ArticlePDF Available

The effect of ginger consumption on glycemic status, lipid profile and some inflammatory markers in patients with type 2 diabetes mellitus

Taylor & Francis
International Journal of Food Sciences and Nutrition
Authors:

Abstract and Figures

Objective: To assess the effect of ginger consumption on glycemic status, lipid profile and some inflammatory markers in patients with type 2 diabetes mellitus. Methods: In a double-blinded, placebo-controlled clinical trial, 70 type 2 diabetic patients were enrolled. They allocated randomly into ginger group and control group. They consumed 1600 mg ginger versus 1600 mg wheat flour placebo daily for 12 weeks. Serum sugar, lipids, CRP, PGE2 and TNFα were measured before and after intervention. Results: Ginger reduced fasting plasma glucose, HbA1C, insulin, HOMA, triglyceride, total cholesterol, CRP and PGE₂ significantly compared with placebo group (p < 0.05). There were no significant differences in HDL, LDL and TNFα between two groups (p > 0.05). Conclusion: Ginger improved insulin sensitivity and some fractions of lipid profile, and reduced CRP and PGE₂ in type 2 diabetic patients. Therefore ginger can be considered as an effective treatment for prevention of diabetes complications.
Content may be subject to copyright.
http://informahealthcare.com/ijf
ISSN: 0963-7486 (print), 1465-3478 (electronic)
Int J Food Sci Nutr, Early Online: 1–6
!2014 Informa UK Ltd. DOI: 10.3109/09637486.2014.880671
RESEARCH ARTICLE
The effect of ginger consumption on glycemic status, lipid profile and
some inflammatory markers in patients with type 2 diabetes mellitus
Tahereh Arablou
1
, Naheed Aryaeian
2
, Majid Valizadeh
3
, Faranak Sharifi
3
, AghaFatemeh Hosseini
4
, and
Mahmoud Djalali
5
1
Department of Nutrition, School of Nutritional Sciences and Dietetics, Tehran University of Medical Sciences, Tehran, Iran,
2
Department of
Nutrition, School of Public Health, Iran University of Medical Sciences, Tehran, Iran,
3
Zanjan Metabolic Disease Research Center, Zanjan University
of Medical Sciences, Zanjan, Iran,
4
Department of Statistics and Mathematics, School of Health Management and Information Science, Iran
University of Medical Sciences, Tehran, Iran, and
5
Department of Nutrition and Biochemistry, School of Nutritional Sciences and Dietetics, Tehran
University of Medical Sciences, Tehran, Iran
Abstract
Objective: To assess the effect of ginger consumption on glycemic status, lipid profile and some
inflammatory markers in patients with type 2 diabetes mellitus. Methods: In a double-blinded,
placebo-controlled clinical trial, 70 type 2 diabetic patients were enrolled. They allocated
randomly into ginger group and control group. They consumed 1600 mg ginger versus
1600 mg wheat flour placebo daily for 12 weeks. Serum sugar, lipids, CRP, PGE2 and TNFawere
measured before and after intervention. Results: Ginger reduced fasting plasma glucose, HbA
1C
,
insulin, HOMA, triglyceride, total cholesterol, CRP and PGE
2
significantly compared with
placebo group (p50.05). There were no significant differences in HDL, LDL and TNFabetween
two groups (p40.05). Conclusion: Ginger improved insulin sensitivity and some fractions of lipid
profile, and reduced CRP and PGE
2
in type 2 diabetic patients. Therefore ginger can be
considered as an effective treatment for prevention of diabetes complications.
Keywords
Blood sugar, ginger, inflammation, insulin
sensitivity, serum lipids, type 2 diabetes
History
Received 14 October 2013
Revised 22 December 2013
Accepted 26 December 2013
Published online 4 February 2014
Introduction
Type 2 diabetes, a growing public health problem, is associated
with increased morbidity and mortality (Mahluji et al., 2013).
Cardiovascular diseases are the leading cause of morbidity and
mortality in diabetic patients. Diabetic patients are 2-to-4-folds
more prone to atherosclerosis. Dyslipidemia is the most important
modifiable risk factor for atherosclerosis in diabetic patients.
Glycemic control moderates diabetes-related dyslipidemia
(Bhandari et al., 2005). Insulin sensitivity can control glycemia,
improve endothelial function and reduce the risk of atheroscler-
osis (Mahluji et al., 2013).
Today, many herbs are used to treat diseases worldwide.
Rhizome of ginger (Zingiber officinale Roscoe, Zingiberaceae),
as a spice, is used widely all around the world. For centuries, this
plant has been an important part of Chinese, Ayurvedic and Tibb
Unani herbal medicine to treat cataract, rheumatism, nervous
diseases, gingivitis, toothache, asthma, stroke, constipation and
diabetes (Ali et al., 2008).
Recent studies have shown that ginger has different pharma-
cological effects, due to its different components such as gingerols
and shogaols. So far, more than 40 antioxidant compounds are
also detected in ginger (Shirdel et al., 2009). The main
pharmacological actions of ginger and its isolated compounds
include immunomodulatory, antitumorogenesis, antiinflamma-
tory, antiapoptosis, glucose and lipid lowering effect and
antiemetic (Ali et al., 2008).
To date, several studies have investigated the effect of ginger
on blood glucose and lipids.
Mahluji et al. (2013) studied the effect of daily intake of 2 g of
powdered ginger in type 2 diabetic patients. After 2 months,
insulin, HOMA, TG and LDL decreased significantly in ginger
group compared with placebo group, no significant changes were
observed in FPG, HbA
1C
, total cholesterol and HDL levels.
Also, in another study (Bordia et al., 1997), 4 g of ginger
powder, were given to healthy subjects, patients with CAD and
type 2 diabetic patients with or without CAD. After 3 months, no
significant changes were observed in blood sugar and serum
lipids.
Diabetes can be considered as an inflammatory disease that is
associated with metabolic disorders (Navarro & Mora, 2006).
Mild chronic inflammation associated with increased levels of
circulating inflammatory cytokines may increase insulin resist-
ance in the liver, skeletal muscles and vascular endothelium
(Simin et al., 2007).
The antiinflammatory effect of ginger is possibly through
inhibition of cyclooxygenase, inducible nitric oxide synthase
and lipoxygenase activity and suppression of prostaglandin
synthesis and interference in cytokine signaling and this is
attributed to gingerols, shogaols and diarylheptanoids in ginger
(Singletary, 2010).
Correspondence: Naheed Aryaeian, PhD, MS, Department of Nutrition,
School of Public Health, Iran University of Medical Sciences, Hemmat
Broadway, Tehran, Iran. Tel: +982186704750. Fax: +982188622707. E-
mail: aryaeian.n@iums.ac.ir, nah_arya2002@yahoo.com
Int J Food Sci Nutr Downloaded from informahealthcare.com by 194.225.187.233 on 04/27/14
For personal use only.
To the best of our knowledge, there is no performed research
assessing the effect of ginger on inflammatory markers in diabetic
patients and because of insufficient information on the effects of
ginger on blood glucose and lipid profile, this study was
performed to investigate the effect of ginger supplementation on
glycemic status, lipid profile and CRP, PGE
2
and TNFaas
inflammatory markers in patients with type 2 diabetes.
Methods and materials
Study design
This is a double-blinded, placebo-controlled clinical trial. The
study was approved by Medical Ethics Committee of Tehran
University of Medical Sciences and Zanjan Metabolic Disease
Research Center which conform to the provision of Helsinki in
1995 (as revised in 2000) and recorded by the identification code
of IRCT201204159472N1 in clinical trials registry of Iran. Prior
to the study, informed written consents were obtained from all of
the participants.
Study population and intervention
Seventy patients 30–70 years old with type 2 diabetes included
after baseline assessments. Eligibility criteria included: treated
with oral hypoglycemic agents, HbA
1C
between 7% and 10%,
BMI between 20 and 35 kg/m
2
, no pregnancy and lactation, no use
of tobacco or alcohol, any renal, liver, thyroid and parathyroid
disorders, cancer, infection, inflammation and fever.
