Risk Factors of Non-alcoholic Fatty Liver Disease in the Iranian Adult Population: A Systematic Review and Meta-analysis

authors:

avatar Ehsan Amini-Salehi ORCID 1 , avatar Soheil Hassanipour 1 , avatar Farahnaz Joukar ORCID 1 , avatar Amir Ali Daryagasht ORCID 1 , avatar Mohammad-Javad Khosousi ORCID 1 , avatar Maryam Sadat Aleali 1 , avatar Malek Moien Ansar 2 , avatar Forough Heidarzad 1 , avatar Elham Abdzadeh 1 , avatar Azin Vakilpour 3 , avatar Fariborz Mansour-Ghanaei ORCID 1 , *

Gastrointestinal and Liver Diseases Research Center, Guilan University of Medical Sciences, Rasht, Iran
Department of Biochemistry, School of Medicine, Guilan University of Medical Sciences, Rasht, Iran
Guilan University of Medical Sciences, Rasht, Iran

how to cite: Amini-Salehi E, Hassanipour S, Joukar F, Daryagasht A A, Khosousi M, et al. Risk Factors of Non-alcoholic Fatty Liver Disease in the Iranian Adult Population: A Systematic Review and Meta-analysis. Hepat Mon. 2023;23(1):e131523. https://doi.org/10.5812/hepatmon-131523.

Abstract

Context:

Non-alcoholic fatty liver disease (NAFLD) is progressing considerably worldwide. Identifying the risk factors of NAFLD is a critical step in preventing its progression.

Methods:

In November 2022, two independent researchers studied seven databases, including PubMed, ISI/WoS, ProQuest, Scopus, SID, Magiran, and Google Scholar, and reference list of relevant articles, searching studies that assessed NAFLD risk factors in the Iranian adult population. Heterogeneity between studies was assessed by Cochran’s test and its composition using I2 statistics. A random-effects model was used when heterogeneity was observed; otherwise, a fixed-effects model was applied. Egger’s regression test and Trim-and-Fill analysis were used to assess publication bias. Comprehensive Meta-analysis software (version 3) was used for the analyses of the present study.

Results:

The results of this study showed significant associations between NAFLD with age (n = 15, odds ratio (OR) = 2.12, 95% CI: 1.79 - 2.51), body mass index (n = 46, OR = 5.00, 95% CI: 3.34 - 7.49), waist circumference (n = 20, OR = 6.37, 95% CI: 3.25 - 12.48), waist-to-hip ratio (n = 17, OR = 4.72, 95% CI: 3.93 - 5.66), total cholesterol (n = 39, OR = 1.80, 95% CI: 1.52 - 2.13), high-density lipoprotein (n = 37, OR = 0.53, 95% CI: 0.44 - 0.65), low-density lipoprotein (n = 31, OR = 1.68, 95% CI: 1.38 - 2.05), triglyceride (n = 31, OR = 3.21, 95% CI: 2.67 - 3.87), alanine aminotransferase (n = 26, OR = 4.06, 95% CI: 2.94 - 5.62), aspartate aminotransferase (n = 27, OR = 2.16, 95% CI: 1.50 - 3.12), hypertension (n = 13, OR = 2.53, 95% CI: 2.32 - 2.77), systolic blood pressure (n = 13, OR = 1.83, 95% CI: 1.53 - 2.18), diastolic blood pressure (n = 14, OR = 1.80, 95% CI: 1.48 - 2.20), fasting blood sugar (n = 31, OR = 2.91, 95% CI: 2.11- 4.01), homeostatic model assessment for insulin resistance (n = 5, OR = 1.92, 95% CI: 1.48 - 2.59), diabetes mellitus (n = 15, OR = 3.04, 95% CI: 2.46 - 3.75), metabolic syndrome (n = 10, OR = 3.56, 95% CI: 2.79 - 4.55), and physical activity (n = 11, OR = 0.32, 95% CI: 0.24 - 0.43) (P < 0.05).

Conclusions:

In conclusion, several factors are significantly associated with NAFLD. However, anthropometric indices had the strongest relationship with NAFLD in the Iranian adult population.

1. Context

Non-alcoholic fatty liver disease (NAFLD) is deemed to be the most common cause of chronic liver disease worldwide (1, 2). The NAFLD will become the leading cause of liver transplantation by 2030 (3). The NAFLD is defined as the accumulation of fat deposition in liver hepatocytes of more than 5% of cells volume without the presence of other conditions, such as excess alcohol usage (> 20 g/day in women, > 30 g/day in men), viral and autoimmune hepatitis, hepatotoxic drugs consumption, and endocrine conditions (4-6).

The NAFLD includes a wide range of conditions, from simple steatosis to non-alcoholic steatohepatitis (NASH) (2). Today NAFLD is thought to be a disease not only specified in the liver but other organs of the human body can be affected (7). The global prevalence of NAFLD is estimated to be 29.8%. South and North America are the regions with the highest prevalence of NAFLD, respectively (8). In a meta-analysis study by Moghaddasifar et al., NAFLD prevalence among Iranian people was estimated at 33.9%. They reported that men, obese individuals, hypertension (HTN), hypertriglyceridemia, and metabolic syndrome (MS) were associated with NAFLD in Iranian patients (9).

Various factors are involved in the development of NAFLD, including genetic features (10), diet (11), physical activity (PA) (12), smoking (13), aging (14), obesity (15), dyslipidemia (16), and HTN (17). It has been shown that there is a relationship between some biochemical blood markers and NAFLD, including alanine transaminase, aspartate transaminase (18), gamma-glutamyl transferase (19), triglyceride (TG) (20), high-density lipoprotein (HDL) (21), low-density lipoprotein (LDL) (22), total cholesterol (TC) (23), uric acid (24), and hemoglobin A1c (25). Anthropometric indices can also be an essential representative of NAFLD progression. There is a direct correlation between NAFLD and body mass index (BMI), waist circumference (WC), and waist-to-hip ratio (WHR) (26).

Several studies have been published on the Iranian population regarding NAFLD risk factors; however, the results were controversial. A comprehensive study was needed to summarize the final effects.

2. Objectives

This study aimed to estimate the risk factors of NAFLD in the Iranian adult population using a meta-analytic systematic review methodology.

3. Methods

3.1. Setting

The current study is a systematic review and meta-analysis of related risk factors of NAFLD in the Iranian adult population. The study was designed and conducted within 2021 - 2022. The Preferred Reporting Items for Systematic Reviews and Meta-analyses (PRISMA) guideline was admired for the study protocol (27).

4. Study Selection

In November 2022, two independent researchers studied seven databases, including PubMed, ISI/WoS, ProQuest, Scopus, SID, Mgiran, and Google Scholar. Two researchers also searched the references of included studies to find any possible missing studies. Table 1 shows the search strategy and keywords. Appendix 40 shows the search formula for each database.

Table 1.

Search Strategy and Keywords to Identify Related Studies in Different Databases

Search QueryKeywords [Searched Through Titles, Abstracts, Medical Subject Headings (MeSH), and General Keywords]
Query 1“Non-alcoholic fatty liver disease” or “nonalcoholic fatty liver disease” or “nonalcoholic fatty liver” or “nonalcoholic steatohepatitis” or “NAFLD”
Query 2“Epidemiology” or “prevalence” or “incidence” or “risk factor” or “related factor” or “odds ratio”
Query 3“Iran” or “Iranian people” or “Iranian population”
Final search queryQueries 1 and 2 and 3

4.1. Inclusion and Exclusion Criteria

This systematic review and meta-analysis enrolled the original studies that surveyed NAFLD patients’ risk factors in the Iranian population. Studies surveyed the adult population, defined as patients older than 18 years. Systematic reviews, meta-analyses, narrative reviews, randomized clinical trials, editorials, and commentaries were excluded. Studies that surveyed NAFLD in underlying conditions, such as Wilson’s disease, hepatitis, and liver cancer, were excluded.

4.2. Quality Assessment

Two researchers evaluated the quality of the included studies separately based on the Joanna Briggs Institute (JBI) Critical Appraisal Checklist for cross-sectional, case-control, and cohort studies (28-30). This checklist has 8 and 10 items for cross-sectional and case-control studies, respectively. The items can be answered as yes, no, unclear, and not applicable. Table 2 shows the total score of quality assessment for the included studies, and Appendix 41 shows detailed information.

Table 2.

Characteristics of Included Studies

Number of StudiesAuthor, YearRegionDesign of StudySample SizeQuality Score
Study 1Abbasalizad Farhangi et al., 2016 (31)TehranCase-control3069/10
Study 2Adibi et al., 2017 (32)IsfahanCross-sectional4836/8
Study 3Alavian et al., 2008 (33)TehranCross-sectional1,1205/8
Study 4Amirkalali et al., 2014 (34)AmolCross-sectional5,0238/8
Study 5Bagheri Lankarani et al., 2013 (35)ShirazCase-control5808/10
Study 6Bagheri Lankarani et al., 2013 (36)ShirazCross-sectional8196/8
Study 7Bahrami et al., 2019 (37)TehranCase-control9999/10
Study 8Birjandi et al., 2016 (38)KavarCross-sectional1,6006/8
Study 9Damavandi and Zeinali, 2021 (39)TehranCase-control2425/10
Study 10Darand et al., 2019 (40)TehranCase-control9598/10
Study 11Dehghan et al., 2015 (41)TehranCross-sectional1705/8
Study 12Dehghanseresht et al., 2020 (42)AhvazCase-control2439/10
Study 13Doustmohammadian et al., 2022 (43)AmolCross-sectional3,2208/8
Study 14Ebrahimi Mousavi et al., 2022 (44)AhvazCase-control2439/10
Study 15Emamat et al., 2021 (45)TehranCase-control9998/10
Study 16Entezari et al., 2021 (46)YazdCase-control2479/10
Study 17Eshraghian et al., 2013 (47)KavarCross-sectional8328/8
Study 18Fadaei et al., 2018 (48)TehranCase-control858/10
Study 19Fattahi et al., 2018 (49)SanandajCross-sectional4106/8
Study 20Ghaemi et al., 2018 (50)TehranCross-sectional1,5005/8
Study 21Hashemian et al., 2021 (51)GonbadCross-sectional1,4648/8
Study 22Hekmatdoost et al., 2016 (52)TehranCase-control3069/10
Study 23Honarvar et al., 2019 (53)ShirazCross-sectional4788/8
Study 24Khoshbaten et al., 2009 (54)TabrizCase-control2049/10
Study 25Kolahi et al., 2015 (55)TehranCase-control1707/10
Study 26Lotfi et al., 2019 (56)IsfahanCase-control6009/10
Study 27Mansour Ghanaei et al., 2018 (57)SoomehesaraCross-sectional9608/8
Study 28Mohammadi et al., 2011 (58)UrmiaCase-control1497/10
Study 29Mohammadifard et al., 2019 (59)BirjandCross-sectional1305/8
Study 30Mohseni et al., 2016 (60)TabrizCase-control1499/10
Study 31Mokhtari et al., 2017 (61)TehranCase-control9515/10
Study 32Moradzad et al., 2022 (62)SanandajCase-control1157/10
Study 33Mosallaei et al., 2015 (63)MashhadCase-control2807/10
Study 34Motamed et al., 2016 (64)AmolCross-sectional5,0527/8
Study 35Motamed et al., 2020 (65)AmolCase-control5,7979/10
Study 36Najafi et al., 2022 (66)TehranCase-control3005/10
Study 37Ostovaneh et al., 2015 (67)AmolCross-sectional5,6458/8
Study 38Ostovaneh et al., 2015 (67)ZahedanCross-sectional2,0788/8
Study 39Pasdar et al., 2016 (68)KermanshahCase-control2169/10
Study 40Pasdar et al., 2017 (69)KermanshahCase-control2508/10
Study 41Pasdar et al., 2019 (70)KermanshahCase-control2108/10
Study 42Radmard et al., 2016 (71)GonbadCross-sectional2018/8
Study 43Rezapour et al., 2021 (72)TabrizCase-control1439/10
Study 44Salehi-Sahlabadi et al., 2021 (73)IsfahanCase-control6759/10
Study 45Savadkoohi et al., 2002 (74)ZahedanCross-sectional2475/8
Study 46Shanaki et al., 2016 (75)TehranCase-control428/10
Study 47Sohouli et al., 2020 (76)TehranCase-control5159/10
Study 48Sohouli et al., 2021 (77)TabrizCase-control3669/10
Study 49Taheri et al., 2022 (78)SabzevarCase-control1,9329/10
Study 50Tutunchi et al., 2021 (79)TabrizCase-control2109/10
Study 51Tutunchi et al., 2021 (80)TabrizCase-control2109/10
Study 52Tutunchi et al., 2021 (81)TabrizCase-control1009/10
Study 53Vahid et al., 2019 (82)TehranCase-control9998/10
Study 54Zarean et al., 2019 (83)ShahrekordCase-control2,3068/10
Study 55Zolfaghari et al., 2016 (84)IsfahanCase-control3177/10

5. Data Extraction

Two independent researchers surveyed the eligible articles and extracted the data based on the current systematic review and meta-analysis objectives. The name of the first author, year of publication, the place of study, the type of study (i.e., cross-sectional, case-control, or cohort), and the sample size were identified (Table 2). The data of several risk factors of NAFLD, including age, gender, BMI, WC, WHR, TC, HDL, LDL, TG, alanine aminotransferase (ALT), aspartate aminotransferase (AST), HTN, systolic blood pressure (SBP), diastolic blood pressure (DBP), fasting blood sugar (FBS), homeostatic model assessment for insulin resistance (HOMA-IR), diabetes mellitus (DM), MS, PA, and smoking, were extracted from selected studies.

