ArticlePDF Available

The effect of DASH diet on glycemic response, meta-inflammation and serum LPS in obese patients with NAFLD: a double-blind controlled randomized clinical trial

Authors:

Abstract and Figures

Background As dietary approaches to stop hypertension (DASH) dietary pattern has been shown to be effective in hypertension and obesity, the present study investigated the effects of following DASH diet on glycemic, meta-inflammation, lipopolysaccharides (LPS) and liver function in obese patients with non-alcoholic fatty liver disease (NAFLD). Methods In this double-blind controlled randomized clinical trial, 40 obese patients with NAFLD were randomly allocated into either “DASH diet” (n = 20) or calorie-restricted diet as "Control” (n = 20) group for 8 weeks. Anthropometric measures, blood pressure, glycemic response, liver enzymes, toll-like reseptor-4 (TLR-4) and monocyte chemoattractant protein (MCP-1) and LPS as well as Dixon's DASH diet index were assessed at baseline and after 8 weeks. Results After 8 weeks, although all obesity indices decreased significantly in both groups, the reduction in all anthropometric measures were significantly greater in DASH vs control group, after adjusting for baseline values and weight change. Fasting glucose level decreased in both group, however, no inter-group significant difference was found at the end of study. Nevertheless, serum levels of hemoglobin A1c (HbA1c), TLR-4, MCP-1 and LPS as well as aspartate aminotransferase (AST) decreased significantly in DASH group, after adjusting for baseline values and weight change (p < 0.001, p = 0.004, p = 0.027, p = 0.011, and p = 0.008, respectively). The estimated number needed to treats (NNTs) for one and two grade reductions in NAFLD severity following DASH diet were 2.5 and 6.67, respectively. Conclusion Adherence to DASH diet could significantly improve weight, glycemia, inflammation and liver function in obese patients with NAFLD.
This content is subject to copyright. Terms and conditions apply.
Rooholahzadeganetal.
Nutrition & Metabolism (2023) 20:11
https://doi.org/10.1186/s12986-023-00733-4
RESEARCH
© The Author(s) 2023. Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which
permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the
original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or
other third party material in this article are included in the article’s Creative Commons licence, unless indicated otherwise in a credit line
to the material. If material is not included in the article’s Creative Commons licence and your intended use is not permitted by statutory
regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this
licence, visit http:// creat iveco mmons. org/ licen ses/ by/4. 0/. The Creative Commons Public Domain Dedication waiver (http:// creat iveco
mmons. org/ publi cdoma in/ zero/1. 0/) applies to the data made available in this article, unless otherwise stated in a credit line to the data.
Open Access
Nutrition & Metabolism
The eect ofDASH diet onglycemic
response, meta-inammation andserum LPS
inobese patients withNAFLD: adouble-blind
controlled randomized clinical trial
Farnaz Rooholahzadegan1, Sara Arefhosseini1, Helda Tutunchi2, Taghi Badali1, Manuchehr Khoshbaten3 and
Mehrangiz Ebrahimi‑Mameghani4*
Abstract
Background As dietary approaches to stop hypertension (DASH) dietary pattern has been shown to be effective
in hypertension and obesity, the present study investigated the effects of following DASH diet on glycemic, meta‑
inflammation, lipopolysaccharides (LPS) and liver function in obese patients with non‑alcoholic fatty liver disease
(NAFLD).
Methods In this double‑blind controlled randomized clinical trial, 40 obese patients with NAFLD were randomly allo‑
cated into either DASH diet (n = 20) or calorie‑restricted diet as "Control” (n = 20) group for 8 weeks. Anthropometric
measures, blood pressure, glycemic response, liver enzymes, toll‑like reseptor‑4 (TLR‑4) and monocyte chemoattract‑
ant protein (MCP‑1) and LPS as well as Dixon’s DASH diet index were assessed at baseline and after 8 weeks.
Results After 8 weeks, although all obesity indices decreased significantly in both groups, the reduction in all anthro‑
pometric measures were significantly greater in DASH vs control group, after adjusting for baseline values and weight
change. Fasting glucose level decreased in both group, however, no inter‑group significant difference was found at
the end of study. Nevertheless, serum levels of hemoglobin A1c (HbA1c), TLR‑4, MCP‑1 and LPS as well as aspartate
aminotransferase (AST) decreased significantly in DASH group, after adjusting for baseline values and weight change
(p < 0.001, p = 0.004, p = 0.027, p = 0.011, and p = 0.008, respectively). The estimated number needed to treats (NNTs)
for one and two grade reductions in NAFLD severity following DASH diet were 2.5 and 6.67, respectively.
Conclusion Adherence to DASH diet could significantly improve weight, glycemia, inflammation and liver function
in obese patients with NAFLD.
Keywords Dietary approaches to stop hypertension, Glycemic control, Inflammation, Lipoploysaccharides, Non‑
alcoholic fatty liver disease
*Correspondence:
Mehrangiz Ebrahimi‑Mameghani
ebrahimimamagani@tbzmed.ac.ir
1 Student Research Committee, Tabriz University of Medical Sciences,
Tabriz, Iran
2 Endocrine Research Center, Tabriz University of Medical Sciences, Tabriz,
Iran
3 Department of Internal Medicine, Faculty of Medicine, Tabriz University
of Medical Sciences, Tabriz, Iran
4 Nutrition Research Center, Department of Biochemistry and Diet
Therapy, Faculty of Nutrition and Food Sciences, Tabriz University
of Medical Sciences, Tabriz, Iran
Content courtesy of Springer Nature, terms of use apply. Rights reserved.
Page 2 of 14
Rooholahzadeganetal. Nutrition & Metabolism (2023) 20:11
Introduction
Exposure to an “obesogenic” environment such as sus-
tained positive energy balance due to increased food
supply and the overconsumption of energy-dense low
nutrient-dense foods as well as modern sedentary life-
style leads to excessive intrahepatic fat accumulation and
increased adiposity known as nonalcoholic fatty liver
disease (NAFLD), as a public health issue [13]. NAFLD
is considered as an umbrella term that includes differ-
ent types of fatty liver diseases unrelated to alcohol con-
sumption [4]. Due to the metabolic roots of NAFLD, it
has been recently proposed that to rename NAFLD as
metabolic dysfunction-associated fatty liver disease or
’MAFLD’ [5]. A recent systematic review and meta-anal-
ysis through the extraction of available epidemiological
data on fatty liver disease demonstrated that MAFLD
has an astonishingly high prevalence rate in overweight
and obese adults [6]. Moreover, the global prevalence of
NAFLD among general population is projected to raise
up to 33.5%, which is largely related to obesity epidemic
[7]. Genetic and epigenetic factors such as insulin resist-
ance (IR), inflammation, oxidative stress and changes
in gut microbiota are involved in the pathophysiology
of NAFLD known as "Multi-Hit" [8]. IR-disturbances in
intra-cellular insulin signaling pathways- plays a funda-
mental role in NAFLD [9]. Furthermore, simple steatosis
could be followed by a number of metabolic abnormali-
ties such as decreased fatty acid oxidation in the liver,
increased de novo lipogenesis and adipose tissue lypoli-
sis resulted in IR and in turn, IR is associated with other
endocrine and metabolic disorders [9, 10]. e inter-
relationship between obesity and NAFLD has been fre-
quently reported and indicating the role of adipose tissue
in regulating endocrine signaling pathways such as hor-
mones, adipokines and pro-inflammatory cytokine [11,
12]. NAFLD is also called as the hepatic manifestation of
metabolic syndrome (Mets) because of the coexistence
of visceral obesity, IR, dyslipidemia, and hypertension [8,
13]. In addition, hypertrophy/ hyperplasia of adipose tis-
sue is considered as one of the contributing factor in a
low-grade chronic inflammation and metabolic dysfunc-
tion known as “metainflammation” [14]. It is caused by
an increased macrophage accumulation and the release
of adipokines, cytokines and chemokines e.g. tumor
necrosis factor α (TNF-α), toll-like receptors-4 (TLR-4),
monocyte chemoattractant protein (MCP1) and some
interleukins (inc. IL6, IL8, IL1β) [14]. ere is evidence
that weight reduction can be effective in the secretion
of inflammatory markers [14]. TLR-4 plays a vital role
in recognizing lipopolysaccharides (LPS)- an indicator
of pathogenic bacteria invasion- and mediates signaling
to produce pro-inflammatory cytokines [14]. Moreover,
excessive adipose tissues releases free fatty acids (FFAs)
due to degradation of triglycerides [15]. LPS and FFAs
bind to TLR4 of monocytes/macrophages and produce
inflammatory mediators, and therefore, lead to the pro-
longed production of inflammatory cytokines [15].
Despite the lack of an approved therapeutic approach
in the treatment of NAFLD, evidence-based guidelines
establish the fundamental role of lifestyle modifica-
tions, particularly calorie restriction, healthy diet, and
regular physical activity in improving hepatic steatosis
and histological features of NAFLD [16, 17]. In clinical
and preclinical studies, several natural compounds have
shown favorable effects in the prevention, inhibition
and treatment of metabolic disorders [1823]. is evi-
dence represents a promising strategy for NALFD, whose
pathogenesis is multifactorial [24]. Patients with NAFLD
mostly consume diet which is low in whole grains, cere-
als, fruits, and vegetables and high in red meat, refined-
grains and sugars, typically named as Western dietary
pattern [2527]. Moreover, results of a meta-analysis
revealed the role of red meat intake and soft drinks in
increased likelihood of NAFLD [28, 29]. One of the die-
tary strategies that has been studied in NAFLD manage-
ment is the Dietary Approaches to Stop Hypertension
(DASH) dietary pattern approved in the prevention and
treatment of hypertension [30]. Previous studies have also
shown that DASH diet has beneficial effects on several
other disorders including obesity, Mets, type 2 diabetes
mellitus (T2DM), cardiovascular disease, and depres-
sion [3133]. DASH diet underlines fruits, vegetables,
low-fat dairy products, whole grains, poultry, fish, nuts,
seeds, and legumes intakes accompanied by reduction in
fats, red meat, sweets, and sugar-containing drinks [30].
Meanwhile, this diet is low in sodium (< 2400mg/day)
and saturated fat while rich in protein, fiber, calcium,
magnesium, potassium, zinc, and folate [34, 35]. Watz-
inger etal. [36] in a cross-sectional study showed that
the DASH score was correlated to lower liver fat content
and NAFLD. Indeed, in a case–control study, an inverse
association was found between adherence to a DASH-
style diet and odds of NAFLD [37]. Similarly, Maskarinec
etal. [38] in the Multiethnic Cohort Study showed that
higher quality diets during mid-to-late adulthood were
associated with a lower risk of NAFLD. Moreover, results
of a recent nested case–control study in the Multiethnic
Cohort revealed that higher DASH score was negatively
correlated with NAFLD risk [39]. e only interventional
study aimed to assess 8-week adherence to DASH diet
compared with low calorie diet showed improvements in
body weight, liver enzymes, IR, lipid profile, inflamma-
tory and oxidative stress biomarkers [40]. As a system-
atic review and meta-analysis of randomized controlled
clinical trials reported that DASH diet seems to be more
suitable dietary approach for weight loss compared
Content courtesy of Springer Nature, terms of use apply. Rights reserved.
Page 3 of 14
Rooholahzadeganetal. Nutrition & Metabolism (2023) 20:11
with low-energy diets and lack of interventional study
in investigating the effect of DASH diet on NAFLD [41,
42], this study compared the effect of adherence to DASH
diet compared with calorie-restricted diet (CRD) on gly-
cemic response, meta-inflammation and serum LPS in
obese patients with NAFLD.
Materials andmethods
Study design
is double-blinded controlled randomized clinical trial
was designed to examine the adherence to DASH diet
compared with CRD on cardiometabolic, inflammatory
biomarkers and LPS in obese patients with NAFLD. e
study was conducted according to the Declaration of Hel-
sinki and approved by the ethics committee of research
vice-chancellor and also registered in the Iranian Registry
of Clinical Trials (IRCT20100209003320N17). In addi-
tion, an informed consent form was read and signed by
the patients at baseline.
