ArticlePDF Available

Effect of Bitter Melon Extracts on Lipid Levels in Japanese Subjects: A Randomized Controlled Study

Authors:

Abstract and Figures

Dyslipidemia is exemplified by high levels of low-density lipoprotein cholesterol (LDL-C) and represents a risk factor for cardiovascular diseases and requires therapeutic intervention. Several experimental studies suggest that bitter melon ( Momordica charantia ) improves lipid metabolism in animal models of dyslipidemia and diabetes. This study evaluated the effects of bitter melon extracts on lipid metabolism following a 30-day treatment period in Japanese adults. This randomized, double-blind, placebo-controlled trial included 43 adult volunteers who received either 100 mg of hot-water extracts of bitter melon ( n = 23) or a placebo ( n = 20) three times daily for 30 days. The body weight, blood pressure, and levels of LDL-C and other blood parameters of each subject were measured before and after the study period. The results showed that the intervention group exhibited significantly lower LDL-C levels ( P = 0.02) as compared with the control group, and there were no significant changes in either group in terms of body weight, body mass index, systolic pressure, diastolic pressure, total cholesterol, high-density lipoprotein cholesterol, triglycerides, or blood glucose. These results suggested that bitter melon extracts might effectively lower LDL-C levels in humans and exhibit potential therapeutic value for the management of dyslipidemic conditions.
This content is subject to copyright. Terms and conditions apply.
Research Article
Effect of Bitter Melon Extracts on Lipid Levels in Japanese
Subjects: A Randomized Controlled Study
Hiroki Kinoshita 1,2 and Yasuyuki Ogata 2
1Department of Public Health, Graduate School of Medicine, e University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 1138654, Japan
2Imagine Global Care Corporation, 3-16-12 8th Fl. Roppongi Minato-ku, Tokyo 1060032, Japan
Correspondence should be addressed to Hiroki Kinoshita; hiroki.kinoshita@gmail.com
Received 9 May 2018; Revised 19 October 2018; Accepted 28 October 2018; Published 8 November 2018
A
c
ademicEditor:PratibhaV.Nerurkar
Copyright ©  Hiroki Kinoshita and Yasuyuki Ogata. is is an open access article distributed under the Creative Commons
Attribution License, which permits unrestricted use, distribution, and reproduction in any medium, provided the original work is
properly cited.
Dyslipidemia is exemplied by high levels of low-density lipoprotein cholesterol (LDL-C) and represents a risk factor for
cardiovascular diseases and requires therapeutic intervention. Several experimental studies suggest that bitter melon (Momordica
charantia) improves lipid metabolism in animal models of dyslipidemia and diabetes. is study evaluated the eects of bitter melon
extracts on lipid metabolism following a -day treatment period in Japanese adults. is randomized, double-blind, placebo-
controlled trial included  adult volunteers who received either  mg of hot-water extracts of bitter melon (n=)oraplacebo
(n= ) three times daily for  days. e body weight, blood pressure, and levels of LDL-C and other blood parameters of each
subject were measured beforeand aer the study period. e results showed that the intervention group exhibited signicantly lower
LDL-C levels (P= .) as compared with the control group, and there were no signicant changes in either group in terms of body
weight, body mass index, systolic pressure, diastolic pressure, total cholesterol, high-density lipoprotein cholesterol, triglycerides,
or blood glucose. ese results suggested that bitter melon extracts might eectively lower LDL-C levels in humans and exhibit
potential therapeutic value for the management of dyslipidemic conditions.
1. Introduction
Cardiovascular diseases (CVDs) remain the leading global
cause of death, with the World Health Organization project-
ing that ,, people died due to ischemic heart disease
in  []. Various studies demonstrated that dyslipidemia
is a risk factor for CVD [], with low-density lipoprotein
cholesterol (LDL-C) having a greater eect than high-density
lipoprotein cholesterol (HDL-C) or triglycerides (TGs). e
US National Cholesterol Education Program Adult Treat-
ment Panel III guidelines found that high LDL-C levels
constitute a major risk factor for coronary heart disease and
require clinical intervention [].
Lipid-lowering drugs, such as statins, are used along
with lifestyle interventions to treat high LDL-C levels,
although the absence of clear symptoms is associated with
poor drug adherence []. Furthermore, many patients have
dyslipidemia and borderline dyslipidemia, which results in
large numbers of patients with inadequately managed high
LDL-C levels. Improvements in diet and exercise can be
enhanced by avoiding saturated fats and cholesterol, as well
as consuming greater proportions of plant stanols/sterols
and soluble ber []. Moreover, lower LDL-C levels can be
achieved by consuming greater prop ortions of oats, avocados,
nuts, soybeans, tomatoes, apples, and prunes [–].
Several studies report that bitter melon (Momordica
charantia)canimprovebloodglucoselevelsandlipid
metabolism in animal models of dyslipidemia and diabetes
[, ]. Bitter melon belongs to the Cucurbitaceae family
and is commonly eaten as a vegetable in Asia, Africa, and
Latin America. Additionally, it has been used as a traditional
herbal medicine for treating diabetes in India and China
for ages []. Studies using mouse models suggest that bitter
melon improves glucose and lipid metabolism by activating
the translocation of GLUT to cell membranes in mouse
L myotubes, T-L adipocytes, skeletal muscle tissue, and
the liver, as well as promoting AMP-activated protein kinase
(AMPK) function [–]. Moreover, bitter melon reduces
Hindawi
Evidence-Based Complementary and Alternative Medicine
Volume 2018, Article ID 4915784, 6 pages
https://doi.org/10.1155/2018/4915784
Evidence-Based Complementary and Alternative Medicine
the mRNA levels of 𝛽-hydroxysteroid dehydrogenase type
(𝛽-HSD) in the mouse liver, which reduces its excessive
glucocorticoid activity and involvement in the development
of obesity and insulin resistance []. Furthermore, patients
with recently diagnosed diabetes and who consumed bitter
melon in herbal supplements exhibited decreased plasma
glycated hemoglobin levels and improved TG levels (vs.
baseline) along with a modest eect on hypoglycemia [].
erefore, the authors of that report concluded that bitter
melon helps ameliorate the diabetes-associated risk of CVD.
Several research groups evaluated the lipid-lowering
eect of bitter melon in mouse and rat models of obesity and
diabetes, providing evidence that its use can improve dyslipi-
demia (e.g., levels of TGs and LDL-C) and hyperglycemia [,
]. Additionally, bitter melon extract suppresses SREBPc
[], which is thought to play an important role in the
expression of lipid- constituting enzymes, synthesis of fatty
acids, and accumulation of TGs. Furthermore, pretreatment
of rats with a bitter melon polysaccharide extract reduced
the size of isoproterenol-induced myocardial infarction, as
well as serum levels of total cholesterol, TGs, and LDL-C.
However, this treatment increased the activity of superoxide
dismutase and catalase along with concomitant increases in
proinammatory cytokines and decreases in inammatory
markers, such as nitric oxide [, ]. ese observations
suggest that bitter melon might have a myocardial-protective
eect, although no human study has examined the eects
ofbittermelononhumanlipidmetabolism.erefore,the
present randomized controlled study examined the eect
of bitter melon extract on lipid metabolism in a group of
Japanese adults.
