separated by SDS-PAGE and
were immunoblotted using anti-Flag antibody. Cytosolic EEA1, peripheral GM130, and
TM protein TfR were used as fractionation controls. Representative blots are shown. C:
Analysis of the maturation and secretion of Flag-tagged CLN5 mutant proteins. HEK293
cells transiently expressing the WT SP-Flag-CLN5, or the EUR, FinM, SWE, or NFL
mutants were pulsed for 5 min with 35S-methionine and were chased for the indicated
periods of time. The Flag-tagged CLN5 proteins were immunoprecipitated from the cell
lysates, and the culture media collected after each chase period. The proteins were
separated by SDS-PAGE and were analyzed by autoradiography. The films were exposed
for 48 hr. *, Nonspecific band; black arrowhead, precursor polypeptide; white arrowhead,
mature polypeptide.
and SP cleavage. Surprisingly, the size and/or amounts of the mature forms of these
mutants were clearly modified following the different chase periods. After a 2-hr chase, the
amount of mature NFL protein was significantly reduced, whereas the MW of SWE
decreased to ∼25 kDa, which was probably caused by partial deglycosylation. Indeed, the
PNGase F treatment indicated that this 25-kDa band is a glycosylated form of SP-cleaved
SWE (Supp. Fig. S3). After a 4-hr chase, both mutants were barely detectable in the cell
lysate. Since the levels of secreted SWE and NFL mutant proteins were no higher than that
of WT CLN5 (Fig. 5C; extracellular), we hypothesized that this was due to the degradation
of the mutated CLN5 proteins by the proteasomal machinery. This was confirmed by the
restoration of SWE and NFL protein levels when the cells were chased for 4 hr in the
presence of proteasomal enzyme inhibitor MG132 (Supp. Fig. S4). Moreover, the amount
of secreted NFL protein was lower than that for the other mutants (Fig. 5C; extracellular),
suggesting that the high level of degradation reduces NFL protein secretion. Lastly, the
lower MW bands detected in the lysates following the different chase periods of the SWE
and NFL mutants most likely correspond to differential glycosylation of the SP-cleaved
proteins and to degradation products. Taken together, these findings indicated that the
EUR, FinM, SWE, and NFL mutations do not affect the processing (cleavage of SP) or
glycosylation of CLN5, whereas the SWE and NFL mutations affect the membrane
association and stability of CLN5. However, all the mutations induced the relocalization of
133
CLN5 to the ER (Supp. Fig. S5). This is the first time that the NFL mutation has been
characterized.
Discussion
Mutations in the CLN5 gene result in NCL, a severe neurodegenerative disorder that
mainly affects children. The basic properties of CLN5 such as its structure and solubility
need to be clarified to better understand the role of this protein in the pathogenesis of NCL.
Previous analyses of the amino acid sequence of CLN5 using various prediction programs
resulted in different representations of the structure of CLN5. The original model predicted
two TM domains and cytoplasmic-oriented N- and C-termini [Savukoski et al., 1998]. In
contrast, current bioinformatic tools predict one TM domain, a cytoplasmic N-terminus,
and a luminal C-terminus. However, some programs have also predicted that CLN5 is a
soluble protein [Holmberg et al., 2004]. The present study is the first to experimentally
define the topology of CLN5. Using proteinase K protection assays on various epitopetagged CLN5 proteins expressed in cells and on untagged CLN5 translated in vitro, we
showed that the N-terminus of CLN5 is located in the cytoplasm, whereas its C-terminus is
located in the lumen of organelles. We thus ruled out the presence of a second TM domain
and demonstrated that CLN5 is synthesized as a type II single pass TM protein. This
precursor form is rapidly cleaved at the SP site located after the TM domain, with the
resulting mature form of CLN5 being entirely located in the lumen of intracellular
compartments.
Mature CLN5 has mostly been reported to be a soluble protein based on the fact that
it is secreted, that it contains mannose-6-phosphate residues, and that mouse CLN5 can be
extracted with TX-114 [Holmberg et al., 2004; Isosomppi et al., 2002; Sleat et al., 2006a;
Vesa et al., 2002]. In the present study, we showed that the mature form of CLN5, which is
devoid of a TM domain, is not a soluble protein but is tightly associated with the membrane
through an AH. Mature CLN5 was resistant to high salt, alkaline sodium carbonate, and
urea membrane extraction whereas TX-114 induced a partial partitioning into the aqueous
fraction, indicating that CLN5 does not behave like a typical peripheral protein or integral
134
membrane protein. This can be explained by the presence of a predicted AH in the aa 353–
392 region composed of two in-plane hydrophobic patches that would directly adsorb at the
interface of the lipidic membrane. We used GFP fusion experiments to show that the
predicted AH of CLN5 is sufficient to tightly hook GFP to the membrane. This is the first
report of the presence of an AH in CLN5 involved in its membrane association. The
presence of this AH may explain the slow trafficking of CLN5 and its localization to
different intracellular compartments. AH is a key structure for the membrane anchoring and
retention of other luminal proteins that lack a TM domain, including the viral Erns
glycoprotein [Burrack et al., 2012; Fetzer et al., 2005], torsin A [Vander Heyden et al.,
2011], and the cyclooxygenase isozymes COX-1 and COX-2 [Simmons et al., 2004].
Furthermore, while CLN3, another NCL protein, has been modeled with one luminal AH
and six TM helices [Nugent et al., 2008], the specific role of AH in the function of CLN3 is
unknown. AH appears to mediate more than just membrane anchoring and has been
variously described as an autoinhibitory protein activity domain, a protein interaction
domain, or a membrane curvature sensor/modulator that can respond to and modulate the
physical properties of membranes [Boucrot et al., 2012; Cornell and Taneva, 2006; Shih et
al., 2011; Stern et al., 2013]. As such, the involvement of the AH of CLN5 in functions
other than membrane anchoring needs to be investigated.
The mature form of overexpressed CLN5 has been detected in the extracellular
medium in several studies [Isosomppi et al., 2002; Kollmann et al., 2005; Vesa et al.,
2002], including the present work. We found that a small amount (∼20%) of mature CLN5
is secreted into the medium after a 2–4-hr chase. However, CLN5 is secreted considerably
more slowly (>1 hr) than most constitutively secreted proteins, which is consistent with our
results indicating that CLN5 is membrane anchored. This raises a question concerning the
functional significance of extracellular CLN5. The secretion of CLN5 may be necessary for
its turnover, or CLN5 may have unknown extracellular functions. In addition,
overexpression may alter the retention of mature CLN5. The secretion of endogenous
CLN5 will thus have to be validated when the appropriate antibodies become available.
135
We noted that the predicted AH domain in CLN5 is highly conserved among species,
suggesting that it has major structural constraints with respect to function and localization.
Since many known NCL pathological missense mutations are located in or immediately
adjacent to the predicted AH domain [Mole et al., 2011], we analyzed the impact of these
mutations on CLN5 membrane anchoring and secretion. The membrane association of two
AH-deficient CLN5 mutants (p.Glu253* [SWE] and p.Glu352* [NFL]) was dramatically
impaired, whereas mutants truncated immediately after the AH (p.Tyr392* [FinM]) and
mutations that did not alter the AH (p.Asp279Asn [EUR]) were not or only slightly
affected. The partial membrane dissociation of FinM might be because the C-terminal
region plays a partial role in membrane association, or because its absence renders the AH
unstable. Surprisingly, the absence of the membrane anchoring AH domain in the
p.Glu253* and p.Glu352* mutants did not lead to increased secretion of CLN5 but, rather,
resulted in their recognition and degradation by the ER quality control system, suggesting
that AH could play a role in protein stability in addition to membrane anchoring. Schmiedt
et al. (2010) also reported that the p.Glu253* and p.Tyr392* mutations resulted in unstable
proteins. However, in our hands, the stability of the p.Tyr392* mutant was not affected.
This discrepancy might be due to the different analytical methods used. As previously
reported, none of the CLN5 disease mutations alter the SP cleavage or glycosylation of
CLN5, but they all result in the mislocalization of CLN5 in the ER, suggesting that the
mutated proteins are misfolded and are retained in the ER [Lyly et al., 2009; Schmiedt et
al., 2010]. However, based on the pulse-chase assay, the mutant CLN5 polypeptides
(except NFL that is rapidly degraded) were secreted at the same level as WT CLN5,
suggesting that they are sufficiently correctly folded to pass the quality control of the
intracellular sorting system in the Golgi but are not sorted to lysosomes. These results are
in line with the observation that CLN5 mutants contain complex sugars, suggesting that
they reach the Golgi but are transported back to the ER [Schmiedt et al., 2010]. Two other
mutations in the AH (p.Leu358Alafs*4 and p.Trp379Cys) have been reported to lead to the
redistribution of CLN5 to the ER [Lebrun et al., 2009]. To better understand the role of the
AH in CLN5 function, it would be interesting to determine whether these mutations, as
well as others identified in the AH [Mole et al., 2011], alter the membrane anchoring,
secretion, and stability of CLN5.
136
Summary and Conclusion
Figure 6 - Schematic model for human CLN5.
A: CLN5 is synthesized as a single pass type II TM precursor glycoprotein. The Nterminus faces the cytoplasm, and the C-terminus is located in the lumen of the
intracellular compartment. B: The SP is rapidly cleaved, and the resulting mature protein
is tightly associated with the lumen of the membrane through an amphipathic anchor
region. The TM domain (aa 76–91) and anchor region (aa 353–392) of human CLN5 are
depicted as a dark rectangle. The predicted SP cleavage site is indicated (aa 96).
Nucleotide numbering is based on GenBank reference sequence NM_006493.2. The
figure is not to scale.
We propose a new topological model for CLN5 in which its structure contains a
cytoplasmic N-terminus and one TM segment, which are cotranslationally cleaved in the
ER by a signal peptidase, and a large luminal C-terminal domain with a previously
unidentified AH, which is involved in the membrane anchoring of the mature form of
CLN5 (Fig. 6). This model provides new molecular insights into the results of previous
studies. It suggests that the reported interactions of mature CLN5 with CLN1, CLN2,
CLN3, CLN6, and CLN8 involve the luminal domains of these proteins [Lyly et al., 2009;
Vesa et al., 2002]. It also indicates that the reported role of CLN5 in the recruitment of
retromers on the cytoplasmic side of the membrane is indirect and is mediated by a TM
interactive partner [Mamo et al., 2012]. Furthermore, the lysosomal transport of CLN5
would not involve a direct interaction with cytosolic trafficking components but would
rather involve a luminal sorting motif or a domain of interaction with a lysosomal sorting
protein. CLN5 lysosomal targeting has been reported to be independent of the mannose-6137
phosphate receptor and sortilin [Mamo et al., 2012; Schmiedt et al., 2010]. Other possible
candidates are LIMP-II, the sorting receptor for glucocerebrosidase [Reczek et al., 2007],
and palmitoyl protein thiosterase 1 (PPT1/CLN1), which facilitates the lysosomal transport
of mutated CLN5 [Lyly et al., 2009]. More work is required to elucidate the sorting and
trafficking mechanisms for CLN5. In conclusion, the elucidation of the proper topology of
CLN5 represents a crucial step toward acquiring a better understanding of the mechanism
of action of CLN5 and clarifying its role in the pathogenesis of NCL.
Acknowledgments
We wish to thank Dr. Stéphane Lefrançois (Université de Montréal, QC, Canada) for
the human 3xHA-CLN5 cDNA and helpful discussions and Dr. Pierre Lavigne (Université
de Sherbrooke, QC) for his valuable advice regarding the analysis of the amphipathic helix.
We also wish to thank Caroline Thériault and Marilène Paquette for their technical
assistance. H.L. holds a fellowship from the National Sciences and Engineering Research
Council of Canada (NSERC). C.L. holds a Canada Research Chair in Cellular
Pharmacology.
138
References
Bessa C, Teixeira CA, Mangas M, Dias A, Sa Miranda MC, Guimaraes A, Ferreira JC,
Canas N, Cabral P, Ribeiro MG. 2006. Two novel CLN5 mutations in a Portuguese
patient with vLINCL: insights into molecular mechanisms of CLN5 deficiency. Mol
Genet Metab 89:245–253.
Bordier C. 1981. Phase separation of integral membrane proteins in Triton X-114 solution.
J Biol Chem 256:1604–1607.
Boucrot E, Pick A, Camdere G, Liska N, Evergren E, McMahon HT, Kozlov MM. 2012.
Membrane fission is promoted by insertion of amphipathic helices and is restricted by
crescent BAR domains. Cell 149:124–136.
Burrack S, Aberle D, Burck J, Ulrich AS, Meyers G. 2012. A new type of intracellular
retention signal identified in a pestivirus structural glycoprotein. FASEB J 26:3292–
3305.
Cornell RB, Taneva SG. 2006. Amphipathic helices as mediators of the membrane
interaction of amphitropic proteins, and as modulators of bilayer physical properties.
Curr Protein Pept Sci 7:539–552.
Fetzer C, Tews BA, Meyers G. 2005. The carboxy-terminal sequence of the pestivirus
glycoprotein E(rns) represents an unusual type of membrane anchor. J Virol
79:11901–11913.
Haltia M. 2006. The neuronal ceroid-lipofuscinoses: from past to present. Biochim Biophys
Acta 1762:850–856.
Holmberg V, Jalanko A, Isosomppi J, Fabritius AL, Peltonen L, Kopra O. 2004. The mouse
ortholog of the neuronal ceroid lipofuscinosis CLN5 gene encodes a soluble
lysosomal glycoprotein expressed in the developing brain. Neurobiol Dis 16:29–40.
Holmberg V, Lauronen L, Autti T, Santavuori P, Savukoski M, Uvebrant P, Hofman I,
Peltonen L, Jarvela I. 2000. Phenotype-genotype correlation in eight patients with
Finnish variant late infantile NCL (CLN5). Neurology 55:579–581.
Isosomppi J, Vesa J, Jalanko A, Peltonen L. 2002. Lysosomal localization of the neuronal
ceroid lipofuscinosis CLN5 protein. Hum Mol Genet 11:885–891.
Kollmann K, Mutenda KE, Balleininger M, Eckermann E, von Figura K, Schmidt B, Lubke
T. 2005. Identification of novel lysosomal matrix proteins by proteome analysis.
Proteomics 5:3966–3978.
Kollmann K, Uusi-Rauva K, Scifo E, Tyynela J, Jalanko A, Braulke T. 2013. Cell biology
and function of neuronal ceroid lipofuscinosis-related proteins. Biochim Biophys
Acta 1832:1866–1881.
Lebrun AH, Storch S, Ruschendorf F, Schmiedt ML, Kyttala A, Mole SE, Kitzmuller C,
Saar K, Mewasingh LD, Boda V, Kohlschütter A, Ullrich K, Braulke T, Schulz A.
2009. Retention of lysosomal protein CLN5 in the endoplasmic reticulum causes
neuronal ceroid lipofuscinosis in Asian sibship. Hum Mutat 30:E651–E661.
Lyly A, von Schantz C, Heine C, Schmiedt ML, Sipila T, Jalanko A, Kyttala A. 2009.
Novel interactions of CLN5 support molecular networking between Neuronal Ceroid
Lipofuscinosis proteins. BMC Cell Biol 10:83.
Mamo A, Jules F, Dumaresq-Doiron K, Costantino S, Lefrancois S. 2012. The role of
ceroid lipofuscinosis neuronal protein 5 (CLN5) in endosomal sorting. Mol Cell Biol
32:1855–1866.
139
Matlin KS, Simons K. 1983. Reduced temperature prevents transfer of a membrane
glycoprotein to the cell surface but does not prevent terminal glycosylation. Cell
34:233–243.
Mole SE, Williams RE, Goebel HH. 2005. Correlations between genotype, ultrastructural
morphology and clinical phenotype in the neuronal ceroid lipofuscinoses.
Neurogenetics 6:107–126.
Mole SE, Williams RE, Goebel HH. 2011. The neuronal ceroid lipofuscinoses (Batten
disease). In: Mole SE, Williams RE, Goebel HH, editors. New York: Oxford
University Press. p 444.
Moore SJ, Buckley DJ, MacMillan A, Marshall HD, Steele L, Ray PN, Nawaz Z, Baskin B,
Frecker M, Carr SM, Ives E, Parfrey PS. 2008. The clinical and genetic epidemiology
of neuronal ceroid lipofuscinosis in Newfoundland. Clin Genet 74:213–222.
Nugent T, Mole SE, Jones DT. 2008. The transmembrane topology of Batten disease
protein CLN3 determined by consensus computational prediction constrained by
experimental data. FEBS Lett 582:1019–1024.
Qoronfleh MW, Benton B, Ignacio R, Kaboord B. 2003. Selective enrichment of membrane
proteins by partition phase separation for proteomic studies. J Biomed Biotechnol
2003:249–255.
Reczek D, Schwake M, Schroder J, Hughes H, Blanz J, Jin X, Brondyk W, Van Patten S,
Edmunds T, Saftig P. 2007. LIMP-2 is a receptor for lysosomal mannose-6phosphate-independent targeting of beta-glucocerebrosidase. Cell 131:770–783.
Santavuori P. 1988. Neuronal ceroid-lipofuscinoses in childhood. Brain Dev 10:80–83.
Savukoski M, Klockars T, Holmberg V, Santavuori P, Lander ES, Peltonen L. 1998. CLN5,
a novel gene encoding a putative transmembrane protein mutated in Finnish variant
late infantile neuronal ceroid lipofuscinosis. Nat Genet 19:286–288.
Schmiedt ML, Bessa C, Heine C, Ribeiro MG, Jalanko A, Kyttala A. 2010. The neuronal
ceroid lipofuscinosis protein CLN5: new insights into cellular maturation, transport,
and consequences of mutations. Hum Mutat 31:356–365.
Shih YL, Huang KF, Lai HM, Liao JH, Lee CS, Chang CM, Mak HM, Hsieh CW, Lin CC.
2011. The N-terminal amphipathic helix of the topological specificity factor MinE is
associated with shaping membrane curvature. PLoS One 6:e21425.
Simmons DL, Botting RM, Hla T. 2004. Cyclooxygenase isozymes: the biology of
prostaglandin synthesis and inhibition. Pharmacol Rev 56:387–437.
Sleat DE, Wang Y, Sohar I, Lackland H, Li Y, Li H, Zheng H, Lobel P. 2006a.
Identification and validation of mannose 6-phosphate glycoproteins in human plasma
reveal a wide range of lysosomal and non-lysosomal proteins. Mol Cell Proteomics
5:1942–1956.
Sleat DE, Zheng H, Qian M, Lobel P. 2006b. Identification of sites of mannose 6phosphorylation on lysosomal proteins. Mol Cell Proteomics 5:686–701.
Stern O, Hung YF, Valdau O, Yaffe Y, Harris E, Hoffmann S, Willbold D, Sklan EH.
2013. An N-terminal amphipathic helix in dengue virus nonstructural protein 4A
mediates oligomerization and is essential for replication. J Virol 87:4080–4085.
Vander Heyden AB, Naismith TV, Snapp EL, Hanson PI. 2011. Static retention of the
lumenal monotopic membrane protein torsinA in the endoplasmic reticulum. EMBO
J 30:3217–3231.
140
Vesa J, Chin MH, Oelgeschlager K, Isosomppi J, DellAngelica EC, Jalanko A, Peltonen L.
2002. Neuronal ceroid lipofuscinoses are connected at molecular level: interaction of
CLN5 protein with CLN2 and CLN3. Mol Biol Cell 13:2410–2420.
Wimley WC, White SH. 1996. Experimentally determined hydrophobicity scale for
proteins at membrane interfaces. Nat Struct Biol 3:842–848.
141
Supporting Information
Supp. Figure S1 - Steady state localization of untagged and tagged human CLN5.
HeLa cells transiently transfected with 3xHA-CLN5 (A–C), CLN5-HA (D-F),SP-FLAGCLN5 (G-I), or untagged CLN5 (J-L) together with LAMP1-GFP (D-I) were fixed with
methanol and immunostained using anti-HA (A, D), anti-FLAG (G), anti-c/32 (J), antiGFP (E, H), anti-Calnexin (B), or anti-LAMP1 (K) antibody. The stained cells were
examined by confocal fluorescence microscopy. The merged images (C, F, I, K) show a
partial overlap between CLN5-HA, SP-FLAG-CLN5, and untagged CLN5 and the
lysosomal marker LAMPI while 3HA-CLN5 colocalized with the ER marker Calnexin.
Scale bar, 10 μm.
142
Supp. Figure S2 - The secretion of CLN5 is inhibited at 20°C.
HEK293 cells transiently expressing the untagged and tagged CLN5 proteins were pulsed
for 5 min with 35S-methionine and were chased for 1 h at 37°C or 20°C. Untagged and
tagged CLN5 proteins were immunoprecipitated from the cell lysates, and culture media
collected after each chase period using anti-C/32 and anti-HA/-Flag antibodies,
respectively. The proteins were separated by SDS-PAGE and were analyzed by
autoradiography.
Supp. Figure S3 - CLN5 pathological mutants are N-glycosylated.
Lysates from HEK293 cells transiently expressing the WT SP-Flag-CLN5 or the EUR
(p.Asp279Asn), FinM (p.Tyr392*), SWE (p.Glu253*), or NFL (p.Glu352*) mutations
were incubated in the absence or presence of PNGaseF. The proteins were separated by
SDS-PAGE and were immunoblotted using anti-Flag antibody.
143
Supp. Figure S4 - Stabilization of the SWE and NFL mutants by proteasome
inhibitors.
HEK293 cells transiently expressing the SWE (p.Glu253*) or NFL (p.Gly352*) mutants
were pulsed for 5 min with 35S-methionine. The cells were then chased for the indicated
periods of time in the absence or presence of the lysosomal enzyme inhibitors leupeptin
(Leu, 1 mg/mL), pepstatin (Pep, 100 μM), and E64 (10 μg/mL), or the proteasomal
enzyme inhibitor MG132 (10 μM). The Flag-tagged CLN5 proteins were
immunoprecipitated from the cell lysates collected after each chase period. The proteins
were separated by SDS-PAGE and were analyzed by autoradiography. The films were
exposed for 48 h.
144
Supp. Figure S5 - Steady state localization of CLN5 pathological mutants in HeLa
cells.
SP-Flag-CLN5 constructs carrying the p.Asp279Asn (EUR, A-C), p.Tyr392* (FinM, D–
F), p.Glu253* (SWE, G-I), or p.Glu352* (NFL, J-L) mutation were transiently
transfected into HeLa cells. The localizations of the CLN5 mutant proteins were studied
by immunofluorescent labeling followed by confocal microscopy using anti-Flag
antibody (A, D, G, J). Calnexin was used to label the ER (B, E, H, K). Colocalization is
indicated in yellow. Scale bar 10 μm.
145
ARTICLE 3
Rôle de Calnuc dans les céroïdes-lipofuscinoses neuronales
Auteurs de l’article: Heidi Larkin, Susan Cotman, Sara Mole et Christine Lavoie
Statut de l’article: En cours de rédaction
Avant-propos: J'ai écrit la première version de l'article et j'ai réalisé toutes les expériences
ainsi que tous les montages. Christine Lavoie a effectué les corrections du texte et a
effectué la prise des photos pour les expériences d'immunofluorescence. Sara Mole a fourni
les fibroblastes issus de patients atteints de céroïdes-lipofuscinoses neuronales et Susan
Cotman les lignées cellulaires stables de cervelet.
146
RÉSUMÉ – ARTICLE 3
Calnuc, un nouveau partenaire d'interaction pour CLN3 et CLN5, est potentiellement
impliqué dans les céroïdes-lipofuscinoses neuronales
Les céroïdes-lipofuscinoses neuronales (NCL), aussi connues sous le nom de maladies
de Batten, forment un groupe de maladies neurodégénératives caractérisées par une
accumulation de lipopigments autofluorescents, nommés lipofuscines, dans les lysosomes.
Jusqu'à présent, 14 gènes (désignés CLN1-14) ont été associés à la pathologie. Récemment,
CLN3 et CLN5 ont été montrées comme étant impliquées dans le transport intracellulaire par
une interaction et une régulation de Rab7, une petite protéine G nécessaire pour le recrutement
endosomial du complexe Rétromère. Nous avons récemment démontré le rôle de Calnuc, une
protéine ubiquitaire qui lie le calcium, dans le transport rétrograde de récepteurs lysosomiaux
des endosomes vers le réseau trans-golgien, en raison de son importance pour l'association
membranaire et l'activité de Rab7. Dans la présente étude, nous examinons les interactions
fonctionnelles entre Calnuc, CLN3 et CLN5, en plus de la potentielle implication de Calnuc
dans les NCL. À l'aide d'analyses de biochimie et de microscopie, nous avons montré que
Calnuc interagit avec CLN3 et CLN5 et colocalise avec ces protéines sur les endosomes. Nous
avons découvert des niveaux réduits de CLN3 et CLN5 dans les cellules déplétées en Calnuc et,
inversement, des niveaux réduits de Calnuc dans les cellules déplétées en CLN3 et CLN5. Ces
résultats suggèrent que Calnuc, CLN3 et CLN5 peuvent former un complexe stabilisé par leurs
interactions intramoléculaires. En microscopie confocale et électronique, nous avons observé
que la déplétion de Calnuc induit l’apparition de caractéristiques morphologiques cellulaires
typiques des NCL, soit un engorgement des lysosomes, une accumulation de matériel
autofluorescent et de la sous-unité C de l’ATP synthase mitochondriale, ainsi qu'une l’induction
accrue de l’autophagie. Dans les lignées de cellules de patients atteints de NCL, le profil
d'expression de Calnuc indique des niveaux protéiques diminués de Calnuc dans les cas
confirmés de type CLN3 et CLN5, de même que dans tous les cas de type orphelin, c'est-à-dire
qui sont génétiquement indéfinis mais présentent des similarités phénotypiques avec NCL.
Dans l'ensemble, ces données suggèrent que la fonction de Calnuc est altérée dans les NCL et
que Calnuc pourrait être impliquée dans la pathogenèse de la maladie.
147
Article 3
Evidence for a role of Calnuc, a novel interacting partner for CLN3 and CLN5, in
neuronal ceroid lipofuscinosis
Heidi Larkin1 and Christine Lavoie1‡
1
Department of Pharmacology, Faculty of Medicine and Health Sciences, Université de
Sherbrooke, Sherbrooke, QC, Canada
‡
Correspondence to: Christine Lavoie,
Department of Pharmacology, Faculty of Medicine, Universite de Sherbrooke,
3001-12e Avenue Nord, Sherbrooke, QC J1H 5N4, Canada
E-mail: [email protected]
Key words: Calnuc, nucleobindin, CLN5, CLN3, lipofuscinosis, lysosomal receptor,
endosomal sorting, lysosome
148
Abstract
The neuronal ceroid lipofuscinoses (NCLs), also known as Batten disease, are a
group of neurodegenerative diseases characterized by accumulation of autofluorescent
lipopigments appointed lipofuscins, in lysosomes. So far, 14 genes (designated CLN1-14)
have been associated with the pathology. Recently, CLN3 and CLN5 have been involved in
the proper trafficking of lysosomal receptors through the interaction and regulation of
Rab7, a small G protein required for the endosomal recruitment of retromers. We
previously highlighted the role of Calnuc, an ubiquitous calcium-binding protein, in the
retrograde transport of lysosomal receptors from endosomes to trans-Golgi network (TGN)
via the activation and membrane association of Rab7 (Larkin et al. 2016). In the present
study, we examined the functional interactions between Calnuc, CLN3 and CLN5 and the
potential implication of Calnuc in NCL pathogenesis. Using biochemical and microscopy
assays, we showed that Calnuc interacts with CLN3 and CLN5 and colocalizes with them
on endosomes. We found reduced levels of CLN3 and CLN5 in Calnuc-depleted cells and
vice-versa, a lower level of Calnuc in CLN3- and CLN5-depleted cells, suggesting that
Calnuc, CLN3 and CLN5 could form a protein complex stabilized by their intramolecular
interactions. Using confocal and electron microscopy, we observed that the depletion of
Calnuc induces cellular morphological features reminiscent of NCL such as lysosome
enlargement, accumulation of autofluorescent material and of subunit C of mitochondrial
ATP synthase (SCMAS), as well as an increased of autophagy induction. Expression
profiling of Calnuc in fibroblasts cell lines from NCL patients indicated a lower proteic
level of Calnuc in CLN3 and CLN5 genetically confirmed cases of NCL as well as in
orphan cases of NCL which remain undefined genetically but display phenotypes similar to
CLN gene deficiencies. Overall, these data suggest that Calnuc function is altered in human
NCL and that Calnuc could be a novel protein involved in NCL pathogenesis.
Introduction
Neuronal Ceroid Lipofuscinoses (NCLs) are severe neurodegenerative lysosomal
storage diseases that manifest in childhood and are characterized by developmental
149
regression, seizures, visual failure and mental retardation, culminating in premature death
(Mole et al. 2005; Jalanko and Braulke 2009). NCLs are relatively rare, with an incidence
of 1 in 100,000 live births worldwide, but appear to be more common (1:12,500 live births)
in northern Europe (Finland and Sweden) and Newfoundland (Santavuori 1988; Palmer et
al. 1989). At the cellular level, an accumulation of autofluorescent ceroid lipopigments (i.e.,
lipofuscin) and specific storage material ultrastructures are morphological indications of
NCL (Goebel and Wisniewski 2004; Mole et al. 2005; Cotman and Staropoli 2012). These
disorders result from mutations in at least 14 Ceroid-Lipofuscinosis Neuronal genes
(CLN1-CLN14) (Jalanko and Braulke 2009) and are classified clinically based on either the
mutation of the CLN gene or the age of onset of disease symptoms (Jalanko and Braulke
2009). Furthermore, there are still several groups of NCL patients that remain undefined
genetically. Infantile NCL (INCL; 0-2 years old) is associated with mutations in CLN1, late
infantile NCL (LINCL; 2-4 years old) is linked to mutations in CLN2, 5, 6, 7 and 8, and
juvenile NCL (JNCL or Batten disease; 5-10 years old) is related to mutations in CLN3.
The precise function and regulation of most of these CLN proteins are unknown. However,
recent studies have implicated CLN3 and CLN5 in the trafficking of lysosomal receptors.
CLN3, a multipass transmembrane (TM) protein localized mainly on endosomes and
lysosomes, has been reported to be involved in both post-Golgi anterograde and endosometo-TGN retrograde trafficking pathways (Cotman and Staropoli 2012). This role of CLN3 is
via its interaction with various proteins involved in different trafficking steps. CLN3 binds
to Rab7 and with various retro- and anterograde microtubular motor complexes responsible
for the movement of late endosomes and lysosomes (Uusi-Rauva et al. 2012). It regulates
Sed5, a Golgi SNARE involved in endosome-to-Golgi retrieval (Kama et al. 2011), as well
as Btn2p, a retromer accessory protein that facilitates specific protein retrieval from the
endosome to the Golgi (Kama et al. 2007). Recently, the activity of CLN5 has also been
linked to Rab7 activity and recruitment to endosomes (Mamo et al. 2012) providing the
first link to lysosomal function. Indeed, CLN5 depletion results in reduced levels of active
Rab7 and retromer membrane recruitment, key elements of the endosmal retrograde sorting
machinery, leading to the misdelivery and degradation of Sortilin and CI-MPR to lysosome
(Mamo et al. 2012). CLN5, is a luminal protein associated with the endolysosomal
membrane (Larkin et al. 2013). This indicates that the reported effect of CLN5 on the
150
lysosomal receptor trafficking is indirect and is mediated by a TM interactive partner.
Interestingly, CLN5 has been shown to directly interact with CLN3 (Vesa et al. 2002; Lyly
et al. 2009), suggesting that CLN3 and CLN5 cooperate on the endosome-to-Golgi
retrograde machinery for lysosomal receptors retrieval. Taken together, these studies imply
that a functional deficiency in CLN3 or CLN5 would impair the retrograde trafficking of
lysosomal receptors and the proper delivery of their cargos, which underlies the lysosomal
dysfunction observed in NCL disorders.
Calnuc (nucleobindin or NUCB1) is a ubiquitous and well-conserved protein
suggesting that it has important biological functions (Wendel, Sommarin et al. 1995; Miura
et al. 1996; Lin et al. 1998; Kawano et al. 2000). Indeed, the modulation of Calnuc
expression is associated with pathologies, such as cancer, lupus and Alzheimer’s, but its
precise cellular functions remained poorly understood. Calnuc is predominantly localized in
the cis-Golgi lumen where it is retained for a long period and constitutes the major Ca2+
storage pool via 2 EF-hand motifs (Lin et al. 1998; Lin et al. 1999). It is then secreted via
constitutive and constitutive-like pathways to the extracellular space where it has been
involved in bone matrix maturation (Lavoie et al. 2002). Calnuc is also found free in
cytosol or associated with the surface of the Golgi, endosomal membranes and secretory
granules (Brodeur et al. 2009; Lin et al. 2009). In addition to being a multicompartmental
protein, Calnuc is also a multifunctional protein comprised of a signal peptide, a putative
DNA-binding domain, a leucine zipper and 2 EF-hand motifs (Lin et al. 1998). These
characteristics potentiates its interactions with a growing list of partners, such as DNA
(Miura et al. 1992), heterotrimeric Gα proteins (Lin et al. 1998), COX (Ballif et al. 1996),
Necdin (Taniguchi et al. 2000) and LRP9 (Brodeur et al. 2009). Known as LRP10 in
humans, this latter is a LDL receptor cycling between the trans-Golgi network (TGN) and
early endosomes (Boucher et al. 2008). We have shown that cytoplasmic pool of Calnuc
plays an essential role in LRP9 retrieving from the endosomes to the TGN (Brodeur et al.
2009). Similarly, Calnuc affects other receptors transiting between the TGN and
endosomes, such as lysosomal receptors Sortilin and CI-MPR (Larkin et al. 2016),
suggesting that Calnuc plays a general role in endosomal receptor sorting. We
demonstrated that Calnuc interacts indirectly and colocalizes with Rab7 on endosome, that
151
Calnuc depletion impairs the activation and membrane association of Rab7 that lead to a
defect in the endosomal recruitment of retromer and the misdelivery and degradation of
MPR and Sortilin in lysosomes (Larkin et al. 2016). Thus Calnuc is novel player involved
in the endosome-to-TGN retrograde transport of lysosomal receptors.
Given that Calnuc affects the same molecular components of the endosomal sorting
machinery as CLN3 and CLN5, and have similar effect on lysosomal receptor retrieval, this
study aim at investigating the functional interactions/cooperation between Calnuc, CLN3
and CLN5. We observed a physical association between Calnuc, CLN3 and CLN5 and that
Calnuc depletion induces cellular morphological features reminiscent of NCL, suggesting
that Calnuc is new protein involved in NCLs.
Materials and methods
Antibodies and reagents
Anti-Flag M2 monoclonal antibodies (mAbs) were purchased from Sigma Aldrich
(Saint Louis, MO, USA), anti-HA mAbs from Covance (Emeryville, CA, USA),, anti-GFP
mAbs from Clontech (Mountain View, CA, USA), anti-Rab7 mAbs from Cell signalling
Technology (Danvers, MA, USA), anti-HA mAbs from Covance (Emeryville, CA, USA),
anti-LAMP 2 (H4B4) m Abs from Developmental Studies Hybridoma Bank at the
University of Iowa (Iowa, IA, USA). Anti-Calnuc polyclonal antibodies (pAbs) were
purchased from Aviva Systems Biology (San Diego, CA, USA), anti-EEA1 pAbs from
Thermo Scientific (Rockford, IL, USA), anti-Flag pAbs from Sigma Aldrich (Saint Louis,
MO, USA), anti-GFP pAbs from Molecular Probes (Eugene, OR, USA) and anti-LC3 pAbs
from Novus Biologicals (Littleton, CO, USA). Anti-Calnuc pAbs against mouse was a gift
from Marilyn Farqhuar (University of California San Diego) and anti-SCMAS pAbs from
Susan Cotman (Harvard Medical School).
DNA constructs
Mammalian expression vector pcDNA3.1 containing Calnuc-GFP fusion protein or
ΔSP-Calnuc (Calnuc without signal peptide) or ΔEF-Calnuc (Calnuc without calcium
152
binding domains) are described elsewhere (Weiss et al. 2001). GST-RILP was a gift from
Aimee L. Edinger (UC Irvine) and LC3-GFP from Sheela Ramanathan (Université de
Sherbrooke). Human CLN1 and CLN3 in pCMV6-XL5 vector as well as 3xHA-CLN5 in
the pReceiver-M06 vector were generous gifts from Stéphane Lefrançois (Université de
Montréal, QC, Canada). CLN1-HA, CLN3-HA and CLN5-HA were generated by
subcloning in the pcDNA3.1 vector. The SP-Flag-CLN5 construct was obtained by the
insertion of the Flag epitope at amino acid position 97.
Cell culture and transfection
HeLa cells were purchased from the American Type Culture Collection (Manassas,
VA, USA) and HEK293T cells were obtained from Alexandra Newton (University of
California, San Diego, CA, USA). The cells were grown at 37°C, with 5% CO2 atmosphere
control in Dulbecco’s modified Eagle’s high glucose medium (DMEM) (Invitrogen,
Carlsbad, CA, USA) containing 10% fetal bovine serum (FBS) (Hyclone Laboratories,
Logan, UT, USA) and 1% penicillin, Glutamine and streptomycin (PSG) (Invitrogen,
Carlsbad, CA, USA). Cerebellar cells stably expressing CLN3+/+ or CLN3Δex7/8/Δex7/8 were
obtained from Susan Cotman (Harvard Medical School) (Fossale 2004, BMC Neurosci. 5,
57). Cb cells maintained between 30 and 90% confluency and were grown at 33°C, with
5% CO2 atmosphere control, in DMEM with 10% heat-inactivated FBS, 24 mM KCl, 1%
PSG, and 200 μg/ml G418 (Invitrogen, Carlsbad, CA, USA). Fibroblasts cells from patients
were obtained from Sara Mole (University College London) and controls fibroblasts were
purchase at Coriell Biorepository. Those cells were culture at 37°C, with 5% CO2
atmosphere control in MEM or DMEM with 10% FBS and 1% PSG accordingly to
instructions. HeLa cells, HEK cells, and Cb cells were transfected with Lipofectamine 2000
transfection reagent (Invitrogen, Carlsbad, CA, USA) according to the manufacturers’
instructions.
Coimmunoprecipitation
HEK293 cells were plated in 60-mm culture dishes and transfected with the various
constructs. After 48 h, the cells were lysed in 50 mM Tris buffer (pH 7.4) containing 100
mM NaCl, 1% NP-40, and protease inhibitors. Cells were incubated for 1 h at 4oC and then
153
centrifuged at 15,800xg for 20 min. The cleared supernatants were incubated with primary
antibodies overnight at 4oC and then with protein A-sepharose (GE Healthcare,
Piscataway, NJ, USA) or protein G-Sepharose (Zymed, San Francisco, CA, USA) for 1 h.
The beads were washed three times in lysis buffer and boiled in Laemmli sample buffer.
Experiments containing multiple transmembrane protein CLN3 were instead incubated 1h
at TP in Laemmli sample buffer. Bound immune complexes were analyzed by SDS-PAGE
and immunoblotting.
Glutathione S-transferase (GST) pull-down assays
GST fusion proteins were expressed in E. coli BL21 and were purified on
glutathione-Sepharose 4B beads (Pharmacia, Piscataway, NJ, USA) according to the
manufacturer’s instructions. Lysate from HeLa cells obtained as described above, was
incubated overnight at 4oC with 20 μg of GST fusion proteins immobilized beads. The
beads were washed three times in lysis buffer and boiled in Laemmli sample buffer. The
bound proteins were separated by SDS-PAGE and detected by autoradiography or
immunoblotting.
Immunoblotting
The protein samples were separated on 10% or 12% SDS-PAGE gels and transferred
to nitrocellulose membranes (Perkin Elmer, Woodbridge, ON, Canada). The membranes
were blocked in Tris-buffered saline (20 mM Tris-HCl, pH 7.5, 150 mM NaCl) containing
0.1% Tween 20 and 5% nonfat dry milk, bovine serum albumin (BSA), or foetal bovine
serum (FBS) and incubated with primary antibodies for 1 h at RT or O/N at 4°C and then
with horseradish peroxidase-conjugated goat anti-rabbit or anti-mouse IgG (Bio-Rad,
Hercules, CA, USA) and enhanced chemiluminescence detection reagent (Pierce Chemical,
Rockford, IL, USA).
RNA Interference and Rescue
Nonspecific (CTL), Calnuc, CLN1, CLN3 and CLN5 siRNA scrambled II duplex
were purchased from Dharmacon Research (Chicago, IL, USA). HeLa cells were
transfected with a final concentration of 100 nM siRNA duplex using Lipofectamine 2000
154
reagent (Invitrogen) according to the manufacturer’s instructions. The cells were analyzed
72 h after the transfection. Tagged proteins were transfected with Fugene 12 h before the
immunofluorescence experiments. Reversal of phenotype (rescue) was performed as
described in (Brodeur et al. 2009). Briefly, HeLa cells were transfected with cDNA
encoding rat Calnuc-GFP, rat ΔSP-Calnuc-GFP, rat ΔEF-Calnuc-GFP or GFP alone using
Fugene 8–10 h after the initial transfection with human Calnuc siRNA, and the cells were
analyzed after 38–40 h.
Immunofluorescence
Cb cells were plated on coverslips. Twelve hours after the transfection, the cells were
fixed for 30 min in 3% paraformaldehyde (PFA) 100 mM phosphate buffer, pH 7.4,
permeabilized with 0.1% Triton X-100 for 10 min, blocked with 10% goat or fetal bovine
serum for 30 min, and incubated with primary antibodies for 1 h at RT, followed by Alexa
Fluor-488, 594 or 647-conjugated antibodies (Molecular Probes, Eugene, OR, USA) for 1 h
at RT. The specimens were visualized using an inverted confocal laser-scanning
microscope (FV1000, Olympus, Tokyo, Japan) equipped with a PlanApo 60x/1.42 oil
immersion objective (Olympus, Tokyo, Japan). Olympus Fluoview software version 1.6a
was used for image acquisition and analysis. The images were further processed using
Adobe Photoshop (Adobe Systems, San Jose, CA, USA). The quantification of the
immunofluorescence signal on endosome was previously described (Mamo et al. 2012).
Electronic microscopy
Cells were rinsed with PBS, prefixed for 15 min with a 1:1 mixture of culture
medium (Dulbecco’s modified Eagle’s medium) and freshly prepared 2.8% glutaraldehyde
in cacodylate buffer (0.1 M cacodylate and 7.5% sucrose) and then fixed for 30 min with
2.8% glutaraldehyde at room temperature. After two rinses, specimens were postfixed for 1
h with 2% osmium tetroxide in cacodylate buffer. Cells were then dehydrated using graded
ethanol concentrations (40, 70, 90, 95, and 100%, three times each) and coated two times
for 3 h with a thin layer of Araldite 502 resin (for ethanol substitution). Finally, the resin
was allowed to polymerize at 60°C for 48 h. The specimens were detached from the plastic
vessels, inverted in embedding molds, immersed in Araldite 502, and polymerized at 60°C
155
for 48 h. Ultramicrotome- prepared thin sections were contrasted with lead citrate and
uranyl acetate and then observed on a Jeol 100 CX transmission electron microscope. All
reagents were purchased from Electron Microscopy Sciences (Cedarlane, Hornby, ON,
Canada).
RNA analysis
RNA analysis were performed by the Plateforme RNomique de l’Université de
Sherbrooke. Total RNA extractions were performed on cell pellets using TRIzol
(Invitrogen, Carlsbad, CA, USA) with chloroform, following the manufacturer’s protocol.
The aqueous layer was recovered, mixed with one volume of 70% ethanol and applied
directly to a RNeasy Mini Kit column (Qiagen, Venlo, Netherlands). DNAse treatment on
the column and total RNA recovery were performed as per the manufacturer’s protocol.
RNA quality and presence of contaminating genomic DNA was verified as previously
described (Brosseau et al. 2010). RNA integrity was assessed with an Agilent 2100
Bioanalyzer (Agilent Technologies, Santa Clara, CA, USA). Reverse transcription was
performed on 750 ng total RNA with Transcriptor reverse transcriptase, random hexamers,
dNTPs (Roche Diagnostics, Basel, Switzerland), and 10 units of RNAseOUT (Invitrogen,
Carlsbad, CA, USA) following the manufacturer’s protocol in a total volume of 20 µl. All
forward and reverse primers were individually resuspended to 20–100 μM stock solution in
Tris-EDTA buffer (IDT) and diluted as a primer pair to 1 μM in RNase DNase-free water
(IDT). Quantitative PCR (qPCR) reactions were performed in 10 µl in 384 well plates on a
CFX-384 thermocycler (Bio-Rad, Hercules, CA, USA) with 5 μl of 2X iTaq Universal
SYBR Green Supermix (Bio-Rad, Hercules, CA, USA), 10 ng (3 µl) cDNA, and 200 nM
final (2 µl) primer pair solutions. The following cycling conditions were used: 3 min at
95°C; 50 cycles: 15 sec at 95°C, 30 sec at 60°C, 30 sec at 72°C. Relative expression levels
were calculated using the qBASE framework (Hellemans et al. 2007) and the housekeeping
genes Gapdh, Lamp1 and Clk2 for human cDNA. Primer design and validation was
evaluated as described elsewhere (Brosseau et al. 2010). In every qPCR run, a no-template
control was performed for each primer pair and these were consistently negative. All primer
sequences are available in Supplemental Table 1.
156
Statistical analysis
Experiments were performed at least in triplicate and results are expressed as means
intensity of protein normalized with loading control ± SD. Statistical significance between
two groups was assessed using the Student t-test whereas ANOVA was used for
comparison with three groups or more. P value < 0.05 was considered significant (*) and p
< 0.001 very significant (**).
Results
Calnuc interacts and colocalizes with CLN3 and CLN5.
Because we previously showed that Calnuc operates on the same molecular
components of the endosomal retrieval machinery (Rab7 and retromer) as CLN5, we first
investigated
whether
Calnuc could
interact
with
CLN5.
We
first
performed
immunoprecipitation experiments using HEK293 cells expressing GFP-tagged Calnuc or
GFP together with HA-tagged CLN5, and the proteins were immunoprecipitated using an
antibody against GFP (Figure 1A). We found an interaction between CLN5-HA and
Calnuc-GFP, but not with GFP alone (Figure 1A). As the cytosolic pool of Calnuc has been
involved in the recruitment of retromers acting on the activity of Rab7 and that CLN5 is a
luminal protein, the interaction between those two proteins must be indirect, probably
through a transmembrane protein. CLN3 is a six multipass transmembrane protein and a
CLN5 and Rab7 interacting partner (Vesa et al. 2002; Lyly et al. 2009; Uusi-Rauva et al.
2012). We thus tested if Calnuc can interact with CLN3. In HEK293 cells expressing GFPtagged Calnuc or GFP together with HA-tagged CLN3, we found an interaction between
CLN3-HA and Calnuc-GFP whereas no unspecific interaction was detect with GFP alone
(Figure 1B). Given that CLN3 interacts directly with CLN5 (Vesa et al. 2002; Lyly et al.
2009), we also determined whether Calnuc can form a complex with both CLN5 and
CLN3. HEK293 cells expressing Calnuc-GFP, CLN3-HA and SP-Flag-CLN5 were
immunonoprecipitated using antibody against Flag or Myc (as negative control). No
interaction was detected with anti-Myc antibody, but CLN3 and Calnuc co157
immunoprecipitated with CLN5 when anti-Flag antibody was used (Figure 1C). These
results suggest that Calnuc form a protein complex with both CLN3 and CLN5.
Figure 1 - Calnuc form a protein complex with CLN3 and CLN5 on endosomes.
Coimmunoprecipitation of Calnuc with (A) CLN5 and (B) CLN3. Lysates from HEK cells
transfected with GFP or Calnuc–GFP together with CLN3-HA or CLN5-HA were
158
immunoprecipitated with anti-GFP antibodies and then immunoblotted with anti-GFP or
anti-HA to detect Calnuc and CLN3 or CLN5 respectively. Calnuc-GFP is observed as two
bands, the lower of which most likely represents an N-terminal cleavage product. (C)
Coimmunoprecipitation of Calnuc with both CLN3 and CLN5. Lysates from HEK cells
transfected with Calnuc-GFP, CLN3-HA and SP-Flag-CLN5 were immunoprecipitated
with anti-Myc of anti-Flag and then immunoblotted with anti-GFP to detect Calnuc, antiHA to detect CLN3 or with anti-HA to detect CLN5. (D) Comparison of the intracellular
distribution of Calnuc, CLN3 and CLN5 in HeLa cells. (a, d, e) Calnuc-GFP, (b, d, f)
CLN3-HA, (c, e, f) SP-Flag-CLN5 are found on enlarged early endosomes created by the
expression of Rab5QL. The merged images (right insets) show a partial overlap (yellow)
between (d) Calnuc-GFP and CLN3-HA, (e) Calnuc-GFP and SP-Flag-CLN5 or (f) CLN3HA and SP-Flag-CLN5. (Central pannel) The three proteins partially colocalize together on
specific membrane microdomains of enlarged early endosomes. HeLa cells were
transfected with Calnuc-GFP, CLN3-HA, SP-Flag-CLN5 and Rab5QL.Twelve hours after
transfection, the cells were fixed, permeabilized and immunostained using anti-GFP, antiHA and anti-Flag antibodies. The labeled cells were examined by confocal fluorescence
microscopy. Scale bar, 10 μm.
To validate the presence of this protein complex in intact cells, we next compared the
intracellular distribution of Calnuc-GFP, CLN3-HA and SP-Flag-CLN5 in HeLa cells using
confocal microscopy. In order to help visualize the colocalization of Calnuc, CLN3 and
CLN5, enlarged endosomes were created by expressing Rab5 GTPase-deficient mutant
(Rab5Q79L) that increases homo- and heterotypic fusion, leading to the formation of
enlarged early endosomes (Stenmark et al. 1994). As previously reported Calnuc-GFP
predominantly localized in the Golgi region (Figure 1D, central panel), but a small amount
is also detected on cytoplasmic face of Rab5-enlarged endosomes (Figure 1Da). CLN3-HA
(Figure 1Db) and SP-Flag-CLN5 (Figure 1Dc) were mainly localized in Rab5-enlarged
endosomes and are already known to colocalize (Figure 1Df). Both proteins also partially
overlap with Calnuc in specific membrane microdomains (Figure 1Dd and 1De,
respectively). When the staining of Calnuc-GFP, CLN3-HA and SP-Flag-CLN5 were
superposed, we observe partial colocalization and close proximity of these protein in early
endosomes
microdomains
(Figure
1D,
central
panel
inset).
Together,
these
immunofluorescence and biochemical interaction assays suggest that Calnuc interacts with
CLN3 and CLN5, on microdomains of early endosomes, and that Calnuc would act on this
complex to regulate endosomal sorting of lysosomal receptors.
159
Calnuc depletion alters the levels of CLN3 and CLN5.
Because various NCL-causing mutations in CLN3 and CLN5 alter their intracellular
distribution and level of expression (Jarvela et al. 1999; Isosomppi et al. 2002; Vesa et al.
2002; Bessa et al. 2006; Lebrun et al. 2009; Schmiedt et al. 2010) and because CLN3 and
CLN5 depletion alter the trafficking of lysosomal receptors (Metcalf et al. 2008; Mamo et
al. 2012), we next examined whether Calnuc knockdown alters the levels of CLN3 and
CLN5. HeLa cells were transfected with control or Calnuc siRNA alone or together with
CLN3-HA or CLN5-HA and were then analyzed by western blotting. CLN1-HA was also
used as a control because it is a cargo molecule of MPR but does not affect its trafficking
(Lefrancois et al. 2003; Jalanko and Braulke 2009). Endogenous Calnuc was strongly (≥
90%) depleted at 72 h after transfection with specific small interfering RNA (siRNA), as
observed in immunoblotting experiments (Figure 2A). The steady-state levels of CLN3-HA
and CLN5-HA, but not CLN1-HA, were reduced in Calnuc knockdown cells compared
with control cells (Figure 2A). Conversely, we next analyzed the effect of CLN3 and CLN5
depletion on Calnuc protein level and distribution. When CLN3 is knockdown in HeLa
cells, we observe a reduction in endogenous Calnuc and CLN5-HA. Levels of Calnuc and
CLN3-HA also decrease in CLN5 depleted cells. The specificity of the effect of Calnuc on
the levels of CLN5 was also validated by a rescue experiment with siRNA-resistant forms
of wild-type, Calnuc without calcium binding domains (ΔEF-Calnuc) and cytosolic Calnuc
(ΔSP-Calnuc). Rat Calnuc was used as a rescue construct because it differs in many
nucleotides from human siRNA target sequences, rendering it resistant to the siRNA. The
results showed that reintroducing rat Calnuc-GFP, ΔEF-Calnuc-GFP or ΔSP-Calnuc-GFP
into Calnuc-depleted cells restored the basal levels of CLN5-HA to levels similar to those
of control cells (Figure 2B). These results suggest that the cytoplasmic pool of Calnuc, but
not the EF-hands, participates in the stabilization of CLN5. We next examined whether the
level of Calnuc is altered in CLN3 pathological culture cell model. We used two
immortalized mouse cerebellar precursor cells lines established from wild-type mice
(CbCln3+/+) (WT1 and WT2, Fig2C) or from a mouse model which contains a genetically
mutation of CLN3 found in the majority of human patients (CbCln3Δex7/8/Δex7/8) (Fossale et
al. 2004; Cao et al. 2006; Cao et al. 2011) (MT1 and MT2, Fig. 2C). This CLN3 mutation
160
Figure 2 - Calnuc knockdown reduces CLN3 and CLN5 levels and vice-versa.