Participants
Sixty two males and females newly diagnosed patients
with mild and moderate NAFLD aged 20–50years with
body mass index (BMI) = 30–40 kg/m2 were enrolled.
NAFLD was confirmed by a single radiologist using
ultrasonography (Sonoace X4 Medisio, South Korea) in a
fasting state and then, liver steatosis severity was catego-
rized into three grades, i.e. grade I as "mild", grade II as
"moderate" based on Hamaguchi etal. [43].
e exclusion criteria were as follow: alcohol con-
sumption, pregnancy, breastfeeding, menopause, regular
exercise, following weight loss diet 3months before the
study, taking medications such as anti-diabetic, anti-
lipidemic, anti-hypertensive, antibiotics, corticosteroids,
oral contraceptives and anti-inflammatory drugs, as well
as suffering from liver, kidney, thyroid, gastro-intestinal,
autoimmune diseases, T2DM, polycystic ovary syndrome
(PCOS), and cancer.
Sample size
According to mean and standard deviation of serum
MCP-1 reported by Wamberg etal. [44] and by consider-
ing 95% confidence interval (CI) and %80 power among
patients with NAFLD using sample size software (PASS;
NCSS, LLC, US), sample size was found 20 for each
group which then increased to 24 by considering 10%
drop-out rate.
Randomization, blinding, andintervention
To randomly allocate the patients into two groups, Ran-
dom Allocation Software (RAS) and randomized block
procedure were used. e patients were assigned into
either DASH or CRD groups (1:1) by an assistant not
involved in the trial. Size 3 randomized block procedure
was applied as follows; gender (female vs male), age (18–
35 yrs. vs 36–55 yrs.) and BMI (< 35 kg/m2 vs 35 kg/
m2)]. Before randomization for treatment, the assign-
ment was concealed.
Energy requirement was estimated individually accord-
ing to Mifflin formula and weight loss diet was planned
by reducing 500 kcal from the estimated energy for
all the patients [45]. Macronutrient distribution was
55–60%, < 30%, and 10–15% of energy from carbohy-
drates, fat, and protein, respectively. Meal plans were
prepared based on these calculations and the food-
based dietary guidelines for Iranians (available at http://
www. fao. org/ nutri tion/ educa tion/ food- based dieta
ry- guide lines/ regio ns/ count ries/ iran/ fr/) for CRD. For
DASH diet, weight loss diet was designed according
to DASH dietary pattern [46]. e DASH diet was rich
in fruits, vegetables, whole grains, and low-fat dairy
products and low in saturated fats, cholesterol, refined
grains, and sweets. Suggested sodium in the DASH diet
was < 2400mg/day.
Food group exchange list and food album were deliv-
ered to each patient to follow the prescribed diet. e
participants were also given a full explanation on how to
use food exchange lists to replace the foods they did not
have access to, by the foods of equal calorie from the cor-
responding food groups as well as delivering food group
exchange list and food album to follow the prescribed
diet.
Assessment ofanthropometric measures, dietary intake,
andphysical activity
At the beginning and end of the trial, personal and dis-
ease details, anthropometric measurements, physical
activity levels and dietary intakes were assessed. Weight
and height were measured using Seca stadiometer (Ham-
burg, Germany) to the nearest 100g and 0.5cm with low
clothes without shoes, respectively. BMI was estimated as
weight (Kg) divided by height squared (m2). e circum-
ferences of waist and hip were also measured at the half-
way between the lower ribs and the iliac crest and around
the widest portion of the buttocks to the nearest 0.1cm,
respectively. en, BMI, Waist-to-hip ratio (WHR) and
waist-to-height ratio (WHtR) were estimated.
A 3-day food record was completed by the patients at
baseline, week 4 and 8 and then, mean of each 3-day food
record was calculated for each food item, converted to
grams and ml, finally energy and macronutrient intakes
were obtained using Nutrition IV software (First Data-
bank: Hearst, San Bruno, CA, USA) at baseline and after
8weeks.
Furthermore, Dixon’s DASH diet index was estimated
to confirm adherence to DASH diet [46]. Foods were
Content courtesy of Springer Nature, terms of use apply. Rights reserved.
Page 4 of 14
Rooholahzadeganetal. Nutrition & Metabolism (2023) 20:11
categorized into 9 components based on the following
daily recommendations: total fruits ( 4 servings), total
vegetables ( 4 servings), whole grains (( 4 servings),
low-fat dairy products ( 2 servings), legumes, seeds
and nuts ( 4 servings), meat/meat equivalents (< 170g),
added sugar (< 3% of total daily energy), alcoholic bev-
erages ( 2 drink) and saturated fat (< 5% of total daily).
Foods were categorized into 9 components based on the
following daily recommendations were scored 1 point
and other than recommendation were scored zero point:
total fruits ( 4 servings), total vegetables ( 4 s ervings),
whole grains (( 4 servings), low-fat dairy products ( 2
servings), legumes, seeds and nuts ( 4 servings), meat/
meat equivalents (< 170 g), added sugar (< 3% of total
daily energy), alcoholic beverages ( 2 drink) and satu-
rated fat (< 5% of total daily). An overall adherence to
DASH diet, therefore, was obtained by summing up the
points ranging from 0 to 9 [4749].
To assess physical activity level, the international physi-
cal activity questionnaire-short form (IPAQ-SF) was
applied through face-to-face interview [50]. e patients
were asked to report the time spent doing each of the
defined intensity-varied activities during the past week to
calculate metabolic equivalent of task (MET-hours/week)
score. en, the participants were categorized into: “low”,
“moderate”, or “high” activity level [50].
Laboratory assays
At baseline and at the end of study, after 12–14h over-
night fasting, blood sample was obtained from each
patient and serum was separated. Metabolic factors
including serum glucose, alanine aminotransferase (ALT)
and aspartate transaminase (AST) concentrations were
determined at the same day while the rest was stored
at 70 °C until assays. Serum alanine aminotransferase
(ALT) and aspartate aminotransferase (AST) concentra-
tions were assessed at baseline and at the end of study
using the International Federation of Clinical Chemistry
(IFCC) approved method. Hemoglobin A1C (HbA1c)
was assessed using photometry in whole blood using Pars
Azmoun Company kit (Pars Azmoun, Iran) and Hitachi
auto analyzer (Hitachi-917, Tokyo, Japan). Furthermore,
serum levels of TLR-4, MCP-1, and LPS were assessed
using enzyme-linked immunosorbent assay (ELISA) kit
(LSBio, Seattle, WA). According to complete blood count
(CBC) results, white blood cell (WBC)-derived inflam-
matory indices including neutrophil to lymphocyte ratio
(NLR), monocyte to lymphocyte ratio (MLR), platelets
to to lymphocyte ratio (PLR), monocyte to high-den-
sity lipoproteincholesterol (HDL-C) ratio (mHDL) and
finally, systemic inflammation response index (SIRI)- as
an index reflecting the host immune and inflammation
balance—was estimated: SIRI = Neutrophil × monocy te/
lymphocyte [5153].
Study outcomes
Changes in energy and macronutrient intakes, serum gly-
cemic indices, MCP-1 and TLR-4, LPS, blood pressure,
and anthropometric indices were considered as the pri-
mary outcomes whereas changes in serum levels of liver
enzymes and NAFLD grade were considered as the sec-
ondary outcomes.
Statistical analysis
All statistical analyses were performed using SPSS Statis-
tics software (IBM SPSS Statistics, Armonk, USA, latest
version). e distribution of continuous variables was
checked using Kolmogorov–Smirnov test. For assess-
ing both primary and secondary outcomes, after treat-
ment approach was applied. Data were expressed as
mean ± standard deviation (SD), median (min, max),
and number (%) for continuous variables with symmetric
and asymmetric distribution and categorical variables,
respectively. Inter-group differences in the continuous
and categorical variables at baseline were performed
using independent samples t- and Chi-square tests,
respectively. Paired samples t- and Sign tests were used
for changes variables. At the end of the trial, the analy-
sis of covariance (ANCOVA) test was used to compare
between group changes in variables by adjusting for the
confounders (i.e., baseline values and weight change).
Absolute risk reduction (ARR) was calculated based on
the difference in the event rate between DASH and con-
trol groups and then number needed to treat (NNT) was
estimated according to the following formula: NNT = 1/
ARR. e significance level was defined at p value lower
than 0.05.
Results
Of totally 62 patients enrolled the trial, 40 subjects (20
patients in each group) completed the trial while 11
patients in each group lost to follow because of not fol-
lowing the prescribed diet (Fig.1).
Table 1 demonstrates baseline characteristics in two
studied groups. More than half of the studied patients
in both groups were women and married. At baseline,
no significant differences were found for not only demo-
graphic characteristics but also for NAFDL severity and
physical activity level between the groups.
Changes in dietary energy and nutrient intakes over
the study in control and DASH group are presented
in Table 2. Apart from monounsaturated fatty acids
(MUFA), dietary fiber, sodium and magnesium at base-
line, no significant differences were found in not only
energy and nutrient intakes but also the proportion of
Content courtesy of Springer Nature, terms of use apply. Rights reserved.
Page 5 of 14
Rooholahzadeganetal. Nutrition & Metabolism (2023) 20:11
macronutrients from energy between the groups before
and after the study. At baseline and end of the study,
saturated fat intake in DASH group was significantly
less than in control group (p < 0.001). Moreover, no sig-
nificant changes were observed in physical activity level
in both groups over the intervention (data not shown).
erefore, physical activity and dietary energy and
nutrient intakes were not considered as confounders in
data analysis.
Table3 demonstrates changes in anthropometric meas-
ures, obesity indices, as well as metabolic and inflam-
matory biomarkers in studied groups. Apart from hip
circumference (HC), the reduction in all anthropomet-
ric measures were significantly greater in DASH group
Fig. 1 Flow chart of the study
Content courtesy of Springer Nature, terms of use apply. Rights reserved.
Page 6 of 14
Rooholahzadeganetal. Nutrition & Metabolism (2023) 20:11
than in control group, after adjusting for baseline val-
ues and weight change. Although fasting blood sugar
(FBS) decreased in both groups, between-group com-
parison did not reveal any significant difference after
8weeks, after adjusting for baseline values and weight
change. Nevertheless, despite significant difference in
serum HbA1c between the groups at baseline, a signifi-
cant reduction in HbA1c was observed in DASH group
(P < 0.001) while there was an increase in serum HbA1c
in control group. Inter-group changes in serum HbA1c
was statistically significant, after adjusting for baseline
values and weight change (p < 0.001) (Table3).
ere were significant reductions in serum levels
of TLR-4, MCP-1 and LPS in DASH group while no
changes were found in these variables in control group
(Table3). After adjusting for baseline values and weight
change, inter-group analysis showed significant differ-
ences in serum concentrations of TLR-4, MCP-1 and
LPS. Among WBC-derived inflammatory indices, only
PLR change was statistically significant between the two
groups, after adjusting for the baseline values and weight
change (p = 0.019).
Regarding serum liver enzymes, significant reduc-
tions in both serum levels of AST and ALT were found in
DASH group whereas serum ALT decreased significantly
in control group (Table3). After adjusting for baseline
values and weight change, there was only significant
between-group difference in serum concentration of AST
(p = 0.008).
Table 4 summarizes the effectiveness of DASH diet
in NAFLD improvement. It was observed significant
improvement in liver steatosis for both group, i.e. the
greatest improvement as being free from NAFLD was
seen for DASH group (80%) and control group (40%),
respectively. e estimated NNTs due to 8-week follow-
ing DASH diet compared with control group for one and
two grade improvements in NAFLD severity were 2.5 and
6.67, respectively.
Discussion
e results of the present study designed to examine the
effect of adherence to DASH diet compared with CRD on
glycemic response, meta-inflammation and serum LPS in
obese patients with NAFLD showed greater reductions in
weight and obesity indices, serum levels of HbA1c, AST,
LPS and inflammatory biomarkers.
