The effect of Irvingia gabonensis seeds on body weight and

Transcription

The effect of Irvingia gabonensis seeds on body weight and
Lipids in Health and Disease
BioMed Central
Open Access
Research
The effect of Irvingia gabonensis seeds on body weight and blood
lipids of obese subjects in Cameroon
Judith L Ngondi, Julius E Oben* and Samuel R Minka
Address: Nutrition, HIV and Health Research Unit, Department of Biochemistry, P.O Box 812, Faculty of Science, University of Yaounde I,
Cameroon
Email: Judith L Ngondi - [email protected]; Julius E Oben* - [email protected]; Samuel R Minka - [email protected]
* Corresponding author
Published: 25 May 2005
Lipids in Health and Disease 2005, 4:12
doi:10.1186/1476-511X-4-12
Received: 26 February 2005
Accepted: 25 May 2005
This article is available from: http://www.lipidworld.com/content/4/1/12
© 2005 Ngondi et al; licensee BioMed Central Ltd.
This is an Open Access article distributed under the terms of the Creative Commons Attribution License (http://creativecommons.org/licenses/by/2.0),
which permits unrestricted use, distribution, and reproduction in any medium, provided the original work is properly cited.
Abstract
Dietary fibres are frequently used for the treatment of obesity. The aim of this study was to
evaluate the efficacy of Irvingia gabonensis seeds in the management of obesity. This was carried out
as a double blind randomised study involving 40 subjects (mean age 42.4 years). Twenty-eight
subjects received Irvingia gabonensis (IG) (1.05 g three time a day for one month) while 12 were on
placebo (P) and the same schedule. During the one-month study period all subjects were on a
normocaloric diet evaluated every week by a dietetic record book. At the end, the mean body
weight of the IG group was decreased by 5.26 ± 2.37% (p < 0.0001) and that of the placebo group
by 1.32 ± 0.41% (p < 0.02). The difference observed between the IG and the placebo groups was
significant (p < 0.01). The obese patients under Irvingia gabonensis treatment also had a significant
decrease of total cholesterol, LDL-cholesterol, triglycerides, and an increase of HDL-cholesterol.
On the other hand, the placebo group did not manifest any changes in blood lipid components.
Irvingia gabonensis seed may find application in weight lose.
Introduction
Obesity is of major primary care concern and is targeted as
an international health objective in Healthy 2000, which
seeks to reduce the prevalence of obesity to less than 20%.
In the last 10 years, the number of overweight people has
increased from 26 to 34% [1]. Conventional dietary and
behavioral treatment have failed in long-term management. Dietary strategies used to manage obesity include
the use of high fibre, low carbohydrates and fats diet [2,3].
Beneficial effect of dietary fibre in the management of
obesity is not well established, since their mechanism of
action is not known. The discovery of new medicinal
plants has led to the creation of potential drugs that modify feeding behavior and metabolism and may therefore
have application in weight management. The aim of the
present study is to investigate the effect of Irvingia gabonensis extract on body weight.
Subjects and methods
A total of 40 obese subjects aged between 19 and 55 years
were selected from a group responding to a radio advertisement. After physical examination and laboratory
screening tests, diabetics, pregnant and lactating women
were excluded. None of these subjects took any weight
reducing medication and none was following any specific
diet. The purpose, nature and potential risks of the study
were explained to all patients and a written informed consent was obtained before their participation. The local
research ethics committee approved the experimental
protocol.
Page 1 of 4
(page number not for citation purposes)
Lipids in Health and Disease 2005, 4:12
http://www.lipidworld.com/content/4/1/12
Table 1: Effect of Irvingia gabonensis crude extract on body weight, body fat, waist and hip circumferences
Treatment period (weeks)
Weight (kg
Active
Placebo
Active
Placebo
Active
Placebo
Active
Placebo
Body fat (%)
Waist (cm)
Hip (cm)
0
2
4
105.10 ± 16.98
79.43 ± 9.83
46.11 ± 4.4848
40.58 ± 3.49
112.76 ± 20.5
81.1 ± 7.1
125.69 ± 11.34
122.2 ± 10.7
102.3 ± 17.06
79.43 ± 9.83
46.5 ± 3.68
40.58 ± 3.9
109.7 ± 20.4
81,91 ± 7,91
122.92 ± 10.67
122.2 ± 10.7
101.01 ± 16.63
79.33 ± 10.63
45.34 ± 3.52
40.3 ± 3.8
106.6 ± 20.79
81.25 ± 7.52
121.15 ± 10.39
121.5 ± 10.9
Table 2: Effect Irvingia gabonensis on systolic (SBP) and diastolic (DBP) blood pressure
Treatment period (weeks)
0
SBP (mmHg)
Active
Placebo
Active
placebo
DBP (mmHg)
2
136.41 ± 19.57
134 ± 5.05
98.5 ± 19.52
93.50 ± 10.31
4
132.66 ± 18.48*
121.5 ± 5.89
97.5 ± 22.80
93.83 ± 7.41
132.83 ± 17.97*
123.83 ± 2.92
94.08 ± 11.07
91.5 ± 6.53
Values are means ± sem. Significant differences were at *P < 0.001 by comparison to the placebo group.
Table 3: The effect of Irvingia gabonensis on blood total cholesterol (TC), triglyceride (TRI), high density lipoprotein cholesterol
(HDL-c), low density lipoprotein cholesterol (LDL-c) and glucose.
Active
Placebo
Initial
Final
Initial
Final
T-cholesterol
TRI
HDL-c
LDL-c
LDL/HDL
T-cho/HDL
GLUCOSE
215 ± 55.12
130.68 ± 39.5
163.70 ±
25.32
158.36 ±
30.46
162 ± 33.15
89.22 ± 55.63
130.65 ±
37.82
100.52 ±
32.55
61.23 ± 20.36
89.9 ± 28.44
31.38 ± 25.21
121.37 ± 36.3
66.08 ± 34.27
105.06 ±
11.86
98.55 ± 27.99
1.98 ± 1.78
0.735 ± 1.20
5.05 ± 3.94
3.51 ± 2.70
1.45 ± 1.38
6.44 ± 3.37
3.8 ± 1.92
2.57 ± 1.03
3.6 ± 0.41
3.19 ± 1.85
4.51 ± 2.07
3.9 ± 0.74
41.20 ± 19.53
Study design
The study was as a randomised, double blind placebocontrolled crossover design, and consisted of a 4-week
treatment period. Subjects were given two different types
of capsules containing 350 mg of Irvingia gabonensis seed
extract (active formulation) or oat bran (placebo). Three
capsules were taken three times daily, one-half hour
before meals (a total daily amount of 3.15 g of Irvingia
gabonensis seed extract) with a glass of warm water. Capsules were identical in shape, colour and appearance, with
neither patients nor researchers knowing what capsule
they received. During the experimental period, subjects
were examined weekly, with their body weight, body fat,
waist and hip circumferences recorded each time. Subjective findings such as increased or decreased appetite, feeling of lightness and gastrointestinal pains were
individually noted. Side effects of the active extract, if any
were also solicited and noted during each visit. The subjects were also interviewed about their physical activity
and food intake during the trial, and were instructed to eat
a low fat diet (1800 Kcal) as well as keep a record for seven
consecutive days (using household measurements).
Page 2 of 4
(page number not for citation purposes)
Lipids in Health and Disease 2005, 4:12
Anthropometric measurements
Anthropometric measurements were done at each visit,
with body weight and body fat (impedance measurement
using a TANITA™ monitor Scale) measurements on fasted
(12 hour) subjects wearing light clothing. Waist and hip
circumferences were measured by soft non-stretchable
plastic tape on the narrowest and the widest parts of the
trunk.
Laboratory methods
Blood samples were collected after a 12 h overnight fast
into heparinized tubes at the beginning of the study, after
two weeks and at the end (4 weeks) of treatment. The concentrations of total cholesterol, triacylglycerol, HDL-cholesterol, and glucose, in plasma were measured using a
commercial diagnostic kit (Cholesterol infinity, triglycerides Int, EZ HDL™ cholesterol, EZ LDL™ cholesterol, Glucose Trinder, respectively) from SIGMA Diagnostics
Statistical Analysis
Results are expressed as mean ± SEM except for anthropometric measurements. Paired Student's t-test was carried
out on the start and end values of placebo and Irvingia
gabonensis capsules and also on the differences between
the placebo and Irvingia gabonensis crude extract.
Results
Effect on body composition
Irvingia gabonensis induced a decrease in weight of 2.91 ±
1.48% (p < 0.0001) after two weeks and 5.6 ± 2.7% (p <
0,0001) after one month. Although the percentage of
body fat was not significantly reduced with both placebo
and IG, the waist circumference (5.07 ± 3.18%; p <
0.0001) and hip circumference (3.42 ± 2.12%; p <
0,0001) were significantly reduced by IG. A reduction of
1.32 ± 0.41% (p < 0.02) and 2.23 ± 1.05% (p < 0.05) was
observed with the placebo after two and four weeks
respectively of treatment.
Effect on blood pressure
From the second week of experimentation, the systolic
blood pressure was significantly reduced by the active
extract (Table 2).
http://www.lipidworld.com/content/4/1/12
Discussion
The soluble fibre of the seed of Irvingia gabonensis like
other forms of water-soluble dietary fibres, are "bulkforming" laxatives. Irvingia gabonensis seeds delay stomach
emptying, leading to a more gradual absorption of dietary
sugar. This effect can reduce the elevation of blood sugar
levels that is typical after a meal [4]. Controlled studies
have found that after-meal blood sugar levels are lower in
people with diabetes given glucomannan in their food [5]
and overall diabetic control is improved with soluble
fibre-enriched diets according to preliminary [6] and controlled [7,8] trials. One double-blind study reported that
glucomannan (8–13 grams per day) stabilized blood
sugar levels in people with the insulin resistance syndrome [9]. Like other soluble fibers, Irvingia gabonensis
seed fibre can bind to bile acids in the gut and carry them
out of the body in the faeces, which requires the body to
convert more cholesterol into bile acids [10]. This can
result in the lowering of blood cholesterol as well as other
blood lipids. Controlled double-blind [11,12] studies
have shown that supplementation with several grams per
day of soluble fibre significantly reduced total blood cholesterol, LDL cholesterol, and triglycerides and in some
cases raised HDL cholesterol, these being comparable
with effects noticed with Irvingia gabonensis.
Considering the wide use of Irvingia gabonensis in the
preparation of various dishes in Cameroon, its use should
be further encouraged for the purposes of control of dietary lipids as well as for weight reduction.
References
1.
2.
3.
4.
5.
Effect on blood lipids components
The plasma total cholesterol cencentration was reduced
by 39.21%, triglyerides by 44.9% (p < 0,05) and LDL by
45.58%. This was accompanied by a significant increase
in HDL-cholesterol of 46.852%. The CT/HDL ratio (p <
0.05) and the blood glucose level were also reduced
(32.36%; p < 0.05). No significant change was observed in
the placebo group.
6.
7.
8.
9.
10.
Bray GA: Complications of obesity.
Ann Int Med 1985,
103:1052-1062.
Harris MI: Epidemiological correlates of NIDDM in Hispanics,
whites, and blacks in the U.S. population. Diabetes Care 1991,
14(suppl 3):639-648.
Weintraub M: Long-term weight control study: conclusions.
Clinic Pharmacol Ther 1992:581. Passaretti S, Franzoni M, Comin U, et
al. Action of glucomannans on complaints in patients affected with
chronic constipation: a multicentric clinical evaluation. Ital J Gastroenterol 1991;23:421-5
Vuksan V, Jenkins DJ, Spadafora P, et al.: Konjac-mannan (glucomannan) improves glycemia and other associated risk factors for coronary heart disease in type 2 diabetes. A
randomized controlled metabolic trial. Diabetes Care 1999,
22:913-9.
Cesa F, Mariani S, Fava A, et al.: The use of vegetable fibers in the
treatment of pregnancy diabetes and/or excessive weight
gain during pregnancy. Minerva Ginecol 1990, 42:271-4. [in Italian]
Vorster HH, Lotter AP, Odendaal I, et al.: Benefits from supplementation of the current recommended diabetic diet with
gel fibre. Int Clin Nutr Rev 1988, 8:140-6.
Hopman WP, Houben PG, Speth PA, Lamers CB: Glucomannan
prevents postprandial hypoglycaemia in patients with previous gastric surgery. Gut 1988, 29:930-4.
Doi K: Effect of konjac fibre (glucomannan) on glucose and
lipids. Eur J Clin Nutr 1995, 49(Suppl 3):S190-7. [review]
Wu J, Peng SS: Comparison of hypolipidemic effect of refined
konjac meal with several common dietary fibers and their
mechanisms of action. Biomed Environ Sci 1997, 10:27-37.
Arvill A, Bodin L: Effect of short-term ingestion of konjac glucomannan on serum cholesterol in healthy men. Am J Clin Nutr
1995, 61:585-9.
Page 3 of 4
(page number not for citation purposes)
Lipids in Health and Disease 2005, 4:12
11.
12.
http://www.lipidworld.com/content/4/1/12
Walsh DE, Yaghoubian V, Behforooz A: Effect of glucomannan on
obese patients: a clinical study. Int J Obes 1984, 8:289-93.
Vido L, Facchin P, Antonello I, et al.: Childhood obesity treatment: double blinded trial on dietary fibres (glucomannan)
versus placebo. Padiatr Padol 1993, 28:133-6.
Publish with Bio Med Central and every
scientist can read your work free of charge
"BioMed Central will be the most significant development for
disseminating the results of biomedical researc h in our lifetime."
Sir Paul Nurse, Cancer Research UK
Your research papers will be:
available free of charge to the entire biomedical community
peer reviewed and published immediately upon acceptance
cited in PubMed and archived on PubMed Central
yours — you keep the copyright
BioMedcentral
Submit your manuscript here:
http://www.biomedcentral.com/info/publishing_adv.asp
Page 4 of 4
(page number not for citation purposes)
Lipids in Health and Disease
BioMed Central
Open Access
Research
IGOB131, a novel seed extract of the West African plant Irvingia
gabonensis, significantly reduces body weight and improves
metabolic parameters in overweight humans in a randomized
double-blind placebo controlled investigation
Judith L Ngondi1, Blanche C Etoundi1, Christine B Nyangono1,
Carl MF Mbofung2 and Julius E Oben*1
Address: 1Laboratory of Nutrition and Nutritional Biochemistry, Faculty of Science, University of Yaounde I, Yaounde, Cameroon and
2Department of Food Science and Nutrition, ENSAI, University of Ngaoundere, BP 686 Ngaoundere, Cameroon
Email: Judith L Ngondi - [email protected]; Blanche C Etoundi - [email protected]; Christine B Nyangono - [email protected];
Carl MF Mbofung - [email protected]; Julius E Oben* - [email protected]
* Corresponding author
Published: 2 March 2009
Lipids in Health and Disease 2009, 8:7
doi:10.1186/1476-511X-8-7
Received: 7 January 2009
Accepted: 2 March 2009
This article is available from: http://www.lipidworld.com/content/8/1/7
© 2009 Ngondi et al; licensee BioMed Central Ltd.