Participants allocated randomly into two groups receiving
ginger or placebo. Neither researcher nor the patients were aware
whether the patients belong to ginger or placebo group. They
consumed 1600 mg of powdered rhizome of ginger or wheat flour
placebo (one 800 mg capsule before lunch and one 800 mg
capsule before dinner) per day. The participants were asked not to
change their regular diet and physical activity during the study
period and report any particular disease or any other abnormal
sensations immediately. The intervention lasted for 12 weeks.
During the study period, two patients in ginger group (one due to
heartburn and one due to unwillingness to cooperation) and five
patients in placebo group (three due to unwillingness to cooper-
ation, one due to pregnancy and one due to recurrence of disease
and the need for insulin therapy) were excluded from the study
and the study ended with 63 patients (33 in ginger group and 30 in
placebo group).
Supplement preparation
Dried rhizomes of Z. officinale were purchased from local market
and ground into fine particles and capsules containing 800 mg of
powdered ginger were prepared. Weight of empty and filled
capsules was measured using high precision balance (HR202i,
A&D company, Japan) at nutrition and biochemistry laboratory
(School of Health, Tehran University of Medical Sciences,
Tehran, Iran). Placebo containing wheat flour was prepared in
the same color and package and they were placed in a container of
ginger powder for 2 weeks to give ginger odor to all placebo
capsules.
Analytical procedures
To control the confounding effects of dietary intake and physical
activity, energy intake, macronutrients and some micronutrients
(vitamins A, C and E, zinc and selenium), 24-h recall question-
naire (1 day) and food diary (2 days) and International physical
activity questionnaire-short form (IPAQ-S), were recorded for
each person at the beginning and end of the study. Dietary
intake were analyzed by Nutritionist IV software (version 3.5.2,
The Hearst Corporation, San Bruno, CA). Body weight was
measured in the fasting state with light clothing and without shoes
using Seca scale (Seca, Hamburg, Germany) and height was
measured without shoes using a stadiometer attached to the scale.
Blood samples (10 ml) were taken in a 12- to 14-h fasting state at
the beginning and after 12 weeks of intervention. Sera were
obtained by high-speed centrifugation and stored at 70 C.
Fasting blood glucose, HbA
1C
, TG, total cholesterol and HDL
levels were determined by enzymatic colorimetric method with
commercial kits (Pars Azmun Co, Iran) on an automatic analyzer
(Abbott, model Alcyon 300, Abbott Park, IL). Serum LDL was
calculated using Friedewald formula:
LDL ¼Total cholestrol HDL TG=5
Serum insulin was measured by radioimmunoassay kit (Mono
bind Inc, 2425-300A, Insulin AccuBind, Lake Forest, CA) using
ELISA method. HOMA was calculated using the US formula:
fasting blood glucose ðmg=dlÞ
fasting serum insulin ðU=mlÞ=405
Plasma PGE
2
was determined by enzyme immunoassay using
ELISA kit (Cayman Chemical, Item Number 514010, Ann Arbor,
MI). Serum TNFaand quantitative CRP were measured by
enzyme-linked immunosorbent assay and enzyme immunoassay
using ELISA kit (Orgenium, product code TNFa021, Vantaa,
Finland and Cayman Chemical, Item Number 10011236, Ann
Arbor, MI, respectively).
Statistical analysis
Statistical analysis was performed using SPSS software (version
16; SPSS Inc., Chicago, IL). Kolmogorov–Smirnov test was used
to determine data compliance with the normal distribution.
Quantitative variables were compared between two groups at
baseline and at the end of the study using an independent t-test or
Mann–Whitney test. Quantitative variables before and after
treatment within each group were compared with paired t-test
or Wilcoxon test. Qualitative variables were analyzed with chi-
squared and McNemar tests. All values are reported based on
mean ± SD. pValue 50.05 was considered as the statistical
significance level.
Results
Baseline characteristics of study participants are presented in
Table 1. Comparisons showed no significant differences in age,
body weight, duration of illness, sex and physical activity levels
between two groups at baseline.
Statistical analysis of energy, macronutrients and some
micronutrients intake showed no significant differences between
two groups and within each group at the beginning and end of the
study (Tables 2 and 3). Also, there is no significant changes in
type and dose of medications and physical activity levels between
two groups before and after intervention (date not shown).
Table 4 shows the mean and standard deviation of anthropo-
metric indices, glycemic status and lipid profile in both groups
before and after intervention. There were no significant changes
in body weight and BMI between two groups before and after
intervention. Results showed that despite no significant differ-
ences in FPG in ginger and placebo groups before and after
intervention, changes in FPG were statistically significant
compared between two groups (p¼0.02).
Ginger also caused a significant decrease in HbA
1C
(p¼0.001), insulin (p¼0.01), HOMA (p¼0.000),TG
(p¼0.001) and total cholesterol (p¼0.02) and significant
increase in HDL/total cholesterol ratio (p¼0.02) in ginger
2T. Arablou et al. Int J Food Sci Nutr, Early Online: 1–6
Int J Food Sci Nutr Downloaded from informahealthcare.com by 194.225.187.233 on 04/27/14
For personal use only.
group compared with placebo group, but had no significant effect
on HDL (p¼0.97), LDL (p¼0.16) and LDL/HDL ratio
(p¼0.29).
Also results showed significant reduction in serum CRP
(p¼0.01) and plasma PGE
2
(p¼0.000) levels in ginger group
compared with placebo group after 12 weeks (Table 5). Although
serum TNFalevels was significantly decreased in ginger group
compared with placebo group after 12 weeks (p¼0.005),
statistical analysis showed no significant changes in serum
TNFalevels within groups (Table 5).
Discussion
In this study, the effect of ginger on glycemic status, plasma lipids
and some inflammatory markers in patients with type 2 diabetes
were examined.
Results showed that ginger reduced FPG and insulin and
increased insulin sensitivity. Mahluji et al. (2013) found that
taking 2 g of ginger per day for 2 months has no effect on FPG and
HbA
1C
, but can reduce serum insulin and HOMA. This difference
is attributable to longer duration of our study.
Bordia et al. (1997) observed no significant changes in blood
sugar levels in healthy individuals and patients with CAD and
type 2 diabetes with or without CAD.
Animal studies had conflicting results. Shanmugam et al.
(2011) showed that ginger decreased blood glucose significantly
in diabetic rats compared with diabetic controls. Although Islam
& Choi (2008) observed no effect of ginger on blood sugar and
insulin levels in diabetic rats, Nammi et al. (2009) and Goyal &
Kadnur (2006), observed the effect of ginger on reduction of
blood glucose and insulin, in their studies on rats receiving the
high-fat diet and obese rats, respectively.
Many researchers speculate that hypoglycemic and other
pharmacological activities of ginger is due to its phenols,
polyphenols and flavonoids (Shanmugam et al., 2011). It seems
that ginger decreases blood glucose by antagonistic activity
against serotonin receptors and its blockage (Al-Amin et al., 2006;
Goyal & Kadnur, 2006). Also ginger may inhibit the intestinal
glucosidase and amylase enzymes activity and thereby reduce
absorption of glucose (Li et al., 2012).
Many studies have shown that treatment with antioxidants
improves glucose transport and tolerance in type 2 diabetic
patients and animals with insulin resistance. Ginger contains lots
of antioxidants including gingerols, shogaols, paradols and
zingerones. It is possible that they act by increasing the GLUT4
protein, insulin receptors and improving b-cells function (Mahluji
et al., 2013).
Table 2. Energy and macronutrients intake in ginger and placebo groups
before and after intervention.