As energy for both DASH diet and CRD had been esti-
mated based on Mifflin formula (around 1550–1650kcal/
day) with similar macronutrient distribution from energy,
no significant differences in energy and macronutrient
Table 1 Baseline characteristic of the study participants
DASH, Dietary Approaches to Stop Hypertension, BMI Body mass index, NAFLD Non-alcoholic fatty liver disease. Data are presented as mean ± SD for numerical data
and number (%) for categorical variables
*p value for Independent sample t-test
**p value for Chi square test
Variable DASH
(N = 20) Control (N = 20) p
Age (yr.) 38.80 ± 9.98 37.10 ± 9.74 0.589*
Weight (Kg) 93.32 ± 19.51 93.49 ± 13.98 0.976*
Height (cm) 166.49 ± 12.0 165.52 ± 7.86 0.764*
BMI (Kg/m2) 33.43 ± 4.09 34.02 ± 3.61 0.632*
N (%) N (%)
Female 13 (65.0) 12 (60.0) 0.594**
Married 17 (85.0) 15 (75.0) 0. 683**
Educational level
Less than Diploma 6 (30.0) 3 (15.0) 0.520**
Diploma 6 (30.0) 9 (45.0)
University degrees 8 (40.0) 8 (40.0)
Physical activity level
Light 10 (50.0) 11 (55.0) 0.674**
Moderate 7 (35.0) 8 (40.0)
Heavy 3 (15.0) 1 (5.0)
NAFLD severity
Mild 10 (50.0) 12 (60.0) 0.751**
Moderate 10 (50.0) 8 (40.0)
Content courtesy of Springer Nature, terms of use apply. Rights reserved.
Page 7 of 14
Rooholahzadeganetal. Nutrition & Metabolism (2023) 20:11
intakes were found between the groups (Table2). Results
of the estimation of Dixon’s DASH diet index revealed
good adherence to DASH diet i.e. score 8 to 9 (ranged
0–9) at baseline and end of the study, respectively. ere-
fore, changes in the study outcomes could be attributed
to the weight loss intervention diet. Apart from saturated
fat intake in DASH group which was approximately half
of that in CRD group (p < 0.001) at baseline and end of
the study, there were no significant differences in micro-
nutrients intakes (being the characteristic of DASH diet
i.e. sodium, potassium, magnesium, calcium and vitamin
D) between the groups at the end of the study.
Our findings also revealed that reductions in weight
and obesity indices were significantly greater in DASH
group than CRD group, after adjusting for the confound-
ers (Table3). ere is evidence indicating that as DASH
diet includes high fruits, vegetables, dietary fiber and cal-
cium as well as low fat intake, particularly in the form of
dairy products, and also simple sugar, following DASH
diet is an effective approach in weight loss in obesity
and a number of metabolic diseases [3133]. For exam-
ple, Asemi et al. [54] showed that adherence to DASH
diet compared with usual low-calorie diet in patients
with PCOS for 8 weeks resulted in greater reductions
in weight and BMI. Similar findings were also reported
Table 2 Daily dietary intakes before and after the study
Variable DASH
(N = 20) Control
(N = 20) P
Energy (Kcal)
Baseline 1585.60 ± 187.67 1629.50 ± 206.54 0.488**
End 1553.00 ± 134.07 1640.10 ± 174.38 0.104***
P* 0.326 0.895
Carbohydrates (g)
Baseline 227.55 ± 31.46 234.40 ± 36.20 0.527**
End
P*219.59 ± 22.94
0.243 232.80 ± 25.70
0.949 0.123***
Protein (g)
Baseline 73.09 ± 8.17 75.72 ± 12.51 0.436**
End 69.52 ± 8.94 70.85 ± 13.68 0.978***
P* 0.192 0.164
Fat (g)
Baseline 46.17 ± 6.18 46.78 ± 10.11 0.841**
End 48.21 ± 5.61 49.84 ± 7.41 0.461***
P* 0.094 0.270
Carbohydrates (%)
Baseline 57.33 ± 2.28 57.07 ± 3.46 0.780**
End 56.54 ± 2.89 56.66 ± 3.60 0.910***
P*0.290 0.692
Protein (%)
Baseline 18.47 ± 1.00 18.99 ± 2.26 0.354**
End 17.90 ± 1.66 17.46 ± 2.70 0.535***
P*0.211 0.054
Fat (%)
Baseline 26.23 ± 2.03 25.96 ± 4.00 0.791**
End 27.94 ± 2.23 27.49 ± 3.13 0.609***
P*0.006 0.109
SFA (g)
Baseline 9.37 ± 2.73 18.41 ± 5.66 < 0.001**
End 9.69 ± 2.00 17.39 ± 3.28 < 0.001***
P*0.242 0.309
MUFA(g)
Baseline 21.09 ± 3.22 14.25 ± 3.99 < 0.001**
End 21.63 ± 3.45 15.60 ± 3.48 0.057***
P*0.415 0.222
PUFA (g)
Baseline 8.89 ± 1.18 7.63 ± 4.45 0.227**
End 10.03 ± 1.93 9.82 ± 4.81 0.868***
P*0.088 0.096
Dietary fiber (g)
Baseline 23.06 ± 6.45 17.89 ± 5.57 0.010**
End 21.93 ± 6.32 17.19 ± 6.32 0.794***
P*0.065 0.346
Sodium (mg)
Baseline 925.69 ± 344.87 1341.12 ± 504.75 0.004**
End 987.39 ± 345.33 1277.44 ± 412.15 0.078***
P* 0.465 0.536
Table 2 (continued)
Variable DASH
(N = 20) Control
(N = 20) P
Magnesium (mg)
Baseline 277.76 ± 65.74 227.29 ± 69.42 0.023**
End 266.94 ± 70.42 211.73 ± 70.25 0.705***
P* 0.340 0.249
Potassium (mg)
Baseline 3181.75 ± 1457.01 2916.95 ± 834.20
End 3510.35 ± 1016.14 2715.85 ± 970.19 0.485**
P* 0.106 0.296 0.530***
Calcium (mg)
Baseline 856.98 ± 308.63 700.03 ± 269.69 0.095**
End 921.32 ± 291.93 592.40 ± 228.59 0.074***
P* 0.357 0.062
Vitamin D (µg)
Baseline 2.14 ± 1.21 1.81 ± 1.34 0.426**
End 2.25 ± 0.84 1.08 ± 1.11 0.091***
P*0.850 0.048
Bold indicates p<0.05 is statistically signicant
DASH Dietary Approaches to Stop Hypertension, SFA Saturated fatty acid, MUFA
Monounsaturated fatty acid, PUFA Polyunsaturated fatty acid. Mean (SD) and
Mean Dierence (95% CI) are presented for data
*p value for paired- t test
**p value for Independent samples t-test
***p value for ANCOVA test (adjusted for baseline values and weight change)
Content courtesy of Springer Nature, terms of use apply. Rights reserved.
Page 8 of 14
Rooholahzadeganetal. Nutrition & Metabolism (2023) 20:11
Table 3 Anthropometric measures, metabolic and inflammatory biomarkers before and after the study
Variable DASH Control P
(N = 20) (N = 20)
Weight (Kg)
Baseline 93.32 ± 19.51 93.49 ± 13.98 0.976**
End 85.57 ± 18.62 87.88 ± 13.88 0.021***
MD (95% CI), P*7.75 (9.34, 6.17), > 0.001 5.61 ( 6.71, 4.50). < 0.001
BMI (Kg/m2)
Baseline 33.43 ± 4.09 34.02 ± 3.61 0.632**
End 30.64 ± 4.06 31.96 ± 3.57 0.025***
MD (95% CI), P*2.79 (3.35, 2.22), < 0.001 2.06 (2.47, 1.64), < 0.001
WC (cm)
Baseline 111.25 ± 12.29 109.92 ± 9.80 0.708**
End 103.32 ± 12.67 105.0 ± 9.60 0.002***
MD (95% CI), P*7.92 (9.58, 6.27), < 0.001 4.92 (5.94, 3.91), < 0.001
HC (cm) 115.42 ± 10.47 115.92 ± 5.84
Baseline 111.07 ± 11.44 112.20 ± 5.99 0.853**
End 4.35 (5.22, 3.48), < 0.001 3.72 (4.63, 2.82), < 0.001 0.323***
MD (95% CI), P*
WHR 0.95 ± 0.07
Baseline 0.96 ± 0.04 0.93 ± 0.07 0.406**
End 0.93 ± 0.05 0.01 (0.02, 0.00), 0.031 0.014***
MD (95% CI), P*0.03 (0.05, 0.02), < 0.001
WHtR
Baseline 0.67 ± 0.06 0.66 ± 0.05 0.821**
End 0.62 ± 0.06 0.63 ± 0.05 0.002***
MD (95% CI), P*0.05 (0.06, 0.04), < 0.001 0.03 (0.04, 0.02), < 0.001
FBS (mg/dl)
Baseline 93.41 ± 9.63 93.12 ± 9.63 0.933**
End 90.76 ± 5.69 90.64 ± 4.69 0.923***
MD (95% CI), P*2.64 (5.82, 0.53), 0.098 0.21 (0.37, 0.05), 0.013
HbA1c (%)
Baseline 5.30 ± 0.35 5.49 ± 0.49 < 0.001**
End 5.09 ± 0.34 5.56 ± 0.32 < 0.001***
MD (95% CI), P*2.49 (7.54, 2.57), 0.314 0.07 (0.08, 0.21), 0.340
TLR-4 (ng/ml)
Baseline 0.82 ± 0.11 0.80 ± 0.17 0.656**
End 0.70 ± 0.13 0.82 ± 0.14 0.004***
MD (95% CI), P*0.13 (0.20, 0.05),0.003 0.02 (0.05, 0.08), 0.634
MCP-1 (pg/ml)
Baseline 110.75 ± 7.56 11.64 ± 16.48 0.828**
End 100.44 ± 10.69 110.01 ± 11.79 0.027***
MD (95% CI), P*10.32 (16.36, 4.27), 0.002 1.64 (8.12, 4.85), 0.603
NLR
Baseline 1.92 ± 0.81 1.81 ± 0.64 0.238**
End 1.94 ± 0.83 1.71 ± 0.60 0.176***
MD (95% CI), P*26.51 (19.08, 33.94), < 0.001 37.44 (29.64, 45.23), < 0.001
MLR
Baseline 0.17 ± 0.09 0.16 ± 0.07 0.732**
End 0.23 ± 0.34 0.16 ± 0.08 0.179***
MD (95% CI), P*0.06 (0.09, 0.21), 0.399 0.004 (0.16, 0.02), 0.701
Content courtesy of Springer Nature, terms of use apply. Rights reserved.
Page 9 of 14
Rooholahzadeganetal. Nutrition & Metabolism (2023) 20:11
by subsequent studies conducted on patients with
NAFLD [40, 55]. Meanwhile, Rifai etal. [56] in patients
with heart failure failed to show any noticeable effect on
weight and BMI after 3months. In a randomized con-
trolled trial in 2021, 12-week following DASH diet with
and without exercise on anthropometric indices, DASH
diet plus exercise resulted in significantly lower weight
and WHR, although at the end of the study, the inter-
group differences were not statistically significant [57].
Moreover, a cross-sectional study on 305 overweight and
obese women showed that adherence to DASH diet was
inversely associated with greater weight reduction [58].
Soltani etal. [42] in a meta-analysis of randomized con-
trolled trials on the effect of low calorie DASH diet on
weight (N = 10), BMI (N = 6) and WC (N = 2) demon-
strated its lowering effect on the studied anthropometric
measures. It appears that DASH diet because of its low
energy density, high content of dietary fiber, particularly
due to the higher intake of whole grains and vegetables
could be effective in delaying carbohydrate absorption
and increasing satiety [42].
Although the results of the present trial failed to
show any difference in micronutrients such as calcium
and magnesium between the two diets at the end of the
study, cummulative evidence shows that DASH diet con-
tains high calcium and magnesium. Studies have dem-
onstrated that dietary calcium increases lipolysis and
plays an important role in weight management [34, 59].
