Review Article | DOI: https://doi.org/IJCRI-RW-25-020
Non-Alcoholic Fatty Liver Disease in India: Mechanisms and Metabolic Signatures
Abstract
Metabolic-associated steatotic liver disease (MASLD), which was formerly referred to as non-alcoholic fatty liver disease (NAFLD), is increasingly recognized as a significant public health concern in India, impacting roughly 16%-32% (~120 million) of the population. Indians often develop NAFLD at lower body mass indices due to a greater accumulation of visceral fat and a heightened level of insulin resistance. While obesity serves as a key risk factor in many Western countries, the variability of the disease (9%-53%) is noteworthy and influenced by genetic, metabolic, and environmental factors. The rise in urbanization and dietary shifts has led to an increased incidence of the disease, affecting even individuals who are not overweight. The condition is characterized by lipid imbalance, oxidative stress, and dysfunction within the gut-liver axis, with involvement from genetic variants (PNPLA3, TM6SF2, MBOAT7) and epigenetic modifications. Diagnosing MASLD presents challenges, as most conventional methods (ultrasound, liver enzymes) lack accuracy, and non-invasive techniques (FLI, NFS, FibroScan) require validation specific to Indian populations. Consequently, other diagnostic methods like the Body Roundness Index (BRI) and microbiome-based assessments should be developed in India to address the rising prevalence of NAFLD. Advances in precision medicine, including genomics, metabolomics, and AI-enhanced risk evaluation, should facilitate earlier detection and treatment strategies for this condition. Therefore, lifestyle modifications must be incorporated into public health strategies to avert disease progression, while a comprehensive approach involving personalized medicine, national screening initiatives, and interdisciplinary research should be vital in managing NAFLDs within the Indian healthcare framework
Introduction
Non-alcoholic fatty liver disease (NAFLD), which is now referred to as metabolic-associated steatotic liver disease (MASLD), has become a silent epidemic that frequently advances undetected to more severe conditions such as cirrhosis and hepatocellular carcinoma (HCC) [1-3]. NAFLD happens when fat builds up in the liver (more than 5% of hepatocytes) even when no alcohol is consumed. It is now seen as a spectrum that ranges from simple steatosis to non-alcoholic steatohepatitis (NASH), with the possibility of progressing to fibrosis, cirrhosis, and liver failure [4-6]. This condition is increasingly affecting lower- and middle-income countries (LMICs), including India, where rapid urbanization, dietary changes, and related genetic factors are exacerbating what was once thought to be a disease associated with wealth [7-9].
Distinct metabolic phenotypes of NAFLD are observed in epidemiological studies conducted in India when compared to those in Western countries [10, 11]. While obesity is a significant contributor in the West, many Indians develop NAFLD at lower BMIs (body mass index) due to higher visceral fat accumulation and increased insulin resistance (IR) [12-14]. NAFLD is commonly found among Indians possessing metabolic risk factors; it has been reported in 55.5%-59.7% of diabetics, 64.6%-95% of overweight or obese individuals, and 73% of patients with metabolic syndrome (MS) [15]. Nonetheless, a community-based epidemiological investigation carried out in rural India found that the overall occurrence of NAFLD is 8.7%, with 164 out of 1911 participants being diagnosed. Interestingly, in individuals with a BMI < 23>
The underlying mechanisms of NAFLD are not yet completely understood, despite its rising prevalence in the general population. The accumulation of lipids in the liver, previously thought to be just a passive effect of metabolic defects, is now acknowledged as a significant factor in causing damage to liver cells through processes like lipotoxicity, oxidative stress, mitochondrial dysfunction (MD), and endoplasmic reticulum (ER) stress [19-22]. These stressors trigger inflammation, which is a critical element in disease progression, with hepatic macrophages (Kupffer cells) and stellate cells serving as primary players in the fibrotic remodeling process [23, 24]. Additionally, there is increasing evidence that the gut-liver axis contributes to the pathogenesis of NAFLD, as changes in bile acid metabolism and endotoxin signaling resulting from dysbiosis further worsen liver inflammation and IR [25-27].
Genetic and epigenetic components influence individuals' vulnerability to NAFLD, particularly within the South Asian demographic [28-31]. Variants in key genes such as PNPLA3 (patatin-like phospholipase domain-containing protein 3) (I148M), TM6SF2 (transmembrane 6 superfamily member 2) (E167K), and MBOAT7 (membrane-bound O-acyltransferase domain-containing 7) have been linked to the likelihood of hepatic fat buildup and fibrosis, with a greater occurrence of PNPLA3 variants among Indian populations, regardless of obesity, adding complexity to the progression of the disease [32-34]. Besides genetic predisposition, epigenetic changes like DNA methylation and histone acetylation play a significant role in the advancement of the disease, as they modify the expression of metabolic and inflammatory genes based on dietary and environmental factors [35-38].