(A) Western blot analysis showing reduced levels of CLN3 and CLN5 in Calnuc-depleted
cells, reduced levels of Calnuc and CLN5 in CLN3-depleted and reduced levels of Calnuc
and CLN3 in CLN5 depleted-cells. Used as a negative control, CLN1-HA level was not
altered upon the depletion of Calnuc. HeLa cells were transfected with control, Calnuc,
CLN1, CLN3, or CLN5 siRNA for 3 days and CLN1-HA, CLN3-HA or CLN5-HA cDNA
was transfected 12 h before the experiment. Proteins were separated by SDS-PAGE and
immunoblotted with anti-HA, anti-Calnuc, and with anti-EEA1 as loading controls. (B)
Steady state levels of CLN5 are irecovered in Calnuc-depleted cells rescued with siRNAresistant full-length Calnuc-GFP or cytoplasmic Calnuc-GFP (ΔSP-Calnuc-GFP) or
calcium-binding deficient Calnuc-GFP (ΔEF-Calnuc-GFP), in contrast to cells rescued with
GFP. HeLa cells treated with human Calnuc siRNA were transfected with GFP, siRNAresistant Calnuc-GFP or ΔSP-Calnuc-GFP or ΔEF-Calnuc-GFP at 5 h after knockdown.
161
The cells were lysed 3 days later, and the proteins were analyzed by western blotting. (C)
Levels of Calnuc in cerebellar culture cells (Cb) from CLN3+/+ or CbCln3Δex7/8/Δex7/8 (MT)
mice. Calnuc expression is reduced in MT compared to WT cells. (D) Lysates from WT
and MT cerebellar cells were incubated with GST or GST-RILP, which specifically
interacts with the active (GTP-bound) form of Rab7. Bound proteins were then analyzed by
western blotting with anti-Rab7.
is a 1.03 kb deletion causing exons 7 and 8 deletion and resulting in a protein harboring a
frame shift at amino acid 153, leading to a truncated and unstable CLN3 protein. Western
blot analysis of these cell lines indicated a lower level of endogenous Calnuc in
CbCln3Δex7/8/Δex7/8 cells compared to CbCln3+/+ cells (Figure 2C). These results suggest that
Calnuc, CLN3 and CLN5 could form a protein complex stabilized by their intramolecular
interactions and that the downregulation of one member of this complex induces the
downregulation of the other ones. These results strengthen our hypothesis that Calnuc,
CLN3 and CLN5 are part of a functional protein complex.
Given that Calnuc levels has been previously shown to alter Rab7 activity (Larkin et
al. 2016) and that Calnuc is reduced in CbCln3Δex7/8/Δex7/8 cells, we next compared the level
of active Rab7 (Rab7-GTP) in CbCln3Δex7/8/Δex7/8 (MT1 and MT2, Fig 2D) to CbCln3+/+
cells (WT1 and WT2, Fig 2D). We used an effector pull-down assay in which the Rab7binding domain of its effector protein RILP (Rab-interacting lysosomal protein) was used
to selectively isolate GTP-loaded Rab7 (Romero Rosales et al. 2009). Recombinant GSTRILP immobilized on glutathione beads was incubated with lysates from CbCln3 +/+ and
CbCln3Δex7/8/Δex7/8 cells, and bound (active) Rab7 proteins were analyzed by
immunoblotting (Figure 2D). GTP-loaded Rab7 was reduced CbCln3Δex7/8/Δex7/8 compared
to CbCln3+/+ cells (Figure 2D). Taken together, these results indicate that the level of
Calnuc and active Rab7 are reduced in CbCln3Δex7/8/Δex7/8 cells, suggesting that this is the
mechanism by which this genetically mutation of CLN3 found in the majority of human
patients alters the lysosomal receptor trafficking.
162
Calnuc-knockdown cells exhibit the characteristic lysosomal features of NCL.
Calnuc depletion causes the misdelivery and degradation of lysosomal receptors CIMPR and Sortilin in lysosomes (Larkin et al. 2016). A consequence of the defective
transport of lysosomal sorting receptors is the missorting of lysosomal enzymes and
consequently, the accumulation of undegraded substrates in lysosomes. In NCL, ceroid
lipofuscins accumulate in lysosomes (Goebel and Wisniewski 2004; Jalanko and Braulke
2009). The accumulation of this material induces the enlargement of lysosomes as well as
specific ultrastructural changes. Based on our observations that Calnuc interacts with and
alters the levels of CLN3 and CLN5, two proteins involved in NCL, we investigated
whether Calnuc depletion generate phenotype typical to lysosomal disease associated to
CLN3 and CLN5 mutations. The morphology of the lysosomes were first be investigated
by confocal microscopy in HeLa cells transfected with control or Calnuc siRNA. Using the
lysosomal marker LAMP2, bright and large lysosomes were observed in Calnuc-depleted
cells compares to control cells (Figure 3A), suggesting an enlargement of the lysosomes. In
concordance, immunoblot analysis indicated that LAMP2 protein levels were increased in
Calnuc-depleted cells compared to control (data not shown). To further characterize the
lysosomal compartments, we next used the greater resolving power of transmission electron
microscopy (EM) to examine the fine ultrastructural details of lysosomes in Calnucdepleted cells. We observed storage-like material that resembled curvilinear as well as
autophagic vacuoles, which are commonly find in CLN3 and CLN5 NCL subtype (Figure
3B).
Autophagy is emerging as a major pathway involved in NCL (Koike et al. 2005; Cao
et al. 2006; Thelen et al. 2012). Autophagic vacuoles accumulate in different forms of NCL
due to a defect in autophagic clearance through the lysosome (Koike et al. 2005; Cao et al.
2006; Thelen al. 2012). We therefore used LC3, considered one of the most specific
markers for autophagosomes (Mizushima 2004) to examine autophagy defects in Calnuc
depleted cells. Under normal conditions, LC3 is reported to be cytosolic; however, after
autophagy induction, LC3 is lipidated and recruited onto autophagosome (Tanida et al.
2008). Control- and Calnuc-depleted HeLa cells transfected with LC3-GFP were treated
163
Figure 3 - Calnuc depletion alters lysosomes morphology, induces curvilinear-like
pattern and activates autophagy.
(A) Comparison of the lysosomes sizes using LAMP2 marker in control versus Calnuc
depleted cells. HeLa cells were immunostained using anti-Calnuc and anti-LAMP2
antibodies. LAMP2 immunostaining showed enhanced signal intensity with less
perinuclear clustering in Calnuc delpleted cells. (B) TEM analysis of inclusions in Calnuc
164
PC12 depleted cells is shown. Curvilinear pattern and large electron-dense inclusion are
present in Calnuc depleted cells. 10,000 × magnification. (C) Level of chloroquine induced
autophagy showed by LC3 in control and Calnuc knockdown cells. Calnuc depleted cells
show increased level of LC3 compared to control cells. HeLa cells were transfected with
control or Calnuc-specific siRNA for 3 days, fixed, permeabilized and immunostained.
Confocal microscopy images were acquired using identical instrument settings. For LC3
experiment cells were also transfected with LC3-GFP cDNA using Fugene HD and were
treated with 50 μM Chloroquine to induces autophagic vacuole formation 12 h before the
experiment. Scale bar, 10 μm.
with choloroquine over night to induce autophagy and analyze by confocal microscopy. In
control siRNA treated HeLa, LC3-GFP was mainly detected in the cytoplasm and on small
puncta that do not colocalize with the lysosomal marker LAMPII (Figure 3Ca-c). In
contrast, Calnuc siRNA treated cells displayed large LC3-positive puncta (Figure 3Cd), that
partially colocalize with LAMP2 (Figure 3Cf). These results indicate an accumulation of
LC3-positive autophagosomes in Calnuc-depleted cells, in concordance with the presence
of dense inclusions bodies associated with autophagy observed by electronic microscopy
(Figure 3B).
The pathological hallmark of NCL is the accumulation of autofluorescent ceroid
lipofuscin deposits that are enriched in saposins or subunit C of the mitochondrial ATP
synthase complex (Palmer et al. 1989; Palmer et al. 1992), in the lysosomes of neurons and
other cell types. These lipofuscin deposits usually appear as small, punctate intracellular
structures that are strongly fluorescent under any excitation ranging from 360 nm to 647
nm (Fossale et al. 2004; Lojewski et al. 2013). Using confocal microscopy with excitation
wavelength of 408 nm, we therefore examined control- and Calnuc-depleted HeLa cells for
the presence of autofluorescent lipofuscin deposits. No autofluorencent material was
observed in the cytoplasm of control cells (Figure 4Aa). However, autofluorescent puncta
that colocalize with lysosomal marker LAMP2 was detected in Calnuc-depleted cells
(Figure 4Af). We next examined the cells for the presence of the pore-forming subunit C of
the mitochondrial ATP synthase complex (SCMAS), considered as a specific markers for
lipofuscin deposits. Control and Calnuc siRNA treated HeLa cells were immunostained
with antibodies against endogenous LAMP2 and SCMAS and analyzed by confocal
microscopy (Figure 4B). An accumulation of SCMAS puncta that partially colocalize with
165
LAMP2 was observed in Calnuc-depleted cells (Fig. 4Bd-f), consistent with the abnormal
accumulation of lipofuscins occurring in a lysosomal compartment.
Figure 4 - Calnuc depletion causes an accumulation of autofluorescent material and of
SCMAS in lysosomes.
166
Autofluorescence and SCMAS immunostaining is shown in HeLa cells transfected with
control or Calnuc siRNA for 3 days, fixed, permeabilized and immunostained. (A) Calnuc
knockdown cells present autofluorescent material which was not observed in control cells
and which strongly, though not perfectly, overlaps with lysosomal marker LAMP2. Cells
were immunostained using anti-LAMP2 and autofluorescence was detected at 408 nm
excitation wavelength. (B) Control cells exhibited limited SCMAS immunostaining
whereas Calnuc depleted cells contained numerous SCMAS puncta wich colocalise with
lysosomal marker LAMP2. Cells were immunostained using anti-LAMP2 and antiSCMAS. The labeled cells were examined by confocal fluorescence microscopy and
images were acquired using identical instrument settings. Scale bar, 10 μm.
Calnuc expression levels are reduced in NCL fibroblasts.
Based on our results suggesting that the depletion of Calnuc induces morphological
features reminiscent of NCL, and that Calnuc interacts and regulates CLN proteins
involved in this disease, we hypothesized that Calnuc can be involved in ceroid
lipofuscinosis pathogenesis. This hypothesis was tested by analyzing the expression level
of Calnuc expression in fibroblast cell lines from NCL patients (Figure 5). Expression
profiling of Calnuc was performed by comparing Calnuc protein and mRNA levels in
fibroblasts from four apparently healthy individual acting as controls (CTL), in one
fibroblast from patients suffering from Niemann Pick disease (NPD), a related lysosomal
storage disease, four genetically confirmed cases of NCL, including one from CLN1
patient, one from CLN3 patient (homozygous for 1.02-kb deletion, the most common
mutation), two from CLN5 patients (p.Leu358AlafsX4 and p.Thr176AsnfsX11 mutants), as
well as five orphan cases of NCL (Orphan) which remain undefined genetically, i.e., not
involving CLN mutation (Mole et al. 2011; Kousi et al. 2012). Immunoblot analysis
indicated that all patients suffering from NCL, both genetically confirmed and orphan,
expressed lower level of Calnuc protein compared to the healthy or NPD controls (Figure
5A). Quantification analysis of Calnuc levels normalized to the loading control early
endosome antigen 1 (EEA1) confirmed that Calnuc expression was approximately 50%
lower in fibroblasts from CLN1, CLN3, CLN5 and orphans patients compared to control
fibroblasts (Figure 5A). Interestingly, no decrease in Calnuc protein levels was observed in
NPD fibroblasts suggesting a Calnuc independent mechanism for the pathogenesis of this
167
Figure 5 - Calnuc levels in healthy and NCL fibroblasts.
Analysis of Calnuc protein and mRNA levels in control (CTL) and Neuronal ceroid
lipofuscinosis (NCL) fibroblasts. (A) Calnuc protein expression in fibroblasts of healthy
(CTL), Niemann pick disease type C1 (NPD) and NCL patients was compared by Western
blotting. Representative data showing lysates subjected to SDS-PAGE and immunoblotted
with antibodies directed against Calnuc and EEA1. (B) Densitometric analysis of Western
blots, such as those shown in (A), normalized to the signal of EEA1. Results are expressed
as means ± SD (n ≥ 3) compared with the mean of healthy patients. (C) Calnuc, CLN1,
CLN3 and CLN5 mRNA levels in fibroblasts of CTL, NPD and NCL patients were
compared by qRT-PCR. Total mRNA was reverse transcribed, and the levels of Calnuc,
CLN1, CLN3 and CLN5 cDNA were analyzed by qPCR with iTaq Universal SYBR Green
168
Supermix and were expressed relative to the housekeeping genes Gapdh, Lamp1 and Clk2
using the qBASE framework. Results are expressed as means ± SD (n = 5 samples, in
duplicate). The difference between CTL and NCL was not significant.
other lysosomal storage disease. To determine whether the decrease in Calnuc protein
levels was due to a change in gene expression, total quantitative RT-PCR assays were next
performed on the same fibroblasts cells lines (Figure 5B). Results were normalized using
the housekeeping genes Gapdh, Lamp1 and Clk2 and expressed relatively to the first
arbitrary establish control. No significant difference was observed between Calnuc mRNA
levels in all the NCL cells, NPD cells and control cells. However, as expected, we noted
that CLN1 fibroblasts shows highly reduced levels of CLN1 mRNA as well as CLN3
mRNA in CLN3 fibroblasts and CLN5 mRNA in one of the CLN5 fibroblast cells line. In
summary, our results indicated that Calnuc protein levels, but not mRNA levels, are lower
in fibroblasts from NCL patients.
Discussion
We previously showed that the cytosolic pool of Calnuc plays an essential role in
endosomes-to-TGN transport of the lysosomal receptors MPR and Sortilin. We reported
that, through an indirect interaction, Calnuc regulates membrane association and activation
of Rab7, a small G protein required for the endosomal recruitment of retromers and crucial
for the retrieval of lysosomal receptors to the TGN (Larkin et al. 2016). In the present
study, we extended our previous findings and provide further mechanistic insight into how
Calnuc regulates Rab7 activity. We demonstrated that Calnuc interacts with and stabilizes
CLN3 and CLN5, two proteins involved in the proper trafficking of lysosomal receptors
through the interaction with and regulation of Rab7. CLN3 and CLN5 are mutated/altered
in NCL, a rare lysosomal storage disease. Interestingly, we observed that the depletion of
Calnuc induced cellular morphological features reminiscent of NCL and that Calnuc
expression is reduced in fibroblasts from NCL patients. Overall, this study suggests that
Calnuc function is altered in human NCL and that Calnuc could be a novel protein involved
in NCL pathogenesis.
169
Our results demonstrate that Calnuc forms a complex with CLN3 and CLN5 on
endosomes. CLN3 is known to interact directly with CLN5 and our immunoprecipitation
results showed that Calnuc can interact with both CLN3 and CLN5, suggesting that they
are in the same protein complex. Using confocal microscopy, all three proteins were found
together on early endosomes microdomains. Furthermore, we showed that knockdown of
one of the protein of the complex (Calnuc, CLN3 or CLN5) induces the downregulation of
the two other partners. Similarly, we observed a reduction of Calnuc level in mouse
cerebellar precursor cell lines established from a mouse model containing a genetically
mutation of CLN3 (CbCln3Δex7/8/Δex7/8) (Fossale et al. 2004; Cao et al. 2006; Cao et al.
2011) in which CLN3 protein level was reported as reduced (Fossale et al. 2004). This codepletion phenomenon is often observed in protein complexes (e.g., retromers,
nucleoporins) (Boehmer et al. 2003; Arighi et al. 2004) and strengthens our hypothesis that
Calnuc, CLN3 and CLN5 are part of a functional protein complex. In concordance, CLN5
could be rescued by the expression of the cytoplasmic form of Calnuc (ΔSP-Calnuc-GFP),
suggesting the implication of the transmembrane CLN3. Based on these findings, we
propose a model (Figure 6) in which cytosolic Calnuc interacts on endolysosomal
membranes with the 6TM protein CLN3, which can in turn associated with the highly
glycosylated luminal protein CLN5. Once assembled, this complex would modulate the
recruitment and activation of Rab7, leading to lysosomal receptor recycling to the TGN.
Indeed, the loss of Calnuc and CLN5 led to a reduction of Rab7-GTP, a decrease in the
recruitment of retromers on endosomal membrane and the missorting of the lysosomal
receptors MPR and Sortilin (Mamo et al. 2012; Larkin et al. 2016). CLN3 has been
reported to bind directly Rab7, preferentially Rab7-GTP, and various retro and anterograde
microtubular motor complexes responsible for the movement of late endosomes and
lysosomes (Uusi-Rauva et al. 2012). However, CLN3 has never been shown to regulate
Rab7 activity. We found that the level of active Rab7 (Rab7-GTP) was reduced in
CbCln3Δex7/8/Δex7/8 cells, suggesting that an alteration of CLN3 levels also modulates Rab7
activity. Thus, it is tempting to speculate the complex Calnuc-CLN3-CLN5 is involved in
the regulation of Rab7 activity. But how is this complex activated in order to promote the
sorting of lysosomal receptors? Based on previous finding by Mamo et al showing that
CLN5 interact with Sortilin (Mamo et al. 2012), we propose that when the cargo-loaded
170
lysosomal sorting receptor arrives at the endosome, the change in pH causes a dissociation
of the cargo from the receptor that subsequently leads to interaction with CLN5. This
would provide a signal to initiate their retrograde transport to the TGN. Through direct
interaction between CLN5 and CLN3, and possibly conformational modifications of CLN3,
this signal would cross the membrane to reach Calnuc. On the cytosolic side, Calnuc would
next participate in the activation of Rab7 and, subsequently, in the recruitment of the
retromer retrograde machinery. However, the manner by which Calnuc alters the amount of
active Rab7 requires further clarification. This model is in accordance with the favored
binding of active GTP-bound form of Rab7 with the N-terminal domain of CLN3 and on
the reported shorter residence of Rab7 on endolysosomal membranes devoided of CLN3
protein. This unbalanced GTP/GDP cycle of Rab7 would be caused by the CLN5-CLN3
signal unable to reach Calnuc.
Figure 6 - Proposed schematic model for Calnuc-CLN3-CLN5 complex influencing
receptors retrograde transport.
CLN5 is tightly associated with the lumen of the endosomal membrane. After cargo
dissociation from the lysosomal sorting receptor, CLN5 binds to it and subsequently to
CLN3, a transmembrane protein. This CLN5-CLN3 interaction would acts as a signal to
initiate the action of Calnuc on the cytosolic side of the endosome, the activation of Rab7
leading to the recruitment of retromer and the retrograde transport of the lysosomal receptor
to the TGN. The figure is not to scale.
171
One question that remains is how the pathological mutations identified in CLN5 and
CLN3 affect the sorting machinery leading to lysosomal disorders. Some CLN5 mutants
such as FinM, SWE and EUR maintain their physical abilities to interact with CLN3 (Lyly
et al. 2009). However, in some cases, they are unstable and rapidly degraded (SWE and
NFL) or mislocated to the endoplasmic reticulum (EUR, FinM, SWE, and NFL) (Schmiedt
et al. 2010; Larkin et al. 2013; Moharir et al. 2013) Thus, it is possible that interactions
with lysosomal receptors may be compromised in some CLN5 mutants. Similarly,
mutations in CLN3 can lead to their incapacity to interact with Calnuc or Rab7 proteins.
For instance, CbCln3Δex7/8/Δex7/8 mutation results in a truncated protein that is retained in the
ER where it cannot interact with endolysosomal proteins. To further understand the
underlying mechanism by which Calnuc-CLN3-CLN5 complex regulates lysosome
receptor trafficking and its potential implication in NCL pathogenesis, the specific regions
of interaction of these proteins need to be characterized as well as how NCL-associated
mutations in CLN5 and CLN3 affect the complex.
Based on our observations that Calnuc form a complex with CLN3 and CLN5, two
proteins involved in NCL, we next investigated whether Calnuc depletion generate
phenotype typical to this lysosomal storage disease. Using confocal and electron
microscopy, we showed that loss of Calnuc induces cellular morphological features
reminiscent of NCL, such as lysosome enlargement, accumulation of autofluorescent
material and of subunit C of mitochondrial ATP synthase (SCMAS), as well as the
presence of storage-like material that resembled curvilinear deposits and an increase of
autophagy vacuoles. To this day, these structure and content of storage material are still
used as diagnostic clues to help define the lysosomal defective function in NCL and
suggest that Calnuc deficiency affect the endosomal/lysosomal system and could cause
lysosomal storage disease. The cellular changes observed upon Calnuc depletion are also
highly consistent with what has been described in animal models and in patients with
mutations in CLN3 and CLN5 genes, suggesting the Calnuc, CLN3 and CLN5 cooperate in
a shared pathway or complex. Indeed, CLN3 and CLN5 mutations are typically associated
with fingerprint, curvilinear or rectilinear lysosomal ultrastructures (Santavuori 1988; Mole
et al. 2005; Pineda-Trujillo et al. 2005), combined with SCMAS as the main component of
172
the storage material (Palmer et al. 1992; Tyynela et al. 1997; Tyynela et al. 1997). In
contrast, CLN1 mutation is associated with the accumulation of sphingolipid-activator
proteins A and D storage material and granular osmiophilic deposits (Carpenter et al. 1973;
Das, Becerra et al. 1998; Wisniewski et al. 1998; Santavuori et al. 2000; Gupta et al. 2001;
Van Diggelen et al. 2001; Ramadan et al. 2007), suggesting its implication in a different
complex/pathway than CLN3 and CLN5. Disrupted autophagic clearance through to the
lysosome is thought to be particularly damaging for vulnerable neuron cells and is rightly
associated with neurodegenerative disorders like NCL and Alzheimer's disease.
Interestingly, recent evidences lead toward lysosomal calcium signalling as a major
regulator of lysosome biogenesis and autophagy through calcineurin-mediated induction of
TFEB (Medina et al. 2015). As Calnuc possesses the ability to bind calcium, it may
respond to such local calcium increase to act in this pathway.
The observation that Calnuc forms a complex with NCL proteins and that loss of
Calnuc induces cellular morphological features reminiscent of NCL suggests a potential
role of Calnuc in ceroid lipofuscinosis pathogenesis. Interestingly, the analysis of Calnuc
expression profile in human fibroblasts from genetically-confirmed cases of NCL and
orphan NCL (NCL phenotypes but undefined genes) indicated a lower level of Calnuc
protein in CLN3, CLN5 as well as all the orphan NCL fibroblasts tested. These results are
in accordance with our data showing a decrease of Calnuc protein level in CLN3 and CLN5
siRNA depleted Hela cells. It is noteworthy that Calnuc is not affected in fibroblasts from
patients suffering from Niemann-Pick disease (NPD) patients, a related lysosomal storage
disease, suggesting the specific involvement of Calnuc in NCL associated lysosomal
storage disease. On the other hand, the reduction of Calnuc protein level in fibroblasts from
CLN1 patient may indicate its implication with the Calnuc-CLN3-CLN5 complex. CLN1 is
an enzyme which removes fatty acids from cysteine residues in lipid-modified proteins
(Camp et al. 1994). CLN1 colocalizes and interacts with CLN5 (Lyly et al. 2009) and has
been proposed to facilitate its lysosomal transport (Schmiedt et al.). Therefore, CLN1
activity may impact the complex formation and Calnuc stability. Although these
preliminary data suggest a link between Calnuc and NCL, the interpretation is limited by
the low amount of samples analyzed due to the limited availability of the samples.
173
Further analysis of Calnuc expression profile using qRT-PCR analysis ruled out the
possibility that the decrease in Calnuc protein level was caused by an alteration of its
mRNA levels. No significant change in Calnuc mRNA was detected in all the analyzed
fibroblasts from LSD patients. However, mutations in Calnuc could be causative of its
defective function without necessarily influencing its level of expression. Indeed, various
mutations that affect the CLN genes lead to hasten mRNA degradation, result in truncated
proteins that are nonfunctional or less stable, alter interaction with partners or cause their
mislocalization (Kousi et al. 2012). For example, in fibrobasts from CLN5
(p.Leu358AlafsX4) patient, the mRNA level of CLN5 was not altered whereas in fibrobasts
from CLN5 (p.Thr176AsnfsX11) patient, in which a STOP codon is inserted at aa 176 due
to a frameshift, the mRNA level is reduced, probably due to the instability of the transcript.
Our data also confirmed previous work reporting a lower level of CLN3 mRNA in CLN3
mutant fibroblasts. CLN1 mRNA was previously reported to be upregulated in a mouse
model of CLN5 FinM mutation (Lyly et al. 2009). No such transcriptional behavior related
to a mutation was observed. However, we noted that the mRNA levels of Calnuc, CLN3
and CLN5 increase together in some orphan cell lines without being reflected as an increase
in Calnuc protein levels. Given that genetically undefined forms of NCL have been
reported, it would be interesting to determine whether Calnuc is mutated in the orphan
cases of NCL, potentially identifying Calnuc as a new gene involved in NCLs. On the other
hand, the direct involvement of Calnuc in genetically-confirmed cases of CLN would
redefine the paradigm of NCL classification: it would not solely be based on the CLN gene
mutated but also on the expression of disease-modifier genes. These genes, such as Calnuc,
could alter the expression of CLN proteins and aggravate the progression of the disease.
These modifier genes could explain the high phenotype variability observed in CLN
patients with an identical CLN genotype.
In summary, herein, we have showed the first evidence for a role of Calnuc in
neuronal ceroid lipofuscinosis through novel interaction with CLN3 and CLN5. The
opportunity to identify new players in the disease provides new insights into the
understanding of the cellular mechanism and physiopathology. Above all, it offers the
possibility to develop new diagnostic tools and best chance of developing treatments.
174
Acknowledgments
We are grateful to Susan Cotman (Harvard Medical School) and Sara Mole
(University College London) for cells and antibodies link to NCL pathology. H.L. holds a
fellowship from the National Sciences and Engineering Research Council of Canada
(NSERC). This work was supported by grants from the Canadian Institutes for Health
Research and a Canada Research Chair to C.L.L. The authors declare that they have no
conflict of interest.
175
References
Arighi, C. N., L. M. Hartnell, et al. (2004). "Role of the mammalian retromer in sorting of
the cation-independent mannose 6-phosphate receptor." J Cell Biol 165(1): 123-133.
Ballif, B. A., N. V. Mincek, et al. (1996). "Interaction of cyclooxygenases with an
apoptosis- and autoimmunity-associated protein." Proc Natl Acad Sci U S A 93(11):
5544-5549.
Bessa, C., C. A. Teixeira, et al. (2006). "Two novel CLN5 mutations in a Portuguese
patient with vLINCL: insights into molecular mechanisms of CLN5 deficiency."
Mol Genet Metab 89(3): 245-253.
Boehmer, T., J. Enninga, et al. (2003). "Depletion of a single nucleoporin, Nup107,
prevents the assembly of a subset of nucleoporins into the nuclear pore complex."
Proc Natl Acad Sci U S A 100(3): 981-985.
Boucher, R., H. Larkin, et al. (2008). "Intracellular trafficking of LRP9 is dependent on two
acidic cluster/dileucine motifs." Histochem Cell Biol 130(2): 315-327.
Brodeur, J., H. Larkin, et al. (2009). "Calnuc binds to LRP9 and affects its endosomal
sorting." Traffic 10(8): 1098-1114.
Brosseau, J. P., J. F. Lucier, et al. (2010). "High-throughput quantification of splicing
isoforms." Rna 16(2): 442-449.
Camp, L. A., L. A. Verkruyse, et al. (1994). "Molecular cloning and expression of
palmitoyl-protein thioesterase." J Biol Chem 269(37): 23212-23219.
Cao, Y., J. A. Espinola, et al. (2006). "Autophagy is disrupted in a knock-in mouse model
of juvenile neuronal ceroid lipofuscinosis." J Biol Chem 281(29): 20483-20493.
Cao, Y., J. F. Staropoli, et al. (2011). "Distinct early molecular responses to mutations
causing vLINCL and JNCL presage ATP synthase subunit C accumulation in
cerebellar cells." PLoS One 6(2): e17118.
Carpenter, S., G. Karpati, et al. (1973). "A type of juvenile cerebromacular degeneration
characterized by granular osmiophilic deposits." J Neurol Sci 18(1): 67-87.
Cotman, S. L. and J. F. Staropoli (2012). "The juvenile Batten disease protein, CLN3, and
its role in regulating anterograde and retrograde post-Golgi trafficking." Clin
Lipidol 7(1): 79-91.
Das, A. K., C. H. Becerra, et al. (1998). "Molecular genetics of palmitoyl-protein
thioesterase deficiency in the U.S." J Clin Invest 102(2): 361-370.
Fossale, E., P. Wolf, et al. (2004). "Membrane trafficking and mitochondrial abnormalities
precede subunit C deposition in a cerebellar cell model of juvenile neuronal ceroid
lipofuscinosis." BMC Neurosci 5: 57.
Goebel, H. H. and K. E. Wisniewski (2004). "Current state of clinical and morphological
features in human NCL." Brain Pathol 14(1): 61-69.
Gupta, P., A. A. Soyombo, et al. (2001). "Disruption of PPT1 or PPT2 causes neuronal
ceroid lipofuscinosis in knockout mice." Proc Natl Acad Sci U S A 98(24): 1356613571.
Hellemans, J., G. Mortier, et al. (2007). "qBase relative quantification framework and
software for management and automated analysis of real-time quantitative PCR
data." Genome Biol 8(2): R19.
176
Isosomppi, J., J. Vesa, et al. (2002). "Lysosomal localization of the neuronal ceroid
lipofuscinosis CLN5 protein." Hum Mol Genet 11(8): 885-891.
Jalanko, A. and T. Braulke (2009). "Neuronal ceroid lipofuscinoses." Biochim Biophys
Acta 1793(4): 697-709.
Jarvela, I., M. Lehtovirta, et al. (1999). "Defective intracellular transport of CLN3 is the
molecular basis of Batten disease (JNCL)." Hum Mol Genet 8(6): 1091-1098.
Kama, R., V. Kanneganti, et al. (2011). "The yeast Batten disease orthologue Btn1 controls
endosome-Golgi retrograde transport via SNARE assembly." J Cell Biol 195(2):
203-215.
Kama, R., M. Robinson, et al. (2007). "Btn2, a Hook1 ortholog and potential Batten
disease-related protein, mediates late endosome-Golgi protein sorting in yeast." Mol
Cell Biol 27(2): 605-621.
Kawano, J., T. Kotani, et al. (2000). "CALNUC (nucleobindin) is localized in the Golgi
apparatus in insect cells." Eur J Cell Biol 79(3): 208-217.
Koike, M., M. Shibata, et al. (2005). "Participation of autophagy in storage of lysosomes in
neurons from mouse models of neuronal ceroid-lipofuscinoses (Batten disease)."
Am J Pathol 167(6): 1713-1728.
Kousi, M., A. E. Lehesjoki, et al. (2012). "Update of the mutation spectrum and clinical
correlations of over 360 mutations in eight genes that underlie the neuronal ceroid
lipofuscinoses." Hum Mutat 33(1): 42-63.
Larkin, H., Costantino, S and Lavoie, C (2016). "Calnuc function in endosomal sorting of
lysosomal receptors." Traffic. 2016 Jan 12. doi:10.1111/ tra.12374. [Epub ahead of
print]
Larkin, H., M. G. Ribeiro, et al. (2013). "Topology and membrane anchoring of the
lysosomal storage disease-related protein CLN5." Hum Mutat 34(12): 1688-1697.
Larkin, H., C. Thériault, et al. (2013). "Topology and membrane anchoring of the
lysosomal storage disease-related membrane protein CLN5." Human mutation
Submitted.
Lavoie, C., T. Meerloo, et al. (2002). "Calnuc, an EF-hand Ca(2+)-binding protein, is
stored and processed in the Golgi and secreted by the constitutive-like pathway in
AtT20 cells." Mol Endocrinol 16(11): 2462-2474.
Lebrun, A. H., S. Storch, et al. (2009). "Retention of lysosomal protein CLN5 in the
endoplasmic reticulum causes neuronal ceroid lipofuscinosis in Asian sibship."
Hum Mutat 30(5): E651-661.
Lefrancois, S., J. Zeng, et al. (2003). "The lysosomal trafficking of sphingolipid activator
proteins (SAPs) is mediated by sortilin." EMBO J 22(24): 6430-6437.
Lin, P., T. Fischer, et al. (2009). "Calnuc plays a role in dynamic distribution of Galphai but
not Gbeta subunits and modulates ACTH secretion in AtT-20 neuroendocrine
secretory cells." Mol Neurodegener 4: 15.
Lin, P., H. Le-Niculescu, et al. (1998). "The mammalian calcium-binding protein,
nucleobindin (CALNUC), is a Golgi resident protein." J Cell Biol 141(7): 15151527.
Lin, P., Y. Yao, et al. (1999). "Overexpression of CALNUC (nucleobindin) increases
agonist and thapsigargin releasable Ca2+ storage in the Golgi." J Cell Biol 145(2):
279-289.
177
Lojewski, X., J. F. Staropoli, et al. (2013). "Human iPSC models of neuronal ceroid
lipofuscinosis capture distinct effects of TPP1 and CLN3 mutations on the
endocytic pathway." Hum Mol Genet.
Lyly, A., C. von Schantz, et al. (2009). "Novel interactions of CLN5 support molecular
networking between Neuronal Ceroid Lipofuscinosis proteins." BMC Cell Biol 10:
83.
Mamo, A., F. Jules, et al. (2012). "The role of ceroid lipofuscinosis neuronal protein 5
(CLN5) in endosomal sorting." Mol Cell Biol 32(10): 1855-1866.
Medina, D. L., S. Di Paola, et al. (2015). "Lysosomal calcium signalling regulates
autophagy through calcineurin and TFEB." Nat Cell Biol 17(3): 288-299.
Metcalf, D. J., A. A. Calvi, et al. (2008). "Loss of the Batten disease gene CLN3 prevents
exit from the TGN of the mannose 6-phosphate receptor." Traffic 9(11): 1905-1914.
Miura, K., M. Hirai, et al. (1996). "Organization of the human gene for nucleobindin
(NUC) and its chromosomal assignment to 19q13.2-q13.4." Genomics 34(2): 181186.
Miura, K., K. Titani, et al. (1992). "Molecular cloning of nucleobindin, a novel DNAbinding protein that contains both a signal peptide and a leucine zipper structure."
Biochem Biophys Res Commun 187(1): 375-380.
Mizushima, N. (2004). "Methods for monitoring autophagy." Int J Biochem Cell Biol
36(12): 2491-2502.
Mole, S. E., R. E. Williams, et al. (2005). "Correlations between genotype, ultrastructural
morphology and clinical phenotype in the neuronal ceroid lipofuscinoses."
Neurogenetics 6(3): 107-126.
Mole, S. E., R. E. Williams, et al. (2011). The neuronal Ceroid Lipofuscinoses (Batten
Disease), Oxford University Press.
Palmer, D. N., I. M. Fearnley, et al. (1989). "Lysosomal storage of the DCCD reactive
proteolipid subunit of mitochondrial ATP synthase in human and ovine ceroid
lipofuscinoses." Adv Exp Med Biol 266: 211-222; discussion 223.
Palmer, D. N., I. M. Fearnley, et al. (1992). "Mitochondrial ATP synthase subunit C storage
in the ceroid-lipofuscinoses (Batten disease)." Am J Med Genet 42(4): 561-567.
Pineda-Trujillo, N., W. Cornejo, et al. (2005). "A CLN5 mutation causing an atypical
neuronal ceroid lipofuscinosis of juvenile onset." Neurology 64(4): 740-742.
Ramadan, H., A. S. Al-Din, et al. (2007). "Adult neuronal ceroid lipofuscinosis caused by
deficiency in palmitoyl protein thioesterase 1." Neurology 68(5): 387-388.
Romero Rosales, K., E. R. Peralta, et al. (2009). "Rab7 activation by growth factor
withdrawal contributes to the induction of apoptosis." Mol Biol Cell 20(12): 28312840.
Santavuori, P. (1988). "Neuronal ceroid-lipofuscinoses in childhood." Brain Dev 10(2): 8083.
Santavuori, P., L. Lauronen, et al. (2000). "Neuronal ceroid lipofuscinoses in childhood."
Neurol Sci 21(3 Suppl): S35-41.
Schmiedt, M. L., C. Bessa, et al. "The neuronal ceroid lipofuscinosis protein CLN5: new
insights into cellular maturation, transport, and consequences of mutations." Hum
Mutat 31(3): 356-365.
178
Schmiedt, M. L., C. Bessa, et al. (2010). "The neuronal ceroid lipofuscinosis protein CLN5:
new insights into cellular maturation, transport, and consequences of mutations."
Hum Mutat 31(3): 356-365.
Stenmark, H., R. G. Parton, et al. (1994). "Inhibition of rab5 GTPase activity stimulates
membrane fusion in endocytosis." Embo J 13(6): 1287-1296.
Tanida, I., T. Ueno, et al. (2008). "LC3 and Autophagy." Methods Mol Biol 445: 77-88.
Taniguchi, N., H. Taniura, et al. (2000). "The postmitotic growth suppressor necdin
interacts with a calcium-binding protein (NEFA) in neuronal cytoplasm." J Biol
Chem 275(41): 31674-31681.
Thelen, M., M. Damme, et al. (2012). "Correction: Disruption of the Autophagy-Lysosome
Pathway Is Involved in Neuropathology of the nclf Mouse Model of Neuronal
Ceroid Lipofuscinosis." PLoS One 7(5).
Tyynela, J., J. Suopanki, et al. (1997). "Sphingolipid activator proteins (SAPs) in neuronal
ceroid lipofuscinoses (NCL)." Neuropediatrics 28(1): 49-52.
Tyynela, J., J. Suopanki, et al. (1997). "Variant late infantile neuronal ceroid-lipofuscinosis:
pathology and biochemistry." J Neuropathol Exp Neurol 56(4): 369-375.
Uusi-Rauva, K., A. Kyttala, et al. (2012). "Neuronal ceroid lipofuscinosis protein CLN3
interacts with motor proteins and modifies location of late endosomal
compartments." Cell Mol Life Sci.
Van Diggelen, O. P., J. L. Keulemans, et al. (2001). "Pre- and postnatal enzyme analysis
for infantile, late infantile and adult neuronal ceroid lipofuscinosis (CLN1 and
CLN2)." Eur J Paediatr Neurol 5 Suppl A: 189-192.
Vesa, J., M. H. Chin, et al. (2002). "Neuronal ceroid lipofuscinoses are connected at
molecular level: interaction of CLN5 protein with CLN2 and CLN3." Mol Biol Cell
13(7): 2410-2420.
Weiss, T. S., C. E. Chamberlain, et al. (2001). "Galpha i3 binding to calnuc on Golgi
membranes in living cells monitored by fluorescence resonance energy transfer of
green fluorescent protein fusion proteins." Proc Natl Acad Sci U S A 98(26):
14961-14966.
Wendel, M., Y. Sommarin, et al. (1995). "Isolation, characterization, and primary structure
of a calcium-binding 63-kDa bone protein." J Biol Chem 270(11): 6125-6133.
Wimley, W. C. and S. H. White (1996). "Experimentally determined hydrophobicity scale
for proteins at membrane interfaces." Nat Struct Biol 3(10): 842-848.
Wisniewski, K. E., F. Connell, et al. (1998). "Palmitoyl-protein thioesterase deficiency in a
novel granular variant of LINCL." Pediatr Neurol 18(2): 119-123.
179
Supplementary figures
Table S1 - Primer sequences for qRT-PCR
180
DISCUSSION
Mise en contexte - Calnuc lie LRP9 et influence son triage endosomial
La présente thèse a pour but de déterminer la fonction de la protéine Calnuc dans le
transport entre le TGN et les endosomes. L'étude fait suite à un premier article dans lequel
notre groupe a montré que Calnuc joue un rôle dans le transport rétrograde de LRP9, un
membre peu caractérisé de la famille des récepteurs au LDL, qui cycle entre le TGN et les
endosomes (Sardiello et al., 2009). Ce dernier est la première protéine transmembranaire à
être identifiée comme partenaire d'interaction direct pour Calnuc. En effet, la partie Nterminale de Calnuc interagit avec une région riche en arginine de la queue cytosolique de
LRP9. Ceci va de pair avec la colocalisation partielle de LRP9 avec la forme cytosolique de
Calnuc au niveau de la surface du TGN et des endosomes précoces. Toutefois, la région
riche en arginines semble insuffisante pour permettre l'interaction indiquant que d'autres
déterminants structuraux pourraient être impliqués. Pour déterminer l'impact de Calnuc sur
la distribution subcellulaire et les niveaux de LRP9, nous avons procédé à la déplétion de
Calnuc par ARN interférent. Il en résulte une redistribution de LRP9 vers les lysosomes où
il est dégradé. Ce phénomène est démontré par le traitement avec des inhibiteurs d'enzymes
lysosomiales qui préviennent la diminution des niveaux des récepteurs. L'expression du
mutant cytosolique de Calnuc contrecarre aussi l'effet ce qui indique que c'est bien cette
fraction qui est en cause dans le triage de LRP9. Cette première étude tend donc à
démontrer que Calnuc pourrait jouer un rôle dans le recyclage de LRP9 vers le TGN de
manière à prévenir sa dégradation. Toutefois, de plus amples travaux étaient nécessaires
afin de déterminer le mécanisme précis par lequel Calnuc régule le transport rétrograde de
LRP9 et potentiellement d'autres récepteurs. En effet, Calnuc interagit également avec
LRP3, qui possède une région riche en arginines, et influence son transport de la même
manière que pour LRP9. À l'opposé, aucune interaction et aucun effet n'est observé avec
LRP1 qui ne contient pas de région riche en arginines. Ces données tendent à démontrer
que Calnuc aurait un effet sur certains récepteurs spécifiques plutôt qu'un effet général.
181
Article 1 - Rôle de Calnuc dans le triage endosomial des récepteurs lysosomiaux
Le premier article qui fait l'objet du présent ouvrage visait donc à déterminer si
Calnuc joue un rôle dans le transport rétrograde d'autres récepteurs et à mettre en lumière le
mécanisme moléculaire qui sous-tend ce rôle. L'étude s'est orientée vers CI-MPR et
Sortiline, deux récepteurs bien caractérisés qui transitent entre le TGN et les endosomes et
qui permettent l'acheminement d'enzymes solubles vers le lysosome. Nous avons montré
que Calnuc interagit avec la queue cytosolique de Sortiline et de CI-MPR et colocalise avec
ces deux récepteurs à la surface des endosomes. À l'image de l'effet observé sur LRP9 et
LRP3, la déplétion de Calnuc cause une redistribution de CI-MPR et Sortiline vers les
lysosomes où ils sont dégradés entraînant une diminution des niveaux intracellulaires des
récepteurs. Cet effet peut être prévenu par un traitement avec des inhibiteurs d'enzymes
lysosomiales et peut être renversé par la surexpression du mutant cytosolique de Calnuc
indiquant, encore une fois, que cette fraction est en cause. De surcroît, la diminution de CIMPR, associées à l'absence de Calnuc, limite le transport de son cargo CTSD à la sortie du
TGN. La maturation intracellulaire de CTSD est alors limitée et il s'en suit une
augmentation de la sécrétion de la forme mature dans le milieu extracellulaire. Ces résultats
indiquent que Calnuc est impliquée dans le transport rétrograde des récepteurs lysosomiaux
CI-MPR et Sortiline et, indirectement, dans la livraison subséquente de leurs cargos dans la
voie endosomiale. Nos études montrent que Calnuc interagit avec de nombreux récepteurs
qui cyclent entre le Golgi et les endosomes, dont CI-MPR, CD-MPR (figure 9E), Sortiline,
LRP3 et LRP9, de façon à prévenir leur acheminement aux lysosomes. Des résultats
préliminaires obtenus par notre groupe suggèrent que la fonction de Calnuc pourrait
s'étendre à des récepteurs internalisés à la membrane plasmique, dont les récepteurs de type
tyrosine-kinase Tropomyosin receptor kinase (Trk)A (figure 9D), TrkB, TrkC, de même
que la protéase à sérine Furine (figure 9A), aussi connue sur le nom de PACE (Paired basic
amino acid cleaving enzyme). Par contre, aucune interaction n'a été décelée avec LRP1,
Neurotrophin receptor (p75/NTR) (figure 9B) et le Epidermal growth factor receptor
(EGFR) (figure 9C). Initialement, un segment amphiphile riche en arginines de la queue
cytosolique de LRP9 a été identifié comme étant important, mais non essentiel pour
l'interaction avec Calnuc, laissant croire que d'autres déterminants structuraux pourraient
182
être en cause. En effet, une brève étude structurale a montré que ce segment n'est pas un
élément commun des queues cytoplasmiques de l'ensemble des récepteurs interagissant
avec Calnuc. De plus, contrairement à LRP9, certains récepteurs, dont CI-MPR et Sortiline,
interagissent indirectement avec Calnuc. Une protéine intermédiaire pourrait alors être
impliquée complexifiant l'identification d'un motif d'interaction. Nos données préliminaires
indiquent également que la déplétion de Calnuc entraîne une diminution des niveaux de
Furine, CD-MPR et EGFR en plus de CI-MPR (figure 10). De plus amples études seront
nécessaires afin de clarifier comment Calnuc affecte ces récepteurs.
Figure 9 - Calnuc interagit avec Furine, TrkA et CD-MPR, mais pas avec p75/NTR ou
EGFR.
Des cellules HEK ont été transfectées avec GFP ou Calnuc-GFP avec (A) Furine-Flag, (B)
p75/NTR-HA, (C) EGFR ou avec HA ou Calnuc-HA avec (D) TrkA et (E) CD-MPR-GFP.
Les cellules ont été lysées et les protéines immunoprécipitées et détectées avec les anticorps
indiqués.
La principale machinerie impliquée dans le recyclage des récepteurs vers le TGN est
le complexe nommé Rétromère. La déplétion des sous-unités Vps26 (Brodeur et al., 2009),
Vps35 (Lin et al., 2007) ou SNX1 (Brodeur et al., 2012) de cet hétéropentamère produit des
effets sur divers récepteurs qui sont similaires à ceux observés à la suite de la déplétion de
Calnuc. Nous avons alors émis l'hypothèse que Calnuc serait impliquée dans le triage
183
endosomiale médié par les rétromères. La déplétion de Calnuc a montré, par l'exemple de
Vps26 et SNX2, que la protéine est nécessaire au recrutement du complexe à la surface des
endosomes sans perturber les niveaux intracellulaires des sous-unités testées, soit Vps26,
Vps35, SNX1 et SNX2. Toutefois, aucune interaction n'a été détectée entre Calnuc et les
différentes sous-unités du complexe Rétromère indiquant que Calnuc n'agit pas comme
adaptateur pour leur recrutement membranaire (données non montrées).
Figure 10 - La déplétion de Calnuc influence les niveaux de CI-MPR, Furine, CDMPR et EGFR.
Les niveaux de CI-MPR, Furine, CD-MPR et EGFR sont réduits dans les cellules déplétées
en Calnuc. Les cellules HeLa ont été transfectées avec un siRNA contrôle ou dirigé contre
Calnuc pendant 3 jours. Les cellules ont aussi été transfectées avec Furine-Flag, CD-MPRGFP ou EGFR 12h avant l'expérience. Les cellules ont été lysées et les protéines séparées
sur gel SDS-PAGE avant d'être détectées avec les anticorps anti-CI-MPR, anti-Flag, antiGFP, anti-EGFR, anti-Calnuc et anti-actine. L'actine sert de contrôle de quantité.
L'initiation et la régulation de l'attachement du complexe Rétromère à la surface
cytosolique de l'endosome dépendent de l'action séquentielle de Rab5 et de Rab7. Nous
avons alors examiné ces deux petites protéines G en tant que potentiels partenaires
d'interaction pour Calnuc. Nos études de microscopie confocale révèlent que Calnuc
cytosolique colocalise partiellement avec Rab5, Rab7 et Rab9 au niveau des endosomes.
Toutefois, en condition de surexpression, Calnuc interagit seulement avec Rab7 et de façon
indépendante de son état d'activation (lié au GDP ou au GTP). Afin de déterminer l'impact
184
Figure 11 - Rôles potentiels de Calnuc dans le cycle d'activation des Rab.
(A) Calnuc pourrait influencer positivement une GEF de manière à favoriser l'activation de
Rab7. Calnuc pourrait également influencer négativement ou (B) séquestrer une GAP de
façon à réduire l'inactivation de Rab7. + signifie influencer positivement et - signifie
influencer négativement. Figure modifiée de RAB11-mediated trafficking in host–pathogen
interactions. Guichard et al. Nat Rev Microbiol. 2014 Sep;12(9):624-34 avec la permission
de Nature Publishing Group ( Macmillan Publishers Ltd).
de Calnuc sur la localisation et l'activité de Rab7 nous avons procédé à la déplétion de
Calnuc. Dans des cellules transfectées de façon stable, il s'en suit une diminution d'environ
28% du signal correspondant à Rab7-GFP alors que les niveaux totaux de la protéine
restent inchangés. Ces résultats sont appuyés par une expérience de fractionnement
cellulaire qui montre un déplacement de Rab7, et subséquemment de Vps26 et SNX2, de la
fraction membranaire vers la fraction cytosolique. Ce défaut de recrutement est
vraisemblablement causé par une réduction de l'activation de Rab7. En effet, la quantité de
Rab7-GTP liant l'effecteur RILP est diminuée d'environ 66% en absence de Calnuc lors de
réactions de liaison par affinité. Calnuc serait donc nécessaire à l'activation de Rab7, qui
permet son recrutement aux endosomes et conséquemment l'assemblage du complexe
Rétromère ainsi que le recyclage de nombreux récepteurs vers le TGN. Toutefois, Calnuc
ne peut pas agir comme activatrice de Rab7, c'est-à-dire comme GEF, étant donné l'absence
d'interaction directe entre les deux protéines. L'action de Calnuc pourrait alors s'expliquer
de deux façons (figure 11) (Guichard et al., 2014). Calnuc pourrait influencer positivement
une GEF de Rab7, comme Mon1-Ccz1 (Small et al., 2005). En l'absence de Calnuc,
l'échange par la GEF du GDP pour le GTP serait moindre limitant l'activation de Rab7. À
l'opposé, Calnuc pourrait séquestrer ou réduire l'activité d'une GAP de Rab7, comme
185
TBC1D5 (Seaman et al., 2009). La déplétion de Calnuc favoriserait alors l'inactivation
précoce et la dissociation membranaire de Rab7.
Calnuc n'est pas la première protéine liant le calcium à influencer le transport de
récepteurs. Calmodulin, une autre protéine à mains EF liant le calcium, interagit de manière
dépendante au calcium avec la queue cytoplasmique d'EGFR (Li and Villalobo, 2002) afin
de réguler son transport (Llado et al., 2008), son activation et sa signalisation (Tebar et al.,
2002a; Tebar et al., 2002b). D'ailleurs, malgré une absence d'interaction, des données
préliminaires de notre groupe montrent que Calnuc jouerait un rôle dans le triage
intracellulaire d'EGFR en modulant la protéine GGA3. EGFR est un récepteur à activité
tyrosine kinase qui, suivant la liaison de son ligand, homodimérise pour permettre son
autophosphorylation et sa mono-ubiquitinylation au niveau de sa queue cytoplasmique.