Hence, calcium and magnesium intakes are inversely
related with obesity due to their roles in the saponifi-
cantion of fatty acids [34]. On the other hand, increased
sodium intake-which is low in DASH diet-results in fat
Table 3 (continued)
Variable DASH Control P
(N = 20) (N = 20)
PLR
Baseline 0.12 ± 0.03 0.11 ± 0.4 0.731**
End 0.12 ± 0.04 0.12 ± 0.05 0.019***
MD (95% CI), P*0.002 (0.010, 0.006), 0.680 0.01 (0.001, 0.02), 0.030
SIRI
Baseline 688.53 ± 437.71 720.85 ± 622.22 0.850**
End 862.13 ± 1139.21 706.92 ± 610.50 0.151***
MD (95% CI), P*173.59 (308.57, 655.76), 0.460 13.92 (68.06, 40.22), 0.597
mHDL
Baseline 7.59 ± 3.67 10.56 ± 8.65 0.166**
End 13.47 ± 29.77 9.44 ± 6.70 0.080***
MD (95% CI), P*5.87 (7.30, 19.04), 0.362 1.12 ( 2.70, 0.45), 0.152
LPS (pg/ml)
Baseline 21.66 ± 1.83 20.72 ± 2.43 0.174**
End 18.91 ± 2.98 20.90 ± 2.36 0.011***
MD (95% CI), P*2.75 (4.17, 1.33), < 0.001 0.18 ( 1.18, 1.54), 0.785
AST (IU/L)
Baseline 24.10 ± 10.91 26.75 ± 9.28 0.413**
End 18.40 ± 6.57 25.05 ± 8.70 0.008***
MD (95% CI), P*5.70 (9.42, 1.98), 0.005 1.70 (3.53, 0.13), 0.067
ALT (IU/L)
Baseline 27.20 ± 14.0 37.35 ± 18.37 0.057**
End 18.75 ± 8.91 31.60 ± 16.24 0.149***
MD (95% CI), P*8.45 (12.89, 4.01), 0.001 5.75 (10.46, 1.04), 0.019
Bold indicates p<0.05 is statistically signicant
DASH Dietary Approaches to Stop Hypertension, BMI Body mass index, WC Waist circumference, HC Hip circumference, WHR Waist to hip ratio, WHtR Waist to height
ratio, FBS Fasting blood sugar, HbA1c hemoglobin A1c, NLR Neutrophil to lymphocyte, MLR Monocyte to lymphocyte, PLR Platelets to lymphocyte, SIRI Systemic
inammation response index, mHDL Monocyte to high-density lipoproteincholesterol ratio, AST Aspartate aminotransferase, ALT Alanine aminotransferase, TLR-4 Toll-
like receptor-4, MCP-1 Monocyte chemoattractant protein-1, LPS Lipopolysaccharides
Mean (SD) and Mean Dierence (95% CI) are presented for data
*p value for paired- t test;
**p value for Independent samples t-test;
***p value for ANCOVA test (adjusted for baseline values and weight change)
Content courtesy of Springer Nature, terms of use apply. Rights reserved.
Page 10 of 14
Rooholahzadeganetal. Nutrition & Metabolism (2023) 20:11
accumulation through increasing leptin [42]. erefore,
following DASH diet for long term appears to help in
weight control.
Previous studies have also demonstrated that high
sodium intake is associated with the risk of NAFLD [60].
Uetake etal. [61]. found that high-salt diet exacerbated
nonalcoholic steatohepatitis in high-fat diet-fed lipopro-
tein receptor-1 (LOX-1) transgenic /apoE knockout mice
and that this effect was associated with the induction of
oxidative and inflammatory processes. Oxidative stress
and chronic inflammation play a major role in patho-
physiology of NAFLD [62]. A high-salt diet also activates
the aldose reductase-fructokinase pathway in the liver
and hypothalamus, which leads to endogenous fructose
production with the development of leptin resistance and
hyperphagia that cause obesity, IR, and NAFLD [63].
Our results also failed to find any significant difference
in serum FBS change after 8weeks between the groups,
however, a significant reduction in serum HbA1c was
observed in DASH group (P < 0.001) compared with an
increase in control group, after adjusting for the con-
founding factors (p < 0.001) (Table 3). ere is evidence
with a great emphasis on the consumption of whole
grains, fruits and vegetables in DASH diet, therefore, the
high content of dietary fiber in DASH diet decreases car-
bohydrate absorption and lowers blood glucose level [54].
Furthermore, DASH diet also includes food items with
low glycemic index and low energy content which is effi-
cient not only in hypertension but also could be consid-
ered as an efficient dietary approach in the management
of IR-related chronic diseases [54, 64]. Shirani etal. [65]
have reported that the adherence to DASH diet is more
likely to be associated with lower risk of hyperglycemia.
In this study, apart from PLR (p = 0.019), there were
no significant inter-group differences in WBC-derived
inflammatory biomarkers at the end of study. Neverthe-
less, serum levels of TLR-4, MCP-1 and LPS decreased
significantly in DASH group while no changes were found
in control group. Even after adjusting for confounding
variables, between-group analysis showed significant dif-
ferences in serum concentrations of TLR-4, MCP-1 and
LPS (Table3). Fung et al. [66] in a prospective cohort
study showed that DASH diet was associated with lower
serum interleukin-6 (IL-6) and C- reactive protein (CRP).
Holt et al. [67] also illustrated that high consumption
of fruits and vegetables decreased IL-6, CRP and TNF-
α. Similar findings have been reported on patients with
metabolic syndrome [68], T2DM [69], Mets [70] and
NAFLD [40]. DASH diet has shown favorable effects on
serum CRP and hs-CRP levels been through previous
studies [55, 71, 72]. However, Asemi etal. [73] on those
with gestational diabetes (24–24 weeks) reported no
effect of DASH diet on CRP level. Studies investigating
the effect of DASH diet with other inflammatory bio-
markers are few. Taheri etal. [58] showed that DASH diet
compared with other dietary pattern did not affect serum
levels of MCP-1. A systematic review on 16 observational
and 13 interventional studies concluded that plant-based
dietary patterns (such as Mediterranean or DASH diet)
reduce inflammatory biomarkers such as serum hs-CRP,
TNF-α and IL-6 [74].
Because DASH diet is rich in fruits and vegetables as
well as flavonoids with antioxidant activity, adherence
to DASH diet results in a decrease in free radicals, lipid
peroxidation and inflammation and in turn, leads to
reduced secretion of leptin and therefore, weight control
[64]. Hence, considering the close link between oxidative
stress and inflammation, inflammatory cells can produce
large amounts of reactive oxygen species (ROS)- as a
part of mechanism for immunological defense-to protect
human organisms against invading pathogens [8].
Moreover, studies havedemonstrated that thelow gly-
cemic index diet may decrease inflammation by slowing
glucose absorption, altering gut microflora and therefore
suppress the production of inflammatory cytokines, stim-
ulate the production of short-chain fatty acids in intra-
lumen which results in lower circulating FFA levels and
thus subsequent inflammation [42]. Furthermore, parallel
to the effect of DASH diet on meta-inflammation observed
Table 4 Changes in liver steatosis
Bold indicate p<0.05 is statistically signicant
* Chi square test
DASH, Dietary Approaches to Stop Hypertension; Non-alcoholic fatty liver
disease; ARR, Attributable risk reduction; NNT, Number needed to treat
Variable DASH
(N = 20) Control
(N = 20) P*
NAFLD severity
Baseline 0.273
Grade I 10(50.0) 8 (40.0)
Grade II 10 (50.0) 12 (60.0)
End
Grade 0 12 (60.0) 4(20.0) 0.012
Grade I 8 (40.0) 12 (60.0)
Grade II 0 (0.0) 4 (20.0)
Liver steatosis severity
No change 1 (5.0) 12 (60.0)
1 grade reduction 16 (80.0) 8(40.0)
2 grade reduction 3 (15.0) 0 (0.0)
ARR
1 grade reduction (%) 40
2 grade reduction (%) 15
NNT
1 grade reduction 2.5
2 grade reduction 6.67
Content courtesy of Springer Nature, terms of use apply. Rights reserved.
Page 11 of 14
Rooholahzadeganetal. Nutrition & Metabolism (2023) 20:11
in serum levels of TLR-4 and MCP-1, the concentration of
LPS decreased. Studies investigating the effect of DASH
diet on serum LPS is limited. Observational and inter-
ventional studies have suggested that phytochemicals and
other compounds present in DASH diet are directly or
indirectly attributed in the modulation of inflammatory
biomarkers as well as intestinal permeability and therefore,
the body’s susceptible to infection [75]. e link between
obesity and high intestinal permeability has been well doc-
umented. e systemic levels of LPS are elevated in obese
individuals through several mechanisms. e.g. impaired
clearance in the liver, alterations in the gut microbiota,
permeability, motility and enzyme levels, and serum levels
of HDL-C [75]. Intercellular tight junctions can regulate
intestinal permeability and factors such as fatty acids and
proinflammatory cytokines are necessary to maintaining
intestinal mucosa integrity [75]. erefore, it appears obe-
sity as well as impaired oxidative stress and chronic inflam-
mation in response to the characteristics of DASH diet are
involved in the improvement of inflammation.
Serum AST and ALT reduced significantly in DASH
group whereas serum ALT decreased significantly in con-
trol group (Table3). After adjusting for the confounders,
there was only significant between-group difference in
serum concentration of AST (p = 0.008) which is in line
with other studies. For example, reduced levels of serum
liver enzymes followed by DASH diet have been reported
in patients with T2DM [69] and NAFLD [40, 76]. Moreo-
ver, Xiao et al. [55] in population-based cohort study
illustrated that following DASH diet was less likely to be
associated with NAFLD risk, particularly in women and
those without abdominal obesity. Mahdavi etal. [71] also
reported following DASH diet led to reductions in liver
steatosis and fibrosis in male adolescents with hemophilia
after 10weeks. In the present study, the NNT was calcu-
lated for assessing the clinical importance of DASH diet.
e estimated NNTs for 8-week following DASH diet com-
pared with RCT for one and two grade improvements in
NAFLD severity were found 2.5 and 6.67, respectively.
Our study had several strengths including studying
NAFLD patients who newly diagnosed without receiv-
ing any medication or treatment, providing an individual-
ized low-calorie diet as an approved strategy for NAFLD
management) on the basis of DASH dietary pattern, good
adherence to DASH diet by the patients and assessing spe-
cific inflammatory biomarkers compared with previous
studies. However, lack of liver biopsy because of ethical
considerations, not assessing other inflammatory factors as
well as insulin resistance indices could be considered as the
study limitations.
Conclusion
It is concluded that following DASH diet for 8 weeks
could significantly improve liver function in patients
with NAFLD due to reduced weight and BMI, glycemic
response, and meta-inflammation.
Abbreviations
ALT Alanine aminotransferase
ANCOVA Analysis of covariance
ARR Absolute risk reduction
AST Aspartate aminotransferase
BMI Body mass index
BP Blood pressure
CBC Complete blood count
CI Confidence interval
CRD Calorie‑restricted diet
CRP C‑ reactive protein
DASH Dietary Approaches to Stop Hypertension
DBP Diastolic blood pressure
ELISA Enzyme‑linked immunosorbent assay
FBS Fasting blood sugar
FFAs Free fatty acids
HbA1c Hemoglobin A1c
HC Hip circumference
HDL‑C High‑density lipoprotein cholesterol
HTN Hypertension
IFCC International Federation of Clinical Chemistry
IPAQ‑SF International physical activity questionnaire‑short form
IR Insulin resistance
LPS Lipopolysaccharide
MAFLD Metabolic associated fatty liver disease
MET Metabolic equivalent of task
Mets Metabolic syndrome
MCP‑1 Monocyte chemoattractant protein
mHDL Monocyte to high‑density lipoprotein cholesterol ratio
MLR Monocyte to lymphocyte ratio
MUFA Monounsaturated fatty acids
NAFLD Non‑alcoholic fatty liver disease
NLR Neutrophil to lymphocyte ratio
NNT Number needed to treat
PCOS Polycystic ovary syndrome
PLR Platelets to to lymphocyte ratio
RAS Random Allocation Software
RCT Randomized clinical trial
SD Standard deviation
SBP Systolic blood pressure
SIRI Systemic inflammation response index
T2DM Type 2 diabetes mellitus
TLR‑4 Toll‑like receptors‑4
TNF‑α Tumor necrosis factor‑α
WBC White blood cell
WC Waist circumference
WHR Waist to hip ratio
WHtR Waist to height ratio
Author contributions
The authors’ responsibilities were as follows: FR and TB help in data collection;
MK contributed in patient selection; FR and SA wrote the original paper; FR
and MEM did statistical analysis; MEM and HT contributed to the conception
of the article as well as to the final revision of the manuscript. All authors read
and approved the final version of the manuscript.