This review thus explores the mechanistic aspects of NAFLD, with a specific emphasis on the molecular triggers, metabolic alterations, and genetic factors prevalent in the Indian population. By examining genomics, lipid metabolism, immune signaling, and gut-liver interactions, we aim to provide a comprehensive understanding of how NAFLD manifests in this demographic and how these findings might inform tailored therapeutic strategies. Gaining insight into additional mechanisms that extend beyond current treatments for NAFLD is essential for developing innovative diagnostics and therapies tailored to specific populations with distinctive metabolic profiles.
NAFLD in India: Diagnosis, Prevalence, Risk Factors, and Unique Metabolic Profiles
Diagnostic methods for NAFLD are increasingly recognized as a significant metabolic disease in India [39]. These methods need to be precise and readily accessible, taking diagnostics to a new level today. Currently, certain modalities may offer benefits or drawbacks compared to others. Ultrasound (US) is the most favored screening technique, as it is inexpensive and user-friendly; however, it lacks sensitivity for detecting mild steatosis and fibrosis [40, 41]. Likewise, liver enzyme levels, including alanine aminotransferase (ALT), aspartate aminotransferase (AST), and gamma-glutamyl transferase (GGT), serve merely as surrogate markers and may remain normal in many individuals, even when there is a substantial fat accumulation in the liver, which restricts their diagnostic utility [42, 43]. Given these considerations and based on these evaluations, several other non-invasive biomarker-based indices, such as the Fatty Liver Index (FLI), NAFLD Fibrosis Score (NFS), and Fibrosis-4 (FIB-4) index, can assist in determining disease severity and identifying individuals at risk for fibrogenesis [44, 45]. Imaging modalities such as transient elastography (FibroScan), MRI-proton density fat fraction (MRI-PDFF), and magnetic resonance elastography (MRE) offer advanced and more precise methods for assessing liver fat levels and fibrosis [46, 47]. However, their use is still limited due to issues with cost and accessibility. Emerging metabolite-based biomarkers, including 20-carboxy arachidonic acid (20-COOH AA) and 13,14-dihydro-15-keto-prostaglandin D2 (dhk PGD2), are showing significant diagnostic accuracy in detecting NAFLD [48, 49]. Furthermore, employing three-dimensional magnetic resonance elastography (3D-MRE), the FM-fibro index alongside liver stiffness measurement (FM-fibro LSM index), and evaluations driven by machine learning algorithms (MLA) enhance the differentiation of liver fibrosis stages with the advancements we have discussed [50, 51]. Nonetheless, large cohort trials must be conducted to establish their clinical relevance. While liver biopsy remains the definitive method for diagnosing non-alcoholic steatohepatitis (NASH) and assessing fibrosis, its invasive nature, high cost, and potential for sampling error limit its routine use [52]. Consequently, there is a growing preference for non-invasive imaging and biomarker strategies, particularly for early identification and risk assessment in high-risk populations, such as individuals with T2D and metabolic syndrome (MS). It is essential to establish standardized, population-specific screening guidelines to enhance the diagnosis of NAFLD and prevent disease progression in India [53, 54].
Regional Disparities and Urban-Rural Trends
The prevalence of NAFLD differs significantly across various regions of India, influenced by genetic factors, eating habits, and socioeconomic status (SES) [7, 55-57]. Higher rates of NAFLD are observed in urban areas, attributed to sedentary lifestyles and diets rich in calories as opposed to rural areas. However, in rural communities, the shift towards increased consumption of processed foods and reduced physical activity may result in a rapid rise in metabolic complications. The differences in NAFLD prevalence between urban and rural locations can be understood through variations in dietary practices, levels of physical activity, and environmental exposures [58, 59]. Urban residents tend to consume calorie-dense processed foods while working in sedentary settings. In contrast, rural residents may follow traditional diets, yet they may be missing out on essential nutrients and lack access to medical care. This may make them susceptible to factors that affect metabolism relating to NAFLD [60, 61]. Additionally, local environmental factors may contribute to a unique metabolic profile that could either increase risk or provide protection against NAFLD. Grasping these differences is essential for developing effective, targeted interventions addressing both lifestyle-related and epigenetically inherited metabolic risks to improve public health [62, 63].
A systematic review and meta-analysis on the prevalence of NAFLD in India indicated a wide range, varying from 9% to 53%, with significant regional differences. A study conducted in North India revealed that the urban prevalence of NAFLD in Delhi was 49.8%. In contrast, rural Ballabhgarh showed a lower prevalence rate of 28.1%, highlighting the impact of urbanization on metabolic health [7, 58]. The community-based cross-sectional study involved participants aged 30 to 60 years, excluding those with alcohol dependence (>140 g/week), a history of cirrhosis, HCC, or viral hepatitis, bedridden individuals, and pregnant women. Alarmingly high prevalence rates of NAFLD are also reported in southern India. Indeed, population-based studies in coastal South India have indicated an overall prevalence of 49.8%. Moreover, it was found that urban residents face an even greater risk of developing NAFLD, with an adjusted odds ratio (OR) of 1.21 (P = 0.048), regardless of gender, BMI, diabetes, and MS considerations. Consequently, these findings underscore the need for a timely and comprehensive longitudinal study to monitor the progression of the disease and emphasize prevention strategies, which may vary by region. A focused public health initiative to address the increasing burden of NAFLD in India could be developed by integrating metabolic markers, epigenetic factors, and environmental influences.