Cette phosphorylation permet l'activation de plusieurs voies de signalisation contrôlant,
entre autres, la croissance cellulaire, la différenciation, l'apoptose et l'expression génique.
Une fois le récepteur internalisé, l'ubiquitine assure son acheminement vers les endosomes
tardifs puis les lysosomes. GGA3 reconnaît et concentre les cargos ubiquitilylés (Staub and
Rotin, 2006). Il a été montré que Calnuc interagit avec GGA3 de manière accrue en
l'absence de calcium. De plus, à l'image des résultats obtenus dans la présente étude, la
déplétion de Calnuc entraîne la redistribution d'EGFR vers les lysosomes où il est dégradé.
Le mécanisme d'action proposé pour cet effet de Calnuc diverge cependant de celui montré
pour CI-MPR et Sortiline. Au niveau du cytosol, Calnuc séquestrerait GGA3 pour
l'empêcher d’interagir avec EGFR. Une fois le récepteur activité, la relâche calcique
intracellulaire engendrée libèrerait GGA3 qui participerait alors à l’acheminement d'EGFR
aux endosomes tardifs. Éventuellement, l'abaissement du niveau de calcium, permettrait à
GGA3 d'être capturée de nouveau par Calnuc empêchant le récepteur de quitter les
endosomes précoces. Calnuc régulerait donc la distribution intracellulaire de GGA3 et, en
son absence, ce dernier serait constamment recruté aux endosomes pour acheminer des
récepteurs aux lysosomes. Il est à noter que ce mécanisme influençant EGFR accorde une
grande importance à l'impact du calcium, alors que, jusqu'à présent, l'ion ne semble pas
affecter le mécanisme influençant CI-MPR et Sortiline. En ce sens, après la déplétion de
Calnuc endogène, l'expression d'un mutant de Calnuc ne pouvant pas lier le calcium (ΔEF186
Calnuc) permet de restaurer les niveaux de CI-MPR au même titre que la forme sauvage
(résultat non montré).
Article 2 - Topologie et ancrage membranaire de CLN5, une protéine associée à une
maladie du lysosome
À l'image de Calnuc, CLN5 a récemment été identifiée comme étant un régulateur du
recrutement endosomial de Rab7. Il a été démontré que l'absence de CLN5 entraîne une
diminution de l'activation de Rab7 qui limite le recrutement du complexe Rétromère aux
endosomes menant à un défaut de recyclage des récepteurs CI-MPR et Sortiline qui sont
alors dégradés aux lysosomes (Mamo et al., 2012). N'ayant jusqu'à présent identifié aucun
partenaire d'interaction direct soutenant le rôle de Calnuc dans le transport rétrograde, nous
avons voulu savoir si Calnuc pourrait agir par l'intermédiaire de CLN5. Cette dernière est
principalement connue pour son implication dans les NCL quoique son rôle exact dans la
pathogenèse et l'impact mécanistique de ses mutants demeurent nébuleux. De plus, la
topologie de CLN5 fait l'objet d'études contradictoires, la protéine ayant été décrite comme
possédant un ou deux domaines transmembranaires ou encore comme étant soluble et
sécrétée. La simple localisation de CLN5 et de ses différents mutants pathologiques a été
compliquée par l'usage d'anticorps variés et d'étiquettes moléculaires localisées à différents
endroits sur la protéine. Dans le but de mieux comprendre la topologie, l'impact des
mutations et le mécanisme d'action de CLN5, nous avons décidé d'étudier d'abord ses
propriétés de bases, à savoir son clivage, sa glycosylation et sa localisation intracellulaire.
Nous avons élaboré différentes constructions présentant des étiquettes localisées en Nterminal, C-terminal et après le site prédit de clivage du SP. Nous avons ainsi confirmé
l'expression d'un précurseur hautement glycosylé de 73 kDa qui subit un clivage,
vraisemblablement en position 96, de manière à produire un fragment de 15 kDa
correspondant au peptide signal ainsi qu'une protéine mature glycosylée de 55-60 kDa
(Larkin et al., 2013). Le précurseur se retrouve coincé au RE alors que la protéine mature,
détectée via un épitope en C-terminal ou un anticorps dirigé contre cette extrémité, se
retrouve au niveau du Golgi, des lysosomes et est partiellement sécrétée en condition de
surexpression (Larkin et al., 2013). Nous avons ensuite voulu vérifier l'existence des deux
187
segments transmembranaires prédits, soit les aa 77-93 et 353-373. Par essai de protection
contre la protéinase K in vitro et in cellulo, nous avons établi que le N-terminal du
précurseur de CLN5 se trouve dans le cytoplasme alors que le C-terminal est dans la
lumière des organites confirmant la présence d'un seul segment transmembranaire et une
topologie de type II (Larkin et al., 2013). Afin de permettre l'accessibilité des enzymes
luminales au site de clivage du SP, le segment transmembranaire doit correspondre à la
région hydrophobe en N-terminal, c'est-à-dire la région la plus souvent prédite. La protéine
mature résultante est donc luminale indiquant que ses partenaires d'interaction directs
doivent se retrouver en totalité ou en partie dans lumière des organites. Ceci implique
également que l'action de CLN5 sur la machinerie cytosolique de transport rétrograde se
fait par un intermédiaire transmembranaire qui pourrait être son partenaire d'interaction
CLN3.
Malgré l'absence de segment transmembranaire dans la forme mature de CLN5, cette
dernière demeure fortement liée à la membrane lors d'expériences de fractionnement
cellulaire. Nous avons alors émis l'hypothèse que les aa 353-373, contenant de nombreux
résidus hydrophobes et non polaires, pourraient permettre l'ancrage de la protéine.
L'analyse de la séquence a révélé un haut niveau de conservation de la région 353-392 au
travers des espèces ainsi qu'une possible organisation en hélice amphipathique présentant
deux surfaces hydrophobes (Larkin et al., 2013). Les hélices amphipathiques sont
généralement localisées aux extrémités N-terminale ou C-terminale des protéines (Cornell
and Taneva, 2006), comme c'est le cas pour CLN5. Elles permettent un ancrage de faible
affinité, contrôlé et réversible, à des lipides généralement anioniques (phosphatidylinositol
et phosphatidylsérine), sans la nécessité d'un motif de sélectivité lipidique (Cornell and
Taneva, 2006). Par fractionnements cellulaires de segments protéiques en fusion avec GFP,
nous avons été en mesure de démontrer que les aa 353–392 suffisent en effet à l'association
membranaire (Larkin et al., 2013). Le rôle de l'hélice amphipathique de CLN5 pourrait
s'étendre au-delà de l'attachement membranaire. En effet, cette structure peut aussi servir de
domaine d'autoinhibition pour prévenir l'activation de protéines à l'état soluble, comme Arf,
ou pour cacher des sites d'interaction, comme c'est le cas chez la Vinculine (Cornell and
Taneva, 2006). Elle permet également de détecter et de moduler les propriétés physiques de
188
bicouches lipidiques. Les protéines à domaines N-BAR, telles que l'Amphiphysine, qui
répondent et induisent la courbure membranaire, en sont de bons exemples (Jao et al.,
2010). De plus amples études fonctionnelles et structurelles seront nécessaires afin de
déterminer le rôle exact de CLN5 et de son hélice amphipathique. Il est toutefois intéressant
de noter que, selon les prédictions informatiques, CLN3 possède une hélice semblable entre
son cinquième et sixième segment transmembranaire (Nugent et al., 2008). Il est alors
tentant de spéculer sur l'implication de cette structure dans l'interaction entre CLN3 et
CLN5 et leur rôle dans le transport intracellulaire.
Pour déterminer l'impact des mutations pathologiques sur la maturation, la sécrétion
et la localisation de CLN5 nous avons étudié les mutants communs EUR, FinM, SWE et
NFL. Ces différentes mutations n'affectent pas la maturation ni la glycosylation générale de
CLN5, mais entraînent leur délocalisation vers le RE (Larkin et al., 2013). De plus, les
mutants pathologiques dépourvus de leur hélice amphipathique, soit SWE et NFL, perdent
leur association membranaire et subissent une dégradation par le protéasome, plutôt qu'une
sécrétion accrue, témoignant de leur instabilité (Larkin et al., 2013). Cette deuxième étude a
permis de clarifier la localisation et la structure de CLN5 ainsi que de quelques mutants
pathologiques. Elle a également démontré que la présence d'une étiquette en C-terminal ne
nuit pas la maturation, la localisation ou encore la sécrétion de CLN5 et permet la détection
de la forme mature.
Au moment où paraissait notre article sur la topologie de CLN5, un autre groupe
publiait une étude des différents sites de glycosylation de CLN5. La mutation en
glutamines des huit sites de séquence consensus N-X-T/S, soit Asn179, 192, 227, 252, 304,
320, 330 et 401, a permis de confirmer leur glycosylation in vivo. Il existe des différences
fonctionnelles dans ces sites affectant le repliement, le transport ou la fonction de CLN5.
Les modifications en position 179, 252, 304, 320 et, plus modestement, 330 sont
nécessaires à la sortie du RE indiquant leur importance dans le repliement protéique alors
que la glycosylation Asn401 est essentielle pour le transport post-Golgi vers les lysosomes
(Moharir et al., 2013). Les glycosylations en Asn192 et Asn227 n'ont pas d'impact sur la
localisation, mais le caractère pathologique de la mutation N192S indique un rôle
189
fonctionnel pour cette modification (Moharir et al., 2013). À l'image de ce que nous avons
observé, les mutants pathologiques EUR (p.D279N), qui introduit un site de glycosylation,
et FinM (p.Y392*), qui perd le potentiel site de glycosylation en position 401, sont retenues
au RE (Moharir et al., 2013; Schmiedt et al., 2010). Par contre, contrairement à nos
résultats, ces deux mutants, de même que ceux retenus au RE, ne sont pas sécrétés (Moharir
et al., 2013). Cette divergence peut s'expliquer par la sensibilité de détection accrue de
notre système de précipitation radioactif par rapport à la capacité de détection par anticorps
sur des lysats cellulaires. Il n'en demeure pas moins que la sécrétion est faible et peut être
attribuée à la saturation de la machinerie cellulaire en condition de surexpression. Les
auteurs ont également remarqué que le mutant FinM est moins stable que le mutant EUR ce
qui est en accord avec la déstabilisation de l'hélice amphipathique et la sensibilité accrue à
l'extraction membranaire rapportée par notre groupe. Bref, la délocalisation de plusieurs
mutants pathologiques, dont p.D279N et p.Y392*, mais aussi p.R112P, p.R112H, p.E253*
et p.W379C, pourrait expliquer leur manque de fonctionnalité (Isosomppi et al., 2002;
Lebrun et al., 2009; Moharir et al., 2013; Schmiedt et al., 2010).
Récemment, la mise au point d'un nouvel anticorps par la compagnie Abcam
(ab170899) a rendu possible la détection efficace de CLN5 endogène dans de nombreux
tissus chez l'humain comme chez la souris. Ce nouvel outil reconnaît une région entre les aa
150 et 250 de CLN5. Il a permis de montrer l'existence de deux formes persistantes de
CLN5 dans la plupart des types cellulaires, soit une proforme de 56 kDa et une forme
mature dominante de 52 kDa (Silva et al., 2015). Cette différence de 4 kDa est attribuable à
la perte, suivant l'action d'une protéase à cystéine, d'environ 10-15 acides aminés en Cterminal (Silva et al., 2015). Dans notre étude de la topologique de CLN5, l'utilisation de
l'étiquette Flag après le peptide signal nous a d'ailleurs permis d'observer ce doublet pour la
forme WT, mais aussi pour le mutant EUR (Article 2 - figure 5) (Larkin et al., 2013). Par
contre, l'anticorps CLN5-C/32 ne permet pas cette détection puisqu'il est dirigé contre la
séquence
terminale
clivée
(EEIPLPIRNKTLSGL).
L'utilisation
d'une
étiquette
intramoléculaire ou d'anticorps reconnaissant la partie centrale de la protéine serait donc à
favoriser pour assurer l'étude de CLN5 mature. L'importance du clivage post-traductionnel
190
en aval de l'hélice amphipathique reste à être déterminée mais pourrait être nécessaire pour
l'ancrage et pour l'activité de CLN5.
La détection endogène de la protéine CLN5 a également permis de clarifier sa
sécrétion. En effet, aucune des formes endogènes de la protéine n'a été détectée dans le
milieu extracellulaire laissant croire que la sécrétion préalablement observée en système de
surexpression serait un artefact causé par la congestion et l'insuffisance de la machinerie
cellulaire (Silva et al., 2015). Ceci va de pair avec le modèle structural proposé par notre
équipe décrivant CLN5 comme étant une protéine soluble ancrée aux membranes luminales
par une hélice amphipathique afin d'assurer sa rétention cellulaire (Larkin et al., 2013).
Bref, la compréhension grandissante de la maturation de CLN5 et la faisabilité des études
endogènes assurent graduellement un consensus dans les études tout en expliquant les
nombreuses discordances antérieures.
Article 3 - Rôle de Calnuc dans les céroïdes-lipofuscinoses neuronales
Après avoir clarifié la topologie de CLN5, nous avons voulu vérifer l'existence d'un
lien fonctionnel entre Calnuc et CLN5. En effet, ces deux protéines influencent le
recrutement de Rab7 au niveau des endosomes et, conséquemment, affectent le transport de
récepteurs qui transitent entre le TGN et les endosomes. Alors que notre modèle dépeint
CLN5 mature comme étant une protéine luminale, notre étude du rôle de Calnuc a montré
l'implication de la fraction cytosolique dans le routage cellulaire. Ces données laissent
croire que Calnuc et CLN5 partagent une mécanistique commune qui implique un
partenaire d'interaction transmembranaire. CLN3, une protéine à six domaines
transmembranaires impliquée dans les lipofuscinoses et interagissant avec CLN5, s'est
avérée être la candidate de choix pour constituer cet intermédiaire. CLN3 serait impliquée
dans le transport intracellulaire puisqu'elle interagit directement avec Rab7 de façon à
influencer son recrutement (Uusi-Rauva et al., 2012). De plus, des études montrent que son
absence mène à une distribution anormale de protéines de la voie endolysosomiale de
même qu'à des défauts morphologiques au niveau des organites de cette voie (Fossale et al.,
2004; Lojewski et al., 2013; Metcalf et al., 2008). Tout comme l'absence de Calnuc et de
191
CLN5, l'expression du mutant pathologique CLN3Δex7/8/Δex7/8 (MT), dépourvu des exons 7 et
8 et arborant un changement de cadre de lecture au 153e acide aminé, entraîne une forte
diminution des niveaux de Rab7 actif. Dans un premier temps, nous avons donc montré que
Calnuc, CLN3 et CLN5 interagissent ensemble et colocalisent aux endosomes précoces.
Ces résultats ont mené à l'élaboration d'un modèle selon lequel Calnuc cytosolique, CLN3
transmembranaire et CLN5 luminale forment un complexe important pour le transport
intracellulaire (figure 12). La dynamique de ce modèle et le rôle exact de chaque partenaire
demeurent pour le moment inconnus. À l'avenir, l'identification des régions d'interaction
protéique impliquées permettra de mieux comprendre la mécanistique et l'impact des
diverses mutations pathologiques.
Figure 12 - Modèle du complexe Calnuc-CLN3-CLN5 influençant le recrutement de
Rab7 et du complexe Rétromère.
Calnuc cytosolique, la protéine à six segments transmembranaires CLN3 et la protéine
luminale ancrée CLN5 forment un complexe. Ce dernier permet le recyclage de récepteurs
en influençant l'activation de Rab7 et conséquemment le recrutement du complexe
Rétromère à la surface des endosomes. Figure créée avec Microsoft Office PowerPoint
2007.
L'implication du calcium dans le complexe Calnuc-CLN3-CLN5 est également une
avenue intéressante. Calnuc est principalement connue pour sa capacité à lier le calcium via
ses mains EF et à former une large réserve mobilisable de l'ion au niveau du Golgi. De son
côté, CLN3 interagit, de façon sensible au calcium, avec Calséniline, une protéine à mains
192
EF qui réprime la transcription (Chang et al., 2007). Ceci suggère la possibilité d'autres
interactions influencées par le calcium, avec des protéines comme Calnuc. Aussi, l'étude de
cellules exprimant des mutants pathologiques de CLN3 a permis d'associer la protéine à
l'homéostasie calcique du réticulum, des mitochondries et des lysosomes (Chandrachud et
al., 2015; Chang et al., 2007). Étant connu pour interagir avec des canaux ioniques (UusiRauva et al., 2008), CLN3 pourrait agir comme régulateur du trafic ou de l'activité de ces
derniers ou constituer un nouveau transporteur ionique. D'ailleurs, le calcium lysosomial
prend de plus en plus d'importance dans les phénomènes intracellulaires permettant, entre
autres, d'activer des protéines en lien avec la biogenèse des lysosomes et l'induction de
l'autophagie (Medina et al., 2015). Ainsi, la relâche de ce calcium pourrait permettre à
Calnuc de lier l'ion dans le cytosol et, possiblement, de moduler son activité. Quoi qu'il en
soit, au niveau des lipofuscinoses, les ions commencent à être considérés en raison de leur
importance pour la transmission synaptique au cerveau, mais surtout de leur neurotoxicité.
Une accumulation et une dérégulation du transport de métaux (zinc, cuivre, manganèse, fer,
cobalt), précédant la neurodégénération, ont d'ailleurs été rapportées chez des moutons de
type CLN6 (Grubman et al., 2014a; Kanninen et al., 2013) et des souris de type CLN1,
CLN3 et CLN5 (Grubman et al., 2014b).
La deuxième partie de l'étude consistait à déterminer si Calnuc, à l'image de ses deux
partenaires, CLN3 et CLN5, pourrait être impliquée dans les maladies du lysosome. Nous
avons vérifié l'impact de la déplétion de Calnuc sur l'apparition de phénotypes cellulaires
couramment retrouvés dans les lipofuscinoses. Par microscopie, nous avons observé la
formation d'ultrastructures de type curviligne, un grossissement des lysosomes, une
accumulation de matériel autofluorescent et de la sous-unité C de l'ATP synthase
mitochondriale (SCMAS) ainsi qu'une augmentation de l'autophagie. Ces évidences tendent
à démontrer le rôle de Calnuc dans les lipofuscinoses. Par contre, cette partie de l'étude
gagnerait en validité par l'usage d'un modèle cellulaire neurologique. C'est pourquoi nous
envisageons de reprendre ces expériences de microscopie en utilisant des cellules de
cervelet de souris. En ce qui a trait à la caractérisation du phénotype, des souris déficientes
en Calnuc, actuellement offertes chez Taconic Biosciences, seraient également un atout pour
la poursuite du projet.
193
Dans l'espoir d'associer une modulation de Calnuc à des cas de lipofuscinoses, nous
avons performé une analyse des niveaux de protéines et d'ARNm dans des lignées de
fibroblastes. Outre les lignées cellulaires contrôles, nous avons obtenu des lignées
provenant de patients atteints de NCL de types CLN1, CLN3 et CLN5, de même que de
type orphelin. En effet, à ce jour, malgré l'identification de 14 gènes porteurs de mutations
pathologiques, il existe encore des patients dont la cause génétique de leur maladie demeure
inconnue. Nous avons observé une réduction d'au moins 40% des niveaux protéiques de
Calnuc chez toutes les lignées de fibroblastes issues de patients atteints de NCL, ce qui
suggère une implication de cette dernière dans la maladie. Malgré l'absence de CLN1 dans
notre modèle, la lignée mutante pour cette protéine présente une forte diminution de
Calnuc. CLN1 ou PPT1 (Palmitoyl-protein thioesterase 1) est une enzyme lysosomiale qui
élimine les groupements lipidiques des protéines et qui a été montrée comme pouvant
interagir avec CLN5 (Lyly et al., 2009) de manière à influencer son transport vers les
lysosomes (Schmiedt et al., 2010). Il se peut que, par ce moyen, CLN1 influence
indirectement Calnuc. Aussi, il est intéressant de noter que Calnuc n'est pas affectée dans la
lignée contrôle Niemann-Pick Disease de type C1 (NPD), qui constitue une maladie du
lysosome en lien avec un défaut d'élimination des lipides intracellulaires et
mécanistiquement distincte des NCL. De son côté, l'analyse de l'ARNm de Calnuc n'a pas
révélé de variations significatives dans les différents échantillons. Par contre, Calnuc
pourrait présenter des mutants pathologiques stables dans des lipofuscinoses de type
orphelin. La recherche de ces mutations, de même que la confirmation des résultats
présentés ici, sont limitées par la faible disponibilité et diversité du matériel biologique issu
de patients. À l'heure actuelle, selon le site 'GeneCards: The Human Gene Database' une
centaine de mutations influençant la séquence de la protéine ont été rapportées, mais
aucune n'est associée à une pathologie.
La fonction de Calnuc au niveau des lysosomes pourrait prochainement être appuyée
par la récente découverte du motif CLEAR (Sardiello et al., 2009). Il s'agit d'une séquence
palindromique de 10 paires de bases (G/C/A-T/C/G-C-A-C/G/T-G/C-T/C-G-A/G-C/T/G ou
GTCACGTGAC) située à proximité du site d'initiation de la transcription et permettant de
coordonner l'expression et la régulation de gènes en lien avec la fonction lysosomiale. Elle
194
est reconnue par le facteur de transcription EB (TFEB) qui agit comme responsable de la
clairance cellulaire via la régulation de 291 gènes, dont plusieurs en lien avec la biogenèse
des lysosomes, la voie de l’autophagosome et les hydrolases lysosomiales (Sardiello et al.,
2009). TFEB constituerait une importante cible thérapeutique puisque sa surexpression
permet d'augmenter la genèse de lysosomes et d'augmenter la dégradation de molécules
complexes (Sardiello et al., 2009). Entre autres, le motif CLEAR est retrouvé au niveau des
gènes codant LAMP1, MPR, CLN1 (PTT1, CLN2 (TPP1), CLN3, CLN5, MFSD8 (CLN7),
CTSD (CLN10), GRN (CLN11), CTSF (CLN13) (Palmieri et al., 2011; Sardiello et al.,
2009). De son côté, Calnuc a été identifiée parmi les 25 gènes dont l'expression varie le
plus en fonction de Psap, CTSA ou CTSD et dans les 90 premiers variant pour CLN2
(Sardiello et al., 2009). Cette coordination d'expression suggère que Calnuc serait aussi
régulée par TFEB. Une rapide analyse de la séquence entourant les promoteurs de Calnuc
révèle en effet la présence d'une séquence GTCAAGAGGC à 101 nucléotides en amont du
site CCAAT. Il serait intéressant de vérifier si l'expression de Calnuc est modulée par
TFEB.
Les lipofuscinoses ne sont pas les premières maladies neurodégénératives dans
lesquelles Calnuc aurait un rôle à jouer. De plus en plus d'évidences soutiennent son
implication dans la maladie d'Alzheimer. Entre autres, elle permet de maintenir les niveaux
intracellulaires de Sortiline et de LRP10 (Brodeur et al., 2009), deux récepteurs d'APP qui
cycle entre le TGN et les endosomes, et protègent contre le clivage amyloïdogénique et
l'accumulation du peptide neurotoxique Aβ. Cela va de pair avec une réduction de Calnuc
(Lin et al., 2007) et de LRP10 (Brodeur et al., 2012) dans les cerveaux de patients atteints
de la maladie d'Alzheimer. Ces mêmes tissus présentent une réduction des rétromères
Vps26 et Vps35 (Small et al., 2005). Il se pourrait donc que l'effet de Calnuc sur
l'activation de Rab7 et le recrutement du complexe Rétromère soit également en cause.
195
CONCLUSION
La présente étude a permis de mettre en lumière un nouveau rôle pour la protéine
Calnuc dans le transport rétrograde de récepteurs entre les endosomes et le TGN. Nos
données démontrent que Calnuc influence positivement l'activité de la petite protéine G
Rab7 qui agit comme régulateur spatio-temporel lors de multiples processus
intracellulaires, dont le transport rétrograde. En fait, Calnuc formerait un complexe avec les
protéines CLN3 et CLN5 qui ont également cette fonction. La mécanistique exacte de la
formation de ce complexe reste inconnue et l'absence d'interactions directes entre les
partenaires laisse supposer l'implication d'autres protéines et la complexification du modèle
proposé ici. Calnuc partage donc une fonction commune avec des protéines impliquées
dans les céroïdes-lipofuscinoses neuronales suggérant son implication dans cette maladie
neurodégénérative. La déplétion de Calnuc entraîne, en effet, l'apparition de phénotypes
cellulaires typiques. La forte modulation de son expression dans toutes les lignées de
fibroblastes de patients atteints de NCL à notre disposition soutient cette hypothèse malgré
l'absence de variation au niveau de l'ARNm. Il se peut donc qu'il existe des mutants
pathologiques stables de Calnuc responsables de formes orphelines de la maladie. La
recherche de ces mutations et l'étude de la fonction exacte de Calnuc dans la pathogenèse
seraient alors primordiales puisque la protéine pourrait constituer un nouvel outil
diagnostique et une nouvelle cible thérapeutique pour le traitement de lipofuscinoses.
196
REMERCIEMENTS
Pour débuter, je tiens à remercier les Pr Alexey Pshezhetsky, Xavier Roucou et Michel
Grandbois pour la révision de ma thèse et pour les commentaires constructifs que vous y
avez apportés. Ma reconnaissance va surtout à ma directrice de recherche Pre Christine
Lavoie qui m'a accueilli dans son laboratoire, d'abord comme stagiaire, puis durant sept
ans. Merci de m'avoir guidé au travers mon doctorat et d'avoir su me pousser en temps et
lieu. Tu es un modèle de travail acharné et de persévérance digne des scientifiques les plus
passionnés. La qualité, l'étendue et la rigueur de la formation que tu m'as offerte me sera un
atout pour ma carrière. J'ai également trouvé auprès de toi une amie qui m'a aidé à passer au
travers les épreuves des études aux cycles supérieurs, mais aussi de la vie.
En cette difficile période pour le financement de la recherche, je crois qu'il est
important de souligner le rôle des organismes subventionnaires. Mon projet a été supporté
par les IRSC et j'ai eu la chance d'obtenir une des rares bourses d’études supérieures du
CRSNG, ce qui a grandement facilité mon parcours. Aussi, à trois occasions, j'ai participé à
des congrès à l'international, entre autres, grâce à la Bourse IPS-RECPUS pour congrès
international et à la Bourse de voyage d’appui communautaire des Instituts (IRSC). Ces
fonds aident à établir des contacts qui peuvent faire toute la différence dans le cheminement
d'un étudiant. Finalement, merci à l'Université de Sherbrooke pour les bourses de stage, de
maîtrise et de doctorat qui constituent un encouragement et un bonus salariale non
négligeable.
Je suis heureuse d'avoir eu le privilège de participer à un projet de collaboration avec
Marc-André Bonin, Hassan Trabulsi et, dernièrement, Étienne Marousseau du laboratoire
de Pr Éric Marsault. Merci les gars d'avoir mis un peu de chimie dans mes études. Grâce à
vous, et à Leonid Volkov (responsable de la Plateforme de biophotonique du Centre de
recherche clinique Étienne-Le Bel), j'ai entre autres appris les rudiments du Fluorescenceactivated cell sorting (FACS). Également, un merci à Pr Pierre Lavigne pour les conseils en
matière de structure protéique qui m'ont été fort utiles pour l'article concernant CLN5.
197
Je voudrais aussi remercier les chercheurs, employés et étudiants des laboratoires
voisins que j'ai côtoyés tout au long de ces années et avec qui j'ai partagé anecdotes,
conseils expérimentaux et matériel. Il m'a fait plaisir de vous connaître. Merci aux membres
présents et passés de mon laboratoire, soit Hugo Gagnon, Julie Brodeur avec qui j'ai
travaillé pour l'article à propos de LRP9, Stéphanie Rosciglione, Catherine Duclos,
Sébastien Grastilleur, Erwan Lanchec, Xuezhi Li, Étienne Marousseau et Marilène
Paquette. Parmi mes collègues certains sont devenus des amis : le très critique Élie Simard,
Yannick Miron, le gastronome, Yannik Réginbal-Dumas, qui m'a montré l'importance de
l'implication sociale et scolaire, Thomas Söllradl, avec qui j'ai siroté moultes bières en
pratiquant mon anglais et mon allemand, Marc-Olivier Frégeau, mon collègue depuis le
baccalauréat, le toujours souriant Alexandre Desroches, Martin Houde l'encyclopédie et
Jean-Sébastien Maltais, mon 'buddy' de salle de culture. En 2006, alors que je faisais mes
premières armes dans le département, j'ai eu le privilège de connaître des étudiants
finissants qui sont devenus mes modèles. Je parle ici de Pascale Lepage et Marc Lussier,
'mon grand-frère académique'.
Au cours de ces années, deux personnes sont devenues des amis indispensables avec
qui je garderai sans doute contact toute ma vie. Stéphanie Rosciglione, alias Darling, ma
collègue doctorante, mon amie et ma confidente. En raison de notre cheminement parallèle,
nous avons partagé nos épreuves, nos réussites, nos peines, nos anecdotes, nos
découragements et nos joies. J'espère que nos chemins se recroiseront et au plaisir de se
faire de nouveau calciner au soleil avec une bouteille de Martini Rosso. Je me souviens
encore du jour où, après qu'il m'ait offert d'utiliser son incubateur, j'ai dit à Dave Boucher
qu'il était devenu 'mon nouveau meilleur ami'. À ce moment-là, je ne savais pas à quel point
c'était vrai. Dave, tu as toujours été l'oreille attentive, le gars qui connaît tous les petits trucs
de lab, le super motivé, l'hyperactif, l'homme capable de tout faire en même temps ; un
futur grand chercheur quoi! Merci pour ces soirées à discuter au laboratoire, pour le sofa et
pour ce merveilleux voyage en Australie.
Le laboratoire fut une grosse portion de ma vie pendant les dernières années et
plusieurs personnes ont veillé à y mettre de l'équilibre. Merci donc à mes amis Luc
198
Paquette, Alexei Nordel-Markovits, Marie-Sol Poirier et Marc-André Daviault. Un merci
aussi à Catherine Bourdon et aux autres personnes du Domaine Équi-Sphère pour m'avoir
fait sortir du monde universitaire ainsi qu'à Tiptoe et Tao pour la zoothérapie de fin de
doctorat.
Pour terminer, je remercie chaleureusement mes parents, Pierre et Francine, de
m'avoir soutenue moralement et financièrement pendant mon long parcours scolaire. Je suis
reconnaissante envers ma fratrie, c'est-à-dire mon frère David qui m'a hébergé dans son
duplex, ma studieuse soeur Annie-Pier ainsi que mon frère Samuel et sa conjointe Nadine.
Finalement, les mots ne suffisent guère pour exprimer ma gratitude envers mon conjoint
Marc Therrien pour ces soirées à m'attendre et ces fins de semaine à m'accompagner au
travail, mais surtout pour son amour, sa compréhension et ses encouragements. Merci à
vous tous de m'avoir montré ce qui est le plus important dans la vie.
199
LISTE DES PUBLICATIONS
Adam, P. J., Boyd, R., Tyson, K. L., Fletcher, G. C., Stamps, A., Hudson, L., Poyser, H. R.,
Redpath, N., Griffiths, M., Steers, G., Harris, A. L., Patel, S., Berry, J., Loader, J.
A., Townsend, R. R., Daviet, L., Legrain, P., Parekh, R., and Terrett, J. A. (2003):
Comprehensive proteomic analysis of breast cancer cell membranes reveals unique
proteins with potential roles in clinical cancer. J Biol Chem 278, 6482-9.
Afshar, N., Black, B. E., and Paschal, B. M. (2005): Retrotranslocation of the chaperone
calreticulin from the endoplasmic reticulum lumen to the cytosol. Mol Cell Biol 25,
8844-53.
Alory, C., and Balch, W. E. (2001): Organization of the Rab-GDI/CHM superfamily: the
functional basis for choroideremia disease. Traffic 2, 532-43.
Alvarez, C., Garcia-Mata, R., Brandon, E., and Sztul, E. (2003): COPI recruitment is
modulated by a Rab1b-dependent mechanism. Mol Biol Cell 14, 2116-27.
Antonin, W., Fasshauer, D., Becker, S., Jahn, R., and Schneider, T. R. (2002): Crystal
structure of the endosomal SNARE complex reveals common structural principles
of all SNAREs. Nat Struct Biol 9, 107-11.
Aradhyam, G. K., Balivada, L. M., Kanuru, M., Vadivel, P., and Vidhya, B. S. (2010):
Calnuc: Emerging roles in calcium signaling and human diseases. IUBMB Life 62,
436-46.
Baldwin, S. A., Beal, P. R., Yao, S. Y., King, A. E., Cass, C. E., and Young, J. D. (2004):
The equilibrative nucleoside transporter family, SLC29. Pflugers Arch 447, 735-43.
Ballabio, A., and Gieselmann, V. (2009): Lysosomal disorders: from storage to cellular
damage. Biochim Biophys Acta 1793, 684-96.
Ballif, B. A., Mincek, N. V., Barratt, J. T., Wilson, M. L., and Simmons, D. L. (1996):
Interaction of cyclooxygenases with an apoptosis- and autoimmunity-associated
protein. Proc Natl Acad Sci U S A 93, 5544-9.
Bandyopadhyay, U., and Cuervo, A. M. (2008): Entering the lysosome through a transient
gate by chaperone-mediated autophagy. Autophagy 4, 1101-3.
Bandyopadhyay, U., Kaushik, S., Varticovski, L., and Cuervo, A. M. (2008): The
chaperone-mediated autophagy receptor organizes in dynamic protein complexes at
the lysosomal membrane. Mol Cell Biol 28, 5747-63.
Barnikol-Watanabe, S., Gross, N. A., Gotz, H., Henkel, T., Karabinos, A., Kratzin, H.,
Barnikol, H. U., and Hilschmann, N. (1994): Human protein NEFA, a novel DNA
binding/EF-hand/leucine zipper protein. Molecular cloning and sequence analysis of
the cDNA, isolation and characterization of the protein. Biol Chem Hoppe Seyler
375, 497-512.
Batten, F. (1903): Cerebral degeneration with symmetrical changes in the maculae in two
members of a family. Trans Ophthalmol Soc UK 23, 386-390.
Batten, F. (1914): Family cerebral degeneration with macular change (so-called juvenile
form of family amaurotic idiocy). Q J Med 7, 444-454.
Belin, D., Bost, S., Vassalli, J. D., and Strub, K. (1996): A two-step recognition of signal
sequences determines the translocation efficiency of proteins. Embo J 15, 468-78.
200
Bellettato, C. M., and Scarpa, M. (2010): Pathophysiology of neuropathic lysosomal
storage disorders. J Inherit Metab Dis 33, 347-62.
Bento, C. F., Puri, C., Moreau, K., and Rubinsztein, D. C. (2013): The role of membranetrafficking small GTPases in the regulation of autophagy. J Cell Sci 126, 1059-69.
Berggard, T., Miron, S., Onnerfjord, P., Thulin, E., Akerfeldt, K. S., Enghild, J. J., Akke,
M., and Linse, S. (2002): Calbindin D28k exhibits properties characteristic of a
Ca2+ sensor. J Biol Chem 277, 16662-72.
Bessa, C., Teixeira, C. A. F., Mangas, M., Dias, A., Sá Miranda, M. C., Guimarães, A.,
Ferreira, J. C., Canas, N., Cabral, P., and Ribeiro, M. G. (2006): Two novel CLN5
mutations in a Portuguese patient with vLINCL: Insights into molecular
mechanisms of CLN5 deficiency. Molecular Genetics and Metabolism 89, 245-253.
Bonifacino, J. S. (2004): The GGA proteins: adaptors on the move. Nat Rev Mol Cell Biol
5, 23-32.
Bonifacino, J. S., and Traub, L. M. (2003): Signals for sorting of transmembrane proteins to
endosomes and lysosomes. Annu Rev Biochem 72, 395-447.
Boucher, R., Larkin, H., Brodeur, J., Gagnon, H., Theriault, C., and Lavoie, C. (2008):
Intracellular trafficking of LRP9 is dependent on two acidic cluster/dileucine
motifs. Histochem Cell Biol 130, 315-27.
Brailoiu, G. C., Dun, S. L., Brailoiu, E., Inan, S., Yang, J., Chang, J. K., and Dun, N. J.
(2007): Nesfatin-1: distribution and interaction with a G protein-coupled receptor in
the rat brain. Endocrinology 148, 5088-94.
Brasaemle, D. L. (2007): Thematic review series: adipocyte biology. The perilipin family
of structural lipid droplet proteins: stabilization of lipid droplets and control of
lipolysis. J Lipid Res 48, 2547-59.
Braulke, T., and Bonifacino, J. S. (2009): Sorting of lysosomal proteins. Biochim Biophys
Acta 1793, 605-14.
Braulke, T., Gartung, C., Hasilik, A., and von Figura, K. (1987): Is movement of mannose
6-phosphate-specific receptor triggered by binding of lysosomal enzymes? J Cell
Biol 104, 1735-42.
Brean, A. (2004): [An account of a strange instance of disease--Stengel-Batten-SpielmayerVogt disease]. Tidsskr Nor Laegeforen 124, 970-1.
Breuer, P., Korner, C., Boker, C., Herzog, A., Pohlmann, R., and Braulke, T. (1997): Serine
phosphorylation site of the 46-kDa mannose 6-phosphate receptor is required for
transport to the plasma membrane in Madin-Darby canine kidney and mouse
fibroblast cells. Mol Biol Cell 8, 567-76.
Brocker, C., Engelbrecht-Vandre, S., and Ungermann, C. (2010): Multisubunit tethering
complexes and their role in membrane fusion. Curr Biol 20, R943-52.
Brodeur, J., Larkin, H., Boucher, R., Theriault, C., St-Louis, S. C., Gagnon, H., and Lavoie,
C. (2009): Calnuc binds to LRP9 and affects its endosomal sorting. Traffic 10,
1098-114.
Brodeur, J., Theriault, C., Lessard-Beaudoin, M., Marcil, A., Dahan, S., and Lavoie, C.
(2012): LDLR-related protein 10 (LRP10) regulates amyloid precursor protein
(APP) trafficking and processing: evidence for a role in Alzheimer's disease. Mol
Neurodegener 7, 31.
201
Brown, J., Esnouf, R. M., Jones, M. A., Linnell, J., Harlos, K., Hassan, A. B., and Jones, E.
Y. (2002): Structure of a functional IGF2R fragment determined from the
anomalous scattering of sulfur. Embo J 21, 1054-62.
Brunk, U. T., and Terman, A. (2002a): Lipofuscin: mechanisms of age-related
accumulation and influence on cell function. Free Radic Biol Med 33, 611-9.
Brunk, U. T., and Terman, A. (2002b): The mitochondrial-lysosomal axis theory of aging:
accumulation of damaged mitochondria as a result of imperfect autophagocytosis.
Eur J Biochem 269, 1996-2002.
Bucci, C., Parton, R. G., Mather, I. H., Stunnenberg, H., Simons, K., Hoflack, B., and
Zerial, M. (1992): The small GTPase rab5 functions as a regulatory factor in the
early endocytic pathway. Cell 70, 715-28.
Bugarcic, A., Zhe, Y., Kerr, M. C., Griffin, J., Collins, B. M., and Teasdale, R. D. (2011):
Vps26A and Vps26B subunits define distinct retromer complexes. Traffic 12, 175973.
Bulankina, A. V., Deggerich, A., Wenzel, D., Mutenda, K., Wittmann, J. G., Rudolph, M.
G., Burger, K. N., and Honing, S. (2009): TIP47 functions in the biogenesis of lipid
droplets. J Cell Biol 185, 641-55.
Burd, C. G. (2011): Physiology and pathology of endosome-to-Golgi retrograde sorting.
Traffic 12, 948-55.
Campellone, K. G., and Welch, M. D. (2010): A nucleator arms race: cellular control of
actin assembly. Nat Rev Mol Cell Biol 11, 237-51.
Cang, C., Zhou, Y., Navarro, B., Seo, Y. J., Aranda, K., Shi, L., Battaglia-Hsu, S., Nissim,
I., Clapham, D. E., and Ren, D. (2013): mTOR regulates lysosomal ATP-sensitive
two-pore Na(+) channels to adapt to metabolic state. Cell 152, 778-90.
Canuel, M., Libin, Y., and Morales, C. R. (2009): The interactomics of sortilin: an ancient
lysosomal receptor evolving new functions. Histol Histopathol 24, 481-92.
Cao, X., and Sudhof, T. C. (2001): A transcriptionally [correction of transcriptively] active
complex of APP with Fe65 and histone acetyltransferase Tip60. Science 293, 11520.
Cao, Y., Espinola, J. A., Fossale, E., Massey, A. C., Cuervo, A. M., MacDonald, M. E., and
Cotman, S. L. (2006): Autophagy is disrupted in a knock-in mouse model of
juvenile neuronal ceroid lipofuscinosis. J Biol Chem 281, 20483-93.
Carlo, A. S., Gustafsen, C., Mastrobuoni, G., Nielsen, M. S., Burgert, T., Hartl, D., Rohe,
M., Nykjaer, A., Herz, J., Heeren, J., Kempa, S., Petersen, C. M., and Willnow, T.
E. (2013): The pro-neurotrophin receptor sortilin is a major neuronal apolipoprotein
E receptor for catabolism of amyloid-beta peptide in the brain. J Neurosci 33, 35870.
Carlton, J., Bujny, M., Peter, B. J., Oorschot, V. M., Rutherford, A., Mellor, H.,
Klumperman, J., McMahon, H. T., and Cullen, P. J. (2004): Sorting nexin-1
mediates tubular endosome-to-TGN transport through coincidence sensing of highcurvature membranes and 3-phosphoinositides. Curr Biol 14, 1791-800.
Carroll, K. S., Hanna, J., Simon, I., Krise, J., Barbero, P., and Pfeffer, S. R. (2001): Role of
Rab9 GTPase in facilitating receptor recruitment by TIP47. Science 292, 1373-6.
Chan, C. H., Ramirez-Montealegre, D., and Pearce, D. A. (2009): Altered arginine
metabolism in the central nervous system (CNS) of the Cln3-/- mouse model of
juvenile Batten disease. Neuropathol Appl Neurobiol 35, 189-207.
202
Chandrachud, U., Walker, M. W., Simas, A. M., Heetveld, S., Petcherski, A., Klein, M.,
Oh, H., Wolf, P., Zhao, W. N., Norton, S., Haggarty, S. J., Lloyd-Evans, E., and
Cotman, S. L. (2015): Unbiased Cell-based Screening in a Neuronal Cell Model of
Batten Disease Highlights an Interaction between Ca2+ Homeostasis, Autophagy,
and CLN3 Protein Function. J Biol Chem 290, 14361-80.
Chang, J. W., Choi, H., Kim, H. J., Jo, D. G., Jeon, Y. J., Noh, J. Y., Park, W. J., and Jung,
Y. K. (2007): Neuronal vulnerability of CLN3 deletion to calcium-induced
cytotoxicity is mediated by calsenilin. Hum Mol Genet 16, 317-26.
Chattopadhyay, S., and Pearce, D. A. (2002): Interaction with Btn2p is required for
localization of Rsglp: Btn2p-mediated changes in arginine uptake in Saccharomyces
cerevisiae. Eukaryot Cell 1, 606-12.
Chavrier, P., Parton, R. G., Hauri, H. P., Simons, K., and Zerial, M. (1990): Localization of
low molecular weight GTP binding proteins to exocytic and endocytic
compartments. Cell 62, 317-29.
Chen, H. J., Remmler, J., Delaney, J. C., Messner, D. J., and Lobel, P. (1993): Mutational
analysis of the cation-independent mannose 6-phosphate/insulin-like growth factor
II receptor. A consensus casein kinase II site followed by 2 leucines near the
carboxyl terminus is important for intracellular targeting of lysosomal enzymes. J
Biol Chem 268, 22338-46.
Chen, H. J., Yuan, J., and Lobel, P. (1997): Systematic mutational analysis of the cationindependent mannose 6-phosphate/insulin-like growth factor II receptor
cytoplasmic domain. An acidic cluster containing a key aspartate is important for
function in lysosomal enzyme sorting. J Biol Chem 272, 7003-12.
Chen, Y., Lin, P., Qiu, S., Peng, X. X., Looi, K., Farquhar, M. G., and Zhang, J. Y. (2007):
Autoantibodies to Ca2+ binding protein Calnuc is a potential marker in colon
cancer detection. Int J Oncol 30, 1137-44.
Cheng, X., Shen, D., Samie, M., and Xu, H. (2010): Mucolipins: Intracellular TRPML1-3
channels. FEBS Lett 584, 2013-21.
Chia, P. Z., Gasnereau, I., Lieu, Z. Z., and Gleeson, P. A. (2011): Rab9-dependent
retrograde transport and endosomal sorting of the endopeptidase furin. J Cell Sci
124, 2401-13.
Chia, P. Z., Gunn, P., and Gleeson, P. A. (2013): Cargo trafficking between endosomes and
the trans-Golgi network. Histochem Cell Biol 140, 307-15.
Choy, R. W., Cheng, Z., and Schekman, R. (2012): Amyloid precursor protein (APP)
traffics from the cell surface via endosomes for amyloid beta (Abeta) production in
the trans-Golgi network. Proc Natl Acad Sci U S A 109, E2077-82.
Christoforidis, S., McBride, H. M., Burgoyne, R. D., and Zerial, M. (1999a): The Rab5
effector EEA1 is a core component of endosome docking. Nature 397, 621-5.
Christoforidis, S., Miaczynska, M., Ashman, K., Wilm, M., Zhao, L., Yip, S. C.,
Waterfield, M. D., Backer, J. M., and Zerial, M. (1999b): Phosphatidylinositol-3OH kinases are Rab5 effectors. Nat Cell Biol 1, 249-52.
Codlin, S., and Mole, S. E. (2009): S. pombe btn1, the orthologue of the Batten disease
gene CLN3, is required for vacuole protein sorting of Cpy1p and Golgi exit of
Vps10p. J Cell Sci 122, 1163-73.
203
Collins, B. M., Skinner, C. F., Watson, P. J., Seaman, M. N., and Owen, D. J. (2005a):
Vps29 has a phosphoesterase fold that acts as a protein interaction scaffold for
retromer assembly. Nat Struct Mol Biol 12, 594-602.
Collins, K. M., Thorngren, N. L., Fratti, R. A., and Wickner, W. T. (2005b): Sec17p and
HOPS, in distinct SNARE complexes, mediate SNARE complex disruption or
assembly for fusion. Embo J 24, 1775-86.
Cooper, G. M., and Hausman, R. E. (2006): The Cell: A Molecular Approach. Sinauer
Associates, Inc.
Cornell, R. B., and Taneva, S. G. (2006): Amphipathic helices as mediators of the
membrane interaction of amphitropic proteins, and as modulators of bilayer
physical properties. Curr Protein Pept Sci 7, 539-52.
Cox, T. M. (2005): Substrate reduction therapy for lysosomal storage diseases. Acta
Paediatr Suppl 94, 69-75; discussion 57.
Cozier, G. E., Carlton, J., McGregor, A. H., Gleeson, P. A., Teasdale, R. D., Mellor, H.,
and Cullen, P. J. (2002): The phox homology (PX) domain-dependent, 3phosphoinositide-mediated association of sorting nexin-1 with an early sorting
endosomal compartment is required for its ability to regulate epidermal growth
factor receptor degradation. J Biol Chem 277, 48730-6.
Cuajungco, M. P., and Lees, G. J. (1997a): Zinc and Alzheimer's disease: is there a direct
link? Brain Res Brain Res Rev 23, 219-36.
Cuajungco, M. P., and Lees, G. J. (1997b): Zinc metabolism in the brain: relevance to
human neurodegenerative disorders. Neurobiol Dis 4, 137-69.
Cullen, P. J. (2011): Phosphoinositides and the regulation of tubular-based endosomal
sorting. Biochem Soc Trans 39, 839-50.
Cullen, P. J., and Korswagen, H. C. (2011): Sorting nexins provide diversity for retromerdependent trafficking events. Nat Cell Biol 14, 29-37.
da Silva, A. C., and Reinach, F. C. (1991): Calcium binding induces conformational
changes in muscle regulatory proteins. Trends Biochem Sci 16, 53-7.
Dahms, N. M., and Hancock, M. K. (2002): P-type lectins. Biochim Biophys Acta 1572,
317-40.
Damen, E., Krieger, E., Nielsen, J. E., Eygensteyn, J., and van Leeuwen, J. E. (2006): The
human Vps29 retromer component is a metallo-phosphoesterase for a cationindependent mannose 6-phosphate receptor substrate peptide. Biochem J 398, 399409.
Daumke, O., Lundmark, R., Vallis, Y., Martens, S., Butler, P. J., and McMahon, H. T.
(2007): Architectural and mechanistic insights into an EHD ATPase involved in
membrane remodelling. Nature 449, 923-7.
de Alba, E., and Tjandra, N. (2004): Structural studies on the Ca2+-binding domain of
human nucleobindin (calnuc). Biochemistry 43, 10039-49.
De Duve, C., Pressman, B. C., Gianetto, R., Wattiaux, R., and Appelmans, F. (1955):
Tissue fractionation studies. 6. Intracellular distribution patterns of enzymes in ratliver tissue. Biochem J 60, 604-17.
De Luca, A., Progida, C., Spinosa, M. R., Alifano, P., and Bucci, C. (2008):
Characterization of the Rab7K157N mutant protein associated with Charcot-MarieTooth type 2B. Biochem Biophys Res Commun 372, 283-7.
204
Derivery, E., and Gautreau, A. (2010): Evolutionary conservation of the WASH complex,
an actin polymerization machine involved in endosomal fission. Commun Integr
Biol 3, 227-30.
Derivery, E., Sousa, C., Gautier, J. J., Lombard, B., Loew, D., and Gautreau, A. (2009):
The Arp2/3 activator WASH controls the fission of endosomes through a large
multiprotein complex. Dev Cell 17, 712-23.
Desnick, R. J. (2004): Enzyme replacement and enhancement therapies for lysosomal
diseases. J Inherit Metab Dis 27, 385-410.
Di Fruscio, G., Schulz, A., De Cegli, R., Savarese, M., Mutarelli, M., Parenti, G., Banfi, S.,
Braulke, T., Nigro, V., and Ballabio, A. (2015): Lysoplex: An efficient toolkit to
detect DNA sequence variations in the autophagy-lysosomal pathway. Autophagy
11, 928-38.
Diaz, E., and Pfeffer, S. R. (1998): TIP47: a cargo selection device for mannose 6phosphate receptor trafficking. Cell 93, 433-43.
Dingwall, C., and Laskey, R. A. (1991): Nuclear targeting sequences--a consensus? Trends
Biochem Sci 16, 478-81.
Dittmer, F., Ulbrich, E. J., Hafner, A., Schmahl, W., Meister, T., Pohlmann, R., and von
Figura, K. (1999): Alternative mechanisms for trafficking of lysosomal enzymes in
mannose 6-phosphate receptor-deficient mice are cell type-specific. J Cell Sci 112 (
Pt 10), 1591-7.
Doray, B., Ghosh, P., Griffith, J., Geuze, H. J., and Kornfeld, S. (2002): Cooperation of
GGAs and AP-1 in packaging MPRs at the trans-Golgi network. Science 297, 17003.
Doray, B., Knisely, J. M., Wartman, L., Bu, G., and Kornfeld, S. (2008): Identification of
acidic dileucine signals in LRP9 that interact with both GGAs and AP-1/AP-2.
Traffic 9, 1551-62.
Ducharme, N. A., and Bickel, P. E. (2008): Lipid droplets in lipogenesis and lipolysis.
Endocrinology 149, 942-9.
Duncan, J. R., and Kornfeld, S. (1988): Intracellular movement of two mannose 6phosphate receptors: return to the Golgi apparatus. J Cell Biol 106, 617-28.
Eaton, S. (2008): Retromer retrieves wntless. Dev Cell 14, 4-6.
Elleder, L., Gobel (1999): Definitions of the ultrastructural patterns found in NCL.
Elleder, M., Sokolova, J., and Hrebicek, M. (1997): Follow-up study of subunit c of
mitochondrial ATP synthase (SCMAS) in Batten disease and in unrelated lysosomal
disorders. Acta Neuropathol 93, 379-90.
Emmerzaal, T. L., and Kozicz, T. (2013): Nesfatin-1; implication in stress and stressassociated anxiety and depression. Curr Pharm Des 19, 6941-8.