5.1. Statistical Analysis

The study’s heterogeneity was investigated by Cochran’s test (a significance level of less than 0.1) and I2 statistics (a significance level of more than 50%). A random-effects model was used for studies with heterogeneity, and a fixed-effects model was used for non-heterogenic studies. The odds ratio (OR) was obtained to combine the results of included studies. This index allowed the researchers to combine the results of included studies reported differently. A subgroup analysis was conducted based on the design of the study. Egger’s regression test was used to evaluate the asymmetry of funnel plots for any possible publication bias. Trim-and-Fill analysis was also utilized to determine the robustness of the results. Comprehensive Meta-analysis Statistical Software (version 3) was used for all the analyses of this study.

6. Results

6.1. Literature Search

After searching all international databases, 328 articles were identified, and after removing 43 duplicates, a total of 285 studies remained. Two independent researchers studied the titles and the abstracts of the remaining studies carefully, and 174 studies were excluded. The remaining studies went for full text reviewing for eligibility criteria, and 56 articles were excluded at this stage. Finally, a total number of 55 articles with a total sample size of 53,847 individuals were selected for analysis (Figure 1). Table 2 shows the information on the selected papers.

Flowchart of included eligible studies in this systematic review
Flowchart of included eligible studies in this systematic review

6.2. Characteristics of Included Studies

Table 2 shows the features and quality scores of the selected studies based on the JBI checklist. Of 55 studies, 19 and 36 studies were cross-sectional case-control, respectively. Of included studies, 43 had high quality (scores higher than 6 and 8 for cross-sectional and case-control studies, respectively). Appendix 41 shows detailed information regarding the quality assessment of the included studies. Ostovaneh et al. surveyed NAFLD epidemiology in two regions that were considered two different studies in the current meta-analysis (67).

6.3. Results of Meta-analysis

This systematic review and meta-analysis study calculated the OR for the risk factors in adults, including age, gender, BMI, WC, WHR, TC, HDL, LDL, TG, ALT, AST, HTN, SBP, DBP, FBS, HOMA-IR, DM, MS, PA, and smoking. Additionally, subgroup analysis was conducted based on the design of the studies.

In total analysis, several factors were significantly associated with NAFLD, including age, BMI, WC, WHR, TC, HDL, LDL, TG, ALT, AST, HTN, SBP, DBP, FBS, HOMA-IR, DM, MS, and PA (Appendices 1 - 18, Table 3). The relationship between gender and smoking with NAFLD was insignificant (Appendices 19 and 20, Table 3). In case-control studies, some factors were significantly associated with NAFLD, including BMI, WC, WHR, TC, HDL, LDL, TG, ALT, AST, HTN, SBP, DBP, FBS, HOMA-IR, DM, MS, PA, and smoking (Appendices 2 - 19, Table 3). In cross-sectional studies, several factors were significantly associated with NAFLD, including age, BMI, WC, WHR, TC, HDL, LDL, TG, ALT, AST, HTN, SBP, DBP, FBS, HOMA-IR, DM, and MS (Appendices 1 - 17, Table 3).

Table 3.

Results of Meta-analysis, Subgroup Analysis, and Heterogeneity for Non-alcoholic Fatty Liver Disease Risk Factors a

Variables and Design of StudiesNo. of Sample SizeNo. of StudiesOR95% CIP-ValueQ-ValueI2; No. (%)P-Value of HeterogeneityModel of Analysis
Age (32, 36, 38, 43, 47, 50, 51, 53, 57, 59, 64, 67, 71, 74)
Cross-sectional24,736152.121.79 - 2.51< 0.001119.8099.34< 0.001Random
BMI (32, 36, 37, 39-48, 50-58, 60-63, 65, 67, 69-73, 75-84)
Case-control18,661335.073.13 - 8.22< 0.0013358.2499.04< 0.001Random
Cross-sectional17,850124.852.33 - 10.09< 0.0012420.6499.54< 0.001Random
Total36,511455.003.34 - 7.49< 0.0016399.1199.32< 0.001Random
WC (34, 36, 42-44, 46, 47, 50, 51, 54, 56, 57, 60, 64, 78, 81, 85)
Case-control5,393126.812.43 - 19.03< 0.001834.7598.68< 0.001Random
Cross-sectional18,05186.072.50 - 14.74< 0.0011171.8299.40< 0.001Random
Total23,444206.373.25 - 12.48< 0.0012212.9799.14< 0.001Random
WHR (36, 38, 42, 44, 47, 56, 57, 60, 67, 68, 70, 71, 78, 79, 81, 85)
Case-control11,316113.131.45 - 6.770.004443.6997.74< 0.001Random
Cross-sectional5,15864.834.01 - 5.82< 0.00117.3871.24< 0.001Random
Total16,474174.723.93 - 5.66< 0.001462.7396.54< 0.001Random
TC (32-34, 36-40, 43, 45-50, 54, 58-63, 66-68, 70-72, 74-81, 83)
Case-control12,504251.651.34 - 2.03< 0.001185.4987.06< 0.001Random
Cross-sectional23,308142.131.60 - 2.84< 0.001311.3695.82< 0.001Random
Total35,812391.801.52 - 2.13< 0.001581.2893.46< 0.001Random
HDL (32, 34, 36-40, 43, 45-50, 54, 58-65, 67, 69-71, 75-81, 83)
Case-control18,102240.510.41 - 0.65< 0.001400.9694.26< 0.001Random
Cross-sectional26,993130.600.41 - 0.870.007593.9397.98< 0.001Random
Total45,095370.530.44 - 0.65< 0.0011590.0697.73< 0.001Random
LDL (31, 32, 36, 37, 39, 40, 43, 45, 48-50, 54, 59, 61-63, 65, 67, 70-72, 75-82)
Case-control15,494221.661.34 - 2.06< 0.001303.6093.83< 0.001Random
Cross-sectional14,48691.801.06 - 3.050.028410.8895.05< 0.001Random
Total29,980311.681.38 - 2.05< 0.0011131.7597.35< 0.001Random
TG (32-34, 36-40, 43, 45-48, 50, 54, 58, 59, 61, 62, 65, 67, 68, 70, 71, 74, 75, 79, 80, 82)
Case-control14,647172.441.63 - 3.66< 0.001739.7197.83< 0.001Random
Cross-sectional17,875143.462.81 - 4.25< 0.001194.0493.30< 0.001Random
Total32,522313.212.67 - 3.87< 0.0013475.2399.13< 0.001Random
ALT (32-34, 36-39, 43, 46, 47, 49, 50, 54, 58-60, 62, 67, 71, 76-78, 80, 82, 83)
Case-control9,008146.673.79 - 11.74< 0.001476.3597.27< 0.001Random
Cross-sectional22,197123.182.14 - 4.73< 0.001433.7197.46< 0.001Random
Total31,205264.062.94 - 5.62< 0.001985.3597.46< 0.001Random
AST (32-34, 36-39, 43, 45-47, 49, 50, 54, 58-60, 62, 67, 71, 76-78, 80, 82, 83)
Case-control10,055152.201.15 - 4.190.016719.3598.05< 0.001Random
Cross-sectional22,197122.151.38 - 3.340.001510.1297.84< 0.001Random
Total32,252272.161.50 - 3.12< 0.0011232.2897.890.175Random
HTN (32, 36, 47, 49, 54, 64, 65, 67-69, 83)
Case-control8,60861.641.14 - 2.350.00782.3393.92< 0.001Random
Cross-sectional19,85972.612.38 - 2.86< 0.0019.8439.060.132Fixed
Total28,467132.532.32 - 2.77< 0.001975.6098.77< 0.001Random
SBP (36, 38, 42-44, 47, 50, 52, 54, 69, 75, 81, 83)
Case-control2,17891.571.26 - 1.97< 0.00114.9046.300.061Fixed
Cross-sectional7,10742.351.76 - 3.14< 0.00124.8487.92< 0.001Random
Total9,285131.831.53 - 2.18< 0.00149.8875.94< 0.001Random
DBP (36, 38, 42-44, 47, 48, 50, 52, 54, 69, 75, 81, 83)
Case-control4,319101.541.23 - 1.91< 0.00120.9757.090.013Random
Cross-sectional7,10743.592.27 - 5.67< 0.00160.6695.05< 0.001Random
Total11,426141.801.48 - 2.20< 0.001177.3792.67< 0.001Random
FBS (32, 34, 36-40, 43, 45, 47, 48, 50, 54, 58, 60, 61, 63, 66-71, 76-78, 80, 81, 83)
Case-control12,033213.102.00 - 4.80< 0.001627.2696.81< 0.001Random
Cross-sectional21,356102.701.68 - 4.33< 0.001593.2598.48< 0.001Random
Total33,389312.912.11 - 4.01< 0.0011225.8197.55< 0.001Random
HOMA-IR (34, 64, 65, 67)
Case-control5,79711.271.19 - 1.35< 0.0010.000.001Fixed
Cross-sectional17,79842.191.44 - 3.34< 0.001217.2898.61< 0.001Random
Total23,59551.921.48 - 2.59< 0.001245.7898.37< 0.001Random
DM (36, 37, 39, 43, 45, 47, 49, 52, 54, 61, 63, 67, 82, 83)
Case-control8,190103.192.52 - 4.02< 0.00118.8752.320.026Random
Cross-sectional12,18552.471.52 - 4.02< 0.00153.3992.50< 0.001Random
Total20,375153.042.46 - 3.75< 0.00194.1385.12< 0.001Random
MS (32, 36, 43, 47, 67, 69, 83)
Case-control3,34642.411.42 - 4.100.00117.8083.14< 0.001Random
Cross-sectional12,59463.963.00 - 5.22< 0.00138.2286.91< 0.001Random
Total15,940103.562.79 - 4.55< 0.001203.6495.58< 0.001Random
PA (37, 40, 44, 45, 56, 61, 73, 76-78, 81)
Case-control6,307110.320.24 - 0.43< 0.00191.7989.10< 0.001Random
Total6,307110.320.24 - 0.43< 0.00191.7989.10< 0.001Random
Smoking (33, 37-39, 41-45, 47, 49-52, 55-57, 61-64, 67, 73, 76-79, 82, 83)
Case-control12,475182.251.13 - 4.480.020745.0397.71< 0.001Random
Cross-sectional18,999121.030.83 - 1.280.76463.04982.67< 0.001Random
Total31,474301.110.90 - 1.360.323866.1896.65< 0.001Random
Gender (male/female) (32-34, 36-38, 40-42, 44, 45, 49-62, 66, 68, 70-74, 76-81, 83, 84)
Case-control14,167271.000.84 - 1.200.948133.9380.58< 0.001Random
Cross-sectional15,201140.940.79 - 1.120.52958.5677.80< 0.001Random
Total29,368410.970.86 - 1.100.688196.8979.68< 0.001Random

6.4. Results of Publication Bias

Egger’s regression test was performed to assess publication bias. When any suspected asymmetry in the funnel plot was observed, Trim-and-Fill analysis was conducted to observe the stability of the results. In summary, age, BMI, HDL, LDL, TG, ALT, HTN, and DM had significant publication bias (Appendices 21 - 39, Table 4).

Table 4.

Results of Publication Bias and Trim-and-Fill Analysis for Non-alcoholic Fatty Liver Disease Risk Factors

Variables Name P-Value of Egger’s Regression TestResults of Trim-and-Fill Analysis (Point Estimate with Lower and Upper Intervals)Number of Imputed Studies in Trim-and-Fill Analysis
Age0.0232.12 (1.79 - 2.51)0
BMI< 0.0015.53 (3.94 - 7.78)0
WC0.7186.51 (3.42 - 12.38)0
WHR0.6103.73 (2.57 - 5.43)0
TC0.3871.82 (1.52 - 2.18)0
HDL< 0.0010.54 (0.44 - 0.66)0
LDL< 0.0011.71 (1.37 - 2.13)0
TG< 0.0012.86 (2.07 - 3.95)0
ALT0.0254.71 (3.41 - 6.50)0
AST0.4671.81 (1.24 - 2.64)3
HTN0.0032.09 (1.46 - 3.00)0
SBP0.4691.86 (1.54 - 2.25)0
DBP0.4442.08 (1.52 - 2.83)0
FBS0.2822.95 (2.19 - 3.98)0
DM0.0033.01 (2.32 - 3.91)0
MS0.4143.19 (2.15 - 4.73)0
PA0.2450.32 (0.24 - 0.43)0
Smoking0.1531.62 (1.14 - 2.29)0
Gender0.6290.98 (0.86 - 1.09)0

7. Discussion

The NAFLD appears as the most common chronic liver disease worldwide, which is associated with insulin resistance (IR) and the characteristics of MS and several factors, including genetic, environmental, and stress determinants. It might also exacerbate liver damage caused by other factors, such as alcohol, viruses, and industrial toxins (86-88). This systematic meta-analysis review was carried out to identify the risk factors of NAFLD in Iran. A total of 55 studies were included, from which 20 risk factors were surveyed. From the investigated factors, 18 factors, namely age, BMI, WC, WHR, TC, HDL, LDL, TG, ALT, AST, HTN, SBP, DBP, FBS, HOMA-IR, DM, MS, and PA, were significantly associated with NAFLD; however, two factors, namely gender and smoking, were not significant.

7.1. Age

This study showed that older individuals had a significantly higher risk of developing NAFLD. Other studies also showed a high prevalence of NAFLD in the older population (89-92). It has also been reported that older individuals have a higher chance of getting NASH and a higher level of hepatic fibrosis (93, 94).

Older individuals are more sensitive to oxidative stress and oxidative damage; nevertheless, the data are still conflicting (95, 96). Oxidative stress plays an important role in the pathogenesis of NAFLD (97). In addition, it is assumed that older individuals have a higher fat accumulation in the liver, muscle, and bone marrow tissues (98). It is believed there is a shift from subcutaneous adipose tissue to visceral adipose tissue in the elderly, leading to IR (99, 100). The IR is a crucial factor in the pathogenesis of NAFLD (discussed later).