The Lean NAFLD Phenomenon: Beyond Obesity
Research on lean NAFLD in the Indian population and the obesity paradox in chronic liver disease highlights the shortcomings of using BMI as a risk indicator [64-66]. Individuals with a normal BMI in India can still develop lean NAFLD, primarily due to the accumulation of visceral fat, IR, and the presence of ectopic fat. At the same time, the obesity paradox [66] suggests that having a slight excess of body fat might be advantageous in severe liver disease, potentially by providing energy reserves or modulating inflammation. These observations underline the intricate role of body fat in liver disease, where lean individuals might face health challenges from visceral fat, while moderate amounts of body fat may enhance survival in advanced cases. These insights suggest the necessity for different approaches in screening and risk assessment, concentrating on metabolic and genetic markers instead of relying only on measurements like BMI [67, 68]. This perspective aligns with the traditional view that not all body fat is harmful, highlighting the importance of tailored strategies in effectively managing the condition. In this context, the Body Roundness Index (BRI) should be included in health evaluations, especially in India, where NAFLD is becoming a significant public health concern [69, 70]. Unlike BMI, which does not consider where fat is distributed, BRI includes waist circumference to offer a more accurate measurement of visceral fat—an important factor in NAFLD. Research indicates that NAFLD is increasing in India, even among individuals with a normal BMI, due to central obesity and genetic factors [8-10, 13, 14]. Given that visceral fat is a better indicator of liver fat accumulation than overall body weight, adding BRI to clinical practices will enhance early detection, enable targeted interventions, and ultimately reduce liver-related health issues [71, 72].
Metabolic and Lifestyle Risk Factors
The metabolic characteristics of India play a crucial role in the prevalence of NAFLD, as they are closely linked to the key elements of MS, including IR, dyslipidemia, hypertension, and central obesity, even among individuals with a normal BMI [73, 74]. These metabolic abnormalities foster a lipotoxic and pro-inflammatory environment, leading to increased liver fat accumulation and disease advancement [75, 76]. Rapid changes in diet and lifestyle driven by urbanization and economic development have further elevated the risk of NAFLD. A growing reliance on ultra-processed foods that are high in refined carbohydrates and unhealthy fats, along with a decline in traditional, fiber-rich diets, has exacerbated metabolic dysfunction [77, 78]. Specific dietary patterns associated with NAFLD show higher consumption of non-vegetarian diets (35% vs. 23%, P = 0.002), regular fried food intake (35% vs. 9%, P = 0.000), frequent consumption of spicy foods (51% vs. 15%, P = 0.001), and tea (55% vs. 39%, P = 0.001) [79].
Urbanization and sedentary habits have emerged as significant metabolic factors contributing to NAFLD. With improved work environments, there is a decline in physical activity and an increase in stress, shifting from physically demanding jobs to predominantly desk-bound roles [80, 81]. This transition has led to reduced daily energy expenditure, promoting the accumulation of visceral fat and prolonged IR. Such changes in metabolic trends highlight the necessity for immediate public health initiatives, which should prioritize lifestyle modifications, early screening programs, and culturally relevant dietary guidelines to address the rising incidence of NAFLD in India. Importantly, the diverse cultural traditions in India have significantly shaped regional eating habits, which influence metabolic and epigenetic profiles that may impact the development of an NAFLD phenotype over time [82, 83]. Traditional diets composed of various whole foods consumed in rural settings create different metabolic profiles. However, this protective effect may be diminished by the increasing trend toward universal diets due to urbanization, alongside a rise in the consumption of high-fat and high-sugar processed foods, which could make individuals more susceptible to metabolic disorders. Chronic dietary patterns can induce epigenetic modifications, such as DNA methylation and histone alterations, which may change the expression of genes involved in lipid metabolism, insulin responsiveness, and inflammation [84, 85]. Epigenetic alterations shaped by dietary habits across generations could lead to population-specific susceptibilities to NAFLD [86, 87]. For instance, pregnant rodents fed a diet high in fat and sugar experience epigenetic changes in the fetal liver [88, 89]. The offspring exhibited changes in the metabolism of liver lipids that increased their risk of developing NAFLD in later life. These changes were associated with modifications in DNA methylation and histone proteins of genes related to lipogenesis and inflammation, suggesting that a mother’s diet can alter the epigenome in a way that affects disease vulnerability in future generations [90, 91]. There are additional animal and human studies linking diet to epigenetic changes, but these fall beyond the scope of this discussion. Hence, understanding how the cultural diversity in diets and their metabolic effects interact with evolving modern diets will be crucial for creating personalized nutrition plans and targeted strategies to address NAFLD in India over time.