Eng, C. M., Banikazemi, M., Gordon, R. E., Goldman, M., Phelps, R., Kim, L., Gass, A.,
Winston, J., Dikman, S., Fallon, J. T., Brodie, S., Stacy, C. B., Mehta, D., Parsons,
R., Norton, K., O'Callaghan, M., and Desnick, R. J. (2001): A phase 1/2 clinical
trial of enzyme replacement in fabry disease: pharmacokinetic, substrate clearance,
and safety studies. Am J Hum Genet 68, 711-22.
Espinosa, E. J., Calero, M., Sridevi, K., and Pfeffer, S. R. (2009): RhoBTB3: a Rho
GTPase-family ATPase required for endosome to Golgi transport. Cell 137, 938-48.
205
Evans, J. H., Spencer, D. M., Zweifach, A., and Leslie, C. C. (2001): Intracellular calcium
signals regulating cytosolic phospholipase A2 translocation to internal membranes.
J Biol Chem 276, 30150-60.
Ezaki, J., Takeda-Ezaki, M., Koike, M., Ohsawa, Y., Taka, H., Mineki, R., Murayama, K.,
Uchiyama, Y., Ueno, T., and Kominami, E. (2003): Characterization of Cln3p, the
gene product responsible for juvenile neuronal ceroid lipofuscinosis, as a lysosomal
integral membrane glycoprotein. J Neurochem 87, 1296-308.
Fasshauer, D., Sutton, R. B., Brunger, A. T., and Jahn, R. (1998): Conserved structural
features of the synaptic fusion complex: SNARE proteins reclassified as Q- and RSNAREs. Proc Natl Acad Sci U S A 95, 15781-6.
Finan, G. M., Okada, H., and Kim, T. W. (2011): BACE1 retrograde trafficking is uniquely
regulated by the cytoplasmic domain of sortilin. J Biol Chem 286, 12602-16.
Fjorback, A. W., Seaman, M., Gustafsen, C., Mehmedbasic, A., Gokool, S., Wu, C., Militz,
D., Schmidt, V., Madsen, P., Nyengaard, J. R., Willnow, T. E., Christensen, E. I.,
Mobley, W. B., Nykjaer, A., and Andersen, O. M. (2012): Retromer binds the
FANSHY sorting motif in SorLA to regulate amyloid precursor protein sorting and
processing. J Neurosci 32, 1467-80.
Ford, M. G., Mills, I. G., Peter, B. J., Vallis, Y., Praefcke, G. J., Evans, P. R., and
McMahon, H. T. (2002): Curvature of clathrin-coated pits driven by epsin. Nature
419, 361-6.
Fossale, E., Wolf, P., Espinola, J. A., Lubicz-Nawrocka, T., Teed, A. M., Gao, H.,
Rigamonti, D., Cattaneo, E., MacDonald, M. E., and Cotman, S. L. (2004):
Membrane trafficking and mitochondrial abnormalities precede subunit c deposition
in a cerebellar cell model of juvenile neuronal ceroid lipofuscinosis. BMC Neurosci
5, 57.
Fukuda, M. (1990): [Lysosomal membrane glycoproteins; structure, biosynthesis, and
trafficking to lysosome]. Seikagaku 62, 1225-40.
Ganley, I. G., Carroll, K., Bittova, L., and Pfeffer, S. (2004): Rab9 GTPase regulates late
endosome size and requires effector interaction for its stability. Mol Biol Cell 15,
5420-30.
Garcia-Galiano, D., Navarro, V. M., Roa, J., Ruiz-Pino, F., Sanchez-Garrido, M. A.,
Pineda, R., Castellano, J. M., Romero, M., Aguilar, E., Gaytan, F., Dieguez, C.,
Pinilla, L., and Tena-Sempere, M. (2010): The anorexigenic neuropeptide, nesfatin1, is indispensable for normal puberty onset in the female rat. J Neurosci 30, 778392.
Garcia-Galiano, D., Pineda, R., Ilhan, T., Castellano, J. M., Ruiz-Pino, F., SanchezGarrido, M. A., Vazquez, M. J., Sangiao-Alvarellos, S., Romero-Ruiz, A., Pinilla,
L., Dieguez, C., Gaytan, F., and Tena-Sempere, M. (2012): Cellular distribution,
regulated expression, and functional role of the anorexigenic peptide,
NUCB2/nesfatin-1, in the testis. Endocrinology 153, 1959-71.
Garcia-Galiano, D., and Tena-Sempere, M. (2013): Emerging roles of NUCB2/nesfatin-1 in
the metabolic control of reproduction. Curr Pharm Des 19, 6966-72.
Garcia-Marcos, M., Kietrsunthorn, P. S., Wang, H., Ghosh, P., and Farquhar, M. G. (2011):
G Protein binding sites on Calnuc (nucleobindin 1) and NUCB2 (nucleobindin 2)
define a new class of G(alpha)i-regulatory motifs. J Biol Chem 286, 28138-49.
206
Garmroudi, F., Devi, G., Slentz, D. H., Schaffer, B. S., and MacDonald, R. G. (1996):
Truncated forms of the insulin-like growth factor II (IGF-II)/mannose 6-phosphate
receptor encompassing the IGF-II binding site: characterization of a point mutation
that abolishes IGF-II binding. Mol Endocrinol 10, 642-51.
Getty, A., Kovacs, A. D., Lengyel-Nelson, T., Cardillo, A., Hof, C., Chan, C. H., and
Pearce, D. A. (2013): Osmotic stress changes the expression and subcellular
localization of the Batten disease protein CLN3. PLoS One 8, e66203.
Getty, A. L., Benedict, J. W., and Pearce, D. A. (2011): A novel interaction of CLN3 with
nonmuscle myosin-IIB and defects in cell motility of Cln3(-/-) cells. Exp Cell Res
317, 51-69.
Geuze, H. J., Stoorvogel, W., Strous, G. J., Slot, J. W., Bleekemolen, J. E., and Mellman, I.
(1988): Sorting of mannose 6-phosphate receptors and lysosomal membrane
proteins in endocytic vesicles. J Cell Biol 107, 2491-501.
Ghai, R., Bugarcic, A., Liu, H., Norwood, S. J., Skeldal, S., Coulson, E. J., Li, S. S.,
Teasdale, R. D., and Collins, B. M. (2013): Structural basis for endosomal
trafficking of diverse transmembrane cargos by PX-FERM proteins. Proc Natl Acad
Sci U S A 110, E643-52.
Gifford, J. L., Walsh, M. P., and Vogel, H. J. (2007): Structures and metal-ion-binding
properties of the Ca2+-binding helix-loop-helix EF-hand motifs. Biochem J 405,
199-221.
Gilchrist, A., Au, C. E., Hiding, J., Bell, A. W., Fernandez-Rodriguez, J., Lesimple, S.,
Nagaya, H., Roy, L., Gosline, S. J., Hallett, M., Paiement, J., Kearney, R. E.,
Nilsson, T., and Bergeron, J. J. (2006): Quantitative proteomics analysis of the
secretory pathway. Cell 127, 1265-81.
Giugliani, R., Federhen, A., Vairo, F., Vanzella, C., Pasqualim, G., da Silva, L. M.,
Giugliani, L., de Boer, A. P., de Souza, C. F., Matte, U., and Baldo, G. (2016):
Emerging drugs for the treatment of mucopolysaccharidoses. Expert Opin Emerg
Drugs, 1-18.
Goda, Y., and Pfeffer, S. R. (1988): Selective recycling of the mannose 6-phosphate/IGF-II
receptor to the trans Golgi network in vitro. Cell 55, 309-20.
Goebel, M., Stengel, A., Wang, L., and Tache, Y. (2011): Central nesfatin-1 reduces the
nocturnal food intake in mice by reducing meal size and increasing inter-meal
intervals. Peptides 32, 36-43.
Gokool, S., Tattersall, D., and Seaman, M. N. (2007): EHD1 interacts with retromer to
stabilize SNX1 tubules and facilitate endosome-to-Golgi retrieval. Traffic 8, 187386.
Golabek, A. A., Kida, E., Walus, M., Kaczmarski, W., Michalewski, M., and Wisniewski,
K. E. (2000): CLN3 protein regulates lysosomal pH and alters intracellular
processing of Alzheimer's amyloid-beta protein precursor and cathepsin D in human
cells. Mol Genet Metab 70, 203-13.
Gomez, T. S., and Billadeau, D. D. (2009): A FAM21-containing WASH complex
regulates retromer-dependent sorting. Dev Cell 17, 699-711.
Gonatas, N. K., Steiber, A., Kim, S. U., Graham, D. I., and Avrameas, S. (1975):
Internalization of neuronal plasma membrane ricin receptors into the Golgi
apparatus. Exp Cell Res 94, 426-31.
207
Grabe, M., and Oster, G. (2001): Regulation of organelle acidity. J Gen Physiol 117, 32944.
Grohovaz, F., Bossi, M., Pezzati, R., Meldolesi, J., and Tarelli, F. T. (1996): High
resolution ultrastructural mapping of total calcium: electron spectroscopic
imaging/electron energy loss spectroscopy analysis of a physically/chemically
processed nerve-muscle preparation. Proc Natl Acad Sci U S A 93, 4799-803.
Grubman, A., Lidgerwood, G. E., Duncan, C., Bica, L., Tan, J. L., Parker, S. J.,
Caragounis, A., Meyerowitz, J., Volitakis, I., Moujalled, D., Liddell, J. R., Hickey,
J. L., Horne, M., Longmuir, S., Koistinaho, J., Donnelly, P. S., Crouch, P. J.,
Tammen, I., White, A. R., and Kanninen, K. M. (2014a): Deregulation of
subcellular biometal homeostasis through loss of the metal transporter, Zip7, in a
childhood neurodegenerative disorder. Acta Neuropathol Commun 2, 25.
Grubman, A., Pollari, E., Duncan, C., Caragounis, A., Blom, T., Volitakis, I., Wong, A.,
Cooper, J., Crouch, P. J., Koistinaho, J., Jalanko, A., White, A. R., and Kanninen,
K. M. (2014b): Deregulation of biometal homeostasis: the missing link for neuronal
ceroid lipofuscinoses? Metallomics 6, 932-43.
Gu, X. (1998): Early metazoan divergence was about 830 million years ago. J Mol Evol 47,
369-71.
Guichard, A., Nizet, V., and Bier, E. (2014): RAB11-mediated trafficking in host-pathogen
interactions. Nat Rev Microbiol 12, 624-34.
Gustafsen, C., Glerup, S., Pallesen, L. T., Olsen, D., Andersen, O. M., Nykjaer, A.,
Madsen, P., and Petersen, C. M. (2013): Sortilin and SorLA display distinct roles in
processing and trafficking of amyloid precursor protein. J Neurosci 33, 64-71.
Haddad, S. E., Khoury, M., Daoud, M., Kantar, R., Harati, H., Mousallem, T., Alzate, O.,
Meyer, B., and Boustany, R. M. (2012): CLN5 and CLN8 protein association with
ceramide synthase: biochemical and proteomic approaches. Electrophoresis 33,
3798-809.
Hales, C. M., Vaerman, J. P., and Goldenring, J. R. (2002): Rab11 family interacting
protein 2 associates with Myosin Vb and regulates plasma membrane recycling. J
Biol Chem 277, 50415-21.
Haltia, M. (2003): The neuronal ceroid-lipofuscinoses. J Neuropathol Exp Neurol 62, 1-13.
Hancock, M. K., Haskins, D. J., Sun, G., and Dahms, N. M. (2002): Identification of
residues essential for carbohydrate recognition by the insulin-like growth factor
II/mannose 6-phosphate receptor. J Biol Chem 277, 11255-64.
Hanna, J., Carroll, K., and Pfeffer, S. R. (2002): Identification of residues in TIP47
essential for Rab9 binding. Proc Natl Acad Sci U S A 99, 7450-4.
Hannover, A. (1843): Mikrokopiske undersögelser af nervesystemet. Det kongelige Danske
Videnskabernes Selskabs Naturvidenskabelige og Mathematiske Afhandlinger 10, 1112.
Harbour, M. E., Breusegem, S. Y., Antrobus, R., Freeman, C., Reid, E., and Seaman, M. N.
(2010): The cargo-selective retromer complex is a recruiting hub for protein
complexes that regulate endosomal tubule dynamics. J Cell Sci 123, 3703-17.
Harterink, M., Port, F., Lorenowicz, M. J., McGough, I. J., Silhankova, M., Betist, M. C.,
van Weering, J. R., van Heesbeen, R. G., Middelkoop, T. C., Basler, K., Cullen, P.
J., and Korswagen, H. C. (2011): A SNX3-dependent retromer pathway mediates
208
retrograde transport of the Wnt sorting receptor Wntless and is required for Wnt
secretion. Nat Cell Biol 13, 914-23.
Haskell, R. E., Carr, C. J., Pearce, D. A., Bennett, M. J., and Davidson, B. L. (2000): Batten
disease: evaluation of CLN3 mutations on protein localization and function. Hum
Mol Genet 9, 735-44.
Haze, K., Yoshida, H., Yanagi, H., Yura, T., and Mori, K. (1999): Mammalian transcription
factor ATF6 is synthesized as a transmembrane protein and activated by proteolysis
in response to endoplasmic reticulum stress. Mol Biol Cell 10, 3787-99.
Heard, J. M., Bruyere, J., Roy, E., Bigou, S., Ausseil, J., and Vitry, S. (2010): Storage
problems in lysosomal diseases. Biochem Soc Trans 38, 1442-7.
Heinonen, O., Salonen, T., Jalanko, A., Peltonen, L., and Copp, A. (2000): CLN-1 and
CLN-5, genes for infantile and variant late infantile neuronal ceroid lipofuscinoses,
are expressed in the embryonic human brain. J Comp Neurol 426, 406-12.
Hierro, A., Rojas, A. L., Rojas, R., Murthy, N., Effantin, G., Kajava, A. V., Steven, A. C.,
Bonifacino, J. S., and Hurley, J. H. (2007): Functional architecture of the retromer
cargo-recognition complex. Nature 449, 1063-7.
Hirst, J., Barlow, L. D., Francisco, G. C., Sahlender, D. A., Seaman, M. N., Dacks, J. B.,
and Robinson, M. S. (2011): The fifth adaptor protein complex. PLoS Biol 9,
e1001170.
Holmberg, V., Jalanko, A., Isosomppi, J., Fabritius, A. L., Peltonen, L., and Kopra, O.
(2004): The mouse ortholog of the neuronal ceroid lipofuscinosis CLN5 gene
encodes a soluble lysosomal glycoprotein expressed in the developing brain.
Neurobiol Dis 16, 29-40.
Holmberg, V., Lauronen, L., Autti, T., Santavuori, P., Savukoski, M., Uvebrant, P.,
Hofman, I., Peltonen, L., and Jarvela, I. (2000): Phenotype-genotype correlation in
eight patients with Finnish variant late infantile NCL (CLN5). Neurology 55, 57981.
Holopainen, J. M., Saarikoski, J., Kinnunen, P. K., and Jarvela, I. (2001): Elevated
lysosomal pH in neuronal ceroid lipofuscinoses (NCLs). Eur J Biochem 268, 58516.
Hong, Z., Yang, Y., Zhang, C., Niu, Y., Li, K., Zhao, X., and Liu, J. J. (2009): The
retromer component SNX6 interacts with dynactin p150(Glued) and mediates
endosome-to-TGN transport. Cell Res 19, 1334-49.
Horiuchi, H., Lippe, R., McBride, H. M., Rubino, M., Woodman, P., Stenmark, H., Rybin,
V., Wilm, M., Ashman, K., Mann, M., and Zerial, M. (1997): A novel Rab5
GDP/GTP exchange factor complexed to Rabaptin-5 links nucleotide exchange to
effector recruitment and function. Cell 90, 1149-59.
Hueck, W. (1912): Pigmentstudien. Beitr. Pathol. Anat., 68–232.
Hunker, C. M., Galvis, A., Kruk, I., Giambini, H., Veisaga, M. L., and Barbieri, M. A.
(2006): Rab5-activating protein 6, a novel endosomal protein with a role in
endocytosis. Biochem Biophys Res Commun 340, 967-75.
Huotari, J., and Helenius, A. (2011): Endosome maturation. Embo J 30, 3481-500.
Ikura, M. (1996): Calcium binding and conformational response in EF-hand proteins.
Trends Biochem Sci 21, 14-7.
209
Islam, A., Adamik, B., Hawari, F. I., Ma, G., Rouhani, F. N., Zhang, J., and Levine, S. J.
(2006): Extracellular TNFR1 release requires the calcium-dependent formation of a
nucleobindin 2-ARTS-1 complex. J Biol Chem 281, 6860-73.
Isosomppi, J., Vesa, J., Jalanko, A., and Peltonen, L. (2002): Lysosomal localization of the
neuronal ceroid lipofuscinosis CLN5 protein. Hum Mol Genet 11, 885-91.
Jahn, R., and Scheller, R. H. (2006): SNAREs--engines for membrane fusion. Nat Rev Mol
Cell Biol 7, 631-43.
Janes, R. W., Munroe, P. B., Mitchison, H. M., Gardiner, R. M., Mole, S. E., and Wallace,
B. A. (1996): A model for Batten disease protein CLN3: functional implications
from homology and mutations. FEBS Lett 399, 75-7.
Jao, C. C., Hegde, B. G., Gallop, J. L., Hegde, P. B., McMahon, H. T., Haworth, I. S., and
Langen, R. (2010): Roles of amphipathic helices and the bin/amphiphysin/rvs
(BAR) domain of endophilin in membrane curvature generation. J Biol Chem 285,
20164-70.
Jarvela, I., Lehtovirta, M., Tikkanen, R., Kyttala, A., and Jalanko, A. (1999): Defective
intracellular transport of CLN3 is the molecular basis of Batten disease (JNCL).
Hum Mol Genet 8, 1091-8.
Jarvela, I., Sainio, M., Rantamaki, T., Olkkonen, V. M., Carpen, O., Peltonen, L., and
Jalanko, A. (1998): Biosynthesis and intracellular targeting of the CLN3 protein
defective in Batten disease. Hum Mol Genet 7, 85-90.
Jay, P., Rougeulle, C., Massacrier, A., Moncla, A., Mattei, M. G., Malzac, P., Roeckel, N.,
Taviaux, S., Lefranc, J. L., Cau, P., Berta, P., Lalande, M., and Muscatelli, F.
(1997): The human necdin gene, NDN, is maternally imprinted and located in the
Prader-Willi syndrome chromosomal region. Nat Genet 17, 357-61.
Jia, D., Gomez, T. S., Billadeau, D. D., and Rosen, M. K. (2012): Multiple repeat elements
within the FAM21 tail link the WASH actin regulatory complex to the retromer.
Mol Biol Cell 23, 2352-61.
Johannes, L., and Wunder, C. (2011): Retrograde transport: two (or more) roads diverged
in an endosomal tree? Traffic 12, 956-62.
Johansson, M., Rocha, N., Zwart, W., Jordens, I., Janssen, L., Kuijl, C., Olkkonen, V. M.,
and Neefjes, J. (2007): Activation of endosomal dynein motors by stepwise
assembly of Rab7-RILP-p150Glued, ORP1L, and the receptor betalll spectrin. J
Cell Biol 176, 459-71.
Johnson, K. F., and Kornfeld, S. (1992a): The cytoplasmic tail of the mannose 6phosphate/insulin-like growth factor-II receptor has two signals for lysosomal
enzyme sorting in the Golgi. J Cell Biol 119, 249-57.
Johnson, K. F., and Kornfeld, S. (1992b): A His-Leu-Leu sequence near the carboxyl
terminus of the cytoplasmic domain of the cation-dependent mannose 6-phosphate
receptor is necessary for the lysosomal enzyme sorting function. J Biol Chem 267,
17110-5.
Jolly, R. D., Douglas, B. V., Davey, P. M., and Roiri, J. E. (1995): Lipofuscin in bovine
muscle and brain: a model for studying age pigment. Gerontology 41 Suppl 2, 28395.
Jung, T., Bader, N., and Grune, T. (2007): Lipofuscin: formation, distribution, and
metabolic consequences. Ann N Y Acad Sci 1119, 97-111.
210
Kaczmarski, W., Wisniewski, K. E., Golabek, A., Kaczmarski, A., Kida, E., and
Michalewski, M. (1999): Studies of membrane association of CLN3 protein. Mol
Genet Metab 66, 261-4.
Kalnina, Z., Silina, K., Bruvere, R., Gabruseva, N., Stengrevics, A., Barnikol-Watanabe, S.,
Leja, M., and Line, A. (2009): Molecular characterisation and expression analysis
of SEREX-defined antigen NUCB2 in gastric epithelium, gastritis and gastric
cancer. Eur J Histochem 53, 7-18.
Kanai, Y., Isonishi, S., Katagiri, T., Koizumi, T., Miura, K., and Kurosawa, Y. (1990):
Early induction of anti-double-stranded DNA antibodies in lupus-prone MRL mice
inoculated with Ly-24+ cells cloned from lymph node cells of an MRL/Mp-lpr/lpr
mouse: possible effect of putative cytokines produced by cloned Ly-24+ cells.
Immunol Lett 24, 49-55.
Kanai, Y., Katagiri, T., Mori, S., and Kubota, T. (1986): An established MRL/Mp-lpr/lpr
cell line with null cell properties produces a B cell differentiation factor(s) that
promotes anti-single-stranded DNA antibody production in MRL spleen cell
culture. Int Arch Allergy Appl Immunol 81, 92-4.
Kanai, Y., Kyuwa, S., Miura, K., and Kurosawa, Y. (1995a): Induction and natural
occurrence of serum nucleosomal DNA in autoimmune MRL/lpr/lpr mice: its
relation to apoptosis in the thymus. Immunol Lett 46, 207-14.
Kanai, Y., Takeda, O., Miura, K., Amagai, M., Kaneko, T., Kubota, T., Tanuma, S., and
Kurosawa, Y. (1995b): Induction of autoantibodies in normal mice by injection of
nucleobindin and natural occurrence of antibodies against nucleobindin in
autoimmune MRL/lpr/lpr mice. Immunol Lett 45, 35-42.
Kanai, Y., Takeda, O., Miura, K., and Kurosawa, Y. (1993): Novel autoimmune
phenomena induced in vivo by a new DNA binding protein Nuc: a study on MRL/n
mice. Immunol Lett 39, 83-9.
Kanai, Y., and Tanuma, S. (1992): Purification of a novel B cell growth and differentiation
factor associated with lupus syndrome. Immunol Lett 32, 43-8.
Kanninen, K. M., Grubman, A., Meyerowitz, J., Duncan, C., Tan, J. L., Parker, S. J.,
Crouch, P. J., Paterson, B. M., Hickey, J. L., Donnelly, P. S., Volitakis, I., Tammen,
I., Palmer, D. N., and White, A. R. (2013): Increased zinc and manganese in parallel
with neurodegeneration, synaptic protein changes and activation of Akt/GSK3
signaling in ovine CLN6 neuronal ceroid lipofuscinosis. PLoS One 8, e58644.
Kanuru, M., Raman, R., and Aradhyam, G. K. (2013): Serine protease activity of calnuc:
regulation by Zn2+ and G proteins. J Biol Chem.
Kanuru, M., Samuel, J. J., Balivada, L. M., and Aradhyam, G. K. (2009): Ion-binding
properties of Calnuc, Ca2+ versus Mg2+--Calnuc adopts additional and unusual
Ca2+-binding sites upon interaction with G-protein. Febs J 276, 2529-46.
Kapoor, N., Gupta, R., Menon, S. T., Folta-Stogniew, E., Raleigh, D. P., and Sakmar, T. P.
(2010): Nucleobindin 1 is a calcium-regulated guanine nucleotide dissociation
inhibitor of G{alpha}i1. J Biol Chem 285, 31647-60.
Karabinos, A., Bhattacharya, D., Morys-Wortmann, C., Kroll, K., Hirschfeld, G., Kratzin,
H. D., Barnikol-Watanabe, S., and Hilschmann, N. (1996): The divergent domains
of the NEFA and nucleobindin proteins are derived from an EF-hand ancestor. Mol
Biol Evol 13, 990-8.
211
Kerr, M. C., Bennetts, J. S., Simpson, F., Thomas, E. C., Flegg, C., Gleeson, P. A.,
Wicking, C., and Teasdale, R. D. (2005): A novel mammalian retromer component,
Vps26B. Traffic 6, 991-1001.
Kida, E., Kaczmarski, W., Golabek, A. A., Kaczmarski, A., Michalewski, M., and
Wisniewski, K. E. (1999): Analysis of intracellular distribution and trafficking of
the CLN3 protein in fusion with the green fluorescent protein in vitro. Mol Genet
Metab 66, 265-71.
Kim, H. S., Kim, E. M., Lee, J. P., Park, C. H., Kim, S., Seo, J. H., Chang, K. A., Yu, E.,
Jeong, S. J., Chong, Y. H., and Suh, Y. H. (2003a): C-terminal fragments of
amyloid precursor protein exert neurotoxicity by inducing glycogen synthase
kinase-3beta expression. Faseb J 17, 1951-3.
Kim, Y., Ramirez-Montealegre, D., and Pearce, D. A. (2003b): A role in vacuolar arginine
transport for yeast Btn1p and for human CLN3, the protein defective in Batten
disease. Proc Natl Acad Sci U S A 100, 15458-62.
Kitzmuller, C., Haines, R. L., Codlin, S., Cutler, D. F., and Mole, S. E. (2008): A function
retained by the common mutant CLN3 protein is responsible for the late onset of
juvenile neuronal ceroid lipofuscinosis. Hum Mol Genet 17, 303-12.
Kjolby, M., Andersen, O. M., Breiderhoff, T., Fjorback, A. W., Pedersen, K. M., Madsen,
P., Jansen, P., Heeren, J., Willnow, T. E., and Nykjaer, A. (2010): Sort1, encoded
by the cardiovascular risk locus 1p13.3, is a regulator of hepatic lipoprotein export.
Cell Metab 12, 213-23.
Koivu, T., Laitinen, S., Riento, K., and Olkkonen, V. M. (1997): Sequence of a human
cDNA encoding Cab45, a Ca2+-binding protein with six EF-hand motifs. DNA Seq
7, 217-20.
Kollmann, K., Mutenda, K. E., Balleininger, M., Eckermann, E., von Figura, K., Schmidt,
B., and Lubke, T. (2005): Identification of novel lysosomal matrix proteins by
proteome analysis. Proteomics 5, 3966-78.
Konczol, K., Bodnar, I., Zelena, D., Pinter, O., Papp, R. S., Palkovits, M., Nagy, G. M., and
Toth, Z. E. (2010): Nesfatin-1/NUCB2 may participate in the activation of the
hypothalamic-pituitary-adrenal axis in rats. Neurochem Int 57, 189-97.
Koneff, H. (1886): Beiträge zur Kenntniss der Nervenzellen den peripheren Ganglien. Mitt.
Naturforsch. Gesellsch 44, 13–14.
Kornfeld, S., and Mellman, I. (1989): The biogenesis of lysosomes. Annu Rev Cell Biol 5,
483-525.
Koumandou, V. L., Klute, M. J., Herman, E. K., Nunez-Miguel, R., Dacks, J. B., and Field,
M. C. (2011): Evolutionary reconstruction of the retromer complex and its function
in Trypanosoma brucei. J Cell Sci 124, 1496-509.
Kretsinger, R. H. (1987): Calcium coordination and the calmodulin fold: divergent versus
convergent evolution. Cold Spring Harb Symp Quant Biol 52, 499-510.
Kroll, K. A., Otte, S., Hirschfeld, G., Barnikol-Watanabe, S., Gotz, H., Sternbach, H.,
Kratzin, H. D., Barnikol, H. U., and Hilschmann, N. (1999): Heterologous
overexpression of human NEFA and studies on the two EF-hand calcium-binding
sites. Biochem Biophys Res Commun 260, 1-8.
Kubota, T., Miyauchi, M., Miura, K., Hirokawa, G., Awaya, A., Miyasaka, N., Kurosawa,
Y., Kanai, Y., and Maruyama, K. (1998): Upregulation of nucleobindin expression
in human-activated lymphocytes and non-Hodgkin's lymphoma. Pathol Int 48, 22-8.
212
Kurten, R. C., Eddington, A. D., Chowdhury, P., Smith, R. D., Davidson, A. D., and Shank,
B. B. (2001): Self-assembly and binding of a sorting nexin to sorting endosomes. J
Cell Sci 114, 1743-56.
Kutay, U., Hartmann, E., and Rapoport, T. A. (1993): A class of membrane proteins with a
C-terminal anchor. Trends Cell Biol 3, 72-5.
Kyttala, A., Ihrke, G., Vesa, J., Schell, M. J., and Luzio, J. P. (2004): Two motifs target
Batten disease protein CLN3 to lysosomes in transfected nonneuronal and neuronal
cells. Mol Biol Cell 15, 1313-23.
LaFerla, F. M. (2002): Calcium dyshomeostasis and intracellular signalling in Alzheimer's
disease. Nat Rev Neurosci 3, 862-72.
Lambright, D. G., Sondek, J., Bohm, A., Skiba, N. P., Hamm, H. E., and Sigler, P. B.
(1996): The 2.0 A crystal structure of a heterotrimeric G protein. Nature 379, 311-9.
Lane, S. C., Jolly, R. D., Schmechel, D. E., Alroy, J., and Boustany, R. M. (1996):
Apoptosis as the mechanism of neurodegeneration in Batten's disease. J Neurochem
67, 677-83.
Laplante, M., and Sabatini, D. M. (2012): mTOR signaling in growth control and disease.
Cell 149, 274-93.
Larkin, H., Ribeiro, M. G., and Lavoie, C. (2013): Topology and membrane anchoring of
the lysosomal storage disease-related protein CLN5. Hum Mutat 34, 1688-97.
Lauffer, B. E., Melero, C., Temkin, P., Lei, C., Hong, W., Kortemme, T., and von Zastrow,
M. (2010): SNX27 mediates PDZ-directed sorting from endosomes to the plasma
membrane. J Cell Biol 190, 565-74.
Laurent-Matha, V., Derocq, D., Prebois, C., Katunuma, N., and Liaudet-Coopman, E.
(2006): Processing of human cathepsin D is independent of its catalytic function
and auto-activation: involvement of cathepsins L and B. J Biochem 139, 363-71.
Lavoie, C., Meerloo, T., Lin, P., and Farquhar, M. G. (2002): Calnuc, an EF-hand Ca(2+)binding protein, is stored and processed in the Golgi and secreted by the
constitutive-like pathway in AtT20 cells. Mol Endocrinol 16, 2462-74.
Lebrun, A. H., Storch, S., Ruschendorf, F., Schmiedt, M. L., Kyttala, A., Mole, S. E.,
Kitzmuller, C., Saar, K., Mewasingh, L. D., Boda, V., Kohlschutter, A., Ullrich, K.,
Braulke, T., and Schulz, A. (2009): Retention of lysosomal protein CLN5 in the
endoplasmic reticulum causes neuronal ceroid lipofuscinosis in Asian sibship. Hum
Mutat 30, E651-61.
Leclerc, P., Biarc, J., St-Onge, M., Gilbert, C., Dussault, A. A., Laflamme, C., and Pouliot,
M. (2008): Nucleobindin co-localizes and associates with cyclooxygenase (COX)-2
in human neutrophils. PLoS One 3, e2229.
Lee, A. Y., Gulnik, S. V., and Erickson, J. W. (1998): Conformational switching in an
aspartic proteinase. Nat Struct Biol 5, 866-71.
Lefrancois, S., Zeng, J., Hassan, A. J., Canuel, M., and Morales, C. R. (2003): The
lysosomal trafficking of sphingolipid activator proteins (SAPs) is mediated by
sortilin. Embo J 22, 6430-7.
Leung, K. F., Baron, R., and Seabra, M. C. (2006): Thematic review series: lipid
posttranslational modifications. geranylgeranylation of Rab GTPases. J Lipid Res
47, 467-75.
Li, H., and Villalobo, A. (2002): Evidence for the direct interaction between calmodulin
and the human epidermal growth factor receptor. Biochem J 362, 499-505.
213
Li, Q. C., Wang, H. Y., Chen, X., Guan, H. Z., and Jiang, Z. Y. (2010): Fasting plasma
levels of nesfatin-1 in patients with type 1 and type 2 diabetes mellitus and the
nutrient-related fluctuation of nesfatin-1 level in normal humans. Regul Pept 159,
72-7.
Lin, P., Fischer, T., Lavoie, C., Huang, H., and Farquhar, M. G. (2009): Calnuc plays a role
in dynamic distribution of Galphai but not Gbeta subunits and modulates ACTH
secretion in AtT-20 neuroendocrine secretory cells. Mol Neurodegener 4, 15.
Lin, P., Fischer, T., Weiss, T., and Farquhar, M. G. (2000): Calnuc, an EF-hand Ca(2+)
binding protein, specifically interacts with the C-terminal alpha5-helix of
G(alpha)i3. Proc Natl Acad Sci U S A 97, 674-9.
Lin, P., Le-Niculescu, H., Hofmeister, R., McCaffery, J. M., Jin, M., Hennemann, H.,
McQuistan, T., De Vries, L., and Farquhar, M. G. (1998): The mammalian calciumbinding protein, nucleobindin (CALNUC), is a Golgi resident protein. J Cell Biol
141, 1515-27.
Lin, P., Li, F., Zhang, Y. W., Huang, H., Tong, G., Farquhar, M. G., and Xu, H. (2007):
Calnuc binds to Alzheimer's beta-amyloid precursor protein and affects its
biogenesis. J Neurochem 100, 1505-14.
Lin, P., Yao, Y., Hofmeister, R., Tsien, R. Y., and Farquhar, M. G. (1999): Overexpression
of CALNUC (nucleobindin) increases agonist and thapsigargin releasable Ca2+
storage in the Golgi. J Cell Biol 145, 279-89.
Lippe, R., Miaczynska, M., Rybin, V., Runge, A., and Zerial, M. (2001): Functional
synergy between Rab5 effector Rabaptin-5 and exchange factor Rabex-5 when
physically associated in a complex. Mol Biol Cell 12, 2219-28.
Liu, T. T., Gomez, T. S., Sackey, B. K., Billadeau, D. D., and Burd, C. G. (2012): Rab
GTPase regulation of retromer-mediated cargo export during endosome maturation.
Mol Biol Cell 23, 2505-15.
Llado, A., Timpson, P., Vila de Muga, S., Moreto, J., Pol, A., Grewal, T., Daly, R. J.,
Enrich, C., and Tebar, F. (2008): Protein kinase Cdelta and calmodulin regulate
epidermal growth factor receptor recycling from early endosomes through Arp2/3
complex and cortactin. Mol Biol Cell 19, 17-29.
Lobel, P., Dahms, N. M., and Kornfeld, S. (1988): Cloning and sequence analysis of the
cation-independent mannose 6-phosphate receptor. J Biol Chem 263, 2563-70.
Lodhi, I. J., Chiang, S. H., Chang, L., Vollenweider, D., Watson, R. T., Inoue, M., Pessin,
J. E., and Saltiel, A. R. (2007): Gapex-5, a Rab31 guanine nucleotide exchange
factor that regulates Glut4 trafficking in adipocytes. Cell Metab 5, 59-72.
Lojewski, X., Staropoli, J. F., Biswas-Legrand, S., Simas, A. M., Haliw, L., Selig, M. K.,
Coppel, S. H., Goss, K. A., Petcherski, A., Chandrachud, U., Sheridan, S. D.,
Lucente, D., Sims, K. B., Gusella, J. F., Sondhi, D., Crystal, R. G., Reinhardt, P.,
Sterneckert, J., Scholer, H., Haggarty, S. J., Storch, A., Hermann, A., and Cotman,
S. L. (2013): Human iPSC models of neuronal ceroid lipofuscinosis capture distinct
effects of TPP1 and CLN3 mutations on the endocytic pathway. Hum Mol Genet 23,
2005-22.
Lombardi, D., Soldati, T., Riederer, M. A., Goda, Y., Zerial, M., and Pfeffer, S. R. (1993):
Rab9 functions in transport between late endosomes and the trans Golgi network.
Embo J 12, 677-82.
214
Luiro, K., Kopra, O., Lehtovirta, M., and Jalanko, A. (2001): CLN3 protein is targeted to
neuronal synapses but excluded from synaptic vesicles: new clues to Batten disease.
Hum Mol Genet 10, 2123-31.
Luiro, K., Yliannala, K., Ahtiainen, L., Maunu, H., Jarvela, I., Kyttala, A., and Jalanko, A.
(2004): Interconnections of CLN3, Hook1 and Rab proteins link Batten disease to
defects in the endocytic pathway. Hum Mol Genet 13, 3017-27.
Lyly, A., Marjavaara, S. K., Kyttala, A., Uusi-Rauva, K., Luiro, K., Kopra, O., Martinez, L.
O., Tanhuanpaa, K., Kalkkinen, N., Suomalainen, A., Jauhiainen, M., and Jalanko,
A. (2008): Deficiency of the INCL protein Ppt1 results in changes in ectopic F1ATP synthase and altered cholesterol metabolism. Hum Mol Genet 17, 1406-17.
Lyly, A., von Schantz, C., Heine, C., Schmiedt, M. L., Sipila, T., Jalanko, A., and Kyttala,
A. (2009): Novel interactions of CLN5 support molecular networking between
Neuronal Ceroid Lipofuscinosis proteins. BMC Cell Biol 10, 83.
MacLeod, P. M., Dolman, C. L., Chang, E., Applegarth, D. A., and Bryant, B. (1976): The
neuronal ceroid lipofuscinoses in British Columbia: a clinical epidemiologic and
ultrastructural study. Birth Defects Orig Artic Ser 12, 289-96.
Maejima, Y., Sedbazar, U., Suyama, S., Kohno, D., Onaka, T., Takano, E., Yoshida, N.,
Koike, M., Uchiyama, Y., Fujiwara, K., Yashiro, T., Horvath, T. L., Dietrich, M.
O., Tanaka, S., Dezaki, K., Oh, I. S., Hashimoto, K., Shimizu, H., Nakata, M., Mori,
M., and Yada, T. (2009): Nesfatin-1-regulated oxytocinergic signaling in the
paraventricular nucleus causes anorexia through a leptin-independent melanocortin
pathway. Cell Metab 10, 355-65.
Mamo, A., Jules, F., Dumaresq-Doiron, K., Costantino, S., and Lefrancois, S. (2012): The
role of Ceroid Lipofuscinosis Neuronal protein-5 (CLN5) in endosomal sorting. Mol
Cell Biol.
Mao, Q., Foster, B. J., Xia, H., and Davidson, B. L. (2003): Membrane topology of CLN3,
the protein underlying Batten disease. FEBS Lett 541, 40-6.
Marcusson, E. G., Horazdovsky, B. F., Cereghino, J. L., Gharakhanian, E., and Emr, S. D.
(1994): The sorting receptor for yeast vacuolar carboxypeptidase Y is encoded by
the VPS10 gene. Cell 77, 579-86.
Mari, M., Bujny, M. V., Zeuschner, D., Geerts, W. J., Griffith, J., Petersen, C. M., Cullen,
P. J., Klumperman, J., and Geuze, H. J. (2008): SNX1 defines an early endosomal
recycling exit for sortilin and mannose 6-phosphate receptors. Traffic 9, 380-93.
Markmann, S., Thelen, M., Cornils, K., Schweizer, M., Brocke-Ahmadinejad, N., Willnow,
T., Heeren, J., Gieselmann, V., Braulke, T., and Kollmann, K. (2015): Lrp1/LDL
Receptor Play Critical Roles in Mannose 6-Phosphate-Independent Lysosomal
Enzyme Targeting. Traffic 16, 743-59.
Marron-Terada, P. G., Hancock, M. K., Haskins, D. J., and Dahms, N. M. (2000):
Recognition of Dictyostelium discoideum lysosomal enzymes is conferred by the
amino-terminal carbohydrate binding site of the insulin-like growth factor
II/mannose 6-phosphate receptor. Biochemistry 39, 2243-53.
Martinez, L. O., Jacquet, S., Esteve, J. P., Rolland, C., Cabezon, E., Champagne, E.,
Pineau, T., Georgeaud, V., Walker, J. E., Terce, F., Collet, X., Perret, B., and
Barbaras, R. (2003): Ectopic beta-chain of ATP synthase is an apolipoprotein A-I
receptor in hepatic HDL endocytosis. Nature 421, 75-9.
215
Maruyama, K., Usami, M., Aizawa, T., and Yoshikawa, K. (1991): A novel brain-specific
mRNA encoding nuclear protein (necdin) expressed in neurally differentiated
embryonal carcinoma cells. Biochem Biophys Res Commun 178, 291-6.
Marz, K. E., Lauer, J. M., and Hanson, P. I. (2003): Defining the SNARE complex binding
surface of alpha-SNAP: implications for SNARE complex disassembly. J Biol
Chem 278, 27000-8.
Matsui, Y., Kikuchi, A., Araki, S., Hata, Y., Kondo, J., Teranishi, Y., and Takai, Y. (1990):
Molecular cloning and characterization of a novel type of regulatory protein (GDI)
for smg p25A, a ras p21-like GTP-binding protein. Mol Cell Biol 10, 4116-22.
Mattson, M. P., and Chan, S. L. (2003): Neuronal and glial calcium signaling in
Alzheimer's disease. Cell Calcium 34, 385-97.
McBride, H. M., Rybin, V., Murphy, C., Giner, A., Teasdale, R., and Zerial, M. (1999):
Oligomeric complexes link Rab5 effectors with NSF and drive membrane fusion via
interactions between EEA1 and syntaxin 13. Cell 98, 377-86.
McCormick, P. J., Dumaresq-Doiron, K., Pluviose, A. S., Pichette, V., Tosato, G., and
Lefrancois, S. (2008): Palmitoylation controls recycling in lysosomal sorting and
trafficking. Traffic 9, 1984-97.
McGough, I. J., and Cullen, P. J. (2011): Recent advances in retromer biology. Traffic 12,
963-71.
McNew, J. A., Parlati, F., Fukuda, R., Johnston, R. J., Paz, K., Paumet, F., Sollner, T. H.,
and Rothman, J. E. (2000): Compartmental specificity of cellular membrane fusion
encoded in SNARE proteins. Nature 407, 153-9.
Medina, D. L., Di Paola, S., Peluso, I., Armani, A., De Stefani, D., Venditti, R.,
Montefusco, S., Scotto-Rosato, A., Prezioso, C., Forrester, A., Settembre, C., Wang,
W., Gao, Q., Xu, H., Sandri, M., Rizzuto, R., De Matteis, M. A., and Ballabio, A.
(2015): Lysosomal calcium signalling regulates autophagy through calcineurin and
TFEB. Nat Cell Biol 17, 288-99.
Meikle, P. J., Hopwood, J. J., Clague, A. E., and Carey, W. F. (1999): Prevalence of
lysosomal storage disorders. Jama 281, 249-54.
Meikle, P. J., Ranieri, E., Simonsen, H., Rozaklis, T., Ramsay, S. L., Whitfield, P. D.,
Fuller, M., Christensen, E., Skovby, F., and Hopwood, J. J. (2004): Newborn
screening for lysosomal storage disorders: clinical evaluation of a two-tier strategy.
Pediatrics 114, 909-16.
Merali, Z., Cayer, C., Kent, P., and Anisman, H. (2008): Nesfatin-1 increases anxiety- and
fear-related behaviors in the rat. Psychopharmacology (Berl) 201, 115-23.
Meresse, S., Ludwig, T., Frank, R., and Hoflack, B. (1990): Phosphorylation of the
cytoplasmic domain of the bovine cation-independent mannose 6-phosphate
receptor. Serines 2421 and 2492 are the targets of a casein kinase II associated to
the Golgi-derived HAI adaptor complex. J Biol Chem 265, 18833-42.
Mermelstein, C. S., Rebello, M. I., Amaral, L. M., and Costa, M. L. (2003): Changes in cell
shape, cytoskeletal proteins and adhesion sites of cultured cells after extracellular
Ca2+ chelation. Braz J Med Biol Res 36, 1111-6.
Metcalf, D. J., Calvi, A. A., Seaman, M., Mitchison, H. M., and Cutler, D. F. (2008): Loss
of the Batten disease gene CLN3 prevents exit from the TGN of the mannose 6phosphate receptor. Traffic 9, 1905-14.
216
Michalak, M., Burns, K., Andrin, C., Mesaeli, N., Jass, G. H., Busaan, J. L., and Opas, M.
(1996): Endoplasmic reticulum form of calreticulin modulates glucocorticoidsensitive gene expression. J Biol Chem 271, 29436-45.
Mitchison, H. M., Lim, M. J., and Cooper, J. D. (2004): Selectivity and types of cell death
in the neuronal ceroid lipofuscinoses. Brain Pathol 14, 86-96.
Miura, K., Hirai, M., Kanai, Y., and Kurosawa, Y. (1996): Organization of the human gene
for nucleobindin (NUC) and its chromosomal assignment to 19q13.2-q13.4.
Genomics 34, 181-6.
Miura, K., Kurosawa, Y., and Kanai, Y. (1994): Calcium-binding activity of nucleobindin
mediated by an EF hand moiety. Biochem Biophys Res Commun 199, 1388-93.
Miura, K., Titani, K., Kurosawa, Y., and Kanai, Y. (1992): Molecular cloning of
nucleobindin, a novel DNA-binding protein that contains both a signal peptide and a
leucine zipper structure. Biochem Biophys Res Commun 187, 375-80.
Mixon, M. B., Lee, E., Coleman, D. E., Berghuis, A. M., Gilman, A. G., and Sprang, S. R.
(1995): Tertiary and quaternary structural changes in Gi alpha 1 induced by GTP
hydrolysis. Science 270, 954-60.
Mochizuki, N., Hibi, M., Kanai, Y., and Insel, P. A. (1995): Interaction of the protein
nucleobindin with G alpha i2, as revealed by the yeast two-hybrid system. FEBS
Lett 373, 155-8.
Moharir, A., Peck, S. H., Budden, T., and Lee, S. Y. (2013): The role of N-glycosylation in
folding, trafficking, and functionality of lysosomal protein CLN5. PLoS One 8,
e74299.
Mole, S., Williams, R., and Goebel, H. (2011): The neuronal ceroid lipofuscinoses (Batten
disease). Oxford University Press.
Moore, S. J., Buckley, D. J., MacMillan, A., Marshall, H. D., Steele, L., Ray, P. N., Nawaz,
Z., Baskin, B., Frecker, M., Carr, S. M., Ives, E., and Parfrey, P. S. (2008): The
clinical and genetic epidemiology of neuronal ceroid lipofuscinosis in
Newfoundland. Clin Genet 74, 213-22.
Morel-Huaux, V. M., Pypaert, M., Wouters, S., Tartakoff, A. M., Jurgan, U., Gevaert, K.,
and Courtoy, P. J. (2002): The calcium-binding protein p54/NEFA is a novel
luminal resident of medial Golgi cisternae that traffics independently of
mannosidase II. Eur J Cell Biol 81, 87-100.
Morris, N. J., Ross, S. A., Lane, W. S., Moestrup, S. K., Petersen, C. M., Keller, S. R., and
Lienhard, G. E. (1998): Sortilin is the major 110-kDa protein in GLUT4 vesicles
from adipocytes. J Biol Chem 273, 3582-7.
Muhammad, A., Flores, I., Zhang, H., Yu, R., Staniszewski, A., Planel, E., Herman, M.,
Ho, L., Kreber, R., Honig, L. S., Ganetzky, B., Duff, K., Arancio, O., and Small, S.
A. (2008): Retromer deficiency observed in Alzheimer's disease causes
hippocampal dysfunction, neurodegeneration, and Abeta accumulation. Proc Natl
Acad Sci U S A 105, 7327-32.
Mullins, C., and Bonifacino, J. S. (2001): The molecular machinery for lysosome
biogenesis. Bioessays 23, 333-43.
Munck Petersen, C., Nielsen, M. S., Jacobsen, C., Tauris, J., Jacobsen, L., Gliemann, J.,
Moestrup, S. K., and Madsen, P. (1999): Propeptide cleavage conditions
sortilin/neurotensin receptor-3 for ligand binding. Embo J 18, 595-604.
217
Munroe, P. B., Mitchison, H. M., O'Rawe, A. M., Anderson, J. W., Boustany, R. M.,
Lerner, T. J., Taschner, P. E., de Vos, N., Breuning, M. H., Gardiner, R. M., and
Mole, S. E. (1997): Spectrum of mutations in the Batten disease gene, CLN3. Am J
Hum Genet 61, 310-6.
Nakada-Tsukui, K., Saito-Nakano, Y., Ali, V., and Nozaki, T. (2005): A retromerlike
complex is a novel Rab7 effector that is involved in the transport of the virulence
factor cysteine protease in the enteric protozoan parasite Entamoeba histolytica. Mol
Biol Cell 16, 5294-303.
Nakayama, S., Moncrief, N. D., and Kretsinger, R. H. (1992): Evolution of EF-hand
calcium-modulated proteins. II. Domains of several subfamilies have diverse
evolutionary histories. J Mol Evol 34, 416-48.
Narayan, S. B., Rakheja, D., Tan, L., Pastor, J. V., and Bennett, M. J. (2006): CLN3P, the
Batten's disease protein, is a novel palmitoyl-protein Delta-9 desaturase. Ann Neurol
60, 570-7.
Narayan, S. B., Tan, L., and Bennett, M. J. (2008): Intermediate levels of neuronal
palmitoyl-protein Delta-9 desaturase in heterozygotes for murine Batten disease.
Mol Genet Metab 93, 89-91.
Nash, P. D., Opas, M., and Michalak, M. (1994): Calreticulin: not just another calciumbinding protein. Mol Cell Biochem 135, 71-8.
Nesselhut, J., Jurgan, U., Onken, E., Gotz, H., Barnikol, H. U., Hirschfeld, G., BarnikolWatanabe, S., and Hilschmann, N. (2001): Golgi retention of human protein NEFA
is mediated by its N-terminal Leu/Ile-rich region. FEBS Lett 509, 469-75.
Ni, X., and Morales, C. R. (2006): The lysosomal trafficking of acid sphingomyelinase is
mediated by sortilin and mannose 6-phosphate receptor. Traffic 7, 889-902.
Nielsen, M. S., Gustafsen, C., Madsen, P., Nyengaard, J. R., Hermey, G., Bakke, O., Mari,
M., Schu, P., Pohlmann, R., Dennes, A., and Petersen, C. M. (2007a): Sorting by
the cytoplasmic domain of the amyloid precursor protein binding receptor SorLA.
Mol Cell Biol 27, 6842-51.
Nielsen, M. S., Jacobsen, C., Olivecrona, G., Gliemann, J., and Petersen, C. M. (1999):
Sortilin/neurotensin receptor-3 binds and mediates degradation of lipoprotein lipase.
J Biol Chem 274, 8832-6.
Nielsen, M. S., Madsen, P., Christensen, E. I., Nykjaer, A., Gliemann, J., Kasper, D.,
Pohlmann, R., and Petersen, C. M. (2001): The sortilin cytoplasmic tail conveys
Golgi-endosome transport and binds the VHS domain of the GGA2 sorting protein.
Embo J 20, 2180-90.
Nielsen, R., Courtoy, P. J., Jacobsen, C., Dom, G., Lima, W. R., Jadot, M., Willnow, T. E.,
Devuyst, O., and Christensen, E. I. (2007b): Endocytosis provides a major
alternative pathway for lysosomal biogenesis in kidney proximal tubular cells. Proc
Natl Acad Sci U S A 104, 5407-12.
Niinobe, M., Koyama, K., and Yoshikawa, K. (2000): Cellular and subcellular localization
of necdin in fetal and adult mouse brain. Dev Neurosci 22, 310-9.
Niphakis, M. J., Lum, K. M., Cognetta, A. B., 3rd, Correia, B. E., Ichu, T. A., Olucha, J.,
Brown, S. J., Kundu, S., Piscitelli, F., Rosen, H., and Cravatt, B. F. (2015): A
Global Map of Lipid-Binding Proteins and Their Ligandability in Cells. Cell 161,
1668-80.
218
Norwood, S. J., Shaw, D. J., Cowieson, N. P., Owen, D. J., Teasdale, R. D., and Collins, B.
M. (2010): Assembly and solution structure of the core retromer protein complex.
Traffic 12, 56-71.
Nothwehr, S. F., Ha, S. A., and Bruinsma, P. (2000): Sorting of yeast membrane proteins
into an endosome-to-Golgi pathway involves direct interaction of their cytosolic
domains with Vps35p. J Cell Biol 151, 297-310.