Funding
This study was funded by the ‘Research Vice‑Chancellor’ of Tabriz University
of Medical Sciences, Tabriz, Iran. This paper is a part of the data obtained from
an MSc dissertation submitted to Tabriz University of Medical Sciences (Farnaz
Rooholahzadegan).
Content courtesy of Springer Nature, terms of use apply. Rights reserved.
Page 12 of 14
Rooholahzadeganetal. Nutrition & Metabolism (2023) 20:11
Availability of data and materials
The datasets used and/or analyzed during the current study are available from
the corresponding author on a reasonable request.
Declarations
Ethics approval and consent to participate
All procedures performed in this study were in accordance with the ethical
standards of the Ethics Committee of Tabriz University of Medical Science.
In this study all subjects signed a consent form and the study protocol was
approved by the ethical committee of Tabriz University of Medical Sciences
(Ethics code: TBZMED. REC. 1398.741) and also registered in the Iranian Regis‑
try of Clinical Trials (IRCT20100209003320N17).
Competing interests
The authors declare that they have no competing interests.
Received: 16 September 2022 Accepted: 8 February 2023
References
1. Swinburn B, Egger G, Raza F. Dissecting obesogenic environments: the
development and application of a framework for identifying and prioritiz‑
ing environmental interventions for obesity. Prev Med. 1999;29(6):563–70.
2. Swinburn BA, Sacks G, Hall KD, McPherson K, Finegood DT, Moodie ML,
et al. The global obesity pandemic: shaped by global drivers and local
environments. The Lancet. 2011;378(9793):804–14.
3. Bedogni G, Nobili V, Tiribelli C. Epidemiology of fatty liver: an update.
World J Gastroenterol: WJG. 2014;20(27):9050.
4. Chalasani N, Younossi Z, Lavine JE, Diehl AM, Brunt EM, Cusi K, et al. The
diagnosis and management of non‑alcoholic fatty liver disease: Practice
Guideline by the American Association for the Study of Liver Diseases,
American College of Gastroenterology, and the American Gastroentero‑
logical Association. Hepatology. 2012;55(6):2005–23.
5. Arefhosseini S, Ebrahimi‑Mameghani M, Najafipour F, Tutunchi H. Non‑
alcoholic fatty liver disease across endocrinopathies: interaction with sex
hormones. Front Endocrinol. 2022;13:1032361.
6. Liu J, Ayada I, Zhang X, Wang L, Li Y, Wen T, et al. Estimating global preva‑
lence of metabolic dysfunction‑associated fatty liver disease in over‑
weight or obese adults. Clin Gastroenterol Hepatol. 2022;20(3):e573–82.
7. Estes C, Razavi H, Loomba R, Younossi Z, Sanyal AJ. Modeling the epi‑
demic of nonalcoholic fatty liver disease demonstrates an exponential
increase in burden of disease. Hepatology. 2018;67(1):123–33.
8. Marchisello S, Di Pino A, Scicali R, Urbano F, Piro S, Purrello F, et al. Patho‑
physiological, molecular and therapeutic issues of nonalcoholic fatty liver
disease: an overview. Int J Mol Sci. 2019;20(8):1948.
9. Marušić M, Paić M, Knobloch M, Liberati Pršo A‑M. NAFLD, insulin
resistance, and diabetes mellitus type 2. Can J Gastroenterol Hepatol.
2021;2021:6613827.
10. Koliaki C, Szendroedi J, Kaul K, Jelenik T, Nowotny P, Jankowiak F, et al.
Adaptation of hepatic mitochondrial function in humans with non‑alco‑
holic fatty liver is lost in steatohepatitis. Cell Metab. 2015;21(5):739–46.
11. Younossi Z, Tacke F, Arrese M, Chander Sharma B, Mostafa I, Bugianesi E,
et al. Global perspectives on nonalcoholic fatty liver disease and nonalco‑
holic steatohepatitis. Hepatology. 2019;69(6):2672–82.
12. Tarantino G, Savastano S, Colao A. Hepatic steatosis, low‑grade chronic
inflammation and hormone/growth factor/adipokine imbalance. World J
Gastroenterol: WJG. 2010;16(38):4773.
13. Younossi ZM, Rinella ME, Sanyal AJ, Harrison SA, Brunt EM, Goodman
Z, et al. From NAFLD to MAFLD: implications of a premature change in
terminology. Hepatology. 2021;73(3):1194–8.
14. Russo S, Kwiatkowski M, Govorukhina N, Bischoff R, Melgert BN.
Meta‑inflammation and metabolic reprogramming of macrophages in
diabetes and obesity: the importance of metabolites. Front Immunol.
2021;12:746151.
15. Suzuki K . Chronic inflammation as an immunological abnormality and
effectiveness of exercise. Biomolecules. 2019;9(6):223.
16. Liver EAftSoT, Diabetes EAftSo. EASL‑EASD‑EASO Clinical Practice Guide‑
lines for the management of non‑alcoholic fatty liver disease. Obesity
facts. 2016;9(2):65–90.
17. Chalasani N, Younossi Z, Lavine JE, Charlton M, Cusi K, Rinella M, et al. The
diagnosis and management of nonalcoholic fatty liver disease: practice
guidance from the American Association for the Study of Liver Diseases.
Hepatology. 2018;67(1):328–57.
18. Palacka P, Kucharska J, Murin J, Dostalova K, Okkelova A, Cizova M, et al.
Complementary therapy in diabetic patients with chronic complications:
a pilot study. Bratisl Lek Listy. 2010;111(4):205–11.
19. Hadi V, Pahlavani N, Malekahmadi M, Nattagh‑Eshtivani E, Navashenaq JG,
Hadi S, et al. Nigella sativa in controlling Type 2 diabetes, cardiovascular,
and rheumatoid arthritis diseases: Molecular aspects. J Res Med Sci: The
Off Jf Isfahan Univ Med Sci. 2021;26:20.
20. Pahlavani N, Roudi F, Zakerian M, Ferns GA, Navashenaq JG, Mashk‑
ouri A, et al. Possible molecular mechanisms of glucose‑lowering
activities of Momordica charantia (karela) in diabetes. J Cell Biochem.
2019;120(7):10921–9.
21. Malekahmadi M, Moradi Moghaddam O, Islam SMS, Tanha K, Nematy
M, Pahlavani N, et al. Evaluation of the effects of pycnogenol (French
maritime pine bark extract) supplementation on inflammatory biomark‑
ers and nutritional and clinical status in traumatic brain injury patients
in an intensive care unit: a randomized clinical trial protocol. Trials.
2020;21(1):1–9.
22. Pahlavani N, Sedaghat A, Moghaddam AB, Kiapey SSM, Navashenaq
JG, Jarahi L, et al. Effects of propolis and melatonin on oxidative stress,
inflammation, and clinical status in patients with primary sepsis:
study protocol and review on previous studies. Clin Nutr ESPEN.
2019;33:125–31.
23. Pahlavani N, Rostami D, Ebrahimi F, Azizi‑Soleiman F. Nuts effects in
chronic disease and relationship between walnuts and satiety: review on
the available evidence. Obes Med. 2020;17: 100173.
24. Salvoza N, Giraudi PJ, Tiribelli C, Rosso N. Natural compounds for
counteracting nonalcoholic fatty liver disease (NAFLD): advantages and
limitations of the suggested candidates. Int J Mol Sci. 2022;23(5):2764.
25. Parra‑Vargas M, Rodriguez‑Echevarria R, Jimenez‑Chillaron JC. Nutritional
approaches for the management of nonalcoholic fatty liver disease: an
evidence‑based review. Nutrients. 2020;12(12):3860.
26. Mirmiran P, Amirhamidi Z, Ejtahed H‑S, Bahadoran Z, Azizi F. Relationship
between diet and non‑alcoholic fatty liver disease: a review article. Iran J
Public Health. 2017;46(8):1007.
27. Kalafati I‑P, Borsa D, Dimitriou M, Revenas K, Kokkinos A, Dedoussis GV.
Dietary patterns and non‑alcoholic fatty liver disease in a Greek case–
control study. Nutrition. 2019;61:105–10.
28. Zelber‑Sagi S, Ivancovsky‑Wajcman D, Isakov NF, Webb M, Orenstein D,
Shibolet O, et al. High red and processed meat consumption is associated
with non‑alcoholic fatty liver disease and insulin resistance. J Hepatol.
2018;68(6):1239–46.
29. He K, Li Y, Guo X, Zhong L, Tang S. Food groups and the likelihood of non‑
alcoholic fatty liver disease: a systematic review and meta‑analysis. Br J
Nutr. 2020;124(1):1–13.
30. Harsha DW, Lin P‑H, Obarzanek E, Karanja NM, Moore TJ, Caballero B, et al.
Dietary Approaches to Stop Hypertension: a summary of study results. J
Am Diet Assoc. 1999;99(8):S35–9.
31. Chiavaroli L, Viguiliouk E, Nishi SK, Blanco Mejia S, Rahelić D, Kahle‑
ová H, et al. DASH dietary pattern and cardiometabolic outcomes: an
umbrella review of systematic reviews and meta‑analyses. Nutrients.
2019;11(2):338.
32. Pahlavani N, Khayyatzadeh SS, Banazadeh V, Bagherniya M, Tayefi M,
Eslami S, et al. Adherence to a dietary approach to stop hypertension
(DASH)‑style in relation to daytime sleepiness. Nat Sci Sleep. 2020;12:325.
33. Valipour G, Esmaillzadeh A, Azadbakht L, Afshar H, Hassanzadeh A, Adibi
P. Adherence to the DASH diet in relation to psychological profile of
Iranian adults. Eur J Nutr. 2017;56(1):309–20.
34. Conlin PR, Chow D, Miller ER, Svetkey LP, Lin P‑H, Harsha DW, et al. The
effect of dietary patterns on blood pressure control in hypertensive
patients: results from the Dietary Approaches to Stop Hypertension
(DASH) trial. Am J Hypertens. 2000;13(9):949–55.
35. Lin P‑H, Aickin M, Champagne C, Craddick S, Sacks FM, McCarron P, et al.
Food group sources of nutrients in the dietary patterns of the DASH‑
Sodium trial. J Am Diet Assoc. 2003;103(4):488–96.
Content courtesy of Springer Nature, terms of use apply. Rights reserved.
Page 13 of 14
Rooholahzadeganetal. Nutrition & Metabolism (2023) 20:11
36. Watzinger C, Nonnenmacher T, Grafetstätter M, Sowah SA, Ulrich CM,
Kauczor H‑U, et al. Dietary factors in relation to liver fat content: a cross‑
sectional study. Nutrients. 2020;12(3):825.
37. Hekmatdoost A, Shamsipour A, Meibodi M, Gheibizadeh N, Eslam
parast T, Poustchi H. Adherence to the dietary approaches to stop
hypertension (DASH) and risk of nonalcoholic fatty liver disease. Int J
Food Sci Nutr. 2016;67(8):1024–9.
38. Maskarinec G, Lim U, Jacobs S, Monroe KR, Ernst T, Buchthal SD,
et al. Diet quality in midadulthood predicts visceral adiposity and
liver fatness in older ages: the Multiethnic Cohort Study. Obesity.
2017;25(8):1442–50.
39. Park S‑Y, Noureddin M, Boushey C, Wilkens LR, Setiawan VW. Diet qual
ity association with nonalcoholic fatty liver disease by cirrhosis status:
the multiethnic cohort. Curr Dev Nutr. 2020;4(3):24.