Variable Ginger group (n¼33) Placebo group (n¼30) p*
Energy (kcal)
Before 1517.3 ± 352.7 1537.7 ± 287.7 0.8
After 1514.2 ± 355.8 1540.7 ± 290.5 0.74
py0.84 0.83
Carbohydrate (g)
Before 194.4 ± 57.9 193.5 ± 46.8 0.95
After 195.2 ± 57.3 192.7 ± 46.1 0.85
py0.87 0.91
Protein (g)
Before 57.6 ± 18.6 61.3 ± 20.1 0.45
After 59.6 ± 18.2 62.7 ± 20.7 0.52
py0.5 0.75
Total fat (g)
Before 56.9 ± 14.5 58.8 ± 18.0 0.65
After 55.3 ± 13.6 58.2 ± 18.5 0.48
py0.49 0.87
SAFA (g)
Before 18.7 ± 1.9 18.9 ± 1.9 0.7
After 18.8 ± 2.0 18.5 ± 2.1 0.73
py0.79 0.35
MUFA (g)
Before 15.2 ± 1.6 14.4 ± 3.6 0.39
After 15.5 ± 1.8 15.4 ± 1.7 0.86
py0.27 0.27
PUFA (g)
Before 16.1 ± 1.5 16.4 ± 1.6 0.63
After 16.2 ± 1.7 16.3 ± 1.6 0.88
py0.76 0.84
Data are presented as mean ± SD.
p* Between group comparison (independent t-test or Mann–Whitney
test).
pyWithin group comparison (paired t-test or Wilcoxon test).
pValue 50.05 is significant.
Table 3. Some micronutrients intake in ginger and placebo groups before
and after intervention.
Variable Ginger group (n¼33) Placebo group (n¼30) p*
Vitamin A (mg)
Before 831.1 ± 773.6 660.0 ± 676.6 0.21
After 831.9 ± 818.3 679.4 ± 661.7 0.39
py0.7 0.057
Vitamin C (mg)
Before 44.6 ± 16.8 39.1 ± 21.6 0.26
After 45.2 ± 16.4 38.9 ± 21.1 0.19
py0.38 0.63
Vitamin E (mg)
Before 16.72 ± 6.8 16.37 ± 5.3 0.82
After 16.76 ± 6.8 16.36 ± 5.3 0.78
py0.62 0.91
Zinc (mg)
Before 7.9 ± 2.7 8.2 ± 2.8 0.55
After 8.0 ± 2.8 8.4 ± 3.2 0.55
py0.22 0.19
Selenium (mg)
Before 62.7 ± 30.6 63.6 ± 31.9 0.95
After 64.0 ± 32.4 63.3 ± 31.5 0.92
py0.32 0.49
Data are presented as mean ± SD.
p* Between group comparison (independent t-test or Mann–Whitney
test).
pyWithin group comparison (paired t-test or Wilcoxon test).
pValue 50.05 is significant.
Table 1. Baseline characteristics of study participants.
Characteristics
Ginger group
(n¼33)
Placebo group
(n¼30) p
Age (year) 52.6 ± 8.4 52.0 ± 9.0 0.78*
Body weight (kg) 66.2 ± 8.2 66.1 ± 7.8 0.95*
Duration of illness (month) 45.8 ± 25.5 48.5 ± 26.5 0.68*
Sex
Female (%) 25 (75.8) 23 (76.7) 0.93y
Male (%) 8 (24.2) 7 (23.3)
Physical activity
Light (%) 15 (45.5) 14 (46.7) 0.82y
Moderate (%) 13 (39.4) 10 (33.3)
Severe (%) 5 (15.2) 6 (20)
Data of age, body weight and duration of illness are presented as mean
± SD.
*Independent t-test.
yChi-squared test.
pValue 50.05 is significant.
DOI: 10.3109/09637486.2014.880671 Effect of ginger consumption in patients with type 2 diabetes mellitus 3
Int J Food Sci Nutr Downloaded from informahealthcare.com by 194.225.187.233 on 04/27/14
For personal use only.
It seems that ginger’s effect on insulin sensitivity is through
the activation of PPARgor up regulation of adiponectin. Isa et al.
(2008) stated that the 6-gingerol and 6-shogaol in ginger,
up regulate adiponectin and 6-shogaol has agonistic activity
with PPARg. Plasma adiponectin levels and its RNA
expression are reduced in obesity and insulin resistance, and
increasing adiponectin, improves insulin sensitivity (Nammi
et al., 2009).
In this study, ginger reduced TG and total cholesterol in
diabetic patients but it had no effect on HDL and LDL.
This is consistent with the findings of Alizadeh et al.’s study
(2008) which investigated the effect of taking 3 g of ginger for
45 days on blood lipids in patients with hyperlipidemia. In
Mahluji et al.’s study (2013), ginger reduced LDL and TG
significantly, but it had no significant effect on total cholesterol
and HDL levels.
In another study, taking 4 g of ginger for 3 months caused no
significant changes in blood lipids in healthy subjects, patients
with CAD and type 2 diabetes with or without CAD (Bordia et al.,
1997).
Our finding on the effectiveness of ginger in decreasing serum
TG is consistent with the findings of several animal studies
(Al-Amin et al., 2006; Bhandari et al., 2005; ElRokh et al., 2010;
Goyal & Kadnur, 2006; Nammi et al., 2009; Sharma et al., 1991;
Shirdel et al., 2009).
The hypotriglyceridemic effect of ginger may be due to
increasing lipoprotein lipase enzyme activity and therefore
hydrolysis of circulatory TG and decreasing serum TG (Shirdel
et al., 2009). Ginger also reduces the ChREBP gene expression in
the liver. ChREBP, a transcriptional regulator of lipid and glucose
metabolism, converts excess carbohydrates into TGs. Reduced
ChREBP expression, decreases expression of ACC1, fatty acid
synthase, SCD1, and glucose-6-phosphatase, glucogenic and
lipogenic proteins and decreases fat accumulation in the liver,
reduces serum TG and improves insulin resistance (Gao et al.,
2012).
Based on the findings of the present study, ginger could
decrease serum total cholesterol in diabetic patients.
This effect also was observed by Alizadeh et al. (2008) and
some animal studies (Al-Amin et al., 2006; Bhandari et al., 2005;
ElRokh et al., 2010; Goyal & Kadnur, 2006; Nammi et al., 2009;
Sharma et al., 1991).
Ginger can increase hepatic cholesterol 7a-hydroxylase enzyme
activity and the conversion of cholesterol into bile acids, resulting
in reduced serum cholesterol concentration. Also there are
observations that the compounds in ginger inhibit the biosynthesis
of cholesterol in the liver of rats (Alizadeh et al., 2008). In addition
to the effect of ginger on increasing bile secretion, increased fecal
excretion of cholesterol and phospholipids by ginger can also
reduce cholesterol levels (Sharma et al., 1991).
The findings showed that ginger has no significant effect on
serum HDL, LDL and LDL/HDL ratio. Findings about the effect
Table 4. Anthropometric indices, glycemic and lipid profile in ginger and
placebo groups before and after intervention.