This is an Open Access article distributed under the terms of the Creative Commons Attribution License (http://creativecommons.org/licenses/by/2.0),
which permits unrestricted use, distribution, and reproduction in any medium, provided the original work is properly cited.
Abstract
Background: A recent in vitro study indicates that IGOB131, a novel seed extract of the traditional West
African food plant Irvingia gabonensis, favorably impacts adipogenesis through a variety of critical metabolic
pathways including PPAR gamma, leptin, adiponectin, and glycerol-3 phosphate dehydrogenase. This study
was therefore aimed at evaluating the effects of IGOB131, an extract of Irvingia gabonensis, on body weight
and associated metabolic parameters in overweight human volunteers.
Methods: The study participants comprised of 102 healthy, overweight and/or obese volunteers (defined
as BMI > 25 kg/m2) randomly divided into two groups. The groups received on a daily basis, either 150 mg
of IGOB131 or matching placebo in a double blinded fashion, 30–60 minutes before lunch and dinner. At
baseline, 4, 8 and 10 weeks of the study, subjects were evaluated for changes in anthropometrics and
metabolic parameters to include fasting lipids, blood glucose, C-reactive protein, adiponectin, and leptin.
Results: Significant improvements in body weight, body fat, and waist circumference as well as plasma
total cholesterol, LDL cholesterol, blood glucose, C-reactive protein, adiponectin and leptin levels were
observed in the IGOB131 group compared with the placebo group.
Conclusion: Irvingia gabonensis administered 150 mg twice daily before meals to overweight and/or obese
human volunteers favorably impacts body weight and a variety of parameters characteristic of the
metabolic syndrome. This is the first double blind randomized placebo controlled clinical trial regarding
the anti-obesity and lipid profile modulating effects of an Irvingia gabonensis extract. The positive clinical
results, together with our previously published mechanisms of gene expression modulation related to key
metabolic pathways in lipid metabolism, provide impetus for much larger clinical studies. Irvingia gabonensis
extract may prove to be a useful tool in dealing with the emerging global epidemics of obesity,
hyperlipidemia, insulin resistance, and their co-morbid conditions.
Trial registration: ClinicalTrials.gov NCT00645775
Page 1 of 7
(page number not for citation purposes)
Lipids in Health and Disease 2009, 8:7
Background
Excess adipose tissue increases the workload of the cardiovascular system, adversely alters immune function, and
dramatically increases the risk of heart disease, non-insulin-dependent diabetes mellitus, obstructive pulmonary
disease, arthritis and a variety of cancers [1]. Although
once considered a major health problem isolated to developed countries, obesity is now recognized as a global
problem with potentially catastrophic consequences for
health economics. In Cameroon, for example, the
reported incidence of obesity in urban areas ranges from
17–21% [2]. This dramatic change reflects the social,
nutritional, and lifestyle changes involved in rural-urban
migration and the urbanization of rural areas. Worthy of
note in this context are the changes from traditional high
fibre diets and high-activity lifestyles to diets rich in saturated fat and low in fibre, further compounded by reduction in physical activity levels [3,4]. Insulin resistance,
caused by the concomitant, complex interaction of environment (e.g. diet, activity level, elevated body mass,
excess visceral adiposity) and genetic factors, is a fundamental pathophysiologic cause of the metabolic syndrome, a recognized risk factor for cardiovascular disease
[5,6]. Like many other obesity-related conditions, the
incidence of the metabolic syndrome is also on the rise in
Cameroon [7].
Nutritional management, especially caloric restriction for
the purpose of weight reduction, can improve insulin
action on target tissues like skeletal muscle, hepatocytes,
and peripheral and visceral adipocytes [8,9].
The consumption of Irvingia gabonensis, a fleshy West African fruit, is common in traditional Nigerian and Cameroonian cuisine. Initial observations suggested beneficial
changes in metabolic parameters were associated with the
high fiber content of Irvingia gabonensis [10]. Results of
subsequent analysis suggested that the earlier observations of the beneficial effects of Irvingia gabonensis could
not be accounted for by fiber content alone [11]. A recent
in vitro study using a validated experimental model
(murine 3T3-L1 adipocytes) provides compelling data
that Irvingia gabonensis seed extract may inhibit adipogenesis through modulation of PPAR gamma and glycerol-3
phosphate dehydrogenase in addition to beneficial
impact upon leptin and adiponectin [12].
The current clinical study was designed to evaluate if the
in vitro data could translate into in vivo clinical efficacy in
overweight and/or obese human volunteers.
Participants and methods
Study population
Participants for the study were recruited from the city of
Yaoundé, Cameroon and surrounding metro region
http://www.lipidworld.com/content/8/1/7
through radio and print media advertisement. Inclusion
criteria for study participation mandated: (1) men and
women ages 19 to 50 years, in general good health free
from significant medical illness that, in the opinion of the
investigator, could adversely compromise study participation for any reason; (2) stable body weight (+/- 2 kg) for
at least three months prior to study randomization without use of medication known or suspected to affect body
weight or appetite; (3) no concomitant or recent (within
the past three months) bacterial or seasonal viral infection
(e.g. influenza); (4) BMI between 26 kg/m2 and 40 kg/m2;
(5) no attempts at weight loss through dietary intervention over the three months prior to trial randomization;
(6) no participation in a structured weight control program for at least three months prior to study randomization; (7) non-smoker; (8) non-drinkers of sugary fizzy
drinks; (9) capability and willingness for multiple blood
sampling procedures; and (10) ability to competently
understand and sign the consent form.
Based on the above criteria, 120 consenting volunteers
(62 males and 58 females) were selected to participate in
the study. The protocol used was approved by the Cameroon National Ethics Committee; the study was conducted in accord with the Helsinki Declaration (1983
version).
Study design/intervention
The study was a randomized, double-blind, placebo-controlled design for 10 weeks. The volunteers were randomly divided into two groups – placebo or IGOB131.
The participants consumed either one capsule of placebo
or one capsule of IGOB131 (containing 150 mg of Irvingia
gabonensis extract) 30–60 minutes before lunch and dinner (BID) throughout the study period.
Test materials
All test materials were supplied by Gateway Health Alliances, Inc. (Fairfield, CA, USA) in individual packets of
capsules. The identical-looking placebo and active formulation capsules contained, respectively, maize-based powder consisting of 150 mg maltodextrin, or 150 mg
IGOB131.
Anthropometric measurements
Height was measured with a locally manufactured wallmounted stadiometer, which was calibrated against the
Cameroon's Department of National Security identification scale. Body weight and percent body fat, were
assessed using a Tanita™ BC-418 Segmental Body Composition Analyzer/Scale that uses bio-electrical impedance
analysis for body composition analysis. Body mass index
(BMI) was calculated using the weight and height measurements. Waist circumference measurements to the nearest 0.1 cm were taken at the mid-point between the
Page 2 of 7
(page number not for citation purposes)
Lipids in Health and Disease 2009, 8:7
bottom rib and the hip bone, without restrictive garments
using a flexible non expandable tape measure.
Dietary Intake
Three days before the collection of baseline measurements and three times during the trial, dietary intake of
subjects was evaluated by a qualified dietician. Energy and
protein intakes were calculated using standard food composition tables based upon the 3-day food intake record.
No major dietary intervention or formal physical activity
program was instituted during the course of the study;
participants were instructed to maintain the current level
of physical activity prior to study randomization during
the course of the study.
Sample collection
Fasting blood samples (5 ml of blood) were collected at
baseline, and at 4, 8, and 10 weeks. Serum obtained from
each blood sample was split into multiple 500 μl aliquots
and stored at -20°C until needed for the measurement of
total cholesterol, LDL cholesterol, fasting blood glucose,
C-reactive protein, adiponectin, and leptin levels.
Analytical methods
Determination of glucose in blood using glucose oxidase
with an alternative oxygen receptor was adopted for this
study [13]. Serum total cholesterol was assayed by the
cholesterol oxidase method [14] while triglycerides as
well as serum glucose levels were assayed following the
method described by Buccolo & David [15]. Quantitative
determinations of C-reactive protein (CRP) was assessed
using a high sensitivity immunoassay (Oxis International,
Foster City, CA. USA) while serum leptin was determined
in duplicate using an enzyme-linked immunosorbent
assay (ELISA) (Diagnostic Systems Laboratory, Webster,
TX. USA) technique within the same assay, with an intraassay variance of 3.2%. Serum adiponectin was measured
in duplicate using an enzyme immunoassay (APLCO
Diagnostics, Salem, NH. USA) within the same assay.
Intra-assay variance was ≤ 5%.
Statistical analysis
Statistical analyses (Student's t-test and ANOVA) were performed using the Statistical Package for the Social Sciences
(SPSS) software. Graphical representations of relative
changes in measured variables were performed using
Sigma Plot statistical package.
Results
The 120 volunteers (mean age 34, range 19–50) were
divided into 2 groups, with 60 randomly assigned to placebo and 60 to IGOB131 (active). Of these initial 120 volunteers, 102 completed the 10 week study; 50 from the
placebo group, and 52 from the IGOB131 group. Eighteen
http://www.lipidworld.com/content/8/1/7
volunteers [placebo (n = 12) and IGOB131 (n = 6)]
dropped out of the study for the following reasons – not
experiencing rapid weight loss (n = 10); influenza attack
(n = 3); dryness of mouth (n = 3); no reason given (n = 2).
Food Intake
The mean daily energy intake in the IGOB131 group was
2767 ± 187 kcal of which 56% was from carbohydrate,
29% from protein, and 15% from fat. In the placebo
group, the mean daily energy intake was 3156 ± 185 kcal
of which 56% was from carbohydrate, 29% was from protein, and 15% was from fat. Seven-day dietary and activity
assessment of the subjects at baseline showed similar food
intake habits and energy levels.
Body weight, waist size and body fat
Baseline characteristics of the two groups were wellmatched and without significant differences at baseline
(Table 1). There were no significant differences in the
baseline body weight, waist circumference and serum leptin measurements between the placebo and experimental
group (Table 1). However, by the tenth week, significant
differences were observed between the placebo and experimental intervention groups, respectively, for body weight
(95.7 kg vs. 85.1 kg, respectively, p < 0.01), waist circumference (101.1 cm vs. 88.1 cm, respectively, p < 0.05).
Body fat decreased over time in both groups but the experimental group lost significantly more body fat (6.3%, p <
0.05) compared to the placebo group (1.9%) (Table 1).
When corrected for placebo response, the pattern of relative changes in weight and waist circumference was found
to be different between the two groups, consistent with a
difference in response to the intake of the extract (Figure
1).
Total and LDL cholesterol, C-reactive proteins, leptin
These variables decreased from baseline, though at different rates and magnitudes associated with duration of the
study, with the experimental group showing statistically
significant changes compared with the placebo group at
week-10 (Table 1).
While baseline levels of serum lipids were similar in the
two groups, significant differences were observed between
the two as the study progressed with the experimental
group showing progressively greater improvement. At
week-10, significant differences were observed for total
cholesterol (placebo: 142.5 mg/dl vs. IGOB131: 111.9
mg/dl, p < 0.05) and LDL cholesterol (placebo: 77.7 mg/
dl vs IGO131: 59.77 mg/dl, p < 0.01). Compared to baseline values, total cholesterol decreased by 1.9% in the placebo group as opposed to 26.2% for the IGOB131 group
while LDL cholesterol levels fell by 4.8% in the placebo
compared to 27.3% in the IGOB131 group. Correcting for
Page 3 of 7
(page number not for citation purposes)
Lipids in Health and Disease 2009, 8:7
http://www.lipidworld.com/content/8/1/7
Table 1: Changes* in body weight, waist, leptin, fat, total cholesterol, LDL cholesterol, glucose and adiponectin measurements in the
course of the study.
Time (weeks)
Variables
Group
Base line
4
8
10
Over change
Weight (Kg)
Placebo
IGOB131
96.4 ± 12.3
97.9 ± 9.1
95.8 ± 8.2
94.3 ± 5.5 a
95.1 ± 10.6
89.7 ± 4.7 a
95.7 ± 15.2
85.1 ± 3.1b
-0.7
-12.8
Waist (cm)
Placebo
IGOB131
106.4 ± 10.8
105.1 ± 6.3
102.7 ± 10.6
98.0 ± 8.4a
101.1 ± 14.8
90.1 ± 8.70a
101.1 ± 15.8b
88.1 ± 7.6
-5.3
-16.19
Leptin (ng/ml)
Placebo
IGOB131
31.3 ± 1.8
32.9 ± 1.6
29.4 ± 1.4
18.1 ± 1.3a
28.1 ± 1.8
16.8 ± 1.2a
28.4 ± 1.8
16.9 ± 1.3b
-2.9
-16.0
Fat (%)
Placebo
IGOB131
34.7 ± 8.6
34.2 ± 7.9
33.31 ± 10.9
31.6 ± 5.4a
32.9 ± 11.9
28.2 ± 6.6a
32.7 ± 15.7
27.9 ± 5.5a
-1.99
-6.3
LDL cholesterol (mg/dL)
Placebo
IGOB131
77.4 ± 9.2
82.2 ± 8.1
76.5 ± 8.9
71.8 ± 5.9b
74.1 ± 9.3
64.7 ± 8.9b
73.7 ± 8.3
59.8 ± 5.0b
-3.75
-22.44
Total cholesterol (mg/dL)
Placebo
IGOB131
145.2 ± 22.4
151.7 ± 18.5
143.9 ± 12.2
133.7 ± 16.6a
142.1 ± 13.6
120.1 ± 11.9 a
142.4 ± 12.1
111.9 ± 5.8a
-2.8
-39.8
C-reactive protein (mg/L)
Placebo
IGOB131
1.46 ± 0.05
1.49 ± 0.041
1.46 ± 0.04
0.91 ± 0.05a
1.445 ± 0.03
0.71 ± 0.04b
1.455 ± 0.05
0.72 ± 0.05b
-0.01
-0.78
81.4 ± 9.6
85.55 ± 5.59
79.5 ± 10.1
76.6 ± 10.3a
77.8 ± 9.4
68.7 ± 9.3a
77.1 ± 7.8
66.3 ± 4.9a
-4.3
-19.3
12.1 ± 3.2
12.2 ± 3.0
15.2 ± 3.5
24.8 ± 3.8a
14.6 ± 3.5
31.0 ± 4.0b
14.94 ± 3.9
31.6 ± 3.9a
+ 2.8
+ 19.4
Glucose (mg/dL)
Adiponectin (mg/L)
aP
Placebo
IGOB131
Placebo
IGOB131
< 0.05; b P < 0.01 compared with Placebo
placebo values, the relative change in the total cholesterol
and LDL cholesterol was observed to follow a similar pattern (Figure 2) in the experimental group, suggesting a
similar response mechanism to IGOB131 intake.