7.2. Gender

Based on the results of the present study, NAFLD was more prevalent in women; however, it was not significant. Some other studies suggest that NAFLD is more common in men (101); nevertheless, other studies believe vice versa (34).

It has been demonstrated that post-menopausal women under hormone replacement therapy (HRT) had lower NAFLD prevalence than post-menopausal women without HRT (87). These phenomena reflect the role of estrogen hormones as a protective factor in developing NAFLD. These hormones have anti-inflammatory, antifibrotic, antiapoptotic, and antioxidant characteristics (102-104).

Estrogen hormones also play an important role in regional fat distribution. It is assumed that estrogen supports subcutaneous fat accumulation rather than visceral fat (105, 106). The direct relationship between the amount of visceral fat and the progression of NAFLD has been demonstrated (107, 108). Gynoid glute-femoral subcutaneous fat reduces the risk of metabolic disorders; however, android visceral fat, which is higher in men, increases the risk of metabolic disorders (109). Glute-femoral adipocytes are less responsive to epinephrine and norepinephrine lipolytic effects and distribute less free fatty acid (FFA) than abdominal adipose tissue (110, 111). Some studies believe estradiol reduces lipolysis and enhances body organ insulin sensitivity (112, 113). Another interesting point that makes NAFLD more prevalent in men is the difference in muscle physiology in both genders. Skeletal muscles are more resistant to insulin in men than women, and IR plays an essential role in the pathogenesis of NAFLD (discussed later) (114, 115).

7.3. Anthropometric Indices

In the current meta-analysis, BMI, WC, and WHR were strong risk factors for NAFLD progression. These anthropometric measurements are related to obesity and can be used in obesity-related health risks (116-119). The association between obesity and NAFLD is a complex phenomenon and is affected by many metabolic pathways. Steatosis has resulted from an imbalance between more FFA input to the liver and less output. The FFA input to the liver depends on liver uptake activity and de novo lipogenesis. Liver uptake activity is affected by liver capacity in uptake and the amount of FFA delivered to the liver. The FFA delivered to the liver originates from adipose tissue and circulating TG (15).

Previous studies have shown an increased liver capacity for FFA uptake in obese individuals. In obese individuals with NAFLD, the gene expression of FAT/CD36, which is an important membrane protein in transporting FFA into tissues, increases in liver and skeletal muscle and decreases in adipose tissue, suggesting a shift in the absorption of FFA from adipose tissue to hepatic tissue (120, 121). Delivery of FFA to the liver also increases in obese individuals. The rates of FFA released from adipose tissue and gene expression of hepatic lipase and hepatic lipoprotein lipase (i.e., an enzyme that changes circulating TG to FFA) are higher in obese individuals (122-124). According to some studies, hepatic de novo lipogenesis is higher in obese individuals due to the overexpression of some involved genes (125, 126).

Another important key in the progression of NAFLD in obese individuals is inflammation. Chemokines [e.g., C-C motif chemokine ligand 2 (CCL2)] and cytokines [e.g., tumor necrosis factor-alpha (TNF-α)] secreted from adipose tissue can lead to IR and inflammation (124). The secretion rate of adipose tissue chemokines and cytokines increases in obese individuals suggesting their role in the pathogenesis of NAFLD (126).

7.4. Dyslipidemia

Atherogenic dyslipidemia, defined as increased plasma TG, small dense LDL, and decreased HDL-C, is noted in patients suffering from NAFLD (16). The fact that NAFLD patients have hypertriglyceridemia is complicated and is affected by several determinants. One of the involved factors in the metabolism is very low-density lipoprotein (VLDL). The VLDLs are TG-carrying particles released from the liver and contain the highest fasting plasma TG (127).

Intrahepatic triglyceride (IHTG) is directly affected by hepatic VLDL production in obese individuals with normal liver fat (128, 129). Studies have shown that in patients with IHTG of more than 10%, the relationship between IHTG and VLDL is disrupted (128, 129). This evidence recommends that VLDL secretion can only compensate for the increased rate of IHTG in a distinct range and might not work correctly in NAFLD subjects. Moreover, IR, observed in NAFLD individuals (130), can decrease the clearance of VLDL from plasma (127). Previous studies have shown that insulin can reduce hepatic VLDL production and increase VLDL clearance by enhancing lipoprotein lipase activity (131). Altogether, the occurrence of these processes simultaneously can justify the coexistence of NAFLD and hypertriglyceridemia.

The current meta-analysis showed that the increased serum LDL was significantly associated with NAFLD development. The LDL is removed from circulatory blood by several receptors on the liver, such as LDL receptor (LDLR) and LDLR related-protein 1 (127). Proprotein convertase subtilisin/kexin type 9 (PCSK9) is a protease that reduces LDLR in the liver leading to increase serum LDL levels (127). Studies have shown that PCSK9 activity has a positive relationship with steatosis status. Additionally, in NAFLD patients, higher PCSK9 activity and higher serum LDL levels are reported (132, 133).

The HDL is a protective determinant for NAFLD patients, and low HDL levels are related to NAFLD progression. Low HDL levels in NAFLD patients can result from hypertriglyceridemia (discussed above). Hypertriglyceridemia can promote cholesterol ester transfer protein activity, producing TG-enriched HDL particles. These particles seem to be removed faster from circulatory blood, leading to lower serum HDL levels (134, 135).

Cholesterol was another critical risk factor in the present meta-analysis. In this study, the relationship between hypercholesterolemia and NAFLD was stronger than in the American population (136). Recent studies claim that hepatic free cholesterol (FC) accumulation is a hallmark of NAFLD pathogenesis and is related to the disease’s histological severity (137, 138). The available evidence demonstrates that cholesterol homeostasis is altered in NAFLD, leading to FC deposition in the liver (139). In animal studies, the accumulation of FC in the liver exacerbates steatohepatitis and liver fibrosis (140-144). The expression and activity of 3-hydroxy-3-methylglutaryl coenzyme A reductase, an essential enzyme in cholesterol biosynthesis (145, 146), are increased in patients with NAFLD leading to increased cholesterol levels (137, 147).

7.5. Hypertension

The HTN is a multifactorial disease influenced by genetic and environmental determinants and is becoming a public health concern (148). The relationship between HTN and NAFLD is a complex issue. It is not clear whether NAFLD is caused by HTN or vice versa. The present study showed that in addition to HTN, high SBP and high DBP are important risk factors for NAFLD.

The mechanisms by which NAFLD can promote HTN are not well understood, and further studies are needed in this regard (149). Several mechanisms can justify the act of NAFLD in HTN progression. There is a relationship between the body’s inflammatory state and sympathetic nervous system stimulation, which plays an important role in the pathogenesis of HTN (150). The NAFLD is considered to be associated with systematic inflammation recognized by the increased serum levels of interleukin 6, TNF-α, and CCL2 (151).

Another mechanism in which NAFLD leads to HTN is IR. In systemic IR, several determinants can result in HTN, such as perivascular and renal sinus fat deposition, endothelial cell dysfunction, and activation of the renin-angiotensin-aldosterone system (RAAS) (149, 152, 153). Other mechanisms leading to HTN in NAFLD include decreased vasodilation and increased vasoconstriction. Several factors can reduce and impair nitric oxide (a vasodilator) activity in NAFLD patients. In addition, as discussed above, the RAAS becomes more active in NAFLD patients, and this hyperactivity can lead to vasoconstriction (154). In NAFLD patients, arterial stiffness develops, affecting contractile vessel function (155).

7.6. Liver Enzymes

In the current study, elevated serum AST and ALT levels were significantly associated with NAFLD; however, some studies reported normal liver enzymes in these individuals (156-159). The present study demonstrated that the relationship between AST and NAFLD was more robust than between ALT and NAFLD. One of the indicative determinants of hepatic injury is ALT which can be considered a maker representing the presence of NAFLD (160). In some studies, AST to platelet ratio index is a prognostic criterion for liver fibrosis in NAFLD patients (161-163). One of the hypotheses about NAFLD pathogenesis that can justify the increased levels of AST and ALT is the two-hit theory. The first hit is defined as fat accumulation in the liver, and the second hit is a liver injury caused by reactive oxygen species-induced necroinflammation (114, 164, 165). The authors of the present study believe that this necroinflammation and hepatocyte injury can result in higher levels of intracellular hepatic enzymes, such as AST and ALT.

7.7. Diabetes Mellitus

The DM was one of the strongest factors associated with NAFLD. The DM and NAFLD have a bilateral relationship, and these two pathologic statuses usually coexist (166-168). Previous studies showed that DM could result in NAFLD progression to NASH, advanced fibrosis, or cirrhosis (169). The pathophysiologic mechanisms of the relationship between NAFLD and DM are not understood well, and debates are going on. The increased chance of getting DM in NAFLD can occur in the early phase (associated with fat accumulation) and late phase (associated with hepatic inflammation) of the disease (169).

Fat accumulation in the liver, which occurs in the first hit regarding the two-hit theory (discussed above), is related to hepatic, adipose, and muscle tissue IR, increasing the chance of DM (3, 166, 169-172). The inflammation of the liver, which occurs in the second hit regarding the two-hit theory (discussed above), is associated with hepatocytes and pro-inflammatory cytokines, such as fetuin-A, fetuin B, angiopoietin-like protein, fibroblast growth factor 21, and selenoprotein, which can increase the risk of DM (3, 167, 173-176). Another mechanism in the pathogenesis of NAFLD and DM is insulin clearance. Lower insulin clearance is shown in patients with NAFLD, and lower hepatic insulin extraction from blood seems to be related to increased hepatic steatosis (169).

In the present study, the IR score (HOMA-IR) was significantly associated with NAFLD. The IR is an important key in the pathogenesis of NAFLD. Several studies have shown that obesity (as discussed above), excess nutrition, and MS play an important role in NAFLD patients; accordingly, a “high-fed status” is expected in these patients (177-180). In high-fed status, FFA is transferred to adipose tissue and, by the effect of insulin, is changed to TG. By this time, the chronic inflammation in adipose tissue leads to peripheral IR (181, 182). In the state of IR, lipogenesis (i.e., the transformation of FFA to TG) does not occur; therefore, circulatory FFA increases (183). These FFA are then transferred to the liver and contribute to de novo lipogenesis (182). A higher de novo lipogenesis is a feature in NAFLD patients (discussed above).

Another phenomenon in the pathogenesis of IR in NAFLD is defined as selective IR. One of the functions of insulin is inhibiting gluconeogenesis and activating de novo lipogenesis in the liver. However, in insulin-resistant NAFLD patients, insulin fails to inhibit gluconeogenesis but can stimulate de novo lipogenesis (182, 184).

7.8. Smoking

The relationship between smoking and NAFLD is controversial, and further studies are needed in this regard. Some studies imply the effect of tobacco on NAFLD progression (185-187). On the other hand, there is some evidence of a non-significant relationship between smoking and NAFLD (188). A significant relationship between NAFLD and smoking was shown in a meta-analysis study by Akhavan Rezayat et al. However, in the present study, this relationship was not significant with a larger sample size (13).

Some witnesses recommend the role of smoking in NAFLD progression. Previous experimental studies have shown the effect of cigarettes on insulin function, which can lead to IR (189, 190). As discussed above, IR is critical in developing NAFLD. The present study talked about the relationship between DM and NAFLD previously. The role of smoking in DM progression and glucose intolerance is also observed in some studies (191-193). Moreover, MS is more prevalent in smokers (194, 195). The links between oxidative stress, pro-inflammatory cytokines, and NAFLD have been discussed. Previous studies also showed the role of smoking in stimulating oxidative stress and inflammation (196-198).

There is also some evidence suggesting the effect of cigarettes on NAFLD prevention. Some studies imply smoking can decrease BMI, and cigarette cessation can lead to following weight gain and DM (199). Moreover, acute energy expenditure is observed with smoking (200). Altogether, some evidence suggests the role of smoking in NAFLD progression; however, other studies reject it. Accordingly, the non-significant effect of smoking on NAFLD development can be justified.

7.9. Limitations

This study identified the risk factors for NAFLD in the Iranian adult population. However, the study had some limitations. There was significant heterogeneity for all variables. For the assessment of the source of heterogeneity, subgroup analysis was performed based on the design of studies (i.e., case-control and cross-sectional). Nevertheless, significant heterogeneity remained for most variables. This review also included studies with low quality, which can affect the pooled effects. In this meta-analysis, the included studies were conducted in 11 cities in Iran, and no study was available for most of the cities; therefore, the results might not be a precise representation of the Iranian population. It was impossible to assess the difference between urban and rural individuals due to a lack of data. It is suggested to perform further studies on the rural population as these individuals have different lifestyles in comparison to urban residents. Not enough information was available regarding the duration of diabetes; therefore, the present study did not assess the effect of this factor on NAFLD. It is recommended to perform future studies to evaluate the duration of DM in diabetic patients who have NAFLD.

8. Conclusions

In conclusion, this study aimed to identify NAFLD risk factors in the Iranian adult population. Among the 18 identified risk factors, WC had the strongest relationship with NAFLD, followed by BMI and WHR. High serum HDL levels and PA were considered two protective factors for NAFLD. Smoking and gender did not have a significant relationship with NAFLD.

References

  • 1.

    Bellentani S, Marino M. Epidemiology and natural history of non-alcoholic fatty liver disease (NAFLD). Ann Hepatol. 2009;8 Suppl 1:S4-8. [PubMed ID: 19381118].

  • 2.

    Younossi ZM, Koenig AB, Abdelatif D, Fazel Y, Henry L, Wymer M. Global epidemiology of nonalcoholic fatty liver disease-Meta-analytic assessment of prevalence, incidence, and outcomes. Hepatology. 2016;64(1):73-84. [PubMed ID: 26707365]. https://doi.org/10.1002/hep.28431.

  • 3.

    Byrne CD, Targher G. NAFLD: a multisystem disease. J Hepatol. 2015;62(1 Suppl):S47-64. [PubMed ID: 25920090]. https://doi.org/10.1016/j.jhep.2014.12.012.