Variable
Ginger group
(n¼33) p*
Placebo group
(n¼30) p*py
Body weight (kg)
Before 66.2 ± 8.2 0.2 66.1 ± 7.8 0.16 0.95
After 66.1 ± 8.2 66.0 ± 7.7 0.95
Differences 0.07 ± 0.3 0.06 ± 0.2 0.9
BMI (kg/m
2
)
Before 26.9 ± 3.6 0.16 26.8 ± 3.4 0.63 0.91
After 26.8 ± 3.5 26.8 ± 3.4 0.93
Differences 0.51 ± 0.2 0.03 ± 0.3 0.82
FPG (mg/dl)
Before 131.0 ± 42.5 0.17 129.0 ± 62.5 0.8 0.88
After 121.9 ± 37.4 145.0 ± 68.4 0.1
Differences 9.1 ± 38.5 16.0 ± 48.5 0.02
HbA
1C
%
Before 8.4 ± 1.6 0.002 8.1 ± 1.5 0.1 0.55
After 7.3 ± 1.3 8.6 ± 2.2 0.01
Differences 1.0 ± 1.7 0.4 ± 1.4 0.001
Insulin (mU/ml)
Before 8.3 ± 8.3 0.000 6.9 ± 4.6 0.66 0.54
After 4.6 ± 1.4 7.0 ± 3.3 0.000
Differences 3.7 ± 8.2 0.1 ± 3.5 0.01
HOMA
Before 3.1 ± 5.1 0.000 2.2 ± 1.5 0.36 0.58
After 1.3 ± 0.5 2.4 ± 1.4 0.01
Differences 1.7 ± 4.9 0.2 ± 1.3 0.000
TG (mg/dl)
Before 187.0 ± 68.9 0.001 187.4 ± 111.4 0.72 0.98
After 141.5 ± 47.4 190.0 ± 115.3 0.03
Differences 45.4 ± 69.6 2.5 ± 38.5 0.001
Total cholesterol (mg/dl)
Before 188.6 ± 35.6 0.01 188.2 ± 45.2 0.4 0.96
After 173.1 ± 28.7 194.0 ± 45.1 0.03
Differences 15.4 ± 34.3 5.8 ± 37.9 0.02
HDL (mg/dl)
Before 44.8 ± 9.1 0.16 42.8 ± 6.4 0.3 0.33
After 46.2 ± 8.3 44.2 ± 7.6 0.32
Differences 1.3 ± 5.5 1.3 ± 7.0 0.97
LDL (mg/dl)
Before 107.7 ± 31.6 0.16 108.6 ± 43.7 0.54 0.93
After 98.9 ± 27.2 112.7 ± 37.0 0.09
Differences 8.8 ± 35.3 4.1 ± 37.3 0.16
HDL/total cholesterol ratio
Before 0.24 ± 0.06 0.005 0.23 ± 0.06 0.92 0.65
After 0.27 ± 0.06 0.23 ± 0.06 0.03
Differences 0.02 ± 0.05 0.0007 ± 0.03 0.02
LDL/HDL ratio
Before 2.4 ± 0.8 0.17 2.58 ± 1.0 0.94 0.67
After 2.2 ± 0.8 2.59 ± 0.9 0.11
Differences 0.2 ± 0.9 0.01 ± 0.9 0.29
Data are presented as mean ± SD.
p* Within group comparison (paired t-test or Wilcoxon test).
pyBetween group comparison (independent t-test or Mann–Whitney
test).
pValue 50.05 is significant.
Table 5. CRP, TNFaand PGE
2
levels in ginger and placebo groups
before and after intervention.
Variable
Ginger group
(n¼33) p*
Placebo group
(n¼30) p*py
CRP (mg/L)
Before 5.2 ± 5.4 0.004 7.1 ± 9.6 0.01 0.99
After 2.6 ± 3.2 6.4 ± 8.4 0.01
Differences 2.5 ± 4.7 0.7 ± 6.5 0.02
TNFa(pg/ml)
Before 17.6 ± 26.3 0.11 23.6 ± 24.6 0.63 0.07
After 11.8 ± 13.5 26.6 ± 26.5 0.005
Differences 5.7 ± 15.8 3.0 ± 18.4 0.12
PGE
2
(pg/ml)
Before 234.9 ± 225.7 0.000 221.9 ± 127.5 0.07 0.42
After 108.1 ± 205.6 200.5 ± 129.6 0.000
Differences 126.7 ± 231.6 21.4 ± 60.4 0.009
Data are presented as mean ± SD.
p* within group comparison (Wilcoxon test).
pybetween group comparison (Mann–Whitney test).
pValue 50.05 is significant.
4T. Arablou et al. Int J Food Sci Nutr, Early Online: 1–6
Int J Food Sci Nutr Downloaded from informahealthcare.com by 194.225.187.233 on 04/27/14
For personal use only.
of ginger on HDL, are similar to findings of Mahluji et al. (2013),
Alizadeh et al. (2008) and Bordia et al. (1997). Increased levels of
insulin and TG are associated with reduced HDL
2
and HDL
particle size (Pascot et al., 2001). Probably ginger increases HDL
2
and HDL particle size and reduces atherogenesis through
decreasing insulin and TG levels.
Although Mahluji et al. (2013) showed that taking 2 g of ginger
per day for 2 months could reduce LDL in diabetic patients, in our
study, LDL reduced in ginger group compared with placebo group
but it was not statistically significant. It is consistent with the
findings of Alizadeh et al. (2008) and Bordia et al. (1997). Ginger
may increase LDL particle size and reduce uptake of oxidized
LDL by macrophages (Fuhrman et al., 2000).
The results showed that ginger reduced plasma PGE
2
signifi-
cantly in diabetic patients. So far, as our knowledge, the effect of
ginger on PGE
2
levels in diabetic humans has not been studied.
Black et al. (2010) studied the effects of daily consumption of 2 g
of raw or heated ginger on muscle pain and plasma PGE
2
in
participants. After 11 days, there were no significant differences
in plasma levels of PGE
2
between two groups. Ginger’s effect on
PGE
2
have addressed in several animal and cellular studies
(Shen et al., 2005; Shimoda et al., 2010; Thomson et al., 2002).
Lantz et al. (2007) demonstrated that PGE
2
production was
inhibited in U937 cell cultures containing standard compounds of
ginger (6, 8, 10 gingerols and 6-shogaol). It also became clear that
inhibition of PGE
2
production in mixed ginger extract was greater
than either of the compounds alone. Although this study did not
show the suppression of TNFaproduction by ginger, several
cellular studies showed the effect of ginger on TNFareduction
(Habib et al., 2008; Tripathi et al., 2007). Levy & Simon (2009)
found that 6-shogaol can reduce TNFalevels in macrophages
activated with lipopolysaccharide.
However, we showed that consumption of ginger powder for
12 weeks can cause a significant reduction in CRP in patients with
type 2 diabetes. Our result is similar to the result of Atashak et al.
(2010). They showed that consumption of 1 g of powdered ginger
daily for 10 weeks made 27.6% reduction in mean CRP levels in
obese men.
Chronic, low grade inflammation and activation of the innate
immune system are closely involved in the pathogenesis of
diabetes (Navarro & Mora, 2006). Anti-inflammatory properties
of ginger have been known for centuries (Ali et al., 2008). Effect
of ginger on PGE
2
takes place through inhibition of cycloox-
ygenase-2 mRNA expression and direct inhibition of this enzyme
activity (Lantz et al., 2007). It seems that the effect of ginger on
inflammation is also due to the effect of certain active compounds
(gingerols and zerumbone) that inhibit NF-kB. Compounds in
ginger inhibit NF-kB and TNFaexpression in liver cancer cells
(Habib et al., 2008). 6-gingerol and 6-paradol have strong anti-
inflammatory activity and suppress TNFaproduction (Kim et al.,
2004; Park et al., 1998). TNFagene inhibition by ginger, reduces
NF-kB activity and thereby other inflammatory cytokines and so
cyclooxygenase 2 and its related products such as PGE
2
. Acute-
phase proteins such as CRP are also inhibited. This reduces
inflammation and its complications in diabetic patients.
Conclusion
The present study showed that ginger can reduce blood glucose
and insulin levels and improve insulin sensitivity, reduce serum
total cholesterol and TG and decrease inflammation by reducing
CRP and PGE
2
levels in type 2 diabetic patients.
Acknowledgements
We thank the efforts of all the teachers and friends who were with us in
this study, especially the staff of Zanjan Metabolic Disease Research
Center. This is a report of a database from thesis entitled ‘‘The effect of
ginger consumption on glycemic status, lipid profile and some inflam-
matory markers in patients with type 2 diabetes mellitus’’.
Declaration of interest
The authors declare no conflict of interest. The study was funded by
Tehran University of Medical Sciences and Zanjan Metabolic Disease
Research Center, Zanjan University of Medical Sciences.
References
Al-Amin ZM, Thomson M, Al-Qattan KK, Peltonen-Shalaby R, Ali M.
2006. Anti-diabetic and hypolipidaemic properties of ginger (Zingiber
officinale) in streptozotocin-induced diabetic rats. Br J Nutr 96:
660–666.
Ali BH, Blunden G, Tanira MO, Nemmar A. 2008. Some phytochemical,
pharmacological and toxicological properties of ginger (Zingiber
officinale Roscoe): a review of recent research. Food Chem Toxicol
46:409–420.
Alizadeh-Navaei R, Roozbeh F, Saravi M, Pouramir M, Jalali F,
Moghadamnia AA. 2008. Investigation of the effect of ginger on the
lipid levels. A double blind controlled clinical trial. Saudi Med J 29:
1280–1284.