Like in the case of the other parameters measured, C-reactive protein and leptin levels decreased over time in both
groups. The rate of decrease was however accelerated in
the IGOB131 relative to the placebo-group. Serum levels
of CRP fell by 1.2% in the placebo group as opposed to
52.0% in the experimental group relative to baseline. On
the other hand, leptin levels decreased by 9.3% in the placebo group compared to a 48.6% in the IGOB131 group
over the 10-week experimental period.
Fasting blood glucose levels
Blood glucose levels in the experimental group (85.6 mg/
dl ± 5.6 mg/dl) and in the placebo group (81.4 ± 9.6 mg/
dl) were similar at baseline but decreased to significantly
different levels (P < 0.05) at week-10 of the study (Table
1). In relative terms, decreases in placebo and treatment
groups were 5.3% vs. 22.5%, respectively (Figure 3). Corrected for the placebo values, the changes in blood glucose levels were similar to that of lipids (Figure 2).
Adiponectin levels
Baseline serum adiponectin levels were comparable
between the two groups (placebo: 12.1 mg/l ± 3.21 mg/l;
IGOB131: 12.16 mg/l ± 3.04 mg/l), but increased with
time (Table 1). By week-10 of the study adiponectin levels
in the placebo group increased by 23.4% compared to
159.8% in the experimental group. Corrected for placebo
values, the rate of increase in serum adiponectin levels in
IGOB131 group followed an exponential curve (Figure 4).
Adverse events
Treatment of overweight and obese subjects with
IGOB131 was well-tolerated. Adverse events with an incidence >n = 3 included headache (n = 5), sleep difficulty (n
= 6), and intestinal flatulence (n = 6). The incidence of all
reported side effects was similar in the placebo group as
well as in the treatment group.
Page 4 of 7
(page number not for citation purposes)
Lipids in Health and Disease 2009, 8:7
http://www.lipidworld.com/content/8/1/7
suggests a favorable impact upon adipogenesis associated
with IGOB131.
102
Weight
Waist size
Variation of baseline(%)
100
Insulin resistance is the hallmark of the metabolic syndrome and is strongly associated with excess adiposity
[16]. A variety of adipocyte-derived biologically active
molecules have been identified, including leptin, resistin,
TNF-α, and IL-6, that may contribute to obesity-linked
metabolic abnormalities [17]. Since plasma leptin levels
are closely correlated with the level of adipose tissue [18],
the decreases in plasma leptin level associated with
IGOB131 treatment may be attributable to the decrease of
adipose tissue induced as a consequence of weight loss.
98
96
94
92
90
88
86
0
2
4
6
8
10
Time (weeks)
Figurein1in
Changes
ments
IGOB131
body weight
groupand
corrected
waist circumference
for placebo values
measureChanges in body weight and waist circumference
measurements in IGOB131 group corrected for placebo values.
Discussion
The present study suggests that IGOB131, a seed extract of
Irvingia gabonensis, safely and significantly reduces body
weight in overweight and/or obese subjects, and has a
favorable impact upon a variety of other metabolic
parameters. The current clinical study shows IGOB131
administration is associated with increases in plasma adiponectin levels and decreases in leptin and CRP levels in
comparison with the placebo group. These observations
support the results of a recent in vitro study [12] which
Body fat
Total cholesterol
LDL Cholesterol
Fasting blood glucose
Leptin
C-reactive protein
100
90
% change
80
y
y0 ae bx
70
According to McTernan et al. [17], adipose tissue plays a
prominent role in the clinical expression of metabolic
syndrome, most likely mediated by the increased release
and peripheral tissue action of non-esterified fatty acids
and by the dysregulated production of adipocyte-secreted
proteins, including leptin, adiponectin, resistin, TNF-α,
and IL-6. Adiponectin, known to correlate with endothelial function and vascular health, is exclusively expressed
in adipose tissue and abundant in human plasma, and
appears to be decreased in individuals with obesity and
type 2 diabetes, risk factors for atherosclerosis[19].
The observation of a significant weight loss, as well as an
increase in adiponectin is consistent with earlier reports in
the literature observing an association between weight
loss and an increase in adiponectin levels [20].
IGOB131 is relatively rich in plant-derived protein and
antioxidants. A study by Baum et al, [21] had earlier
shown a reduction of both total cholesterol and triglycerides levels in a hypercholesterolemic man who was fed a
plant protein diet. Other studies have shown metabolic
syndrome-preventive activity of antioxidant components
(e.g. vitamin C, polyphenols). A number of polyphenols
such as epigallocatechin gallate have anti-obesity activity
and may improve metabolic disorders via modulation of
adipokines and growth factors, including metabolic
improvement of leptin function [22-24]. Although the
active principles of IGOB131 have not yet been fully identified, it is possible that it contains some of the above
mentioned compounds.
60
50
40
0
2
4
6
8
10
Figurefor
Changes
rected
2inplacebo
measured
values
characteristics in IGOB131 group corChanges in measured characteristics in IGOB131
group corrected for placebo values.
In conclusion, Irvingia gabonensis extract administered
twice a day to healthy, overweight and obese individuals
resulted in both weight reduction (body weight, body fat,
waist size) and an improvement in metabolic parameters
associated with insulin resistance. The current results suggest that IGOB131 may be a helpful adjunct in the management of overweight and/or obesity, supporting
previous suggestions from our laboratory [25].
Page 5 of 7
(page number not for citation purposes)
Lipids in Health and Disease 2009, 8:7
http://www.lipidworld.com/content/8/1/7
30
20
10
% Change
0
WT
WS
FAT
TCHOT
LDL
GLU
LEP
CRP
ADIP
Placebo
-10
IGOB131
-20
-30
-40
-50
Glucose
Figure
Percentage
3(GLU),
decrease
Leptin
in (LEP),
body weight
C-reactive
(WT),
protein
Waist(CRP)
size (WS),
and Adiponectin
Fat (FAT), Total
(ADIP)
Cholesterol
after 10 weeks
(TCHOL),
of useLDL
of extract
cholesterol
IGO131
(LDL),
Percentage decrease in body weight (WT), Waist size (WS), Fat (FAT), Total Cholesterol (TCHOL), LDL
cholesterol (LDL), Glucose (GLU), Leptin (LEP), C-reactive protein (CRP) and Adiponectin (ADIP) after 10
weeks of use of extract IGO131.
140
Adiponectin
120
% of change
100
80
y
y0 ae(1 e bx )
60
40
20
0
0
2
4
6
8
10
Time (week)
Figure 4in the adiponectin levels in IGOB131 group corrected for placebo
Changes
Changes in the adiponectin levels in IGOB131 group corrected for placebo.
Page 6 of 7
(page number not for citation purposes)
Lipids in Health and Disease 2009, 8:7
http://www.lipidworld.com/content/8/1/7
Competing interests
16.
Part of the funds for carrying out this study was provided
by Gateway Health Alliances Inc., through their "Encouragement of Research in developing countries Initiative".
17.
Authors' contributions
18.
JEO conceived, designed and coordinated the work, as
well as prepared the manuscript; CMFM was involved in
the co-design of the work as well as the draft of the manuscript. JLN carried out analytical work, BCE carried out
analytical work and contributed in drafting the manuscript; CBN carried out analytical and statistical analysis.
All authors read and approved the final manuscript.
19.
20.
21.
Acknowledgements
The LNNB is grateful to Gateway Health Alliances Inc. (Fairfield, California,
USA) for preparing and supplying the Irvingia gabonensis extract (IGOB131).
The authors are extremely grateful to Dr. Kenneth Blum of Wake Forest
University School of Medicine, North Carolina for reviewing this article.
22.
23.
References
1.
2.
3.
4.
5.
6.
7.
8.
9.
10.
11.
12.
13.
14.
15.
Conway B, Rene A: Obesity as a disease: no lightweight matter.
Obes Rev 2004, 3:145-51.
Pasquet P, Temgoua LS, Melaman SF, Froment A, Rikong AH: Prevalence of obesity and overweight for urban adults in Cameroon. Annal of Human Biology 2003, 30:551-562.
Caballero B: Introduction Symposium. Obesity in developing
countries, Biological and ecological factors. J Nutr 2001,
131(3):866-870.
Lee K, Sobal J: Socio-economic, dietary, activity, nutrition and
body weight transitions in South Korea. Public Health Nutr 2003,
6(7):665-674.
Reaven GM: Role of insulin resistance in human disease. Diabetes 1989, 37:1595-1607.
NCEP – National Cholesterol Education Program: Expert Panel on
Detection, Evaluation, and Treatment of High Blood Cholesterol in Adults (Adult Treatment Panel III) final report. Circulation 2002, 106:3143-3121.
Mandob DE, Ngondi JL, Fomekong GI, Agbor GA, Oben JE: Prediction and prevalence of metabolic syndrome in overweight
and obese subjects in Cameroon. International J of Biomedical and
Pharmaceutical Sciences 2008, 2:117-121.
Goodpaster BH, Kelley DE, Wing RR, Meier A, Thaete FL: Effects of
weight loss on regional fat distribution and insulin sensitivity
in obesity. Diabetes 1999, 48:839-847.
Knowler WC, Barrett-Connor E, Fowler SE, Hamman RF, Lachin JM,
Walker EA, Nathan DM: Reduction in the incidence of type 2
diabetes with lifestyle intervention or metformin. N Engl J
Med 2002, 346:393-403.
Ngondi J, Oben J, Minka R: The effect of Irvingia gabonensis
seeds on body weight and blod lipids of obese subjects in
Cameroon. Lipids in Health & Disease 2005, 4:12.
Ngondi JL, Djiotsa EJ, Fossouo Z, Oben J: Hypoglycaemic effect of
the methanol extract of Irvingia gabonensis seeds on streptozotocin diabetic rates. Afr J Trad Cam 2006, 3(4):74-77.
Oben J, Ngondi JL, Blum K: Inhibition of Irvingigia gabonensis
seed extract (OB131) on adipogenesis as mediated via down
regulation of the PPARy and Leptin genes and up-regulation
of the adiponectic gene. Lipids in Health and Disease 2008, 7:44.
Trinder P: Determination of glucose in blood using glucose
oxidase with an alternative oxygen acceptor. Ann Clin Biochem
1969, 6:24.
Richmond W: Preparation and properties of a cholesterol oxidase from Nocardia sp. and its application to the enzymatic
assay of total cholesterol in serum.
Clin Chem 1973,
19:1350-1356.
Buccolo G, David H: Quantitative determination of serum triglycerides by the use of enzymes. Clin Chem 1973, 19:476-482.
24.
25.
Maison P, Byrne CD, Hales CN, Day NE, Wareham NJ: Do different
dimensions of the metabolic syndrome change together
over time? Evidence supporting obesity as the central feature. Diabetes Care 2001, 24:1758-1763.
McTernan CL, McTernan PG, Harte AL, Levick PL, Barnett AH,
Kumar S: Resistin, central obesity, and type 2 diabetes. Lancet
2002, 359:46-47.
Friedman JM, Halaas JL: Leptin and the regulation of body
weight in mammals. Nature 1998, 395:763-770.
Weyer C, Funahashi T, Tanaka S, Hotta K, Matsuzawa Y, Pratley RE,
Tataranni PA: Hypoadiponectinemia in obesity and type 2 diabetes, close association with insulin resistance and hyperinsulinemia. J Clin Endocrinol Metab 2001, 86:1930-1935.
Yang WS, Lee WJ, Funahashi T, Tanaka S, Matsuzawa Y, Chao CL,
Chen CL, Tai TY, Chuang LM: Weight reduction increases
plasma levels of an adipose-derived anti-inflammatory protein, adiponectin. J Clin Endocrinol Metab 2001, 86:3815-3819.
Baum JA, Teng H, Erdman JW Jr, Weigel RM, Klein BP, Persky VW,
Freels S, Surya P, Bakhit RM, Ramos E, Shay NF, Potter SM: Longterm intake of soy protein improves blood lipid profiles and
increases mononuclear cell low-density-lipoprotein receptor
messenger RNA in hypercholesterolemic, postmenopausal
women. Am J Clin Nutr 1998, 68:545-551.
Kao YH, Hiipakka RA, Liao S: Modulation of obesity by a green
tea catechin. Am J Clin Nutr 2000, 72(5):1232-1234.
Okamoto Y, Kihara S, Ouchi N, Nishida M, Arita Y, Kumada M,
Ohashi K, Sakai N, Shimomura I, Kobayashi H, Terasaka N, Inaba T,
Funahashi T, Matsuzawa Y: Adiponectin reduces atherosclerosis
in apolipoprotein E-deficient mice.
Circulation 2002,
106:2767-2770.
Yamauchi T, Kamon J, Minokoshi Y, Ito Y, Waki H, Uchida S, Yamashita S, Noda M, Kita S, Ueki K, Eto K, Akanuma Y, Froguel P, Foufelle
F, Ferre P, Carling D, Kimura S, Nagai R, Kahn BB, Kadowaki T: Adiponectin stimulates glucose utilization and fatty-acid oxidation by activating AMP-activated protein kinase. Nat Med
2002, 8:1288-1295.
Ngondi JL, Fossouo Z, Djiotsa EJ, Oben J: Glycaemic variations
after administration of Irvingia gabonensis seeds fractions in
normoglycemic rats. Afri J Trad Cam 2006, 4:94-10.
Publish with Bio Med Central and every
scientist can read your work free of charge
"BioMed Central will be the most significant development for
disseminating the results of biomedical researc h in our lifetime."
Sir Paul Nurse, Cancer Research UK
Your research papers will be:
available free of charge to the entire biomedical community
peer reviewed and published immediately upon acceptance
cited in PubMed and archived on PubMed Central
yours — you keep the copyright
BioMedcentral
Submit your manuscript here:
http://www.biomedcentral.com/info/publishing_adv.asp
Page 7 of 7
(page number not for citation purposes)
Online Submissions: wjg.wjgnet.com
[email protected]
doi:10.3748/wjg.15.3073
World J Gastroenterol 2009 July 7; 15(25): 3073-3085
World Journal of Gastroenterology ISSN 1007-9327
© 2009 The WJG Press and Baishideng. All rights reserved.