Nugent, T., Mole, S. E., and Jones, D. T. (2008): The transmembrane topology of Batten
disease protein CLN3 determined by consensus computational prediction
constrained by experimental data. FEBS Lett 582, 1019-24.
Nykjaer, A., Lee, R., Teng, K. K., Jansen, P., Madsen, P., Nielsen, M. S., Jacobsen, C.,
Kliemannel, M., Schwarz, E., Willnow, T. E., Hempstead, B. L., and Petersen, C.
M. (2004): Sortilin is essential for proNGF-induced neuronal cell death. Nature
427, 843-8.
Oh, I. S., Shimizu, H., Satoh, T., Okada, S., Adachi, S., Inoue, K., Eguchi, H., Yamamoto,
M., Imaki, T., Hashimoto, K., Tsuchiya, T., Monden, T., Horiguchi, K., Yamada,
M., and Mori, M. (2006): Identification of nesfatin-1 as a satiety molecule in the
hypothalamus. Nature 443, 709-12.
Olsnes, S., and Pihl, A. (1972): Ricin - a potent inhibitor of protein synthesis. FEBS Lett
20, 327-329.
Olson, L. J., Castonguay, A. C., Lasanajak, Y., Peterson, F. C., Cummings, R. D., Smith,
D. F., and Dahms, N. M. (2015): Identification of a fourth mannose 6-phosphate
binding site in the cation-independent mannose 6-phosphate receptor. Glycobiology.
Orsel, J. G., Sincock, P. M., Krise, J. P., and Pfeffer, S. R. (2000): Recognition of the 300kDa mannose 6-phosphate receptor cytoplasmic domain by 47-kDa tail-interacting
protein. Proc Natl Acad Sci U S A 97, 9047-51.
Osei-Hyiaman, N., Encinas (2011): Fasting co-supresses Nesfatin-1 and GPR12 in Mouse
Hypothlamic Appetite Center: Implications for Energy Metabolism. Endocrine
Reviews 32, 2-300.
Padilla-Lopez, S., and Pearce, D. A. (2006): Saccharomyces cerevisiae lacking Btn1p
modulate vacuolar ATPase activity to regulate pH imbalance in the vacuole. J Biol
Chem 281, 10273-80.
Palmer, D. N., Fearnley, I. M., Medd, S. M., Walker, J. E., Martinus, R. D., Bayliss, S. L.,
Hall, N. A., Lake, B. D., Wolfe, L. S., and Jolly, R. D. (1989): Lysosomal storage of
the DCCD reactive proteolipid subunit of mitochondrial ATP synthase in human
and ovine ceroid lipofuscinoses. Adv Exp Med Biol 266, 211-22; discussion 223.
Palmieri, M., Impey, S., Kang, H., di Ronza, A., Pelz, C., Sardiello, M., and Ballabio, A.
(2011): Characterization of the CLEAR network reveals an integrated control of
cellular clearance pathways. Hum Mol Genet 20, 3852-66.
Pan, X., Eathiraj, S., Munson, M., and Lambright, D. G. (2006): TBC-domain GAPs for
Rab GTPases accelerate GTP hydrolysis by a dual-finger mechanism. Nature 442,
303-6.
Pankiv, S., Alemu, E. A., Brech, A., Bruun, J. A., Lamark, T., Overvatn, A., Bjorkoy, G.,
and Johansen, T. (2010): FYCO1 is a Rab7 effector that binds to LC3 and PI3P to
mediate microtubule plus end-directed vesicle transport. J Cell Biol 188, 253-69.
Pearce, D. A., Ferea, T., Nosel, S. A., Das, B., and Sherman, F. (1999a): Action of BTN1,
the yeast orthologue of the gene mutated in Batten disease. Nat Genet 22, 55-8.
219
Pearce, D. A., Nosel, S. A., and Sherman, F. (1999b): Studies of pH regulation by Btn1p,
the yeast homolog of human Cln3p. Mol Genet Metab 66, 320-3.
Pearce, D. A., and Sherman, F. (1997): BTN1, a yeast gene corresponding to the human
gene responsible for Batten's disease, is not essential for viability, mitochondrial
function, or degradation of mitochondrial ATP synthase. Yeast 13, 691-7.
Pereira-Leal, J. B., and Seabra, M. C. (2001): Evolution of the Rab family of small GTPbinding proteins. J Mol Biol 313, 889-901.
Persaud-Sawin, D. A., and Boustany, R. M. (2005): Cell death pathways in juvenile Batten
disease. Apoptosis 10, 973-85.
Persaud-Sawin, D. A., McNamara, J. O., 2nd, Rylova, S., Vandongen, A., and Boustany, R.
M. (2004): A galactosylceramide binding domain is involved in trafficking of
CLN3 from Golgi to rafts via recycling endosomes. Pediatr Res 56, 449-63.
Peter, B. J., Kent, H. M., Mills, I. G., Vallis, Y., Butler, P. J., Evans, P. R., and McMahon,
H. T. (2004): BAR domains as sensors of membrane curvature: the amphiphysin
BAR structure. Science 303, 495-9.
Peter, F., Nuoffer, C., Pind, S. N., and Balch, W. E. (1994): Guanine nucleotide
dissociation inhibitor is essential for Rab1 function in budding from the
endoplasmic reticulum and transport through the Golgi stack. J Cell Biol 126, 1393406.
Petersen, C. M., Nielsen, M. S., Nykjaer, A., Jacobsen, L., Tommerup, N., Rasmussen, H.
H., Roigaard, H., Gliemann, J., Madsen, P., and Moestrup, S. K. (1997): Molecular
identification of a novel candidate sorting receptor purified from human brain by
receptor-associated protein affinity chromatography. J Biol Chem 272, 3599-605.
Petersson, U., Somogyi, E., Reinholt, F. P., Karlsson, T., Sugars, R. V., and Wendel, M.
(2004): Nucleobindin is produced by bone cells and secreted into the osteoid, with a
potential role as a modulator of matrix maturation. Bone 34, 949-60.
Platt, F. M., Boland, B., and van der Spoel, A. C. (2012): The cell biology of disease:
lysosomal storage disorders: the cellular impact of lysosomal dysfunction. J Cell
Biol 199, 723-34.
Plemel, R. L., Lobingier, B. T., Brett, C. L., Angers, C. G., Nickerson, D. P., Paulsel, A.,
Sprague, D., and Merz, A. J. (2011): Subunit organization and Rab interactions of
Vps-C protein complexes that control endolysosomal membrane traffic. Mol Biol
Cell 22, 1353-63.
Pocha, S. M., Wassmer, T., Niehage, C., Hoflack, B., and Knust, E. (2011): Retromer
controls epithelial cell polarity by trafficking the apical determinant Crumbs. Curr
Biol 21, 1111-7.
Porta, E. A. (1991): Advances in age pigment research. Arch Gerontol Geriatr 12, 303-20.
Porta, E. A. (2002): Pigments in aging: an overview. Ann N Y Acad Sci 959, 57-65.
Poteryaev, D., Datta, S., Ackema, K., Zerial, M., and Spang, A. (2010): Identification of
the switch in early-to-late endosome transition. Cell 141, 497-508.
Praefcke, G. J., and McMahon, H. T. (2004): The dynamin superfamily: universal
membrane tubulation and fission molecules? Nat Rev Mol Cell Biol 5, 133-47.
Prasad, K., Barouch, W., Greene, L., and Eisenberg, E. (1993): A protein cofactor is
required for uncoating of clathrin baskets by uncoating ATPase. J Biol Chem 268,
23758-61.
220
Pullarkat, R. K., and Morris, G. N. (1997): Farnesylation of Batten disease CLN3 protein.
Neuropediatrics 28, 42-4.
Puranam, K. L., Guo, W. X., Qian, W. H., Nikbakht, K., and Boustany, R. M. (1999):
CLN3 defines a novel antiapoptotic pathway operative in neurodegeneration and
mediated by ceramide. Mol Genet Metab 66, 294-308.
Puthenveedu, M. A., Lauffer, B., Temkin, P., Vistein, R., Carlton, P., Thorn, K., Taunton,
J., Weiner, O. D., Parton, R. G., and von Zastrow, M. (2010): Sequence-dependent
sorting of recycling proteins by actin-stabilized endosomal microdomains. Cell 143,
761-73.
Qian, M., Sleat, D. E., Zheng, H., Moore, D., and Lobel, P. (2008): Proteomics analysis of
serum from mutant mice reveals lysosomal proteins selectively transported by each
of the two mannose 6-phosphate receptors. Mol Cell Proteomics 7, 58-70.
Quistgaard, E. M., Madsen, P., Groftehauge, M. K., Nissen, P., Petersen, C. M., and
Thirup, S. S. (2009): Ligands bind to Sortilin in the tunnel of a ten-bladed betapropeller domain. Nat Struct Mol Biol 16, 96-8.
Ramanjaneya, M., Chen, J., Brown, J. E., Tripathi, G., Hallschmid, M., Patel, S., Kern, W.,
Hillhouse, E. W., Lehnert, H., Tan, B. K., and Randeva, H. S. (2010): Identification
of nesfatin-1 in human and murine adipose tissue: a novel depot-specific adipokine
with increased levels in obesity. Endocrinology 151, 3169-80.
Ramesh, N., Mohan, H., and Unniappan, S. (2015): Nucleobindin-1 encodes a nesfatin-1like peptide that stimulates insulin secretion. Gen Comp Endocrinol.
Ramirez-Montealegre, D., and Pearce, D. A. (2005): Defective lysosomal arginine transport
in juvenile Batten disease. Hum Mol Genet 14, 3759-73.
Rawlings, N. D., and Barrett, A. J. (1995): Families of aspartic peptidases, and those of
unknown catalytic mechanism. Methods Enzymol 248, 105-20.
Ren, J., Wen, L., Gao, X., Jin, C., Xue, Y., and Yao, X. (2009): DOG 1.0: illustrator of
protein domain structures. Cell Res 19, 271-3.
Rider, J. A., and Rider, D. L. (1988): Batten disease: past, present, and future. Am J Med
Genet Suppl 5, 21-6.
Riederer, M. A., Soldati, T., Shapiro, A. D., Lin, J., and Pfeffer, S. R. (1994): Lysosome
biogenesis requires Rab9 function and receptor recycling from endosomes to the
trans-Golgi network. J Cell Biol 125, 573-82.
Risselada, H. J., Kutzner, C., and Grubmuller, H. (2011): Caught in the act: visualization of
SNARE-mediated fusion events in molecular detail. Chembiochem 12, 1049-55.
Roberts, D. L., Weix, D. J., Dahms, N. M., and Kim, J. J. (1998): Molecular basis of
lysosomal enzyme recognition: three-dimensional structure of the cation-dependent
mannose 6-phosphate receptor. Cell 93, 639-48.
Rohrer, J., and Kornfeld, R. (2001): Lysosomal hydrolase mannose 6-phosphate uncovering
enzyme resides in the trans-Golgi network. Mol Biol Cell 12, 1623-31.
Rojas, A. M., Fuentes, G., Rausell, A., and Valencia, A. (2012): The Ras protein
superfamily: evolutionary tree and role of conserved amino acids. J Cell Biol 196,
189-201.
Rojas, R., Kametaka, S., Haft, C. R., and Bonifacino, J. S. (2007): Interchangeable but
essential functions of SNX1 and SNX2 in the association of retromer with
endosomes and the trafficking of mannose 6-phosphate receptors. Mol Cell Biol 27,
1112-24.
221
Rojas, R., van Vlijmen, T., Mardones, G. A., Prabhu, Y., Rojas, A. L., Mohammed, S.,
Heck, A. J., Raposo, G., van der Sluijs, P., and Bonifacino, J. S. (2008): Regulation
of retromer recruitment to endosomes by sequential action of Rab5 and Rab7. J Cell
Biol 183, 513-26.
Rosenthal, J. A., Chen, H., Slepnev, V. I., Pellegrini, L., Salcini, A. E., Di Fiore, P. P., and
De Camilli, P. (1999): The epsins define a family of proteins that interact with
components of the clathrin coat and contain a new protein module. J Biol Chem
274, 33959-65.
Rosorius, O., Mieskes, G., Issinger, O. G., Korner, C., Schmidt, B., von Figura, K., and
Braulke, T. (1993): Characterization of phosphorylation sites in the cytoplasmic
domain of the 300 kDa mannose-6-phosphate receptor. Biochem J 292 ( Pt 3), 8338.
Rotty, J. D., Wu, C., and Bear, J. E. (2013): New insights into the regulation and cellular
functions of the ARP2/3 complex. Nat Rev Mol Cell Biol 14, 7-12.
Roux, A., Cuvelier, D., Nassoy, P., Prost, J., Bassereau, P., and Goud, B. (2005): Role of
curvature and phase transition in lipid sorting and fission of membrane tubules.
Embo J 24, 1537-45.
Rusyn, E., Mousallem, T., Persaud-Sawin, D. A., Miller, S., and Boustany, R. M. (2008):
CLN3p impacts galactosylceramide transport, raft morphology, and lipid content.
Pediatr Res 63, 625-31.
Saadipour, K., Yang, M., Lim, Y., Georgiou, K., Sun, Y., Keating, D., Liu, J., Wang, Y. R.,
Gai, W. P., Zhong, J. H., Wang, Y. J., and Zhou, X. F. (2013): Amyloid beta(1)()(4)(2) (Abeta(4)(2)) up-regulates the expression of sortilin via the p75(NTR)/RhoA
signaling pathway. J Neurochem 127, 152-62.
Saftig, P., and Klumperman, J. (2009): Lysosome biogenesis and lysosomal membrane
proteins: trafficking meets function. Nat Rev Mol Cell Biol 10, 623-35.
Sagne, C., and Gasnier, B. (2008): Molecular physiology and pathophysiology of
lysosomal membrane transporters. J Inherit Metab Dis 31, 258-66.
Sancak, Y., Bar-Peled, L., Zoncu, R., Markhard, A. L., Nada, S., and Sabatini, D. M.
(2010): Ragulator-Rag complex targets mTORC1 to the lysosomal surface and is
necessary for its activation by amino acids. Cell 141, 290-303.
Sandvig, K., Garred, O., Prydz, K., Kozlov, J. V., Hansen, S. H., and van Deurs, B. (1992):
Retrograde transport of endocytosed Shiga toxin to the endoplasmic reticulum.
Nature 358, 510-2.
Santavuori, P. (1988): Neuronal ceroid-lipofuscinoses in childhood. Brain Dev 10, 80-3.
Sardiello, M., Palmieri, M., di Ronza, A., Medina, D. L., Valenza, M., Gennarino, V. A., Di
Malta, C., Donaudy, F., Embrione, V., Polishchuk, R. S., Banfi, S., Parenti, G.,
Cattaneo, E., and Ballabio, A. (2009): A gene network regulating lysosomal
biogenesis and function. Science 325, 473-7.
Sarret, P., Krzywkowski, P., Segal, L., Nielsen, M. S., Petersen, C. M., Mazella, J., Stroh,
T., and Beaudet, A. (2003): Distribution of NTS3 receptor/sortilin mRNA and
protein in the rat central nervous system. J Comp Neurol 461, 483-505.
Savukoski, M., Klockars, T., Holmberg, V., Santavuori, P., Lander, E. S., and Peltonen, L.
(1998): CLN5, a novel gene encoding a putative transmembrane protein mutated in
Finnish variant late infantile neuronal ceroid lipofuscinosis. Nat Genet 19, 286-8.
222
Scherer, P. E., Lederkremer, G. Z., Williams, S., Fogliano, M., Baldini, G., and Lodish, H.
F. (1996): Cab45, a novel (Ca2+)-binding protein localized to the Golgi lumen. J
Cell Biol 133, 257-68.
Schlossman, D. M., Schmid, S. L., Braell, W. A., and Rothman, J. E. (1984): An enzyme
that removes clathrin coats: purification of an uncoating ATPase. J Cell Biol 99,
723-33.
Schmidt, B., Kiecke-Siemsen, C., Waheed, A., Braulke, T., and von Figura, K. (1995):
Localization of the insulin-like growth factor II binding site to amino acids 15081566 in repeat 11 of the mannose 6-phosphate/insulin-like growth factor II receptor.
J Biol Chem 270, 14975-82.
Schmiedt, M. L. (2012): The CLN5 disease - protein maturation, trafficking and pathology:
Medical Faculty, University of Helsinki.
Schmiedt, M. L., Bessa, C., Heine, C., Ribeiro, M. G., Jalanko, A., and Kyttala, A. (2010):
The neuronal ceroid lipofuscinosis protein CLN5: new insights into cellular
maturation, transport, and consequences of mutations. Hum Mutat 31, 356-65.
Schmiedt, M. L., Blom, T., Kopra, O., Wong, A., von Schantz-Fant, C., Ikonen, E.,
Kuronen, M., Jauhiainen, M., Cooper, J. D., and Jalanko, A. (2012): Cln5deficiency in mice leads to microglial activation, defective myelination and changes
in lipid metabolism. Neurobiol Dis 46, 19-29.
Schu, P. V., Takegawa, K., Fry, M. J., Stack, J. H., Waterfield, M. D., and Emr, S. D.
(1993): Phosphatidylinositol 3-kinase encoded by yeast VPS34 gene essential for
protein sorting. Science 260, 88-91.
Schultz, M. L., Tecedor, L., Chang, M., and Davidson, B. L. (2011): Clarifying lysosomal
storage diseases. Trends Neurosci.
Schweizer, A., Kornfeld, S., and Rohrer, J. (1996): Cysteine34 of the cytoplasmic tail of
the cation-dependent mannose 6-phosphate receptor is reversibly palmitoylated and
required for normal trafficking and lysosomal enzyme sorting. J Cell Biol 132, 57784.
Schweizer, A., Kornfeld, S., and Rohrer, J. (1997): Proper sorting of the cation-dependent
mannose 6-phosphate receptor in endosomes depends on a pair of aromatic amino
acids in its cytoplasmic tail. Proc Natl Acad Sci U S A 94, 14471-6.
Seabra, M. C., Goldstein, J. L., Sudhof, T. C., and Brown, M. S. (1992): Rab
geranylgeranyl transferase. A multisubunit enzyme that prenylates GTP-binding
proteins terminating in Cys-X-Cys or Cys-Cys. J Biol Chem 267, 14497-503.
Seaman, M. N. (2004): Cargo-selective endosomal sorting for retrieval to the Golgi
requires retromer. J Cell Biol 165, 111-22.
Seaman, M. N. (2007): Identification of a novel conserved sorting motif required for
retromer-mediated endosome-to-TGN retrieval. J Cell Sci 120, 2378-89.
Seaman, M. N. (2012): The retromer complex - endosomal protein recycling and beyond. J
Cell Sci 125, 4693-702.
Seaman, M. N., Harbour, M. E., Tattersall, D., Read, E., and Bright, N. (2009): Membrane
recruitment of the cargo-selective retromer subcomplex is catalysed by the small
GTPase Rab7 and inhibited by the Rab-GAP TBC1D5. J Cell Sci 122, 2371-82.
Seaman, M. N., Marcusson, E. G., Cereghino, J. L., and Emr, S. D. (1997): Endosome to
Golgi retrieval of the vacuolar protein sorting receptor, Vps10p, requires the
function of the VPS29, VPS30, and VPS35 gene products. J Cell Biol 137, 79-92.
223
Seaman, M. N., McCaffery, J. M., and Emr, S. D. (1998): A membrane coat complex
essential for endosome-to-Golgi retrograde transport in yeast. J Cell Biol 142, 66581.
Seidah, N. G., and Chretien, M. (1999): Proprotein and prohormone convertases: a family
of subtilases generating diverse bioactive polypeptides. Brain Res 848, 45-62.
Semerdjieva, S., Shortt, B., Maxwell, E., Singh, S., Fonarev, P., Hansen, J., Schiavo, G.,
Grant, B. D., and Smythe, E. (2008): Coordinated regulation of AP2 uncoating from
clathrin-coated vesicles by rab5 and hRME-6. J Cell Biol 183, 499-511.
Sharpe, H. J., Stevens, T. J., and Munro, S. (2010): A comprehensive comparison of
transmembrane domains reveals organelle-specific properties. Cell 142, 158-69.
Shimizu, H., Oh, I. S., Okada, S., and Mori, M. (2009): Nesfatin-1: an overview and future
clinical application. Endocr J 56, 537-43.
Silva, B., Adams, J., and Lee, S. Y. (2015): Proteolytic processing of the neuronal ceroid
lipofuscinosis related lysosomal protein CLN5. Exp Cell Res.
Sincock, P. M., Ganley, I. G., Krise, J. P., Diederichs, S., Sivars, U., O'Connor, B., Ding,
L., and Pfeffer, S. R. (2003): Self-assembly is important for TIP47 function in
mannose 6-phosphate receptor transport. Traffic 4, 18-25.
Sivars, U., Aivazian, D., and Pfeffer, S. R. (2003): Yip3 catalyses the dissociation of
endosomal Rab-GDI complexes. Nature 425, 856-9.
Skanland, S. S., Walchli, S., Brech, A., and Sandvig, K. (2009): SNX4 in complex with
clathrin and dynein: implications for endosome movement. PLoS One 4, e5935.
Sleat, D. E., Sohar, I., Pullarkat, P. S., Lobel, P., and Pullarkat, R. K. (1998): Specific
alterations in levels of mannose 6-phosphorylated glycoproteins in different
neuronal ceroid lipofuscinoses. Biochem J 334 ( Pt 3), 547-51.
Sleat, D. E., Zheng, H., Qian, M., and Lobel, P. (2006): Identification of sites of mannose
6-phosphorylation on lysosomal proteins. Mol Cell Proteomics 5, 686-701.
Small, S. A., Kent, K., Pierce, A., Leung, C., Kang, M. S., Okada, H., Honig, L., Vonsattel,
J. P., and Kim, T. W. (2005): Model-guided microarray implicates the retromer
complex in Alzheimer's disease. Ann Neurol 58, 909-19.
Sollner, T., Bennett, M. K., Whiteheart, S. W., Scheller, R. H., and Rothman, J. E. (1993):
A protein assembly-disassembly pathway in vitro that may correspond to sequential
steps of synaptic vesicle docking, activation, and fusion. Cell 75, 409-18.
Somogyi, E., Petersson, U., Hultenby, K., and Wendel, M. (2003): Calreticulin--an
endoplasmic reticulum protein with calcium-binding activity is also found in the
extracellular matrix. Matrix Biol 22, 179-91.
Somogyi, E., Petersson, U., Sugars, R. V., Hultenby, K., and Wendel, M. (2004):
Nucleobindin--a Ca2+-binding protein present in the cells and mineralized tissues
of the tooth. Calcif Tissue Int 74, 366-76.
Spielmeyer, W. (1905a): über familiäre amaurotische Idioten. Neurol Cbl 24, 620-621.
Spielmeyer, W. (1905b): Weitere Mitteilungen über eine besondere Form von familiärer
amaurotischer Idiotie. Neuro Cbl 24, 1131-1132.
Sprang, S. R. (1997): G protein mechanisms: insights from structural analysis. Annu Rev
Biochem 66, 639-78.
St-Arnaud, R., Prud'homme, J., Leung-Hagesteijn, C., and Dedhar, S. (1995): Constitutive
expression of calreticulin in osteoblasts inhibits mineralization. J Cell Biol 131,
1351-9.
224
Staub, O., and Rotin, D. (2006): Role of ubiquitylation in cellular membrane transport.
Physiol Rev 86, 669-707.
Stein, C. S., Yancey, P. H., Martins, I., Sigmund, R. D., Stokes, J. B., and Davidson, B. L.
(2010): Osmoregulation of ceroid neuronal lipofuscinosis type 3 in the renal
medulla. Am J Physiol Cell Physiol 298, C1388-400.
Stein, M. P., Feng, Y., Cooper, K. L., Welford, A. M., and Wandinger-Ness, A. (2003):
Human VPS34 and p150 are Rab7 interacting partners. Traffic 4, 754-71.
Steinberg, F., Gallon, M., Winfield, M., Thomas, E. C., Bell, A. J., Heesom, K. J., Tavare,
J. M., and Cullen, P. J. (2013): A global analysis of SNX27-retromer assembly and
cargo specificity reveals a function in glucose and metal ion transport. Nat Cell Biol
15, 461-71.
Stengel, A., Goebel, M., and Tache, Y. (2010): Nesfatin-1: a novel inhibitory regulator of
food intake and body weight. Obes Rev 12, 261-71.
Stengel, A., Goebel, M., Yakubov, I., Wang, L., Witcher, D., Coskun, T., Tache, Y., Sachs,
G., and Lambrecht, N. W. (2009): Identification and characterization of nesfatin-1
immunoreactivity in endocrine cell types of the rat gastric oxyntic mucosa.
Endocrinology 150, 232-8.
Stengel, O. (1826a): Beretning om et maerkeligt Sygdomstilfaelde hos fire Sodskende i
Naerheden af Roraas. Eyr Medicinsk Tidskrift (Christiana) 1, 347-352.
Stenmark, H. (2009): Rab GTPases as coordinators of vesicle traffic. Nat Rev Mol Cell Biol
10, 513-25.
Storch, S., Pohl, S., and Braulke, T. (2004): A dileucine motif and a cluster of acidic amino
acids in the second cytoplasmic domain of the batten disease-related CLN3 protein
are required for efficient lysosomal targeting. J Biol Chem 279, 53625-34.
Storch, S., Pohl, S., Quitsch, A., Falley, K., and Braulke, T. (2007): C-terminal prenylation
of the CLN3 membrane glycoprotein is required for efficient endosomal sorting to
lysosomes. Traffic 8, 431-44.
Stow, J. L., de Almeida, J. B., Narula, N., Holtzman, E. J., Ercolani, L., and Ausiello, D. A.
(1991): A heterotrimeric G protein, G alpha i-3, on Golgi membranes regulates the
secretion of a heparan sulfate proteoglycan in LLC-PK1 epithelial cells. J Cell Biol
114, 1113-24.
Su, Y., Zhang, J., Tang, Y., Bi, F., and Liu, J. N. (2010): The novel function of nesfatin-1:
anti-hyperglycemia. Biochem Biophys Res Commun 391, 1039-42.
Sugiyama, T., Kumagai, H., Morikawa, Y., Wada, Y., Sugiyama, A., Yasuda, K., Yokoi,
N., Tamura, S., Kojima, T., Nosaka, T., Senba, E., Kimura, S., Kadowaki, T.,
Kodama, T., and Kitamura, T. (2000): A novel low-density lipoprotein receptorrelated protein mediating cellular uptake of apolipoprotein E-enriched beta-VLDL
in vitro. Biochemistry 39, 15817-25.
Sunahara, R. K., Tesmer, J. J., Gilman, A. G., and Sprang, S. R. (1997): Crystal structure of
the adenylyl cyclase activator Gsalpha. Science 278, 1943-7.
Sutton, R. B., Fasshauer, D., Jahn, R., and Brunger, A. T. (1998): Crystal structure of a
SNARE complex involved in synaptic exocytosis at 2.4 A resolution. Nature 395,
347-53.
Swarbrick, J. D., Shaw, D. J., Chhabra, S., Ghai, R., Valkov, E., Norwood, S. J., Seaman,
M. N., and Collins, B. M. (2011): VPS29 is not an active metallo-phosphatase but is
225
a rigid scaffold required for retromer interaction with accessory proteins. PLoS One
6, e20420.
Tabuchi, M., Yanatori, I., Kawai, Y., and Kishi, F. (2010): Retromer-mediated direct
sorting is required for proper endosomal recycling of the mammalian iron
transporter DMT1. J Cell Sci 123, 756-66.
Taniguchi, N., Taniura, H., Niinobe, M., Takayama, C., Tominaga-Yoshino, K., Ogura, A.,
and Yoshikawa, K. (2000): The postmitotic growth suppressor necdin interacts with
a calcium-binding protein (NEFA) in neuronal cytoplasm. J Biol Chem 275, 3167481.
Taniura, H., Matsumoto, K., and Yoshikawa, K. (1999): Physical and functional
interactions of neuronal growth suppressor necdin with p53. J Biol Chem 274,
16242-8.
Taylor, R. S., Jones, S. M., Dahl, R. H., Nordeen, M. H., and Howell, K. E. (1997):
Characterization of the Golgi complex cleared of proteins in transit and examination
of calcium uptake activities. Mol Biol Cell 8, 1911-31.
Tebar, F., Llado, A., and Enrich, C. (2002a): Role of calmodulin in the modulation of the
MAPK signalling pathway and the transactivation of epidermal growth factor
receptor mediated by PKC. FEBS Lett 517, 206-10.
Tebar, F., Villalonga, P., Sorkina, T., Agell, N., Sorkin, A., and Enrich, C. (2002b):
Calmodulin regulates intracellular trafficking of epidermal growth factor receptor
and the MAPK signaling pathway. Mol Biol Cell 13, 2057-68.
Temkin, P., Lauffer, B., Jager, S., Cimermancic, P., Krogan, N. J., and von Zastrow, M.
(2011): SNX27 mediates retromer tubule entry and endosome-to-plasma membrane
trafficking of signalling receptors. Nat Cell Biol 13, 715-21.
Teng, H. K., Teng, K. K., Lee, R., Wright, S., Tevar, S., Almeida, R. D., Kermani, P.,
Torkin, R., Chen, Z. Y., Lee, F. S., Kraemer, R. T., Nykjaer, A., and Hempstead, B.
L. (2005): ProBDNF induces neuronal apoptosis via activation of a receptor
complex of p75NTR and sortilin. J Neurosci 25, 5455-63.
Terman, A., and Brunk, U. T. (1998): Lipofuscin: mechanisms of formation and increase
with age. Apmis 106, 265-76.
The International Batten Disease Consortium (1995): Isolation of a novel gene underlying
Batten disease, CLN3. The International Batten Disease Consortium. Cell 82, 94957.
Tsukumo, Y., Tomida, A., Kitahara, O., Nakamura, Y., Asada, S., Mori, K., and Tsuruo, T.
(2007): Nucleobindin 1 controls the unfolded protein response by inhibiting ATF6
activation. J Biol Chem 282, 29264-72.
Tsukumo, Y., Tsukahara, S., Saito, S., Tsuruo, T., and Tomida, A. (2009): A novel
endoplasmic reticulum export signal: proline at the +2-position from the signal
peptide cleavage site. J Biol Chem 284, 27500-10.
Tuxworth, R. I., Vivancos, V., O'Hare, M. B., and Tear, G. (2009): Interactions between the
juvenile Batten disease gene, CLN3, and the Notch and JNK signalling pathways.
Hum Mol Genet 18, 667-78.
Tyynela, J., Palmer, D. N., Baumann, M., and Haltia, M. (1993): Storage of saposins A and
D in infantile neuronal ceroid-lipofuscinosis. FEBS Lett 330, 8-12.
226
Tyynela, J., Suopanki, J., Santavuori, P., Baumann, M., and Haltia, M. (1997): Variant late
infantile neuronal ceroid-lipofuscinosis: pathology and biochemistry. J Neuropathol
Exp Neurol 56, 369-75.
Ullrich, O., Horiuchi, H., Bucci, C., and Zerial, M. (1994): Membrane association of Rab5
mediated by GDP-dissociation inhibitor and accompanied by GDP/GTP exchange.
Nature 368, 157-60.
Uusi-Rauva, K., Kyttala, A., van der Kant, R., Vesa, J., Tanhuanpaa, K., Neefjes, J.,
Olkkonen, V. M., and Jalanko, A. (2012): Neuronal ceroid lipofuscinosis protein
CLN3 interacts with motor proteins and modifies location of late endosomal
compartments. Cell Mol Life Sci.
Uusi-Rauva, K., Luiro, K., Tanhuanpaa, K., Kopra, O., Martin-Vasallo, P., Kyttala, A., and
Jalanko, A. (2008): Novel interactions of CLN3 protein link Batten disease to
dysregulation of fodrin-Na+, K+ ATPase complex. Exp Cell Res 314, 2895-905.
Uvebrant, P., and Hagberg, B. (1997): Neuronal ceroid lipofuscinoses in Scandinavia.
Epidemiology and clinical pictures. Neuropediatrics 28, 6-8.
Valencia, C. A., Cotten, S. W., Duan, J., and Liu, R. (2008): Modulation of nucleobindin-1
and nucleobindin-2 by caspases. FEBS Lett 582, 286-90.
van Weering, J. R., Verkade, P., and Cullen, P. J. (2012): SNX-BAR-mediated endosome
tubulation is co-ordinated with endosome maturation. Traffic 13, 94-107.
Vesa, J., Chin, M. H., Oelgeschlager, K., Isosomppi, J., DellAngelica, E. C., Jalanko, A.,
and Peltonen, L. (2002): Neuronal ceroid lipofuscinoses are connected at molecular
level: interaction of CLN5 protein with CLN2 and CLN3. Mol Biol Cell 13, 241020.
Vesa, J., Hellsten, E., Verkruyse, L. A., Camp, L. A., Rapola, J., Santavuori, P., Hofmann,
S. L., and Peltonen, L. (1995): Mutations in the palmitoyl protein thioesterase gene
causing infantile neuronal ceroid lipofuscinosis. Nature 376, 584-7.
Vitiello, S. P., Benedict, J. W., Padilla-Lopez, S., and Pearce, D. A. (2010): Interaction
between Sdo1p and Btn1p in the Saccharomyces cerevisiae model for Batten
disease. Hum Mol Genet 19, 931-42.
Vitner, E. B., Platt, F. M., and Futerman, A. H. (2010): Common and uncommon
pathogenic cascades in lysosomal storage diseases. J Biol Chem 285, 20423-7.
von Blume, J., Alleaume, A. M., Kienzle, C., Carreras-Sureda, A., Valverde, M., and
Malhotra, V. (2012): Cab45 is required for Ca(2+)-dependent secretory cargo
sorting at the trans-Golgi network. J Cell Biol 199, 1057-66.
von Rotz, R. C., Kohli, B. M., Bosset, J., Meier, M., Suzuki, T., Nitsch, R. M., and
Konietzko, U. (2004): The APP intracellular domain forms nuclear multiprotein
complexes and regulates the transcription of its own precursor. J Cell Sci 117, 443548.
von Schantz, C., Saharinen, J., Kopra, O., Cooper, J. D., Gentile, M., Hovatta, I., Peltonen,
L., and Jalanko, A. (2008): Brain gene expression profiles of Cln1 and Cln5
deficient mice unravels common molecular pathways underlying neuronal
degeneration in NCL diseases. BMC Genomics 9, 146.
Wagner, U. G., Petersen, E. I., Schwab, H., and Kratky, C. (2002): EstB from Burkholderia
gladioli: a novel esterase with a beta-lactamase fold reveals steric factors to
discriminate between esterolytic and beta-lactam cleaving activity. Protein Sci 11,
467-78.
227
Walenta, J. H., Didier, A. J., Liu, X., and Kramer, H. (2001): The Golgi-associated hook3
protein is a member of a novel family of microtubule-binding proteins. J Cell Biol
152, 923-34.
Wang, S. N., Miyauchi, M., Koshikawa, N., Maruyama, K., Kubota, T., Miura, K.,
Kurosawa, Y., Awaya, A., and Kanai, Y. (1994): Antigen expression associated
with lymph node metastasis in gastric adenocarcinomas. Pathol Int 44, 844-9.
Wassmer, T., Attar, N., Bujny, M. V., Oakley, J., Traer, C. J., and Cullen, P. J. (2007): A
loss-of-function screen reveals SNX5 and SNX6 as potential components of the
mammalian retromer. J Cell Sci 120, 45-54.
Wassmer, T., Attar, N., Harterink, M., van Weering, J. R., Traer, C. J., Oakley, J., Goud,
B., Stephens, D. J., Verkade, P., Korswagen, H. C., and Cullen, P. J. (2009): The
retromer coat complex coordinates endosomal sorting and dynein-mediated
transport, with carrier recognition by the trans-Golgi network. Dev Cell 17, 110-22.
Weimbs, T., Low, S. H., Chapin, S. J., Mostov, K. E., Bucher, P., and Hofmann, K. (1997):
A conserved domain is present in different families of vesicular fusion proteins: a
new superfamily. Proc Natl Acad Sci U S A 94, 3046-51.
Weimer, J. M., Custer, A. W., Benedict, J. W., Alexander, N. A., Kingsley, E., Federoff, H.
J., Cooper, J. D., and Pearce, D. A. (2006): Visual deficits in a mouse model of
Batten disease are the result of optic nerve degeneration and loss of dorsal lateral
geniculate thalamic neurons. Neurobiol Dis 22, 284-93.
Weiss, T. S., Chamberlain, C. E., Takeda, T., Lin, P., Hahn, K. M., and Farquhar, M. G.
(2001): Galpha i3 binding to calnuc on Golgi membranes in living cells monitored
by fluorescence resonance energy transfer of green fluorescent protein fusion
proteins. Proc Natl Acad Sci U S A 98, 14961-6.
Wen, L., Tang, F. L., Hong, Y., Luo, S. W., Wang, C. L., He, W., Shen, C., Jung, J. U.,
Xiong, F., Lee, D. H., Zhang, Q. G., Brann, D., Kim, T. W., Yan, R., Mei, L., and
Xiong, W. C. (2011): VPS35 haploinsufficiency increases Alzheimer's disease
neuropathology. J Cell Biol 195, 765-79.
Wendel, M., Sommarin, Y., Bergman, T., and Heinegard, D. (1995): Isolation,
characterization, and primary structure of a calcium-binding 63-kDa bone protein. J
Biol Chem 270, 6125-33.
Whitley, P., Grahn, E., Kutay, U., Rapoport, T. A., and von Heijne, G. (1996): A 12residue-long polyleucine tail is sufficient to anchor synaptobrevin to the
endoplasmic reticulum membrane. J Biol Chem 271, 7583-6.
Williams, P., Tulke, S., Ilegems, E., Berggren, P. O., and Broberger, C. (2014): Expression
of nucleobindin 1 (NUCB1) in pancreatic islets and other endocrine tissues. Cell
Tissue Res 358, 331-42.
Williams, R. E., Aberg, L., Autti, T., Goebel, H. H., Kohlschutter, A., and Lonnqvist, T.
(2006): Diagnosis of the neuronal ceroid lipofuscinoses: an update. Biochim
Biophys Acta 1762, 865-72.
Williams, R. E., Gardiner, R. M., and Goebel, H. H. (1999): The European Concerted
Action NCL Clinical Case Registry. Mol Genet Metab 66, 407-8.
Wilson, B. S., Komuro, M., and Farquhar, M. G. (1994): Cellular variations in
heterotrimeric G protein localization and expression in rat pituitary. Endocrinology
134, 233-44.
228
Winchester, B. (2013): Lysosomal diseases: diagnostic update. J Inherit Metab Dis 37,
599-608.
Wolins, N. E., Rubin, B., and Brasaemle, D. L. (2001): TIP47 associates with lipid
droplets. J Biol Chem 276, 5101-8.
Xin, W., Mullen, T. E., Kiely, R., Min, J., Feng, X., Cao, Y., O'Malley, L., Shen, Y., ChuShore, C., Mole, S. E., Goebel, H. H., and Sims, K. (2010): CLN5 mutations are
frequent in juvenile and late-onset non-Finnish patients with NCL. Neurology 74,
565-71.
Yamashiro, D. J., Fluss, S. R., and Maxfield, F. R. (1983): Acidification of endocytic
vesicles by an ATP-dependent proton pump. J Cell Biol 97, 929-34.
Yamashiro, D. J., and Maxfield, F. R. (1987): Acidification of morphologically distinct
endosomes in mutant and wild-type Chinese hamster ovary cells. J Cell Biol 105,
2723-33.
Yang, Y., and Cook, D. G. (2004): Presenilin-1 deficiency impairs glutamate-evoked
intracellular calcium responses in neurons. Neuroscience 124, 501-5.
Ye, J., Rawson, R. B., Komuro, R., Chen, X., Dave, U. P., Prywes, R., Brown, M. S., and
Goldstein, J. L. (2000): ER stress induces cleavage of membrane-bound ATF6 by
the same proteases that process SREBPs. Mol Cell 6, 1355-64.
Yoshida, N., Maejima, Y., Sedbazar, U., Ando, A., Kurita, H., Damdindorj, B., Takano, E.,
Gantulga, D., Iwasaki, Y., Kurashina, T., Onaka, T., Dezaki, K., Nakata, M., Mori,
M., and Yada, T. (2010): Stressor-responsive central nesfatin-1 activates
corticotropin-releasing hormone, noradrenaline and serotonin neurons and evokes
hypothalamic-pituitary-adrenal axis. Aging (Albany NY) 2, 775-84.
Yosten, G. L., and Samson, W. K. (2009): Nesfatin-1 exerts cardiovascular actions in brain:
possible interaction with the central melanocortin system. Am J Physiol Regul
Integr Comp Physiol 297, R330-6.
Yosten, G. L., and Samson, W. K. (2010): The anorexigenic and hypertensive effects of
nesfatin-1 are reversed by pretreatment with an oxytocin receptor antagonist. Am J
Physiol Regul Integr Comp Physiol 298, R1642-7.
Yosten, G. L., and Samson, W. K. (2013): Cardiovascular and antidipsogenic effects of
nesfatin-1. Curr Pharm Des 19, 6973-5.
Yosten, G. L., and Samson, W. K. (2014): Neural circuitry underlying the central
hypertensive action of nesfatin-1: melanocortins, corticotropin-releasing hormone,
and oxytocin. Am J Physiol Regul Integr Comp Physiol 306, R722-7.
Zelazny, E., Santambrogio, M., Pourcher, M., Chambrier, P., Berne-Dedieu, A., FobisLoisy, I., Miege, C., Jaillais, Y., and Gaude, T. (2013): Mechanisms governing the
endosomal membrane recruitment of the core retromer in Arabidopsis. J Biol Chem
288, 8815-25.
Zeman, W., and Dyken, P. (1969): Neuronal ceroid-lipofuscinosis (Batten's disease):
relationship to amaurotic family idiocy? Pediatrics 44, 570-83.
Zimprich, A., Benet-Pages, A., Struhal, W., Graf, E., Eck, S. H., Offman, M. N.,
Haubenberger, D., Spielberger, S., Schulte, E. C., Lichtner, P., Rossle, S. C., Klopp,
N., Wolf, E., Seppi, K., Pirker, W., Presslauer, S., Mollenhauer, B., Katzenschlager,
R., Foki, T., Hotzy, C., Reinthaler, E., Harutyunyan, A., Kralovics, R., Peters, A.,
Zimprich, F., Brucke, T., Poewe, W., Auff, E., Trenkwalder, C., Rost, B.,
Ransmayr, G., Winkelmann, J., Meitinger, T., and Strom, T. M. (2011): A mutation
229
in VPS35, encoding a subunit of the retromer complex, causes late-onset Parkinson
disease. Am J Hum Genet 89, 168-75.
Zoncu, R., Bar-Peled, L., Efeyan, A., Wang, S., Sancak, Y., and Sabatini, D. M. (2011):
mTORC1 senses lysosomal amino acids through an inside-out mechanism that
requires the vacuolar H(+)-ATPase. Science 334, 678-83.
230
ANNEXE 1
Intracellular trafficking of LRP9 is dependent on two
acidic cluster/dileucine motifs
Boucher, R., Larkin, H., Brodeur, J., Gagnon, H., Thériault, C., and
Lavoie, C.
Histochem Cell Biol 130(2):315-27 (2008)
Travaux de stage
IF=2,32
231
Intracellular trafficking of LRP9 is dependent on two acidic cluster/dileucine motifs
Boucher, R., Larkin, H., Brodeur, J., Gagnon, H., Thériault, C., and Lavoie, C.
Abstract
LDL receptor-related protein 9 (LRP9) is a distant member of the low-density
lipoprotein receptor (LDLR) superfamily. To date, there are no reports on the cellular
distribution of LRP9 or the signals responsible for its localization. Here, we investigated
the intracellular localization and trafficking of LRP9. Using confocal microscopy, we
demonstrated that LRP9 was not present at the plasma membrane but co-localized with
various markers of the trans-Golgi network (TGN) and endosomes. This co-localization
was dependent on the presence of two acidic cluster/dileucine (DXXLL) motifs in the
cytoplasmic tail of LRP9, which interact with GGA proteins, clathrin adaptors involved in
transport between the TGN and endosomes. LRP9 is the first example of a transmembrane
protein with an internal GGA-binding sequence in addition to the usual C-terminal motif.
An inactivating mutation (LL → AA) in both DXXLL motifs, which completely inhibited
the interaction of LRP9 with GGA proteins, led to an intracellular redistribution of LRP9
from the TGN to early endosomes and the cell surface, indicating that the two DXXLL
motifs are essential sorting determinants of LRP9. In conclusion, our results suggest that
LRP9 cycles between the TGN, endosomes and the plasma membrane through a GGA
dependent-trafficking mechanism.
Keywords
LDLR related protein 9 (LRP9), GGA (Golgi-localized, γ ear-containing, ADP
ribosylation factor binding protein), trans-Golgi network, Endosome, Acidic
cluster/dileucine (DXXLL) motif, LDL receptor
Introduction
Introduction
The low-density lipoprotein receptor (LDLR) family comprises a large number of
members that share common structural features as the LDLRA repeats, LDLRB repeats,
epidermal growth factor precursor-type repeats and a cytoplasmic region which includes
internalization motifs. The variable numbers and combinations of these elements
correspond with the huge variety of ligands and functions of these receptors. A new
member of this growing family, termed LDLR-related protein 9 (LRP9), was recently
identified (Sugiyama et al. 2000). LRP9 is part of a new subfamily of the LDLR
superfamily that includes two other receptors, LRP3 and LRP12 (previously named st7)
(Battle et al. 2003). Like all LDLR family members, this new subgroup contains LDLRA
domain repeats in their extracellular domains (Hussain et al. 1999; Strickland et al. 2002)
that function as ligand binding sites (Yamamoto et al. 1984). However, several unique
232
structural features distinguish this new LRP subfamily from prototypical members of the
LDLR superfamily (Battle et al. 2003): (1) the absence of EGF-like repeats and YWTDcontaining domains in their extracellular domain; (2) the presence of extracellular CUB
domains that are found in a diverse array of functionally unrelated extracellular proteins
(Bork and Beckmann 1993) and that are involved in ligand binding (Christensen and Birn
2002); and (3) a large cytoplasmic tail that contains a proline-rich domain.
The important role of many members of the LDLR subfamily like LDL, LRP,
megalin, VDL and ApoER receptors in endocytosis and lipid metabolism is well known
(Willnow 1999). However, several other biological functions are becoming more and more
evident. Members of the LDLR family are involved in the regulation of cell surface
protease activity, control of cellular entry of bacterial toxins and viruses, the transport and
activation of steroid hormones, the regulation of Ca2+ homeostasis and also in cellular
signaling events (Sugiyama et al. 2000; Herz 2001; Strickland et al. 2002; Battle et al.
2003; Kikuchi et al. 2007; May et al. 2007)
Little is known about the function of LRP9. It has been reported to be ubiquitously
expressed and to be involved in the internalization of apolipoprotein E (ApoE)-enriched
βVLDL (Sugiyama et al. 2000). Additionally, putative signals for endocytosis and
intracellular trafficking (YXXϕ, dileucine, acidic cluster/dileucine [DXXLL] motifs) are
present in the cytoplasmic tail of LRP9 (Sugiyama et al. 2000). Moreover, several putative
signal transduction motifs (e.g., PDZ-binding domain, proline-rich region) as well as
potential phosphorylation sites have also been identified in the cytoplasmic tail of LRP9
(Battle et al. 2003). LRP9 thus has the molecular characteristics of a trafficking and
signalling member of the LDLR family.
GGA proteins (Golgi localized, γ ear-containing, ADP ribosylation factor-binding
proteins) are monomeric adaptor proteins that mediate the transport of cargo proteins
between the trans-Golgi Network (TGN) and endosomes (reviewed by Bonifacino 2004;
Ghosh and Kornfeld 2004). They have a modular structure consisting of an amino-terminal
VHS (Vps27, Hrs, STAM) domain, a GAT (GGA and TOM1) domain, a hinge region and
a C-terminal γ-adaptin “ear” (GAE) domain. The VHS domain binds to acidic
cluster/dileucine (DXXLL (where X is any amino acid)) sorting signals that are present in
the cytoplamic tail of various transmembrane proteins such as mannose-6-phosphate
receptor (MPR) (Puertollano et al. 2001; Zhu et al. 2001), sortilin (Nielsen et al. 2001), βsecretase (He et al. 2002) and the LDLR member, LRP3 (Takatsu et al. 2001). The GAT
domain binds to the Arf family of GTP-binding protein, the hinge region binds to clathrin
and the GAE domain binds to several accessory proteins (reviewed by Bonifacino 2004).
Through these multiple interactions, GGAs are involved in the sorting of DXXLLcontaining receptors into coated vesicles.
In this study, we describe for the first time the intracellular localization and
trafficking of LRP9. We observed by confocal microscopy that LRP9 was predominantly
localized at the TGN, where it co-localized with GGA proteins, and to a lesser extent, in
endosomes. We also demonstrated that the VHS domain of GGA proteins bound to two
functional DXXLL motifs in the cytoplasmic tail of LRP9. LRP9 is the first reported
233
example of a transmembrane protein with an internal GGA-binding sequence in addition to
the usual C-terminal motif. Interestingly, mutation (LL → AA) of both DXXLL motifs
prevented the interaction of LRP9 with GGA proteins and caused a redistribution of LRP9
in early endosomes and at the plasma membrane. These data demonstrated that LRP9
cycles between the TGN, endosomes and the plasma membrane and suggested that GGA
proteins participate in the regulation of LRP9 trafficking.
Materials and methods
Antibodies and reagents
Rabbit antibodies against mannosidase II were generously provided by Dr. Marilyn
Gist Farquhar (University of California, San Diego, CA, USA). Anti-HA monoclonal
antibodies (mAbs) were purchased from Covance (Berkley, CA, USA), anti-Myc mAbs
from Cell Signaling Technology (Danvers, MA, USA), anti-FLAG mAbs from Sigma (St
Louis, MO, USA) and anti-GM130 mAbs from BD Transduction Laboratories (Franklin
Lakes, NJ, USA). Anti-GFP polyclonal antibodies (pAbs) were purchased from Molecular
Probes (Eugene, OR, USA), anti-HA pAbs from Covance, anti-EEA1 pAbs from Affinity
Bioreagents (Golden, CO, USA), and anti-TGN46 pAbs from Novus Biologicals (Littleton,
CO, USA). Brefeldin A and monensin were obtained from Sigma.
DNA constructs
Expression vectors for GGA1-Myc, GGA2-Myc and GGA3-Myc were kindly
provided by Dr. Juan Bonifacino (National Institutes of Health, Bethesda, MD, USA).
pcDNA-FLAG-tagged furin was kindly provided by Dr. Richard Leduc (Université de
Sherbrooke, QC, Canada). Mammalian expression vector pMKITNeo encoding the murine
LRP9-HA was kindly provided by Dr. T. Kitamura (University of Tokyo, Japan). PCRbased mutagenesis was used to generate the LRP9 mutant constructs. To generate the
LRP9-proximal mutant, Leu692, Leu693 and Leu694 in the C-terminal sequence DDVLLL
were replaced by alanines. To generate the LRP9-distal mutant, Leu711 and Leu712 in the
C-terminal sequence DEPLLA were replaced by alanines. To generate the double LRP9
mutant, Leu692, Leu693, Leu694, Leu711 and Leu712 in the two DXXLL motifs in the Cterminal sequences (DDVLLL and DEPLLA) were replaced by alanines. The sequences of
the DNA constructs generated by PCR were systematically verified.
Cell culture and transfection
COS7 cells were obtained from Dr. Klaus Hahn (University of North Carolina,
Chapel Hill, NC, USA) and HEK293T cells were obtained from Dr. Alexandra Newton
(University of California, San Diego, CA, USA). The cells were grown in Dulbecco’s
modified Eagle’s high glucose medium (Invitrogen, Carlsbad, CA, USA) containing 10%
foetal bovine serum (FBS) (Hyclone Laboratories, Logan, UT, USA), penicillin, and
streptomycin. COS7 cells were transfected using Fugene6 transfection reagent (Roche
Diagnostics, Indianapolis, IN, USA), while HEK cells were transfected using
Lipofectamine 2000 transfection reagent (Invitrogen), both according to the manufacturers’
instructions.