40. Razavi Zade M, Telkabadi MH, Bahmani F, Salehi B, Farshbaf S, Asemi Z.
The effects of DASH diet on weight loss and metabolic status in adults
with non‑alcoholic fatty liver disease: a randomized clinical trial. Liver
Int. 2016;36(4):563–71.
41. Soltani S, Shirani F, Chitsazi MJ, Salehi‑Abargouei A. The effect of
dietary approaches to stop hypertension (DASH) diet on weight and
body composition in adults: a systematic review and meta‑analysis of
randomized controlled clinical trials. Obes Rev. 2016;17(5):442–54.
42. Soltani S, Chitsazi MJ, Salehi‑Abargouei A. The effect of dietary
approaches to stop hypertension (DASH) on serum inflammatory
markers: a systematic review and meta‑analysis of randomized trials.
Clin Nutr. 2018;37(2):542–50.
43. Hamaguchi M, Kojima T, Itoh Y, Harano Y, Fujii K, Nakajima T, et al. The
severity of ultrasonographic findings in nonalcoholic fatty liver disease
reflects the metabolic syndrome and visceral fat accumulation. Off J
Am Coll Gastroenterol| ACG. 2007;102(12):2708–15.
44. Wamberg L, Cullberg K, Rejnmark L, Richelsen B, Pedersen S. Investiga
tions of the anti‑inflammatory effects of vitamin D in adipose tissue:
results from an in vitro study and a randomized controlled trial. Horm
Metab Res. 2013;45(06):456–62.
45. Mifflin MD, St Jeor ST, Hill LA, Scott BJ, Daugherty SA, Koh YO. A new
predictive equation for resting energy expenditure in healthy individu
als. Am J Clin Nutr. 1990;51(2):241–7.
46. Dixon LB, Subar AF, Peters U, Weissfeld JL, Bresalier RS, Risch A, et al.
Adherence to the USDA Food Guide, DASH Eating Plan, and Mediter
ranean dietary pattern reduces risk of colorectal adenoma. J Nutr.
2007;137(11):2443–50.
47. Sacks FM, Svetkey LP, Vollmer WM, Appel LJ, Bray GA, Harsha D,
et al. Effects on blood pressure of reduced dietary sodium and the
Dietary Approaches to Stop Hypertension (DASH) diet. N Engl J Med.
2001;344(1):3–10.
48. Heidari Z, Mohammadi E, Aghamohammadi V, Jalali S, Rezazadeh A,
Sedaghat F, et al. Dietary Approaches to Stop Hypertension (DASH)
diets and breast cancer among women: a case control study. BMC
Cancer. 2020;20(1):1–10.
49. Miller PE, Cross AJ, Subar AF, Krebs‑Smith SM, Park Y, Powell‑Wiley
T, et al. Comparison of 4 established DASH diet indexes: examining
associations of index scores and colorectal cancer. Am J Clin Nutr.
2013;98(3):794–803.
50. Committee IR. Guidelines for data processing and analysis of the Inter
national Physical Activity Questionnaire (IPAQ)‑short and long forms.
http:// www. ipaqk ise/ scori ng pdf. 2005.
51. Zhao M, Duan X, Mi L, Shi J, Li N, Yin X, et al. Prognosis of hepatocel
lular carcinoma and its association with immune cells using systemic
inflammatory response index. Future Oncol. 2022;18(18):2269–88.
52. Alkhouri N, Morris‑Stiff G, Campbell C, Lopez R, Tamimi TAR, Yerian L,
et al. Neutrophil to lymphocyte ratio: a new marker for predicting stea
tohepatitis and fibrosis in patients with nonalcoholic fatty liver disease.
Liver Int. 2012;32(2):297–302.
53. Aktas G, Duman TT, Kurtkulagi O, Tel BMA, Bilgin S, Kahveci G, et al.
Liver steatosis is associated both with platelet distribution width,
neutrophil/lymphocyte and monocyte/lymphocyte ratios. Prim Health
Care: Open Access. 2020;10(4):1–4.
54. Asemi Z, Esmaillzadeh A. DASH diet, insulin resistance, and serum
hs‑CRP in polycystic ovary syndrome: a randomized controlled clinical
trial. Horm Metab Res. 2015;47(03):232–8.
55. Xiao M‑L, Lin J‑S, Li Y‑H, Liu M, Deng Y‑Y, Wang C‑Y, et al. Adherence to
the Dietary Approaches to Stop Hypertension (DASH) diet is associated
with lower presence of non‑alcoholic fatty liver disease in middle‑aged
and elderly adults. Public Health Nutr. 2020;23(4):674–82.
56. Rifai L, Pisano C, Hayden J, Sulo S, Silver MA, editors. Impact of the
DASH diet on endothelial function, exercise capacity, and quality of life
in patients with heart failure. Baylor University Medical Center Proceed
ings; 2015: Taylor & Francis
57. Hassanian‑Fard S, Jalali‑Dehkordi K, Rahimi H. The effect of combined
training with dietary approaches to stop hypertension (DASH) on liver
damage indices in patients with non‑alcoholic fatty liver disease. J
Isfahan Med School. 2021;39(634):533–41.
58. Taheri A, Mirzababaei A, Setayesh L, Yarizadeh H, Shiraseb F, Imani H,
et al. The relationship between Dietary approaches to stop hyperten
sion diet adherence and inflammatory factors and insulin resistance in
overweight and obese women: a cross‑sectional study. Diabetes Res
Clin Pract. 2021;182: 109128.
59. Mansouri M, Pahlavani N, Sharifi F, Varmaghani M, Shokri A, Yaghubi
H, et al. Dairy consumption in relation to hypertension among a large
population of university students: the MEPHASOUS study. Diabetes,
Metab Synd Obes: Targets Therapy. 2020;13:1633.
60. Shojaei‑Zarghani S, Safarpour AR, Fattahi MR, Keshtkar A. Sodium
in relation with nonalcoholic fatty liver disease: a systematic
review and meta‑analysis of observational studies. Food Sci Nutr.
2022;10(5):1579–91.
61. Uetake Y, Ikeda H, Irie R, Tejima K, Matsui H, Ogura S, et al. High‑salt in
addition to high‑fat diet may enhance inflammation and fibrosis in
liver steatosis induced by oxidative stress and dyslipidemia in mice.
Lipids Health Dis. 2015;14(1):1–8.
62. Masarone M, Rosato V, Dallio M, Gravina AG, Aglitti A, Loguercio C, et al.
Role of oxidative stress in pathophysiology of nonalcoholic fatty liver
disease. Oxid Med Cell Longev. 2018;2018:9547613.
63. Lanaspa MA, Kuwabara M, Andres‑Hernando A, Li N, Cicerchi C, Jensen
T, et al. High salt intake causes leptin resistance and obesity in mice by
stimulating endogenous fructose production and metabolism. Proc
Natl Acad Sci. 2018;115(12):3138–43.
64. Suri S, Kumar V, Kumar S, Goyal A, Tanwar B, Kaur J, et al. DASH dietary
pattern: a treatment for non‑communicable diseases. Curr Hypertens
Rev. 2020;16(2):108–14.
65. Shirani F, Salehi‑Abargouei A, Azadbakht L. Effects of Dietary
Approaches to Stop Hypertension (DASH) diet on some risk for
developing type 2 diabetes: a systematic review and meta‑analysis on
controlled clinical trials. Nutrition. 2013;29(7–8):939–47.
66. Fung TT, Chiuve SE, McCullough ML, Rexrode KM, Logroscino G, Hu FB.
Adherence to a DASH‑style diet and risk of coronary heart disease and
stroke in women. Arch Intern Med. 2008;168(7):713–20.
67. Holt EM, Steffen LM, Moran A, Basu S, Steinberger J, Ross JA, et al.
Fruit and vegetable consumption and its relation to markers of
inflammation and oxidative stress in adolescents. J Am Diet Assoc.
2009;109(3):414–21.
68. North C, Venter C, Jerling J. The effects of dietary fibre on C‑reactive
protein, an inflammation marker predicting cardiovascular disease. Eur
J Clin Nutr. 2009;63(8):921–33.
69. Azadbakht L, Surkan PJ, Esmaillzadeh A, Willett WC. The Dietary
Approaches to Stop Hypertension eating plan affects C‑reactive
protein, coagulation abnormalities, and hepatic function tests among
type 2 diabetic patients. J Nutr. 2011;141(6):1083–8.
70. Saneei P, Hashemipour M, Kelishadi R, Esmaillzadeh A. The Dietary
Approaches to Stop Hypertension (DASH) diet affects inflammation in
childhood metabolic syndrome: a randomized cross‑over clinical trial.
Ann Nutr Metab. 2014;64(1):20–7.
71. Mahdavi A, Mohammadi H, Bagherniya M, Foshati S, Clark CC, Moafi
A, et al. The effect of the Dietary Approaches to Stop Hypertension
(DASH) diet on body composition, complete blood count, prothrom
bin time, inflammation, and liver function in hemophilic adolescents.
Br J Nutr. 2021:128(9):1771–9.
72. Sakhaei R, Shahvazi S, Mozaffari‑Khosravi H, Samadi M, Khatibi N,
Nadjarzadeh A, et al. The dietary approaches to stop hypertension
(DASH)‑style diet and an alternative Mediterranean diet are differently
associated with serum inflammatory markers in female adults. Food
Nutr Bull. 2018;39(3):361–76.
Content courtesy of Springer Nature, terms of use apply. Rights reserved.
Page 14 of 14
Rooholahzadeganetal. Nutrition & Metabolism (2023) 20:11
fast, convenient online submission
thorough peer review by experienced researchers in your field
rapid publication on acceptance
support for research data, including large and complex data types
gold Open Access which fosters wider collaboration and increased citations
maximum visibility for your research: over 100M website views per year
At BMC, research is always in progress.
Learn more biomedcentral.com/submissions
Ready to submit your research
Ready to submit your research
? Choose BMC and benefit from:
? Choose BMC and benefit from:
73. Asemi Z, Samimi M, Tabassi Z, Sabihi S‑s, Esmaillzadeh A. A randomized
controlled clinical trial investigating the effect of DASH diet on insulin
resistance, inflammation, and oxidative stress in gestational diabetes.
Nutrition. 2013;29(4):619–24.
74. Aleksandrova K, Koelman L, Rodrigues CE. Dietary patterns and bio
markers of oxidative stress and inflammation: a systematic review of
observational and intervention studies. Redox Biol. 2021;42: 101869.
75. Huang Z, Kraus VB. Does lipopolysaccharide‑mediated inflammation
have a role in OA? Nat Rev Rheumatol. 2016;12(2):123–9.
76. Doustmohammadian A, Clark CC, Maadi M, Motamed N, Sobhrakh‑
shankhah E, Ajdarkosh H, et al. Favorable association between Mediter‑
ranean diet (MeD) and DASH with NAFLD among Iranian adults of the
Amol Cohort Study (AmolCS). Sci Rep. 2022;12(1):1–9.
Publisher’s Note
Springer Nature remains neutral with regard to jurisdictional claims in pub‑
lished maps and institutional affiliations.
Content courtesy of Springer Nature, terms of use apply. Rights reserved.
1.
2.
3.
4.
5.
6.
Terms and Conditions
Springer Nature journal content, brought to you courtesy of Springer Nature Customer Service Center GmbH (“Springer Nature”).
Springer Nature supports a reasonable amount of sharing of research papers by authors, subscribers and authorised users (“Users”), for small-
scale personal, non-commercial use provided that all copyright, trade and service marks and other proprietary notices are maintained. By
accessing, sharing, receiving or otherwise using the Springer Nature journal content you agree to these terms of use (“Terms”). For these
purposes, Springer Nature considers academic use (by researchers and students) to be non-commercial.
These Terms are supplementary and will apply in addition to any applicable website terms and conditions, a relevant site licence or a personal
subscription. These Terms will prevail over any conflict or ambiguity with regards to the relevant terms, a site licence or a personal subscription
(to the extent of the conflict or ambiguity only). For Creative Commons-licensed articles, the terms of the Creative Commons license used will
apply.