EDITORIAL
A systematic review of the efficacy and safety of herbal
medicines used in the treatment of obesity
Shirin Hasani-Ranjbar, Neda Nayebi, Bagher Larijani, Mohammad Abdollahi
Shirin Hasani-Ranjbar, Neda Nayebi, Bagher Larijani,
Endocrinology and Metabolism Research Center, and Faculty
of Medicine, Tehran University of Medical Sciences, Tehran
1411413137, Iran
Mohammad Abdollahi, Faculty of Pharmacy, and Pharmaceutical
Sciences Research Centre, Tehran University of Medical
Sciences, Tehran 1417614411, Iran
Author contributions: Hasani-Ranjbar S completed the
bibliography and drafted the paper; Nayebi N carried out the
literature search and provided tables; Larijani B read the paper
and commented; Abdollahi M supervised, reviewed and edited
the paper.
Correspondence to: Mohammad Abdollahi, Professor,
Faculty of Pharmacy, and Pharmaceutical Sciences Research Center,
Tehran University of Medical Sciences, Tehran 1417614411,
Iran. [email protected]
Telephone: +98-21-66959104 Fax: +98-21-66959104
Received: March 17, 2009 Revised: May 1, 2009
Accepted: May 8, 2009
Published online: July 7, 2009
Abstract
This review focuses on the efficacy and safety of
effective herbal medicines in the management of
obesity in humans and animals. PubMed, Scopus,
Google Scholar, Web of Science, and IranMedex
databases were searched up to December 30,
2008. The search terms were “obesity” and (“herbal
medicine” or “plant”, “plant medicinal” or “medicine
traditional”) without narrowing or limiting search
elements. All of the human and animal studies on the
effects of herbs with the key outcome of change in
anthropometric measures such as body weight and
waist-hip circumference, body fat, amount of food
intake, and appetite were included. In vitro studies,
reviews, and letters to editors were excluded. Of the
publications identified in the initial database, 915
results were identified and reviewed, and a total of
77 studies were included (19 human and 58 animal
studies). Studies with Cissus quadrangularis (CQ),
Sambucus nigra , Asparagus officinalis , Garcinia
atroviridis , ephedra and caffeine, Slimax (extract
of several plants including Zingiber officinale and
Bofutsushosan) showed a significant decrease in body
weight. In 41 animal studies, significant weight loss
or inhibition of weight gain was found. No significant
adverse effects or mortality were observed except
in studies with supplements containing ephedra,
caffeine and Bofutsushosan. In conclusion, compounds
containing ephedra, CQ, ginseng, bitter melon, and
zingiber were found to be effective in the management
of obesity. Attention to these natural compounds would
open a new approach for novel therapeutic and more
effective agents.
© 2009 The WJG Press and Baishideng. All rights reserved.
Key words: Animal; Herbal medicine; Human; Obesity
Peer reviewers: Cheng Ji, Professor of Research, Department
of Medicine, University of Southern California, 2011 Zonal
Ave., HMR-101, Los Angeles, CA 90033, United States;
Anders E Lehmann, PhD, Associate Professor, Senior Principal
Scientist, Bioscience, AstraZeneca R&D Mölndal, Mölndal,
Sweden
Hasani-Ranjbar S, Nayebi N, Larijani B, Abdollahi M. A
systematic review of the efficacy and safety of herbal medicines
used in the treatment of obesity. World J Gastroenterol 2009;
15(25): 3073-3085 Available from: URL: http://www.wjgnet.
com/1007-9327/15/3073.asp DOI: http://dx.doi.org/10.3748/
wjg.15.3073
INTRODUCTION
The prevalence of obesity is increasing worldwide [1]
resulting in an association with major health problems
such as type 2 diabetes, ischemic heart disease, stroke,
and cancer. It is necessary to treat obese individuals by
both lifestyle interventions and/or pharmacological
therapy. Phar macologic treatment and surgical
interventions used in some circumstances are not always
appropriate[2]. Unfortunately, drug treatment of obesity
despite short-term benefits, is often associated with
rebound weight gain after the cessation of drug use, side
effects from the medication, and the potential for drug
abuse[3]. Pharmacologic options include sibutramine,
orlistat, phentermine, diethylpropion, and fluoxetine
or bupropion. Phentermine and diethylpropion have
potential for abuse and are only approved for shortterm use. Approved medications for long term use in
the treatment of obesity are sibutramine and orlistat,
however, these agents should be used with caution in
patients with a history of cardiovascular disorders [4].
The general public uses many other methods for weight
www.wjgnet.com
3074 ISSN 1007-9327
CN 14-1219/R
World J Gastroenterol
loss including herbs, vitamins, nutritional supplements,
and meal replacement preparations. Rigorous scientific
studies have not been carried out on these products,
and in many cases safety and efficacy take a back seat to
marketing.
Complementary and alternative therapies have
long been used in the Eastern world but recently these
therapies are being used increasingly worldwide[5]. When
conventional medicine fails to treat chronic diseases
and conditions such as obesity efficaciously and without
adverse events, many people seek unconventional
therapies including herbal medicine [6]. Although the
number of randomized trials on complementary therapies
has doubled every 5 years and the Cochrane library
included 100 systematic reviews of unconventional
inter ventions [7] , none of these studies specifically
mentioned herbal therapy in obesity.
This review aimed to evaluate the current science
on the efficacy and safety of herbal medicines in the
management of obesity.
DATA SOURCES AND STUDY
SELECTIONS
PubMed, Scopus, Google Scholar, Web of Science, and
IranMedex databases were searched up to December 30,
2008 for all human and animal studies investigating the
effects (both harmful and beneficial) of treating obesity
with herbal medicines. The search terms were “obesity”
and (“herbal medicine” or “plant”, “plant medicinal” or
“medicine traditional”) without narrowing or limiting
search elements. Only publications with available
abstracts were reviewed. The main outcome measures
sought at the end of treatments as anti-obesity effects,
were body weight, body fat including fat mass/fat weight
or fat percentage/visceral adipose tissue weight, triceps
skin fold thickness, waist or hip circumference, and
appetite or amount of food intake.
Herbal medicines are defined in this review as raw or
refined products derived from plants or parts of plants
(e.g. leaves, stems, buds, flowers, roots, or tubers) used
for the treatment of diseases. The synonyms of herbal
medicines are herbal remedies, herbal medications,
herbal products, herbal preparations, medicinal herbs,
and phytopharmaceuticals, etc.
All of the abstracts from human and animal studies
with the main outcome of change in anthropometric
measures such as body weight and waist-hip circumference,
body fat (weight or mass of visceral adipose tissue, fat
mass or percent), amount of food intake, and appetite in
participants were included. Even studies on other relevant
diseases such as diabetes were also reviewed and included
if the appropriate outcomes were shown. In vitro studies,
review articles, and letters to the editor were excluded.
Unpublished data such as theses were also excluded. Two
reviewers independently examined the title, abstract and
references of each article meeting the inclusion criteria
and eliminated duplications and those showing exclusion
criteria.
July 7, 2009
Volume 15
Number 25
FINDINGS
Of the publications identified from the initial database
search, 915 results were identified and reviewed for
inclusion or exclusion. A total of 77 studies were
included (19 human and 58 animal studies). Human
studies included 17 randomized clinical trials (RCTs)
and two before-after clinical trials[8-26]. RCTs reported
random allocation of humans to herbal medicines vs
(placebo/another plant/combination of plants) with or
without specific dietary and exercise programs outlined in
Tables 1 and 2 as weight loss programs. Human subjects
were healthy overweight, obese or with impaired glucose
tolerance test volunteers. Animal studies included healthy,
genetically or experimentally obese or diabetic mice, rats
and other rodents. The route of administration of herbs
in almost all studies was oral intake with the exception of
some animal studies as indicated in Table 2.
HUMAN STUDIES
Change in human body weight
All studies showed loss of body weight except one[21]
which seemed to have problems with the study design,
and one other study [10] which showed a significant
decrease only in body fat. Studies with Cissus quadrangularis
(CQ)[26] or combined with Irvingia gabonensis (IG)[15], a
combination of Sambucus nigra and Asparagus officinalis[16],
calcium hydroxycitrate in Garcinia atroviridis[18], supplements
containing ephedra and caffeine[9,13,20], and Slimax as an
extract of several plants including Zingiber officinale[8] and
Bofutsushosan[14] showed significant decreases in body
weight.
Body fat
A significant decrease in body fat was shown with CQ[26],
supplements containing ephedra and caffeine[9,13], a natural
compound containing capsicum and some lipotropic
nutrients[10], Bofutsushosan[14], and calcium hydroxycitrate
in Garcinia atroviridis [18]. These phytopharmaceuticals
showed a significant decrease in triceps skin fold thickness
indicating significant loss of fat.
Waist and hip circumference
Efficient decreases in both waist and hip circumferences
in trials with a supplement containing ephedra and
caffeine[9] and Slimax (extract of several plants including
Zingiber officinale [8] were shown whereas Caralluma
fimbriata[19] and CQ with or without IG[15] significantly
decreased waist size.
Food intake
Decreases in appetite or amount of food or energy
intake with a supplement containing ephedra and
caffeine[20] and Caralluma fimbriata[19] were shown (not
significant) but hydroxycitric acid (HCA-SX) with or
without Gymnema sylvestre[23] decreased the amount of
food intake efficiently. A natural compound containing
capsicum and other lipotropic nutrients [10] did not
significantly change energy intake.
www.wjgnet.com
Hasani-Ranjbar S et al . Herbal medicines used in the management of obesity
3075
Table 1 Human studies considering the anti-obesity effects of herbal medicines
Authors
Ignjatovic
et al[8] 2000
Boozer et al[9]
2001
Hoeger
et al[10] 1998
Target
Herbs (scientific name)
Healthy
Slimax: extract of
volunteers several plants: Hordeum
vulgare, Polygonatum
multiflorum,
Dimocarpus longan,
Ligusticum sinense,
Lilium brownie, and
Zingiber officinale
Over wt.
An herbal supplement:
(n = 35 )
(Ma Huang & Guarana)
Healthy
A natural dietary
compound of
chromium picolinate,
inulin, capsicum, Lphenylalanine, and other
lipotropic nutrients
Study
Dose/duration
Groups
Main outcome
Other relevant effects
& complications
RCT
6 wk
C: Placebo
I: Compound
Sig. decrease in
body wt. & waist &
hip Cir. & BMI
Modification of lipid
metabolism with
sig. effect on the
accumulation & the
release of lipid from
adipose tissue
RCT
(doubleblind)
72 mg (ephedra)
240 mg
(caffeine)/8 wk
C: Placebo
(n = 24)
I: Compound
(n = 24)
RCT
(doubleblind)
4 wk
C: wt. loss
program
(n = 67)
I: wt. loss
program +
compound
(n = 56)
Greater reduction in
Sig. decrease in
serum TG, potentially
body wt. & total
treatment-related
body fat & sig.
greater reduction in dropouts (23%) in the
active group and none
hip & waist Cir.
in the placebo group.
Dry mouth, insomnia
& headache were
reported
Sig. decrease in
body fat percent, fat
mass & FFM, but
no sig. difference in
body wt. BMI and
energy intake
Ziauddin
et al[11] 2004
Hhyperlip- Terminalia arjuna Roxb
idemic
(n = 30)
Beforeafter CT
Abidov
et al[12] 2006
Obese non- A compound of Aralia
diabetic
mandshurica (A) and
women
Engelhardtia chrysolepis
(n = 32)
(E) extracts named
ARALOX
RCT
450 mg (A) &
450 mg (E)/d
C: Diet +
placebo
I: Diet
+ compound
Greenway
et al[13] 2004
Human
(obese &
over wt.)
healthy
Obese
women with
IGT (n = 80)
Herbal supplement
containing caffeine
and ephedra
RCT
(doubleblind)
210 mg (e) &
72 mg (c)/12 wk
C: Placebo
I: Compound
Bofu-tsusho-san
containing (Ephedrae
Herba, Glycyrrhizae
Radix, Forsythiae
Fructus, Schizonepetae
Spica &…)
RCT
(doubleblind)
Equivalent of
(24 mg/ephedrine
& 280 mg
caffeine/24 wk)
C: wt. loss
program
I: wt. loss
program +
compound
Hioki et al[14]
2004
Oben et al[15]
2008
Human
(obese &
over wt.)
A combination of Cissus
quadrangularis (CQ) &
Irvingia gabonensis (IG)
RCT
(doubleblind)
300 mg (CQ) &
500 mg (IG)
per day/10 wk
C: Placebo
I: CQ
CQ + IG
Chrubasik
et al[16] 2008
Healthy
(n = 80)
A combination of
Sambucus nigra (S) and
Asparagus officinalis (A)
Beforeafter CT
(S): 1 mg
anthocyanin,
370 mg flavonol,
150 mg
hydroxycinnamate
(A): 19 mg
saponin per day
-
www.wjgnet.com
Sig. improvement Sig. decrease in serum
in obesity.
total lipid levels. Sig.
Reduction in body
relief of palpitation,
wt. in some cases dyspnea, chest & joint
pain. Reduction in BP
in some cases
Decrease in total Reduction in perilipin
body wt. & fat wt. content in adipocytes
and plasma TG.
Stimulate activity of
hormone sensitive
lipase
Sig. decrease in
No differences in lipid
body wt. & the
levels, or BP were
percentage of fat
shown. No serious
adverse effect
No decrease in RMR.
Compared to
Sig. improvement
baseline the I group
in insulin resistance
lost significantly
compared to
more body wt. &
abdominal visceral week 0. Loose bowel
fat & the placebo movements resulted in
three withdrawals
group lost sig.
body wt. & had
no sig. change in
abdominal visceral
fat
Sig. decrease in Chol
Sig. decrease in
& LDL of plasma and
body wt. & body
fat percent & waist fasting blood glucose
levels
size in both I groups
but the combination
group (CQ + IG)
resulted in larger
reductions
Sig. decrease in
Sig. improvement
mean of the wt.
of BP, physical and
emotional well-being
and quality of life
3076 ISSN 1007-9327
CN 14-1219/R
World J Gastroenterol
Udani et al[17]
2007
Healthy
(n = 25)
Proprietary fractionated
white bean extract
RCT
(doubleblind)
2000 mg/14 wk
Roongpisu­
thipong
et al[18] 2007
Obese
women
Calcium hydroxycitrate
in Garcinia atroviridis
RCT
2 mo
Kuriyan
et al[19] 2007
Over wt.
(n = 50)
Caralluma fimbriata
RCT
1 g/60 d
Hackman
et al[20] 2007
Obese &
over wt.
women
(n = 41)
Multinutrient
supplement containing
ephedra (e) and
caffeine (c)
RCT
(doubleblind)
40 mg (e) and
100 mg (c)/9 mo
Garrison
[21]
et al 2006
Over wt.
women
Proprietary extracts of
Magnolia officinalis and
phellodendron amurense
RCT
750 mg/6 wk
Coffey et al[22]
2004
Human
(over wt.