234
Immunofluorescence
COS7 cells were plated on coverslips. Twelve hours after transfection, the cells
were fixed in 3% paraformaldehyde (PFA) in 100 mM phosphate buffer, pH 7.4 for 30 min,
permeabilized with 0.1% Triton X-100 for 10 min, blocked with 10% goat serum for 30
min, and incubated with primary antibodies for 1 h at RT, followed by Alexa Fluor-594 or
488-conjugated antibodies (Molecular Probes) for 1 h at RT. Specimens were visualized
using an inverted confocal laser-scanning microscope (FV1000, Olympus, Tokyo, Japan)
with a PlanApo 60x/1.42 oil immersion objective (Olympus). Olympus Fluoview software
version 1.6a was used for image acquisition and analysis. The images were further
processed using Adobe Photoshop (Adobe Systems, San Jose, CA, USA).
Glutathione S-transferase (GST) pull-down assays
GST constructs encoding GGA1-VHSGAT (residues 5–314), GGA1-VHS (residues
5–147), GGA1-GAT (residues 148–314), GGA1-Hinge (residues 315–514), GGA1-GAE
(residues 515–639) and GGA3-VHS (residues 1–146) were generated by standard PCR
methods and subcloned into pGEX-KG (Amersham-Pharmacia Biotech, Piscataway, NJ,
USA). GST fusion proteins were expressed in E. Coli BL21 and purified on glutathioneSepharose 4B beads (GE Healthcare, Piscataway, NJ, USA) according to the
manufacturer’s instructions. Cells were lysed in buffer (50 mM Tris pH 7.4, 200 mM NaCl,
1% NP40 and complete protease inhibitors (Roche)) for 1 h at 4°C and then centrifuged at
13,000×g for 20 min. One mg of cleared lysate was incubated overnight at 4°C with 10 µg
GST fusion proteins immobilized on beads. Beads were washed three times in lysis buffer
and boiled in Laemmli sample buffer. Bound proteins were analyzed by SDS-PAGE and
detected by immunoblotting.
Coimmunoprecipitation
HEK cells were plated in 60 mm culture dishes and transfected with the various
constructs. After 48 h, the cells were lysed in 50 mM Tris buffer (pH 7.4) containing 100
mM NaCl, 1% NP40 and protease inhibitors for 1 h at 4°C and then centrifuged at 13,000
rpm for 20 min. Cleared supernatants were incubated with primary antibodies overnight at
4° and then with protein A-sepharose (GE Healthcare, Piscataway, NJ, USA) or protein GSepharose (Zymed, San Francisco, CA, USA) for 1 h. The beads were washed three times
in lysis buffer and boiled in Laemmli sample buffer. Bound immune complexes were
analyzed by SDS-PAGE and immunoblotting.
Immunoblotting
Protein samples were separated on 10% SDS-PAGE gels and transferred to
nitrocellulose membranes (Perkin Elmer, Woodbridge, ON, Canada). Membranes were
blocked in Tris-buffered saline (20 mM Tris–HCl, pH 7.5, 150 mM NaCl) containing 0.1%
Tween 20 and 5% nonfat dry milk and incubated with primary antibodies for 2 h at RT and
then with horseradish peroxidase-conjugated goat anti-rabbit or anti-mouse IgG (Bio-Rad,
Richmond, ON, Canada) and enhanced chemiluminescence detection (Pierce Chemical,
Rockford, IL, USA).
235
Results
LRP9 localizes in the trans-Golgi network and early endosomes in COS7 cells
The cellular distribution of LRP9 has never been investigated. To characterize the
intracellular localization of this receptor, we compared the distribution of LRP9 with
several specific intracellular markers by confocal microscopy. HA-tagged LRP9 expressed
in COS7 cells was not detected at the cell surface but was primarily localized in the
juxtanuclear compartment as well as in some scattered vesicles (Fig. 1a, d, g). The
juxtanuclear distribution of LRP9 did not significantly co-localize with α-mannosidase II
(Fig. 1a–c) or galactosyl transferase (Fig. 1d–f), which are markers of the medial- and
trans-Golgi cisternae, respectively. However, extensive co-localization with furin (Fig. 1g–
i) and TGN46 (Fig. 2e–h), which are markers of the TGN, was observed. To determine
whether LRP9 is a Golgi resident protein or a cargo protein in transit, COS7 cells
transfected with LRP9-HA were treated for 1 h with cycloheximide, a protein synthesis
inhibitor. This treatment did not alter the distribution of LRP9-HA, which suggested that
LRP9, which has a half-life of 4 h, remains in the Golgi for a long period of time and is not
just in transit to the plasma membrane (data not shown).
Figure 1 - Comparison of LRP9 distribution with different Golgi markers in COS7
cells.
COS7 cells transfected with LRP9-HA alone (a–c) or together with galactosyl transferaseYFP (d–f) or furin-FLAG (G-I) were fixed, permeabilized and immunostained using antiHA (a, d, g), anti-mannosidase II (b), anti-GFP (e), anti-FLAG (h) antibodies. Stained cells
were examined by confocal fluorescence microscopy. Yellow colour in the merged images
(c, f, i) indicates co-localization. Scale bar 10 µm.
236
Figure 2 - Effect of brefeldin A and monensin treatments on LRP9 intracellular
localization.
237
COS7 cells transfected with LRP9-HA were incubated in either the absence (a–h) or
presence of 10 µg/µl brefeldin A (i–p) or 10 µM monensin (q–x) for 60 min. Cells were
fixed, permeabilized and immunostained using anti-HA (b, f, j, n, r, v), anti-GM130 (a, i, q)
and anti-TGN46 (e, m, u) antibodies. Stained cells were examined by confocal fluorescence
microscopy. Yellow colour in the merged images indicates co-localization. Last column
show ×2.5 magnified views of the boxes. Scale bar 10 µm.
To confirm that LRP9-HA specifically localized to the TGN compartment,
experiments were performed with brefeldin A (BFA) and monensin, two Golgi perturbing
agents. BFA is a fungal metabolite known to inhibit coat formation on the Golgi
membranes, thereby disrupting the cisternal stacks leading to a redistribution of the cis-,
medial- and trans-cisternae in the endoplasmic reticulum, leaving the TGN to coalesce into
a spherical mass near the microtubule organizing centre (MTOC; (Banting and
Ponnambalam 1997; Chardin and McCormick 1999; Watson and Pessin 2000). After BFA
treatment, LRP9-HA showed a concentrated vesicular distribution that colocalized with the
TGN marker TGN46 (Fig. 2m–p). This differs from the cis-Golgi marker GM130 (Fig. 2i–
l) which remained associated with dispersed punctate structures as previously described
(Nakamura et al. 1995; Xu et al. 2002). Monensin is an ionophore that disrupts the acidic
pH of luminal compartments and causes the accumulation of some Golgi proteins such as
TGN38/46 and GPP130 in swollen vesicles that has been defined as derivative of the TGN
(Schaub et al. 2006) or endosomes (Linstedt et al. 1997; Puri et al. 2002). Monensin
treatment of COS7 cells led to the relocalization of LRP9-HA and TGN46 to swollen
vesicles (Fig. 2u–x) while GM130 staining remained in a characteristic perinuclear Golgi
pattern (Fig. 2q–t). These results support the TGN localization of LRP9. Confocal
microscopy analysis of the intracellular localization of LRP9-HA also showed a partial colocalization with EEA1, an early endosome marker (Fig. 3a–c). These LRP9-EEA1labelled vesicles mainly localized in the Golgi area and not at the periphery of the cells
(Fig. 3a–c). The presence of LRP9 in EEA1-labelled endosomes was confirmed in COS7
cells transfected with an established Rab5 GTPase-deficient mutant (Rab5Q79L) that
greatly enlarges early endosomes due to increased homo- and heterotypic fusion (Stenmark
et al. 1994) (Fig. 3d–f). These observations suggest that LRP9 predominantly localizes to
the TGN and, to a lesser extent, to early endosomes at steady state in COS7 cells.
LRP9 co-localizes with GGA proteins in COS7 cells
GGA proteins play a role of molecular adaptors in the transport and retrieval of
cargo proteins between the TGN and endosomal compartments (Puertollano et al. 2001;
Zhu et al. 2001; Ghosh et al. 2003; Bonifacino 2004). GGA proteins directly bind to acidic
cluster/dileucine (DXXLL) motifs located at the extreme end of the C-termini of cargo
proteins (Bonifacino 2004; He et al. 2005). Since LRP9 localized to the TGN and
endosomes (Figs. 1–3) and since the C-terminal domain of LRP9 possesses a DXXLL
motif similar to the one found in transmembrane proteins reported to bind GGAs (LRP9distal; Fig. 5a, b), we postulated that GGA proteins might interact with LRP9 and
consequently regulate its intracellular trafficking. To test this hypothesis, we first examined
238
the co-localization of GGA isoforms with LRP9 by confocal microscopic imaging. This
was performed using COS7 cells expressing HA-tagged LRP9 together with Myc-tagged
GGA1, GGA2 or GGA3. GGA1-Myc displayed both juxtanuclear and vesicular
distributions that substantially overlapped those of LRP9-HA (Fig. 4a–c). A similar
distribution pattern was observed for GGA3-Myc (Fig. 4g–i) whereas partial colocalization was observed between GGA2-Myc and HA-LRP9 (Fig. 4d–f). These results
demonstrated that GGA proteins broadly co-localize with LRP9 and suggested that GGA
may be involved in the TGN-endosome transport of LRP9 through a potential interaction
with its C-terminal domain.
Figure 3 - LRP9 partially co-localizes with EEA1 on endosomes.
COS7 cells were co-transfected with HA-tagged LRP9 alone (a–c) or together with
Rab5Q79L, a Rab5 GTPase-deficient mutant that creates enlarged early endosomes (d–f).
Cells were fixed, permeabilized and immunostained using anti-HA (a, d) and anti-EEA1 (b,
e) antibodies. Stained cells were examined by confocal fluorescence microscopy. Yellow
colour in the merged images (c, f) indicates co-localization. Scale bar 10 µm.
239
Figure 4 - LRP9 co-localizes with GGA proteins at the TGN in COS7 cells.
COS7 cells co-transfected with HA-tagged LRP9 (a, d, g) together with Myc-tagged GGA1
(b), GGA2 (e) or GGA3 (h) were fixed, permeabilized and immunostained using anti-HA
(a, d, g) or anti-Myc (b, e, h) antibodies. Stained cells were examined by confocal
fluorescence microscopy. Yellow colour in the merged images (c, f, i) indicates colocalization. Scale bar 10 µm.
240
Figure 5 - LRP9 interacts with GGA proteins via a proximal and distal DXXLL motif
in its cytoplasmic tail.
a Schematic representation of the cytoplasmic tail of LRP9 containing two DXXLL motifs:
proximal and distal (bold face letters). Underlined leucines were substituted by alanines. b
Alignment of the C-terminal amino acids of transmembrane proteins that contain acidic
cluster/dileucine signals in their cytosolic tails. The two crucial leucine residues are shown
in red and the crucial aspartate is shown in blue. The position of the transmembrane domain
(Tm) and the number of residues before and/or after the signals is indicated. CD-MPR,
cation-dependent mannose-6-phosphate receptor; CI-MPR, cation-independent mannose-6phosphate receptor; LRP, low-density lipoprotein receptor related protein. c–d Coimmunoprecipitations of wild-type and mutant LRP9 with GGA proteins. Lysates from
HEK cells transfected with wild-type (WT) or DXXLL mutant LRP9-HA [proximal (Pr),
241
distal (Di) or double (Do)] and GGA1-Myc, GGA2-Myc or GGA3-Myc were
immunoprecipitated with anti-HA (c, lanes 2–13), anti-Myc (d, lanes 2–13) or control (Ctl)
(c, d, lanes 1 and 14) antibodies and then immunoblotted with anti-Myc or anti-HA to
detect GGA proteins and LRP9, respectively. Asterisk denotes longer exposure to show
GGA1-Myc bands with same intensity as GGA3-Myc and GGA2-Myc. It highlights the
fact that the interaction between LRP9-WT and GGA1-Myc is weaker than with GGA2Myc and GGA3-Myc. Note that the level of expression of Myc-GGA3 is approximatively
six times the level of endogenous GGA3 proteins.
GGA proteins interact with two DXXL motifs in the cytoplasmic tail of LRP9
To determine whether GGA proteins bind to LRP9, we first tested whether GGA
proteins co-immunoprecipitate with LRP9 in HEK293 cells transfected with both HAtagged wild-type LRP9 (WT) and Myc-tagged GGA1, GGA2 or GGA3. When the
immunoprecipitations were carried out using anti-HA IgGs, GGA1, GGA2 and GGA3 coimmunoprecipitated with LRP9-HA (Fig. 5c, lanes 2, 6, 10). However, GGA2-Myc and
GGA3-Myc bind better to LRP9-HA than GGA1-Myc. The same results were obtained
when the immunoprecipitation was carried out using anti-Myc IgGs. HA-LRP9 coimmunoprecipitated with GGA1-Myc, GGA2-Myc and GGA3-Myc (Fig. 5d, lanes 2, 6,
10), thus confirming the interaction of GGA proteins and LRP9 in cultured cells.
Since an inactivating mutation of the dileucine residues of the DXXLL sequence
has previously been reported to abrogate the interaction with GGA proteins (Nielsen et al.
2001; Puertollano et al. 2001; Takatsu et al. 2001; Zhu et al. 2001; He et al. 2002), leucines
711 and 712 were replaced by alanines in the DXXLL motif at the extreme C-terminus of
the cytoplasmic tail of HA-LRP9 (named distal DXXLL, Fig. 5a). Interactions between the
various GGA-Myc proteins and this LRP9 mutant were then investigated using the coimmunoprecipitation assays described above with either anti-HA (Fig. 5c) or anti-Myc
IgGs (Fig. 5d). The interaction between LRP9 and GGA1 was almost completely abolished
(Fig. 5c, d, lane 4) by this mutation. However, the interactions between LRP9 and GGA2
and GGA3 were not affected (Fig. 5c, d, lanes 8, 12). These results suggested that the distal
DXXLL motif is not the only key element that contributes to the interaction with GGA
proteins. After further examination of the C-terminal sequence of LRP9, another DXXLL
motif was identified thirteen amino acids upstream from the distal DXXLL (Fig. 5a). This
new DXXLL motif was composed of two overlapping DXXLL motifs (D689 D690 V691
L692 L693 L694) and was called the proximal DXXLL motif (Fig. 5a, b). Alanines were
substituted for Leu692, Leu693 and Leu694 in the proximal DXXLL motif and interactions
between GGA proteins and the mutated LRP9 were analyzed by immunoprecipitation using
either anti-HA IgGs (Fig. 5c) or anti-Myc IgGs (Fig. 5d). As shown in Fig. 5c and d (lanes
3, 7, 11), the mutations introduced into the proximal DXXLL did not abrogate the
interaction of LRP9 with GGA1, GGA2 or GGA3. This indicated that both the proximal
and distal DXXLL motifs might be responsible for the binding of GGA proteins to LRP9.
To confirm this, leucines (Leu692, Leu693, Leu694, Leu711 and Leu712) were substituted
by alanines in both the proximal and distal DXXLL motifs of LRP9 cytoplasmic tail (Fig.
5a). This double DXXLL mutation completely inhibited the interaction of LRP9 with the
242
three GGA proteins (Fig. 5c, d; lanes 5, 9, 13), indicating that the C-terminal domain of
LRP9 contained two DXXLL motifs that were able to bind to GGA proteins. This
discovery is of particular interest because LRP9 is the first example of a transmembrane
protein with an internal GGA-binding sequence in addition to the usual C-terminal motif.
Figure 6 - LRP9 interacts with the VHS domain of GGA proteins.
a Diagram of GGA1 showing the VHS (Vps27, Hrs, Stam) domain, the GAT (GGA and
TOM) domain, the hinge segment and the GAE (γ-adaptin ear) domain (Bonifacino 2004).
b–c The indicated GST-fusion proteins immobilized on glutathione beads were incubated
with lysates from HEK cells expressing wild-type (WT) LRP9-HA or cation dependant
mannose-6-phosphate receptor–GFP (CD-MPR-GFP, b) or LRP9-HA in which the two
DXXLL motifs were mutated (Double, c). Bound proteins were separated by SDS-PAGE
and immunoblotted with anti-HA or anti-GFP antibodies to detect LRP9 and CD-M6PR,
respectively.
We next determined which domain of GGA proteins mediates the interaction with
LRP9 using pull-down assays. Glutathione-S-transferase (GST) fusion proteins comprising
the various GGA1 domains (Fig. 6a) were incubated with lysates from HEK293 cells
expressing HA-tagged wild-type LRP9 (Fig. 6b). As a positive control, the pull-down-assay
were also performed with lysates from cells expressing GFP-tagged cation-dependant
mannose-6-phosphate receptor (CD-M6PR-GFP) that contains a DXXLL motif reported to
interact with the VHS domain of GGA1 (Puertollano et al. 2001). Similar to CD-M6PR243
Figure 7 - Mutations in both the proximal and distal DXXLL motifs of LRP9 affect its
intracellular distribution.
a–i In COS7 cells transfected with GGA1-Myc together with wild-type LRP9-HA (WT, a)
or LRP9-HA in which the distal DXXLL motif (Distal, d) or proximal DXXLL motif
(Prox., g) were mutated, LRP9-HA co-localized extensively with GGA1-Myc (b, e, h) in
the juxtanuclear Golgi region (arrowheads, c, f, i) and on vesicular structures (inset, c, f, i).
j–l In cells transfected with GGA1-Myc and LRP9-HA in which the two DXXLL motifs
(double, j) were mutated, LRP9 was mainly present on the cell surface (arrows, j) and in
peripheral punctate structures (inset, J) and did not significantly co-localize with GGA1244
Myc in the Golgi region (arrowhead, l) or on vesicular structures (inset, l). Cells were fixed,
permeabilized and immunostained using anti-HA (a, d, g, j) or anti-Myc (b, e, h, k)
antibodies. Stained cells were examined by confocal fluorescence microscopy. Yellow
colour in the merged images (c, f, i, l) indicates co-localization. Insets show ×2 magnified
views of cytoplasmic regions. Scale bar 10 µm.
GFP, LRP9-HA bound strongly to GST-GGA1-VHS or GST-GGA1-VHSGAT and weakly
to GST-GGA1-GAT (Fig. 6b). In contrast, no interaction was observed with GST, GSTGGA1-Hinge and GST-GGA1-GAE (Fig. 6b). Similar results were obtained with GST
fusion proteins comprising the various GGA3 domains (data not shown). These results
demonstrated that LRP9 interacts predominantly with the VHS domain of GGA proteins.
The importance of the DXXLL motifs in LRP9 interaction with the VHS domain of GGA
was next examined. To this end, interactions of GST-GGA1-VHS with the double DXXLL
mutants of LRP9-HA were compared to wild-type LRP9-HA. As shown in Fig. 6c,
mutation of both DXXLL motifs completely inhibited the interaction of LRP9 with the
VHS domain of GGA1. Similar results were obtained with GST-GGA3-VHS (data not
shown). These results indicate that the DXXLL motifs of LRP9 interact specifically with
the VHS domain of GGA proteins. This is in agreement with previous studies showing that
VHS is the specific GGA domain that binds to the DXXLL motif of various of
transmembrane proteins such as sortilin, LRP3, β-secretase and M6PR (Puertollano et al.
2001; Takatsu et al. 2001; He et al. 2002; Bonifacino 2004).
Mutations in both the proximal and distal DXXLL motifs induce a redistribution of
LRP9 to the plasma membrane and peripheral endosomes
To investigate the role of the DXXLL motifs in LRP9 trafficking, the subcellular
distributions of the proximal, distal and double DXXLL mutants were compared to wildtype LRP9. COS7 cells were transfected with Myc-tagged GGA1 together with HA-tagged
wild-type LRP9 or the mutant DXXLLs and were analyzed by confocal microscopy. The
distributions of LRP9-HA mutated in the distal- (Fig. 7d) or proximal-DXXLL (Fig. 7g)
motifs were identical to the wild-type LRP9-HA (Fig. 7a). They all extensively colocalized with GGA1 in the juxtanuclear Golgi region (Fig. 7a–i, arrowheads) and in
cytoplasmic vesicles (insets, Fig. 7a–i). In contrast, the double DXXLL mutant mainly
localized at the cell surface and in peripheral punctate structures (Fig. 7j) and did not colocalize with GGA1 (Fig. 7l, inset). Similar data were obtained with cells expressing Myctagged GGA2 and Myc-tagged GGA3 (data not shown). We noticed that the localization of
Myc-GGA1 was also modified in cells expressing the double DXXLL mutant. GGA1 was
less concentrated in the Golgi region in these cells (Fig. 7k) compared to cells expressing
LRP9-WT (Fig. 7b), distal (Fig. 7e) or proximal mutants (Fig. 7h). This could be explained
by the fact that cargo proteins help recruit GGAs onto membranes (Hirst et al. 2007).
Indeed, it has been reported that the overexpression of CD8 chimeras with cytoplasmic tails
containing DXXLL-sorting signals, which bind to GGAs, increases the localization of all
three GGAs to juxtanuclear membranes (Hirst et al. 2007). In the same vein, we suggest
that the overexpression of LRP9 double DXXLL mutants that cannot bind GGA proteins
would decrease GGA labelling in the Golgi region compared to the cells that overexpress
LRP9 that can bind GGA proteins.
245
To better define the subcellular localization of the double DXXLL mutant, the cells
were transfected with HA-tagged wild-type LRP9 or the DXXLL mutants and were doublelabelled with endogenous EEA1, an early endosome marker. Figure 8a–i shows that the
wild-type LRP9 (Fig. 8a–c) and the distal DXXLL (Fig. 8d–f) and proximal DXXLL (Fig.
8g–i) mutants did not co-localize with peripheral EEA1-labelled structures and were not
present at the plasma membrane. In contrast, the double DXXLL mutant was present at the
plasma membrane and co-localized with EEA1 in peripheral punctate structures (Fig. 8j–l).
In summary, mutations of both DXXLL motifs, which inhibited the interaction with GGA
proteins, shifted the distribution of LRP9 from the TGN to the plasma membrane and
peripheral EEA1-labelled endosomes. These observations suggested that GGA proteins
participate in the intracellular trafficking of LRP9.
Figure 8 - Redistribution of LRP9 double DXXLL mutant to the plasma membrane
and peripheral EEA1-positive compartments.
246
a–i In COS7 cells transfected with wild-type LRP9-HA (WT, a–c) or LRP9-HA in which
the distal DXXLL motif (Distal, d–f) or proximal DXXLL motif (Prox., g–i) were mutated,
LRP9-HA was not present in peripheral EEA1-labelled endosomes (inset, c, f, i). j–l In
cells transfected with LRP9-HA in which the two DXXLL motifs were mutated (Double, j–
l), LRP9-HA was mainly present on the cell surface (arrows, j, l) and in peripheral punctate
structures (arrowheads, j) that significantly co-localized with EEA1 (inset, l). Cells were
fixed, permeabilized and immunostained using anti-HA (a, d, g, j) and anti-EEA1 (b, e, h,
k) antibodies. Stained cells were examined by confocal fluorescence microscopy. Yellow
colour in the merged images indicates co-localization. Merged images (c, f, i, l) show ×2
magnified views of the boxes in a–l. Scale bar 10 µm.
Discussion
LRP9 is a distant member of the LDLR superfamily that has been reported to be
expressed in a number of tissues, notably kidney, liver, lung and heart tissue and to be
involved in the internalization of ApoE-enriched βVLDL (Sugiyama et al. 2000). However,
the intracellular distribution, subcellular targeting signals and physiological role of LRP9
remain to be clearly demonstrated. In this study, we characterized for the first time the
intracellular localization and trafficking of LRP9. Confocal microscopy studies showed that
LRP9 was not present at the PM [as confirmed by biotinylation assays (data not shown)]
but was predominantly distributed to the juxtanuclear region where it co-localized with
TGN markers such as TGN46 and GGA proteins. Localization to the TGN was supported
by the action of brefeldin A and monensin, two agents that promote Golgi disorganization.
These drugs induced a co-redistribution of LRP9 and TGN46 into the same intracellular
structures, consistent with a TGN localization of LRP9. In addition, a part of LRP9 staining
was detected in vesicles that overlapped with early endosome markers. Thus, our data
suggest that LRP9 predominantly localizes to the TGN and, to a lesser extent to early
endosomes, at steady state.
GGA proteins are known to interact with acidic cluster/dileucine (DXXLL) motifs
in the cytoplamic tail of transmembrane proteins (Nielsen et al. 2001; Puertollano et al.
2001; Takatsu et al. 2001; Zhu et al. 2001; He et al. 2002; Misra et al. 2002). Interestingly,
we noted that the cytoplasmic tail of LRP9 contained two DXXLL sequences, one at the
extreme end of the cytoplasmic tail (distal motif relative to the transmembrane domain,
residues 708–712) and another, thirteen amino acids upstream (proximal motif relative to
the transmembrane domain, residues 689–694). Using immunoprecipitation assays, we
demonstrated that LRP9 interacts strongly with GGA2 and GGA3 and more weakly with
GGA1. Simultaneous mutations in both DXXLL motifs completely inhibited the interaction
of LRP9 with the three GGA proteins. However, the substitution of LL by AA in the
proximal DXXLL motif did not abrogate the interaction of LRP9 with the three GGA
proteins. In addition, the LL → AA mutation in the distal DXXLL motif did not alter the
interaction of LRP9 with GGA2 and GGA3, although it almost completely abrogated the
interaction with GGA1. Therefore, both DXXLL motifs bound to GGA proteins but had
different affinities for GGA1. Indeed, the distal motif bound the three GGA proteins (albeit
247
more weakly to GGA1) whereas the proximal DXXLL motif bound mainly GGA2 and
GGA3. Significant differences in the affinity of GGA proteins for acidic dileucine motifs
have been previously reported. Although both CD-M6PR and CI-M6PR contain similar
DXXLL motifs near the C-terminus of their cytoplasmic tails (Fig. 5b), CI-M6PR binds to
GGA proteins with higher affinity than the CD-M6PR (Doray et al. 2002). There is also a
difference in the relative affinity of the DXXLL motif in the CD-M6PR cytoplasmic tail for
the different GGAs. Puertollano et al. (2001) showed that CD-MPR bound well to GGA1,
but only poorly to GGA3, and not at all to GGA2. In contrast, the cytoplasmic tail of CIMPR bound to the VHS domains of all three GGAs with about equal affinity. Comparison
of the C-terminal residues of these two M6PR (Fig. 5b) revealed a number of nonconservative differences in the acidic cluster/dileucine motifs and the flanking residues.
Mutation analysis revealed that several residues immediately upstream and downstream
from the DXXLL motif, as well as the XX residues of this motif, significantly impact the
specificity of the interactions of GGA proteins with these motifs (Puertollano et al. 2001;
Zhu et al. 2001; Doray et al. 2002; He et al. 2002; He et al. 2003). Alignment of the
proximal and distal acidic cluster dileucine motifs of LRP9 with the amino acid sequences
of the cytoplasmic tail of other transmembrane proteins containing a DXXLL motif (Fig.
5b) showed differences in residues in position +1 and +2 as well as in several residues
upstream and downstream from the proximal and distal DXXLL motifs. The importance of
these residues is now under investigation, and should provide new insights into selectivity
towards specific interactions with GGA proteins and the trafficking mechanisms of
DXXLL-mediated sorting signals.
LRP9 is the first example of a transmembrane protein with an internal GGA-binding
sequence in addition to the usual C-terminal motif. Using various protein sequence data
banks such as TrEMBL and Swiss-Prot, we were unable to identify other proteins with two
DXXLL motifs, suggesting that LRP9 might be the only transmembrane protein with two
functional DXXLL motifs. The VHS domain of GGA proteins has been identified as the
binding site of the DXXLL motifs of LRP9. This is in agreement with previous reports
defining VHS as the specific GGA domain that binds to the DXXLL motif (Bonifacino
2004; He et al. 2002; Nielsen et al. 2001; Puertollano et al. 2001; Takatsu et al. 2001).
LRP9 can now be added to the long list of proteins with DXXLL-type sorting signals that
bind to the VHS domain of the GGAs including sortilin, LRP3, β-secretase and M6PR
(Bonifacino 2004).
Previous studies have reported that interactions of GGA proteins with a DXXLL
motif in the cytoplasmic tail of various receptors mediate their anterograde or retrograde
transport between the TGN and endosomes (Huse et al. 2000; Tikkanen et al. 2000;
Tortorella et al. 2007). These studies, together with our observation that LRP9 interacted
and co-localized with GGA proteins, prompted us to investigate whether GGA proteins
were involved in the intracellular transport of LRP9. Mutations (LL → AA) in the proximal
or distal DXXLL motif did not affect the intracellular distribution of LRP9. Therefore,
proximal and distal DXXLL motifs alone seemed to be sufficient for the proper trafficking
of LRP9 since they both had the capacity to bind GGA proteins. In addition, since we did
not detect any substantial differences between the single DXXLL mutants, we concluded
that these two motifs might play redundant roles in the trafficking of LRP9. Mutations in
248
both DXXLL motifs that completely inhibited the interaction of LRP9 with the three GGA
proteins resulted in a redistribution of the receptor from the TGN to EEA1-labelled
endosomes and the plasma membrane. Similar mutations in the DXXLL motifs in the
cytoplasmic tails of other receptors such as MPR and mepapsin (BACE1) have also been
reported to cause a redistribution of these receptors from the TGN to endosomes and the
plasma membrane (Huse et al. 2000; Tikkanen et al. 2000; He et al. 2005; Tortorella et al.
2007), thus reinforcing our observations. Furthermore, downregulation of GGA proteins or
overexpression of dominant negative GGA mutants cause an accumulation of MPR in
endosomal compartments (Ghosh et al. 2003; Puertollano and Bonifacino 2004; He et al.
2005; Wahle et al. 2005). These receptor redistributions have been explained by a sorting
defect at different levels: (1) TGN sorting: GGA proteins normally retain DXXLL
receptors in the TGN and prevent premature exit from the Golgi apparatus to early
endosomes or from the Golgi apparatus via the secretory pathway to the cell surface where
they would then be rapidly internalized into early endosomes or (2) endosomal sorting:
GGA proteins are involved in early endosome-to-TGN retrieval of receptors containing a
DXXLL motif. These explanations and our results provide support for a potential role for
GGA adaptor proteins in sorting LRP9 between the TGN and endosomal compartments in
both anterograde and retrograde directions.
Our data thus suggested that LRP9 cycles between the TGN and early endosomes at
steady state. LRP9 can also reach the cell surface since the double DXXLL mutant relocalized at the plasma membrane. However, since there was no significant labelling of
wild-type LRP9 at the plasma membrane, we propose that either only a minor fraction of
wild-type LRP9 is transported to the cell surface or its presence at the plasma membrane is
very transient because the receptor is quickly internalized. Interestingly, Doray et al. (2007)
have recently reported that a chimera protein containing the cytoplasmic tail of LRP9
undergoes rapid endocytosis. Furthermore, like our observations, these authors showed that
a double DXXLL mutant (LL → AA) accumulates at the cell surface, impairing LRP9
internalization. They reported that the proximal and distal dileucine motifs of LRP9 bind
γ/σ1 AP1 and α/σ2 AP2 hemicomplexes and that the intracellular distribution of an LRP9
mutant that is unable to bind AP1 is not affected whereas the LRP9 mutant that is unable to
bind to both AP2 and AP1 is redistributed to the plasma membrane (Doray et al. 2007).
They concluded that there is a correlation between α/σ2 AP2 hemicomplex binding and
LRP9 chimera receptor internalization mediated by dileucine-based sorting signals. Since
current evidence does not support the involvement of GGA proteins in the internalization
steps, we suggest that the retention of the double DXXLL mutant at the cell surface we
observed (Figs. 7, 8) might be due to the loss of the interaction between LRP9 and AP2.
The DXXLL motifs of LRP9 thus appear to be required for sorting at two different steps in
LRP9 trafficking: (1) internalization from the plasma membrane via an interaction with
AP2 and (2) transport between the endosomes and the TGN via an interaction with the
GGA proteins. Further studies are now underway to characterize the exact trafficking
itinerary and kinetics of LRP9 and to identify the precise molecular machinery involved in
the intracellular sorting of LRP9.
Only a few other members of the LDLR family have been reported to bind GGA
proteins. LRP3 and LRP12, the two other members of the new LRP9 subgroup of the
249
LDLR superfamily, possess a single DXXLL motif in their cytoplasmic tail, suggesting that
they have the capacity to interact with GGA proteins. Indeed, LRP3 has already been
shown to interact with GGA proteins (Takatsu et al. 2001). However, the intracellular
trafficking and function of LRP3 and LRP12 remain uncharacterized. SorLA (sorting
protein related receptor; LR11) is another distant member of the LDLR family shown to
mediate the uptake of ApoE-rich lipoproteins in vitro (Taira et al. 2001), to interact with
GGA1 and GGA2 (Jacobsen et al. 2002) and to shuttle between the Golgi and endosomes
(Jacobsen et al. 2001; Andersen et al. 2005; Offe et al. 2006). Interestingly, SorLA/LR11
has been recently described as a sorting receptor that regulates the intracellular transport
and processing of the amyloid precursor protein (APP) (Andersen et al. 2005; Shah and Yu
2006; Schmidt et al. 2007) pointing to an involvement in Alzheimer’s disease.
To date, the only ligand identified for LRP9 is apoE-βVLDL (Sugiyama et al.
2000). However, since LRP9 is not mainly localized at the cell surface at steady state, it is
likely to serve other functions inside the cell. Different lines of evidence give hints towards
a possible function of LRP9. First, the intracellular distribution of LRP9 at steady state. We
observed that only a minor fraction of the receptor is expressed on the cell surface, whereas
the majority is found in TGN and endosomes. Secondly, LRP9 cytoplasmic domain
contains signal sequence involved in adaptor protein binding for endocytosis and Golgiendosome sorting. We and others have recently demonstrated that LRP9 interacts with
AP1, AP2 (Doray et al. 2007) and GGA proteins (this paper). Thirdly, many receptors
containing a DXXLL motif or a GGA-binding site, such as MPR (Tikkanen et al. 2000;
Puertollano et al. 2001; Tortorella et al. 2007), Sortilin (Nielsen et al. 2001) and SorLA
(Andersen et al. 2005; Nielsen et al. 2001), are sorting receptors that traffic between the
TGN, endosomes and the cell surface. It would appear from the above that LRP9 might be
a candidate sorting receptor, targeted for transport by ligands in the synthetic pathway as
well as on the surface membrane. Investigations are presently underway to test this
hypothesis and identify intracellular cargos for LRP9. Furthermore, there is increasing
evidence of a prominent role for certain members of the LDLR superfamily in signal
transduction pathways and cell physiology regulation (Strickland et al. 2002; May et al.
2007). This signalling occurs through cooperation with other cell surface molecules that
associate directly or indirectly with LDL receptor family members and cytoplasmic adaptor
molecules that interact to their cytoplasmic tails. The fact that signalling domains are
present in the cytoplasmic tail of LRP9 suggests that it might also play a role in signal
transduction. This hypothesis is supported by the work of Battle et al. (2003), who reported
that various proteins involved in signal transduction interact with a juxtamembrane region
of LRP12 that is conserved in LRP3 and LRP9. Further investigation is needed to clarify
the biological functions of LRP9 and the other members of this LDLR subfamily.
In conclusion, our study is the first to characterize the intracellular localization and
trafficking of LRP9, an unusual member of the LDLR family. Its predominantly subcellular
localization in the TGN and endosomes, together with its capacity to bind GGA proteins
via two DXXLL motifs, suggest that LRP9 cycles between the TGN and endosomes
through a GGA dependent-trafficking mechanism. These observations are important first
steps in understanding the role of this uncharacterized receptor and constitute evidences
pointing to a function of LRP9 in protein sorting and transport.
250
Acknowledgments
We would like to thank Juan Bonifacino, Richard Leduc and T. Kitamura for DNA
constructs and Marilyn Gist Farquhar for the generous gifts of reagents. We would also like
to thank Eric Chevet, Catherine Denicourt and Richard Leduc for their constructive
comments on the manuscript. This work was supported by grants from the Canadian
Institutes for Health Research and a Canada Research Chair to C.L.
References
Andersen OM, Reiche J, Schmidt V, Gotthardt M, Spoelgen R, Behlke J, von Arnim CA,
Breiderhoff T, Jansen P, Wu X (2005) Neuronal sorting protein-related receptor
sorLA/LR11 regulates processing of the amyloid precursor protein. Proc Natl Acad
Sci USA 102:13461–13466
Banting G, Ponnambalam S (1997) TGN38 and its orthologues: roles in post-TGN
vesicle formation and maintenance of TGN morphology. Biochim Biophys Acta
1355:209–217
Battle MA, Maher VM, McCormick JJ (2003) ST7 is a novel low-density lipoprotein
receptor-related protein (LRP) with a cytoplasmic tail that interacts with proteins
related to signal transduction pathways. Biochemistry 42:7270–7282
Bonifacino JS (2004) The GGA proteins: adaptors on the move. Nat Rev Mol Cell Biol
5:23–32
Bork P, Beckmann G (1993) The CUB domain. A widespread module in
developmentally regulated proteins. J Mol Biol 231:539–545
Chardin P, McCormick F (1999) Brefeldin A: the advantage of being uncompetitive.
Cell 97:153–155
Christensen EI, Birn H (2002) Megalin and cubilin: multifunctional endocytic receptors.
Nat Rev Mol Cell Biol 3:256–266PubMed
Doray B, Bruns K, Ghosh P, Kornfeld S (2002) Interaction of the cation-dependent
mannose 6-phosphate receptor with GGA proteins. J Biol Chem 277:18477–18482
Doray B, Lee I, Knisely J, Bu G, Kornfeld S (2007) The gamma/sigma1 and
alpha/sigma2 hemicomplexes of clathrin adaptors AP-1 and AP-2 harbor the
dileucine recognition site. Mol Biol Cell 18:1887–1896
Ghosh P, Kornfeld J (2004) The GGA proteins: key players in protein sorting at the
trans-Golgi network. Eur J Cell Biol 83:257–262
Ghosh P, Griffith J, Geuze H, Kornfeld S (2003) Mammalian GGAs act together to sort
mannose-6-phosphate receptors. J Cell Biol 163:755–766
He X, Chang WP, Koelsch G, Tang J (2002) Memapsin 2 (beta-secretase) cytosolic
domain binds to the VHS domains of GGA1 and GGA2: implications on the
endocytosis mechanism of memapsin 2. FEBS Lett 524:183–187PubMed
He X, Li F, Chang WP, Tang J (2005) GGA proteins mediate the recycling pathway of
memapsin 2 (BACE). J Biol Chem 280:11696–11703
He X, Zhu G, Koelsch G, Rodgers KK, Zhang XC, Tang J (2003) Biochemical and
structural characterization of the interaction of memapsin 2 (beta-secretase)
cytosolic domain with the VHS domain of GGA proteins. Biochemistry 42:12174–
12180
251
Herz J (2001) The LDL receptor gene family: (un) expected signal transducers in the
brain. Neuron 29:571–581
Hirst J, Seaman MN, Buschow SI, Robinson MS (2007) The role of cargo proteins in
GGA recruitment. Traffic 8:594–604
Huse JT, Pijak DS, Leslie GJ, Lee VM, Doms RW (2000) Maturation and endosomal
targeting of beta-site amyloid precursor protein-cleaving enzyme. The Alzheimer’s
disease beta-secretase. J Biol Chem 275:33729–33737
Hussain MM, Strickland DK, Bakillah A (1999) The mammalian low-density lipoprotein
receptor family. Annu Rev Nutr 19:141–172
Jacobsen L, Madsen P, Jacobsen C, Nielsen MS, Gliemann J, Petersen CM (2001)
Activation and functional characterization of the mosaic receptor SorLA/LR11. J
Biol Chem 276:22788–22796
Jacobsen L, Madsen P, Nielsen MS, Geraerts WP, Gliemann J, Smit AB, Petersen CM
(2002) The sorLA cytoplasmic domain interacts with GGA1 and -2 and defines
minimum requirements for GGA binding. FEBS Lett 511:155–158
Kikuchi A, Yamamoto H, Kishida S (2007) Multiplicity of the interactions of Wnt
proteins and their receptors. Cell Signal 19:659–671
Linstedt AD, Mehta A, Suhan J, Reggio H, Hauri HP (1997) Sequence and
overexpression of GPP130/GIMPc: evidence for saturable pH-sensitive targeting of
a type II early Golgi membrane protein. Mol Biol Cell 8:1073–1087PubMed
May P, Woldt E, Matz RL, Boucher P (2007) The LDL receptor-related protein (LRP)
family: an old family of proteins with new physiological functions. Ann Med
39:219–228
Misra S, Puertollano R, Kato Y, Bonifacino JS, Hurley JH (2002) Structural basis for
acidic-cluster-dileucine sorting-signal recognition by VHS domains. Nature
415:933–937
Nakamura N, Rabouille C, Watson R, Nilsson T, Hui N, Slusarewicz P, Kreis TE,
Warren G (1995) Characterization of a cis-Golgi matrix protein, GM130. J Cell
Biol 131:1715–1726
Nielsen MS, Madsen P, Christensen EI, Anders Nykjær Gliemann J, Kasper D,
Pohlmann R, Petersen CM (2001) The sortilin cytoplasmic tail conveys Golgi–
endosome transport and binds the VHS domain of the GGA2 sorting protein.
EMBO J 20:2180–2190
Offe K, Dodson SE, Shoemaker JT, Fritz JJ, Gearing M, Levey AI, Lah JJ (2006) The
lipoprotein receptor LR11 regulates amyloid beta production and amyloid precursor
protein traffic in endosomal compartments. J Neurosci 26:1596–1603
Puertollano R, Bonifacino JS (2004) Interactions of GGA3 with the ubiquitin sorting
machinery. Nat Cell Biol 6:244–251
Puertollano R, Aguilar RC, Gorshkova I, Crouch RJ, Bonifacino JS. (2001) Sorting of
mannose 6-phosphate receptors mediated by the GGAs. Science 292:1712–1716
Puri S, Bachert C, Fimmel CJ, Linstedt AD (2002) Cycling of early Golgi proteins via
the cell surface and endosomes upon lumenal pH disruption. Traffic 3:641–653
Schaub BE, Berger B, Berger EG, Rohrer J (2006) Transition of galactosyltransferase 1
from trans-Golgi cisterna to the trans-Golgi network is signal mediated. Mol Biol
Cell 17:5153–5162
252
Schmidt V, Sporbert A, Rohe M, Reimer T, Rehm A, Andersen OM, Willnow TE.
(2007) SorLA/LR11 regulates processing of amyloid precursor protein via
interaction with adaptors GGA and PACS-1. J Biol Chem 282:32956–32964
Shah S, Yu G (2006). sorLA: sorting out APP. Mol Interv 6(74–76):58
Stenmark H, Parton RG, Steele-Mortimer O, Lutcke A, Gruenberg J, Zerial M (1994)
Inhibition of rab5 GTPase activity stimulates membrane fusion in endocytosis.
Embo J 13:1287–1296PubMed
Strickland DK, Gonias SL, Argraves WS. (2002) Diverse roles for the LDL receptor
family. Trends Endocrinol Metab 13:66–74
Sugiyama T, Kumagai H, Morikawa Y, Wada Y, Sugiyama A, Yasuda K, Yokoi N,
Tamura S, Kojima T, Nosaka T et al (2000) A novel low-density lipoprotein
receptor-related protein mediating cellular uptake of apolipoprotein E-enriched
beta-VLDL in vitro. Biochemistry 39:15817–15825
Taira K, Bujo H, Hirayama S, Yamazaki H, Kanaki T, Takahashi K, Ishii I, Miida T,
Schneider WJ, Saito Y (2001) LR11, a mosaic LDL receptor family member,
mediates the uptake of ApoE-rich lipoproteins in vitro. Arterioscler Thromb Vasc
Biol 21:1501–1506
Takatsu H, Katoh Y, Shiba Y, Nakayama K. (2001) Golgi-localizing, gamma-adaptin ear
homology domain, ADP-ribosylation factor-binding (GGA) proteins interact with
acidic dileucine sequences within the cytoplasmic domains of sorting receptors
through their Vps27p/Hrs/STAM (VHS) domains. J Biol Chem 276:28541–28545
Tikkanen R, Obermuller S, Denzer K, Pungitore R, Geuze HJ, von Figura K, Honing S
(2000) The dileucine motif within the tail of MPR46 is required for sorting of the
receptor in endosomes. Traffic 1:631–640
Tortorella LL, Schapiro FB, Maxfield FR. (2007) Role of an acidic cluster/dileucine
motif in cation-independent mannose 6-phosphate receptor traffic. Traffic 8:402–
413
Wahle T, Prager K, Raffler N, Haass C, Famulok M, Walter J (2005) GGA proteins
regulate retrograde transport of BACE1 from endosomes to the trans-Golgi
network. Mol Cell Neurosci 29:453–461
Watson RT, Pessin JE. (2000) Functional cooperation of two independent targeting
domains in syntaxin 6 is required for its efficient localization in the trans-golgi
network of 3T3L1 adipocytes. J Biol Chem 275:1261–1268
Willnow TE (1999) The low-density lipoprotein receptor gene family: multiple roles in
lipid metabolism. J Mol Med 77:306–315
Xu Y, Martin S, James DE, Hong W (2002) GS15 forms a SNARE complex with
syntaxin 5, GS28, and Ykt6 and is implicated in traffic in the early cisternae of the
Golgi apparatus. Mol Biol Cell 13:3493–3507
Yamamoto TDC, Brown MS, Schneider WJ, Casey ML, Goldstein JL, Russell DW
(1984) The human LDL receptor: a cysteine-rich protein with multiple Alu
sequences in its mRNA. Cell 39:27–38
Zhu Y, Doray B, Poussu A, Lehto VP, Kornfeld S. (2001) Binding of GGA2 to the
lysosomal enzyme sorting motif of the mannose 6-phosphate receptor. Science
292:1716–1718
253
ANNEXE 2
Calnuc Binds to LRP9 and Affects its Endosomal
Sorting.
Brodeur, J., Larkin, H., Boucher, R., Gagnon, H., Chayer St-Louis,
S., and Lavoie, C.
Traffic 10(8):1098-114 (2009)
Travaux de maîtrise
IF = 6,255
254
Calnuc Binds to LRP9 and Affects its Endosomal Sorting
Brodeur, J., Larkin, H., Boucher, R., Gagnon, H., Chayer St-Louis, S., and Lavoie, C.
Abstract
Calnuc is an ubiquitous Ca2+-binding protein found in the cytoplasm where it binds
different Gα subunits, in the Golgi lumen where it constitutes a major Ca2+ storage pool,
and outside the cell. We identified LDLR-related protein 9 (LRP9) as the first
transmembrane protein shown to interact directly with Calnuc. LRP9 is a member of a new
subfamily of the LDLR superfamily that cycles between the trans-Golgi network (TGN)
and endosomes through a mechanism dependent on clathrin adaptor GGA proteins. The
aim of the present study was to characterize the interaction between Calnuc and LRP9.
Various biochemical assays showed that the N-terminus of Calnuc interacts with an
arginine-rich region in the cytosolic tail of LRP9. Confocal microscopy showed that Calnuc
colocalizes with LRP9 at the surface of the TGN and early endosomes. Depletion of Calnuc
by small interfering RNA (siRNA) missorted LRP9 in the late endosome/lysosome
compartments and enhanced its lysosomal degradation. This phenotype was rescued by the
expression of siRNA-resistant wild-type Calnuc as well as cytoplasmic Calnuc, indicating
that the cytoplasmic pool of Calnuc is involved in LRP9 endosomal sorting to prevent the
delivery of LRP9 to lysosomes. This is the first report showing that Calnuc plays a role in
receptor trafficking.
Keywords
Calnuc; endosomal sorting; GGA; LDLR-related protein 9; lysosome; nucleobindin;
trans-Golgi network
Introduction
Calnuc, also known as nucleobindin, is an ubiquitous (1–3), EF-hand calciumbinding protein that is conserved from flies to humans, suggesting that it has important
biological functions (3,4). Indeed, the modulation of Calnuc expression is associated with
various pathologies, including cancer (5–10), lupus (11,12) and Alzheimer's (13). Calnuc is
a multifunctional protein that contains a signal peptide (SP), a putative DNA-binding
domain, a leucine zipper and two EF-hand motifs that possess the ability to bind Ca2+ (14).
It is also a multicompartmental protein, with Golgi luminal, extracellular and cytosolic
pools. This is not unique to Calnuc because other proteins with a SP, such as calreticulin,
also localize in different compartments (15). The Golgi pool is localized in the lumen of
cis-Golgi cisternae, where it constitutes the major Ca2+-binding protein (3) and a major
Ca2+ storage pool (16). After a long period of retention in the Golgi, Calnuc is secreted via
constitutive and constitutive-like pathways (17). The role of the extracellular pool is poorly
characterized but has been involved in bone matrix maturation (18). Information on the
functions of cytosolic Calnuc is just beginning to emerge. To date, a few partners that
interact with this pool of Calnuc have been identified (Gαi subunits, necdin) (3,13,19), but
the functional roles of these interactions are unknown.
255
In the present study, we carried out a yeast-two-hybrid screening to identify new
partners that interact with Calnuc to better understand the intracellular roles of Calnuc. One
of these new interacting proteins was LDLR-related protein 9 (LRP9). LRP9 is a type I
membrane protein and a member of a new subfamily of the LDLR superfamily that
includes two other members, LRP3 and LRP12 (previously named ST7) (20). Like all
LDLR superfamily proteins, this new subgroup contains LDLRA domain repeats in their
extracellular domains (21,22) that function as ligand-binding sites (23). However, several
unique structural features distinguish that the new LRP subfamily from prototypical
members of the LDLR superfamily (20), including a relatively large cytoplasmic tail that
contains a proline-rich (P-rich) domain and acidic dileucine (DXXLL) motif, does not
possess EGF-like repeats and YWTD-containing domains and has CUB domains in the
extracellular region. These structures are found in a diverse array of functionally unrelated
extracellular proteins (24) and are involved in ligand binding (25).
Little is known about LRP9 apart from the fact that it is expressed in various tissues,
may be involved in apolipoprotein E internalization (26), and is mainly localized in the
trans-Golgi network (TGN) and endosomes rather than the plasma membrane (27,28). Two
acidic cluster/dileucine (DXXLL) motifs in the cytoplasmic tail of LRP9 interact with
clathrin adaptor GGA proteins and are involved in transporting it between the TGN and
endosomes (27,28). This suggests that LRP9 may play a role in the trafficking of ligands
between these intracellular compartments much like the mannose 6-phosphate receptor,
which also contains a GGA-binding site and traffics between the TGN and endosomes (29–
31).
LRP9 is the first transmembrane protein shown to interact directly with Calnuc. In
the present study, we investigated the nature of the interaction between Calnuc and LRP9
and characterized the role of Calnuc in LRP9 intracellular trafficking.
Results
LDLR-related protein 9 (LRP9) is a novel Calnuc-interacting protein
A yeast-two-hybrid system was employed to screen for new protein(s) that interact
with Calnuc. We used the conserved N-terminus of Calnuc (residues 25–172, Figure 1A) as
bait and an adult human kidney cDNA library as the source of interacting proteins. Several
clones were isolated, sequenced and analyzed. Two had an identical 1400-bp insert and
shared 100% homology with a portion of the cytoplasmic tail of human LDL-related
protein 9 (LRP9) (Figure 1B). To confirm the interaction between Calnuc and LRP9, we
performed yeast one-on-one interaction experiments. Yeast strain AH109 was transformed
with vectors expressing various portions of Calnuc fused to the Gal4-DNA-binding domain
together with a vector expressing the Gal4-DNA activation domain fused to the C-terminal
portion of human LRP9 fished out by the yeast-two-hybrid screen (residues 556–713)
(Table 1). The clones containing the N-terminal region of Calnuc (Figure 1A) and
LRP9556−−713 grew on histidine-deficient plates and had β-galactosidase activity (Table
1). However, little or no interaction was detected when C-terminal portions of Calnuc (with
or without the EF-hands ; Figure 1A) or the PXA domain (control) were used as bait,
256
indicating that the interaction was specific to the N-terminal region of Calnuc (Table 1).
Full-length Calnuc could not be used in this assay because it self-activated the yeast
reporter genes.
Figure 1. Domains and motifs of Calnuc and the cytoplasmic tail of LRP9.
A) Schematic representation of Calnuc showing the N-terminal SP, the putative DNAbinding domain (basic region), the two EF-hands (EF−1 and EF−2) and the leucine zipper
motif. B) Comparison of the amino acid sequences of the cytoplasmic tail of LRP9 and the
clone fished out by yeast-two-hybrid (TH52p). YXXϕ and the acidic cluster dileucine
(DXXLL) motifs, two putative sorting signals, an R-rich domain and a P-rich, which are
potential protein–protein interactions sites, are indicated in the sequences. m, mouse; h,
human; TM, transmembrane.