We collect and use personal data to provide access to the Springer Nature journal content. We may also use these personal data internally within
ResearchGate and Springer Nature and as agreed share it, in an anonymised way, for purposes of tracking, analysis and reporting. We will not
otherwise disclose your personal data outside the ResearchGate or the Springer Nature group of companies unless we have your permission as
detailed in the Privacy Policy.
While Users may use the Springer Nature journal content for small scale, personal non-commercial use, it is important to note that Users may
not:
use such content for the purpose of providing other users with access on a regular or large scale basis or as a means to circumvent access
control;
use such content where to do so would be considered a criminal or statutory offence in any jurisdiction, or gives rise to civil liability, or is
otherwise unlawful;
falsely or misleadingly imply or suggest endorsement, approval , sponsorship, or association unless explicitly agreed to by Springer Nature in
writing;
use bots or other automated methods to access the content or redirect messages
override any security feature or exclusionary protocol; or
share the content in order to create substitute for Springer Nature products or services or a systematic database of Springer Nature journal
content.
In line with the restriction against commercial use, Springer Nature does not permit the creation of a product or service that creates revenue,
royalties, rent or income from our content or its inclusion as part of a paid for service or for other commercial gain. Springer Nature journal
content cannot be used for inter-library loans and librarians may not upload Springer Nature journal content on a large scale into their, or any
other, institutional repository.
These terms of use are reviewed regularly and may be amended at any time. Springer Nature is not obligated to publish any information or
content on this website and may remove it or features or functionality at our sole discretion, at any time with or without notice. Springer Nature
may revoke this licence to you at any time and remove access to any copies of the Springer Nature journal content which have been saved.
To the fullest extent permitted by law, Springer Nature makes no warranties, representations or guarantees to Users, either express or implied
with respect to the Springer nature journal content and all parties disclaim and waive any implied warranties or warranties imposed by law,
including merchantability or fitness for any particular purpose.
Please note that these rights do not automatically extend to content, data or other material published by Springer Nature that may be licensed
from third parties.
If you would like to use or distribute our Springer Nature journal content to a wider audience or on a regular basis or in any other manner not
expressly permitted by these Terms, please contact Springer Nature at
onlineservice@springernature.com
... The group with a mediterranean diet showed weight loss, fat content of up to 40.9%, and increased antioxidant capacity. Rooholahzadegan et al. (2023). ...
... The implementation of a Dash diet is also beneficial for improving inflammatory markers. Rooholahzadegan et al. (2023) showed a decrease in body weight, BMI, serum triglycerides, very low-density lipoprotein cholesterol (VLDL-C), MCP-1, and improvements in total antioxidant capacity (TAC) and glutathione (GSH). ...
... Calcium and magnesium are inversely related to obesity because of their role in increasing lipolysis, thus playing an important role in weight management. Calcium and magnesium are also known to be protective agents against DNA damage and antioxidant enzymes because they influence oxidative stress levels by increasing TAC and GSH as free radical scavengers (Rooholahzadegan et al., 2023). The mechanism of the benefits of the MedDiet and dash diet in reducing inflammation and oxidative stress is presented in Figure 1. ...
Article
Full-text available
Obesity is also associated with increased inflammation and oxidative stress. Mediterranean and dash diets, as well as antioxidants, can reduce inflammation and oxidative stress levels in obesity. This study aimed to determine the effects of dietary modifications and antioxidants on inflammatory and oxidative stress levels in obesity. This study was carried out using the literature review method using PubMed, Science Direct, and Google Scholar databases with the keywords Mediterranean Diet, DASH Diet, Antioxidant, Obesity, Inflammation, and Oxidative Stress. The inclusion criteria in this literature are articles published in the last 5 years (2018-2023) using RCT, in English, subjects >18 years old with obesity, interventions providing Mediterranean and dash diet, antioxidants derived from natural ingredients, and articles that can be accessed online. Articles were selected individually through a double process, considering the three main research quality criteria. This study showed that a modified diet and antioxidants can reduce inflammatory biomarkers and oxidative stress in obesity by suppressing NF-κB signaling and protecting against DNA damage. In conclusion, a modified diet and antioxidants can reduce inflammatory and oxidative stress in obesity.
... (2) Studies on the treatment of NAFLD. Many pieces of evidence showed that calorie restriction (18,19), fat restriction (20,21), carbohydrate reduction (22,23), and appropriate vitamin supplementation (24,25) in diet therapy, aerobic exercise (26,27) resistance exercise (28,29) and high-intensity interval training (30) in exercise therapy had good therapeutic effects. Moreover, clinical experiments have proved that combining diet and exercise may achieve better therapeutic efficacy than a single one (31,32). ...
Article
Full-text available
Introduction The primary treatment for non-alcoholic fatty liver disease (NAFLD) is modifying lifestyle through dietary or exercise interventions. In recent decades, it has received increasing attention. However, the lack of bibliometric analysis has posed a challenge for researchers seeking to understand the overall trends in this field. Methods As of February 3rd, 2024, 876 articles on treating NAFLD through diet or exercise therapy from 2013 to 2023 had been retrieved. Two software tools, VOSviewer and CiteSpace, were utilized to analyze the growth of publications, countries, institutions, authors, journals, citations, and keywords. Additionally, the keywords with strong citation burstiness were identified to determine the changes and future trends of research hotspots in this field. Results China had the highest number of articles, followed by the United States and South Korea. Yonsei University and Nutrients were the institutions and journals with the most significant contributions. Professor Younossi Zobair M, from the United States, is the most prolific author in this field. Through analyzing the keywords, three research hotspots were identified: research on the pathogenesis of NAFLD, research on the treatment modalities of NAFLD, and research on the risk factors and diagnosis methods of NAFLD. In recent years, the research emphasis in this field has changed, suggesting that future research will focus on two frontier keywords: “oxidative stress” and “aerobic capacity.” Conclusion In the past eleven years, the attention in this field was still rising, and the authors, journals, countries and so on had formed a considerable cooperative relationship. There were also many highly influential and productive researchers in this field. It is speculated that new research will continue around “aerobic exercise” and “oxidative stress” in the future.
... Yet, there are no approved treatments for NAFLD. However, lifestyle modifcations and weight reduction seem to be the most appropriate strategies for NAFLD management [2,17]. ...
Article
Full-text available
Background: This trial assessed the effects of a calorie-restricted diet (CRD) with hydroxycitric acid (HCA) supplementation on appetite-regulating hormones, obesity indices, body composition, and appetite in women with nonalcoholic fatty liver disease (NAFLD). Methods: This study was carried out on 44 overweight/obese women with NAFLD. The patients were randomly assigned into two groups, namely, "Intervention group" (receiving individual CRD plus HCA tablets per day) and "Control group" (receiving only CRD) for eight weeks. Obesity indices, body composition, appetite status, and serum levels of leptin and adiponectin were assessed before and after the intervention. Results: Forty patients completed the trial. At the end of the trial, although significant reductions were found in most of the studied obesity indices in the intervention group, there was only a significant decrease in waist circumference and waist-to-height ratio in the control group. Fat mass and muscle mass significantly decreased in the intervention group (p=0.044 and p=0.024, respectively), and the reduction in visceral fat in the intervention group was significantly greater than that in the control group (-0.49 kg vs -0.37 kg, p=0.024). Intra- and intergroup differences in serum leptin and adiponectin levels and their ratios before and after the trial were not significant. We found a negative and marginally significant correlation between percent of changes in serum adiponectin level and percent of changes in visceral adipose tissue (VAT) (r = -0.429, p=0.067) and BMI (r = -0.440, p=0.059) as well as an inverse relationship between percent of changes in leptin/adiponectin with VAT (r = -0.724, p < 0.001) in the intervention group. Conclusion: HCA plus weight loss diet could significantly reduce visceral adipose tissue without any significant changes in serum leptin and adiponectin levels.
Article
Full-text available
Metabolic-associated fatty liver disease (MAFLD) includes several metabolic dysfunctions caused by dysregulation in the brain–gut–liver axis and, consequently, increases cardiovascular risks and fatty liver dysfunction. In MAFLD, type 2 diabetes mellitus, obesity, and metabolic syndrome are frequently present; these conditions are related to liver lipogenesis and systemic inflammation. This study aimed to review the connection between the brain–gut–liver axis and MAFLD. The inflammatory process, cellular alterations in hepatocytes and stellate cells, hypercaloric diet, and sedentarism aggravate the prognosis of patients with MAFLD. Thus, to understand the modulation of the physiopathology of MAFLD, it is necessary to include the organokines involved in this process (adipokines, myokines, osteokines, and hepatokines) and their clinical relevance to project future perspectives of this condition and bring to light new possibilities in therapeutic approaches. Adipokines are responsible for the activation of distinct cellular signaling in different tissues, such as insulin and pro-inflammatory cytokines, which is important for balancing substances to avoid MAFLD and its progression. Myokines improve the quantity and quality of adipose tissues, contributing to avoiding the development of MAFLD. Finally, hepatokines are decisive in improving or not improving the progression of this disease through the regulation of pro-inflammatory and anti-inflammatory organokines.
Article
This study compares two diets, Dietary Approaches to Stop Hypertension (DASH) and a Low-Calorie Diet on Trimethylamine N-oxide (TMAO) levels and gut microbiota. 120 obese adults were randomly allocated to these three groups: a low-calorie DASH diet, a Low-Calorie diet, or a control group for 12 weeks. Outcomes included plasma TMAO, lipopolysaccharides (LPS), and gut microbiota profiles. After the intervention, the low-calorie DASH diet group demonstrated a greater decrease in TMAO levels (-20 ± 8.1 vs. -10.63 ± 4.6 μM) and a significant decrease in LPS concentration (-19.76 ± 4.2 vs. -5.68 ± 2.3) compared to the low-calorie diet group. Furthermore, the low-calorie DASH diet showed a higher decrease in the Firmicutes and Bactericides (F/B) ratio, which influenced TMAO levels, compared to the Low-Calorie diet (p = 0.028). The current study found the low-calorie DASH diet improves TMAO and LPS in comparison to a Low-Calorie diet.
Article
Full-text available
Nonalcoholic fatty liver disease (NAFLD) has emerged as the most frequent chronic liver disease globally. NAFLD is strongly associated with metabolic syndrome and it has been recently suggested that to rename NAFLD as metabolic dysfunction-associated fatty liver disease (MAFLD). NAFLD has been studied in different endocrine axes and accumulating body of clinical and experimental studies have suggested that NAFLD is associated with polycystic ovarian syndrome (PCOS), hypopituitarism, growth hormone deficiency (GHD), hypogonadism and other endocrine disorders. In fact, endocrine dysfunction may be considered as the major contributor for the development, progression, and severity of NAFLD. In the present comprehensive review, we discussed the epidemiological and clinical evidence on the epidemiology, pathophysiology, and management of NAFLD in endocrine disorders, with an emphasis on the effects of sex-specific hormones/conditions as well as molecular basis of NAFLD development in these endocrine diseases.
Article
Full-text available
The booming prevalence of nonalcoholic fatty liver disease (NAFLD) in adults and children will threaten the health system in the upcoming years. The “multiple hit” hypothesis is the currently accepted explanation of the complex etiology and pathophysiology of the disease. Some of the critical pathological events associated with the development of NAFLD are insulin resistance, steatosis, oxidative stress, inflammation, and fibrosis. Hence, attenuating these events may help prevent or delay the progression of NAFLD. Despite an increasing understanding of the mechanisms involved in NAFLD, no approved standard pharmacological treatment is available. The only currently recommended alternative relies on lifestyle modifications, including diet and physical activity. However, the lack of compliance is still hampering this approach. Thus, there is an evident need to characterize new therapeutic alternatives. Studies of food bioactive compounds became an attractive approach to overcome the reticence toward lifestyle changes. The present study aimed to review some of the reported compounds with beneficial properties in NAFLD; namely, coffee (and its components), tormentic acid, verbascoside, and silymarin. We provide details about their protective effects, their mechanism of action in ameliorating the critical pathological events involved in NAFLD, and their clinical applications.