& obese)
(n = 102)
Product containing
ephedrine, caffeine &
other ingredients.
RCT
(doubleblind)
12 wk
[23]
Obese
(n = 60)
Hydroxycitric acid (HCA
-SX) and a combination
of HCA-SX and niacinbound chromium
(NBC) and Gymnema
sylvestre extract (GSE)
RCT
(doubleblind)
HCA-SX:
4667 mg
GSE: 400 mg
NBC: 4 mg/8 wk
[24]
Obese
(n = 24)
A proprietary
fractionated white bean
(Phaseolus vulgaris)
RCT
(doubleblind)
3000 mg/8 wk
Bhatt et al[25]
1995
Healthy
(n = 58)
Guggulu (Medohar)
RCT
1/5, 3 g/30 d
Oben et al[26]
2007
Over wt. &
obese
Cissus quadrangularis
RCT
(doubleblind)
300, 1028 mg
Preuss et al
2004
Udani et al
2004
July 7, 2009
Volume 15
Number 25
C: Placebo + wt.
loss program
I: Extract + wt.
loss program
In both groups,
There were no
decrease in body
adverse effect
wt. & waist size
from baseline was
sig. but no sig value
between groups
C: Diet
Sig. decrease in
I: Diet + extract body wt. & greater
reduction in BMI.
Sig. decrease in the
triceps skin fold
thickness
C: wt. loss
Sig. decrease in
program
waist Cir. & hunger
I: wt. loss
levels. Greater
program +
decrease in body
extract
wt., BMI, hip Cir.,
body fat & energy
intake but not sig.
C: Control
Sig. decrease in
Sig. decline in serum
supplement
body wt. decrease
chol, TG, glucose,
I: Multinutrient
in appetite
fasting insulin & leptin
supplement
levels & minor adverse
effects like dry mouth,
insomnia, nervousness
and palpitation were
reported
C: Placebo
No sig. wt. gain
The I groups tended
I: Extract
for the I group
to have lower levels of
but sig. wt. gain
cortisol in the evening
in C. groups
C: Placebo
Additional wt. loss No difference in pulse,
I: Compound
(1/5 kg) & greater diastolic & systolic BP
reduction in BMI
& adverse events
& waist Cir. No
difference in body
fat & fat mass
percent was shown
C: Placebo
5%-6% decrease in Sig. decrease in serum
I1 = HCA-SX
body wt. & BMI
lipids & leptin &
I2 = GSE + NBC
& sig. decrease in
increase in HDL &
+ HCA-SX
food intake in
excretion of urinary
All groups
both I groups
fat metabolites in both
had wt. loss
I groups. There were
program
mild adverse effects
but not significant
between groups
C: Placebo
Decrease of body Reduction of TG three
I: Extract
wt. with 129%
times greater than
difference
C. group. No adverse
effect was shown
C: wt. loss
Higher mean wt.
program
reduction
I: wt. loss
in I group.
program +
In I group, all
extract
patients > 90 kg lost
wt. but 3 in C group
did not lose wt.
C: Placebo
Sig. decrease in
Sig. decrease in serum
I: Two extract
body wt &
lipids and glucose.
formulation:
body fat
Sig. increase in HDL-C
CQR-300, CORE
plasma 5-HT and
creatinine levels
Cir: Circumference; BP: Blood pressure; BMI: Body mass index; sig.: Significant; C: Control; I: Intervention; RCT: Randomized control trial; CT: Clinical
trial; TG: Triglyceride; HDL: High density lipoprotein; LDL: Low density lipoprotein; Chol: Cholesterol; IGT: Impaired glucose tolerance.
Other effects
Anti-hyperlipidemic, antihyperglycemic, and other
relevant anti-obesity effects of medicinal plants in
human studies are summarized in Table 1.
Adverse effects
No significant adverse effects compared to controls
were mentioned and no mortality was reported, except
in studies with supplements containing ephedra and
www.wjgnet.com
Hasani-Ranjbar S et al . Herbal medicines used in the management of obesity
3077
Table 2 Animal studies on the anti-obesity effects of herbal medicines
Authors
Target
Herbs (scientific name)
Dose/duration
Groups
Main outcome
Other relevant effects
& complications
Wang
et al[27] 2000
Rat (obese)
(2.5 g/kg)
-
Sig. decrease in
Lee’s index &
size of fat cells
Did not influence the function
of thyroid gland & metabolism
of water & salt
Jeon
et al[28] 2003
Mouse
Haidonghua powder:
Laminaria japonica
Aresch & Benincasa
hispida (Thunb.) Cogn.
etc
Rhus vemiciflua
Stokes
8 wk
C: HFD
I: HFD + extract
AlarconAguilar
et al[29] 2007
Mouse
Hibiscus sabdariffa
120 mg/kg
60 d
Urías-Silvas
et al[30] 2008
Mouse
Fructans extracted from
Agave tequilana (TEQ)
and Dasylirion spp
(DAS)
10%
supplement
Park et al[31]
2007
Rat
Platycodon
grandiflorum
150 mg/kg
7 wk
Jongwon
et al[32] 2005
Rat (obese
by HFD)
Allium victorialis var.
platyphyllum leaves
100 mg/kg
2 wk
Kobayashi
et al[33] 2001
Rat
Evodiamine an alkaloid
of a fruit: Evodia
rutaecarpa
0/02%, 0/03%
of the diet
12 wk
Jin et al[34]
1994
Rat
Jiang-zhi jian-fei yao:
the refined Rhubarb
Injected
intragastrically
Kim et al[35]
2008
Rat
Juniperus chinensis
1% supplement
/79 d
Shih et al[36]
2008
Mouse (obese
by HFD)
Momordica charantia
(bitter melon)
4 wk
Pang
et al[37] 2008
Rat (obese
by HFD)
Ilex paraguariensis
Bruno
et al[38] 2008
Mouse
Green tea
0%, 1%, 2%
(wt.:wt.)/6 wk
Sig. suppression
of body wt. gain
and lower wt. of
subcutaneous
adipose tissue
C: Healthy &
Sig. decrease in body
obese (by MSG) + wt. gain in obese mice
placebo
& increased liquid
I: Same groups + intake in both groups
extract
C: STD
Sig. decrease in body
I: STD +
wt. gain & food intake.
Raftilose/
The (TEQ) group had
DAS/TEQ
the lowest value
Lowered plasma TG
No sig. change in TG & Chol
levels. Increase in ALT levels
was shown but was not sig.
Lower serum glucose & Chol
level but Sig. decrease in TG
levels was shown in Raftilose
group. Higher concentration
of GLP-1 & it's precursor &
proglucagon mRNA in I groups
C: NLD/HFD
Sig. decrease in body
Sig. decrease in plasma TG
wt & subcutaneous
I: Same groups
& Chol concentrations, upadipose tissue wt. &
+ extract
regulation of FABP mRNA
adipocytes size in I
expression induced by HFD
group
Considerable reduction Sig. decrease in hyperlipidemia
of retroperitoneal,
and increased lipid content in
epididymal and total
feces
abdominal fat pad wt.
C: Control
Sig. decrease in
Sig. decrease of lipid in liver
& serum FFAs. Sig. increase of
I: Extract
perirenal fat wt. &
decrease of epididymal lipolytic activity in perirenal
fat tissue & specific GDP
fat mass
binding in brown adipose tissue
mitochondria as the biological
index of heat production
No sig. increase in
Prolongation of stomach
body wt. but reduction
evacuation time and
of food intake.
acceleration of intestinal
Decreased size of
movements
abdominal adipose
cells
C: NLD/HFD
Sig. decrease in body
Sig. decrease in blood lipid,
leptin & insulin levels. Sig.
I: HFD + extract wt gain & visceral fat
reversal of the down-regulation
pad wt.
of genes implicated in
adipogenesis & increased gene
expressions & phosphorylations
related to FABO
C: Control
Sig. decrease in
Sig. improvement in blood
epididymal white
glucose, leptin, and FFA.
I: Rosiglitazone/
adipose tissue wt. &
Influenced PPARα/
extract
visceral fat wt.
PPARγ expression
Sig. decrease in body
Sig. decrease in blood and
wt. of visceral fat-pad hepatic lipid, glucose, insulin
and leptin levels. Reversed
wt.
the down-regulation of genes
implicated adipogenesis,
thermogenesis & enhanced
expression of uncoupling
proteins in adipose tissue
C: Obese/lean
Sig. decrease in body In obese I group, sig. decrease
in hepatic steatosis was
I: Same groups
wt. of both I groups
observed dose dependently.
+ extract
Liver enzymes decreased.
30%-41% and 22%-33% lower
serum ALT and AST activities
were shown, respectively
www.wjgnet.com
3078 ISSN 1007-9327
CN 14-1219/R
World J Gastroenterol
Lee et al[39]
2008
Mouse
A combination of Morus
alba, Melissa officinalis
and Artemisia capillaries
12 wk
-
Choi et al[40]
2008
Mouse
(obese by
HFD)
Cucurbita moschata
500 mg/kg
8 wk
_
Huang
et al 2008[41]
Rat
Lemaure
et al[42] 2007
Rat (obese)
Salacia reticulata
0/5% 8 wk
in mice
0/2% 35 d
in rats
1 g/kg (A),
500 mg/kg (u)
8 wk
C: HFD
I: HFD + plant
1% (wt:wt)
/12 wk
C: HFD
I: HFD + extract
Lei et al[43]
2007
Mouse
Aoki et al[44]
2007
Mouse
(obese by
HFD)
Oluyemi
et al[45] 2007
Rat
Han et al[46]
2006
Mouse
(obese by
HFD)
Goyal
et al[47] 2006
Mouse
(obese gold
thioglucose)
Rat and
mouse
Kishino
et al[48] 2006
Jayaprakasam
et al[49] 2006
Mouse
July 7, 2009
Volume 15
Sig. decrease in body
wt. gain & adipose
mass
Number 25
Decreased serum levels of TG,
Chol & inhibited hepatic lipid
accumulation, and increased
hepatic mRNA levels of
enzymes responsible for FABO
Sig. suppression
of body wt. &
fat storage increase
but amount of food
intake was not
affected
Momordira charantia L.
5%
C: HFD
Sig. decrease in the
Sig. decrease in enzymes of
(Bitter melon)
I1: HFD + plant
number of large
adipose tissue implicating
I2: HFD +
adipocytes in both I
reduction of insulin resistance
thiazolidinedione groups. Sig. decrease
in I group as compared to
in adipose tissue mass
C group
in I1 group compared
to I2 group
Cyperus rotundus
45, 220 mg/kg
_
Sig. decrease in wt.
L. tubers
60 d
gain without affecting
food consumption
Pomegranate leaf
400/800 mg
C: HFD/NLD
Sig. decrease in body
Sig. decrease in serum TG,
per kilogram
I: Same groups
wt. & energy intake
Chol, glucose levels & Chol/
5 wk
+ extract
and adipose pad wt.
HDL ratio, inhibition of
percents in I. group.
intestinal fat absorption
Sig. decrease in
appetite of obese mice
on NLD was shown
Licorice flavonoids
0/5%, 1%, 2%
C: Placebo
Sig. decrease of
Improvement of fatty
oil (LFO)
8 wk
I: Extract
abdominal white
degeneration of hepatocytes and
adipose tissue & body changes in genes implicating
wt. gain with 1% & 2% regulation of lipid metabolism
LFO groups, decrease
with 2% concentration
of adipocyte size
Garcinia cambogia
200, 400 mg/kg
C: Placebo
Sig. decrease in
Sig. decrease in TG pool of
seed (bitter cola)
5 wk
I: Extract
body wt.
adipose tissue & liver but sig.
increase of HDL & decreased
LDL
Kochia scoparia
1%, 3%/3 d
Prevented the
Sig. increase the fecal content &
increases in body &
fecal TG levels in day 3
parametrial adipose
tissue wt.
Zingiber officinale
250 mg/kg
C: Placebo
Sig. decrease in
Sig. decrease in serum Chol,
8 wk
I: Extract
body wt.
TG, glucose, and insulin
Cornelian cherry
(cornus mas) (Purified
anthocyanins (A) &
ursolic acid (u)
Arachis hypogaea
nutshell
Moreno
et al[50] 2006
Rat
Galisteo
et al[51] 2005
Rat (obese)
Plantago Ovata
Zhao et al[52]
2005
Mouse (obese
by
hyperalimentation)
Phillyrin (Fructose
forsythia)
Chen et al[53]
2005
Rat
Bitter melon (Momordica
charantia)
3/5%
25 wk
0/75% or
7/5 g per
kilogram
7 wk
C: HFD
I: HFD + A/A
+u
Sig. decrease in the
Sig. decrease in plasma TG,
body wt. and visceral 4 h after ingestion; Sig. decrease
fat mass increase
in energy efficiency, plasma
leptin and adiponectine levels
24% decrease in wt.
Elevated insulin levels; Sig.
gain in (A) group
decrease of liver TG in A + u
group
Increased fecal lipid excretion.
Reduced TG content of liver
and serum glucose and insulin
C: STD
Sig. decrease in body
Sig. improvement of lipid
I: STD + extract
wt. gain
profile, FFA & insulin & TNF-α
& hypoadinectinemia
Sig. decrease in wet Decrease in jejunum microvillus
wt. of fat & fat index &
area, and serum levels of
diameter of fat cells &
TG & Chol
lee index
C: LFD/HFD
Lower energy
Reduced plasma glucose and
I: LFD/HFD
efficiency and visceral hepatic TG but higher serum
+ extract
fat mass after 4 wk in FFA after 4 wk; Higher plasma
I group
catecholamine after 7 wk in
I group; Sig. decrease in hepatic
TG & steatosis and sig. increase
of serum epinephrine & FFA in
HFD group of I
www.wjgnet.com
Sig. decrease in body
wt. gain and liver size
Hasani-Ranjbar S et al . Herbal medicines used in the management of obesity
Han et al[54]
2005
Rat
Coleus forskohlii
50 g/kg
Han et al[55]
2005
Mouse
Chikusetsu saponins
isolated from Panax
japonicus rhizomes
1%, 3%/9 wk
Han et al[56]
2005
Mouse
Zingiber officinale
Roscoe
1%, 3%/8 wk
Cha et al[57]
2004
Mouse
Kim et al[58]
2005
Rat
Yun et al[59]
2004
Mouse
Junbao
et al[60] 2004
Kim et al[61]
2004
Rat (obese)
Kwon et al[62]
2003
Lu et al[63]
1999
Rat
Rodent
C: Sham
operated/
ovariectomized
+ control diet
I: Same groups
+ extract
C: HFD
I: HFD + extract
Reduced body wt.&
food intake & fat
accumulation
Prevented body wt.
gain & increase of
parametrial adipose
tissue wt.