2. Materials and Methods
2.1. Materials. is study used capsules containing bitter
melon extract from Okinawa prefecture or placebo. e bitter
melons were subjected to hot-water extraction and ltration
along with the addition of starch hydrolysate as an excipient
before being autoclaved and dried. Based on our research,
the constituent (derived from bitter gourd) extracted with
this method is considered a type of pectin (a plant cell-wall
constituent). Because the extract has a unique appearance
andtaste,thestudydose(mgofextract,approximately
equivalent to  g of melon) was placed in white capsules that
also contained microcrystalline cellulose, calcium stearate,
and ne silicon dioxide as llers. e placebo capsules were
lled with starch hydrolysate.
2.2. Subjects. For this randomized controlled trial,  healthy
Japanese adults ( men and  women) were recruited
according to the method described by Ursoniu et al. [].
Subject recruitment was coordinated by Huma Corporation
(Minato Ward, Tokyo, Japan). e inclusion criteria were ()
age between  and  years and () willingness to provide
written informed consent to participate aer receiving a
sucient explanation regarding the purpose and procedures
of the study. e exclusion criteria were () receiving contin-
uous drug therapy (e.g., pranlukast hydrate, metformin, and
lipid-lowering drugs), () exhibiting an allergic response to
the study materials, () consuming supplements that might
aect the study parameters (based on the discretion of the
attending physician), () having had digestive organs surgi-
cally removed, () having had the presence or possibility of
pregnancy and/or breast feeding, and () having participated
in another clinical study within the previous  months.
2.3. Study Design and Parameters. e study protocol was
approved by the Ethics committee of Imagine Global Care
Corporation and was pre-registered in the University Hos-
pital Medical Information Network Clinical Trials Reg-
istry (UMIN). All participants provided written
informed consent, and the study was performed in accor-
dance with the  Declaration of Helsinki.
e subjects were randomly assigned. is was specif-
ically done by placing participants in either the control
group or the intervention group using a computer-generated
randomized number table. e subjects, study doctor, and
data analyzer were blinded to subject assignments. Starting
on day , the subjects consumed three capsules daily ( mg)
for  days, with each capsule containing either bitter melon
extract or placebo. Blood testing and measurements of weight
and blood pressure were performed on days  (the rst visit)
and  (the second visit).
Subjects were instructed to not consume sweet food or
drinks aer : on the night before blood testing. e
values of total cholesterol, HDL-C, LDL-C, TGs, glucose, and
glycated hemoglobin were also determined. All biochemical
tests were conducted by LSI Medience Corporation (Tokyo,
Japan) using Stacia, an automated clinical testing machine.
2.4. Statistical Analysis. Pearson's chi-square test was used
to compare the male: female ratios between the control and
intervention groups. An unpaired ttest was used to compare
baseline and post intervention body weight, blood pressure,
and biochemical parameters between the control and inter-
ventiongroupsaswellasthechangesinthesevaluesforeach
group. Confounding eects were evaluated using multiple
regression analysis (forced entry method), with the change
in LDL-C levels used as the dependent variable, and sex, age,
and baseline body mass index used as independent variables.
e Japan Atherosclerosis Society denes hypercholes-
terolemia as  mg/dL LDL-C and borderline hypercholes-
terolemia as – mg/dL LDL-C. erefore, we addition-
ally analyzed subjects whose baseline LDL-C was equal to
or exceeded  mg/dL and determined the dierence in
changes in LDL-C between the control and intervention
subjects. Dierences were considered statistically signicant
at P<., and all analyses were performed using SPSS
soware (v.; SPSS, Inc., Chicago, IL, USA).
3. Results
Among the  recruited subjects, two were excluded because
they were receiving continuous drug therapy (pranlukast
hydrate and metformin), and two other were excluded aer
failing to attend the second visit. ere was no signicant
dierence in the male: female ratios of the control (n=)
and intervention (n= ) groups (Table ). Table  shows the
Evidence-Based Complementary and Alternative Medicine
T:Numberandageofmaleandfemalesubjectsinthecontrolandinterventiongroups.
Control group Intervention group Overall
No. Age No. Age No. Age
Male . ±.  . ±.  . ±.
Female  . ±.  . ±.  . ±.
Tot a l .±.      .  ±.      .  ±. 
T : Change in metabolic parameters aer intervention with bitter melon.
Control group (n = ) Intervention group (n = ) p-value
Body weight Baseline . ±. . ±. .
Post intervention . ±. . ±. .
Change -. ±. -. ±. .
Body mass index, kg/m2Baseline . ±. . ±. .
Post intervention . ±. . ±. .
Change -. ±. -. ±. .
Systolic pressure, mmHg Baseline . ±. . ±. .
Post intervention . ±. . ±. .
Change -. ±. -. ±. .
Diastolic pressure, mmHg Baseline . ±. . ±. .
Post intervention . ±. . ±. .
Change . ±.   .  ±. .
Total cholesterol, mg/dL Baseline . ±. . ±. .
Post intervention . ±. . ±. .
Change . ±. . ±. .
LDL-C, mg/dL Baseline . ±. . ±. .
Post intervention . ±. . ±. .
Change . ±. -. ±. .
HDL-C, mg/dL Baseline . ±. . ±. .
Post intervention . ±. . ±. .
Change . ±. -. ±. .
Triglycerides, mg/dL Baseline . ±. . ±. .
Post intervention . ±. . ±. .
Change . ±. . ±. .
Glucose, mg/dL Baseline . ±. . ±. .
Post intervention . ±. . ±. .
Change -. ±. .±. .
Glycated hemoglobin, % Baseline . ±. . ±. .
Post intervention . ±. . ±. .
Change ±. ±. .
changes in metabolic parameters of the subjects from baseline
to post intervention. No signicant dierences were obser ved
between the control and intervention groups at baseline. e
intervention group showed signicantly decreased LDL-C
levels at the second visit (-. ±. mg/dL) as compared
with the control group (+ ±., P= .). ere was
no signicant dierence in changes in body weight, BMI,
blood pressure, total cholesterol, HDL-C, TGs, glucose, and
glycated hemoglobin between the control and intervention
groups. Multiple regression analysis revealed that the inter-
vention group had a signicantly increased likelihood of
lower LDL-C levels aer adjusting for sex, age, and baseline
body mass index (Table ). Baseline LDL-C  mg/dL was
observed for  subjects in the control group and  subjects
in the intervention group compared with  subjects in each
group at the second visit. Furthermore, baseline LDL-C 
mg/dL was observed for  subjects in the control group
and  subjects in the intervention group compared with 
subjects in each group at the second visit. Among the subjects
with baseline LDL-C equal to or exceeding  mg/dL, the
mean changes were +. ±. mg/dL in the control group
and -. ±. mg/dL in the intervention group (P= .).
Figure  shows the changes in LDL-C levels in all subjects in
the control and intervention groups and those with baseline
LDL-C  mg/dL.
4. Discussion
Bitter melon has been used in traditional Indian and Chinese
medicine since ancient times for the treatment of various
Evidence-Based Complementary and Alternative Medicine
T : Multiple linear regression analysis of factors related to changes in LDL-C.
𝐵SE 𝛽𝑃
Sex (male = ; female = ) –. . –. .
Age –. . –. .
Group (control = ; intervention = ) –. . –. 0.006
Body mass index –. . –. .