Metabolic and Molecular Mechanisms of Hepatic Lipid Dysregulation in NAFLD
The liver plays a vital role in lipid metabolism, managing the synthesis, storage, breakdown, and distribution of lipids to ensure a proper lipid balance [92, 93]. Various tissues use lipids for energy while preventing excessive accumulation. The liver is responsible for synthesizing fatty acids from scratch, controlling the intake of lipids from food and the blood, and overseeing their distribution to other tissues for either storage in fat cells or use through β-oxidation [94, 95]. Four interrelated metabolic pathways govern the lipid equilibrium in the liver: (1) absorption of lipids from the bloodstream, (2) production of fats through de novo synthesis, (3) breakdown of fatty acids, and (4) the creation and secretion of triglyceride-rich very low-density lipoproteins (VLDLs) [96]. The free fatty acids (FFAs) and circulating lipoproteins are derived from either dietary intake or adipose tissue (AT). Excess carbohydrates are converted into fatty acids through a process known as de novo lipogenesis, which uses acetyl-CoA and malonyl-CoA as intermediates and depends significantly on enzymes including fatty acid synthase (FAS) and acetyl-CoA carboxylase (ACC) [97, 98]. The process of fatty acid oxidation (FAO)—which mainly takes place in mitochondria and peroxisomes—produces ATP and ketone bodies [99]. The formation and secretion of VLDLs then facilitate the transport of surplus lipids into the bloodstream, helping to avert metabolic overload in hepatocytes. The pathway to metabolic imbalance is characterized by the accumulation of lipids in liver cells and the onset of NAFLD [100]. IR causes the liver to increase de novo lipogenesis, primarily influenced by the activities of sterol regulatory element-binding protein-1c (SREBP-1c) and enhanced functions of fatty acid synthase (FAS) and acetyl-CoA carboxylase (ACC) [101, 102]. This leads to the generation of more FFAs than the liver can metabolize and export, causing fat to build up within its cells. Dysfunctional AT increases lipolysis, resulting in the release of FFAs into the bloodstream [103, 104]. When the FFAs are taken up by the liver, they can produce toxic lipid intermediates such as ceramides and diacylglycerols (DAGs), which initiate a cascade of MD, ER stress, and inflammation via the JNK (c-Jun N-terminal kinase) and NF-κB (Nuclear Factor-kappa B) signaling pathways [105, 106]. This cellular damage further harms liver parenchymal cells, facilitating the transition from simple steatosis to NASH [107, 108]. Lower levels of adiponectin released by AT, resulting from metabolic dysregulation, diminish insulin sensitivity and its anti-inflammatory role [109, 110]. The levels of pro-inflammatory cytokines, such as TNF-α (Tumor Necrosis Factor Alpha) and IL-6 (interleukin-6), increase, thereby promoting liver inflammation and fibrosis. This establishes a detrimental cycle of excessive lipid accumulation, the presence of harmful saturated lipids, and chronic inflammation, which exacerbates the advancement of NAFLD [111, 112]. Consequently, these findings highlight the urgent need for the development of therapeutic approaches that focus on metabolic pathways to impede disease progression.
Gut-Liver Axis and Dysbiosis, and NAFLD
Emerging evidence suggests that the gut microbiome significantly influences the development and progression of NAFLD by affecting the gut-liver axis, which facilitates bidirectional communication between the gut and liver through portal circulation, bile duct, and systemic circulation [113, 114]. Dysbiosis, or microbial imbalance, interferes with communication between the gut and liver and heightens intestinal permeability, allowing many harmful microbial byproducts to reach the liver [115, 116]. This effect particularly targets tight junction proteins, including occludin, claudin-1, and ZO-1 (Zonula Occludens-1), thereby compromising the gut barrier—this is attributed to decreased mucus production as well as tight junction dysfunction, which impairs the intestines' defense mechanisms [117, 118]. As a result, pathogenic bacterial byproducts can translocate into portal circulation and exacerbate the effects of NAFLD [119, 120].