Atashak S, Piri M, Jafari A, Azarbayjani MA. 2010. Effects of 10 week
resistance training and ginger consumption on C-reactive protein and
some cardiovascular risk factors in obese men. PPJ (Iranian Society of
Physiology and Pharmacology) 14:318–328.
Bhandari U, Kanojia R, Pillai KK. 2005. Effect of ethanolic extract of
Zingiber officinale on dyslipidaemia in diabetic rats. J Ethnopharmacol
97:227–230.
Black C, Herring M, Hurley D, O’Connor P. 2010. Ginger (Zingiber
officinale) reduces muscle pain caused by eccentric exercise. Pain 11:
894–903.
Bordia A, Verma SK, Srivastava KC. 1997. Effect of ginger (Zingiber
officinale Rosc.) and fenugreek (Trigonella foenumgraecum L.) on
blood lipids, blood sugar and platelet aggregation in patients with
coronary artery disease. Prostaglandins Leukot Essent Fatty Acids 56:
379–384.
ElRokh el-SM, Yassin NA, El-Shenawy SM, Ibrahim BM. 2010.
Antihypercholesterolaemic effect of ginger rhizome (Zingiber offici-
nale) in rats. Inflammopharmacology 18:309–315.
Fuhrman B, Rosenblat M, Hayek T, Coleman R, Aviram M. 2000.
Ginger extract consumption reduces plasma cholesterol, inhibits LDL
oxidation and attenuates development of atherosclerosis in atheroscler-
otic, apolipoprotein E-deficient mice. J Nutr 130:1124–1131.
Gao H, Guan T, Li C, Zuo G, Yamahara J, Wang J, Li Y. 2012. Treatment
with ginger ameliorates fructose-induced fatty liver and hypertrigly-
ceridemia in rats: modulation of the hepatic carbohydrate response
element-binding protein-mediated pathway. Evid Based Complement
Altern Med 2012: article ID 570948, doi:10.1155/2012/570948.
Goyal RK, Kadnur SV. 2006. Beneficial effects of Zingiber officinale on
goldthioglucose induced obesity. Fitoterapia 77:160–163.
Habib SHM, Makpol S, Hamid NAA, Das S, Ngah WZW, YusofI YAM.
2008. Ginger extract (Zingiber Officinale) has anti-cancer and anti-
inflammatory effects on ethionine-induced hepatoma rats. Clinics 63:
807–813.
Isa Y, Miyakawa Y, Yanagisawa M, Goto T, Kang M, Kawada T. 2008.
6-Shogaol and 6-gingerol, the pungent of ginger, inhibit TNF-a
mediated down regulation of adiponectin expression via different
mechanisms in 3T3-L1 adipocytes. Biochem Biophys Res Commun
373:429–434.
Islam MS, Choi H. 2008. Comparative effects of dietary ginger (Zingiber
officinale) and garlic (Allium sativum) investigated in a type 2 diabetes
model of rats. J Med Food 11:152–159.
Kim S, Chun K, Kundu J, Surh Y. 2004. Inhibitory effects pg 6-gingerol
on PMA-induced COX-2 expression and activation of NFkB and p38
MAPK in skin mouse. Biofactors 21:27–31.
Lantz RC, Chen GJ, Sarihan M, Solyom AM, Jolad SD, Timmermann
BN. 2007. The effect of extracts from ginger rhizome on inflammatory
mediator production. Phytomedicine 14:123–128.
Levy A, Simon O. 2009. Six-shogaol inhibits production of tumour
necrosis factor alpha, interleukin-1 beta and nitric oxide from
lipopolysaccharide-stimulated RAW 264.7 macrophages. West Indian
Med J 58:295–300.
DOI: 10.3109/09637486.2014.880671 Effect of ginger consumption in patients with type 2 diabetes mellitus 5
Int J Food Sci Nutr Downloaded from informahealthcare.com by 194.225.187.233 on 04/27/14
For personal use only.
Li Y, Tran VH, Duke CC, Roufogalis BD. 2012. Preventive and protective
properties of Zingiber officinale (ginger) in diabetes mellitus, diabetic
complications, and associated lipid and other metabolic disorders: a
brief review. Evid Based Complement Altern Med 2012: article ID
516870, doi:10.1155/2012/516870.
Mahluji S, Attari VE, Mobasseri M, Payahoo L, Ostadrahimi A, Golzari
SE. 2013. Effects of ginger (Zingiber officinale) on plasma glucose
level, HbA1c and insulin sensitivity in type 2 diabetic patients. Int J
Food Sci Nutr 2013. 64:682–686.
Nammi S, Sreemantula S, Roufogalis BD. 2009. Protective effects of
ethanolic extract of Zingiber officinale rhizome on the development of
metabolic syndrome in high-fat diet-fed rats. Basic Clin Pharmacol
Toxicol 104:366–373.
Navarro JF, Mora C. 2006. Diabetes, inflammation, proinflammatory
cytokines, and diabetic nephropathy. Sci World J 6:908–917.
Park K, Chun K, Lee S, Surh Y. 1998. Inhibitory effect of [6]-gingerol, a
major pungent principle of ginger, on phorbol ester-induced inflam-
mation, epidermal ornithine decarboxylase activity and skin tumor
promotion in ICR mice. Cancer Lett 129:139–144.
Pascot A, Lemieux I, Prud’homme D, Tremblay A, Nadeau A, Couillard
C, Bergeron J, et al. 2001. Reduced HDL particle size as an additional
feature of the atherogenic dyslipidemia of abdominal obesity. J Lipid
Res 42:2007–2014.
Shanmugam KR, Mallikarjuna K, Kesireddy N, Sathyavelu Reddy K.
2011. Neuroprotective effect of ginger on anti-oxidant enzymes
in streptozotocin-induced diabetic rats. Food Chem Toxicol 49:
893–897.
Sharma I, Gusain D, Dixit VP. 1991. Hypolipidaemic and antiathero-
sclerotic effects of plumbagin in rabbits. Indian J Physiol Pharmacol
35:10–14.
Shen C-L, Hong K-J, Kim SW. 2005. Comparative effects of ginger root
(Zingiber officinale Rosc.) on the production of inflammatory medi-
ators in normal and osteoarthrotic sow chondrocytes. J Med Food 8:
149–153.
Shimoda H, Shan SJ, Tanaka J, Seki A, Seo JW, Kasajima N, Tamura S,
et al. 2010. Anti-inflammatory properties of red ginger (Zingiber
officinale var. Rubra) extract and suppression of nitric oxide production
by its constituents. J Med Food 13:156–162.
Shirdel Z, Mirbadalzadeh R, Madani H. 2009. Antidiabetic and
antilipidemic effect of ginger in alloxan monohydrate diabetic rats in
comparison with glibenclamide. Iran J Diabetes Lipid Disord 9:7–15.
Simin L, Tinker L, Song Y, Rifai N, Bonds DE, Cook NR, Heiss G, et al.
2007. A prospective study of inflammatory cytokines and diabetes
mellitus in a multiethnic cohort of postmenopausal women. Arch
Intern Med 167:1676–1685.
Singletary K. 2010. Ginger an overview of health benefits. Nutr Today
45:171–183.
Thomson M, Al-Qattan KK, Al-Sawan SM, Alnaqeeb MA, Khan I,
Ali M. 2002. The use of ginger (Zingiber officinale Rosc.) as a
potential anti-inflammatory and antithrombotic agent. Prostaglandins
Leukot Essent Fatty Acids 67:475–478.
Tripathi S, Bruch D, Kittur DS. 2007. Ginger extract inhibits LPS induced
macrophage activation and function. BMC Complement Altern Med 8:
1–7.
6T. Arablou et al. Int J Food Sci Nutr, Early Online: 1–6
Int J Food Sci Nutr Downloaded from informahealthcare.com by 194.225.187.233 on 04/27/14
For personal use only.