R2= .. Bold values represent signicant dierences.
𝛽: standardized regression coecient; B: unstandardized regression coecient; LDL-C: low-density lipoprotein cholesterol; SE: standard error.
All subjects
20
10
0
1
10
20
Baseline LDL‐C ≥ 120
20
10
0
1
10
20
F : Changes in LDL-C. p=. for both all subjects and those with baseline LDC-C .
ailments, including gastrointestinal complaints, constipation,
dermatitis, cough, and diabetes []. However, several clinical
studies have failed to show clear pharmacological eects.
Our research revealed that the component extracted from
bitter melon in this study was one form of pectin. It has long
been known that soluble ber, including pectin, eectively
lowers LDL-C. Brown et al. conducted a meta-analysis of
four types of soluble ber (pectin, oat bran, guar gum, and
psyllium) and found that all reduce LDL-C to the same extent
[]. Namely, intake of  g of soluble ber per day reduced
LDL-C by roughly  mg/dL. ough soluble ber also lowers
HDL-C,thedecreaseisextremelyslight,andthereisnoeect
on TGs. is study was consistent with the results of previous
research on soluble ber, demonstrating that although bitter
melon extract reduced LDL-C, there was no eect on HDL-
C or TGs. e daily dose of bitter melon extract used in
the present study was  mg—a small amount compared
with that in most previous studies, in which quantities were
in grams—suggesting that soluble ber from bitter melon
extract is eective at reducing LDL-C even at a lower dose.
ere are several possible mechanisms involved in the
reductioninHDL-Cbysolubleber.First,thetheorythat
ber promotes bile acid excretion, thereby reducing choles-
terol, has been advocated for years []. e idea is that
because blood cholesterol is used in the synthesis of bile
acid, highly viscous soluble ber adheres to and helps excrete
bile acid enveloping cholesterol. However, some scholars
argue that the amount of bile acid excretion is insucient
to explain the reduction in cholesterol []. It was further
reported that soluble ber increases the number of apo B/E
receptors, which bind to LDL-C and accelerate the LDL-
C metabolic turnover rate [, ]. Eects from improved
insulin sensitivity [] and inhibition of cholesterol synthesis
in the liver due to the formation of short-chain fatty acids
through fermentation of ber in the large intestine have also
been indicated [, ]. Although we observed signicant
decrease in LDL-C levels in the present study, there was no
change in blood glucose levels, suggesting that reduced LDL-
C levels might not be mediated by insulin resistance.
Previous studies evaluated the eects of bitter melon on
dyslipidemia using rats. Bitter melon treatment of diabetic
rats normalized the increase in nonesteried cholesterol,
TGs, LDL, and phospholipids [, , ]. Additionally,
increased mitochondrial biogenesis could be a pathway
associated with increased lipid metabolism and utilization,
with several genes, including PPAR𝛾,involvedinthereg-
ulation of this process [, ]. Bitter melon treatment of
rats increased the expression of PPAR𝛾coactivator (PGC𝛼)
and fatty acid-binding protein , and in this context, PGC
family members inuence hepatic metabolism by stimulating
mitochondrial biogenesis and respiration in several types
of cells while also altering biological pathways involved in
oxidative metabolism []. In rat-based studies, test animals
that received a high-fat diet and bitter melon extract displayed
reduced levels of plasma TGs, cholesterol, and free fatty acids
[]. ese results suggest that bitter melon extract might
improve dyslipidemia in humans.
is study had several limitations. First, the small sample
size of the study suggests that caution is needed in gener-
alizing the applicability of the study results. Using puried
Evidence-Based Complementary and Alternative Medicine
samples of bitter melon extract might improve the interest
of volunteers in enrolling in follow-up studies. Second, the
dose of  mg per day used in this study may be too low
compared with that in previous studies, in which several
grams were administered daily. In addition, the administered
dose in this study was not adjusted for individual body weight
and was unrelated to the dose used in tradomedical use of
bitter melon. Previous studies used doses of  mg/kg []
and between  mg/kg and , mg/kg [, ].
erefore, it is important to perform additional studies
to determine the mechanisms through which bitter melon
lowers LDL-C levels, as well as its eects on other indices
related to the lipid prole.
5. Conclusion
e water-soluble extract of bitter melon signicantly
decreased LDL-C levels as compared with the control
(placebo) group in humans. erefore, bitter melon might be
useful in reducing the risks of cholesterol-mediated diseases,
including CVDs.
Data Availability
e data used to support the ndings of this study are
included within the article.
Disclosure
Editage provided editorial support in the form of medical
writing, assembling tables, creating high-resolution images
based on the authors’ detailed directions, collating author
comments, copyediting, fact checking, and referencing.
Conflicts of Interest
Hiroki Kinoshita is a board member of Imagine Global Care
Corporation, and Yasuyuki Ogata is an employee of the
company.
References
[] World Health Organization, Geneva, , http://www.who
.int/mediacentre/factsheets/fs/en/.
[] R.H.Eckel,J.M.Jakicic,andJ.D.Ard,“AHA/ACCguide-
line on lifestyle management to reduce cardiovascular risk: a
report of the American College of Cardiology/American Heart
Association Task Force on Practice Guidelines,Circulation,vol.
, no. , supplement , pp. S–S, .
[] Expert Panel on Detection Evaluation and Treatment of High
Blood Cholesterol in Adults, “Executive summary of the third
report of the National Cholesterol Education Program (NCEP)
expert panel on detection, evaluation, and treatment of high
blood cholesterol in adults (adult treatment panel III),Journal
of the American Medical Association,vol.,no.,pp.
, .
[]H.Caspard,A.K.Chan,andA.M.Walker,“Compliance
with a statin treatment in a usual-care setting: Retrospective
database analysisover  years aer treatment initiation in health
maintenance organization enrollees with dyslipidemia,Clinical
erapeutics,vol.,no.,pp.,.
[] H.V.T.Ho,J.L.Sievenpiper,A.Zurbauetal.,“eeectofoat
𝛽-glucan on LDL-cholesterol, non-HDL-cholesterol and apoB
for CVD risk reduction: A systematic review and meta-analysis
of randomised-controlled trials,British Journal of Nutrition,
vol. , no. , pp. –, .
[] X.Liu,A.M.Hill,S.G.Westetal.,“AcutePeanutConsumption
Alters Postprandial Lipids and Vascular Responses in Healthy
Overweight or Obese Men,Journal of Nutrition,vol.,no.,
pp.,.
[] S. Peou, B. Milliard-Hasting, and S. A. Shah, “Impact of
avocado-enriched diets on plasma lipoproteins: A meta-
analysis,” Journal of Clinical Lipidology,vol.,no.,pp.,
.
[] E.Mah,J.A.Schulz,V.N.Kadenetal.,“Cashewconsumption
reduces total and LDL cholesterol: A randomized, crossover,
controlled-feeding trial,American Journal of Clinical Nutrition,
vol. , no. , pp. –, .
[] A. R. Gonciulea and D. E. Sellmeyer, “e eect of dietary
protein source on serum lipids: Secondary data analysis from
arandomizedclinicaltrial,Journal of Clinical Lipidology,vol.
, no. , pp. –, .