Sig. decrease in body
wt. gain at 2-8 wk
with 3% & in final
parametrial adipose
tissue wt. with 1%
concentration
HFD group of I had
C: NLD/HFD
Acanthopanax
0/5 g per
lower wt. gain but no
I: NLD/HFD
senticosus
kilogram
difference in food
+ extract
12 wk
consumption was
shown
Reduced body wt.,
Crude saponin of
200 mg/kg
C: NLD/HFD
food intake & fat
Korean red ginseng
3 wk, ip
I: NLD/HFD
content in HFD group
+ extract
of I similar to those
fed with NLD
Wild Ginseng
250, 500 mg/kg
C: HFD
Sig. inhibition of
I: HFD + extract
body wt. gain dose
dependently.
Decrease of white &
brown adipocytes
diameters
Semen cassiae
6%
Sig. decrease in body
wt. & lee index
Sig. decrease in body
50 mg/100 g
C1: NLD
Adlay seed
wt. & food intake
of body wt.
C2: HFD + saline
(CoixLachrymajobi var.
& epididymal and
(sham group)
mayuen)
I: HFD + plant peritoneal fat & white
adipose tissue size as
compared to sham
group
Dioscorea nipponica
5%/8 wk
C: HFD
Sig. decrease in body
Makino
I: HFD + plant
wt. & adipose gain
Inspissation tea
(Guangdong
kudingcha)
Yoshikawa
et al[64] 2002
Rat (obese)
Salacia reticulata
125 mg/kg
27 d
Xie et al[65]
2002
Mouse
Ginseng berry
150 mg/kg
12 d, ip
Yamamoto
et al[66] 2000
Rat
CT-Ⅱ, an extract from
Nomame Herba
8 wk, 12 wk,
6 mo
Han et al[67]
1999
Mouse
Oolong tea
10 wk
Sig. increase of the fecal content
& TG level in day 3; reduction
of plasma TG 2 h after oral
lipid intake & inhibition of
pancreatic lipase activity
C: HFD
I: HFD + plant
Stronger modulation
on lymphocytes
hypertrophy and
quantity was shown in
I1 group
Suppression of body
wt. and periuterine
fat storage increase
in female rats but no
effect on male rats
C: Diabetic/lean Sig. decrease in body
wt. as compared to day
diabetic
0 in diabetic group of
+ placebo
I: Same groups I. wt. loss in lean mice
was shown
+ extract
Sig. inhibition of
C: Lean/obese
body wt. gain dose
+ HFD
I: Same groups dependently without
+ HFD + plant affecting food intake in
lean rats after 12 wk.
Sig. decrease in body
wt. gain in obese mice
after 24 wk
C: HFD
No sig. difference
I: HFD + extract
in food intake but
prevented obesity &
liver induced by a HFD
C: Control
I1: Extract
I2: Fenfluramine
Rat (obese by
hyperalimentation)
3079
www.wjgnet.com
In HFD group of I, lower serum
LDL and restoration of liver TG
at the same level as fed by LFD
was shown; No alteration in
carnitine status
Reduction of hypothalamic
NPY expression and serum
leptin level in HFD group of I
Sig. inhibition of FBG, TG, and
FFAs dose-dependently; insulin
resistance improved
Reduction of fasting serum TG,
insulin & malondialdehyde
Increase of brown adipocytes
as compared to NLD group
but not significant
Suppression of time dependent
increase of serum TG level after
lipid intake
Only fenfluramine showed sig.
difference in small intestine
villus model
Sig. increase in glucose
tolerance in diabetic mice but
no sig. decrease of FBG in lean
mice.
Sig. inhibition of TG elevation
Enhancement of noradrenalin
induced lipolysis & inhibition
of pancreatic lipase activity
3080 ISSN 1007-9327
CN 14-1219/R
World J Gastroenterol
Pusztai
et al[68] 1998
Rat
Kidney bean
(Phaseolus vulgaris)
130, 150,
280 g/kg
10-70 d
Nagasawa
et al[69] 1991
Mouse
(obese)
Tree peony root
(Paenia suffruticosa)
0/5%
30 wk
Wang
et al[70] 2008
Mouse
Parasitic loranthus from
Loranthacea or Viscaceae
20 d
Hu et al[71]
2008
Mouse (female)
2%/11 wk
Ohkoshi
et al[72] 2007
Mouse
Escins extracted
from Aesculus
turbinata Blume
(Hippocastanaceae)
Nelumbo nucifera
Gaertn leaves
(Nymphaceae)
Kang et al[73]
2004
Rat
6 wk
Mary et al[74]
2003
Rabbit
PM-F2-OB composed
of Lycii Fructus,
Rehmanniae Radix,
Coicis Semen,
Carthami Flos, Hoelen,
Angelicae Radix,
Nelumbinis Semen,
Radix Dioscorea
and Aurantii
Fructus
Caps HT2
A herbal formulation
Wu et al[75]
2005
Rat (diabetic
by STZ )
Xie et al[76]
2005
Mouse
(Genetically
obese)
Palit et al[77]
1999
Mouse
Oi et al[78]
1999
50%
July 7, 2009
Volume 15
Number 25
Sig. decrease of body protein
in lean I group. Sig. decrease in
plasma insulin levels in obese
I group. Sig. pancreatic growth
after long term feeding in all
I groups
C: Control
Sig. decrease in food
Improvement in glucose
I: Extract
intake and Lee index tolerance. No sig. difference in
serum FFA levels
Sig. decrease in body High inhibitory ability on FASwt. & food intake
Loran thacea was nearly 400
fold stronger than that from
the viscaceae
Suppressed the increase of liver
I: HFD
Suppressed the
TG content; increased TG in
C: HFD + extract
increase in body &
feces after fat ingestion
parametrial adipose
tissue wt.
Sig. suppression of
Exhibition of lipolytic activity
C: STD/HFD
body wt. gain
especially in visceral adipose
I: Same groups
tissue; β adrenergic receptor
+ extract
pathway was partly involved
Sig. decrease in serum Chol/
No sig. difference in
C: STD/HFD
LDL and total lipids; reduction
wt. change if STD
I: Same groups
of kidney fat wt./FFA/PL &
was used but in HFD
+ plant
group of I resulted in TG to levels equal or below the
normal diet
sig. decrease in body
wt. gain but showed
no sig. difference in
amount of food intake
C: Lean/obese
+ LFD/HFD
I: Same groups
+ extract
-
5 mg/kg (iv)
30 d
100/200/300/
400/mg per
kilogram
orally
400 mg/kg
Astragalus
(APS) 5 wk
polysaccharide (APS) a
component of Astragalus
membranaceus roots
C: Placebo
Total, Ginsenosides in 100, 200 mg/kg
I: Extract
(ip) 12 wk
Chinese ginseng (TG
& 150,
CG), from leaves and the
300 mg/kg
stem of Panax ginseng
(oral)/12 wk
Galega officinalis
10% (w/w) of C: Diabetic/NL
the diet 28 d
I: Same groups
+ plant
The growth was
retarded dosedependently lower
body fat
Sig. decrease in body Sig. increase in HDL after oral
wt.
administration and decrease
in atherogenic index in oral
administration; Sig. increase of
the release of LPL enzyme and
sig. hypolipidemic effect in
Ⅳ groups
Sig. decrease in
Sig. decrease in plasma glucose;
body wt.
improved insulin sensitivity
Sig. decrease in
body wt.
Sig. decrease in body
wt. in both I groups,
sig. wt. loss in normal
mice independent of
a reduction in food
intake but in diabetic
mice wt. loss was with
reduced food intake
C: HFD
Sig. decrease in
I: HFD + extract body wt. & perirenal
adipose tissue wt. &
epididymal fat pad
Rat
Garlic
8 g/kg of diet
28 d
Yoshida
et al[79] 1995
Mouse (obese
and lean)
Bofu-tsusho-san
1/4%, 4/7% of
wt. of food
8 wk
-
He et al[80]
2008
Rat (obese by
STZ & HFD
Yi-Qi-Yang-Yin-Ye
2, 4, 8 g/kg
4 wk
-
www.wjgnet.com
Sig. decrease in body
wt. & retroperitoneal
white adipose tissue
wt. and no change in
food intake
Body wt. decreased
Sig. decrease in FBG in
200 mg/kg dose after injection
Sig. decrease in FBG in
300 mg/kg dose
Striking loss of body fat in both
groups; Sig. decrease in serum
glucose in both groups but Sig.
decrease in serum insulin in
diabetic mice
Sig. decrease in plasma
TG levels; sig. decrease in
mitochondrial protein and
(UCP) in brown adipose tissue,
and in urinary noradrenaline
and adrenaline excretion
Sig. increase in GDP binding
dose dependently
Decrease in TG/Chol/
LDL/FFA/FBG/insulin;
improvement of glucose
tolerance
Hasani-Ranjbar S et al . Herbal medicines used in the management of obesity
Jeong et al[81]
2008
Park
et al[82] 2005
Akagiri
et al[83] 2008
Kim et al[84]
2005
Attele et al[85]
2002
Rat (fatty)
Gyeongshang
angjeehwan: Liriope
platyphylla F.T./Wang
& T. Tang (Liliaceae),
Platycodongrandiflorum
A. DC. (Campanulaceae).
Schisandrachinensis K.
Koch (Magnoliaceae).
Ephedra sinica Stapf
(Ephedraceae)
Rat (obese by diet)
Platycodon
grandiflorum
3081
8 wk
C: Placebo
I: Compound
Sig. decrease in food
intake & body wt. gain
& abdominal fat
Sig. decrease in plasma leptin
levels; decrease in circulating
TG and inhibition of lipid
accumulation in liver; increase
of mRNA of genes responsible
for FABO
150 mg/kg
7 wk
C: Convert to
NLD/HFD
I: Same groups
+ extract
Sig. decrease in wt. of
body & adipose tissues
in rats converted to
NLD as compared to
those remained
on HFD
The wt. of WAT and
increase in size of
adipocytes inhibited
Sig. decrease in
body wt.
Sig. decrease in fat cell number
& size in both I groups as
compared to their state before
intervention; decrease of FABP
expression in HFD group of I
Mouse
(obese by
HFD)
Mouse (diabetic)
Bofutsushosan
(BOF)
1%/4 wk
C: Placebo
I: Compound
Pine extract
(bark and
needle)
21 d
C: Control
I: Extract
Mouse
(obese
diabetic)
Panax ginseng
berry
150 mg/kg
(ip) 12 d
C: Control
I: Extract
Sig. loss of wt. with a
sig. reduction in food
intake & a very sig.
increase in energy
expenditure & body
temperature
Expression of UCP1 mRNA in
WAT was found but not sig.
Effectively suppressed the
increase of postprandial blood
glucose level by delaying
absorption of diet
Sig. improvement in glucose
tolerance & sig. reduction in
serum insulin levels & plasma
chol levels
MSG: Monosodium glutamate; FABO: Fatty acid β oxidation; STD: Standard diet; LFD: Low fat diet; NLD: Normal diet; HFD: High fat diet; FABP: Fatty
acid binding protein; FFM: Fat free mass; sig.: Significant; AST: Aspartate transaminase; ALT: Alanine transaminase; C: Control; I: Intervention; FAS: Fatty
acid synthetase; UCP: Uncoupling protein; GDP: Guanosine 5' diphosphate; FAS: Fatty acid synthetase; TG: Triglyceride; HDL: High density lipoprotein;
LDL: Low density lipoprotein; FBG: Fasting blood glucose; ip: Intraperitoneal; iv: Intravenous. Caps HT2 is a herbal formulation containing methanolic
extract of selected parts of plants: commiphora mukul; Allium Sativum; Plumbago indica/some carpus anacardium/Hemidesmus indicus/Terminalia
arjuna/Tinospora cordifolia/Withania somnifera ocimum sanctum.
caffeine[9,20] which caused minor adverse effects such
as dry mouth, insomnia, ner vousness, palpitation
and headache. Bofutsushosan [14] caused loose bowel
movements.
ANIMAL STUDIES
Change in body weight and body fat
The majority of animal studies (41 out of 58) showed
significant weight loss or inhibition of weight gain when
supplemented with high fat diets containing extracts
of plants, with or without an efficient decrease in fat
mass[27-85] (Table 2).
Food intake
Clinical trials with Agave tequilana (TEQ) and Dasylirion spp
(DAS)[30], Pomegranate leaf[43], Korean red ginseng[58], Tree
peony[69], Gyeongshang angjeehwan containing a variety
of plants including platycodongrandiflorum, Magnoliaceae
and Ephedra[81], Parasitic loranthus[70], and Panax ginseng
berry[85] showed significant reductions in food intake
or appetite. In studies with Cucurbita moschata[40], Cyperus
rotundus[42], Nomame Herba[66], Acanthopanax senticosus[57] PMF2-OB (a traditional herbal medicine used for the treatment
of obesity in Korea composed of Lycii Fructus), and
several other plants[73], bofu-tsusho-san[79], Galega officinalis[77],
and Oolong tea[67], no change in the amount of food intake
or appetite was observed.
DISCUSSION
In many studies[8-10,12-16,20-23,27,39,73,74,79-81,83], a combination
of plants or compounds containing minerals and or
chemical extracts of plants were investigated and the
scientific names are summarized in Tables 1 and 2. Most
of these studies showed anti-obesity effects such as decreasing body weight in humans or body weight gain in
animals with or without changes in body fat.
Currently available anti-obesity medications attack
the body fat dilemma in three different ways. They can
stimulate metabolism, suppress appetite, affect serotonin,
or they can impede digestion of fat. In this review, we
can categorize the target effects of herbal medicines in
the same way.
Arachis hypogaea[50] decreased body weight gain, liver
triglyceride content and liver size in association with
increased fecal lipid excretion, suggesting an inhibitory
mechanism on lipid absorption. Phillyrin [52], Allium
victorialis[32], Pomegranate leaf[43], Kochia scoparia[46], Panax
japonicus[55], Oolong tea[67], and Aesculus turbinata Blume[71]
also had the same effect.
A decrease in food intake as a result of a decrease
in appetite and an influence on hormonal status was
observed with TEQ and DAS[30], Pomegranate leaf[43],
Korean red ginseng [58], Tree peony [69], Gyeongshang
angjeehwan containing a variety of plants including
platycodon g randif lor um and Magnoliaceae and
www.wjgnet.com
3082 ISSN 1007-9327
CN 14-1219/R
World J Gastroenterol
ephedra[81], and Parasitic loranthus[70], refined Rhubarb[34],
Caralluma fimbriata[19] and Panax ginseng berry[85]. Possible
stimulation of metabolism has been reported as a
mechanism of action for compounds such as Slimax[8],
supplements containing ephedra[9,13,14,20] and Terminalia
arjuna Roxb [11] which showed modification of lipid
metabolism and a reduction in serum lipid levels.