... Mahluji et al. reported no significant effect of ginger powder on the FBG, TG, HDL, and HbA1c levels when investigating the effect of 2 g/d of ginger for 12 weeks on blood glucose and lipid profiles [70]. In contrast, Arablou et al. assessed the effect of 1.6 g/day of ginger powder for 12 weeks on inflammatory markers, blood glucose, and lipid profiles to find that ginger powder decreased the FBG, HbA1c, TG, total cholesterol, and CRP levels significantly, while it had no significant effect on HDL and LDL [72]. Moreover, Mozaffari and his colleagues proved that the consumption of 3 g/day of ginger powder for 8 weeks caused a significant decrease in FBG and HbA1c levels, while no significant effect was noted for weight and BMI [73]. ...
... Qidwai et al. concluded no significant effect of black seed powder on the previously mentioned parameters [76]. Previous studies that involved less than 12 weeks or 2 g or less of the spice powders in their intervention recommended longer interventional periods and a higher dose of the spice powders to exhibit a significant effect of spice powder consumption on the lipid profile, blood glucose, WC, weight, and BMI [63,[72][73][74][75]. ...
Article
Full-text available
Non-communicable diseases (NCDs) place a significant burden on global health and the healthcare systems which support it. Metabolic syndrome is a major risk factor for a large number of NCDs; however, treatments remain limited. Previous research has shown the protective benefits of edible dietary spices on key components of metabolic syndrome. Therefore we performed a 12-week double-blind, placebo-controlled, randomized, clinical trial to evaluate the effect of ginger (Zingiber officinale), cinnamon (Cinnamomum), and black seed (Nigella sativa) consumption on blood glucose, lipid profiles, and body composition in 120 participants with, or at risk of, metabolic syndrome. Each participant consumed 3 g/day of powder (spice or placebo). Data related to different parameters were collected from participants at the baseline, midpoint, and endpoint of the intervention. Over the 12-week interventions, there was an improvement in a number of biochemical indices of metabolic syndrome, including fasting blood glucose, HbA1c, LCL, and total cholesterol associated with supplementation with the spices when compared to a placebo. This study provides evidence to support the adjunct use of supplementation for those at risk of metabolic syndrome and its sequelae.
... In particular, in two studies, T2DM patients decreased FBG, HbA1c, insulin and HOMA-IR after receive a daily capsule with 1.6 or 1 g/day of ginger, respectively. In the first study, 33 subjects received the treatment and 30 were controls (60), meanwhile in the second one, the sample were 39 subjects and the control 31 (63). Moreover, specifically, the three gingerol derivatives, namely paradol, (E)-shogaol, and (5R)-acetoxy-gingerol, were able to inhibit human and mouse 11b-HSD1 activities (IC 50 values ranged between 1.09 and 1.30 mM) (120). ...
Article
Full-text available
Type 2 diabetes Mellitus (T2DM) prevalence has significantly increased worldwide in recent years due to population age, obesity, and modern sedentary lifestyles. The projections estimate that 439 million people will be diabetic in 2030. T2DM is characterized by an impaired β-pancreatic cell function and insulin secretion, hyperglycemia and insulin resistance, and recently the epigenetic regulation of β-pancreatic cells differentiation has been underlined as being involved. It is currently known that several bioactive molecules, widely abundant in plants used as food or infusions, have a key role in histone modification and DNA methylation, and constituted potential epidrugs candidates against T2DM. In this sense, in this review the epigenetic mechanisms involved in T2DM and protein targets are reviewed, with special focus in studies addressing the potential use of phytochemicals as epidrugs that prevent and/or control T2DM in vivo and in vitro. As main findings, and although some controversial results have been found, bioactive molecules with epigenetic regulatory function, appear to be a potential replacement/complementary therapy of pharmacological hypoglycemic drugs, with minimal side effects. Indeed, natural epidrugs have shown to prevent or delay the T2DM development and the morbidity associated to dysfunction of blood vessels, eyes and kidneys due to sustained hyperglycemia in T2DM patients.
... This gingerol gives a spicy taste to fresh ginger which is a phenolic compound (69). Gingerol is very susceptible to thermal decomposition (70). The compound 6-gingerol is reported to have antioxidant and antiinflammatory properties (60, 71). ...
Article
Full-text available
Ginger (Zingiber officinale) is a rhizome that has been used as a healthy herbal plant for years. Ginger’s chemical components are recognized to provide beneficial health effects, namely as antioxidants and anti-inflammatory agents with the potential to operate as immunomodulators. This literature review covers numerous publications concerning ginger’s immunomodulatory potential, associated with antioxidant and anti-inflammatory effects in modifying the body’s immune system. Pathophysiology of oxidative stress and inflammation were introduced before diving deep down into the herbal plants as an immunomodulator. Ginger’s antioxidant and anti-inflammatory properties are provided by gingerol, shogaols, paradol, and zingerone. Ginger’s antioxidant mechanism is linked to Nrf2 signaling pathway activation. Its anti-inflammatory mechanism is linked to Akt inhibition and NF-KB activation, triggering the release of anti-inflammatory cytokines while reducing proinflammatory cytokines. Ginger consumption as food and drink was also explored. Overall, ginger and its active components have been shown to have strong antioxidant properties and the potential to reduce inflammation. Challenges and future prospects of ginger are also elaborated for future development. Future collaborations between researchers from various fields, including chemists, biologists, clinicians, pharmacists, and the food industry, are required further to investigate the effect of ginger on human immunity. Collaboration between researchers and industry can help accelerate the advancement of ginger applications.
... Furthermore, some beneficial activities like antioxidant activities or reduction in triglycerides and insulin resistance are also observed in ginger [16][17][18]. The analgesic action of ginger seems to come from several mechanisms, including cyclooxygenase (COX) pathways which inhibit arachidonic acid metabolism that is similar to the non-steroidal anti-inflammatory drugs [17]. ...
Conference Paper
Full-text available
Migraine is a common neuromuscular disorder characterized by recurrent headaches and is known as one of the most common causes of disability and a major public health problem. Objective: To study the effects of ginger in the management of migraine, the efficacy of using ginger on patients with migraine headaches was assessed in the present systematic review. To source the studies, data search was performed through the following databases using the relevant terms: PubMed/Medline, Scopus, EMBASE, and Science Direct. We identified 5 studies. The overall data suggest some favorable effects of ginger in migraine treatment. Ginger could promote a reduction in pain severity and migraine-associated symptoms. Also, ginger was well tolerated in the most patients. Our study suggests that ginger administration has beneficial effects on different aspects of this complicated disorder. However, the results are insufficient to make a firm conclusion.
... Although clinical studies on gingerol's efficacy in diabetes management are limited, emerging research indicates its potential in reducing cholesterol and eicosanoid levels, with doses ranging from 100 milligrams to 3 grams (49). Clinical trials have further demonstrated that ginger supplementation can significantly improve insulin sensitivity and lower levels of insulin, HOMA index, haemoglobin A1C, prostaglandin E2, and C-reactive protein in diabetic patients (50,51). However, the impact of ginger on fasting plasma glucose (FPG) and HbA1C levels has shown variability, indicating the need for extended research to ascertain its full therapeutic potential (52,53). ...
Article
Full-text available
Background: Diabetes mellitus and atherosclerosis are significant contributors to global morbidity and mortality, with their prevalence escalating alongside the increasing incidence of obesity and sedentary lifestyles. Traditional therapeutic approaches have had limited success in fully managing these conditions, prompting interest in natural compounds such as capsaicin and gingerol. These compounds have been identified for their potential therapeutic effects, including anti-inflammatory, antioxidant, and metabolic regulatory properties. Objective: This review aims to critically assess the impact of capsaicin and gingerol on the management of diabetes and atherosclerosis, highlighting their mechanisms of action, therapeutic potential, and clinical applications. Methods: A comprehensive literature search was conducted across multiple databases, including PubMed, Scopus, Web of Science, and Google Scholar, focusing on studies that evaluated the effects of capsaicin and gingerol on diabetes and atherosclerosis. The search strategy incorporated a combination of keywords and MeSH terms related to the compounds and diseases of interest. Studies were selected based on predefined inclusion and exclusion criteria, with data extraction and quality assessment conducted according to standardized protocols. Results: The review synthesized evidence from both animal models and human clinical trials, demonstrating that capsaicin and gingerol exhibit significant potential in improving metabolic health, reducing inflammation, and mitigating the risk factors associated with diabetes and atherosclerosis. Capsaicin was found to enhance weight management and cardiovascular health through mechanisms involving the sympathetic nervous system and TRPV1 activation. Gingerol showed potent anti-inflammatory effects and improved glucose and lipid metabolism. However, the clinical applicability of these findings requires further investigation to determine optimal dosages and long-term effects. Conclusion: Capsaicin and gingerol present promising natural therapeutic options for the management of diabetes and atherosclerosis, with potential benefits extending beyond traditional treatment modalities. Further clinical trials are necessary to fully elucidate their therapeutic potential and integrate them into clinical practice.