[] H. M. Cheng, G. Koutsidis, J. K. Lodge, A. Ashor, M. Siervo,
and J. Lara, “Tomato and lycopene supplementation and cardio-
vascular risk factors: A systematic review and meta-analysis,”
Atherosclerosis, vol. , pp. –, .
[]S.C.Chai,S.Hooshmand,R.L.Saadat,M.E.Payton,K.
Brummel-Smith, and B. H. Arjmandi, “Daily Apple versus
Dried Plum: Impact on Cardiovascular Disease Risk Factors in
Postmenopausal Women,Journal of the Academy of Nutrition
and Dietetics, vol. , no. , pp. –, .
[] M. A. Alam, R. Uddin, N. Subhan et al., “Benecial role of bitter
melon supplementation in obesity and related complications in
metabolic syndrome,Journal of Lipids, vol. ,  pages, .
[] M. J. Tan, J. M. Ye, and N. Turner, “Antidiabetic activities
of triterpenoids isolated from bitter melon associated with
activation of the AMPK pathway,Chemistry & Biology,vol.,
no. , pp. –, .
[] P. Chaturvedi, S. George, M. Milinganyo, and Y. B. Tripathi,
“Eect of Momordica charantia on lipid prole and oral glucose
tolerance in diabetic rats, Phytotherapy Research,vol.,no.,
pp. –, .
[] M. G. Sridhar, R. Vinayagamoorthi, V. A. Suyambunathan,
Z. Bobby, and N. Selvaraj, “Bitter gourd (Momordica charan-
tia) improves insulin sensitivity by increasing skeletal muscle
insulin-stimulated IRS- tyrosine phosphorylation in high-fat-
fed rats,” British Journal of Nutrition,vol.,no.,pp.,
.
[] I. U. Rahman, R. U. Khan, K. U. Rahman, and M. Bashir,
“Lower hypoglycemic but higher antiatherogenic eects of
bitter melon than glibenclamide in type  diabetic patients,
Nutrition Journal,vol.,no.,.
[] Q. Chen and E. T. S. Li, “Reduced adiposity in bitter melon
(Momordica charantia) fed rats is associated with lower tissue
triglyceride and higher plasma catecholamines,British Journal
of Nutrition,vol.,no.,pp.,.
[] F.Bano,N.Akthar,andH.Naz,“Eectoftheaqueousextracts
of Momordica charantia on body weight of rats,Journal of Basic
Applied Science,v
ol.,pp.,.
[] M. Raish, “Momordica charantia polysaccharides ameliorate
oxidative stress, hyperlipidemia, inammation, and apoptosis
Evidence-Based Complementary and Alternative Medicine
during myocardial infarction byinhibiting the NF-𝜅Bsignaling
pathway,International Journal of Biological Macromolecules,
vol. , pp. –, .
[] S. Ursoniu, A. Sahebkar, M.-C. Serban, and M. Banach, “Lipid
prole and glucose changes aer supplementation with astax-
anthin: A systematic review and meta-analysis of randomized
controlled trials,Archives of Medical Science,vol.,no.,pp.
–, .
[] J. K. Grover and S. P. Yadav, “Pharmacological actions and
potential uses of Momordica charantia: a review,Journal of
Ethnopharmacology, vol. , no. , pp. –, .
[]L.Brown,B.Rosner,W.W.Willett,andF.M.Sacks,
“Cholesterol-lowering eects of dietary ber: a meta-analysis,
American Journal of Clinical Nutrition,vol.,no.,pp.,
.
[] J. W. Anderson and J. Tietyen-Clark, “Dietary Fiber: Hyperlipi-
demia, Hypertension, and Coronary Heart Disease,American
Journal of Gastroenterology,vol.,no.,pp.,.
[] Federation of American Societies for Experimental Biology,
Physiological effects and health consequences of dietary fiber,US
Department of Health and Human Services, Washington DC,
Wash , U S A , .
[] M. Vergara-Jimenez, K. Conde, S. K. Erickson, and M. L.
Fernandez, “Hypolipidemic mechanisms of pectin and psyllium
in guinea pigs fed high fat-sucrose diets: Alterations on hepatic
cholesterol metabolism, Journal of Lipid Research,vol.,no.,
pp. –, .
[] H. Shen, L. He, R. L. Price, and M. L. Fernandez, “Dietary
soluble ber lowers plasma LDL cholesterol concentrations by
altering lipoprotein metabolism in female guinea pigs,Journal
of Nutrition,vol.,no.,pp.,.
[] B. O. Schneeman, “Dietary Fiber and Gastrointestinal Func-
tion,Nutrition Reviews,vol.,no.,pp.,.
[] P. M. Nishina and R. A. Freedland, “e eects of dietary ber
feeding on cholesterol metabolism in rats,Journal of Nutrition,
vol. , no. , pp. –, .
[] B. O. Schneeman and D. Gallaher, “Eects of Dietary Fiber
on Digestive Enzyme Activity and Bile Acids in the Small
Intestine,” Proceedings of the Society for Experimental Biology
and Medicine,vol.,no.,pp.,.
[] I. Ahmed, M. S. Lakhani, M. Gillett, A. John, and H.
Raza, “Hypotriglyceridemic and hypocholesterolemic eects
of anti-diabetic Momordica charantia (karela) fruit extract in
streptozotocin-induced diabetic rats,Diabetes Research and
Clinical Practice, vol. , no. , pp. –, .
[] P. Chaturvedi and S. George, “Momordica charantia maintains
normal glucose levels and lipid proles and prevents oxidative
stress in diabetic rats subjected to chronic sucrose load, Journal
of Medicinal Food,vol.,no.,pp.,.
[] P. Puigserver and B. M. Spiegelman, “Peroxisome proliferator-
activated receptor-𝛾coactivator 𝛼(PGC-𝛼): transcriptional
coactivator and metabolic regulator,Endocrine Rev iews,vol.,
no.,pp.,.
[] R.B.Vega,J.M.Huss,andD.P.Kelly, “ecoactivatorPGC-
 cooperates with peroxisome proliferator-activated receptor
𝛼in transcriptional control of nuclear genes encoding mito-
chondrial fatty acid oxidation enzymes,Molecular and Cellular
Biology,vol.,no.,pp.,.
[] B. N. Finck and D. P. Kelly, “PGC- coactivators: inducible
regulators of energy metabolism in health and disease,e
Journal of Clinical Investigation,vol.,no.,pp.,
.
[] A.Fuangchan,P. Sonthisombat, T.Seubnukarnetal.,“Hypo-
glycemic eect of bitter melon compared with metformin in
newly diagnosed type  diabetes patients,Journal of Ethnophar-
macology,v
ol.,no.,pp.,.
... In the full-text section stage, most articles were excluded due to the lack of placebo control (57,58), lack of blinding (59), lack of numerically reported results (32,60,61), and since other plant parts (leaves) than M. charantia fruits were used as study (59,(62)(63)(64)(65). We identified nine trials eligible for our review and meta-analysis (33,(66)(67)(68)(69)(70)(71)(72)(73). The selection process is presented in Figure 1 (47). ...
... We analyzed nine randomized placebo-controlled trials (33,(66)(67)(68)(69)(70)(71)(72)(73). Table 1 contains the summary of the main study and patient characteristics. ...