Additionally, the gut microbiome plays a crucial role in maintaining metabolic balance by regulating bile acid metabolism, insulin sensitivity, and immune responses [121, 122]. Bile acids, which are generated from cholesterol in the liver, facilitate lipid digestion and serve as metabolic signaling molecules [123]. Gut bacteria alter the composition of bile acids through processes like deconjugation, dehydroxylation, and arabinose epimerization, leading to various effects on host metabolism and insulin sensitivity [124, 125]. Bacteroides and Clostridium bacteria are known to deconjugate primary bile acids, thereby reducing the level at which they are reabsorbed [126, 127]. The conversion of primary bile acids into secondary ones by bacteria such as Clostridium and Eubacterium results in varying signaling capabilities. Bile-acid-regulated receptors, namely the farnesoid X receptor (FXR) and the Takeda G-protein-coupled receptor 5 (TGR5), influence lipid metabolism, glucose regulation, and inflammation [128, 129]. Activation of FXR reduces liver fat production and enhances insulin sensitivity, while TGR5 activation boosts energy expenditure and lowers inflammation [130, 131]. Dysbiosis can disturb bile acid balance and hinder FXR/TGR5 signaling, consequently leading to liver fat accumulation and IR [132, 133]. Patients with NASH exhibit increased bile acid production, and preclinical studies indicate that antibiotic treatment may affect the bile acid/intestinal FXR pathway, subsequently influencing fat buildup in the liver [134, 135]. Furthermore, germ-free (GF) mice, which are resistant to obesity caused by diet, illustrate the microbiota's contribution to the progression of NAFLD through FXR-dependent mechanisms [136, 137].
Dysbiosis leads to heightened intestinal permeability, which subsequently triggers metabolic endotoxemia and results in systemic inflammation [138, 139]. The gut barrier typically prevents microbial products like lipopolysaccharides (LPS), peptidoglycans, and bacterial DNA from entering the bloodstream [140, 141]. A counteraction to this is provided by beneficial bacterial metabolites, such as short-chain fatty acids (SCFAs), which contribute to maintaining barrier integrity and ensuring homeostasis within the gut [142, 143]. The main SCFAs—acetate, propionate, and butyrate—are produced through the fermentation of dietary fibers by bacteria and have a range of systemic effects [144, 145]. Butyrate helps maintain the integrity of the gut barrier, stimulates the differentiation of regulatory T cells (Tregs), and exhibits anti-inflammatory properties [146, 147]. Propionate influences the metabolism of glucose in the liver by affecting gluconeogenesis and the accumulation of lipids, whereas acetate acts as an energy source but can also trigger lipogenesis in conditions of microbial imbalance [148, 149]. On the other hand, harmful bacterial products like LPS can increase permeability and inflammation [139, 140]. When LPS and other microbial elements enter the portal circulation, they activate TLR4 (Toll-like receptor 4) on hepatocytes and Kupffer cells, causing NF-κB signaling and the activation of inflammasomes, which leads to the secretion of proinflammatory cytokines such as TNF-α, IL-6, and IL-1β [150-152]. These cytokines contribute to hepatic IR, steatosis, and the advancement of NASH and cirrhosis. Approaches such as probiotics, prebiotics, dietary modifications, or bile acid therapy show promise in re-establishing the balance of microflora and ensuring gut-liver harmony [153, 154]. A thorough understanding of these interactions is crucial for developing therapeutic strategies that may help in preventing or managing metabolic disorders.
CHALLENGES & FUTURE DIRECTIONS
NAFLD in India presents a complex interaction of genetic, metabolic, and environmental factors, making its diagnosis and treatment challenging. The disease is frequently underdiagnosed due to its asymptomatic nature and the limited availability of noninvasive diagnostic methods. Currently, there are no established screening protocols tailored for the diverse Indian population. Genetic factors, such as variations in PNPLA3 or TM6SF2, combined with epigenetic changes influenced by diet and environmental and socioeconomic pressures, further complicate the disease's progression. The importance of the gut-liver relationship is also notable; however, the lack of microbiome studies conducted in India hinders the development of targeted therapies. As urbanization progresses, a diet high in carbohydrates and the presence of obesity alongside micronutrient deficiencies worsen metabolic issues, while socioeconomic inequalities prevent timely interventions. The burden of NAFLD may be intensified in these communities due to another factor, specifically consanguineous marriages, which could lead to a higher inheritance of genetic variants that are detrimental to metabolic functioning, insulin sensitivity, and lipid accumulation in the liver. Additionally, the lack of longitudinal studies using multi-omics approaches complicates our understanding of how NAFLD progresses to more severe conditions like NASH and HCC.
A precision medicine strategy that combines genomics, microbiomics, and metabolomics is essential for early risk evaluation and tailored therapies [155-157]. Innovative non-invasive metabolic and epigenetic biomarkers for diagnostics have the potential to transform early detection while significantly lowering the reliance on liver biopsies [158, 159]. Microbiome-focused strategies, such as the use of probiotics and nutritional changes, should be evaluated scientifically within the Indian population. Predictive models powered by AI enhance the assessment of risk and treatment approaches [160, 161]. At a broader level, there is a need for national screening recommendations for NAFLD, integration into primary healthcare, and public health campaigns promoting healthier lifestyles and diets. Addressing the rising incidence of NAFLD in India will require an interdisciplinary research approach that integrates systems biology with healthcare and policy. There is an urgent need for a precision medicine approach that assesses risk and offers personalized therapies at initial stages, incorporating genomics, metabolomics, and microbiome analysis. New non-invasive metabolic and epigenetic biomarkers for diagnostics have the potential to dramatically improve early detection while significantly reducing the need for liver biopsies. Evaluating microbiome-focused approaches such as probiotics and dietary changes should take place within the Indian context. AI-driven predictive models can improve risk assessment and treatment strategies. Broadly, there should be a framework for national guidelines on NAFLD screening, integration into primary healthcare, and public health efforts that advocate for healthier diets and lifestyles. Solutions will necessitate a systems biology-oriented interdisciplinary research and policy strategy that links research and healthcare to tackle the growing challenge of NAFLD in India [162-164].