Article
Full-text available
Background: The Mediterranean Diet (MedDiet) is the dietary pattern par excellence for managing and preventing metabolic diseases, such as Type 2 Diabetes (T2DM). The MedDiet incorporates spices and aromatic herbs, which are abundant sources of bioactive compounds. The aim of this study was to analyze the effect of all aromatic herbs and spices included in the MedDiet, such as black cumin, clove, parsley, saffron, thyme, ginger, black pepper, rosemary, turmeric, basil, oregano, and cinnamon, on the glycemic profile in T2DM subjects. Methods: PubMed, Web of Science, and Scopus databases were searched for interventional studies investigating the effect of these aromatic herbs and spices on the glycemic profile in T2DM subjects. Results: This systematic review retrieved 6958 studies, of which 77 were included in the qualitative synthesis and 45 were included in the meta-analysis. Our results showed that cinnamon, turmeric, ginger, black cumin, and saffron significantly improved the fasting glucose levels in T2DM subjects. The most significant decreases in fasting glucose were achieved after supplementation with black cumin, followed by cinnamon and ginger, which achieved a decrease of between 27 and 17 mg/dL. Conclusions: Only ginger and black cumin reported a significant improvement in glycated hemoglobin, and only cinnamon and ginger showed a significant decrease in insulin.
Article
Full-text available
Ginger is rich in secondary metabolites such as 6-gingerol, 6-shogaol, 10-gingerol, gingerdiones, gingerdiols, paradols, 6-dehydrogingerols, 5-acetoxy-6-gingerol, 3,5-diacetoxy-6-gingerdiol, and 12-gingerol. These compounds are largely responsible for the evidenced pharmacological properties of ginger. Among them, 6-shogaol and 6-gingerol are considered the most crucial active ingredients. Research supports ginger's medicinal properties, particularly its antioxidant and anti-inflammatory activities. On the other hand, the neuroprotective benefits of ginger, while intriguing, remain relatively unexplored and understudied. Neurodegenerative disorders (NDs), characterized by increased oxidative stress, neuro-inflammation, and protein misfolding, are becoming increasingly prevalent as life expectancy rises. Ginger's phytochemicals show promise in treating NDs due to their ability to target specific ligand sites, as suggested by their structure-activity relationships. The bioactive compounds in ginger may alleviate neurological symptoms and pathological disorders by influencing markers associated with cell death or survival. Furthermore, ginger's potential to enhance cognitive clarity could be attributed to its impact on the monoamine and cholinergic systems in different brain regions. It also appears to decrease the incidence of inflammation-related components. This review aims to highlight how ginger could be leveraged to treat major neurological conditions such as multiple sclerosis, Alzheimer's disease, and Parkinson's disease.
Article
Context Ginger consumption may have an inverse relationship with obesity and metabolic syndrome parameters; however, clinical trials have reported contradictory results. Objective To systematically review and analyze randomized controlled trials (RCTs) assessing the effects of ginger on body weight and body composition parameters. Methods Databases were searched for appropriate articles up to August 20, 2022. All selected RCTs investigated the impact of ginger on glycemic indices in adults. A random effects model was used to conduct a meta-analysis, and heterogeneity was assessed using the I2 statistic. Net changes in body weight, body mass index (BMI), waist circumference (WC), and percent body fat were used to calculate the effect size, which was reported as a weighted mean difference (WMD) and 95% confidence intervals (CIs). The risk of bias was assessed. Results A total of 27 RCTs involving 1309 participants were included. The certainty in the evidence was very low for WC and BMI, and low for body weight and percent body fat as assessed by the GRADE evidence profiles. The meta-analysis showed a significant association between ginger supplementation and a reduction in body weight (WMD, –1.52 kg; 95%CI, –2.37, –0.66; P < 0.001), BMI (WMD, –0.58 kg/m2; 95%CI, –0.87, –0.30; P < 0.001), WC (WMD, –1.04 cm; 95%CI: –1.93, –0.15; P = 0.021), and percent body fat consumption (WMD, –0.87%; 95%CI, –1.71, –0.03; P = 0.042). The results of the nonlinear dose-response analysis showed a significant association between the ginger dose with body weight (Pnonlinearity = 0.019) and WC (Pnonlinearity = 0.042). The effective dose of ginger intervention for body mass reduction was determined to be 2 g/d in dose-response analysis. The duration of ginger intervention had a significant nonlinear relationship with body weight (Pnonlinearity = 0.028) with an effective duration of longer than 8 weeks. Conclusions Our findings provide evidence that ginger consumption may significantly affect body composition parameters nonlinearly. More, well-constructed RCTs are needed.
Article
Full-text available
Introduction: The use of exercise along with herbal supplements is one of the recommended methods for controlling obesity and its complications, but its effects have been controversial due to the diversity of training programs and also herbal supplements. Hence, the objective of this study was to investigate the effects of 10 week ginger and progressive resistance training on C-reactive protein (CRP) and other cardiovascular risk factors in obese men. Methods: In a double-blind design, 32 obese men (BMI≥ 30) were selected and randomized to four groups (each group comprised of 8 subjects): 1- ginger (GI) 2- resistance training plus placebo (PLRT) 3- resistance training plus ginger (RTPL) and 4- placebo (PL). Subjects of groups 1 and 3 consumed 1 gr ginger/d for 10 weeks, while subjects of groups 2 and 3 performed progressive resistance training at the same time. To evaluate lipid profiles, insulin resistance and CRP, blood samples were collected at the beginning of the first week and after the last week. Moreover, body composition and anthropometric indices were measured simultaneously. Results: After 10 weeks of interventions, both GRT and PLRT groups showed a significant decrease in WC, WHR, body fat percent, body fat mass, total cholesterol and insulin resistance, while these remained unchanged in two PL and GI groups (P>0.05). Moreover, significant decreases in the mean values of CRP were observed in all groups except the placebo group (P<0.05). Conclusion: According to our results, resistance training was a preventive approach to reduce the cardiovascular risk in obese men. Moreover, ginger supplementation did not have any influence on the lipid profile and insulin resistance at a dose of 1 gr/day, however, it exerted favorable effects on CRP in obese men.
Article
Full-text available
The present study was aimed to evaluate the effects of Zingiber officinale on some biochemical parameters in type 2 diabetic (DM2) patients. In a randomized double-blind placebo controlled trial, 64 patients with DM2 were assigned to ginger or placebo groups (receiving 2 g/d of each). A 3 d diet record, anthropometric measurements and concentrations of fasting blood glucose (FPG), HbA1c, lipid profile (including total cholesterol, triglyceride, low density lipoprotein and high density lipoprotein) and also the homeostasis model assessment (HOMA) and quantitative insulin-sensitivity check index (QUICKI) were determined before and after 2 months of intervention. Ginger supplementation significantly lowered the levels of insulin (11.0 AE 2.3 versus 12.1 AE 3.3; p ¼ 0.001), LDL-C (67.8 AE 27.2 versus 89.2 AE 24.9; p ¼ 0.04), TG (127.7 AE 43.7 versus 128.2 AE 37.7; p ¼ 0.03) and the HOMA index (3.9 AE 1.09 versus 4.5 AE 1.8; p ¼ 0.002) and increased the QUICKI index (0.313 AE 0.012 versus 0.308 AE 0.012; p ¼ 0.005) in comparison to the control group; while, there were no significant changes in FPG, TC, HDL-C and HbA1c (p40.05). In summary, ginger supplementation improved insulin sensitivity and some fractions of lipid profile in DM2 patients. Therefore it may be considered as a useful remedy to reduce the secondary complications of DM2.