... Random sequence generation was described in six studies (33,(67)(68)(69)(70)72); however, the measures taken to ensure allocation concealment were given in only one trial (67). Performance and detection biases were unclear in five studies (33,(68)(69)(70)(71) because the authors of these studies failed to report whether the intervention and the comparator were identical in size, shape, color, and odor; and it remained unclear whether the outcomes were assessed in a blinded manner or not. ...
Article
Full-text available
Several studies have shown that Momordica charantia L. (Cucurbitaceae, bitter melon) has beneficial effects on metabolic syndrome (MetS) parameters and exerts antidiabetic, anti-hyperlipidemic, and anti-obesity activities. Since the findings of these studies are contradictory, the goal of this systematic review and meta-analysis was to assess the efficacy of bitter melon in the treatment of metabolic syndrome, with special emphasis on the anti-diabetic effect. Embase, Cochrane, PubMed, and Web of Science databases were searched for randomized controlled human trials (RCTs). The meta-analysis was reported according to the PRISMA statement. The primary outcomes of the review are body weight, BMI, fasting blood glucose, glycated hemoglobin A1c, systolic blood pressure, diastolic blood pressure, serum triglyceride, HDL, LDL, and total cholesterol levels. Nine studies were included in the meta-analysis with 414 patients in total and 4-16 weeks of follow-up. In case of the meta-analysis of change scores, no significant effect could be observed for bitter melon treatment over placebo on fasting blood glucose level (MD = −0.03; 95% CI: −0.38 to 0.31; I 2 = 34%), HbA1c level (MD = −0.12; 95% CI: −0.35 to 0.11; I 2 = 56%), HDL (MD = −0.04; 95% CI: −0.17 to 0.09; I 2 = 66%), LDL (MD = −0.10; 95% CI: −0.28 to 0.08; I 2 = 37%), total cholesterol (MD = −0.04; 95% CI: −0.17 to 0.09; I 2 = 66%,), body weight (MD = −1.00; 95% CI: −2.59-0.59; I 2 = 97%), BMI (MD = −0.42; 95% CI: −0.99-0.14; I 2 = 95%), systolic blood pressure (MD = 1.01; 95% CI: −1.07-3.09; I 2 = 0%) and diastolic blood pressure levels (MD = 0.24; 95% CI: −1.04-1.53; I 2 = 0%). Momordica treatment was not associated with a notable change in ALT, AST, and creatinine levels compared to the placebo, which supports the safety of this plant. However, the power was overall low and the meta-analyzed studies were also too short to reliably detect long-term metabolic effects. This highlights the need for additional research into this plant in carefully planned clinical trials of longer duration.
... [8][9][10][11][12] Previous clinical trials have reported the effect of MC administration on BW, BMI, WC, and fat percentage as secondary outcomes in different populations with contradictory results. [13][14][15][25][26][27][28] To the best of our knowledge, this is the first study that evaluates the effect of MC administration on anthropometric measures in patients with obesity. Findings of this study showed after 12 weeks of MC administration, and reductions in BW, BMI, WC, and body fat percentage were observed; however, these differences were not statistically significant. ...
... 25 Nonsignificant differences were found in BW and BMI after MC administration (300 mg of MC extract) for 30 days in healthy patients. 26 Similarly, no significant reduction in BW (2000 and 4000 mg/day of MC fruit powder) was reported in T2DM patients after the administration of MC for 10 weeks. 27 BMI was also not modified in another clinical trial conducted in T2DM patients after MC administration for 3 months (3000 mg/day of MC fruit and seed powder). ...
Article
The aim of this study was to evaluate the effect of Momordica charantia (MC) administration on anthropometric measures in patients with obesity. A randomized, double-blind, placebo-controlled pilot clinical trial was carried out in 24 patients with obesity. Twelve patients randomly received MC (2000 mg/day) for 12 weeks, and 12 patients received placebo. Body weight (BW), body mass index (BMI), waist circumference (WC), body fat percentage, as well as clinical and laboratory determinations, were evaluated before and after the intervention. Results showed that while reductions in BW, BMI, WC, and body fat percentage were observed in the MC group, these differences did not reach statistical significance. Significant decreases in triglycerides (TG) (1.9 ± 0.6 mM vs. 1.7 ± 0.7 mM, P ≤ .05) and very low-density lipoprotein (VLDL) (0.4 ± 0.1 mM vs. 0.3 ± 0.1 mM, P ≤ .05) levels were found after the intervention with MC. In contrast, significant increases in BW (83.0 ± 10.7 kg vs. 84.6 ± 9.1 kg, P ≤ .05) and BMI (31.9 ± 1.5 kg/m2 vs. 33.0 ± 1.3 kg/m2, P ≤ .05) were observed in the placebo group. In conclusion, no significant reductions in BW, BMI, WC, and body fat percentage were observed after MC administration; however, MC significantly decreased TG and VLDL levels. The protocol was registered at ClinicalTrials.gov with the identifier NCT04916379.
... Indeed, higher levels of triglycerides measured in a fasting specimen indicate a lack of clearance or over-production; it could increase the risk of developing cardiovascular disease. The decrease in LDL cholesterol level led by Momordica foetida extract confirms the results of Yanmei et al. [29] or Hiroki and Yasuyuki [30] which showed that a plant of the same genus Momordica charantia is effective against lipid metabolism disorder associated with an increased blood level in Low-Density Lipoprotein (LDL). Serum urea, serum and urinary creatinine, as well as urinary protein levels are used to evaluate the kidney's function [31]. ...
... Even though the differences in the lipid profile after the intervention with MC were not significant, the MC group exhibited lower LDL-c levels compared with the control group (P = .02). 44 The effect of MC on the lipid profile has also been reported as a secondary objective in several clinical trials in patients with T2DM and MetS. In the study conducted by Rahman et al. in T2DM patients, significant differences in LDL-c and TG concentrations were found in the group that received 4 g/day of MC (group II) for 10 weeks (154.0 ...
Article
The global rise in the prevalence of metabolic diseases such as diabetes, obesity, and dyslipidemia is a serious public health issue. The search for safe and effective complementary and alternative therapies to treat metabolic disorders is a key field of research. Momordica charantia (MC) is a tropical and subtropical vine of the Cucurbitaceae family used as a medicinal plant since ancient times. Although MC has been widely studied for its hypoglycemic potential, hypolipidemic and antiobesity effects have also been reported in preclinical studies and clinical trials. This study aims to review the metabolic effects of MC reported in clinical trials as well as its mechanisms of action.
... Besides, multiple reports have expounded the hypolipidemic activity of Momordica saponin [43], MC ethanol extract [32], and MC leaf extract [119]. A clinical RCT also confirms the improvement of dyslipidemia by bitter melon hot water extract [120]. All of the above indicate that MC has antiatherosclerosis potential. ...
Article
Full-text available
In recent years, many studies of Momordica charantia (MC) in the treatment of diabetes mellitus (DM) and its complications have been reported. This article reviewed the effect and mechanism of MC against diabetes, including the results from in vitro and in vivo experiments and clinical trials. The common side effects of MC were also summarized. We hope that it might open up new ideas for further mechanism exploration and clinical application as well as provide a scientific theoretical basis for the development of drugs or foods derived from MC.