Table 1. Calnuc and LRP9 interaction in yeast-two-hybrid.
257
LRP9 interacts with the N-terminus of Calnuc
We next verified the in vitro and in vivo interactions between Calnuc and LRP9
using pull-down and immunoprecipitation assays. 35S-labeled in vitro-translated
LRP9556−−713 bound to glutathione S-transferase (GST)-Calnuc-full-length and GSTCalnuc-N-terminus but not to GST-Calnuc-C-terminus or GST alone (Figure 2A).
Similarly, GST-Calnuc-full-length and GST-Calnuc-N-terminus were able to pull-down
full-length LRP9 from the lysate of HEK293 cells that overexpressed hemagglutinin (HA)tagged LRP9 (Figure 2B). However, no interactions were detected with GST-Calnuc-Cterminus or GST alone (Figure 2B). To confirm that the interaction between Calnuc and
LRP9 also occurs in vivo, we performed immunoprecipitation experiments on HEK293
cells transfected with green fluorescent protein (GFP)-tagged Calnuc and HA-tagged LRP9.
When we performed the immunoprecipitation with anti-HA immunoglobulin (IgG),
Calnuc-GFP coprecipitated with HA-LRP9 (Figure 2C). Similarly, when we used anti-GFP
IgG, LRP9-HA co-precipitated with Calnuc-GFP in cells cotransfected with both proteins
(Figure 2C). Calnuc-GFP in the cell lysate is visible as two bands, the lower of which likely
represents an N-terminal cleavage product (Figure 2C). It is interesting to note that the
lower band did not bind LRP9 (Figure 2C), which provides further support for the
importance of the N-terminal portion of Calnuc in the interaction with LRP9. The
conformation of Calnuc has been reported to change when it associates with calcium.
However, the amount of Calnuc that coimmunoprecipitated with LRP9 was the same
whether calcium was present or not (data not shown). These findings suggest that the Nterminal region of Calnuc (residues 25–172) interacts with the cytoplasmic tail of LRP9,
both in vitro and in vivo and that the interaction is calcium independent.
Figure 2. Calnuc interacts with LRP9.
A) In vitro interaction of Calnuc N-terminus with the cytoplasmic tail of LRP9. The in
vitro-translated 35S-labeled C-tail of LRP9 bound to GST-Calnuc-N-terminus (N-term) and
GST-Calnuc-full-length (FL) but not to GST-Calnuc-C-terminus (C-term) or GST alone.
GST proteins (10 μg each) immobilized on glutathione beads were incubated with in vitrotranslated human LRP9 (residues 556–713). Bound proteins were separated by SDS-PAGE
258
and detected by autoradiography. Input equals 7% of the total in vitro-translated product.
B) In vivo interaction of Calnuc N-terminus with HA-tagged LRP9-full-length. LRP9-HA
bound to GST-Calnuc-N-terminus (N-term) and GST-Calnuc-full-length (FL) but not to
GST-Calnuc-C-terminus (C-term) or GST alone. GST proteins (30 μg each) immobilized
on glutathione beads were incubated with lysate from HEK cells transfected with mouse
LRP9-HA. Bound proteins were separated by SDS-PAGE and detected with anti-HA
antibodies. C) Coimmunoprecipitations of LRP9-HA with Calnuc-GFP protein. Lysates
from HEK cells transfected with LRP9-HA and Calnuc-GFP were immunoprecipitated
with anti-HA (lane 2), anti-GFP (lane 4) or control (lanes 1 and 3) antibodies and then
immunoblotted with anti-HA or anti-GFP to detect LRP9 and Calnuc, respectively. CalnucGFP is seen as two bands, the lower of which probably represents an N-terminal cleavage
product. Note that only the 95 kDa band is coimmunoprecipitated. Quantification of the
band intensity revealed that approximately 2% of LRP9-HA immunoprecipitated with
Calnuc-GFP and <1% of the total Calnuc-GFP were recovered in the LRP9-HA
immunoprecipitate. Nevertheless, coimmunoprecipitations in both directions were highly
specific.
Figure 3. Calnuc interacts with LRP3 but not with LRP1.
Lysates from HEK cells transfected with GFP or Calnuc-GFP together with (A) LRP3FLAG or (B) LRP1-Myc were immunoprecipitated with anti-GFP antibodies and then
immunoblotted with anti-FLAG, anti-Myc, or anti-GFP to detect LRP3, LRP1 and Calnuc,
respectively.
259
To verify the specificity of the Calnuc–LRP9 interaction, we next looked at whether
Calnuc bound to LRP1 and LRP3, two other members of the LDLR family. LRP3 is a
member of the same LDLR subfamily as LRP9, whereas LRP1 is a member of a different
subfamily (20). Immunoprecipitations with anti-GFP IgG were performed on HEK293 cells
expressing GFP or GFP-Calnuc together with FLAG-tagged LRP3 or Myc-tagged LRP1.
As shown in Figure 3, LRP3 bound to Calnuc, whereas LRP1 did not display any
appreciable binding to Calnuc. These results suggest that Calnuc interacts with LRP9 and
LRP3, a related family member, perhaps via a common binding domain.
LRP9 interacts with endogenous Calnuc
We next looked at whether endogenous Calnuc interacts with LRP9 in HEK293
cells transfected with HA-tagged LRP9. When we performed the immunoprecipitation with
anti-Calnuc, a fraction of LRP9-HA coimmunoprecipitated with Calnuc, confirming the
occurrence and specificity of the LRP9 interaction with endogenous Calnuc in intact cells
(Figure 4A). Because endogenous Calnuc is found in both cytosolic and membrane
fractions (3), we next looked at whether the interaction with LRP9 occurs specifically with
the cytosolic pool of endogenous Calnuc. Using subcellular fractionation, we prepared a
cytosolic fraction from HeLa cells. As observed previously (3), endogenous Calnuc was
distributed in both membrane (P100) and cytosolic (S100) fractions (Figure 4B, lanes 1–2).
However, the transmembrane receptor mannose−6-phosphate receptor (MPR) was detected
mainly in the P100 fraction, whereas the cytosolic protein EEA1 was only detected in the
S100 fraction (Figure 4B, lanes 1–2). The ability of Calnuc in the S100 fraction to interact
with the C-tail of LRP9 was next assessed using pull-down binding assays. GST-LRP9-Ctail (GST-LRP9-CT1 in Figure 5A) was able to pull-down endogenous cytosolic Calnuc
(Figure 4B, lane 4). However, only little interaction was detected with GST alone (Figure
4B, lane 3). These results indicated that endogenous cytosolic Calnuc interacts with the
LRP9 C-tail.
Calnuc interacts with an arginine-rich (R-rich) region in the cytoplasmic tail of LRP9
The cytoplasmic tail of LRP9 contains many putative protein–protein interaction
sites (Figure 5A). To identify the specific region within LRP9 required for the interaction
with Calnuc, several deletion mutants of the C-terminal tail of LRP9 were generated
(Figure 5A). The ability of these mutants to interact with Calnuc was assessed using pulldown binding assays. GST-LRP9-CT1, CT3 and CT4 pulled down Calnuc from the lysate
of HEK293 cells that expressed Calnuc-GFP (Figure 5B). In contrast, no interaction was
detected with GST-LRP9-CT2, CT5, CT6, or GST alone (Figure 5B). The complete Cterminus of LRP9 (GST-CT) could not be used because the fusion protein is degraded
when expressed in bacteria. The only domain present in all the deletion mutants that bound
to Calnuc was the R-rich domain of the cytoplasmic tail of LRP9, indicating that it is major
determinant for the interaction between LRP9 and Calnuc. Interestingly, this R-rich region
is also present in LRP3 but not in LRP1. To confirm the importance of this region for the
interaction with Calnuc in vivo, a LRP9 mutant lacking the R-rich domain (ΔR-LRP9) was
generated and tested for its ability to coimmunoprecipitate with Calnuc. Surprisingly, we
observed that the ΔR-LRP9 mutant had not lost its capacity to interact with Calnuc (data
260
not shown). This suggests that the R-rich domain of LRP9 might be important but not
crucial for the interaction with Calnuc in vivo.
Figure 4. Endogenous Calnuc binds to the cytoplasmic tail of LRP9.
A) Coimmunoprecipitation of LRP9-HA with endogenous Calnuc. Lysates from HEK cells
transfected with LRP9-HA or a control vector (pCMV-HA) were immunoprecipitated with
anti-Calnuc antibodies and then immunoblotted with anti-HA (LRP9) or anti-Calnuc
antibody. Quantification of the band intensity revealed that <1% of the total LRP9-HA
coprecipitated with endogenous Calnuc. B) Interaction of cytoplasmic endogenous Calnuc
with LRP9-C-tail. Endogenous Calnuc is found in approximately equal amounts in both
membrane (P100, lane 1) and cytosolic (S100, lane 2) fractions obtained by subcellular
fractionation of HeLa cells. Calnuc from the S100 fraction bound to GST-LRP9-C-tail
(GST-CT1 described in Figure 5) (lane 4) but bound only poorly to GST alone (lane 3).
GST proteins (10 μg each) immobilized on glutathione beads were incubated with the S100
fraction from HeLa cells. Input (lane 2) equals 4% of the total S100 used for pull-down.
Bound proteins were separated by SDS-PAGE and detected with anti-Calnuc antibody.
Calnuc and LRP9 colocalize on Golgi cisternae
To determine whether LRP9 colocalizes with Calnuc, we compared the intracellular
distribution of HA-tagged LRP9 with endogenous Calnuc and GFP-tagged Calnuc in COS7
cells using confocal microscopy. LRP9-HA was mainly detected in the juxtanuclear region
and surrounding vesicles (Figure 6A,D,G). This is in agreement with our previous findings
showing that LRP9 localizes mainly in the TGN and, to a lesser extent, in endosomes (27).
261
Endogenous Calnuc was concentrated in the Golgi region (Figure 6B). Indeed, previous
immunofluorescence (IF) and immunoelectronmicroscopy studies precisely localized
Calnuc in the cis-Golgi cisternae (3). This suggests that endogenous Calnuc is mainly
concentrated and detected in the lumen of the cis-Golgi, whereas the cytoplasmic pool of
endogenous Calnuc is undetectable by IF. There was only partial colocalization between
LRP9-HA and endogenous Calnuc because both proteins were located in different stacks of
the Golgi apparatus (Figure 6C). In cells overexpressing Calnuc-GFP (Figure 6D–F), two
intracellular pools of Calnuc were detected by IF, as previously reported (3,17,32). A major
pool was localized in the lumen of the cis-Golgi cisternae and a minor pool in the
cytoplasm (Figure 6E). After double labeling, LRP9-HA partially overlapped with CalnucGFP in the Golgi cisternae (Figure 6F) as observed with endogenous Calnuc (Figure 6C).
Given that Calnuc interacted with the cytoplasmic tail of LRP9, we next asked whether the
specific cytosolic pool of Calnuc and LRP9 colocalized. To address this question, we
conducted confocal analyses of COS7 cells transfected with LRP9-HA and Calnuc-GFP
without its SP (ΔSP-Calnuc-GFP). This deletion resulted in the expected defects in
targeting Calnuc in the lumen of the Golgi and its accumulation in the cytoplasm (32).
ΔSP-Calnuc-GFP was detected in the cytoplasm and on the plasma membrane (Figure 6H)
and clearly colocalized with LRP9-HA on the surface of the Golgi cisternae and
surrounding vesicles (Figure 6I). Together, these IF and biochemical interaction assays
suggested that the cytoplasmic pool of Calnuc interacts with the C-terminal tail of LRP9 on
the cytoplasmic side of the TGN cisternae.
Figure 5. Calnuc interacts with the R-rich region of LRP9.
A) Schematic representation of the various GST-LRP9 proteins used for the pull-down
assays. R-rich, arginine-rich domain; P-rich, proline-rich domain; PDZb, PDZ-binding
domain motif; DXXLL, acidic dileucine motif. B) The deletion mutants of LRP9 shown in
A were bound to glutathione beads and incubated with lysate from HEK cells transfected
with Calnuc-GFP. Bound proteins were separated by SDS-PAGE and detected with antiGFP antibody. Calnuc-GFP bound to the deletion mutants spanning residues 92–115
containing the R-rich region (CT1, CT3, CT4) but not to the mutants lacking this region
(CT2, CT5, CT6) or to GST alone. The additional Calnuc-GFP bands detected in the cell
lysate by the polyclonal anti-GFP antibody may probably correspond to an N-terminal
cleavage product. Note that only the 95 kDa band was pulled down.
262
Figure 6. Calnuc and LRP9 colocalize on the Golgi apparatus.
In COS7 cells transfected with (A–C) LRP9-HA alone or with (D–I) wild-type CalnucGFP, LRP9-HA was found in the juxtanuclear region and surrounding vesicles (inset,
A,D,G). A–C) Endogenous Calnuc was found mainly in the Golgi region (inset, B) where it
partially overlapped with LRP9-HA (inset, C). D–F) Calnuc-GFP was found mainly inside
the Golgi cisternae (inset, E) and, to a lesser extent, in the cytoplasm (E). The merged
image (yellow) shows a partial overlap between LRP9-HA and Calnuc-GFP in the Golgi
cisternae (inset, F). G–I) COS7 cells transfected with LRP9-HA and Calnuc devoid of its
SP (ΔSP-Calnuc-GFP). Deleting the signal sequence of Calnuc (residues 2–25) resulted in
the expected defect in targeting in the Golgi lumen and an accumulation in the cytoplasm.
ΔSP-Calnuc-GFP was also detected on the plasma membrane (arrows, H) and the cytosolic
side of the Golgi cisternae (arrowhead and inset, H) where it colocalized with LRP9-HA
(yellow, inset I). Twelve hours after the transfection, the cells were fixed, permeabilized
and immunostained using (A, D, G) anti-HA, (B) anti-Calnuc and (E, H) anti-GFP
antibodies. The labeled cells were examined by confocal fluorescence microscopy. Scale
bar, 10 μm.
263
Calnuc colocalizes with LRP9 on early endosomes
A minor pool of LRP9 has been previously reported to be present in early
endosomes (27,28). To determine whether Calnuc is present on the surface of endosomes
and whether it colocalizes with LRP9 on this organelle, we performed confocal IF on COS7
cells expressing ΔSP-Calnuc-GFP alone or with HA-LRP9. We started by comparing the
subcellular localization of ΔSP-Calnuc-GFP with the early endosome marker EEA1 (Figure
7A–C). ΔSP-Calnuc-GFP was detected on the plasma membrane and in the Golgi region as
well as on some fine, punctate structures distributed throughout the cytoplasm (Figure 7A)
that partially overlapped with EEA1 (Figure 7C). This colocalization was consistent with
the presence of Calnuc on early endosomes.
Figure 7. Calnuc and LRP9 colocalize on early endosomes.
A–C) In COS7 cells transfected with ΔSP-Calnuc-GFP (cytoplasmic form of Calnuc)
alone, ΔSP-Calnuc-GFP was distributed on the plasma membrane (arrow, A), the cytosolic
side of the Golgi (arrowhead, A), and small vesicular structures in the cytoplasm (inset, A).
EEA1 was found on early endosomes distributed throughout the cell (B). Merged image
showing occasional overlap in the vesicular distribution of ΔSP-Calnuc-GFP and EEA1
(inset C). D–F) COS7 cells were cotransfected with ΔSP-Calnuc-GFP, HA-tagged LRP9
and Rab5Q79L, a Rab5 GTPase-deficient mutant that creates enlarged early endosomes.
264
LRP9-HA was present in enlarged endosomes (E). ΔSP-Calnuc-GFP was present in the
cytoplasm (D), on the plasma membrane (arrow, D) and on the enlarged endosomes (inset,
D) labeled with LRP9-HA (inset, F). G–I) COS7 cells were transfected with ΔSP-CalnucGFP and LRP9-HA in which the two DXXLL motifs were mutated (LRP9−2DXXAA).
The mutation induced a redistribution of LRP9 to the plasma membrane (arrow, H) and
early endosomes (inset, H). ΔSP-Calnuc-GFP was present in the cytoplasm, on the
cytosolic side of the Golgi (arrowhead, G), and together with the HA-LRP9−2DXXAA
mutant on the plasma membrane (arrow, I) and endosomes (inset, I). Twelve hours after the
transfection, the cells were fixed, permeabilized and immunostained using (E, H) anti-HA,
(B) anti-EEA1 and (A,D,G) anti-GFP antibodies. The stained cells were examined by
confocal fluorescence microscopy. The yellow color in the merged images (C, F, I)
indicates colocalization. Scale bar, 10 μm.
To determine whether Calnuc colocalized with LRP9 on endosomes, we enriched
LRP9 in early endosomes and compared its localization with that of Calnuc. We first
trapped LRP9-HA in enlarged endosomes (Figure 7E) by transfecting COS7 cells with an
established Rab5 GTPase-deficient mutant (Rab5Q79L) that greatly enlarges early
endosomes because of increased homo- and heterotypic fusion (33). LRP9-labeled enlarged
vesicles have been previously shown to colocalize with EEA1 (27). ΔSP-Calnuc-GFP
colocalized with LRP9-HA on subregions of the enlarged endosomes (Figure 7F),
suggesting that they are present together on early endosomes. To confirm this finding, we
next transfected COS7 cells with HA-tagged LRP9−2DXXAA, an LRP9 mutant in which
the leucines of both DXXLL motifs (Figure 1B) are substituted by alanines. This mutant,
which is incapable of binding GGA proteins, mislocalized on the plasma membrane and
early endosomes (Figure 7H) (27,28). Figure 7(G–I) shows that ΔSP-Calnuc-GFP
colocalized with LRP9−2DXXAA on the plasma membrane and peripheral early
endosomes. From these experiments, we concluded that a fraction of Calnuc is present on
endosomes where it partially colocalizes with LRP9, suggesting that the cytoplasmic pool
of Calnuc can also bind LRP9 on early endosomes.
265
Figure 8. Knocking down Calnuc affects the intracellular distribution of LRP9.
LRP9 localized mainly in the TGN region in control cells (B), but knocking down Calnuc
induced a redistribution of LRP9 to peripheral vesicles (D). HeLa cells were transfected
with (A, B) control or (C, D) Calnuc siRNA for 3 days. LRP9-HA cDNA was transfected
for 12 h before the IF experiment. The cells were fixed, permeabilized and immunostained
using (A, C) anti-Calnuc and (B, D) anti-HA antibodies. Scale bar, 10 μm.
Calnuc knockdowns redistribute LRP9 to late endosomes/lysosomes
To investigate the functional role of Calnuc in LRP9 trafficking, we studied the
subcellular distribution of LRP9 in Calnuc-depleted cells. HeLa cells were transfected with
control or Calnuc small interfering RNA (siRNA) together with HA-tagged LRP9 and were
analyzed by confocal microscopy. Calnuc was significantly (≥ 90%) depleted 72 h after
transfection with specific siRNA, as observed in the IF (Figure 8) and immunoblotting
experiments (Figure 10). In Calnuc knockdown cells, LRP9 was mainly localized in
peripheral punctate structures (Figure 8D) rather than in the TGN (Figure 8B). This did not
appear to be because of gross changes in TGN morphology, because the localization of
TGN46, a TGN marker, was not altered (Figure S1).
266
Figure 9. LRP9 redistributes in late endosomes/lysosomes in Calnuc-depleted cells.
267
A) Comparison of LRP9 and EEA1 distribution in Calnuc-depleted cells. LRP9-HA was
localized in the juxtanuclear region and surrounding vesicles in HeLa cells transfected with
control siRNA (a). Some of the LRP9-containing vesicles colocalized with EEA1 in the
Golgi area (inset, c) but not at the periphery of the cells. In cells transfected with Calnuc
siRNA (d–f), LRP9-HA redistributed into peripheral vesicles (d) but did not colocalize
mainly with EEA1 (inset, f). Arrowheads indicate the Golgi region. B) Comparison of
LRP9 and LAMP−2 distribution in Calnuc-depleted cells. LRP9-HA was found in the
Golgi region and surrounding vesicles in HeLa cells transfected with control siRNA (a) and
was relatively rare in the LAMP−2-labeled compartment (b–c). In Calnuc siRNAtransfected cells (d–f), LRP9-HA was mainly found in peripheral punctate structures (d)
and significantly colocalized with LAMP−2 (inset, f). The cells were fixed, permeabilized
and immunostained using anti-HA, anti-EEA1 and anti-LAMP−2 antibodies. The stained
cells were examined by confocal fluorescence microscopy. The yellow color in the merged
images indicates colocalization. Scale bar, 10 μm. C) Quantification of the degree of
overlap between LRP9-HA and EEA1 or LAMP−2 in cells treated with control or Calnuc
siRNA as described in (A) and (B). The histogram depicts relative colocalization R values
calculated as described in Materials and Methods. Values are significant at p < 0.001 (**)
for control versus Calnuc knockdown cells. Data represent means ± SD (n≥10) for each
condition.
Figure 10. Knocking down Calnuc affects the levels of LRP3 and LRP9.
The steady-state levels of LRP3 and LRP9 but not LRP1 were reduced in Calnuc-depleted
cells. HeLa cells were transfected with control or Calnuc siRNA for 3 days. The cells were
also transfected with LRP1-Myc, LRP3-FLAG or LRP9-HA cDNA for 12 h before the
experiment. Cells were lysed and proteins were separated by SDS-PAGE and detected with
specific anti-Myc, anti-FLAG, anti-HA, anti-Calnuc and anti-actin antibodies. Actin served
as a loading control. Calnuc was below the detection level in samples treated with Calnucspecific siRNA.
268
To better define the specific vesicular compartment in which LRP9 redistributes in
the absence of Calnuc, the cells were transfected with control or Calnuc siRNAs and the
localization of LRP9-HA was compared to endosomal markers by confocal microscopy
(Figure 9). Colocalization was evaluated quantitatively by measuring Pearson correlation
coefficient (R) values, which are standard measures of colocalization (34). Because we
previously observed that LRP9 can redistribute to early endosomes (27), we first compared
the localization of LRP9 to that of the early endosome marker EEA1 (Figure 9A). In
control cells, as previously reported (27), LRP9 partially colocalized with EEA1 on
vesicles in the Golgi area (R = 0.29 ± 0.07) [Figure 9A(a–c),C]. In Calnuc-depleted cells,
the colocalization of EEA1 and LRP9-HA did not increase (R = 0.26 ± 0.05) [Figure 9A(d–
f),C]. We next compared the colocalization of LRP9-HA and LAMP−2, a late
endosome/lysosome marker. Figure 9B(a–c) shows that LRP9 partially colocalized with
LAMP−2-labeled structures in control cells (R = 0.33 ± 0.06). However, the colocalization
of LRP9 with LAMP−2 on vesicles significantly increased in Calnuc knockdown cells (R =
0.53 ± 0.09) [Figure 9B(d–f),C]. In summary, the depletion of Calnuc redistributed LRP9
from the TGN to peripheral LAMP−2 labeled vesicles. Knocking down Calnuc thus caused
missorting of LRP9 to late endosomes/lysosomes. These observations suggested that
Calnuc participates in the intracellular trafficking or sorting of LRP9.
Calnuc knockdown increases the lysosomal turnover of LRP9
Because Calnuc depletion shifted LRP9 towards late endosomes/lysosomes, the
level of LRP9 present in cell lysates of control- and siRNA-treated cells was next examined
by immunoblotting (Figure 10). Following a 72-h treatment with Calnuc siRNA, the levels
of LRP9 were significantly reduced compared with control cells (Figure 10). Because
Calnuc interacted with LRP3 but not with LRP1 (Figure 3), we also examined the effect of
knocking down Calnuc on the level of these two LRPs. Interestingly, the depletion of
Calnuc decreased the amount of LRP3 but had no effect on LRP1 (Figure 10). These results
suggest that the interaction with Calnuc is important for the proper sorting of LRP3 and
LRP9.
We reasoned that the reduction in LRP9 levels might be because of the degradation
of the receptor in lysosomes. To test this hypothesis, we performed cycloheximide chase
experiments in which the levels of LRP9 in control- and siRNA-treated cells were
examined at different times after inhibiting protein synthesis (Figure 11A). In mock-treated
cells, LRP9 had a half-life of 4 h, whereas in Calnuc siRNA-treated cells, the half-life of
LRP9 was reduced to <2 h (Figure 11A). To determine whether this degradation occurred
in lysosomes, we incubated the siRNA-treated cells with the lysosomal inhibitors leupeptin
and E64. In the absence of lysosomal inhibitors, 48% of the LRP9 was degraded in Calnuc
knockdown cells, whereas, in the presence of lysosomal inhibitors, only 13% of the LRP9
was degraded (Figure 11B). Treatments with lysosomal inhibitors thus significantly
prevented a decrease in LRP9 levels (Figure 11B). Taken together, these observations
suggest that depleting Calnuc increases the delivery of LRP9 to lysosomes.
269
Figure 11. LRP9 is degraded in lysosomes when Calnuc is depleted.
A) The half-life of LRP9 is shorter in Calnuc-depleted cells than in control cells. HeLa
cells were transfected with control or Calnuc siRNA for 3 days, and with LRP9-HA cDNA
for 12 h before the experiment. Cells were incubated with 40 μg/mL of cycloheximide, an
inhibitor of protein synthesis, for different periods of time. Cells were lysed and analyzed
by immunoblotting. B) LRP9 was degraded in lysosomes in Calnuc-depleted cells. Control
and Calnuc-suppressed cells transfected with LRP9-HA [as described in (A)] were treated
or not with the lysosomal inhibitors leupeptin (1 mg/mL) and E64 (10 μg/mL) for 3 h. The
cells were lysed and the proteins were separated by SDS-PAGE and detected with specific
anti-Calnuc, anti-HA (LRP9), anti actin or anti-tubulin antibodies. Tubulin and actin were
used as loading controls. (Bottom) Bar graph showing the quantification of LRP9
degradation in cells treated as described in (B) and expressed as a percentage of LRP9
present in control cells. Results are shown as means ±sd (n = 3). *p < 0.05 (compared with
control cells).
Calnuc siRNA phenotype is rescued by Calnuc and ΔSP-Calnuc overexpression
To confirm that the consequence of Calnuc depletion on LRP9 sorting is because of
a lack of Calnuc binding to the LRP9 cytoplasmic domain, we performed rescue
experiments with siRNA-resistant forms of wild-type Calnuc and cytosolic Calnuc (ΔSP270
Calnuc). Rat Calnuc (rCalnuc) was used as a rescue construct because it differs in many
nucleotides from human siRNA target sequences, rendering it siRNA resistant. We first
compared the distribution of LRP9-HA in Calnuc siRNA-treated cells overexpressing GFP,
GFP-tagged wild-type rCalnuc, or GFP-tagged ΔSP-rCalnuc (Figure 12A,B). In control
(GFP) cells, LRP9 was mainly localized in peripheral punctate structures that strongly
colocalized with LAMP−2 (R = 0.53 ± 0.08) [Figure 12A(a–d),B]. However, in cells
rescued with rCalnuc-GFP or ΔSP-rCalnuc-GFP, LRP9-HA was mainly concentrated in the
TGN region and very little LRP9 was observed in LAMP−2-labeled vesicles [Figure
12A(e–h,i–l)]. The colocalization values (R) of LRP9 with LAMP−2 decreased
significantly to 0.28 ± 0.02 and 0.30 ± 0.05 in rCalnuc-GFP- and ΔSP-rCalnuc-GFPrescued cells, respectively (Figure 12B). These values were similar to those observed in
control siRNA-treated cells (R = 0.33 ± 0.06, Figure 9C). As expected, partial
colocalization between LRP9-HA and Calnuc-GFP or ΔSP-Calnuc-GFP was observed in
the Golgi region of these rescued cells (Figure S2)
Figure 12. Expression of cytoplasmic Calnuc restores the normal distribution of
LRP9.
271
A) Comparison of LRP9 and LAMP−2 distribution in Calnuc-depleted cells rescued with
GFP (a–d), siRNA-resistant versions of full-length Calnuc-GFP (e–h) or cytoplasmic
Calnuc-GFP (ΔSP-Calnuc-GFP) (i–l). LRP9-HA was mainly found in peripheral vesicles
(b) that colocalized with LAMP−2 in cells transfected with GFP (inset, d). However,
LRP9-HA was found in the Golgi region and surrounding vesicles (f and j) that were
occasionnaly labeled with anti-LAMP−2 in cells rescued with Calnuc-GFP (inset, h) or
ΔSP-Calnuc-GFP (inset, l). The cells were fixed, permeabilized and immunostained using
anti-HA and anti-LAMP−2 antibodies. The expression of GFP proteins was monitored
using GFP fluorescence and was pseudocolored blue. The stained cells were examined by
confocal fluorescence microscopy. The yellow color in the merged images indicates
colocalization between LRP9-HA and LAMP−2. Scale bar, 10 μm. B) Quantification of the
degree of overlap between LRP9-HA and LAMP−2 in cells treated as described above. The
histogram depicts relative colocalization R values calculated as described in Materials and
Methods. Values are significant at p < 0.001 (**) for control (GFP) versus Calnuc rescue
cells. Data represent means ± SD (n≥10) for each condition.
We next looked at whether the degradation of LRP9 was reversed when Calnuc was
restored in Calnuc siRNA-treated cells. In cells rescued with rCalnuc-GFP or ΔSP-rCalnucGFP, the levels of LRP9 increased by 44.8 and 48.9%, respectively, compared with cells
rescued with control GFP (Figure 13A,B). In summary, the redistribution and degradation
of LRP9 in lysosomes observed in Calnuc-depleted cells could be significantly reversed by
expressing siRNA-resistant wild-type Calnuc or cytosolic Calnuc. These results suggested
that the cytoplasmic pool of Calnuc participates in the intracellular trafficking/sorting of
LRP9 and provided support for a direct role for the interaction of Calnuc with the
cytoplasmic tail of LRP9.
Figure 13. Expression of cytoplasmic Calnuc rescues the lysosomal degradation of
LRP9.
272
Steady-state levels of LRP9 are increased in Calnuc-depleted cells rescued with siRNAresistant full-length Calnuc-GFP or cytoplasmic Calnuc-GFP (ΔSP-Calnuc-GFP) unlike
cells rescued with control GFP. HeLa cells treated with Calnuc siRNA were transfected
with GFP, full-length rCalnuc-GFP or ΔSP-rCalnuc-GFP for 3 days, and then with LRP9HA cDNA for 12 h before the experiment. Cells were lysed, and proteins were separated by
SDS-PAGE and detected with specific anti-HA, anti-GFP, anti-Calnuc and anti-actin
antibodies. Actin served as a loading control. Endogenous human Calnuc was below the
detection level in samples treated with Calnuc-specific siRNA. B) Bar graph showing the
quantification of LRP9 degradation in cells treated as described in A) and expressed as a
percentage of LRP9 present in control (GFP) cells. Results are shown as means ± SD (n =
3). *p< 0.05 [compared with control (GFP) cells].
Discussion
In the present study, we used a yeast-two-hybrid assay to search for new interacting
partners for Calnuc and to better understand its intracellular functions. We identified
LDLR-related protein 9 (LRP9) as the first transmembrane protein that directly interacts
with Calnuc. We also demonstrated that Calnuc plays a role in the retrieving of LRP9 from
endosomes to the TGN.
Our results demonstrated that the interaction between Calnuc and LRP9 involved
the N-terminus domain of Calnuc and an R-rich region in the cytoplasmic domain of LRP9.
In vivo and in vitro pull-down and yeast-two-hybrid assays indicated that the conserved Nterminal region of Calnuc was essential for the interaction with the cytoplasmic tail of
LRP9. Furthermore, an analysis of LRP9 C-terminal tail truncation mutants identified an Rrich domain as the Calnuc-binding determinant. Our yeast-two-hybrid results are in
agreement with this because the R-rich region was present in both clones fished out by the
yeast-two-hybrid approach. Interestingly, we noted that an R-rich region was present in the
cytoplasmic tail of LRP3, a member of the same LRP subfamily as LRP9 (20) that
interacted with Calnuc, but was absent from LRP1, a member of another subfamily of LRP
that did not interact with Calnuc. This is consistent with the notion that the region bearing
the R-rich domain is responsible for the interaction with Calnuc. However, an LRP9 mutant
lacking the R-rich region (ΔR-LRP9) still coimmunoprecipitated Calnuc in vivo,
suggesting that the R-rich domain of LRP9 is important but not crucial for the interaction
with Calnuc in vivo. Calnuc might also interact indirectly, via other partners, with LRP9HA, or with endogenous LRP9 or LRP3, which might form oligomers with ΔR-LRP9.
Indeed, we have data suggesting that LRP9 and LRP3 form homo- and hetero-oligomers
(C. Lavoie, Université de Sherbrooke, Sherbrooke, unpublished results). R-rich domains,
also called basic amphiphilic segments, have been previously identified in the cytoplasmic
tail of other receptors such as EGFR. This region has been reported to be the binding site
for calmodulin (CaM), another EF-hand calcium-binding protein (35). CaM regulates
EGFR tyrosine kinase activity, trafficking and signaling (36–39). The CaM-binding site is
an R-helical structure composed of a cluster of positively charged amino acids arranged on
one side of the R-helix, whereas most of the hydrophobic amino acids are on the opposite
273
side (35). A helical-wheel projection predicted that the R-rich regions of LRP9 and LRP3
fold in the same kind of helical structure (C. Lavoie, unpublished results). These
observations suggest that Calnuc might bind and modulate the trafficking and signaling of
other receptors (e.g., EGFR or other members of the LDLR superfamily). This is presently
under investigation.
The interaction between Calnuc and LRP9 is likely physiological because our
confocal microscopic studies showed that the cytosolic form of Calnuc colocalized with
LRP9 predominantly on the TGN and, to a lesser extent, on early endosomes. Indeed, in
cells expressing the Rab5QL or LRP9−2DXXAA mutants, which both induced the
redistribution of LRP9 on early endosomes, Calnuc clearly colocalized with LRP9 on this
compartment. Because Calnuc colocalized with LRP9 on the TGN and early endosomes,
we propose that it is at this location that Calnuc participates in the trafficking of LRP9.
The involvement of Calnuc in the retrieval of LRP9 from endosomes to the TGN is
supported by the phenotype elicited by siRNA-mediated interference of Calnuc expression.
This treatment resulted in the redistribution of LRP9 into late endosomes/lysosomes,
enhanced lysosomal degradation of LRP9 and, consequently, decreased steady-state levels
of this receptor. Importantly, treatment with lysosomal protease inhibitors restored LRP9
levels in Calnuc-depleted cells, unequivocally demonstrating that the reduction in LRP9
levels was because of mistargeting and degradation in lysosomes. These data point to a role
for Calnuc in the recycling of LRP9 from endosomes. These effects were specific because
the silencing of Calnuc had no effect on the distribution and levels of TGN46, another TGN
protein, indicating no obvious perturbation in Golgi organization and increased the turnover
of LRP3 but not of LRP1, two members of different LRP subfamilies that do and do not
interact with Calnuc, respectively. These results strongly indicate that Calnuc is directly
involved in retrograde transport from endosomes of specific receptors rather than a
generalized effect on membrane protein dynamics. The specificity of the phenotype
observed with Calnuc depletion was also corroborated by rescue experiments using an
siRNA-resistant version of Calnuc. Indeed, the expression of siRNA-resistant wild-type
and cytoplasmic Calnuc restored the distribution of LRP9 in the TGN and decreased its
redistribution and degradation in lysosomes in Calnuc-depleted cells. Therefore, the
cytoplasmic pool of Calnuc emerged as the functional pool involved in the endosomal
sorting of LRP9. This provided further support for a direct role in the interaction of Calnuc
with LRP9 cytoplasmic tail. The most logical interpretation of these observations is that
Calnuc keeps LRP9 from going to lysosomes by mediating its removal from maturing
endosomes. The cytoplasmic LRP9 determinant recognized by Calnuc might encompass a
‘lysosomal avoidance’ signal functionally similar to that in the cytosolic domain of the
MPR (40). However, the Trp/Phe-Leu-Met/Val or Phe-Trp motifs defined as endosome-toTGN retrieval motifs for MPR and sortilin (40,41) are not present in LRP9. This indicates
that other determinants within LRP9 direct its sorting from endosomes. Our results suggest
that Calnuc recognizes determinants in the R-rich region of the cytoplasmic domain of
LRP9 and facilitates LRP9 sorting into transport vesicles destined for the TGN.
Calnuc, GGA and adaptor protein (AP1/AP2) bind to distinct domains in the
cytoplasmic tail of LRP9 and are involved in different trafficking steps. Deletion of
274
dileucine signals that interact with both GGAs and AP1/AP2 cause an accumulation of
LRP9 on early endosomes and the plasma membrane (27,28), whereas siRNA depletion of
Calnuc increases the lysosomal delivery of LRP9. Interestingly, the effects of Calnuc
depletion are similar to the retromer subunits depletion on MPR trafficking (42). Retromers
are molecules that retrieve receptors such as MPR, sortilin and SorLA from maturing
endosomal intermediates or late endosomes to the TGN and whose depletion results in a
dramatic reduction of the half-life of these receptors (42–44). This suggests that Calnuc
function is similar to retromers in the efficient endosomal sorting and retrograde transport
of LRP9. These findings provide evidence that GGA and Calnuc may act in concert to
promote the transport of LRP9 from the TGN and their return from endosomes.
LRP9 shares trafficking similarities with the sortilin, SorLA and MPR receptors,
which transport cargo from the TGN to the endosomal/lysosomal system (29,43–46). These
receptors predominate in the TGN and endosomes, whereas only a minor fraction (5–10%)
is present on the cell surface at any given time (28,44,47). They contain GGA and
AP1/AP2-binding sites, which are crucial for endocytosis and Golgi-endosome sorting (27–
29,43,48–51) and are delivered from early to late endosomes prior to recycling to TGN for
another round of transport. Their passage through late endosomal intermediates is
important for the dissociation of cargo from the receptors that occurs at low pH (52,53).
Proteins such as retromers (54,55), AP1 (56), TIP47 (57,58) and PACS−1 (59) participate
in this retrieval step. We have identified Calnuc as a key player in the endosomal sorting of
LRP9 for proper retrieval to the TGN. The physiological role(s) of LRP9 are far from clear
but, given the finding described above, it likely involved in trafficking of ligands between
the TGN and endosomes. Investigations are presently underway to identify potential
intracellular cargos for LRP9.
LRP9 thus most likely cycles between the TGN, endosomes and lysosomes through
a GGA- and Calnuc-dependent trafficking mechanism. Further work is currently in
progress to define the precise mechanism by which Calnuc regulates LRP9 trafficking and
to determine whether Calnuc regulates the transport of other receptors. These studies will
help clarify the biological functions of Calnuc and LRP9.
Materials and Methods
Antibodies and reagents
Anti-HA monoclonal antibodies (mAbs) and anti-LAMP−2 mAbs were purchased
from Covance and the Developmental Studies Hybridoma Bank, respectively. Anti-GFP
polyclonal antibodies (pAbs) were purchased from Molecular Probes, anti-HA pAbs from
Covance, anti-EEA1 pAbs from Affinity Bioreagents, anti-TGN46 pAbs from Novus
Biologicals, and anti-Calnuc pAbs from Aviva System Biology. Rabbit antibodies against
CI-MPR were generously provided by Dr Thomas Braulke (University of Hamburg,
Hambourg, Germany).
DNA constructs
Mammalian expression vector pMKITNeo encoding the murine LRP9-HA was
kindly provided by Dr T. Kitamura (University of Tokyo, Japan). The LRP9 double
275
DXXLL mutant construct has been described previously (27). Mammalian expression
vector pcDNA3.1 containing rCalnuc-GFP fusion protein or rat ΔSP-Calnuc (Calnuc
without SP) is described elsewhere (32).
Yeast-two-hybrid screen and mating
The MATCHMAKER GAL4 Two-Hybrid System 3 (Clontech) was used. The Nterminus of Calnuc cDNA (residues 25–168) was cloned into the pGBKT7 vector to
generate the bait construct expressing the Calnuc-GAL4 DNA-binding domain fusion
protein. Saccharomyces cerevisiae strain AH109, which contains the HIS3, ADE2 and
MEL1 reporter genes, was transformed with the bait plasmid using the lithium acetate
method described in the MATCHMAKER instruction manual (Clontech). Transformants
containing the bait plasmid were selected by plating the cells on minimal SD agar lacking
tryptophan (SD/-trp). A MATCHMAKER human kidney cDNA library (Clontech)
pretransformed in yeast strain Y187 was used for mating. To detect Calnuc-interacting
proteins, AH109-Calnuc-N-term cells were incubated overnight with a Y187-kidney
library. The mating mixture was plated on medium stringency SD/-leu/-trp/-his/5 mm 3amino triazole agar plates. After incubating the plates at 30^C for several days, the colonies
were streaked on SD/-leu/-trp and SD/-leu/-trp/-his agar plates. These yeast colonies were
subjected to a higher stringency test (i.e., they were replica plated onto SD/-leu/-trp/-his/ade agar plates). The colonies that grew well were restreaked to assess β-galactosidase (βGal) activity using a colony lift assay (CLONTECH Laboratories). Plasmid DNA was
isolated from the colonies using Zymoprep yeast plasmid miniprep kits (Zymo Research).
Prey plasmids were purified by transforming E. coli KC8 by electroporation with the
plasmid DNA isolated from yeast. The prey plasmids were isolated using QIAprep spin
plasmid DNA miniprep kits (Qiagen) and were sequenced. cDNA fragments were
identified by BLAST analysis against GenBank. One of the clones (TH52) contained a
portion of the C-terminus of LRP9 (residues 556–713).
To confirm the interaction of Calnuc with LRP9 C-tail (TH52 clone), bait plasmids
encoding truncated forms of Calnuc (N-term, C-term or EF-hand+C-term) were engineered,
and AH109 was transformed with the bait plasmids together with the LRP9 C-tail pACT2
prey plasmids. AH109 transformed with empty prey and bait plasmid (pACT2 and
pGBKT7, respectively) were used as negative controls. To assess the specificity of the
interaction between the prey proteins and Calnuc, the interactions of an unrelated bait
protein (the PXA domain) with the preys was also tested. All the transformants were plated
on quadruple drop-out agar (SD/-leu/-trp/-his/-ade). Growth on this medium indicated that
the bait proteins interacted with the prey protein of interest. The colonies were also tested
for β-galactosidase (β-Gal) activity using a colony lift assay to confirm a positive
interaction.
Cell culture and transfection
COS7 cells were obtained from Dr Klaus Hahn (University of North Carolina,
Chapel Hill, NC, USA) and HEK293T cells were obtained from Dr Alexandra Newton
(University of California, San Diego, CA, USA). HeLa cells were purchased from ATCC.
The cells were grown in Dulbecco's modified Eagle's high glucose medium (Invitrogen)
containing 10% FBS (Hyclone Laboratories), penicillin and streptomycin. COS7 cells were
276
transfected using Fugene6 transfection reagent (Roche Diagnostics), and the HEK and
HeLa cells were transfected with Lipofectamine 2000 transfection reagent (Invitrogen),
both according to the manufacturers’ instructions.
Immunofluorescence
COS7 cells were plated on coverslips. Twelve hours after the transfection, the cells
were fixed in 3% paraformaldehyde in 100 mm phosphate buffer, pH 7.4, for 30 min,
permeabilized with 0.1% Triton X−100 for 10 min, blocked with 10% goat serum for 30
min, and incubated with primary antibodies for 1 h at room temperature, followed by Alexa
Fluor−594 or 488-conjugated antibodies (Molecular Probes) for 1 h at room temperature.
The specimens were visualized using an inverted confocal laser-scanning microscope
(FV1000, Olympus) equipped with a PlanApo 60×/1.42 oil immersion objective
(Olympus). Olympus Fluoview software version 1.6b was used for image acquisition and
analysis. The images were further processed using Adobe Photoshop (Adobe Systems). To
quantify the relative colocalization in multiple images (n = 10–15 cells), the Pearson
correlation coefficients (R values) were calculated for each image set for pixels above the
calculated thresholds using Olympus Fluoview v1.6b colocalization software. The Pearson
coefficient is a statistical appraisal of how well a linear equation describes the relationship
between two variables for a measured function and is commonly used in image
colocalization analysis.
Cell fractionation (S100/P100 centrifugation)
Cytosolic and membrane fractions were prepared as described previously (3). HeLa
cells were washed in cold PBS, scraped into cold homogenization buffer [50 mm Tris, pH
7.4, 150 mm NaCl, 5 mm ethylenediaminetetraacetic acid (EDTA) with protease
inhibitors], and centrifuged for 5 min at 500 ×g. The pellet was homogenized by 40
passages through a 25 G 5/8 needle. Nuclei and unbroken cells were removed by
centrifugation (1000 ×g for 10 min). The post-nuclear supernatant was collected and
centrifuged at 100,000 ×g for 1 h at 4^C. The soluble or cytosolic fraction (S100) was
collected, and the pellet or membrane fraction (P100) was resuspended using a pestle in an
equal volume of buffer. The same volumes (60 μL) of membrane and cytosolic fractions
were analyzed by SDS-PAGE and immunoblotting.
GST pull-down assays
Full-length rCalnuc cDNA or Calnuc fragments containing the N-terminus (residues
1–168) or C-terminus (residues 335–459) were amplified and subcloned in pGEX-KG
(Amersham Biosciences). GST fusion proteins were expressed in E. coli BL21 and purified
on glutathione-Sepharose 4B beads (Pharmacia) according to the manufacturer's
instructions. 35S-labeled in vitro translation products of pCDNA3.1-human LRP9 Cterminus (residues 556–713) were prepared using the TNT T7 rabbit reticulocyte Quick
Coupled Transcription/Translation system (Promega) in the presence of [35S]methionine
(1000 Ci/mmol, in vivo cell labeling grade; Amersham Biosciences). For the pull-down
assays, GST fusion proteins (10 μg) immobilized on beads were incubated with in vitrotranslated products in 20 mm Tris–HCl buffer (pH 7.4) containing 150 mm NaCl, 3 mm
EDTA and 0.1% NP−40 in the presence of protease inhibitors for 3 h at 4^C and were
washed four times with the same buffer. For the lysate GST pull-down assays, 1 mg of cell
277
lysate or cytosolic fraction (S100) in buffer [50 mm Tris, pH 7.4, 150 mm NaCl, 1%
NP−40, 5 mm EDTA and complete protease inhibitors (Roche)] was incubated overnight at
4^C with 10 or 30 μg of GST fusion proteins immobilized on glutathione-Sepharose 4B
beads. The beads were washed three times in lysis buffer and boiled in Laemmli sample
buffer. The bound proteins were separated by SDS-PAGE and detected by autoradiography
or immunoblotting.
Coimmunoprecipitation
HEK cells were plated in 60-mm culture dishes and transfected with the various
constructs. After 48 h, the cells were lysed in 50 mm Tris buffer (pH 7.4) containing 150
mm NaCl, 1% NP−40 and protease inhibitors for 1 h at 4^C and then centrifuged at 13,000
×g for 20 min. The cleared supernatants were incubated with primary antibodies overnight
at 4^C and then with protein A-sepharose (GE Healthcare) or protein G-Sepharose (Zymed)
for 1 h. The beads were washed three times in lysis buffer and boiled in Laemmli sample
buffer. Bound immune complexes were analyzed by SDS-PAGE and immunoblotting.
Immunoblotting
The protein samples were separated on 8 or 10% SDS-PAGE gels and transferred to
nitrocellulose membranes (Perkin Elmer). The membranes were blocked in Tris-buffered
saline (20 mm Tris–HCl, pH 7.4, 150 mm NaCl) containing 0.1% Tween 20 and 5% nonfat
dry milk and incubated with primary antibodies for 1 h at room temperature and then with
horseradish peroxidase-conjugated goat anti-rabbit or anti-mouse IgG (Bio-Rad) for 45 min
at room temperature and enhanced chemiluminescence detection reagent (Pierce Chemical).
RNA interference
Scrambled RNA oligos (scramble II duplex) and Calnuc siRNA were purchased
from Dharmacon Research. HeLa cells were transfected with a final concentration of 100
nM siRNA duplex using Lipofectamine 2000 reagent (Invitrogen) according to the
manufacturer's instructions. The cells were analyzed 72 h after the transfection. HA-LRP9
was transfected with Fugene 12 h before the IF experiments. Reversal of the phenotype was
attempted by transfecting HeLa cells with cDNA encoding rCalnuc-GFP, rat ΔSP-CalnucGFP, or GFP alone using Fugene 5 h after they were transfected with human Calnuc
siRNA.
Cycloheximide chase
Control- and siRNA-treated cells were incubated at 37^C in complete DMEM
containing 25 mm Hepes buffer, pH 7.4 and 40 μg/mL of cycloheximide (Sigma-Aldrich).
At the corresponding time points, the cells were lysed as described above and analyzed by
SDS-PAGE and immunoblotting.
Treatment with lysosomal inhibitors
Control- and siRNA-treated cells were incubated for 3–5 h at 37^C in complete
DMEM containing 1 mg/mL of leupeptin (Roche Diagnostics) and 10 μg/mL of E64
(Sigma-Aldrich). The cells were collected, lysed and analyzed by SDS-PAGE and
immunoblotting.
278
Statistical analysis
Experiments were performed in triplicate and results are expressed as means ± SD.
Statistical significance between various samples was assessed using the Student t-test. A p
< 0.05 was considered significant.
Acknowledgments
We are grateful to Toshio Kitamura for the LRP9 construct, Takahiro Fujino for the
LRP3 construct, to Bradley Hyman for the LRP1 construct, to Thomas Braulke for the
rabbit CI-MPR antibodies, and to Marilyn Gist Farquhar for the generous gifts of reagents.
We would also like to thank Radu Stan for the advices with the yeast-two-hybrid screening.
The LAMP−2 monoclonal antibody developed by J. Thomas August and James E.K.
Hildreth was obtained from the Developmental Studies Hybridoma Bank developed under
the auspices of the NICHD and maintained by The University of Iowa. This work was
supported by grants from the Canadian Institutes for Health Research and a Canada
Research Chair to C.L.
References
1
2
3
4
5
6
7
8
9
Wendel M, Sommarin Y, Bergman T, Heinegard D. Isolation, characterization and
primary structure of a calcium-binding 63-kDa bone protein. J Biol Chem 1995;270:
6125–6133.
Miura K, Hirai M, Kanai Y, Kurosawa Y. Organization of the human gene for
nucleobindin (NUC) and its chromosomal assignment to 19q13.2–q13.4. Genomics
1996;34: 181–186.
Lin P, Le-Niculescu H, Hofmeister R, McCaffery JM, Jin M, Hennemann H,
McQuistan T, De Vries L, Farquhar MG. The mammalian calcium-binding protein,
nucleobindin (CALNUC), is a Golgi resident protein. J Cell Biol 1998;141: 1515–
1527.
Kawano J, Kotani T, Ogata Y, Ohtaki S, Takechi S, Nakayama T, Sawaguchi A,
Nagaike R, Oinuma T, Suganuma T. CALNUC (nucleobindin) is localized in the
Golgi apparatus in insect cells. Eur J Cell Biol 2000;79: 208–217.
Kubota T, Miyauchi M, Miura K, Hirokawa G, Awaya A, Miyasaka N, Kurosawa Y,
Kanai Y, Maruyama K. Upregulation of nucleobindin expression in human-activated
lymphocytes and non-Hodgkin's lymphoma. Pathol Int 1998;48: 22–28.
De Vos S, Hofmann WK, Grogan TM, Krug U, Schrage M, Miller TP, Braun JG,
Wachsman W, Koeffler HP, Said JW. Gene expression profile of serial samples of
transformed B-cell lymphomas. Lab Invest 2003;83: 271–285.
Howell BG, Solish N, Lu C, Watanabe H, Mamelak AJ, Freed I, Wang B, Sauder
DN. Microarray profiles of human basal cell carcinoma: insights into tumor growth
and behavior. J Dermatol Sci 2005;39: 39–51.
Chen Y, Lin P, Qiu S, Peng XX, Looi K, Farquhar MG, Zhang JY. Autoantibodies to
Ca2+ binding protein Calnuc is a potential marker in colon cancer detection. Int J
Oncol 2007;30: 1137–1144.
Gronborg M, Kristiansen TZ, Iwahori A, Chang R, Reddy R, Sato N, Molina H,
Jensen ON, Hruban RH, Goggins MG, Maitra A, Pandey A. Biomarker discovery
279
10
11
12
13
14
15
16
17
18
19
20
21
22
23
24
25
from pancreatic cancer secretome using a differential proteomic approach. Mol Cell
Proteomics 2006;5: 157–171.