  • 4.

    Aller R, Izaola O, Gomez S, Tafur C, Gonzalez G, Berroa E, et al. Effect of silymarin plus vitamin E in patients with non-alcoholic fatty liver disease. A randomized clinical pilot study. Eur Rev Med Pharmacol Sci. 2015;19(16):3118-24. [PubMed ID: 26367736].

  • 5.

    National Guideline Centre (UK). Non-alcoholic fatty liver disease: Assessment and management. London; 2016.

  • 6.

    European Association for the Study of the L, European Association for the Study of D, European Association for the Study of O. EASL-EASD-EASO Clinical Practice Guidelines for the management of non-alcoholic fatty liver disease. J Hepatol. 2016;64(6):1388-402. [PubMed ID: 27062661]. https://doi.org/10.1016/j.jhep.2015.11.004.

  • 7.

    Armstrong MJ, Adams LA, Canbay A, Syn WK. Extrahepatic complications of nonalcoholic fatty liver disease. Hepatology. 2014;59(3):1174-97. [PubMed ID: 24002776]. https://doi.org/10.1002/hep.26717.

  • 8.

    Le MH, Yeo YH, Li X, Li J, Zou B, Wu Y, et al. 2019 Global NAFLD prevalence: A systematic review and meta-analysis. Clin Gastroenterol Hepatol. 2022;20(12):2809-2817 e28. [PubMed ID: 34890795]. https://doi.org/10.1016/j.cgh.2021.12.002.

  • 9.

    Moghaddasifar I, Lankarani KB, Moosazadeh M, Afshari M, Ghaemi A, Aliramezany M, et al. Prevalence of non-alcoholic fatty liver disease and its related factors in Iran. Int J Organ Transplant Med. 2016;7(3):149-60. [PubMed ID: 27721961]. [PubMed Central ID: PMC5054138].

  • 10.

    Dongiovanni P, Anstee QM, Valenti L. Genetic predisposition in NAFLD and NASH: impact on severity of liver disease and response to treatment. Curr Pharm Des. 2013;19(29):5219-38. [PubMed ID: 23394097]. [PubMed Central ID: PMC3850262]. https://doi.org/10.2174/13816128113199990381.

  • 11.

    Perdomo CM, Fruhbeck G, Escalada J. Impact of nutritional changes on nonalcoholic fatty liver disease. Nutrients. 2019;11(3). [PubMed ID: 30901929]. [PubMed Central ID: PMC6470750]. https://doi.org/10.3390/nu11030677.

  • 12.

    Li Y, He F, He Y, Pan X, Wu Y, Hu Z, et al. Dose-response association between physical activity and non-alcoholic fatty liver disease: A case-control study in a Chinese population. BMJ Open. 2019;9(3). e026854. [PubMed ID: 30928957]. [PubMed Central ID: PMC6475196]. https://doi.org/10.1136/bmjopen-2018-026854.

  • 13.

    Akhavan Rezayat A, Dadgar Moghadam M, Ghasemi Nour M, Shirazinia M, Ghodsi H, Rouhbakhsh Zahmatkesh MR, et al. Association between smoking and non-alcoholic fatty liver disease: A systematic review and meta-analysis. SAGE Open Med. 2018;6:2050312117745220. [PubMed ID: 29399359]. [PubMed Central ID: PMC5788091]. https://doi.org/10.1177/2050312117745223.

  • 14.

    Gan L, Chitturi S, Farrell GC. Mechanisms and implications of age-related changes in the liver: Nonalcoholic Fatty liver disease in the elderly. Curr Gerontol Geriatr Res. 2011;2011:831536. [PubMed ID: 21918648]. [PubMed Central ID: PMC3171768]. https://doi.org/10.1155/2011/831536.

  • 15.

    Fabbrini E, Sullivan S, Klein S. Obesity and nonalcoholic fatty liver disease: Biochemical, metabolic, and clinical implications. Hepatology. 2010;51(2):679-89. [PubMed ID: 20041406]. [PubMed Central ID: PMC3575093]. https://doi.org/10.1002/hep.23280.

  • 16.

    Chatrath H, Vuppalanchi R, Chalasani N. Dyslipidemia in patients with nonalcoholic fatty liver disease. Semin Liver Dis. 2012;32(1):22-9. [PubMed ID: 22418885]. [PubMed Central ID: PMC3654545]. https://doi.org/10.1055/s-0032-1306423.

  • 17.

    Wang Y, Zeng Y, Lin C, Chen Z. Hypertension and non-alcoholic fatty liver disease proven by transient elastography. Hepatol Res. 2016;46(13):1304-10. [PubMed ID: 26932594]. https://doi.org/10.1111/hepr.12688.

  • 18.

    Hadizadeh F, Faghihimani E, Adibi P. Nonalcoholic fatty liver disease: Diagnostic biomarkers. World J Gastrointest Pathophysiol. 2017;8(2):11-26. [PubMed ID: 28573064]. [PubMed Central ID: PMC5437499]. https://doi.org/10.4291/wjgp.v8.i2.11.

  • 19.

    Kasapoglu B, Turkay C, Yalcin KS, Carlioglu A, Koktener A. Role of gamma-glutamyl transferase levels in prediction of high cardiovascular risk among patients with non-alcoholic fatty liver disease. Indian J Med Res. 2016;143(1):30-6. [PubMed ID: 26997011]. [PubMed Central ID: PMC4822365]. https://doi.org/10.4103/0971-5916.178585.

  • 20.

    Tomizawa M, Kawanabe Y, Shinozaki F, Sato S, Motoyoshi Y, Sugiyama T, et al. Triglyceride is strongly associated with nonalcoholic fatty liver disease among markers of hyperlipidemia and diabetes. Biomed Rep. 2014;2(5):633-6. [PubMed ID: 25054002]. [PubMed Central ID: PMC4106613]. https://doi.org/10.3892/br.2014.309.

  • 21.

    Trojak A, Walus-Miarka M, Wozniakiewicz E, Malecki MT, Idzior-Walus B. Nonalcoholic fatty liver disease is associated with low HDL cholesterol and coronary angioplasty in patients with type 2 diabetes. Med Sci Monit. 2013;19:1167-72. [PubMed ID: 24336007]. [PubMed Central ID: PMC3871489]. https://doi.org/10.12659/MSM.889649.

  • 22.

    Sun DQ, Liu WY, Wu SJ, Zhu GQ, Braddock M, Zhang DC, et al. Increased levels of low-density lipoprotein cholesterol within the normal range as a risk factor for nonalcoholic fatty liver disease. Oncotarget. 2016;7(5):5728-37. [PubMed ID: 26735337]. [PubMed Central ID: PMC4868717]. https://doi.org/10.18632/oncotarget.6799.

  • 23.

    Peng K, Mo Z, Tian G. Serum lipid abnormalities and nonalcoholic fatty liver disease in adult males. Am J Med Sci. 2017;353(3):236-41. [PubMed ID: 28262209]. https://doi.org/10.1016/j.amjms.2017.01.002.

  • 24.

    Moon SS. Relationship between serum uric acid level and nonalcoholic fatty liver disease in pre- and postmenopausal women. Ann Nutr Metab. 2013;62(2):158-63. [PubMed ID: 23406781]. https://doi.org/10.1159/000346202.

  • 25.

    Chen C, Zhu Z, Mao Y, Xu Y, Du J, Tang X, et al. HbA1c may contribute to the development of non-alcoholic fatty liver disease even at normal-range levels. Biosci Rep. 2020;40(1). [PubMed ID: 31940026]. [PubMed Central ID: PMC6997109]. https://doi.org/10.1042/BSR20193996.

  • 26.

    Reis SS, Callejas GH, Marques RA, Gestic MA, Utrini MP, Chaim FDM, et al. Correlation Between Anthropometric Measurements and Non-alcoholic Fatty Liver Disease in Individuals With Obesity Undergoing Bariatric Surgery: Cross-Sectional Study. Obes Surg. 2021;31(8):3675-85. [PubMed ID: 33982243]. https://doi.org/10.1007/s11695-021-05470-2.

  • 27.

    Liberati A, Altman DG, Tetzlaff J, Mulrow C, Gotzsche PC, Ioannidis JP, et al. The PRISMA statement for reporting systematic reviews and meta-analyses of studies that evaluate health care interventions: Explanation and elaboration. PLoS Med. 2009;6(7). e1000100. [PubMed ID: 19621070]. [PubMed Central ID: PMC2707010]. https://doi.org/10.1371/journal.pmed.1000100.

  • 28.

    Joanna Briggs Institute. Checklist for analytical cross sectional studies. Adelaide, South Australia: Joanna Briggs Institute; 2017.

  • 29.

    Joanna Briggs Institute. Ckecklist for case control studies 2017. Adelaide, South Australia: Joanna Briggs Institute; 2017.

  • 30.

    Joanna Briggs Institute. Checklist for cohort studies 2017. Adelaide, South Australia: Joanna Briggs Institute; 2017.

  • 31.

    Abbasalizad Farhangi M, Mohseni F, Farajnia S, Jafarabadi MA. Major components of metabolic syndrome and nutritional intakes in different genotype of UCP2 -866G/A gene polymorphisms in patients with NAFLD. J Transl Med. 2016;14(1):177. [PubMed ID: 27301474]. [PubMed Central ID: PMC4908770]. https://doi.org/10.1186/s12967-016-0936-3.

  • 32.

    Adibi A, Maleki S, Adibi P, Etminani R, Hovsepian S. Prevalence of nonalcoholic fatty liver disease and its related metabolic risk factors in Isfahan, Iran. Adv Biomed Res. 2017;6:47. [PubMed ID: 28503502]. [PubMed Central ID: PMC5414408]. https://doi.org/10.4103/2277-9175.204590.

  • 33.

    Alavian SM, Ramezani MAJID, Bazzaz A, Azuzabadi Farahani M, Behnava B, et al. [Frequency of fatty liver and some of its risk factors in asymptomatic carriers of HBV attending the Tehran blood transfusion organization hepatitis clinic]. Iran J Endocrinol Metab. 2008;10(2):99-106. Persian.

  • 34.

    Amirkalali B, Poustchi H, Keyvani H, Khansari MR, Ajdarkosh H, Maadi M, et al. Prevalence of non-alcoholic fatty liver disease and its predictors in North of Iran. Iran J Public Health. 2014;43(9):1275-83. [PubMed ID: 26175982]. [PubMed Central ID: PMC4500430].

  • 35.

    Bagheri Lankarani K, Mahmoodi M, Lotfi M, Zamiri N, Heydari ST, Ghaffarpasand F, et al. Common carotid intima-media thickness in patients with non-alcoholic fatty liver disease: A population-based case-control study. Korean J Gastroenterol. 2013;62(6):344-51. [PubMed ID: 24365733]. https://doi.org/10.4166/kjg.2013.62.6.344.

  • 36.

    Bagheri Lankarani K, Ghaffarpasand F, Mahmoodi M, Lotfi M, Zamiri N, Heydari ST, et al. Non alcoholic fatty liver disease in southern Iran: A population based study. Hepat Mon. 2013;13(5). e9248. [PubMed ID: 23922564]. [PubMed Central ID: PMC3734894]. https://doi.org/10.5812/hepatmon.9248.

  • 37.

    Bahrami A, Teymoori F, Eslamparast T, Sohrab G, Hejazi E, Poustchi H, et al. Legume intake and risk of nonalcoholic fatty liver disease. Indian J Gastroenterol. 2019;38(1):55-60. [PubMed ID: 30796701]. https://doi.org/10.1007/s12664-019-00937-8.

  • 38.

    Birjandi M, Ayatollahi SM, Pourahmad S, Safarpour AR. Prediction and diagnosis of non-alcoholic fatty liver disease (NAFLD) and identification of its associated factors using the classification tree method. Iran Red Crescent Med J. 2016;18(11). e32858. [PubMed ID: 28191344]. [PubMed Central ID: PMC5292777]. https://doi.org/10.5812/ircmj.32858.

  • 39.

    Damavandi N, Zeinali S. Association of xenobiotic-metabolizing enzymes (GSTM1 and GSTT 1), and pro-inflammatory cytokines (TNF-alpha and IL-6) genetic polymorphisms with non-alcoholic fatty liver disease. Mol Biol Rep. 2021;48(2):1225-31. [PubMed ID: 33492571]. https://doi.org/10.1007/s11033-021-06142-1.

  • 40.

    Darand M, Darabi Z, Yari Z, Hekmatdoost A. Fructose consumption is associated with non-alcoholic fatty liver disease risk: A case-control study from Iran. Hepat Mon. 2019;19(4). https://doi.org/10.5812/hepatmon.88283.

  • 41.

    Dehghan P, Miwechi M, Izadi E, Mohammadi F, Sohrabi MR. Comparison of physical activity and body mass index in patients with and without non-alcoholic fatty liver disease. COMMONITY HEALTH. 2015.

  • 42.

    Dehghanseresht N, Jafarirad S, Alavinejad SP, Mansoori A. Association of the dietary patterns with the risk of non-alcoholic fatty liver disease among Iranian population: A case-control study. Nutr J. 2020;19(1):63. [PubMed ID: 32605646]. [PubMed Central ID: PMC7329390]. https://doi.org/10.1186/s12937-020-00580-6.

  • 43.

    Doustmohammadian A, Clark CCT, Maadi M, Motamed N, Sobhrakhshankhah E, Ajdarkosh H, et al. Favorable association between Mediterranean diet (MeD) and DASH with NAFLD among Iranian adults of the Amol Cohort Study (AmolCS). Sci Rep. 2022;12(1):2131. [PubMed ID: 35136128]. [PubMed Central ID: PMC8825797]. https://doi.org/10.1038/s41598-022-06035-8.

  • 44.