Chapter
Momordica charantia L. (MC, bitter melon) is a cultivated plant from the family Cucurbitaceae. Regarding metabolomics and phytochemical studies, it has phenolic compounds, terpenoids, saponins, peptides and proteins, and polysaccharides as main constituents with pharmacological effects. Preclinical and clinical studies exhibited numerous biological activities attributed to MC or its constituents. Antidiabetic, cardioprotective, antidyslipidemia, antiobesity hypotensive, antioxidant, anti-inflammatory, hepatoprotective, renoprotective, neuroprotective, anticancer antiviral, antibacterial, antifungal, anthelmintic, antimalarial, and wound healing are significant beneficial properties of MC and its ingredients. Although its safety and toxicity are not vastly studied in clinical trials, some adverse clinical manifestations have been reported afterward its consumption. Modification of its bioavailability by fabrication of nanotechnology-based formulations and conducting more clinical trials for investigation of its efficacy and toxicity are the future prospects.KeywordsCucurbitaceaePhytochemicals Momordica charantia Bitter melonBitter gourdPharmacological applicationsChemical components
Chapter
Extensive research has provided much scientific evidence on the nutrient compositions and beneficial medicinal effects of the ash gourd, bottle gourd, and bitter gourd and to a lesser extent the snake gourd, ivy gourd, pointed gourd, and ridge gourd. These gourds have been found to possess several functional components that provide beneficial effects on health. Unfortunately, research on the ridge gourd, snake gourd, ivy gourd, and pointed gourd is limited. Therefore further research is needed to validate the beneficial effects of these gourds’ functional components on our health. The fat content is low in gourds, contributing less than 0.3% by weight of edible portion. Along with macronutrients, proteins, carbohydrates, and fibers, gourds also contain vitamins and minerals, which are the essential nutrients found in all the gourds. Flavonoids and polyphenolics are also protective phytochemicals that are present in all the gourds. These phytochemicals in plants are generated to fight environmental factors including insects and infecting agents: bacteria, viruses, and fungi. Recently, these agents have been found to be protective because their phytochemicals can inhibit oxidative stress and inflammation, which appear to be much more pronounced in bitter gourd. Randomized, controlled trials have indicated that consumption of gourd vegetables can decrease blood glucose, blood lipids, blood pressure, and body weight and can inhibit cancer growth. Gourd vegetables, in particular bitter gourd and bottle gourd, have also been found to be protective against gastric ulcer, bacterial and viral infections, arthritis, diabetes, hypertension, and heart diseases. Bitter gourd has been studied by various researchers widely, showing potential antidiabetic, antiulcerogenic, antimutagenic, antioxidant, antitumor, antilipolytic, analgesic, abortifacient, antiviral, hypoglycemic, and immunomodulatory effects. Flavonoids and polyphenolics and fiber can interact with the vitamins and minerals and the gut microbiome that are required by the body to normalize physiological functions. This review describes the nutritional benefits, composition, and medicinal effects of three selected gourds, namely, ash, bottle, and bitter gourds, in health and diseases with particular reference to the treatment of non-communicable diseases.
Article
Full-text available
Cardiovascular disease continues to rise at an alarming rate, and research focuses on possible therapies to reduce the risk and slow down its progression. Several epidemiological studies have indicated that dietary modifications, such as increased consumption of fruits and vegetables play an important role in reducing cardiovascular disease risk factors. Food sources rich in antioxidants, anti-inflammatory, hypolipidemic, and hypoglycemic properties are thought to ameliorate the progression of cardiovascular disease and serve as a potential treatment mode. Many in vivo and in vitro studies using turmeric, cinnamon, mango, blueberries, red wine, chocolate, and extra virgin olive oil have demonstrated significant improvements in cholesterol profiles, toxic reactive oxygen species, inflammation, obesity, and hypertension. In this review, we summarize recent evidence on the cardioprotective effect of different food groups, outline their potential mechanisms involved in slowing down the progression of cardiovascular disease, and highlight the beneficial effects associated with increased consumption.
Chapter
Momordica charantia (bitter gourd) is utilized as traditional medicinal plant and food in Indo-China and Southeast Asia. This nutrient plant-based food contains abundant of bioactive components such as polypeptide, minerals vitamins, alkaloids, flavonoids, isoflavones, terpenes, anthroquinones, and glucosinolates. In the present study, physicochemical properties, nutritional values, and health promoting phytochemicals, as well as value added products of bitter gourd are described. Majority of the bioactive compounds in bitter gourd confer bitter taste. A large number of value-added products can be prepared from bitter gourd such as bitter gourd juice, slices, pickle, dried rings, and chips. These valued products in addition to being healthy are more palatable than raw fruit, thus increasing consumption of this bitter fruit. Hence, besides having the health-enhancing properties, it might be considered proficient option in value-added foodstuffs.
Article
Full-text available
Background: Optimizing plasma lipoproteins is the primary goal of pharmacotherapy and diet interventions in people at risk for cardiovascular diseases. Avocados offer a rich source of monounsaturated fat and may pose beneficial effects on the lipid profile. Objective: We aimed to perform a meta-analysis of randomized clinical trials assessing the impact of avocados on TC, low-density lipoprotein cholesterol (LDL-C), high-density lipoprotein cholesterol, and/or triglycerides (TG). Methods: We searched PUBMED, Cumulative Index to Nursing and Allied Health Literature, Index to Nursing and Allied Health Literature, and the Cochrane Database of Systemic Reviews from their inception to February 2015. The weighted mean difference from baseline was calculated for all endpoints. Subgroup analyses were performed to assess heterogeneity, and funnel plots inspected to assess publication bias. Results: Ten unique studies (n = 229) were included. Avocado consumption significantly reduced TC, LDL-C, and TG by -18.80 mg/dL (95% confidence interval [CI], -24.56 to -13.05; I(2), 46.9%), -16.50 mg/dL (95% CI, -22.91 to -10.10; I(2), 72.5%), -27.20 mg/dL (95% CI, -44.41 to -9.99; I(2), 91.1%) respectively. High-density lipoprotein cholesterol decreased nonsignificantly by -0.18 mg/dL (95% CI, -3.23 to 2.88; I(2), 84.8%). Conclusion: Avocado-substituted diets significantly decrease TC, LDL-C, and TG levels. Substituting dietary fats with avocados versus adding to the free diet should be the primary recommendation strategy. Larger trials looking at the impact of avocados on major adverse cardiovascular events are warranted.
Article
Full-text available
Introduction: Many studies have shown that oral supplementation with astaxanthin may be a novel potential treatment for inflammation and oxidative stress in cardiovascular diseases, but evidence of the effects on lipid profile and glucose is still inconclusive. Therefore, we performed a meta-analysis to evaluate the efficacy of astaxanthin supplementation on plasma lipid and glucose concentrations. Material and methods: The search included PubMed, Cochrane Library, Scopus, and EMBASE (up to November 27, 2014) to identify randomized controlled trials (RCTs) investigating the effects of astaxanthin supplementation on lipid profile and glucose levels. Two independent reviewers extracted data on study characteristics, methods and outcomes. Results: Seven studies meeting inclusion criteria with 280 participants were selected for this meta-analysis; 163 participants were allocated to the astaxanthin supplementation group and 117 to the control group. A random-effect meta-analysis of data from 7 RCTs (10 treatment arms) did not show any significant effect of supplementation with astaxanthin on plasma concentrations of total cholesterol (weighted mean difference (WMD): –1.52 mg/dl, 95% CI: –8.69 to –5.66, p = 0.679), LDL-C (WMD: +1.25 mg/dl, 95% CI: –6.70 to +9.21, p = 0.758), HDL-C (WMD: +1.75 mg/dl, 95% CI: –0.92 to +4.42, p = 0.199), triglycerides (WMD: –4.76 mg/dl, 95% CI: –21.52 to +12.00, p = 0.578), or glucose (WMD: –2.65 mg/dl, 95% CI: –5.84 to +0.54, p = 0.103). All these effect sizes were robust, and omission of any of the included studies did not significantly change the overall estimate. Conclusions: This meta-analysis of data from 10 RCT arms did not indicate a significant effect of supplementation with astaxanthin on plasma lipid profile, but a slight glucose-lowering effect was observed. Further, well-designed trials are necessary to validate these results.