Tamura K, Furihata M, Tsunoda T, Ashida S, Takata R, Obara W, Yoshioka H,
Daigo Y, Nasu Y, Kumon H, Konaka H, Namiki M, Tozawa K, Kohri K, Tanji N et
al. Molecular features of hormone-refractory prostate cancer cells by genome-wide
gene expression profiles. Cancer Res 2007;67: 5117–5125.
Kanai Y, Miura K, Uehara T, Amagai M, Takeda O, Tanuma S, Kurosawa Y. Natural
occurrence of Nuc in the sera of autoimmune-prone MRL/lpr mice. Biochem Biophys
Res Commun 1993;196: 729–736.
Iizuka H, Kubota T, Satake F, Hirokawa G, Miura K, Kurosawa Y, Miyasaka N,
Kanai Y. Enhanced expression of nucleobindin in lymphatic organs of lupus-prone
mice. Lupus 1997;6: 365–370.
Lin P, Li F, Zhang YW, Huang H, Tong G, Farquhar MG, Xu H. Calnuc binds to
Alzheimer's beta-amyloid precursor protein and affects its biogenesis. J Neurochem
2007;100: 1505–1514.
De Alba E, Tjandra N. Structural studies on the Ca2+-binding domain of human
nucleobindin (calnuc). Biochemistry 2004;43: 10039–10049.
Johnson S, Michalak M, Opas M, Eggleton P. The ins and outs of calreticulin: from
the ER lumen to the extracellular space. Trends Cell Biol 2001;11: 122–129.
Lin P, Yao Y, Hofmeister R, Tsien RY, Farquhar MG. Overexpression of CALNUC
(nucleobindin) increases agonist and thapsigargin releasable Ca2+ storage in the
Golgi. J Cell Biol 1999;145: 279–289.
Lavoie C, Meerloo T, Lin P, Farquhar MG. Calnuc, an EF-hand Ca(2+)-binding
protein, is stored and processed in the Golgi and secreted by the constitutive-like
pathway in AtT20 cells. Mol Endocrinol 2002;16: 2462–2474.
Petersson U, Somogyi E, Reinholt FP, Karlsson T, Sugars RV, Wendel M.
Nucleobindin is produced by bone cells and secreted into the osteoid, with a potential
role as a modulator of matrix maturation. Bone 2004;34: 949–960.
Taniguchi N, Taniura H, Niinobe M, Takayama C, Tominaga-Yoshino K, Ogura A,
Yoshikawa K. The postmitotic growth suppressor necdin interacts with a calciumbinding protein (NEFA) in neuronal cytoplasm. J Biol Chem 2000;275: 31674–
31681.
Battle MA, Maher VM, McCormick JJ. ST7 is a novel low-density lipoprotein
receptor-related protein (LRP) with a cytoplasmic tail that interacts with proteins
related to signal transduction pathways. Biochemistry 2003;42: 7270–7282.
Hussain MM, Strickland DK, Bakillah A. The mammalian low-density lipoprotein
receptor family. Annu ReV Nutr 1999;19: 141–172.
Strickland DK, Gonias SL, Argraves WS. Diverse roles for the LDL receptor family.
Trends Endocrinol Metab 2002;13: 66–74.
Yamamoto T, Davis CG, Brown MS, Schneider WJ, Casey ML, Goldstein JL,
Russell DW. The human LDL receptor: a cysteine-rich protein with multiple Alu
sequences in its mRNA. Cell 1984;39: 27–38.
Bork P, Beckmann G. The CUB domain. A widespread module in developmentally
regulated proteins. J Mol Biol 1993;231: 539–545.
Christensen EI, Birn H. Megalin and cubilin: multifunctional endocytic receptors. Nat
Rev Mol Cell Biol 2002;3: 256–266.
280
26
27
28
29
30
31
32
33
34
35
36
37
38
39
40
Sugiyama T, Kumagai H, Morikawa Y, Wada Y, Sugiyama A, Yasuda K, Yokoi N,
Tamura S, Kojima T, Nosaka T, Senba E, Kimura S, Kadowaki T, Kodama T,
Kitamura T. A novel low-density lipoprotein receptor-related protein mediating
cellular uptake of apolipoprotein E-enriched beta-VLDL in vitro. Biochemistry
2000;39: 15817–15825.
Boucher R, Larkin H, Brodeur J, Gagnon H, Thériault C, Lavoie C. Intracellular
trafficking of LRP9 is dependent on two acidic cluster/dileucine motifs. Histochem
Cell Biol 2008;130: 317–327.
Doray B, Knisely JM, Wartman L, Bu G, Kornfeld S. Identification of acidic
dileucine signals in LRP9 that interact with both GGAs and AP−1/AP−2. Traffic
2008;9: 1551–1562.
Puertollano R, Aguilar RC, Gorshkova I, Crouch RJ, Bonifacino JS. Sorting of
mannose 6-phosphate receptors mediated by the GGAs. Science 2001;292: 1712–
1716.
Tikkanen R, Obermuller S, Denzer K, Pungitore R, Geuze H, Von Figura K, Honing
S. The dileucine motif within the tail of MPR46 is required for sorting of the receptor
in endosomes. Traffic 2000;1: 631–640.
Tortorella LL, Schapiro FB, Maxfield FR. Role of an acidic cluster/dileucine motif in
cation-independent mannose 6-phosphate receptor traffic. Traffic 2007;8: 402–413.
Weiss TS, Chamberlain CE, Takeda T, Lin P, Hahn KM, Farquhar MG. Galpha i3
binding to calnuc on Golgi membranes in living cells monitored by fluorescence
resonance energy transfer of green fluorescent protein fusion proteins. Proc Natl
Acad Sci USA 2001;98: 14961–14966.
Stenmark H, Parton RG, Steele-Mortimer O, Lutcke A, Gruenberg J, Zerial M.
Inhibition of rab5 GTPase activity stimulates membrane fusion in endocytosis.
EMBO J 1994;13: 1287–1296.
Manders E, Verbeek F, Aten J. Measurement of colocalization of objects in dualcolour confocal images. J Microsc 1993;169: 375–382.
Martin-Nieto J, Villalobo A. The human epidermal growth factor receptor contains a
juxtamembrane calmodulin-binding site. Biochemistry 1998;37: 227–236.
Li E, Hristova K. Role of receptor tyrosine kinase transmembrane domains in cell
signaling and human pathologies. Biochemistry 2006;45: 6241–6251.
Tebar F, Villalonga P, Sorkina T, Agell N, Sorkin A, Enrich C. Calmodulin regulates
intracellular trafficking of epidermal growth factor receptor and the MAPK signaling
pathway. Mol Biol Cell 2002;13: 2057–2068.
Aifa S, Aydin J, Nordvall G, Lundstrom I, Svensson SP, Hermanson O. A basic
peptide within the juxtamembrane region is required for EGF receptor dimerization.
Exp Cell Res 2005;302: 108–114.
Llado A, Timpson P, Vila de Muga S, Moreto J, Pol A, Grewal T, Daly RJ, Enrich C,
Tebar F. Protein kinase Cdelta and calmodulin regulate epidermal growth factor
receptor recycling from early endosomes through Arp2/3 complex and cortactin. Mol
Biol Cell 2008;19: 17–29.
Rohrer J, Schweizer A, Johnson KF, Kornfeld S. A determinant in the cytoplasmic
tail of the cation-dependent mannose 6-phosphate receptor prevents trafficking to
lysosomes. J Cell Biol 1995;130: 1297–1306.
281
41
42
43
44
45
46
47
48
49
50
51
52
53
54
55
56
Seaman MN. Identification of a novel conserved sorting motif required for retromermediated endosome-to-TGN retrieval. J Cell Sci 2007;120: 2378–2389.
Arighi CN, Hartnell LM, Aguilar RC, Haft CR, Bonifacino JS. Role of the
mammalian retromer in sorting of the cation-independent mannose 6-phosphate
receptor. J Cell Biol 2004;165: 123–133.
Nielsen MS, Gustafsen C, Madsen P, Nyengaard JR, Hermey G, Bakke O, Mari M,
Schu P, Pohlmann R, Dennes A, Petersen CM. Sorting by the cytoplasmic domain of
the amyloid precursor protein binding receptor SorLA. Mol Cell Biol 2007;27: 6842–
6851.
Mari M, Bujny MV, Zeuschner D, Geerts WJ, Griffith J, Petersen CM, Cullen PJ,
Klumperman J, Geuze HJ. SNX1 defines an early endosomal recycling exit for
sortilin and mannose 6-phosphate receptors. Traffic 2008;9: 380–393.
Nielsen MS, Madsen P, Christensen EI, Nykjaer A, Gliemann J, Kasper D, Pohlmann
R, Petersen CM. The sortilin cytoplasmic tail conveys Golgi-endosome transport and
binds the VHS domain of the GGA2 sorting protein. EMBO J 2001;20: 2180–2190.
Ghosh P, Dahms NM, Kornfeld S. Mannose 6-phosphate receptors: new twists in the
tale. Nat Rev Mol Cell Biol 2003;4: 202–212.
Jacobsen L, Madsen P, Jacobsen C, Nielsen MS, Gliemann J, Petersen CM.
Activation and functional characterization of the mosaic receptor SorLA/LR11. J Biol
Chem 2001;276: 22788–22796.
Honing S, Sosa M, Hille-Rehfeld A, Von Figura K. The 46-kDa mannose 6phosphate receptor contains multiple binding sites for clathrin adaptors. J Biol Chem
1997;272: 19884–19890.
Nielsen MS, Madsen P, Christensen EI, Nykjær A, Gliemann J, Kasper D, Pohlmann
R, Petersen1 CM. The sortilin cytoplasmic tail conveys Golgi-endosome transport
and binds the VHS domain of the GGA2 sorting protein. EMBO J 2001;20: 2180–
2190.
Jacobsen L, Madsen P, Nielsen MS, Geraerts WP, Gliemann J, Smit AB, Petersen
CM. The sorLA cytoplasmic domain interacts with GGA1 and −2 and defines
minimum requirements for GGA binding. FEBS Lett 2002;511: 155–158.
Canuel M, Lefrancois S, Zeng J, Morales CR. AP−1 and retromer play opposite roles
in the trafficking of sortilin between the Golgi apparatus and the lysosomes. Biochem
Biophys Res Commun 2008;366: 724–730.
Sahagian GG, Distler J, Jourdian GW. Characterization of a membrane-associated
receptor from bovine liver that binds phosphomannosyl residues of bovine testicular
beta-galactosidase. Proc Natl Acad Sci USA 1981;78: 4289–4293.
Hoflack B, Kornfeld S. Purification and characterization of a cation-dependent
mannose 6-phosphate receptor from murine P388D1 macrophages and bovine liver. J
Biol Chem 1985;260: 12008–12014.
Collins BM. The structure and function of the retromer protein complex. Traffic
2008;9: 1811–1822.
Bonifacino JS, Hurley JH. Retromer. Curr Opin Cell Biol 2008;20: 427–436.
Meyer C, Zizioli D, Lausmann S, Eskelinen EL, Hamann J, Saftig P, Von Figura K,
Schu P. mu1A-adaptin-deficient mice: lethality, loss of AP−1 binding and rerouting
of mannose 6-phosphate receptors. EMBO J 2000;19: 2193–2203.
282
57
58
59
Diaz E, Pfeffer SR. TIP47: a cargo selection device for mannose 6-phosphate
receptor trafficking. Cell 1998;93: 433–443.
Krise JP, Sincock PM, Orsel JG, Pfeffer SR. Quantitative analysis of TIP47-receptor
cytoplasmic domain interactions: implications for endosome-to-trans Golgi network
trafficking. J Biol Chem 2000;275: 25188–25193.
Scott GK, Fei H, Thomas L, Medigeshi GR, Thomas G. A PACS−1, GGA3 and CK2
complex regulates CI-MPR trafficking. EMBO J 2006;25: 4423–4435.
283
ANNEXE 3
Macrocyclic cell penetrating peptides: a study of
structure-penetration properties.
Traboulsi, H., Larkin, H., Bonin, M.-A., Volkov, L., Lavoie, C., and
Marsault, É.
Bioconjug Chem. (2015)
Travaux en collaboration
IF= 4,58
284
Macrocyclic cell penetrating peptides: a study of structure-penetration properties.
Traboulsi, H., Larkin, H., Bonin, M.-A., Volkov, L., Lavoie, C., and Marsault, É.
Abstract
Arginine-rich cell penetrating peptides are short cationic peptides able to cross
biological membranes despite their peptidic character. In order to optimize their penetration
properties and further elucidate their mechanisms of cellular entry, these peptides have
been intensively studied for the last two decades. Although several parameters are
simultaneously involved in the internalization mechanism, recent studies suggest that
structural modifications influence cellular internalization. Particularly, backbone
rigidification, including macrocyclization, was found to enhance proteolytic stability and
cellular uptake. In the present work, we describe the synthesis of macrocyclic arginine-rich
cell penetrating peptides and study their cellular uptake properties using a combination of
flow cytometry and confocal microscopy. By varying ring size, site of cyclization, and
stereochemistry of the arginine residues, we studied their structure–uptake relationship and
showed that the mode and site of cyclization as well as the stereochemistry influence
cellular uptake. This study led to the identification of a hepta-arginine macrocycle as
efficient as its linear nona-arginine congener to enter cells.
Introduction
Cell-penetrating peptides (CPPs), originally known as protein transduction domains,
are peptides consisting of fewer than 35 amino acids capable of penetrating cells despite
their peptidic character. Initially identified as specific transduction domains embedded in
protein sequences, they represent a natural mechanism of cellular targeting and entry.(1-4)
Since their discovery, the cell penetrating properties of CPPs have been harnessed to
internalize otherwise impermeable cargos into cells, including small molecules,
macromolecules (e.g., proteins, nucleic acids), and nanoparticles.(5-11) The majority of
CPPs contain a high relative abundance of positively charged amino acids such as lysine
285
(Lys) or arginine (Arg), or display alternating patterns of polar/charged amino acids and
nonpolar, hydrophobic amino acids.(9, 12, 13)
Arginine-rich CPPs have been used successfully to deliver a broad diversity of
biologically active macromolecules intracellularly, although their mechanisms of cellular
entry are still under investigation.(9, 12, 14-17) In the process, the guanidinium cationic
groups of the Arg residues are crucial for efficient cell penetration, a role attributed to their
capacity to interact with the various anion types localized on the plasma membrane such as
the polar heads of phospholipids or the sulfate groups of glycosaminoglycans.(13, 18, 19)
Polyarginines are readily synthesized and represent the structurally simplest CPPs.
They have been studied extensively for their ability to penetrate through cell membranes. It
was found that cellular uptake can be achieved with oligoarginines composed of 5–15
residues.(14, 20) In particular, nona-arginine (R9) was shown to possess improved cell
penetration efficiency compared to TAT peptides.(9, 20) Thus, several studies have utilized
R8 and R9 as reference tools to import into cells a variety of biological molecules including
siRNA, anticancer drugs, small molecules, proteins, peptides, and oligonucleotides.(10, 11)
The mechanisms by which CPPs translocate across cell membranes remain a subject
of investigation. Certain CPPs, when attached to small cargos, translocate passively across
the plasma membrane of cells.(21) However, an increasing number of studies have shown
that cellular uptake of cationic CPPs, when conjugated to macromolecules or used at low
concentrations, is temperature-sensitive, suggesting the involvement of energy-dependent
processes.(17, 22-25) To better delineate the mechanisms at play, Matsushita et al. recently
applied siRNA library screening to identify potential partners involved in the cellular entry
of polyarginine peptides. This led to the identification of the involvement of cotransporter
gene SLC4A4 and the trafficking regulator gene COPA in cellular entry. Knowing that
COPA plays an important role in early endosome maturation, these results suggest that
cellular entry of polyarginine involves at least two steps, such as initial binding to the cell
surface, followed by endosomal entry.(26)
Since polyarginines are the most commonly used CPPs, their optimization has been
investigated. Replacement of (l)-Arg residues by (d) analogues and backbone modifications
have produced protease-resistant analogues with improved translocation properties.(12, 13)
Major structural modifications aimed at improving cellular penetration and proteolytic
stability, while retaining acceptable cytotoxicity, have met with various degrees of success.
In particular, it has been demonstrated that increased rigidity and static display of guanidine
groups is beneficial for cellular entry properties.(12, 13, 27-32)
In this context, macrocyclization is a well established approach to simultaneously
rigidify a peptide backbone, modulate its structure–activity, improve its proteolytic
stability, and reduce the entropic cost of macrocycle–target interactions.(33-36) These
attributes are attractive for improving the properties of CPPs;(37-44) thus, macrocyclization
and backbone rigidification of CPPs was expected to improve cellular uptake and stability,
as well as to provide a means to discriminate among different cell types. Indeed, it has been
proposed that guanidinium groups are forced into maximally distant positions by
286
cyclization, increasing membrane contacts and leading to enhanced cellular penetration.(28,
31, 45, 46)
To date, there is no systematic study of the features favorable for cellular uptake in
macrocyclic polyarginines (Figure 1). In order to better understand the potential structure–
penetration relationship, we synthesized several analogues in this series, with the goal of
better delineating how structural variations like cyclization, stereochemistry, or endocyclic
vs exocyclic display of arginine residues influence cellular uptake.
Compounds presented in this study were functionalized with a fluorescent label in the
form of a fluorescein isothiocyanate (FITC) moiety. Their cellular penetration properties
were assessed using flow cytometry and results were confirmed using fluorescence
confocal microscopy, with the hypothesis that stereochemistry and mode of cyclization
could influence cellular uptake.
Figure 1. Representative structure of cyclic polyarginines (n = 4 and 6–9); (l)
stereochemistry given as an example.
Results
Synthesis
Analogues were synthesized as described in Figure 2 and Supporting Information (SI)
Figure S1. Briefly, synthesis relied on Fmoc chemistry and macrocyclization was
performed via the formation of a lactam bridge between a Lys and a Glu residue. These two
residues were previously protected as Alloc and Allyl ester, respectively, then deprotected
under Pd catalysis immediately prior to cyclization. Every macrocycle was functionalized
with FITC and a spacer (β-Ala, except for R9 which was functionalized as an
aminohexanoate as per previous works).
287
Figure 2. Synthetic scheme for the preparation of cyclic polyarginine analogues.
A total of 23 analogues (4 linear and 19 cyclic) were synthesized for this study, as
summarized in Table 1. Entries 1–4 (Table 1) are reference compounds aimed at comparing
cellular uptake with published results; entries 5–15 are side-chain to side-chain
macrocycles containing 4–9 homochiral (l)Arg or (d)Arg residues or heterochiral (l,d)Arg
residues. It should be kept in mind that between homo- and heterochiral macrocycles, the
projection of side chains with respect to the plane of the macrocycle differs. Entries 16–19
are bicyclic macrocycles built as described in SI Figure S1, with the goal of assessing the
influence of additional rigidification on cellular uptake; finally, entries 20–23 are
macrocyclic polyarginines (3–6 residues) bearing 1–4 exocyclic arginine residues built to
assess the influence of endocyclic vs exocyclic residues on cellular uptake. Compounds
were synthesized on solid phase synthesis and purified to >95% purity (UPLC-UV).
The cellular uptake of new compounds was first assessed by flow cytometry, then
confirmed using confocal microscopy as described below.
Table 1. Sequences of Linear and Macrocyclic Peptides Used in This Study. Lower case
letters indicate (d) and upper case letters (l) stereochemistry. Residues between brackets are
included in the macrocycle, closed by macrolactamization between the Glu (E) and Lys (K)
side chains.
288
Flow Cytometry
Flow cytometry analysis was used to quantify the cellular uptake of the new
constructs in HeLa cells. Uptake measurements were based on changes in the mean cellular
fluorescence following incubation with FITC-labeled peptides, relatively to linear control
R9. Knowing that conventional flow cytometry cannot distinguish between intracellular
and cell surface fluorescence, trypsin treatment was applied (Trypsin-EDTA 0.05%, 5 min)
to remove cell surface-bound peptides prior to analysis. The efficiency of trypsin treatment
for macrocycles was confirmed since treatment of Cyc-R4, which contains only 4 Arg
residues, showed a very weak emission signal comparable to the signal of autofluorescence,
confirming that membrane-bound cyclic peptides were indeed detached from the cell
surface (Figure 3B). Additionally, it was confirmed that the uptake of Cyc-R9 is
concentration-dependent (Supporting Information, Figure S2).
Figure 3. Flow cytometry analysis of HeLa cells treated with (A) linear reference
compounds; (B) all (l) macrocycles; (C) all (d) macrocycles; and (D) alternating (l,d)
macrocycles. Each value represents the average of three experiments, and values are
normalized with respect to control compound R9.
Linear peptides R6–R9 were used as reference compounds and demonstrated an
increasing uptake with increasing arginine contents (Figure 3A), consistently with previous
literature reports.(14) In order to better understand the influence of structural variations on
cellular uptake of macrocyclic constructs, we next analyzed the uptake of the cyclic
analogues of the polyarginine peptides (Figure 3B). Cyc-R4 and Cyc-R6 showed a very
weak emission signal (commensurate with the autofluorescence signal), suggesting little or
no uptake. A significant increase in uptake was noted for Cyc-R8 and Cyc-R9. Importantly,
Cyc-R9 displayed 40% higher uptake than its linear analogue R9.
In order to better understand the influence of stereochemistry on internalization by
HeLa cells, we prepared both homochiral (l) macrocycles, homochiral (d) macrocycles, as
well as heterochiral (l,d) macrocycles, then compared their uptake by flow cytometry
(Figure 3B–D). It is interesting to note that cycloocta-arginine macrocycles were
289
particularly sensitive to the influence of stereochemistry, as opposed to their cycloheptaarginine counterparts. Indeed, homochiral (l) analogue Cyc-R8 displayed higher levels of
uptake (125% the value obtained for linear R9) than analogues Cyc-r8 and Cyc-(l,d)-R8
(63% and 75% the value reached by R9, respectively). In fact, Cyc-R8 demonstrated
improved penetration compared to the standard linear R9 peptide. Similar observations
were made for cyclonona-arginines. For example, Cyc-R9 was internalized twice as much
as Cyc-r9 and 50% more than its linear analogue. Finally, Cyc-R9 macrocycle displayed an
improved uptake compared to all the other cyclic or linear analogues (Figure 3B).
To confirm that the fluorescence observed in flow cytometry is intracellular and not
associated with cell surface, uptake experiments of R9, Cyc-R9, and Cyc-r9 were repeated
in the presence of Trypan Blue to quench extracellular fluorescence (Figure 4, white vs
black bars).(47) No major loss of fluorescence was observed in the presence of Trypan
Blue in the case of R9 and Cyc-R9, indicating that the observed fluorescence is indeed
intracellular. In the case of Cyc-r9, 20% of the fluorescence was quenched in the presence
of Trypan Blue, suggesting that a small portion of the compounds remain membranebound. However, when Triton-X100 was added to permeabilize the cells, extensive
quenching was observed, as expected if the fluorescence is intracellular (Figure 4, gray
bars).
Figure 4. Flow cytometry analysis of HeLa cells treated with R9, Cyc-R9, and Cyc-r9
peptides in the presence and absence of Trypan blue and Triton-X100. Each value
represents the average of three experiments.
In order to better understand how further rigidification influences cellular uptake, we
next explored the effect of double cyclization, spacing between the two macrocycles, and
the respective contribution of macrocyclic and linear groups on cellular uptake. This was
performed on analogues bearing 6 Arg derivatives, to assess whether increased structural
rigidification could bring these analogues to levels of uptake similar to those observed with
linear R8 or R9 (see Table 1 for structures). Indeed, it was demonstrated that rigid scaffolds
bearing as little as 4 or 5 guanidine groups could be as efficient as linear R9 to mediate
cellular uptake.(27-29, 31, 32) Macrobicyclic hexa-arginine derivatives were synthesized
using a similar approach to that of macrocycles with a single ring (see SI Figure S1). In
some cases, spacers composed of 1–3 βAla residues were incorporated between the two
290
rings, in order to introduce some flexibility to the peptide and potentially impart some αhelical contents.(48) For comparison, we present in Figure 5A the results obtained by flow
cytometry of Cyc-R6 and the different bicyclic peptides bearing 6 Arg residues. One can
observe that in the whole series of hexa-arginine derivatives, the mode of cyclization and
the presence of a linker had little influence on cellular uptake, which remained low in all
cases. Bicyclization of hexa-arginine had either no effect, or a small tendency toward a
negative effect.
We next investigated the respective impact of endo- vs exocyclic Arg residues on
cellular uptake. Given the poor results obtained in the previous hexa-arginine bicyclic
derivatives, this was performed on hepta-arginine derivatives (Figure 5B). In this series, we
compared a macrocycle containing 7 endocyclic Arg groups (Cyc-R7) to analogues bearing
6 endocyclic and 1 exocyclic Arg (Cyc-R6-R1), 5 endocyclic and 2 exocyclic Arg (CycR5-R2), 4 endocyclic and 3 exocyclic Arg (Cyc-R4-R3), and finally 3 endocyclic and 4
exocyclic Arg (Cyc-R3-R4) groups. Thus, the number of Arg residues was kept constant in
this series. Flow cytometry results (Figure 5B) confirmed the possibility to significantly
increase cellular uptake via the structural modification of these peptides. The optimal
structure, Cyc-R4-R3, contains 4 endocyclic and 3 exocyclic Arg residues. Ultimately, the
latter reached the same level of internalization as the standard linear R9 derivative, despite
the fact that it possesses 7 instead of 9 Arg residues. In the linear series (Figure 3A), the
internalization of linear R7 was ∼40% of that of linear R9. These results were consistent
with those observed in confocal microscopy (see below).
Figure 5. Flow cytometry analysis of HeLa cells treated with (A) bicyclic and (B) 7
arginine monocyclic peptides. Each value represents an average of three experiments, and
values are normalized with respect to control compound R9.
Confocal Microscopy
The cellular uptake of selected linear and macrocyclic peptides was subsequently
assessed by confocal microscopy on HeLa cells. Toward this end, cells were incubated with
compounds (5 μM) for 30 min at 37 °C, then imaged by confocal microscopy after fixation
on coverslips, or using live cells. The images of fixed HeLa cells treated with linear
291
peptides R6–R9 are shown in Figure 6 (top panels) as controls. Cells treated with linear R6
polyarginine showed little, if any, intracellular fluorescence, while treatment with R7–R9
showed increasing levels of fluorescence with increasing number of Arg residues under the
same imaging conditions, as previously reported.(14)
Similarly to linear peptides, internalization of macrocyclic peptides showed strongly
diffused signals for peptides containing 7, 8, and 9 arginine residues (Cyc-R7, Cyc-R8,
Cyc-R9), consistent with a predominantly cytoplasmic distribution (Figure 6, middle
panels). As observed with linear peptides, increasing the number of Arg residues was
associated with increased uptake from Cyc-R6 to Cyc-R8. However, there appeared to be
no significant increase between Cyc-R8 to Cyc-R9 (Figure 6, middle panels). In parallel,
the macrocyclic peptides possessing an exocyclic arginine chain showed significant
variations in cellular uptake, whereby Cyc-R3-R4 displayed a stronger diffused signal
(Figure 6, bottom panels) compared with the other peptides of this family (Cyc-R4-R3,
Cyc-R5-R2, Cyc-R6-R1).
Figure 6. Confocal microscopy images of HeLa cells treated with FITC-labeled linear and
macrocyclic peptides (in green). Nuclei are marked in blue. Scale bar, 10 μm.
The results of internalization of Cyc-r7, Cyc-r8, and Cyc-r9, as well as those of Cyc(l,d)-R6, Cyc-(l,d)-R7, and Cyc-(l,d)-R9, were also confirmed by confocal microscopy (SI
Figures S3, S4).
Selected linear and macrocyclic peptides were also tested using live cell imaging (SI
Figure S5), given that fixation may lead to artifactual cell permeabilization and uptake.(49)
292
Essentially, results obtained with live cell imaging were consistent with those obtained with
fixed cells, indicating that fixation did not alter cellular penetration.
Discussion
The aim of this study was to better understand how the mode of macrocyclization, the
stereochemistry, and the endo vs exocyclic display of Arg in macrocyclic polyarginines
influence cellular uptake. Indeed, it was previously reported, on one hand, that a minimal
number of 8–9 arginine residues is required for efficient cell entry of linear arginine-rich
peptides, yet it is also known that structural rigidification of the molecule could improve
cellular penetration and allow a reduction in the number of exposed guanidine motifs, down
to 4 or 5 guanidine units.(27-29, 31, 32) Flow cytometry was used to quantify the extent of
cellular uptake in the various series, and confocal microscopy was used to confirm uptake
and assess preliminarily the intracellular distribution. It should be kept in mind that the only
charged units in these peptides are the guanidinium groups since Rink amide resin was used
to deliver C-terminal amide peptides.
First, results from flow cytometry of the control linear peptides confirmed that
increasing the number of arginine increased cell internalization by 20% to 30% per arginine
residue, as previously reported (Figure 3A).(14)
We further asked how structural modifications to the macrocycles could influence
cellular uptake. Indeed, reported molecular modeling suggested that a subset of the side
chain guanidinium groups of these transporters might be required for transport involving
contact with a common surface such as a plasma membrane or cell surface receptor,
suggesting the possibility to develop the structure–uptake relationship and possibly cellular
specificity.(27-29, 31, 46, 50) Spacers such as glycine have been introduced between
arginine amino acids, showing that the spacing between the arginine residues in a set of
linear peptides could influence uptake.(50) Consequently, by cyclizing polyarginine
peptides, we expected to increase the distance between the guanidinium groups by way of
conformational restrictions, without the addition of spacer elements and increase in
molecular weight. Uptake measurements using flow cytometry on monocyclic peptides
(Figure 3B) showed a dramatic effect on the cellular uptake of octa- and nona-arginine
derivatives. The cyclic form of octa-arginine was not only able to penetrate into cells more
efficiently than the linear form, it also exhibited superior uptake than control linear nonaarginine. This result is in agreement with literature describing the cyclic arginine-rich cellpenetrating peptides showing enhanced cellular uptake relative to their linear and more
flexible counterpart.(31, 38) It has been proposed that guanidinium groups are forced into
maximally distant positions by cyclization and this orientation increases membrane
contacts leading to enhanced cell penetration. Interestingly, macrocyclic analogues of (l)Arg gave much higher penetration than those of (d)-Arg congeners or those alternating (l)
and (d) stereochemistry (Figure 3B–D). This result is in agreement with the works of
Verdurmen et al., who reported that cationic (l)-cell-penetrating peptides are taken up more
efficiently than their (d)-counterparts in MC57 fibrosarcoma and HeLa cells but not in
Jurkat T leukemia cells.(46)
293
To further rigidify structure and improve cellular uptake, we examined bicyclic hexaarginine analogues (Figure 5). Despite increased structural rigidification, cellular uptake of
these variously double-cyclized analogues remained very low (Figure 5A).
Subsequently, the position of cyclization within the macrocycle was varied. Based on
the disappointing results obtained in the hexa-arginine series, we focused this part of the
study on hepta-arginine derivatives. Thus, hepta-arginine macrocycles containing 7, 6, 5, 4,
or 3 endocyclic and 0, 1, 2, 3, or 4 exocyclic Arg residues, respectively, were synthesized
(Table 1). Flow cytometry data showed an important effect of changing the position of
macrocyclization on cellular uptake. Indeed, all the hepta-arginine cyclic peptides with
exocyclic arginine residues possessed enhanced cellular internalization compared to CycR7 with 7 endocyclic Arg residues. Interestingly, the cyclic form Cyc-R4-R3 was able to
penetrate cells more efficiently than the other hepta-arginine derivatives, and possessed an
uptake comparable to that displayed by linear nona-arginine (Figure 5B). This important
result confirms that structural modification of the peptide enhances cellular uptake per Arg
residue in this macrocyclic series; however, a clear relationship remains to be identified.
We performed circular dichroism analyses of the peptides presented in this manuscript (SI
Figures S6–S8); however, they did not lead to any conclusive relationship between
apparent secondary structure and cellular uptake.
Results from confocal microscopy performed on fixed or live cells (Figure 6 and SI
Figures S3–S5) were similar and demonstrated qualitatively that 7 Arg residues are
necessary to reach a decent level of uptake.
Conclusion
We have synthesized diversely cyclized macrocyclic oligoarginine analogues in order
to better understand the influence of conformational restrictions on cellular penetration in
HeLa cells. Similarly to linear peptides, our results showed that increasing arginine content
in the macrocycles enhanced cellular uptake. We also demonstrated that incorporation of 6
arginines in bicyclic peptides was not sufficient to enhance cellular uptake. Finally, we
explored the effect of stereochemistry and mode of cyclization and provided the evidence
of an important effect on cellular uptake. In this series, the most efficient macrocycle
contained 4 endocyclic and 3 exocyclic Arg residues. We are currently investigating
additional macrocyclic analogues of the hepta-arginine family, as well as further details on
the mechanisms of cellular uptake and the potential of this new macrocycle to mediate the
cellular uptake of functional cargos.
Experimental Section
Peptide Synthesis
Linear and cyclic peptides were synthesized using standard solid-phase synthesis with
Fmoc chemistry on a Tribute peptide synthesizer (Protein Technologies Inc.) with IR
activation. The protected amino acids Fmoc-l-Arg(Pbf)-OH, Fmoc-d-Arg(Pbf)-OH, Fmocl-Glu(OAll)-OH, Fmoc-l-Lys(Alloc)-OH, and Fmoc-βAla-OH were purchased from
ChemImpex International and used with no further purification. The protected peptide
294
resins used to synthesize the macrocycles were prepared using 0.25 g of Rink amide resin
from Rapp Polymere (0.23 mmol of NH2/g of resin) by first coupling to the resin,
previously deprotected with piperidine/DMF (1:1). Couplings were performed using HATU
(1-[Bis(dimethylamino)methylene]-1H-1,2,3-triazolo[4,5-b]pyridinium
3-oxide
hexafluorophosphate), using ∼20 min for all amino acids as monitored by UV. Upon
complete formation of the protected linear peptide, resins were washed with
dimethylformamide (DMF), isopropanol, and dichloromethane (DCM) and dried in vacuo.
At this step, the Alloc and Allyl protecting groups of Lys and Glu residues were
simultaneously removed. A solution of dimedone (15 equiv) in 10 mL dry tetrahydrofuran
(THF) was added under argon to the resin. Subsequently, 0.45 equiv of Pd(PPh3)4 was
added to the resin and the reaction was mechanically stirred for 3 h under Ar. Then, the
resin was washed with DMF, isopropanol, and DCM.
To perform macrocyclization, the resin was suspended in 20 mL DMF, followed by
side-chain to side-chain cyclization of the peptide by addition of PYBOP (Benzotriazol-1yl-oxytripyrrolidinophosphonium hexafluorophosphate, 10 equiv), and DIPEA (4 equiv)
overnight. The last step of synthesis was to attach the FITC fluorophore by deprotecting the
peptide resin with piperidine/DMF (1:1), followed by reaction with FITC in DCM/Pyridine
(7:3) during 30 min. Because FITC conjugates are susceptible to Edman degradation, we
used β-alanine as a spacer.(51) The bicyclic peptides were prepared using the standard solid
phase method described in SI Figure S1.
Peptides were cleaved from the resin and the Pbf protecting groups were
simultaneously removed using a mixture of TFA/TIPS/H2O (95:2.5:2.5) for 5 h. The resin
was filtered, washed with 1 mL of the cleavage solution, and then the crude peptide
recovered by precipitation with cold diethyl ether to give an orange powder that was
purified by preparative HPLC (Waters Autosampler 2707, Quaternary gradient module
2535, UV detector 2489, fraction collector WFCIII) equipped with an ACE5 C18 column
(250 × 21.2 mm, 5 μm spherical particle size) and water + 0.1% TFA and acetonitrile as
eluents. The purification was monitored at 245 nm and the fractions corresponding to the
major peak were collected, pooled, and lyophilized. Purities and molecular weights of the
corresponding peptides were determined using an Acquity H-Class UPLC-MS system with
PDA UV and SQD2Mass detectors equipped with a C18 column (2.1 × 50 mm, 1.7 μm
spherical particle size column). All peptides possessed UV purity >92% (for
characterization details, please refer to Supporting Information).
Confocal Microscopy
HeLa cells were purchased from the American Type Culture Collection (Manassas,
VA, USA). Approximately 5 × 104 cells were plated on coverslips in 35 mm culture dishes.
The cells were grown for 24 h in Dulbecco’s modified Eagle’s high glucose medium
(Invitrogen, Carlsbad, CA, USA) supplemented with 10% fetal bovine serum (FBS)
(Hyclone Laboratories, Logan, UT, USA), and 1% Penicillin–Streptomycin–Glutamine
solution (Invitrogen, Carlsbad, CA, USA). The day of the experiment, cells were treated by
adding 5 μM FITC-labeled peptide to the culture medium and incubated for 30 min at 37
°C in the presence of 5% CO2. Cells were then washed twice with phosphate buffered
saline (PBS) and fixed for 30 min in 3% paraformaldehyde (PFA) in 100 mM phosphate
295
buffer, pH 7.4. PFA was quenched for 10 min using a solution of 50 mM ammonium
chloride and nuclei were marked using 1 μg/mL Hoechst 33342 stain for 10 min. The
specimens mounted on slides using mounting medium (1% n-propyl gallate in
glycerol/PBS (1:1)) and visualized using an inverted confocal laser-scanning microscope
(FV1000, Olympus, Tokyo, Japan) equipped with a PlanApo 60×/1.42 oil immersion
objective (Olympus, Tokyo, Japan). Olympus Fluoview software version 1.6a was used for
image acquisition and analysis. The images were further processed using Adobe Photoshop
(Adobe Systems, San Jose, CA, USA). FITC-labeled peptides and nuclei were excited at
488 and 405 nm and detected with 500–600 and 425–475 nm band-pass filters,
respectively.
Flow Cytometry
Approximately 5 × 104 HeLa cells were plated on 35 mm culture dishes. After 48 h,
cells were treated with the FITC-labeled peptides (5 mM) as described above. Cells were
washed once with PBS and taken off using Trypsin-EDTA 0.05% (GIBCO, Invitrogen,
Carlsbad, CA, USA). Finally, cells were resuspended in PBS buffer containing 10 mg/mL
propidium iodide (PI) to establish the live gate to exclude debris and dead cells. A
minimum of 10 000 gated events by sample were acquired and analyzed by a FACScan
cytometer (Becton Dickinson, Mountain View, CA) equipped with a 15 mW argon ion
laser tuned at 488 nm. The emitted fluorescences were split and collected as follows: FITC
530 ± 15 nm (green), PI 585 ± 21 nm (orange).
Flow cytometry analyses were performed in the presence of Trypan Blue to ensure
extracellular fluorescence quenching. Cells were treated with peptides in the conditions
described above. Signal was then acquired a first time in cytometry. Cells were then
incubated with Trypan Blue (0.05% w/v in PBS) for 3 min before the second signal
acquisition. Finally, the same cells were permeabilized with 0.05% (v/v) Triton-X100 and
fluorescence was measured one last time by cytometry.
For live cell experiments, cells were cultured and treated with peptides as described
above. The coverslips were mounted directly onto slides using mounting medium and
visualized within the next 10 min.
Acknowledgment
This work was financially supported by the Natural Sciences and Engineering Research
Council of Canada (NSERC). H.L. acknowledges NSERC for a graduate fellowship. E.
Marsault is a member of the FRQNT-funded Proteo network and the FRQS-funded Réseau
Québécois de Recherche sur le Médicament (RQRM).
References
1.
Frankel, A. D. and Pabo, C. O. (1988) Cellular uptake of the tat protein from human
immunodeficiency virus Cell 55, 1189– 1193
296
2.
3.
4.
5.
6.
7.
8.
9.
10.
11.
12.
13.
14.
15.
16.
17.
Green, M. and Loewenstein, P. M. (1988) Autonomous functional domains of
chemically synthesized human immunodeficiency virus tat trans-activator protein
Cell 55, 1179– 1188
Rudolph, C., Plank, C., Lausier, J., Schillinger, U., Muller, R. H., and Rosenecker, J.
(2003) Oligomers of the arginine-rich motif of the HIV-1 TAT protein are capable of
transferring plasmid DNA into cells J. Biol. Chem. 278, 11411– 11418
Perez, F., Joliot, A., Bloch-Gallego, E., Zahraoui, A., Triller, A., and Prochiantz, A.
(1992) Antennapedia homeobox as a signal for the cellular internalization and nuclear
addressing of a small exogenous peptide J. Cell. Sci. 102 (Pt 4) 717– 722
Derossi, D., Chassaing, G., and Prochiantz, A. (1998) Trojan peptides: the penetratin
system for intracellular delivery Trends Cell. Biol. 8, 84– 87
Kondo, E., Saito, K., Tashiro, Y., Kamide, K., Uno, S., Furuya, T., Mashita, M.,
Nakajima, K., Tsumuraya, T., Kobayashi, N., and M 2012, Tumour lineage-homing
cell-penetrating peptides as anticancer molecular delivery systems Nat. Commun. 3,
951– 954
Kwon, E. J., Liong, S., and Pun, S. H. (2010) A truncated HGP peptide sequence that
retains endosomolytic activity and improves gene delivery efficiencies Mol.
Pharmacol. 7, 1260– 1265
Fonseca, S. B., Pereira, M. P., and Kelley, S. O. (2009) Recent advances in the use of
cell-penetrating peptides for medical and biological applications Adv. Drug Delivery
Rev. 61, 953– 964
Tung, C. H. and Weissleder, R. (2003) Arginine containing peptides as delivery
vectors Adv. Drug Delivery Rev. 55, 281– 294
Cerrato, C. P., Lehto, T., and Langel, U. (2014) Peptide-based vectors: recent
developments Biomol. Concepts 5, 479– 488
Copolovici, D. M., Langel, K., Eriste, E., and Langel, U. (2014) Cell-penetrating
peptides: design, synthesis, and applications ACS Nano 8, 1972– 1994
Wender, P. A., Mitchell, D. J., Pattabiraman, K., Pelkey, E. T., Steinman, L., and
Rothbard, J. B. (2000) The design, synthesis, and evaluation of molecules that enable
or enhance cellular uptake: peptoid molecular transporters Proc. Natl. Acad. Sci.
U.S.A. 97, 13003– 13008
Wender, P. A., Galliher, W. C., Goun, E. A., Jones, L. R., and Pillow, T. H. (2008)
The design of guanidinium-rich transporters and their internalization mechanisms
Adv. Drug Delivery Rev. 60, 452– 472
Mitchell, D. J., Kim, D. T., Steinman, L., Fathman, C. G., and Rothbard, J. B. (2000)
Polyarginine enters cells more efficiently than other polycationic homopolymers J.
Pept. Res. 56, 318– 325
Eiriksdottir, E., Mager, I., Lehto, T., El Andaloussi, S., and Langel, U. (2010)
Cellular internalization kinetics of (luciferin-) cell-penetrating peptide conjugates
Bioconjugate Chem. 21, 1662– 72
Madani, F., Lindberg, S., Langel, U., Futaki, S., and Graslund, A. (2011)
Mechanisms of cellular uptake of cell-penetrating peptides J. Biophys. 2011, 1– 10
Duchardt, F., Fotin-Mleczek, M., Schwarz, H., Fischer, R., and Brock, R. A. (2007)
comprehensive model for the cellular uptake of cationic cell-penetrating peptides
Traffic 8, 848– 866
297
18.
19.
20.
21.
22.
23.
24.
25.
26.
27.
28.
29.
30.
31.
Naik, R. J., Chandra, P., Mann, A., and Ganguli, M. (2011) Exogenous and cell
surface glycosaminoglycans alter DNA delivery efficiency of arginine and lysine
homopeptides in distinctly different ways J. Biol. Chem. 286, 18982– 18993
Wallbrecher, R., Verdurmen, W. P., Schmidt, S., Bovee-Geurts, P. H., Broecker, F.,
Reinhardt, A., van Kuppevelt, T. H., Seeberger, P. H., and Brock, R. (2014) The
stoichiometry of peptide-heparan sulfate binding as a determinant of uptake
efficiency of cell-penetrating peptides Cell. Mol. Life Sci. 71, 2717– 2729
Futaki, S., Suzuki, T., Ohashi, W., Yagami, T., Tanaka, S., Ueda, K., and Sugiura, Y.
(2001) Arginine-rich peptides. An abundant source of membrane-permeable peptides
having potential as carriers for intracellular protein delivery J. Biol. Chem. 276,
5836– 5840
Rydstrom, A., Deshayes, S., Konate, K., Crombez, L., Padari, K., Boukhaddaoui, H.,
Aldrian, G., Pooga, M., and Divita, G. (2011) Direct translocation as major cellular
uptake for CADY self-assembling peptide-based nanoparticles PLoS One 6, e25924
Fittipaldi, A., Ferrari, A., Zoppe, M., Arcangeli, C., Pellegrini, V., Beltram, F., and
Giacca, M. (2003) Cell membrane lipid rafts mediate caveolar endocytosis of HIV-1
Tat fusion proteins J. Biol. Chem. 278, 34141– 34149
Mager, I., Eiriksdottir, E., Langel, K., El Andaloussi, S., and Langel, U. (2010)
Assessing the uptake kinetics and internalization mechanisms of cell-penetrating
peptides using a quenched fluorescence assay Biochim. Biophys. Acta 1798, 338–
343
Vives, E. (2003) Cellular uptake [correction of uptake] of the Tat peptide: an
endocytosis mechanism following ionic interactions J. Mol. Recog. 16, 265– 271
Pae, J., Saalik, P., Liivamagi, L., Lubenets, D., Arukuusk, P., Langel, U., and Pooga,
M. (2014) Translocation of cell-penetrating peptides across the plasma membrane is
controlled by cholesterol and microenvironment created by membranous proteins J.
Controlled Release 192, 103– 113
Tsumuraya, T. and Matsushita, M. (2014) COPA and SLC4A4 are required for
cellular entry of arginine-rich peptides PLoS One 9, e86639
Elson-Schwab, L., Garner, O. B., Schuksz, M., Crawford, B. E., Esko, J. D., and Tor,
Y. (2007) Guanidinylated neomycin delivers large, bioactive cargo into cells through
a heparan sulfate-dependent pathway J. Biol. Chem. 282, 13585– 13591
Geisler, I. and Chmielewski, J. (2009) Cationic amphiphilic polyproline helices: sidechain variations and cell-specific internalization Chem. Biol. Drug Des. 73, 39– 45
Okuyama, M., Laman, H., Kingsbury, S. R., Visintin, C., Leo, E., Eward, K. L.,
Stoeber, K., Boshoff, C., Williams, G. H., and Selwood, D. L. (2007) Small-molecule
mimics of an alpha-helix for efficient transport of proteins into cells Nat. Meth. 4,
153– 159
Wender, P. A., Cooley, C. B., and Geihe, E. I. (2012) Beyond Cell Penetrating
Peptides: Designed Molecular Transporters Drug Discovery Today Technol. 9, e49–
e55
Lattig-Tunnemann, G., Prinz, M., Hoffmann, D., Behlke, J., Palm-Apergi, C.,
Morano, I., Herce, H. D., and Cardoso, M. C. (2011) Backbone rigidity and static
presentation of guanidinium groups increases cellular uptake of arginine-rich cellpenetrating peptides Nat. Commun. 2, 453– 458
298
32.
33.
34.
35.
36.
37.
38.
39.
40.
41.
42.
43.
44.
45.
Prochiantz, A. (2007) For protein transduction, chemistry can win over biology Nat.
Meth. 4, 119– 120
White, C. J. and Yudin, A. K. (2011) Contemporary strategies for peptide
macrocyclization Nat. Chem. 3, 509– 524
Hill, T. A., Shepherd, N. E., Diness, F., and Fairlie, D. P. (2014) Constraining cyclic
peptides to mimic protein structure motifs Angew. Chem., Int. Ed. 53, 13020– 13041
Delorbe, J. E., Clements, J. H., Whiddon, B. B., and Martin, S. F. (2010)
Thermodynamic and structural effects of macrocyclization as a constraining method
in protein-ligand interactions ACS Med. Chem. Lett. 1, 448– 452
Marsault, E. and Peterson, M. L. (2011) Macrocycles are great cycles: applications,
opportunities, and challenges of synthetic macrocycles in drug discovery J. Med.
Chem. 54, 1961– 2004
Qian, Z., Liu, T., Liu, Y. Y., Briesewitz, R., Barrios, A. M., Jhiang, S. M., and Pei, D.
(2013) Efficient delivery of cyclic peptides into mammalian cells with short sequence
motifs ACS Chem. Biol. 8, 423– 431
Mandal, D., Nasrolahi Shirazi, A., and Parang, K. (2011) Cell-penetrating homochiral
cyclic peptides as nuclear-targeting molecular transporters Angew. Chem., Int. Ed.
50, 9633– 9637
Hu, Y., Liu, X., Sinha, S. K., and Patel, S. (2014) Translocation thermodynamics of
linear and cyclic nonaarginine into model DPPC bilayer via coarse-grained molecular
dynamics simulation: implications of pore formation and nonadditivity J. Phys.
Chem. B 118, 2670– 2682
Nischan, N., Herce, H. D., Natale, F., Bohlke, N., Budisa, N., Cardoso, M. C., and
Hackenberger, C. P. (2014) Covalent attachment of cyclic TAT peptides to GFP
results in protein delivery into live cells with immediate bioavailability Angew.
Chem., Int. Ed. 53, 1– 5
Oh, D., Nasrolahi Shirazi, A., Northup, K., Sullivan, B., Tiwari, R. K., Bisoffi, M.,
and Parang, K. (2014) Enhanced cellular uptake of short polyarginine peptides
through fatty acylation and cyclization Mol. Pharmacol. 11, 2845– 2854
Qian, Z., LaRochelle, J. R., Jiang, B., Lian, W., Hard, R. L., Selner, N. G.,
Luechapanichkul, R., Barrios, A. M., and Pei, D. (2014) Early endosomal escape of a
cyclic cell-penetrating peptide allows effective cytosolic cargo delivery Biochemistry
53, 4034– 4046
Cascales, L., Henriques, S. T., Kerr, M. C., Huang, Y. H., Sweet, M. J., Daly, N. L.,
and Craik, D. J. (2011) Identification and characterization of a new family of cellpenetrating peptides: cyclic cell-penetrating peptides J. Biol. Chem. 286, 36932–
36943
Wallbrecher, R., Depre, L., Verdurmen, W. P., Bovee-Geurts, P. H., van Duinkerken,
R. H., Zekveld, M. J., Timmerman, P., and Brock, R. (2014) Exploration of the
design principles of a cell-penetrating bicylic peptide scaffold Bioconjugate Chem.
25, 955– 964
Duchardt, F., Ruttekolk, I. R., Verdurmen, W. P., Lortat-Jacob, H., Burck, J.,
Hufnagel, H., Fischer, R., van den Heuvel, M., Lowik, D. W., and Vuister, G. W.
2009, Cell-penetrating peptide derived from human lactoferrin with conformationdependent uptake efficiency J. Biol. Chem. 284, 36099– 36108
299
46.
Verdurmen, W. P., Bovee-Geurts, P. H., Wadhwani, P., Ulrich, A. S., Hallbrink, M.,
van Kuppevelt, T. H., and Brock, R. (2011) Preferential uptake of L- versus D-amino
acid cell-penetrating peptides in a cell type-dependent manner Chem. Biol. 18, 1000–
1010
47. Hed, J., Hallden, G., Johansson, S. G., and Larsson, P. (1987) The use of fluorescence
quenching in flow cytofluorometry to measure the attachment and ingestion phases in
phagocytosis in peripheral blood without prior cell separation J. Immunol. Methods
101, 119– 125
48. Oh, D., Darwish, S. A., Shirazi, A. N., Tiwari, R. K., and Parang, K. (2014)
Amphiphilic bicyclic peptides as cellular delivery agents ChemMedChem 9, 2449–
2453
49. Richard, J. P., Melikov, K., Vives, E., Ramos, C., Verbeure, B., Gait, M. J.,
Chernomordik, L. V., and Lebleu, B. (2003) Cell-penetrating peptides. A
reevaluation of the mechanism of cellular uptake J. Biol. Chem. 278, 585– 590
50. Rothbard, J. B., Kreider, E., VanDeusen, C. L., Wright, L., Wylie, B. L., and Wender,
P. A. (2002) Arginine-rich molecular transporters for drug delivery: role of backbone
spacing in cellular uptake J. Med. Chem. 45, 3612– 3618
51. Julien, M. H. A., Mauras, A., Puget, K., Amblard, M., Martinez, J., and Subra, G.
(2009) N-terminus FITC labeling of peptides on solid support: the truth behind the
spacer Tetrahedron Lett. 50, 260– 263
300