Ephedra known as Ma Huang is a well known
natural product with amphetamine-like stimulation
effects. Although it’s efficacy in weight loss need more
investigations, its adverse effects are well established in
the literature. In this review, nine studies investigated the
effects of ephedra as one of the major components in
the combinations with caffeine[9,13,22] or with several other
plants[14,20,79,81,83] 5 of which were human studies[9,13,14,20,22].
In one study[13], efficient decreases in body weight
and fat were observed with the administration of 210 mg
caffeine and 72 mg ephedra per day for 12 wk with an
improvement in lipid metabolism and blood pressure
without serious adverse effects. In this study, the weight
loss at 12-wk was -3.5 ± 0.6 kg with the test compound
which was significantly (P < 0.02) higher than that of
the placebo. The percentage fat loss shown by DXA was
-7.9% ± 2.9% and -1.9% ± 1.1%, respectively (P < 0.05).
In another study[20], ephedra at a dose of 40 mg/d and
caffeine at a dose of 100 mg/d for a longer time (9 mo)
was found to be more efficient than the previous study in
lowering body fat and weight, improving lipid metabolism
and blood pressure and had no serious adverse effects.
The treatment group lost significantly more body weight
(-7.18 kg) and body fat (-5.33 kg) than the control group
(-2.25 and -0.99 kg, respectively). The difference in
data from these two studies possibly resulted from the
different dosages and duration of interventions.
In a human study[9], a significantly greater weight loss
was observed (-4.0 ± 3.4 kg or 3.5% of baseline) in the
test group vs (-0.8 ± 2.4 kg or 0.09% of baseline) in the
placebo group. Changes were significantly greater for body
fat and percentage of body fat in the active group (-3.5 ±
3.3 kg and -2.1% ± 3.0%) in comparison to the placebo
group (-0.7 ± 2.9 kg and -0.2% ± 2.3%). The tested product also produced several untoward side effects, leading to
some actively treated subjects withdrawing from the study.
Additional long-term studies are needed to elucidate the
effects of chronic treatment. Thus further examinations
in healthy individuals using scientific combinations and
dose/duration adjustments are required.
Four studies[58,59,65,76] investigated different doses and
types of ginseng which is a very popular Chinese herbal
medicine. Ginseng significantly decreased weight gain
and efficiently improved glucose tolerance[59,76].
It has been reported [58] that hormonal influences
can reduce food intake and decrease serum leptin and
neuropeptide Y in the brain hypothalamus although not
significantly. Thus the anti-obesity effect of this plant
requires further investigation.
CQ, a succulent vine native to West Africa and
Southeast Asia, has been used in traditional African and
Ayurvedic medicine for more than a century. Although
some studies have examined other uses for CQ, its
role in fighting against obesity and for symptoms of
July 7, 2009
Volume 15
Number 25
metabolic syndrome has recently attracted interest in
other parts of the world, because of its milder adverse
effects comparing to ephedra. In this review, two studies
focused on this herb [15,26] . CQ in combination with
IG[15] induced marked reductions in body weight and
fat. In addition, a reduction in waist size of 1.0 cm in
the placebo group vs 21.9 cm in the CQ-IG group was
observed.
As we focused on herbal medicines, all dietary
interventions such as the consumption of fruits and
vegetables, whole grains, different types of fibers,
functional food components including omega three
fatty acids or phytochemicals such as flavonoids were
omitted. Lifestyle modification is still the safest and
efficacious method of inducing a persistent weight loss.
In this review, some of the studies were carried out on
subjects who simultaneously received diet and exercise
programs (mentioned as weight loss programs in Tables).
These results demonstrated that specific phytochemical
supplements increase the effectiveness of weight loss
programs and additional significant anti-obesity effects
are observed.
Although few studies mentioned adverse effects, it
should be noted that many serious adverse events which
would have stopped a trial of a pharmaceutical agent
would likely not have been identified by the authors’
search methods. Moreover, important safety issues
including significant adverse events or supplement-drug
interactions relevant to many clinical populations may
not be fully addressed by the trials available for review.
CONCLUSION
Compliance with conventional weight-management
programs, which often include increasing energ y
expenditure via physical activity, is low. It is not surprising
to see the marketing of many new dietary slimming
aids aimed at satisfying the need for palatable (as well as
safe, effective, and therapeutic) options. In accord with
this approach there are numerous investigations on the
effectiveness of medicinal plants as natural supplements
to reduce body weight. In this paper a variety of herbal
supplements had beneficial effects on obesity especially
compounds containing ephedra, CQ, ginseng, bitter
melon (Momordica charantia), and zingiber. Most of the
introduced herbals (Tables 1 and 2) have also been shown
to have antioxidant effects, and with regard to the role of
oxidative stress in the pathophysiology of some diseases
and conditions, their further positive effects may be very
promising[86-95]. Attention to these natural compounds
and further work on the isolation and characterization of
their constituents is highly recommended.
REFERENCES
1
2
World Health Organisation. Obesity: preventing
and managing the global epidemic. Report of a WHO
consultation on obesity. Geneva: World Health Organisation,
1997
Hardeman W, Griffin S, Johnston M, Kinmonth AL, Wareham
NJ. Interventions to prevent weight gain: a systematic review
www.wjgnet.com
Hasani-Ranjbar S et al . Herbal medicines used in the management of obesity
3
4
5
6
7
8
9
10
11
12
13
14
15
16
17
18
19
20
21
of psychological models and behaviour change methods. Int J
Obes Relat Metab Disord 2000; 24: 131-143
Abdollahi M, Afshar-Imani B. A review on obesity and
weight loss measures. Middle East Pharmacy 2003; 11: 6-10
Mahan LK, Escott-Stump S. Krause's food, nutrition, and
diet therapy. 12th ed. Philadelphia: WB Saunders, 2008
Hasani-Ranjbar S, Larijani B, Abdollahi M. A systematic
review of Iranian medicinal plants useful in diabetes
mellitus. Arch Med Sci 2008; 4: 285-292
Liu JP, Zhang M, Wang WY, Grimsgaard S. Chinese herbal
medicines for type 2 diabetes mellitus. Cochrane Database
Syst Rev 2004; CD003642
Liu JP, Yang M, Du XM. Herbal medicines for viral
myocarditis. Cochrane Database Syst Rev 2004; CD003711
Ignjatovic V, Ogru E, Heffernan M, Libinaki R, Lim Y, Ng
F. Studies on the use of 'slimax', a chinese herbal mixture, in
the treatment of human obesity. Pharm Biol 2000; 38: 30-35
Boozer CN, Nasser JA, Heymsfield SB, Wang V, Chen G,
Solomon JL. An herbal supplement containing Ma HuangGuarana for weight loss: a randomized, double-blind trial.
Int J Obes Relat Metab Disord 2001; 25: 316-324
Hoeger WW, Harris C, Long EM, Hopkins DR. Fourweek supplementation with a natural dietary compound
produces favorable changes in body composition. Adv Ther
1998; 15: 305-314
Ziauddin KS, Anwar M, Mannan A, Khan AB. Clinical
efficacy of Terminalia arjuna Roxb. in the management of
hyperlipidaemia. Hamdard Med 2004; 47: 15-18
Abidov MT, del Rio MJ, Ramazanov TZ, Klimenov AL,
Dzhamirze Sh, Kalyuzhin OV. Effects of Aralia mandshurica
and Engelhardtia chrysolepis extracts on some parameters
of lipid metabolism in women with nondiabetic obesity.
Bull Exp Biol Med 2006; 141: 343-346
Greenway FL, De Jonge L, Blanchard D, Frisard M, Smith
SR. Effect of a dietary herbal supplement containing
caffeine and ephedra on weight, metabolic rate, and body
composition. Obes Res 2004; 12: 1152-1157
Hioki C, Yoshimoto K, Yoshida T. Efficacy of bofu-tsushosan, an oriental herbal medicine, in obese Japanese women
with impaired glucose tolerance. Clin Exp Pharmacol Physiol
2004; 31: 614-619
Oben JE, Ngondi JL, Momo CN, Agbor GA, Sobgui CS.
The use of a Cissus quadrangularis/Irvingia gabonensis
combination in the management of weight loss: a doubleblind placebo-controlled study. Lipids Health Dis 2008; 7: 12
Chrubasik C, Maier T, Dawid C, Torda T, Schieber A,
Hofmann T, Chrubasik S. An observational study and
quantification of the actives in a supplement with Sambucus
nigra and Asparagus officinalis used for weight reduction.
Phytother Res 2008; 22: 913-918
Udani J, Singh BB. Blocking carbohydrate absorption and
weight loss: a clinical trial using a proprietary fractionated
white bean extract. Altern Ther Health Med 2007; 13: 32-37
Roongpisuthipong C, Kantawan R, Roongpisuthipong
W. Reduction of adipose tissue and body weight: effect of
water soluble calcium hydroxycitrate in Garcinia atroviridis
on the short term treatment of obese women in Thailand.
Asia Pac J Clin Nutr 2007; 16: 25-29
Kuriyan R, Raj T, Srinivas SK, Vaz M, Rajendran R, Kurpad
AV. Effect of Caralluma fimbriata extract on appetite, food
intake and anthropometry in adult Indian men and women.
Appetite 2007; 48: 338-344
Hackman RM, Havel PJ, Schwartz HJ, Rutledge JC, Watnik
MR, Noceti EM, Stohs SJ, Stern JS, Keen CL. Multinutrient
supplement containing ephedra and caffeine causes weight
loss and improves metabolic risk factors in obese women:
a randomized controlled trial. Int J Obes (Lond) 2006; 30:
1545-1556
Garrison R, Chambliss WG. Effect of a proprietary Magnolia
and Phellodendron extract on weight management: a pilot,
double-blind, placebo-controlled clinical trial. Altern Ther
Health Med 2006; 12: 50-54
22
23
24
25
26
27
28
29
30
31
32
33
34
35
36
37
38
3083
Coffey CS, Steiner D, Baker BA, Allison DB. A randomized
double-blind placebo-controlled clinical trial of a product
containing ephedrine, caffeine, and other ingredients from
herbal sources for treatment of overweight and obesity in
the absence of lifestyle treatment. Int J Obes Relat Metab
Disord 2004; 28: 1411-1419
Preuss HG, Bagchi D, Bagchi M, Rao CV, Dey DK,
Satyanarayana S. Effects of a natural extract of (-)-hydroxycitric
acid (HCA-SX) and a combination of HCA-SX plus niacinbound chromium and Gymnema sylvestre extract on weight
loss. Diabetes Obes Metab 2004; 6: 171-180
Udani J, Hardy M, Madsen DC. Blocking carbohydrate
absorption and weight loss: a clinical trial using Phase 2
brand proprietary fractionated white bean extract. Altern
Med Rev 2004; 9: 63-69
Bhatt AD, Dalal DG, Shah SJ, Joshi BA, Gajjar MN, Vaidya
RA, Vaidya AB, Antarkar DS. Conceptual and methodologic
challenges of assessing the short-term efficacy of Guggulu
in obesity: data emergent from a naturalistic clinical trial. J
Postgrad Med 1995; 41: 5-7
Oben JE, Enyegue DM, Fomekong GI, Soukontoua YB,
Agbor GA. The effect of Cissus quadrangularis (CQR-300)
and a Cissus formulation (CORE) on obesity and obesityinduced oxidative stress. Lipids Health Dis 2007; 6: 4
Wang W, Wang WX, Sun BH, Zhao DZ, Gao P. [Effect of
haidonghua powder(HDHP) on hypothalamic obesity in
rats] Zhongguo Zhongyao Zazhi 2000; 25: 490-492
Jeon WK, Kim JH, Lee HW, Ko BS, Kim HK. Effects of Rhus
verniciflua Stokes (RVS) extract on diet-induced obesity in
C57BL/6 mouse. Kor J Pharmacognosy 2003; 34: 339-343
Alarcon-Aguilar FJ, Zamilpa A, Perez-Garcia MD, AlmanzaPerez JC, Romero-Nuñez E, Campos-Sepulveda EA,
Vazquez-Carrillo LI, Roman-Ramos R. Effect of Hibiscus
sabdariffa on obesity in MSG mice. J Ethnopharmacol 2007;
114: 66-71
Urías-Silvas JE, Cani PD, Delmée E, Neyrinck A, López
MG, Delzenne NM. Physiological effects of dietary fructans
extracted from Agave tequilana Gto. and Dasylirion spp. Br
J Nutr 2008; 99: 254-261
Park YS, Yoon Y, Ahn HS. Platycodon grandiflorum
extract represses up-regulated adipocyte fatty acid binding
protein triggered by a high fat feeding in obese rats. World J
Gastroenterol 2007; 13: 3493-3499
Jongwon C, KyungTae L, WonBae K, KwangKyun P,
WonYun C, JinHa L, SangCheol L, HyunJu J, HeeJuhn
P. Effect of Allium victorialis var. platyphyllum leaves
on triton WR-1339-induced and poloxamer-407-induced
hyperlipidemic rats and on diet-induced obesity rats. Kor J
Pharmacognosy 2005; 36: 109-115
Kobayashi Y, Nakano Y, Kizaki M, Hoshikuma K, Yokoo Y,
Kamiya T. Capsaicin-like anti-obese activities of evodiamine
from fruits of Evodia rutaecarpa, a vanilloid receptor
agonist. Planta Med 2001; 67: 628-633
Jin HM, Jiao DH. [Effect of jiang-zhi jian-fei yao on gastrointestinal movement and adipose cell of abdominal wall]
Zhongguo Zhongxiyi Jiehe Zazhi 1994; 14: 230-231, 198
Kim SJ, Jung JY, Kim HW, Park T. Anti-obesity effects of
Juniperus chinensis extract are associated with increased
AMP-activated protein kinase expression and phosphorylation
in the visceral adipose tissue of rats. Biol Pharm Bull 2008; 31:
1415-1421
Shih CC, Lin CH, Lin WL. Effects of Momordica charantia
on insulin resistance and visceral obesity in mice on high-fat
diet. Diabetes Res Clin Pract 2008; 81: 134-143
Pang J, Choi Y, Park T. Ilex paraguariensis extract
ameliorates obesity induced by high-fat diet: potential role
of AMPK in the visceral adipose tissue. Arch Biochem Biophys
2008; 476: 178-185
Bruno RS, Dugan CE, Smyth JA, DiNatale DA, Koo SI.