Conclusion
India faces a rising burden of NAFLD, influenced by urbanization, metabolic shifts, and genetic predispositions. The high prevalence among both obese and lean individuals highlights the need for improved diagnostic strategies beyond BMI, such as the Body Roundness Index (BRI) and non-invasive biomarkers. Addressing regional dietary patterns, lifestyle changes, and socioeconomic disparities is crucial for effective prevention and management. Given the complex interplay between metabolic, genetic, and gut-liver axis factors, a precision medicine approach integrating genomics, metabolomics, and microbiome research holds promise for personalized interventions. Additionally, national screening guidelines, public health initiatives, and interdisciplinary research efforts are essential to mitigate NAFLD’s growing impact on India’s healthcare system.
Conflict of Interest:
The authors do not have anything to declare.
Acknowledgments
This research is supported by Bandhan, India.
Author Contribution
Conceptualization and supervision: S. K. C.; Formal analysis: S. K. C.; Original draft preparation: S. K. C.; Writing—review and editing: S. K. C. and D. C.; Project administration: S. K. C.; Funding acquisition: S. K. C.
References
-
Rinella ME, Sookoian S. From NAFLD to MASLD: updated naming and diagnosis criteria for fatty liver disease. J Lipid Res. 2024 Jan;65(1):100485. doi: 10.1016/j.jlr.2023.100485.
View at Publisher | View at Google Scholar -
Chan WK, Chuah KH, Rajaram RB, Lim LL, Ratnasingam J, Vethakkan SR. Metabolic Dysfunction-Associated Steatotic Liver Disease (MASLD): A State-of-the-Art Review. J Obes Metab Syndr. 2023 Sep 30;32(3):197-213. doi: 10.7570/jomes23052.
View at Publisher | View at Google Scholar -
Eskridge W, Cryer DR, Schattenberg JM, Gastaldelli A, Malhi H, Allen AM, et al. Metabolic Dysfunction-Associated Steatotic Liver Disease and Metabolic Dysfunction-Associated Steatohepatitis: The Patient and Physician Perspective. J Clin Med. 2023 Sep 26;12(19):6216. doi: 10.3390/jcm12196216.
View at Publisher | View at Google Scholar -
Grander C, Grabherr F, Tilg H. Non-alcoholic fatty liver disease: pathophysiological concepts and treatment options. Cardiovasc Res. 2023 Aug 7;119(9):1787-1798. doi: 10.1093/cvr/cvad095.
View at Publisher | View at Google Scholar -
Francque SM, Marchesini G, Kautz A, Walmsley M, Dorner R, Lazarus JV, et al. Non-alcoholic fatty liver disease: A patient guideline. JHEP Rep. 2021 Sep 17;3(5):100322. doi: 10.1016/j.jhepr.2021.100322. Erratum in: JHEP Rep. 2024 May 18;6(6):101097. doi: 10.1016/j.jhepr.2024.101097.
View at Publisher | View at Google Scholar -
Benedict M, Zhang X. Non-alcoholic fatty liver disease: An expanded review. World J Hepatol. 2017 Jun 8;9(16):715-732. doi: 10.4254/wjh.v9.i16.715.
View at Publisher | View at Google Scholar -
Shalimar, Elhence A, Bansal B, Gupta H, Anand A, Singh TP, Goel A. Prevalence of Non-alcoholic Fatty Liver Disease in India: A Systematic Review and Meta-analysis. J Clin Exp Hepatol. 2022 May-Jun;12(3):818-829. doi: 10.1016/j.jceh.2021.11.010.
View at Publisher | View at Google Scholar -
Kannan S, Nelliyanil M, Mendagudli R, Rajeshwari S, Kona C, Kundapur R, et al. Evaluation of risk factors for non-alcoholic fatty liver disease in India: A systematic review and meta-analysis. J Educ Health Promot. 2024 Jan 22;12:435. doi: 10.4103/jehp.jehp_208_23.
View at Publisher | View at Google Scholar -
De A, Duseja A. Nonalcoholic Fatty Liver Disease: Indian Perspective. Clin Liver Dis (Hoboken). 2021 Sep 13;18(3):158-163. doi: 10.1002/cld.1141.