    Ebrahimi Mousavi S, Dehghanseresht N, Dashti F, Khazaei Y, Salamat S, Asbaghi O, et al. The association between Dietary Diversity Score and odds of nonalcoholic fatty liver disease: a case-control study. Eur J Gastroenterol Hepatol. 2022;34(6):678-85. [PubMed ID: 35352692]. https://doi.org/10.1097/MEG.0000000000002344.

  • 45.

    Emamat H, Ghalandari H, Totmaj AS, Tangestani H, Hekmatdoost A. Calcium to magnesium intake ratio and non-alcoholic fatty liver disease development: a case-control study. BMC Endocr Disord. 2021;21(1):51. [PubMed ID: 33736626]. [PubMed Central ID: PMC7972345]. https://doi.org/10.1186/s12902-021-00721-w.

  • 46.

    Entezari MR, Talenezhad N, Mirzavandi F, Rahimpour S, Mozaffari-Khosravi H, Fallahzadeh H, et al. Mediterranean dietary pattern and non-alcoholic fatty liver diseases: a case-control study. J Nutr Sci. 2021;10. e55. [PubMed ID: 34367629]. [PubMed Central ID: PMC8327389]. https://doi.org/10.1017/jns.2021.43.

  • 47.

    Eshraghian A, Dabbaghmanesh MH, Eshraghian H, Fattahi MR, Omrani GR. Nonalcoholic fatty liver disease in a cluster of Iranian population: Thyroid status and metabolic risk factors. Arch Iran Med. 2013;16(10):584-9. [PubMed ID: 24093139].

  • 48.

    Fadaei R, Poustchi H, Meshkani R, Moradi N, Golmohammadi T, Merat S. Impaired HDL cholesterol efflux capacity in patients with non-alcoholic fatty liver disease is associated with subclinical atherosclerosis. Sci Rep. 2018;8(1):11691. [PubMed ID: 30076407]. [PubMed Central ID: PMC6076293]. https://doi.org/10.1038/s41598-018-29639-5.

  • 49.

    Fattahi N, Sharifi K, Moradi G, Iri R, Reshadat R, Ataee P, et al. Prevalence of non-alcoholic fatty liver disease in Kurdistan province, Iran, 2013–2014: A population based study. Govaresh. 2018;23(2):107-13.

  • 50.

    Ghaemi A, Hosseini N, Osati S, Naghizadeh MM, Dehghan A, Ehrampoush E, et al. Waist circumference is a mediator of dietary pattern in Non-alcoholic fatty liver disease. Sci Rep. 2018;8(1):4788. [PubMed ID: 29555959]. [PubMed Central ID: PMC5859081]. https://doi.org/10.1038/s41598-018-23192-x.

  • 51.

    Hashemian M, Merat S, Poustchi H, Jafari E, Radmard AR, Kamangar F, et al. Red meat consumption and risk of nonalcoholic fatty liver disease in a population with low meat consumption: The golestan cohort study. Am J Gastroenterol. 2021;116(8):1667-75. [PubMed ID: 33767101]. [PubMed Central ID: PMC8460710]. https://doi.org/10.14309/ajg.0000000000001229.

  • 52.

    Hekmatdoost A, Shamsipour A, Meibodi M, Gheibizadeh N, Eslamparast 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. [PubMed ID: 27436528]. https://doi.org/10.1080/09637486.2016.1210101.

  • 53.

    Honarvar B, Bagheri Lankarani K, Keshani P, Rafiee T. Dietary determinants of non-alcoholic fatty liver disease in lean and non-lean adult patients: A population-based study in Shiraz, Southern Iran. Hepatitis Monthly. 2017;17(4). https://doi.org/10.5812/hepatmon.44962.

  • 54.

    Khoshbaten M, Fatahi E, Soomi H, Farhang S, Majidi G, Fatahi V. [Clinico-Biochemical comparison of patients with nonalcoholic fatty liver disease and healthy populace]. Zahedan j res med sci. 2009;11(1). Persian.

  • 55.

    Kolahi AA, Pakdaman R, Mivehchi M, Dehghan P. Comparison of nutritional behaviors and body mass index in patients with and without non-alcoholic fatty liver diseases. community healthcare. 2015.

  • 56.

    Lotfi A, Saneei P, Hekmatdost A, Salehisahlabadi A, Shiranian A, Ghiasvand R. The relationship between dietary antioxidant intake and physical activity rate with nonalcoholic fatty liver disease (NAFLD): A case - Control study. Clin Nutr ESPEN. 2019;34:45-9. [PubMed ID: 31677710]. https://doi.org/10.1016/j.clnesp.2019.09.004.

  • 57.

    Mansour-Ghanaei R, Mansour-Ghanaei F, Naghipour M, Joukar F, Atrkar-Roushan Z, Tabatabaii M, et al. The role of anthropometric indices in the prediction of non-alcoholic fatty liver disease in the PERSIAN Guilan Cohort study (PGCS). J Med Life. 2018;11(3):194-202. [PubMed ID: 30364682]. [PubMed Central ID: PMC6197514]. https://doi.org/10.25122/jml-2018-0031.

  • 58.

    Mohammadi A, Sedani HH, Ghasemi-Rad M. Evaluation of carotid intima-media thickness and flow-mediated dilatation in middle-aged patients with nonalcoholic fatty liver disease. Vasc Health Risk Manag. 2011;7:661-5. [PubMed ID: 22140316]. [PubMed Central ID: PMC3225348]. https://doi.org/10.2147/VHRM.S26011.

  • 59.

    Mohammadifard M, Saremi Z, Rastgoo M, Akbari E. Relevance between Helicobacter pylori Infection and Non-Alcoholic Fatty Liver Disease in Birjand, Iran. J Med Life. 2019;12(2):168-72. [PubMed ID: 31406519]. [PubMed Central ID: PMC6685302]. https://doi.org/10.25122/jml-2019-0012.

  • 60.

    Mohseni F, Rashvand Z, Najafipour R, Hadizadeh S, Moghbelinejad S. Evaluating -238 G>A polymorphism association in TNF-alpha gene with metabolic parameters and nutritional intakes among the Iranian NAFLD patients. Biochem Genet. 2016;54(5):685-95. [PubMed ID: 27344153]. https://doi.org/10.1007/s10528-016-9747-8.

  • 61.

    Mokhtari Z, Poustchi H, Eslamparast T, Hekmatdoost A. Egg consumption and risk of non-alcoholic fatty liver disease. World J Hepatol. 2017;9(10):503-9. [PubMed ID: 28443155]. [PubMed Central ID: PMC5387362]. https://doi.org/10.4254/wjh.v9.i10.503.

  • 62.

    Moradzad M, Abdi M, Sheikh Esmaeili F, Ghaderi D, Rahmani K, Moloudi MR, et al. Possible correlation between high circulatory levels of trimethylamine-N-oxide and 2177G>C polymorphisms of hepatic flavin containing monooxygenase 3 in Kurdish Population with non-alcoholic fatty liver disease. Mol Biol Rep. 2022;49(7):5927-37. [PubMed ID: 35348964]. https://doi.org/10.1007/s11033-022-07375-4.

  • 63.

    Mosallaei Z, Mazidi M, Safariyan M, Norouzy A, Mohajeri SAR, Esmaily H, et al. Dietary intake and its relationship with non-alcoholic fatty liver disease (NAFLD). Mediterranean Journal of Nutrition and Metabolism. 2015;8(2):139-48. https://doi.org/10.3233/mnm-150032.

  • 64.

    Motamed N, Maadi M, Sohrabi M, Keyvani H, Poustchi H, Zamani F. Rural residency has a protective effect and marriage is a risk factor for NAFLD. Hepat Mon. 2016;16(7). e38357. [PubMed ID: 27642349]. [PubMed Central ID: PMC5018359]. https://doi.org/10.5812/hepatmon.38357.

  • 65.

    Motamed N, Khoonsari M, Panahi M, Rezaie N, Maadi M, Safarnezhad Tameshkel F, et al. The incidence and risk factors of non-alcoholic fatty liver disease: A cohort study from Iran. Hepatitis Monthly. 2020;20(2). https://doi.org/10.5812/hepatmon.98531.

  • 66.

    Najafi M, Rafiei A, Ghaemi A, Hosseini V. Association between rs738408, rs738409 and rs139051polymorphisms in PNPLA3 gene and non-alcoholic fatty liver disease. Gene Reports. 2022;26. https://doi.org/10.1016/j.genrep.2021.101472.

  • 67.

    Ostovaneh MR, Zamani F, Ansari-Moghaddam A, Sharafkhah M, Saeedian FS, Rohani Z, et al. Nonalcoholic Fatty Liver: The Association with Metabolic Abnormalities, Body Mass Index and Central Obesity--A Population-Based Study. Metab Syndr Relat Disord. 2015;13(7):304-11. [PubMed ID: 26042518]. https://doi.org/10.1089/met.2014.0131.

  • 68.

    Pasdar Y, Rahimi F, Gharib Salehi M, Darbandi M, Moradi Nazar M, Niazi P, et al. The relationship between anthropometric indexes and blood biomarkers with fatty liver-a case-control study; Kermanshah, Iran. Int J Pharm Technol. 2016;8(2):12837-46.

  • 69.

    Pasdar Y, Darbandi M, Niazi P, Bagheri A, Mohajeri SAR, Norouzy A, et al. The risk factors of metabolic syndrome and nutritional status in patients with non-alcoholic fatty liver disease: A case-control study in Kermanshah, Iran. Acta Medica Mediterranea. 2017;33:715.

  • 70.

    Pasdar Y, Moradi S, Moludi J, Darbandi M, Niazi P, Nachvak SM, et al. Risk of metabolic syndrome in non-alcoholic fatty liver disease patients. Med J Nutrition Metab. 2019;12(4):353-63. https://doi.org/10.3233/mnm-190290.

  • 71.

    Radmard AR, Rahmanian MS, Abrishami A, Yoonessi A, Kooraki S, Dadgostar M, et al. Assessment of abdominal fat distribution in non-alcoholic fatty liver disease by magnetic resonance imaging: A population-based Study. Arch Iran Med. 2016;19(10):693-9. [PubMed ID: 27743433].

  • 72.

    Rezapour S, Khosroshahi SA, Farajnia H, Mohseni F, Khoshbaten M, Farajnia S. Association of 45-bp ins/del polymorphism of uncoupling protein 2 (UCP2) and susceptibility to nonalcoholic fatty liver and type 2 diabetes mellitus in North-west of Iran. BMC Res Notes. 2021;14(1):169. [PubMed ID: 33957975]. [PubMed Central ID: PMC8101211]. https://doi.org/10.1186/s13104-021-05586-9.

  • 73.

    Salehi-Sahlabadi A, Sadat S, Beigrezaei S, Pourmasomi M, Feizi A, Ghiasvand R, et al. Dietary patterns and risk of non-alcoholic fatty liver disease. BMC Gastroenterol. 2021;21(1):41. [PubMed ID: 33509112]. [PubMed Central ID: PMC7844966]. https://doi.org/10.1186/s12876-021-01612-z.

  • 74.

    Savadkouhi F, HosseiniTabatabaei MT, Nezhad SS. [The frequency of fatty liver in sonography of patients without liver diseases background and its correlation with blood cholesterol and triglyceride]. Zahedan Journal of Research in Medical Sciences. 2003;5(3). Persian.

  • 75.

    Shanaki M, Fadaei R, Moradi N, Emamgholipour S, Poustchi H. The Circulating CTRP13 in Type 2 Diabetes and Non-Alcoholic Fatty Liver Patients. PLoS One. 2016;11(12). e0168082. [PubMed ID: 27936230]. [PubMed Central ID: PMC5148106]. https://doi.org/10.1371/journal.pone.0168082.

  • 76.

    Sohouli MH, Fatahi S, Sayyari A, Olang B, Shidfar F. Associations between dietary total antioxidant capacity and odds of non-alcoholic fatty liver disease (NAFLD) in adults: a case-control study. J Nutr Sci. 2020;9. e48. [PubMed ID: 33244400]. [PubMed Central ID: PMC7681134]. https://doi.org/10.1017/jns.2020.39.

  • 77.

    Sohouli MH, Sayyari AA, Lari A, Nameni G, Lotfi M, Fatahi S, et al. Association of dietary insulinaemic potential and odds of non-alcoholic fatty liver disease among adults: A case-control study. J Hum Nutr Diet. 2021;34(5):901-9. [PubMed ID: 33586811]. https://doi.org/10.1111/jhn.12865.

  • 78.

    Taheri E, Pourhoseingholi MA, Moslem A, Hassani AH, Mousavi Jarrahi A, Asadzadeh Aghdaei H, et al. The triglyceride-glucose index as a clinical useful marker for metabolic associated fatty liver disease (MAFLD): A population-based study among Iranian adults. J Diabetes Metab Disord. 2022;21(1):97-107. [PubMed ID: 35673435]. [PubMed Central ID: PMC9167320]. https://doi.org/10.1007/s40200-021-00941-w.

  • 79.

    Tutunchi H, Saghafi-Asl M, Ebrahimi-Mameghani M, Ostadrahimi A. Food insecurity and lipid profile abnormalities are associated with an increased risk of nonalcoholic fatty liver disease (NAFLD): A case-control study. Ecol Food Nutr. 2021;60(4):508-24. [PubMed ID: 33573415]. https://doi.org/10.1080/03670244.2021.1875453.

  • 80.

    Tutunchi H, Saghafi-Asl M, Asghari-Jafarabadi M, Ostadrahimi A. Association between dietary patterns and non-alcoholic fatty liver disease: Results from a Case-Control Study. Arch Iran Med. 2021;24(1):35-42. [PubMed ID: 33588566]. https://doi.org/10.34172/aim.2021.06.

  • 81.