Article
Full-text available
Findings on the association of sodium with nonalcoholic fatty liver disease (NAFLD) are conflicting. The present systematic review and meta‐analysis study aimed to assess the association between salt or sodium intake or serum sodium levels and NAFLD risk. Relevant articles were identified by searching PubMed, Web of Knowledge, Scopus, Proquest, and Embase databases through May 1, 2021, without language restriction. The pooled odds ratio (OR) and 95% confidence interval (CI) were estimated using Der‐Simonian and Laird method and random‐effects meta‐analysis. The certainty of the evidence was rated using the GRADE method. Out of 6470 documents, 7 epidemiological/observational (1 cohort, 1 case–control, and 5 cross‐sectional) studies on the relationship between dietary salt/sodium intakes and NAFLD risk met our inclusion criteria. The meta‐analysis of all studies showed a significant positive association between the highest salt/sodium intake and NALFD risk (OR = 1.60, 95% CI: 1.19–2.15) with a meaningful heterogeneity among studies (I2 = 96.70%, p‐value <.001). The NAFLD risk was greater in the studies with higher quality (OR = 1.81, 95% CI: 1.24–2.65) or using the equation‐based methods for NAFLD ascertainment (OR = 2.02, 95% CI: 1.29–3.17) or urinary sodium collection as a sodium intake assessment (OR = 2.48, 95% CI: 1.52–4.06). The overall certainty of the evidence was very low. In conclusion, high sodium intake seems to be related to increased NAFLD risk. Further well‐designed studies are needed to clarify this association and shed light on the underlying mechanisms. High sodium intake was related to increased NAFLD risk. The NAFLD risk was greater in the studies with higher quality or using the equation‐based methods for NAFLD ascertainment or urinary sodium collection as a sodium intake assessment. The overall certainty of the evidence was very low.
Article
Full-text available
Nonalcoholic fatty liver disease (NAFLD) is an emerging cause of chronic liver diseases and a major health problem worldwide. Dietary patterns may play a critical role in controlling and preventing this disease, but the available evidence is scarce. The current study aims to ascertain the association of adherence to the Dietary Approach to Stop Hypertension (DASH) diet and Mediterranean diet (MeD) with nonalcoholic fatty liver disease (NAFLD) among Iranian adults of the Amol Cohort Study (AmolCS). In a cross-sectional analysis among 3220 adults (55.3% men), age ≥ 18 years (46.96 ± 14.67), we measured usual dietary intake with a validated food frequency questionnaire (FFQ) and then calculated dietary pattern scores for DASH and MeD. Sociodemographic and lifestyle factors were collected by a structured questionnaire. The presence and degree of NAFLD were also determined by abdominal sonography. Multiple regression models were used to estimate NAFLD odds across tertiles of DASH and Mediterranean dietary scores. Dietary DASH and Mediterranean components were adjusted for total energy intake, based on the residual methods. After adjusting for multiple potential confounders, we found an inverse association of DASH and MeD with NAFLD (Ptrend = 0.02, and Ptrend = 0.002, respectively). Those in the highest tertiles of adherence to the DASH and MeD had the lowest risk for NAFLD (OR = 0.80, 95%CI = 0.66–0.96, OR = 0.64, 95%CI = 0.52–0.78, respectively). The results of logistic analysis of MeD, stratified by gender and abdominal obesity, revealed the favorable association was more pronounced in women (OR = 0.42, 95%CI = 0.29–0.61, Ptrend = 0.004), and in participants with or without abdominal obesity (OR = 0.62, 95% CI = 0.47–0.81, Ptrend = 0.03, OR = 0.64, 95%CI = 0.475–0.91, Ptrend = 0.04, respectively). Similar results were obtained for the adherence to DASH diet score with the prevalence of NAFLD patients with abdominal obesity (OR = 0.75, 95% CI = 0.57–0.97, Ptrend = 0.04). The findings suggested the favorable association between DASH and MeD with NAFLD in Iranian adults, especially women and subjects with or without abdominal obesity. Further prospective investigations are needed to confirm the integrity of our findings.
Article
Full-text available
Diabetes mellitus type II and obesity are two important causes of death in modern society. They are characterized by low-grade chronic inflammation and metabolic dysfunction (meta-inflammation), which is observed in all tissues involved in energy homeostasis. A substantial body of evidence has established an important role for macrophages in these tissues during the development of diabetes mellitus type II and obesity. Macrophages can activate into specialized subsets by cues from their microenvironment to handle a variety of tasks. Many different subsets have been described and in diabetes/obesity literature two main classifications are widely used that are also defined by differential metabolic reprogramming taking place to fuel their main functions. Classically activated, pro-inflammatory macrophages (often referred to as M1) favor glycolysis, produce lactate instead of metabolizing pyruvate to acetyl-CoA, and have a tricarboxylic acid cycle that is interrupted at two points. Alternatively activated macrophages (often referred to as M2) mainly use beta-oxidation of fatty acids and oxidative phosphorylation to create energy-rich molecules such as ATP and are involved in tissue repair and downregulation of inflammation. Since diabetes type II and obesity are characterized by metabolic alterations at the organism level, these alterations may also induce changes in macrophage metabolism resulting in unique macrophage activation patterns in diabetes and obesity. This review describes the interactions between metabolic reprogramming of macrophages and conditions of metabolic dysfunction like diabetes and obesity. We also focus on different possibilities of measuring a range of metabolites intra-and extracellularly in a precise and comprehensive manner to better identify the subsets of polarized macrophages that are unique to diabetes and obesity. Advantages and disadvantages of the currently most widely used metabolite analysis approaches are highlighted. We further describe how their combined use may serve to provide a comprehensive overview of the metabolic changes that take place intracellularly during macrophage activation in conditions like diabetes and obesity.
Article
Full-text available
Oxidative stress is an important factor in the etiology of several chronic diseases that include cardiovascular disease (CVD), Type 2 diabetes (T2D), and rheumatoid arthritis (RA). Oxidative stress can lead to inflammation, and this can contribute to these chronic diseases. Reducing inflammation and oxidative stress may, therefore, be useful in the prevention and treatment of these conditions. One of the treatment options for chronic diseases is the use of traditional medicine and herbs, such as Nigella sativa. This is one of the herbs that have recently been assessed for its ability to reduce inflammation and oxidative stress. We have reviewed the reported effects of N. sativa on risk factors of chronic diseases (CVD, DM, and RA) with emphasis on molecular and cellular mechanisms in controlling inflammation and oxidative stress. Various mechanisms have been proposed to contribute to the beneficial properties of N. sativa, including a reduction of lipid peroxidation via its antioxidant properties; agonist of peroxisome proliferator-activated receptor gamma in adipose tissue; activation of AMP-activated protein kinase, increased antioxidants, inhibition of nuclear factor-kappa B pathway; increased in interleukin-10 expression, CD4+ T-cell percentage, T regulatory cell percentage (CD4+ CD25+ T-cell) in peripheral blood, and CD4+/CD8+ ratio, but to prove this claim, it is necessary to conduct experimental and well-designed clinical trial studies with a larger sample size on the effects of N. sativa on these chronic diseases.
Article
Full-text available
Background & Aims Metabolic dysfunction-associated fatty liver disease (MAFLD) is a new terminology updated from non-alcoholic fatty liver disease (NAFLD). In this study, we aim to estimate the global prevalence of MAFLD specifically in overweight and obese adults from the general population by performing a systematic review and meta-analysis through mining the existing epidemiological data on fatty liver disease. Methods We searched Medline, Embase, Web of Science, Cochrane and google scholar database from inception to November, 2020. DerSimonian-Laired random-effects model with Logit transformation was performed for data analysis. Sensitivity analysis and meta-regression were used to explore predictors of MAFLD prevalence in pooled statistics with high heterogeneity. Results We identified 116 relevant studies comprised of 2667052 participants in general population with an estimated global MAFLD prevalence as 50.7% (95% CI 46.9-54.4) among overweight/obese adults regardless of diagnostic techniques. Ultrasound was the most commonly used diagnostic technique generating prevalence rate of 51.3% (95% CI 49.1-53.4). Male (59.0%, 95% CI 52.0-65.6) had a significantly higher MAFLD prevalence than female (47.5%, 95% CI 40.7-54.5). Interestingly, MAFLD prevalence rates are comparable based on classical NAFLD and non-NAFLD studies in general population. The pooled estimate prevalence of comorbidities such as type 2 diabetes and metabolic syndrome was 19.7% (95% CI 12.8-29.0) and 57.5% (95% CI 49.9-64.8), respectively. Conclusion MAFLD has an astonishingly high prevalence rate in overweight and obese adults. This calls for attention and dedicated action from primary care physicians, specialists, health policy makers and the general public alike.
Article
Aim: To explore the prognostic value of the systemic inflammatory response index (SIRI) and peripheral blood T-cell subsets in patients with hepatocellular carcinoma (HCC) and the relationship between them. Materials and Methods: We treated 352 patients with HCC with sorafenib and/or immune checkpoint inhibitors (ICIs) and analyzed SIRI and peripheral blood T cells. Results: SIRI was an independent prognostic factor for patients with HCC receiving systemic therapy. Patients with high SIRI and low baseline peripheral blood T-cell counts showed a poor response to ICIs. SIRI was significantly and negatively correlated with CD3+, CD4+ and CD8+ T-cell counts. Conclusion: SIRI markers can be employed to noninvasively assess the presence of cancer-promoting inflammation in the tumor microenvironment and predict the efficacy of targeted therapy and immunotherapy.
Article
There is no dietary strategy that has yet been specifically advocated for hemophilia. Therefore, we sought to assess the effect of the Dietary Approaches to Stop Hypertension (DASH) diet in adolescents with hemophilia. In this parallel trial, 40 male adolescents with hemophilia were dichotomized into the DASH group or control group for 10 weeks. The serum high sensitivity C-reactive protein (hs-CRP), interleukin 6 (IL-6), complete blood count (CBC), serum glutamic oxaloacetic transaminase (SGOT), serum glutamic pyruvic transaminase (SGPT), partial thromboplastin time (PTT), waist circumference (WC), percentage of body fat, fat-free mass (FFM), and liver steatosis were measured at the beginning and end of the study. Serum vitamin C was measured as a biomarker of compliance with the DASH diet. The DASH diet was designed to include high amounts of whole grains, fruits, vegetables, and low-fat dairy products, as well as low amounts of saturated fats, cholesterol, refined grains, sweets and red meat. Serum vitamin C in the DASH group was significantly increased compared to the control (P=0.001). There was a significant reduction in WC (P=0.005), fat mass (P=0.006), hepatic fibrosis (P=0.02), and PTT (P=0.008) in the DASH group, compared with the control. However, there were no significant differences regarding other selected outcomes between groups. Patients in the DASH group had significantly greater increase in the levels of red blood cell, hemoglobin, and hematocrit, as compared to control. Adherence to the DASH diet in children with hemophilia yielded significant beneficial effects on body composition, complete blood count, inflammation, and liver function.
Article
Background Although there is abundant evidence for an association between dietary pattern, weight, and other related factors, such as homeostatic model assessment for insulin resistance (HOMA-IR) and inflammatory markers; there is limited information pertaining to levels of plasminogen activator inhibitor-1 (PAI-1) and Monocyte Chemoattractant Protein -1 (MCP-1). Therefore, this study sought to examine the association between adherence to the dietary approaches to stop hypertension (DASH) diet and levels of inflammatory factors PAI-1, MCP-1, and HOMA-IR. Methods This cross-sectional study was performed on 305 obese and overweight women. The typical food intake of individuals was assessed using the 147 items semi-quantitative food frequency questionnaire (FFQ). Body components were measured for all participants. Insulin resistance was estimated by homeostasis model assessment (HOMA), and biochemical parameters were examined. Results No significant relationship was observed between the DASH diet and MCP-1 (P-trend = 0.70), PAI-1 (P-trend = 0.92), or HOMA-IR (P-trend = 0.08) in the crude model. However, there was a significant inverse relationship between the DASH diet and HOMA-IR (P-trend = 0.03) after adjusting for age, BMI, and physical activity. Conclusion This study showed that higher adherence to DASH pattern is inversely correlated with HOMA-IR in overweight and obese women.