View at Publisher | View at Google Scholar -
Singh SP, Panigrahi MK, Patel A, Viswanathan L, Rath MM, Kar SK, Harrison SA. Comparison of Clinical, Biochemical, and Histopathologic Profiles between NAFLD in Asian-Indians and United States Adults. Euroasian J Hepatogastroenterol. 2022 Jul;12(Suppl 1):S15-S18. doi: 10.5005/jp-journals-10018-1362.
View at Publisher | View at Google Scholar -
Mitra S, De A, Chowdhury A. Epidemiology of non-alcoholic and alcoholic fatty liver diseases. Transl Gastroenterol Hepatol. 2020 Apr 5;5:16. doi: 10.21037/tgh.2019.09.08.
View at Publisher | View at Google Scholar -
Fabbrini E, Sullivan S, Klein S. Obesity and nonalcoholic fatty liver disease: biochemical, metabolic, and clinical implications. Hepatology. 2010 Feb;51(2):679-89. doi: 10.1002/hep.23280.
View at Publisher | View at Google Scholar -
Albhaisi S, Chowdhury A, Sanyal AJ. Non-alcoholic fatty liver disease in lean individuals. JHEP Rep. 2019 Aug 30;1(4):329-341. doi: 10.1016/j.jhepr.2019.08.002.
View at Publisher | View at Google Scholar -
Pati GK, Singh SP. Nonalcoholic Fatty Liver Disease in South Asia. Euroasian J Hepatogastroenterol. 2016 Jul-Dec;6(2):154-162. doi: 10.5005/jp-journals-10018-1189.
View at Publisher | View at Google Scholar -
Shalimar, Elhence A, Bansal B, Gupta H, Anand A, Singh TP, Goel A. Prevalence of Non-alcoholic Fatty Liver Disease in India: A Systematic Review and Meta-analysis. J Clin Exp Hepatol. 2022 May-Jun;12(3):818-829. doi: 10.1016/j.jceh.2021.11.010.
View at Publisher | View at Google Scholar -
Kumar R, Mohan S. Non-alcoholic Fatty Liver Disease in Lean Subjects: Characteristics and Implications. J Clin Transl Hepatol. 2017 Sep 28;5(3):216-223. doi: 10.14218/JCTH.2016.00068.
View at Publisher | View at Google Scholar -
DiStefano JK, Gerhard GS. NAFLD in normal weight individuals. Diabetol Metab Syndr. 2022 Mar 24;14(1):45. doi: 10.1186/s13098-022-00814-z.
View at Publisher | View at Google Scholar -
Kuchay MS, Martínez-Montoro JI, Choudhary NS, Fernández-García JC, Ramos-Molina B. Non-Alcoholic Fatty Liver Disease in Lean and Non-Obese Individuals: Current and Future Challenges. Biomedicines. 2021 Sep 28;9(10):1346. doi: 10.3390/biomedicines9101346.
View at Publisher | View at Google Scholar -
Ipsen DH, Lykkesfeldt J, Tveden-Nyborg P. Molecular mechanisms of hepatic lipid accumulation in non-alcoholic fatty liver disease. Cell Mol Life Sci. 2018 Sep;75(18):3313-3327. doi: 10.1007/s00018-018-2860-6.
View at Publisher | View at Google Scholar -
Prasun P, Ginevic I, Oishi K. Mitochondrial dysfunction in nonalcoholic fatty liver disease and alcohol related liver disease. Transl Gastroenterol Hepatol. 2021 Jan 5;6:4. doi: 10.21037/tgh-20-125.
View at Publisher | View at Google Scholar -
Karkucinska-Wieckowska A, Simoes ICM, Kalinowski P, Lebiedzinska-Arciszewska M, Zieniewicz K, Milkiewicz P, et al. Mitochondria, oxidative stress and nonalcoholic fatty liver disease: A complex relationship. Eur J Clin Invest. 2022 Mar;52(3):e13622. doi: 10.1111/eci.13622.
View at Publisher | View at Google Scholar -
Lebeaupin C, Vallée D, Hazari Y, Hetz C, Chevet E, Bailly-Maitre B. Endoplasmic reticulum stress signalling and the pathogenesis of non-alcoholic fatty liver disease. J Hepatol. 2018 Oct;69(4):927-947. doi: 10.1016/j.jhep.2018.06.008.
View at Publisher | View at Google Scholar -
Thibaut R, Gage MC, Pineda-Torra I, Chabrier G, Venteclef N, Alzaid F. Liver macrophages and inflammation in physiology and physiopathology of non-alcoholic fatty liver disease. FEBS J. 2022 Jun;289(11):3024-3057. doi: 10.1111/febs.15877.
View at Publisher | View at Google Scholar -
Wang M, Li L, Xu Y, Du J, Ling C. Roles of hepatic stellate cells in NAFLD: From the perspective of inflammation and fibrosis. Front Pharmacol. 2022 Oct 13;13:958428. doi: 10.3389/fphar.2022.958428.