    Tutunchi H, Mobasseri M, Aghamohammadzadeh N, Hooshyar J, Naeini F, Najafipour F. Serum neuregulin 4 (NRG-4) level and non-alcoholic fatty liver disease (NAFLD): A case-control study. Int J Clin Pract. 2021;75(10). e14555. [PubMed ID: 34159710]. https://doi.org/10.1111/ijcp.14555.

  • 82.

    Vahid F, Hekmatdoost A, Mirmajidi S, Doaei S, Rahmani D, Faghfoori Z. Association between index of nutritional quality and nonalcoholic fatty liver disease: The role of vitamin D and B group. Am J Med Sci. 2019;358(3):212-8. [PubMed ID: 31326093]. https://doi.org/10.1016/j.amjms.2019.06.008.

  • 83.

    Zarean E, Goujani R, Rahimian G, Ahamdi A. Prevalence and risk factors of non-alcoholic fatty liver disease in southwest Iran: A population-based case-control study. Clin Exp Hepatol. 2019;5(3):224-31. [PubMed ID: 31598559]. [PubMed Central ID: PMC6781826]. https://doi.org/10.5114/ceh.2019.87635.

  • 84.

    Zolfaghari H, Askari G, Siassi F, Feizi A, Sotoudeh G. Intake of nutrients, fiber, and sugar in patients with nonalcoholic fatty liver disease in comparison to healthy individuals. Int J Prev Med. 2016;7:98. [PubMed ID: 27625763]. [PubMed Central ID: PMC4995850]. https://doi.org/10.4103/2008-7802.188083.

  • 85.

    Salehi-Sahlabadi A, Teymoori F, Jabbari M, Momeni A, Mokari-Yamchi A, Sohouli M, et al. Dietary polyphenols and the odds of non-alcoholic fatty liver disease: A case-control study. Clin Nutr ESPEN. 2021;41:429-35. [PubMed ID: 33487302]. https://doi.org/10.1016/j.clnesp.2020.09.028.

  • 86.

    Adams LA, Angulo P, Lindor KD. Nonalcoholic fatty liver disease. CMAJ. 2005;172(7):899-905. [PubMed ID: 15795412]. [PubMed Central ID: PMC554876]. https://doi.org/10.1503/cmaj.045232.

  • 87.

    Clark JM, Brancati FL, Diehl AM. Nonalcoholic fatty liver disease. Gastroenterology. 2002;122(6):1649-57. [PubMed ID: 12016429]. https://doi.org/10.1053/gast.2002.33573.

  • 88.

    Bagheri Lankarani K, Ghaffarpasand F, Mahmoodi M, Lotfi M, Zamiria N, Heydari ST, et al. Non alcoholic fatty liver disease in Southern Iran: A population based study. Hepatitis Monthly. 2013;13(5). https://doi.org/10.5812/hepatmon.9248.

  • 89.

    Okanoue T, Umemura A, Yasui K, Itoh Y. Nonalcoholic fatty liver disease and nonalcoholic steatohepatitis in Japan. J Gastroenterol Hepatol. 2011;26 Suppl 1:153-62. [PubMed ID: 21199527]. https://doi.org/10.1111/j.1440-1746.2010.06547.x.

  • 90.

    Angulo P. Nonalcoholic fatty liver disease. N Engl J Med. 2002;346(16):1221-31. [PubMed ID: 11961152]. https://doi.org/10.1056/NEJMra011775.

  • 91.

    Argo CK, Caldwell SH. Epidemiology and natural history of non-alcoholic steatohepatitis. Clin Liver Dis. 2009;13(4):511-31. [PubMed ID: 19818302]. https://doi.org/10.1016/j.cld.2009.07.005.

  • 92.

    Arshad T, Paik JM, Biswas R, Alqahtani SA, Henry L, Younossi ZM. Nonalcoholic Fatty Liver Disease Prevalence Trends Among Adolescents and Young Adults in the United States, 2007-2016. Hepatol Commun. 2021;5(10):1676-88. [PubMed ID: 34558817]. [PubMed Central ID: PMC8485885]. https://doi.org/10.1002/hep4.1760.

  • 93.

    Koehler EM, Plompen EP, Schouten JN, Hansen BE, Darwish Murad S, Taimr P, et al. Presence of diabetes mellitus and steatosis is associated with liver stiffness in a general population: The Rotterdam study. Hepatology. 2016;63(1):138-47. [PubMed ID: 26171685]. https://doi.org/10.1002/hep.27981.

  • 94.

    Noureddin M, Yates KP, Vaughn IA, Neuschwander-Tetri BA, Sanyal AJ, McCullough A, et al. Clinical and histological determinants of nonalcoholic steatohepatitis and advanced fibrosis in elderly patients. Hepatology. 2013;58(5):1644-54. [PubMed ID: 23686698]. [PubMed Central ID: PMC3760979]. https://doi.org/10.1002/hep.26465.

  • 95.

    Kim IH, Kisseleva T, Brenner DA. Aging and liver disease. Curr Opin Gastroenterol. 2015;31(3):184-91. [PubMed ID: 25850346]. [PubMed Central ID: PMC4736713]. https://doi.org/10.1097/MOG.0000000000000176.

  • 96.

    Marchisello S, Di Pino A, Scicali R, Urbano F, Piro S, Purrello F, et al. Pathophysiological, Molecular and Therapeutic Issues of Nonalcoholic Fatty Liver Disease: An Overview. Int J Mol Sci. 2019;20(8). [PubMed ID: 31010049]. [PubMed Central ID: PMC6514656]. https://doi.org/10.3390/ijms20081948.

  • 97.

    Chen Z, Tian R, She Z, Cai J, Li H. Role of oxidative stress in the pathogenesis of nonalcoholic fatty liver disease. Free Radic Biol Med. 2020;152:116-41. [PubMed ID: 32156524]. https://doi.org/10.1016/j.freeradbiomed.2020.02.025.

  • 98.

    Kuk JL, Saunders TJ, Davidson LE, Ross R. Age-related changes in total and regional fat distribution. Ageing Res Rev. 2009;8(4):339-48. [PubMed ID: 19576300]. https://doi.org/10.1016/j.arr.2009.06.001.

  • 99.

    Tran TT, Yamamoto Y, Gesta S, Kahn CR. Beneficial effects of subcutaneous fat transplantation on metabolism. Cell Metab. 2008;7(5):410-20. [PubMed ID: 18460332]. [PubMed Central ID: PMC3204870]. https://doi.org/10.1016/j.cmet.2008.04.004.

  • 100.

    Petta S, Amato M, Cabibi D, Camma C, Di Marco V, Giordano C, et al. Visceral adiposity index is associated with histological findings and high viral load in patients with chronic hepatitis C due to genotype 1. Hepatology. 2010;52(5):1543-52. [PubMed ID: 20799355]. https://doi.org/10.1002/hep.23859.

  • 101.

    Non-alcoholic Fatty Liver Disease Study G, Lonardo A, Bellentani S, Argo CK, Ballestri S, Byrne CD, et al. Epidemiological modifiers of non-alcoholic fatty liver disease: Focus on high-risk groups. Dig Liver Dis. 2015;47(12):997-1006. [PubMed ID: 26454786]. https://doi.org/10.1016/j.dld.2015.08.004.

  • 102.

    Lu G, Shimizu I, Cui X, Itonaga M, Tamaki K, Fukuno H, et al. Antioxidant and antiapoptotic activities of idoxifene and estradiol in hepatic fibrosis in rats. Life Sci. 2004;74(7):897-907. [PubMed ID: 14659978]. https://doi.org/10.1016/j.lfs.2003.08.004.

  • 103.

    Yasuda M, Shimizu I, Shiba M, Ito S. Suppressive effects of estradiol on dimethylnitrosamine-induced fibrosis of the liver in rats. Hepatology. 1999;29(3):719-27. [PubMed ID: 10051473]. https://doi.org/10.1002/hep.510290307.

  • 104.

    Ballestri S, Nascimbeni F, Baldelli E, Marrazzo A, Romagnoli D, Lonardo A. NAFLD as a Sexual dimorphic disease: Role of Gender and reproductive status in the development and progression of nonalcoholic fatty liver disease and inherent cardiovascular risk. Adv Ther. 2017;34(6):1291-326. [PubMed ID: 28526997]. [PubMed Central ID: PMC5487879]. https://doi.org/10.1007/s12325-017-0556-1.

  • 105.

    Despres JP. Body fat distribution and risk of cardiovascular disease: An update. Circulation. 2012;126(10):1301-13. [PubMed ID: 22949540]. https://doi.org/10.1161/CIRCULATIONAHA.111.067264.

  • 106.

    Varlamov O, Bethea CL, Roberts CT. Sex-specific differences in lipid and glucose metabolism. Front Endocrinol (Lausanne). 2014;5:241. [PubMed ID: 25646091]. [PubMed Central ID: PMC4298229]. https://doi.org/10.3389/fendo.2014.00241.

  • 107.

    Tiniakos DG, Vos MB, Brunt EM. Nonalcoholic fatty liver disease: Pathology and pathogenesis. Annu Rev Pathol. 2010;5:145-71. [PubMed ID: 20078219]. https://doi.org/10.1146/annurev-pathol-121808-102132.

  • 108.

    Van der Poorten D, Milner KL, Hui J, Hodge A, Trenell MI, Kench JG, et al. Visceral fat: A key mediator of steatohepatitis in metabolic liver disease. Hepatology. 2008;48(2):449-57. [PubMed ID: 18627003]. https://doi.org/10.1002/hep.22350.

  • 109.

    Fried SK, Lee MJ, Karastergiou K. Shaping fat distribution: New insights into the molecular determinants of depot- and sex-dependent adipose biology. Obesity (Silver Spring). 2015;23(7):1345-52. [PubMed ID: 26054752]. [PubMed Central ID: PMC4687449]. https://doi.org/10.1002/oby.21133.

  • 110.

    Leibel RL, Edens NK, Fried SK. Physiologic basis for the control of body fat distribution in humans. Annu Rev Nutr. 1989;9:417-43. [PubMed ID: 2669880]. https://doi.org/10.1146/annurev.nu.09.070189.002221.

  • 111.

    Lonardo A, Nascimbeni F, Ballestri S, Fairweather D, Win S, Than TA, et al. Sex Differences in Nonalcoholic Fatty Liver Disease: State of the Art and Identification of Research Gaps. Hepatology. 2019;70(4):1457-69. [PubMed ID: 30924946]. [PubMed Central ID: PMC6766425]. https://doi.org/10.1002/hep.30626.

  • 112.

    Pedersen SB, Kristensen K, Hermann PA, Katzenellenbogen JA, Richelsen B. Estrogen controls lipolysis by up-regulating alpha2A-adrenergic receptors directly in human adipose tissue through the estrogen receptor alpha. Implications for the female fat distribution. J Clin Endocrinol Metab. 2004;89(4):1869-78. [PubMed ID: 15070958]. https://doi.org/10.1210/jc.2003-031327.

  • 113.

    Park YM, Pereira RI, Erickson CB, Swibas TA, Cox-York KA, Van Pelt RE. Estradiol-mediated improvements in adipose tissue insulin sensitivity are related to the balance of adipose tissue estrogen receptor alpha and beta in postmenopausal women. PLoS One. 2017;12(5). e0176446. [PubMed ID: 28472101]. [PubMed Central ID: PMC5417515]. https://doi.org/10.1371/journal.pone.0176446.

  • 114.

    Utzschneider KM, Kahn SE. Review: The role of insulin resistance in nonalcoholic fatty liver disease. J Clin Endocrinol Metab. 2006;91(12):4753-61. [PubMed ID: 16968800]. https://doi.org/10.1210/jc.2006-0587.

  • 115.

    Hoeg LD, Sjoberg KA, Lundsgaard AM, Jordy AB, Hiscock N, Wojtaszewski JF, et al. Adiponectin concentration is associated with muscle insulin sensitivity, AMPK phosphorylation, and ceramide content in skeletal muscles of men but not women. J Appl Physiol (1985). 2013;114(5):592-601. [PubMed ID: 23305978]. https://doi.org/10.1152/japplphysiol.01046.2012.

  • 116.

    Cho GJ, Yoo HJ, Hwang SY, Choi J, Lee KM, Choi KM, et al. Differential relationship between waist circumference and mortality according to age, sex, and body mass index in Korean with age of 30-90 years; a nationwide health insurance database study. BMC Med. 2018;16(1):131. [PubMed ID: 30092838]. [PubMed Central ID: PMC6085614]. https://doi.org/10.1186/s12916-018-1114-7.

  • 117.

    Sonmez HE, Canpolat N, Agbas A, Tasdemir M, Ekmekci OB, Alikasifoglu M, et al. The relationship between the waist circumference and increased carotid intima thickness in obese children. Child Obes. 2019;15(7):468-75. [PubMed ID: 31246513]. https://doi.org/10.1089/chi.2019.0022.

  • 118.

    Goh LG, Dhaliwal SS, Welborn TA, Lee AH, Della PR. Anthropometric measurements of general and central obesity and the prediction of cardiovascular disease risk in women: A cross-sectional study. BMJ Open. 2014;4(2). e004138. [PubMed ID: 24503301]. [PubMed Central ID: PMC3918987]. https://doi.org/10.1136/bmjopen-2013-004138.

  • 119.

    Naghipour M, Joukar F, Amini Salehi E, Hassanipour S, Mansour Ghanaei F. [The association between age at first pregnancy and number of deliveries with metabolic syndrome and its components: Results from Persian Guilan Cohort Study (PGCS)]. Iran J Obstet Gynecol Infertil. 2022;25(6):1-11. Persian. https://doi.org/10.22038/ijogi.2022.21046.

  • 120.

    Greco D, Kotronen A, Westerbacka J, Puig O, Arkkila P, Kiviluoto T, et al. Gene expression in human NAFLD. Am J Physiol Gastrointest Liver Physiol. 2008;294(5):G1281-7. [PubMed ID: 18388185]. https://doi.org/10.1152/ajpgi.00074.2008.