Article
Full-text available
Zingiber officinale (ginger) has been used as herbal medicine to treat various ailments worldwide since antiquity. Recent evidence revealed the potential of ginger for treatment of diabetes mellitus. Data from in vitro, in vivo , and clinical trials has demonstrated the antihyperglycaemic effect of ginger. The mechanisms underlying these actions are associated with insulin release and action, and improved carbohydrate and lipid metabolism. The most active ingredients in ginger are the pungent principles, gingerols, and shogaol. Ginger has shown prominent protective effects on diabetic liver, kidney, eye, and neural system complications. The pharmacokinetics, bioavailability, and the safety issues of ginger are also discussed in this update.
Article
Full-text available
Ginger has been demonstrated to improve lipid derangements. However, its underlying triglyceride-lowering mechanisms remain unclear. Fructose overconsumption is associated with increase in hepatic de novo lipogenesis, thereby resulting in lipid derangements. Here we found that coadministration of the alcoholic extract of ginger (50 mg/kg/day, oral gavage, once daily) over 5 weeks reversed liquid fructose-induced increase in plasma triglyceride and glucose concentrations and hepatic triglyceride content in rats. Plasma nonesterified fatty acid concentration was also decreased. Attenuation of the increased vacuolization and Oil Red O staining area was evident on histological examination of liver in ginger-treated rats. However, ginger treatment did not affect chow intake and body weight. Further, ginger treatment suppressed fructose-stimulated overexpression of carbohydrate response element-binding protein (ChREBP) at the mRNA and protein levels in the liver. Consequently, hepatic expression of the ChREBP-targeted lipogenic genes responsible for fatty acid biosynthesis was also downregulated. In contrast, expression of neither peroxisome proliferator-activated receptor- (PPAR-) alpha and its downstream genes, nor PPAR-gamma and sterol regulatory element-binding protein 1c was altered. Thus the present findings suggest that in rats, amelioration of fructose-induced fatty liver and hypertriglyceridemia by ginger treatment involves modulation of the hepatic ChREBP-mediated pathway.
Article
Full-text available
Many herbal medicinal products have potential hypocholesterolaemic activity and encouraging safety profiles. However, only a limited amount of clinical research exists to support their efficacy. The present study was designed to evaluate the antihypercholesterolaemic effects of aqueous ginger (Zingiber officinale) infusion in hypercholesterolaemic rat models. 48 rats were used throughout the experiment, which were divided into six groups, eight animals each as follows: normal control group (normal rats which fed with standard diet). After induction of hypercholesterolaemia by feeding rats with high cholesterol diet, the remaining rats were divided into five groups: group 1, hypercholesterolaemic control group (hypercholesterolaemic rats group); groups 2, 3 and 4, rats were given aqueous infusion of ginger (100, 200 and 400 mg/kg, respectively) orally; and group 5, rats were given atorvastatin (0.18 mg/kg) orally as a reference antihypercholesterolaemic drug. The blood was obtained from all groups of rats after being lightly anaesthetized with ether and the following lipid profile [serum total cholesterol (TC), HDL-cholesterol (HDL-C), LDL-C and triglyceride levels] was measured at zero time and 2 and 4 weeks after ginger and atorvastatin treatment, and the risk ratio (TC/HDL-cholesterol) was assessed. The results revealed that the hypercholesterolaemic rats treated with aqueous ginger infusion in the three doses used after 2 and 4 weeks of treatment induce significant decrease in all lipid profile parameters which were measured and improved the risk ratio.
Article
[6]-Gingerol, a major pungent ingredient of ginger (Zingiber officinale Roscoe, Zingiberaceae), has a wide array of pharmacologic effects. Previous studies have demonstrated that [6]-gingerol inhibits mouse skin tumor promotion and anchorage-independent growth of cultured mouse epidermal cells stimulated with epidermal growth factor. Cyclooxygenase-2 (COX-2), a key enzyme in the prostaglandin biosynthesis, has been recognized as a molecular target for many anti-inflammatory as well as chemopreventive agents. Topical application of [6]-gingerol inhibited phorbol 12-myristate 13-acetate -induced COX-2 expression. One of the essential transcription factors responsible for COX-2 induction is NF-κB. [6]-Gingerol suppressed NF-κB DNA binding activity in mouse skin. In addition, [6]-gingerol inhibited the phoshorylation of p38 mitogen-activated protein kinase which may account for its inactivation of NF-κB and suppression of COX-2 expression.
Article
Ginger (Zingiber officinale Roscoe) is a member of the Zingiberaceae family of plants. It has been a part of healing strategies in Asia, India, Europe, and the Middle East for centuries for treatment of such disorders as arthritis, stomach upset, asthma, diabetes, and menstrual irregularities, to name a few. There is scientific support that ginger may alleviate the symptoms of nausea and vomiting following pregnancy, surgery, cancer therapy, or motion sickness and suggestive evidence that ginger reduces inflammation and pain. Cell culture studies show that ginger has antioxidant properties. However, it is not known whether ginger antioxidant constituents are bioavailable in humans once ingested and whether they can affect markers of oxidative stress in human in vivo. There are preliminary data that ginger has antimicrobial potential, although there is little evidence supporting ginger's practical usefulness in combating infections in humans. Based on evidence primarily from animal and in vitro studies, ginger may have beneficial effects toward cardiovascular disease through its multiple actions counteracting inflammation, hyperlipidemia, platelet aggregation, and hypertension. Overall, based on the current body of scientific literature, more information is needed from clinical studies to confirm these promising multiple health benefits of ginger in human subjects and the doses that are most efficacious
Article
The aim of the present study was to investigate the effect of ginger on oxidative stress markers in the mitochondrial fractions of cerebral cortex (CC), cerebellum (CB), hippocampus (HC) and hypothalamus (HT) of diabetic rats. Diabetes exacerbates neuronal injury induced by hyperglycemia mediated oxidative damage. A marked decrease in anti-oxidant marker enzymes, superoxide dismutase (SOD), catalase (CAT), glutathione peroxidase (GPx), glutathione reductase (GR), reduced glutathione (GSH) and increase in malondialdehyde (MDA) was observed in the diabetic rats. Decreased activities of anti-oxidant enzymes in diabetic rats were augmented on oral administration of ginger. Moreover, ginger administration depleted the MDA level, which was earlier increased in the diabetic rats. These results suggest that ginger exhibit a neuroprotective effect by accelerating brain anti-oxidant defense mechanisms and down regulating the MDA levels to the normal levels in the diabetic rats. Thus, ginger may be used as therapeutic agent in preventing complications in diabetic patients.
Article
Unlabelled: Ginger has been shown to exert anti-inflammatory effects in rodents, but its effect on human muscle pain is uncertain. Heat treatment of ginger has been suggested to enhance its hypoalgesic effects. The purpose of this study was to examine the effects of 11 days of raw (study 1) and heat-treated (study 2) ginger supplementation on muscle pain. Study 1 and 2 were identical double-blind, placebo controlled, randomized experiments with 34 and 40 volunteers, respectively. Participants consumed 2 grams of either raw (study 1) or heated (study 2) ginger or placebo for 11 consecutive days. Participants performed 18 eccentric actions of the elbow flexors to induce pain and inflammation. Pain intensity, perceived effort, plasma prostaglandin E(2), arm volume, range-of-motion and isometric strength were assessed prior to and for 3 days after exercise. Results Raw (25%, -.78 SD, P = .041) and heat-treated (23%, -.57 SD, P = .049) ginger resulted in similar pain reductions 24 hours after eccentric exercise compared to placebo. Smaller effects were noted between both types of ginger and placebo on other measures. Daily supplementation with ginger reduced muscle pain caused by eccentric exercise, and this effect was not enhanced by heat treating the ginger. Perspective: This study demonstrates that daily consumption of raw and heat-treated ginger resulted in moderate-to-large reductions in muscle pain following exercise-induced muscle injury. Our findings agree with those showing hypoalgesic effects of ginger in osteoarthritis patients and further demonstrate ginger's effectiveness as a pain reliever.