Article
Full-text available
Since antiquity bitter melon has been in use for treating diabetes but clinical trials show conflicting results about its usefulness. The present study aims to asses and compare the hypoglycemic and antiatherogenic effects as well as the safety of two different doses of bitter melon with glibenclamide. A total of 95 participants were randomized into 3 groups; group I and group II received bitter melon (2 g/day and 4 g/day respectively) and group III received glibenclamide (5 mg/day) for 10 weeks. Glycemic control and antiatherogenic effects were determined by assessing glycohemoglobin (HbA1-c), fasting plasma glucose (FPG), 2 hour oral glucose tolerance test (OGTT), plasma sialic acid (PSA), systolic blood pressure (SBP), blood lipids and atherogenic index at different time periods. Compared to baseline, mean reduction in HbA1-c at the endpoint was significant among patients of group I, group II and group III (p <= 0.05, p <= 0.02 and p < 0.005 respectively) and same was the case for FPG (p <= 0.05, p < 0.04, p < 0.003 respectively), but the improvement in 2 hour OGTT was significant only in group III (p < 0.03). The decrease in PSA was observed only among group I and group II with the later showing significant reduction from baseline (p < 0.01). In group III, the level slightly increased. Parameters including blood lipids, atherogenic index, body weight and SBP improved among patients of group I and group II but deteriorated among group III patients. Our study concludes that bitter melon has a weaker hypoglycemic effect but ameliorates the diabetes associated cardiovascular (CV) risk factors more effectively than glibenclamide.Trial registration: The trial was registered with Naseer Teaching Hospital Clinical Trials Registry number GU2014492233.
Article
Full-text available
Diabetes, obesity, and metabolic syndrome are becoming epidemic both in developed and developing countries in recent years. Complementary and alternative medicines have been used since ancient era for the treatment of diabetes and cardiovascular diseases. Bitter melon is widely used as vegetables in daily food in Bangladesh and several other countries in Asia. The fruits extract of bitter melon showed strong antioxidant and hypoglycemic activities in experimental condition both in vivo and in vitro. Recent scientific evaluation of this plant extracts also showed potential therapeutic benefit in diabetes and obesity related metabolic dysfunction in experimental animals and clinical studies. These beneficial effects are mediated probably by inducing lipid and fat metabolizing gene expression and increasing the function of AMPK and PPARs, and so forth. This review will thus focus on the recent findings on beneficial effect of Momordica charantia extracts on metabolic syndrome and discuss its potential mechanism of actions.
Article
Background: Cashews are the third most-consumed tree nut in the United States and are abundant with monounsaturated fatty acids and polyunsaturated fatty acids, which are associated with reduced cardiovascular disease risk. Although a qualified Food and Drug Administration health claim exists for nuts and heart health, cashews have been exempt from its use because cashews exceed the disqualifying amount of saturated fatty acids. Approximately one-third of the saturated fat in cashews is stearic acid, which is relatively neutral on blood lipids, thereby suggesting that cashews could have effects that are similar to those of other nuts. However, clinical data on cashews and blood lipids have been limited. Objective: We investigated the effect of reasonable intakes of cashews on serum lipids in adults with or at risk of high LDL cholesterol. Design: In a randomized, crossover, isocaloric, controlled-feeding study, 51 men and women (aged 21–73 y) with a median LDL-cholesterol concentration of 159 mg/dL (95% CI: 146, 165 mg/dL) at screening consumed typical American diets with cashews (28–64 g/d; 50% of kilocalories from carbohydrate, 18% of kilocalories from protein, and 32% of kilocalories from total fat) or potato chips (control; 54% of kilocalories from carbohydrate, 18% of kilocalories from protein, and 29% of kilocalories from total fat) for 28 d with a ≥2-wk washout period. Results: Consumption of the cashew diet resulted in a significantly greater median change from baseline (compared with the control, all P < 0.05) in total cholesterol [−3.9% (95% CI: −9.3%, 1.7%) compared with 0.8% (95% CI: −1.5%, 4.5%), respectively], LDL cholesterol [−4.8% (95% CI: −12.6%, 3.1%) compared with 1.2% (95% CI: −2.3%, 7.8%), respectively], non-HDL cholesterol [−5.3% (95% CI: −8.6%, 2.1%) compared with 1.7% (95% CI: −0.9%, 5.6%), respectively], and the total-cholesterol:HDL-cholesterol ratio [−0.0% (95% CI: −4.3%, 4.8%) compared with 3.4% (95% CI: 0.6%, 5.2%), respectively]. There were no significant differences between diets for HDL cholesterol and triglyceride. Conclusions: In comparison with a control diet, the incorporation of cashews into typical American diets decreases total cholesterol and LDL cholesterol. Results from this study provide support that the daily consumption of cashews, when substituted for a high-carbohydrate snack, may be a simple dietary strategy to help manage total cholesterol and LDL cholesterol. This study was registered at clinicaltrials.gov as NCT02769741.
Article
Background: Postprandial hyperlipidemia is associated with impaired endothelial function. Peanut consumption favorably affects the lipid and lipoprotein profile; however, the effects on endothelial function remain unclear. Objective: The purpose of the study was to evaluate the effects of acute peanut consumption as part of a high-fat meal on postprandial endothelial function. Methods: We conducted a randomized, controlled, crossover postprandial study to evaluate the effect of acute peanut consumption on postprandial lipids and endothelial function as assessed by flow-mediated dilatation (FMD) of the brachial artery in 15 healthy overweight or obese men [mean age: 26.7 y; mean body mass index (in kg/m²): 31.4]. Participants consumed, in a randomized order, a peanut meal containing 3 ounces (85 g) ground peanuts (1198 kcal; 40.0% carbohydrate, 47.7% fat, 19.4% saturated fat, 13.2% protein) and a control meal matched for energy and macronutrient content. Meals were in the form of a shake, scheduled ≥1 wk apart. Lipids, lipoproteins, glucose, and insulin were measured at baseline (0 min) and at 30, 60, 120, and 240 min after shake consumption. FMD was measured at baseline and at 240 min. Results: Acute peanut consumption blunted the serum triglyceride (TG) response 120 and 240 min after consumption compared with the control meal (means ± SEMs—120 min: 188.9 ± 19.4 compared with 197.5 ± 20.7 mg/dL; 240 min: 189.9 ± 24.3 compared with 197.3 ± 18.4 mg/dL; P < 0.05 for both). Total, LDL, and HDL cholesterol and glucose and insulin responses were similar between the test meals. Compared with baseline, only the control meal significantly decreased FMD at 240 min (control: −1.2% ± 0.5%; P = 0.029; peanut: −0.6% ± 0.5%; P = 0.3). Participants with higher baseline total (>150 mg/dL) and LDL (>100 mg/dL)-cholesterol concentrations showed a significant decrease in FMD after the control meal (−1.8%, P = 0.017; −2.0%, P = 0.038), whereas the peanut meal maintained endothelial function in all participants irrespective of total- and LDL-cholesterol concentrations. Conclusion: The inclusion of 85 g peanuts (3 ounces) as part of a high-fat meal improved the postprandial TG response and preserved endothelial function in healthy overweight or obese men. This trial was registered at clinicaltrials.gov as NCT01405300.