  • 121.

    Fabbrini E, Magkos F, Mohammed BS, Pietka T, Abumrad NA, Patterson BW, et al. Intrahepatic fat, not visceral fat, is linked with metabolic complications of obesity. Proc Natl Acad Sci U S A. 2009;106(36):15430-5. [PubMed ID: 19706383]. [PubMed Central ID: PMC2741268]. https://doi.org/10.1073/pnas.0904944106.

  • 122.

    Mittendorfer B, Magkos F, Fabbrini E, Mohammed BS, Klein S. Relationship between body fat mass and free fatty acid kinetics in men and women. Obesity (Silver Spring). 2009;17(10):1872-7. [PubMed ID: 19629053]. [PubMed Central ID: PMC3319738]. https://doi.org/10.1038/oby.2009.224.

  • 123.

    Pardina E, Baena-Fustegueras JA, Catalan R, Galard R, Lecube A, Fort JM, et al. Increased expression and activity of hepatic lipase in the liver of morbidly obese adult patients in relation to lipid content. Obes Surg. 2009;19(7):894-904. [PubMed ID: 18972174]. https://doi.org/10.1007/s11695-008-9739-9.

  • 124.

    Westerbacka J, Kolak M, Kiviluoto T, Arkkila P, Siren J, Hamsten A, et al. Genes involved in fatty acid partitioning and binding, lipolysis, monocyte/macrophage recruitment, and inflammation are overexpressed in the human fatty liver of insulin-resistant subjects. Diabetes. 2007;56(11):2759-65. [PubMed ID: 17704301]. https://doi.org/10.2337/db07-0156.

  • 125.

    Mitsuyoshi H, Yasui K, Harano Y, Endo M, Tsuji K, Minami M, et al. Analysis of hepatic genes involved in the metabolism of fatty acids and iron in nonalcoholic fatty liver disease. Hepatol Res. 2009;39(4):366-73. [PubMed ID: 19054139]. https://doi.org/10.1111/j.1872-034X.2008.00464.x.

  • 126.

    Kohjima M, Enjoji M, Higuchi N, Kato M, Kotoh K, Yoshimoto T, et al. Re-evaluation of fatty acid metabolism-related gene expression in nonalcoholic fatty liver disease. Int J Mol Med. 2007;20(3):351-8. [PubMed ID: 17671740].

  • 127.

    Deprince A, Haas JT, Staels B. Dysregulated lipid metabolism links NAFLD to cardiovascular disease. Mol Metab. 2020;42:101092. [PubMed ID: 33010471]. [PubMed Central ID: PMC7600388]. https://doi.org/10.1016/j.molmet.2020.101092.

  • 128.

    Mittendorfer B, Yoshino M, Patterson BW, Klein S. VLDL triglyceride kinetics in lean, overweight, and obese men and women. J Clin Endocrinol Metab. 2016;101(11):4151-60. [PubMed ID: 27588438]. [PubMed Central ID: PMC5095238]. https://doi.org/10.1210/jc.2016-1500.

  • 129.

    Fabbrini E, Mohammed BS, Magkos F, Korenblat KM, Patterson BW, Klein S. Alterations in adipose tissue and hepatic lipid kinetics in obese men and women with nonalcoholic fatty liver disease. Gastroenterology. 2008;134(2):424-31. [PubMed ID: 18242210]. [PubMed Central ID: PMC2705923]. https://doi.org/10.1053/j.gastro.2007.11.038.

  • 130.

    Manco M. Metabolic syndrome in childhood from impaired carbohydrate metabolism to nonalcoholic fatty liver disease. J Am Coll Nutr. 2011;30(5):295-303. [PubMed ID: 22081615]. https://doi.org/10.1080/07315724.2011.10719972.

  • 131.

    Ormazabal V, Nair S, Elfeky O, Aguayo C, Salomon C, Zuniga FA. Association between insulin resistance and the development of cardiovascular disease. Cardiovasc Diabetol. 2018;17(1):122. [PubMed ID: 30170598]. [PubMed Central ID: PMC6119242]. https://doi.org/10.1186/s12933-018-0762-4.

  • 132.

    Lebeau PF, Byun JH, Platko K, MacDonald ME, Poon SV, Faiyaz M, et al. Diet-induced hepatic steatosis abrogates cell-surface LDLR by inducing de novo PCSK9 expression in mice. J Biol Chem. 2019;294(23):9037-47. [PubMed ID: 31004037]. [PubMed Central ID: PMC6556582]. https://doi.org/10.1074/jbc.RA119.008094.

  • 133.

    Ruscica M, Ferri N, Macchi C, Meroni M, Lanti C, Ricci C, et al. Liver fat accumulation is associated with circulating PCSK9. Ann Med. 2016;48(5):384-91. [PubMed ID: 27222915]. https://doi.org/10.1080/07853890.2016.1188328.

  • 134.

    Sniderman A, Couture P, de Graaf J. Diagnosis and treatment of apolipoprotein B dyslipoproteinemias. Nat Rev Endocrinol. 2010;6(6):335-46. [PubMed ID: 20421882]. https://doi.org/10.1038/nrendo.2010.50.

  • 135.

    DeFilippis AP, Blaha MJ, Martin SS, Reed RM, Jones SR, Nasir K, et al. Nonalcoholic fatty liver disease and serum lipoproteins: The Multi-Ethnic Study of Atherosclerosis. Atherosclerosis. 2013;227(2):429-36. [PubMed ID: 23419204]. [PubMed Central ID: PMC4049078]. https://doi.org/10.1016/j.atherosclerosis.2013.01.022.

  • 136.

    Sarkar S, Lipworth L, Kabagambe EK, Bian A, Stewart TG, Blot WJ, et al. A Description of Risk Factors for Non-alcoholic Fatty Liver Disease in the Southern Community Cohort Study: A Nested Case-Control Study. Front Nutr. 2020;7:71. [PubMed ID: 32671089]. [PubMed Central ID: PMC7326146]. https://doi.org/10.3389/fnut.2020.00071.

  • 137.

    Min HK, Kapoor A, Fuchs M, Mirshahi F, Zhou H, Maher J, et al. Increased hepatic synthesis and dysregulation of cholesterol metabolism is associated with the severity of nonalcoholic fatty liver disease. Cell Metab. 2012;15(5):665-74. [PubMed ID: 22560219]. [PubMed Central ID: PMC3361911]. https://doi.org/10.1016/j.cmet.2012.04.004.

  • 138.

    Caballero F, Fernandez A, De Lacy AM, Fernandez-Checa JC, Caballeria J, Garcia-Ruiz C. Enhanced free cholesterol, SREBP-2 and StAR expression in human NASH. J Hepatol. 2009;50(4):789-96. [PubMed ID: 19231010]. https://doi.org/10.1016/j.jhep.2008.12.016.

  • 139.

    Arguello G, Balboa E, Arrese M, Zanlungo S. Recent insights on the role of cholesterol in non-alcoholic fatty liver disease. Biochim Biophys Acta. 2015;1852(9):1765-78. [PubMed ID: 26027904]. https://doi.org/10.1016/j.bbadis.2015.05.015.

  • 140.

    Mari M, Caballero F, Colell A, Morales A, Caballeria J, Fernandez A, et al. Mitochondrial free cholesterol loading sensitizes to TNF- and Fas-mediated steatohepatitis. Cell Metab. 2006;4(3):185-98. [PubMed ID: 16950136]. https://doi.org/10.1016/j.cmet.2006.07.006.

  • 141.

    Savard C, Tartaglione EV, Kuver R, Haigh WG, Farrell GC, Subramanian S, et al. Synergistic interaction of dietary cholesterol and dietary fat in inducing experimental steatohepatitis. Hepatology. 2013;57(1):81-92. [PubMed ID: 22508243]. [PubMed Central ID: PMC5341743]. https://doi.org/10.1002/hep.25789.

  • 142.

    Van Rooyen DM, Larter CZ, Haigh WG, Yeh MM, Ioannou G, Kuver R, et al. Hepatic free cholesterol accumulates in obese, diabetic mice and causes nonalcoholic steatohepatitis. Gastroenterology. 2011;141(4):1393-403. 1403 e1-5. [PubMed ID: 21703998]. [PubMed Central ID: PMC3186822]. https://doi.org/10.1053/j.gastro.2011.06.040.

  • 143.

    Wouters K, van Bilsen M, van Gorp PJ, Bieghs V, Lutjohann D, Kerksiek A, et al. Intrahepatic cholesterol influences progression, inhibition and reversal of non-alcoholic steatohepatitis in hyperlipidemic mice. FEBS Lett. 2010;584(5):1001-5. [PubMed ID: 20114046]. https://doi.org/10.1016/j.febslet.2010.01.046.

  • 144.

    Zhao L, Chen Y, Tang R, Chen Y, Li Q, Gong J, et al. Inflammatory stress exacerbates hepatic cholesterol accumulation via increasing cholesterol uptake and de novo synthesis. J Gastroenterol Hepatol. 2011;26(5):875-83. [PubMed ID: 21488946]. https://doi.org/10.1111/j.1440-1746.2010.06560.x.

  • 145.

    Turley S. Dietschy (1986) The metabolism and excretion of cholesterol by the liver. The liver: biology and pathobiology. 2nd ed. New York: Raven; 1386.

  • 146.

    Cohen DE. Lipoprotein metabolism and cholesterol balance. The Liver: Biology and Pathobiology. 2009:271-85.

  • 147.

    Beg ZH, Stonik JA, Brewer HJ. Phosphorylation of hepatic 3-hydroxy-3-methylglutaryl coenzyme A reductase and modulation of its enzymic activity by calcium-activated and phospholipid-dependent protein kinase. J Biol Chem. 1985;260(3):1682-7. [PubMed ID: 3155737].

  • 148.

    Whelton PK, Carey RM, Aronow WS, Casey DJ, Collins KJ, Dennison Himmelfarb C, et al. 2017 ACC/AHA/AAPA/ABC/ACPM/AGS/APhA/ASH/ASPC/NMA/PCNA guideline for the prevention, detection, evaluation, and management of high blood pressure in adults: A report of the American College of Cardiology/American Heart Association Task Force on Clinical Practice Guidelines. J Am Coll Cardiol. 2018;71(19):e127-248. [PubMed ID: 29146535]. https://doi.org/10.1016/j.jacc.2017.11.006.

  • 149.

    Zhao YC, Zhao GJ, Chen Z, She ZG, Cai J, Li H. Nonalcoholic Fatty Liver Disease: An Emerging Driver of Hypertension. Hypertension. 2020;75(2):275-84. [PubMed ID: 31865799]. https://doi.org/10.1161/HYPERTENSIONAHA.119.13419.

  • 150.

    Carnagarin R, Matthews V, Zaldivia MTK, Peter K, Schlaich MP. The bidirectional interaction between the sympathetic nervous system and immune mechanisms in the pathogenesis of hypertension. Br J Pharmacol. 2019;176(12):1839-52. [PubMed ID: 30129037]. [PubMed Central ID: PMC6534787]. https://doi.org/10.1111/bph.14481.

  • 151.

    Haukeland JW, Damas JK, Konopski Z, Loberg EM, Haaland T, Goverud I, et al. Systemic inflammation in nonalcoholic fatty liver disease is characterized by elevated levels of CCL2. J Hepatol. 2006;44(6):1167-74. [PubMed ID: 16618517]. https://doi.org/10.1016/j.jhep.2006.02.011.

  • 152.

    Ferrara D, Montecucco F, Dallegri F, Carbone F. Impact of different ectopic fat depots on cardiovascular and metabolic diseases. J Cell Physiol. 2019;234(12):21630-41. [PubMed ID: 31106419]. https://doi.org/10.1002/jcp.28821.

  • 153.

    Artunc F, Schleicher E, Weigert C, Fritsche A, Stefan N, Haring HU. The impact of insulin resistance on the kidney and vasculature. Nat Rev Nephrol. 2016;12(12):721-37. [PubMed ID: 27748389]. https://doi.org/10.1038/nrneph.2016.145.

  • 154.

    Fountain JH, Lappin SL. Physiology, renin angiotensin system. Treasure Island: StatPearls; 2017.

  • 155.

    Li N, Zhang GW, Zhang JR, Jin D, Li Y, Liu T, et al. Non-alcoholic fatty liver disease is associated with progression of arterial stiffness. Nutr Metab Cardiovasc Dis. 2015;25(2):218-23. [PubMed ID: 25456154]. https://doi.org/10.1016/j.numecd.2014.10.002.

  • 156.

    Mofrad P, Contos MJ, Haque M, Sargeant C, Fisher RA, Luketic VA, et al. Clinical and histologic spectrum of nonalcoholic fatty liver disease associated with normal ALT values. Hepatology. 2003;37(6):1286-92. [PubMed ID: 12774006]. https://doi.org/10.1053/jhep.2003.50229.

  • 157.

    Browning JD, Szczepaniak LS, Dobbins R, Nuremberg P, Horton JD, Cohen JC, et al. Prevalence of hepatic steatosis in an urban population in the United States: Impact of ethnicity. Hepatology. 2004;40(6):1387-95. [PubMed ID: 15565570]. https://doi.org/10.1002/hep.20466.

  • 158.

    Bugianesi E, Manzini P, D'Antico S, Vanni E, Longo F, Leone N, et al. Relative contribution of iron burden, HFE mutations, and insulin resistance to fibrosis in nonalcoholic fatty liver. Hepatology. 2004;39(1):179-87. [PubMed ID: 14752836]. https://doi.org/10.1002/hep.20023.

  • 159.

    Cobbina E, Akhlaghi F. Non-alcoholic fatty liver disease (NAFLD-(pathogenesis, classification, and effect on drug metabolizing enzymes and transporters. Drug Metab Rev. 2017;49(2):197-211. [PubMed ID: 28303724]. [PubMed Central ID: PMC5576152]. https://doi.org/10.1080/03602532.2017.1293683.