Green tea extract protects leptin-deficient, spontaneously
obese mice from hepatic steatosis and injury. J Nutr 2008;
138: 323-331
www.wjgnet.com
3084 39
40
41
42
43
44
45
46
47
48
49
50
51
52
53
54
55
ISSN 1007-9327
CN 14-1219/R
World J Gastroenterol
Lee J, Chae K, Ha J, Park BY, Lee HS, Jeong S, Kim MY,
Yoon M. Regulation of obesity and lipid disorders by
herbal extracts from Morus alba, Melissa officinalis, and
Artemisia capillaris in high-fat diet-induced obese mice. J
Ethnopharmacol 2008; 115: 263-270
Choi H, Eo H, Park K, Jin M, Park EJ, Kim SH, Park JE, Kim
S. A water-soluble extract from Cucurbita moschata shows
anti-obesity effects by controlling lipid metabolism in a high
fat diet-induced obesity mouse model. Biochem Biophys Res
Commun 2007; 359: 419-425
Huang HL, Hong YW, Wong YH, Chen YN, Chyuan JH,
Huang CJ, Chao PM. Bitter melon (Momordica charantia
L.) inhibits adipocyte hypertrophy and down regulates
lipogenic gene expression in adipose tissue of diet-induced
obese rats. Br J Nutr 2008; 99: 230-239
Lemaure B, Touché A, Zbinden I, Moulin J, Courtois D,
Macé K, Darimont C. Administration of Cyperus rotundus
tubers extract prevents weight gain in obese Zucker rats.
Phytother Res 2007; 21: 724-730
Lei F, Zhang XN, Wang W, Xing DM, Xie WD, Su H, Du
LJ. Evidence of anti-obesity effects of the pomegranate leaf
extract in high-fat diet induced obese mice. Int J Obes (Lond)
2007; 31: 1023-1029
Aoki F, Honda S, Kishida H, Kitano M, Arai N, Tanaka
H, Yokota S, Nakagawa K, Asakura T, Nakai Y, Mae
T. Suppression by licorice flavonoids of abdominal fat
accumulation and body weight gain in high-fat diet-induced
obese C57BL/6J mice. Biosci Biotechnol Biochem 2007; 71:
206-214
Oluyemi KA, Omotuyi IO, Jimoh OR, Adesanya OA, Saalu
CL, Josiah SJ. Erythropoietic and anti-obesity effects of
Garcinia cambogia (bitter kola) in Wistar rats. Biotechnol
Appl Biochem 2007; 46: 69-72
Han LK, Nose R, Li W, Gong XJ, Zheng YN, Yoshikawa M,
Koike K, Nikaido T, Okuda H, Kimura Y. Reduction of fat
storage in mice fed a high-fat diet long term by treatment
with saponins prepared from Kochia scoparia fruit.
Phytother Res 2006; 20: 877-882
Goyal RK, Kadnur SV. Beneficial effects of Zingiber
officinale on goldthioglucose induced obesity. Fitoterapia
2006; 77: 160-163
Kishino E, Ito T, Fujita K, Kiuchi Y. A mixture of the
Salacia reticulata (Kotala himbutu) aqueous extract and
cyclodextrin reduces the accumulation of visceral fat mass
in mice and rats with high-fat diet-induced obesity. J Nutr
2006; 136: 433-439
Jayaprakasam B, Olson LK, Schutzki RE, Tai MH, Nair MG.
Amelioration of obesity and glucose intolerance in highfat-fed C57BL/6 mice by anthocyanins and ursolic acid in
Cornelian cherry (Cornus mas). J Agric Food Chem 2006; 54:
243-248
Moreno DA, Ilic N, Poulev A, Raskin I. Effects of Arachis
hypogaea nutshell extract on lipid metabolic enzymes and
obesity parameters. Life Sci 2006; 78: 2797-2803
Galisteo M, Sánchez M, Vera R, González M, Anguera A,
Duarte J, Zarzuelo A. A diet supplemented with husks of
Plantago ovata reduces the development of endothelial
dysfunction, hypertension, and obesity by affecting
adiponectin and TNF-alpha in obese Zucker rats. J Nutr
2005; 135: 2399-2404
Zhao Y, Li F, Yang J, An X, Zhou M. [Effect of phillyrin on
the anti-obesity in nutritive obesity mice] Zhongyaocai 2005;
28: 123-124
Chen Q, Li ET. Reduced adiposity in bitter melon
(Momordica charantia) fed rats is associated with lower
tissue triglyceride and higher plasma catecholamines. Br J
Nutr 2005; 93: 747-754
Han LK, Morimoto C, Yu RH, Okuda H. Effects of Coleus
forskohlii on fat storage in ovariectomized rats. Yakugaku
Zasshi 2005; 125: 449-453
Han LK, Zheng YN, Yoshikawa M, Okuda H, Kimura Y.
Anti-obesity effects of chikusetsusaponins isolated from
56
57
58
59
60
61
62
63
64
65
66
67
68
69
70
71
72
73
74
July 7, 2009
Volume 15
Number 25
Panax japonicus rhizomes. BMC Complement Altern Med
2005; 5: 9
Han LK, Gong XJ, Kawano S, Saito M, Kimura Y, Okuda H.
[Antiobesity actions of Zingiber officinale Roscoe] Yakugaku
Zasshi 2005; 125: 213-217
Cha YS, Rhee SJ, Heo YR. Acanthopanax senticosus extract
prepared from cultured cells decreases adiposity and
obesity indices in C57BL/6J mice fed a high fat diet. J Med
Food 2004; 7: 422-429
Kim JH, Hahm DH, Yang DC, Kim JH, Lee HJ, Shim I.
Effect of crude saponin of Korean red ginseng on highfat diet-induced obesity in the rat. J Pharmacol Sci 2005; 97:
124-131
Yun SN, Moon SJ, Ko SK, Im BO, Chung SH. Wild ginseng
prevents the onset of high-fat diet induced hyperglycemia
and obesity in ICR mice. Arch Pharm Res 2004; 27: 790-796
Junbao Y, Long J, Jiangbi W, Yonghui D, Tianzhen Z,
Songyi Q, Wei L. [Inhibitive effect of Semen Cassiae on the
weight gain in rats with nutritive obesity] Zhongyaocai 2004;
27: 281-284
Kim SO, Yun SJ, Lee EH. The water extract of adlay seed
(Coix lachrymajobi var. mayuen) exhibits anti-obesity
effects through neuroendocrine modulation. Am J Chin Med
2007; 35: 297-308
Kwon CS, Sohn HY, Kim SH, Kim JH, Son KH, Lee JS,
Lim JK, Kim JS. Anti-obesity effect of Dioscorea nipponica
Makino with lipase-inhibitory activity in rodents. Biosci
Biotechnol Biochem 2003; 67: 1451-1456
Lu J, Liu H. [Electron microscope observation on effect of
kudingcha inspissation tea on small intestine villus in the
adiposity rats] Zhongyaocai 1999; 22: 641-642
Yoshikawa M, Shimoda H, Nishida N, Takada M, Matsuda
H. Salacia reticulata and its polyphenolic constituents
with lipase inhibitory and lipolytic activities have mild
antiobesity effects in rats. J Nutr 2002; 132: 1819-1824
Xie JT, Zhou YP, Dey L, Attele AS, Wu JA, Gu M, Polonsky
KS, Yuan CS. Ginseng berry reduces blood glucose and
body weight in db/db mice. Phytomedicine 2002; 9: 254-258
Yamamoto M, Shimura S, Itoh Y, Ohsaka T, Egawa M, Inoue
S. Anti-obesity effects of lipase inhibitor CT-II, an extract
from edible herbs, Nomame Herba, on rats fed a high-fat
diet. Int J Obes Relat Metab Disord 2000; 24: 758-764
Han LK, Takaku T, Li J, Kimura Y, Okuda H. Anti-obesity
action of oolong tea. Int J Obes Relat Metab Disord 1999; 23:
98-105
Pusztai A, Grant G, Buchan WC, Bardocz S, de Carvalho
AF, Ewen SW. Lipid accumulation in obese Zucker rats
is reduced by inclusion of raw kidney bean (Phaseolus
vulgaris) in the diet. Br J Nutr 1998; 79: 213-221
Nagasawa H, Iwabuchi T, Inatomi H. Protection by treepeony (Paeonia suffruticosa Andr) of obesity in (SLN x
C3H/He) F1 obese mice. In Vivo 1991; 5: 115-118
Wang Y, Deng M, Zhang SY, Zhou ZK, Tian WX. Parasitic
loranthus from Loranthaceae rather than Viscaceae potently
inhibits fatty acid synthase and reduces body weight in
mice. J Ethnopharmacol 2008; 118: 473-478
Hu JN, Zhu XM, Han LK, Saito M, Sun YS, Yoshikawa
M, Kimura Y, Zheng YN. Anti-obesity effects of escins
extracted from the seeds of Aesculus turbinata BLUME
(Hippocastanaceae). Chem Pharm Bull (Tokyo) 2008; 56:
12-16
Ohkoshi E, Miyazaki H, Shindo K, Watanabe H, Yoshida
A, Yajima H. Constituents from the leaves of Nelumbo
nucifera stimulate lipolysis in the white adipose tissue of
mice. Planta Med 2007; 73: 1255-1259
Kang M, Oh JW, Lee HK, Chung HS, Lee SM, Kim C, Lee
HJ, Yoon DW, Choi H, Kim H, Shin M, Hong M, Bae H.
Anti-obesity effect of PM-F2-OB, an anti-obesity herbal
formulation, on rats fed a high-fat diet. Biol Pharm Bull 2004;
27: 1251-1256
Mary NK, Babu BH, Padikkala J. Antiatherogenic effect
of Caps HT2, a herbal Ayurvedic medicine formulation.
www.wjgnet.com
Hasani-Ranjbar S et al . Herbal medicines used in the management of obesity
75
76
77
78
79
80
81
82
83
Phytomedicine 2003; 10: 474-482
Wu Y, Ou-Yang JP, Wu K, Wang Y, Zhou YF, Wen CY.
Hypoglycemic effect of Astragalus polysaccharide and its
effect on PTP1B. Acta Pharmacol Sin 2005; 26: 345-352
Xie JT, Wang CZ, Wang AB, Wu J, Basila D, Yuan CS.
Antihyperglycemic effects of total ginsenosides from leaves
and stem of Panax ginseng. Acta Pharmacol Sin 2005; 26:
1104-1110
Palit P, Furman BL, Gray AI. Novel weight-reducing
activity of Galega officinalis in mice. J Pharm Pharmacol 1999;
51: 1313-1319
Oi Y, Kawada T, Shishido C, Wada K, Kominato Y,
Nishimura S, Ariga T, Iwai K. Allyl-containing sulfides in
garlic increase uncoupling protein content in brown adipose
tissue, and noradrenaline and adrenaline secretion in rats. J
Nutr 1999; 129: 336-342
Yoshida T, Sakane N, Wakabayashi Y, Umekawa T, Kondo
M. Thermogenic, anti-obesity effects of bofu-tsusho-san
in MSG-obese mice. Int J Obes Relat Metab Disord 1995; 19:
717-722
He GW, Qu WJ, Fan B, Jing R, He R. The protective effect
of Yi-Qi-Yang-Yin-Ye, a compound of traditional Chinese
herbal medicine in diet-induced obese rats. Am J Chin Med
2008; 36: 705-717
Jeong S, Chae K, Jung YS, Rho YH, Lee J, Ha J, Yoon KH,
Kim GC, Oh KS, Shin SS, Yoon M. The Korean traditional
medicine Gyeongshingangjeehwan inhibits obesity through
the regulation of leptin and PPARalpha action in OLETF
rats. J Ethnopharmacol 2008; 119: 245-251
Park YS, Cha MH, Yoon YS, Ahn HS. Effects of low
calorie diet and Platycodon grandiflorum extract on fatty
acid binding protein expression in rats with diet-induced
obesity. Nutritional Sciences 2006; 8: 3-9
Akagiri S, Naito Y, Ichikawa H, Mizushima K, Takagi
T, Handa O, Kokura S, Yoshikawa T. Bofutsushosan, an
Oriental Herbal Medicine, Attenuates the Weight Gain of
White Adipose Tissue and the Increased Size of Adipocytes
Associated with the Increase in Their Expression of
Uncoupling Protein 1 in High-Fat Diet-Fed Male KK/Ta
84
85
86
87
88
89
90
91
92
93
94
95
3085
mice. J Clin Biochem Nutr 2008; 42: 158-166
Kim YM, Jeong YK, Wang MH, Lee WY, Rhee HI. Inhibitory
effect of pine extract on alpha-glucosidase activity and
postprandial hyperglycemia. Nutrition 2005; 21: 756-761
Attele AS, Zhou YP, Xie JT, Wu JA, Zhang L, Dey L, Pugh
W, Rue PA, Polonsky KS, Yuan CS. Antidiabetic effects of
Panax ginseng berry extract and the identification of an
effective component. Diabetes 2002; 51: 1851-1858
Hasani-Ranjbar S, Larijani B, Abdollahi M. A systematic
review of the potential herbal sources of future drugs
effective in oxidant-related diseases. Inflamm Allergy Drug
Targets 2009; 8: 2-10
Mohseni Salehi Monfared SS, Larijani B, Abdollahi M. Islet
transplantation and antioxidant management: a comprehensive
review. World J Gastroenterol 2009; 15: 1153-1161
Rahimi R, Mozaffari S, Abdollahi M. On the use of herbal
medicines in management of inflammatory bowel diseases:
a systematic review of animal and human studies. Dig Dis
Sci 2009; 54: 471-480
Salari P, Rezaie A, Larijani B, Abdollahi M. A systematic
review of the impact of n-3 fatty acids in bone health and
osteoporosis. Med Sci Monit 2008; 14: RA37-RA44
Rezaie A, Parker RD, Abdollahi M. Oxidative stress
and pathogenesis of inflammatory bowel disease: an
epiphenomenon or the cause? Dig Dis Sci 2007; 52: 2015-2021
Rahimi R, Abdollahi M. A review on the mechanisms
involved in hyperglycemia induced by organophosphorus
pesticides. Pestic Biochem Physiol 2007; 88: 115-121
Kajbaf F, Mojtahedzadeh M, Abdollahi M. Mechanisms
underlying stress-induced hyperglycemia in critically ill
patients. Therapy 2007; 4: 97-106
Rahimi R, Nikfar S, Larijani B, Abdollahi M. A review on
the role of antioxidants in the management of diabetes and
its complications. Biomed Pharmacother 2005; 59: 365-373
Abdollahi M, Larijani B, Rahimi R, Salari P. Role of
oxidative stress in osteoporosis. Therapy 2005; 2: 787-796
Abdollahi M, Ranjbar A, Shadnia S, Nikfar S, Rezaie A.
Pesticides and oxidative stress: a review. Med Sci Monit
2004; 10: RA141-RA147
S- Editor Li LF
www.wjgnet.com
L- Editor Webster JR
E- Editor Zheng XM