Article
The polysaccharide extract of Momordica charantia has various biological activities; however, its effect on endothelial dysfunction in myocardial infarction remains unclear. To elucidate this, myocardial infarction was induced in rats using isoproterenol (ISP). Pretreatment with M. charantia polysaccharides (MCP; 150 or 300 mg/kg) for 25 days significantly inhibited increases in heart weight, the heart-weight-to-body-weight ratio, and infarction size, and ameliorated the increased serum levels of aspartate transaminase, creatine kinase, lactate dehydrogenase, total cholesterol, triglycerides, very-low-density lipoprotein cholesterol, low-density lipoprotein cholesterol, and high-density lipoprotein cholesterol. In addition, MCP enhanced the activity of superoxide dismutase, catalase, and non-protein sulfhydryls, and decreased the level of lipid peroxidation. Moreover, MCP pretreatment downregulated the expression of proinflammatory cytokines (tumor necrosis factor alpha, interleukin (IL)-6, and IL-10), inflammatory markers (nitric oxide, myeloperoxidase, and inducible nitric oxide synthase), and apoptotic markers (caspase-3 and BAX), and upregulated Bcl-2 expression. Pretreatment with MCP reduced myonecrosis, edema, and inflammatory cell infiltration, and restored cardiomyocytes architecture. This myocardial protective effect could be related to the enhancement of the antioxidant defense system through the nuclear factor kappa B (NF-kB) pathways, and to anti-apoptosis through regulation of Bax, caspase-3, and Bcl-2.
Article
Background and aims: Epidemiological evidence suggests an association between consumption of tomato products or lycopene and lower risk for cardiovascular diseases (CVD). Our aim was to evaluate the state of the evidence from intervention trials on the effect of consuming tomato products and lycopene on markers of cardiovascular (CV) function. We undertook a systematic review and meta-analysis on the effect of supplementing tomato and lycopene on CV risk factors. Methods: Three databases including Medline, Web of science, and Scopus were searched from inception to August 2016. Inclusion criteria were: intervention trials reporting effects of tomato products and lycopene supplementation on CV risk factors among adult subjects >18 years of age. The outcomes of interest included blood lipids (total-, HDL-, LDL-cholesterol, triglycerides, oxidised-LDL), endothelial function (flow-mediated dilation (FMD), pulse wave velocity (PWV)) and blood pressure (BP) inflammatory factors (CRP, IL-6) and adhesion molecules (ICAM-1). Random-effects models were used to determine the pooled effect sizes. Results: Out of 1189 publications identified, 21 fulfilled inclusion criteria and were meta-analysed. Overall, interventions supplementing tomato were associated with significant reductions in LDL-cholesterol (-0.22 mmol/L; p = 0.006), IL-6 (standardised mean difference -0.25; p = 0.03), and improvements in FMD (2.53%; p = 0.01); while lycopene supplementation reduced systolic-BP (-5.66 mmHg; p = 0.002). No other outcome was significantly affected by these interventions. Conclusions: The available evidence on the effects of tomato products and lycopene supplementation on CV risk factors supports the view that increasing the intake of these has positive effects on blood lipids, blood pressure and endothelial function. These results support the development of promising individualised nutritional strategies involving tomatoes to tackle CVD.
Article
Oats are a rich source of β -glucan, a viscous, soluble fibre recognised for its cholesterol-lowering properties, and are associated with reduced risk of CVD. Our objective was to conduct a systematic review and meta-analysis of randomised-controlled trials (RCT) investigating the cholesterol-lowering potential of oat β -glucan on LDL-cholesterol, non-HDL-cholesterol and apoB for the risk reduction of CVD. MEDLINE, Embase, CINAHL and Cochrane CENTRAL were searched. We included RCT of ≥3 weeks of follow-up, assessing the effect of diets enriched with oat β -glucan compared with controlled diets on LDL-cholesterol, non-HDL-cholesterol or apoB. Two independent reviewers extracted data and assessed study quality and risk of bias. Data were pooled using the generic inverse-variance method with random effects models and expressed as mean differences with 95 % CI. Heterogeneity was assessed by the Cochran’s Q statistic and quantified by the I² -statistic. In total, fifty-eight trials ( n 3974) were included. A median dose of 3·5 g/d of oat β -glucan significantly lowered LDL-cholesterol (−0·19; 95 % CI −0·23, −0·14 mmol/l, P <0·00001), non-HDL-cholesterol (−0·20; 95 % CI −0·26, −0·15 mmol/l, P <0·00001) and apoB (−0·03; 95 % CI −0·05, −0·02 g/l, P <0·0001) compared with control interventions. There was evidence for considerable unexplained heterogeneity in the analysis of LDL-cholesterol ( I² =79 %) and non-HDL-cholesterol ( I² =99 %). Pooled analyses showed that oat β -glucan has a lowering effect on LDL-cholesterol, non-HDL-cholesterol and apoB. Inclusion of oat-containing foods may be a strategy for achieving targets in CVD reduction.
Article
Background: Plant-based diets can lower serum lipids. Whether soy foods offer additional benefits remains controversial. Objective: To determine the effect of different protein sources on serum lipids and glucose metabolism, emphasizing comparisons between soy and nonsoy plant-based diets. Methods: Secondary data analysis. A total of 173 postmenopausal women were randomized to 1 of 4 weighed metabolic diets for 6 weeks. Diets were equivalent in energy, protein, and fat with at least 80% of protein from either nondairy animal, dairy, nonsoy plant, or soy foods. At baseline and week 6, total cholesterol (TC), low-density lipoprotein (LDL), high-density lipoprotein (HDL), triglycerides, glucose, and insulin were measured. Changes in variables during the diet period were compared within and among groups using t tests and analysis of variance. Results: TC decreased 38.8 mg/dL in soy group (P < .001 vs dairy and animal) and 30.5 mg/dL in nonsoy plant group (P = .003 vs dairy, .03 vs animal). LDL decreased 28.3 mg/dL in soy group (P < .001 vs dairy and animal) and 20.6 mg/dL in nonsoy plant group (P = .005 vs dairy, .06 vs animal). HDL decreased 12 mg/dL in soy group (P = .003 vs dairy, .0008 vs animal) and 10 mg/dL in nonsoy plant group (P = .05 vs dairy, .04 vs animal). There were no significant differences in lipid changes between soy and nonsoy plant-based diets. No differences among groups in changes in triglycerides, glucose, or insulin were seen. Conclusions: Soy and nonsoy plant-based diets reduced TC and LDL with no significant difference between them. Further studies are needed to determine the specific lipid-lowering components of both soy and nonsoy plant foods.