  • 160.

    Schindhelm RK, Diamant M, Dekker JM, Tushuizen ME, Teerlink T, Heine RJ. Alanine aminotransferase as a marker of non-alcoholic fatty liver disease in relation to type 2 diabetes mellitus and cardiovascular disease. Diabetes Metab Res Rev. 2006;22(6):437-43. [PubMed ID: 16832839]. https://doi.org/10.1002/dmrr.666.

  • 161.

    Peleg N, Issachar A, Sneh-Arbib O, Shlomai A. AST to Platelet Ratio Index and fibrosis 4 calculator scores for non-invasive assessment of hepatic fibrosis in patients with non-alcoholic fatty liver disease. Dig Liver Dis. 2017;49(10):1133-8. [PubMed ID: 28572039]. https://doi.org/10.1016/j.dld.2017.05.002.

  • 162.

    De Matteis C, Cariello M, Graziano G, Battaglia S, Suppressa P, Piazzolla G, et al. AST to Platelet Ratio Index (APRI) is an easy-to-use predictor score for cardiovascular risk in metabolic subjects. Sci Rep. 2021;11(1):14834. [PubMed ID: 34290320]. [PubMed Central ID: PMC8295377]. https://doi.org/10.1038/s41598-021-94277-3.

  • 163.

    Loaeza-del-Castillo A, Paz-Pineda F, Oviedo-Cardenas E, Sanchez-Avila F, Vargas-Vorackova F. AST to platelet ratio index (APRI) for the noninvasive evaluation of liver fibrosis. Ann Hepatol. 2008;7(4):350-7. [PubMed ID: 19034235].

  • 164.

    Chen ZW, Chen LY, Dai HL, Chen JH, Fang LZ. Relationship between alanine aminotransferase levels and metabolic syndrome in nonalcoholic fatty liver disease. J Zhejiang Univ Sci B. 2008;9(8):616-22. [PubMed ID: 18763311]. [PubMed Central ID: PMC2491691]. https://doi.org/10.1631/jzus.B0720016.

  • 165.

    Bugianesi E, McCullough AJ, Marchesini G. Insulin resistance: a metabolic pathway to chronic liver disease. Hepatology. 2005;42(5):987-1000. [PubMed ID: 16250043]. https://doi.org/10.1002/hep.20920.

  • 166.

    Targher G, Lonardo A, Byrne CD. Nonalcoholic fatty liver disease and chronic vascular complications of diabetes mellitus. Nat Rev Endocrinol. 2018;14(2):99-114. [PubMed ID: 29286050]. https://doi.org/10.1038/nrendo.2017.173.

  • 167.

    Tilg H, Moschen AR, Roden M. NAFLD and diabetes mellitus. Nat Rev Gastroenterol Hepatol. 2017;14(1):32-42. [PubMed ID: 27729660]. https://doi.org/10.1038/nrgastro.2016.147.

  • 168.

    Valenti L, Bugianesi E, Pajvani U, Targher G. Nonalcoholic fatty liver disease: cause or consequence of type 2 diabetes? Liver Int. 2016;36(11):1563-79. [PubMed ID: 27276701]. https://doi.org/10.1111/liv.13185.

  • 169.

    Targher G, Corey KE, Byrne CD, Roden M. The complex link between NAFLD and type 2 diabetes mellitus - mechanisms and treatments. Nat Rev Gastroenterol Hepatol. 2021;18(9):599-612. [PubMed ID: 33972770]. https://doi.org/10.1038/s41575-021-00448-y.

  • 170.

    Sung KC, Jeong WS, Wild SH, Byrne CD. Combined influence of insulin resistance, overweight/obesity, and fatty liver as risk factors for type 2 diabetes. Diabetes Care. 2012;35(4):717-22. [PubMed ID: 22338098]. [PubMed Central ID: PMC3308286]. https://doi.org/10.2337/dc11-1853.

  • 171.

    Sung KC, Wild SH, Byrne CD. Resolution of fatty liver and risk of incident diabetes. J Clin Endocrinol Metab. 2013;98(9):3637-43. [PubMed ID: 23873989]. https://doi.org/10.1210/jc.2013-1519.

  • 172.

    Yki-Jarvinen H. Ceramides: A Cause of Insulin Resistance in Nonalcoholic Fatty Liver Disease in Both Murine Models and Humans. Hepatology. 2020;71(4):1499-501. [PubMed ID: 31899812]. https://doi.org/10.1002/hep.31095.

  • 173.

    Misu H. Identification of hepatokines involved in pathology of type 2 diabetes and obesity. Endocr J. 2019;66(8):659-62. [PubMed ID: 31366824]. https://doi.org/10.1507/endocrj.EJ19-0255.

  • 174.

    Gao RY, Hsu BG, Wu DA, Hou JS, Chen MC. Serum Fibroblast Growth Factor 21 levels are positively associated with metabolic syndrome in patients with type 2 diabetes. Int J Endocrinol. 2019;2019:5163245. [PubMed ID: 31582974]. [PubMed Central ID: PMC6754922]. https://doi.org/10.1155/2019/5163245.

  • 175.

    Ix JH, Wassel CL, Kanaya AM, Vittinghoff E, Johnson KC, Koster A, et al. Fetuin-A and incident diabetes mellitus in older persons. JAMA. 2008;300(2):182-8. [PubMed ID: 18612115]. [PubMed Central ID: PMC2779582]. https://doi.org/10.1001/jama.300.2.182.

  • 176.

    Kralisch S, Hoffmann A, Lossner U, Kratzsch J, Bluher M, Stumvoll M, et al. Regulation of the novel adipokines/ hepatokines fetuin A and fetuin B in gestational diabetes mellitus. Metabolism. 2017;68:88-94. [PubMed ID: 28183456]. https://doi.org/10.1016/j.metabol.2016.11.017.

  • 177.

    Assy N, Kaita K, Mymin D, Levy C, Rosser B, Minuk G. Fatty infiltration of liver in hyperlipidemic patients. Dig Dis Sci. 2000;45(10):1929-34. [PubMed ID: 11117562]. https://doi.org/10.1023/a:1005661516165.

  • 178.

    Beymer C, Kowdley KV, Larson A, Edmonson P, Dellinger EP, Flum DR. Prevalence and predictors of asymptomatic liver disease in patients undergoing gastric bypass surgery. Arch Surg. 2003;138(11):1240-4. [PubMed ID: 14609874]. https://doi.org/10.1001/archsurg.138.11.1240.

  • 179.

    Leite NC, Salles GF, Araujo AL, Villela-Nogueira CA, Cardoso CR. Prevalence and associated factors of non-alcoholic fatty liver disease in patients with type-2 diabetes mellitus. Liver Int. 2009;29(1):113-9. [PubMed ID: 18384521]. https://doi.org/10.1111/j.1478-3231.2008.01718.x.

  • 180.

    Prashanth M, Ganesh HK, Vima MV, John M, Bandgar T, Joshi SR, et al. Prevalence of nonalcoholic fatty liver disease in patients with type 2 diabetes mellitus. J Assoc Physicians India. 2009;57:205-10. [PubMed ID: 19588648].

  • 181.

    Hirosumi J, Tuncman G, Chang L, Gorgun CZ, Uysal KT, Maeda K, et al. A central role for JNK in obesity and insulin resistance. Nature. 2002;420(6913):333-6. [PubMed ID: 12447443]. https://doi.org/10.1038/nature01137.

  • 182.

    Samala N, Tersey SA, Chalasani N, Anderson RM, Mirmira RG. Molecular mechanisms of nonalcoholic fatty liver disease: Potential role for 12-lipoxygenase. J Diabetes Complications. 2017;31(11):1630-7. [PubMed ID: 28886991]. [PubMed Central ID: PMC5643240]. https://doi.org/10.1016/j.jdiacomp.2017.07.014.

  • 183.

    Samuel VT, Shulman GI. Mechanisms for insulin resistance: Common threads and missing links. Cell. 2012;148(5):852-71. [PubMed ID: 22385956]. [PubMed Central ID: PMC3294420]. https://doi.org/10.1016/j.cell.2012.02.017.

  • 184.

    Brown MS, Goldstein JL. Selective versus total insulin resistance: A pathogenic paradox. Cell Metab. 2008;7(2):95-6. [PubMed ID: 18249166]. https://doi.org/10.1016/j.cmet.2007.12.009.

  • 185.

    Azzalini L, Ferrer E, Ramalho LN, Moreno M, Dominguez M, Colmenero J, et al. Cigarette smoking exacerbates nonalcoholic fatty liver disease in obese rats. Hepatology. 2010;51(5):1567-76. [PubMed ID: 20432253]. https://doi.org/10.1002/hep.23516.

  • 186.

    Okamoto M, Miyake T, Kitai K, Furukawa S, Yamamoto S, Senba H, et al. Cigarette smoking is a risk factor for the onset of fatty liver disease in nondrinkers: A longitudinal cohort study. PLoS One. 2018;13(4). e0195147. [PubMed ID: 29664906]. [PubMed Central ID: PMC5903610]. https://doi.org/10.1371/journal.pone.0195147.

  • 187.

    Liu Y, Dai M, Bi Y, Xu M, Xu Y, Li M, et al. Active smoking, passive smoking, and risk of nonalcoholic fatty liver disease (NAFLD): A population-based study in China. J Epidemiol. 2013;23(2):115-21. [PubMed ID: 23399520]. [PubMed Central ID: PMC3700247]. https://doi.org/10.2188/jea.je20120067.

  • 188.

    Chavez-Tapia NC, Lizardi-Cervera J, Perez-Bautista O, Ramos-Ostos MH, Uribe M. Smoking is not associated with nonalcoholic fatty liver disease. World J Gastroenterol. 2006;12(32):5196-200. [PubMed ID: 16937532]. [PubMed Central ID: PMC4088019]. https://doi.org/10.3748/wjg.v12.i32.5196.

  • 189.

    Attvall S, Fowelin J, Lager I, Von Schenck H, Smith U. Smoking induces insulin resistance--a potential link with the insulin resistance syndrome. J Intern Med. 1993;233(4):327-32. [PubMed ID: 8463765]. https://doi.org/10.1111/j.1365-2796.1993.tb00680.x.

  • 190.

    Janzon L, Berntorp K, Hanson M, Lindell SE, Trell E. Glucose tolerance and smoking: A population study of oral and intravenous glucose tolerance tests in middle-aged men. Diabetologia. 1983;25(2):86-8. [PubMed ID: 6354814]. https://doi.org/10.1007/BF00250893.

  • 191.

    Wannamethee SG, Shaper AG, Perry IJ, British Regional Heart S. Smoking as a modifiable risk factor for type 2 diabetes in middle-aged men. Diabetes Care. 2001;24(9):1590-5. [PubMed ID: 11522704]. https://doi.org/10.2337/diacare.24.9.1590.

  • 192.

    Houston TK, Person SD, Pletcher MJ, Liu K, Iribarren C, Kiefe CI. Active and passive smoking and development of glucose intolerance among young adults in a prospective cohort: CARDIA study. BMJ. 2006;332(7549):1064-9. [PubMed ID: 16603565]. [PubMed Central ID: PMC1458534]. https://doi.org/10.1136/bmj.38779.584028.55.

  • 193.

    Will JC, Galuska DA, Ford ES, Mokdad A, Calle EE. Cigarette smoking and diabetes mellitus: Evidence of a positive association from a large prospective cohort study. Int J Epidemiol. 2001;30(3):540-6. [PubMed ID: 11416080]. https://doi.org/10.1093/ije/30.3.540.

  • 194.

    Chiang PH, Chang TY, Chen JD. Synergistic effect of fatty liver and smoking on metabolic syndrome. World J Gastroenterol. 2009;15(42):5334-9. [PubMed ID: 19908343]. [PubMed Central ID: PMC2776862]. https://doi.org/10.3748/wjg.15.5334.

  • 195.

    Slagter SN, van Vliet-Ostaptchouk JV, Vonk JM, Boezen HM, Dullaart RP, Kobold AC, et al. Associations between smoking, components of metabolic syndrome and lipoprotein particle size. BMC Med. 2013;11:195. [PubMed ID: 24228807]. [PubMed Central ID: PMC3766075]. https://doi.org/10.1186/1741-7015-11-195.

  • 196.

    Agarwal R. Smoking, oxidative stress and inflammation: Impact on resting energy expenditure in diabetic nephropathy. BMC Nephrol. 2005;6:13. [PubMed ID: 16303055]. [PubMed Central ID: PMC1308817]. https://doi.org/10.1186/1471-2369-6-13.

  • 197.

    Malfertheiner P, Schutte K. Smoking--a trigger for chronic inflammation and cancer development in the pancreas. Am J Gastroenterol. 2006;101(1):160-2. [PubMed ID: 16405549]. https://doi.org/10.1111/j.1572-0241.2006.00402.x.

  • 198.

    El-Zayadi AR. Heavy smoking and liver. World J Gastroenterol. 2006;12(38):6098-101. [PubMed ID: 17036378]. [PubMed Central ID: PMC4088100]. https://doi.org/10.3748/wjg.v12.i38.6098.

  • 199.

    Bush T, Lovejoy JC, Deprey M, Carpenter KM. The effect of tobacco cessation on weight gain, obesity, and diabetes risk. Obesity (Silver Spring). 2016;24(9):1834-41. [PubMed ID: 27569117]. [PubMed Central ID: PMC5004778]. https://doi.org/10.1002/oby.21582.

  • 200.

    Walker JF, Collins LC, Rowell PP, Goldsmith LJ, Moffatt RJ, Stamford BA. The effect of smoking on energy expenditure and plasma catecholamine and nicotine levels during light physical activity. Nicotine Tob Res. 1999;1(4):365-70. [PubMed ID: 11072434]. https://doi.org/10.1080/14622299050011501.