View at Publisher | View at Google Scholar -
Kobayashi T, Iwaki M, Nakajima A, Nogami A, Yoneda M. Current Research on the Pathogenesis of NAFLD/NASH and the Gut-Liver Axis: Gut Microbiota, Dysbiosis, and Leaky-Gut Syndrome. Int J Mol Sci. 2022 Oct 2;23(19):11689. doi: 10.3390/ijms231911689.
View at Publisher | View at Google Scholar -
Pezzino S, Sofia M, Faletra G, Mazzone C, Litrico G, La Greca G, Latteri S. Gut-Liver Axis and Non-Alcoholic Fatty Liver Disease: A Vicious Circle of Dysfunctions Orchestrated by the Gut Microbiome. Biology (Basel). 2022 Nov 6;11(11):1622. doi: 10.3390/biology11111622.
View at Publisher | View at Google Scholar -
Ohtani N, Kamiya T, Kawada N. Recent updates on the role of the gut-liver axis in the pathogenesis of NAFLD/NASH, HCC, and beyond. Hepatol Commun. 2023 Aug 28;7(9):e0241. doi: 10.1097/HC9.0000000000000241.
View at Publisher | View at Google Scholar -
Singh S, Kuftinec GN, Sarkar S. Non-alcoholic Fatty Liver Disease in South Asians: A Review of the Literature. J Clin Transl Hepatol. 2017 Mar 28;5(1):76-81. doi: 10.14218/JCTH.2016.00045.
View at Publisher | View at Google Scholar -
Kovalic AJ, Banerjee P, Tran QT, Singal AK, Satapathy SK. Genetic and Epigenetic Culprits in the Pathogenesis of Nonalcoholic Fatty Liver Disease. J Clin Exp Hepatol. 2018 Dec;8(4):390-402. doi: 10.1016/j.jceh.2018.04.001.
View at Publisher | View at Google Scholar -
Sulaiman SA, Dorairaj V, Adrus MNH. Genetic Polymorphisms and Diversity in Nonalcoholic Fatty Liver Disease (NAFLD): A Mini Review. Biomedicines. 2022 Dec 30;11(1):106. doi: 10.3390/biomedicines11010106.
View at Publisher | View at Google Scholar -
Njei B, Al-Ajlouni YA, Ugwendum D, Abdu M, Forjindam A, Mohamed MF. Genetic and epigenetic determinants of non-alcoholic fatty liver disease (NAFLD) in lean individuals: a systematic review. Transl Gastroenterol Hepatol. 2023 Dec 6;9:11. doi: 10.21037/tgh-23-31.
View at Publisher | View at Google Scholar -
Rüschenbaum S, Schwarzkopf K, Friedrich-Rust M, Seeger F, Schoelzel F, Martinez Y, et al. Patatin-like phospholipase domain containing 3 variants differentially impact metabolic traits in individuals at high risk for cardiovascular events. Hepatol Commun. 2018 Jun 6;2(7):798-806. doi: 10.1002/hep4.1183.
View at Publisher | View at Google Scholar -
Luo F, Oldoni F, Das A. TM6SF2: A Novel Genetic Player in Nonalcoholic Fatty Liver and Cardiovascular Disease. Hepatol Commun. 2022 Mar;6(3):448-460. doi: 10.1002/hep4.1822.
View at Publisher | View at Google Scholar -
Carlsson B, Lindén D, Brolén G, Liljeblad M, Bjursell M, Romeo S, Loomba R. Review article: the emerging role of genetics in precision medicine for patients with non-alcoholic steatohepatitis. Aliment Pharmacol Ther. 2020 Jun;51(12):1305-1320. doi: 10.1111/apt.15738.
View at Publisher | View at Google Scholar -
Zaiou M, Amrani R, Rihn B, Hajri T. Dietary Patterns Influence Target Gene Expression through Emerging Epigenetic Mechanisms in Nonalcoholic Fatty Liver Disease. Biomedicines. 2021 Sep 18;9(9):1256. doi: 10.3390/biomedicines9091256.
View at Publisher | View at Google Scholar -
Rodríguez-Sanabria JS, Escutia-Gutiérrez R, Rosas-Campos R, Armendáriz-Borunda JS, Sandoval-Rodríguez A. An Update in Epigenetics in Metabolic-Associated Fatty Liver Disease. Front Med (Lausanne). 2022 Jan 11;8:770504. doi: 10.3389/fmed.2021.770504.
View at Publisher | View at Google Scholar -
Aggeletopoulou I, Kalafateli M, Tsounis EP, Triantos C. Epigenetic Regulation in Lean Nonalcoholic Fatty Liver Disease. Int J Mol Sci. 2023 Aug 16;24(16):12864. doi: 10.3390/ijms241612864.
View at Publisher | View at Google Scholar -
Hyun J, Jung Y. DNA Methylation in Nonalcoholic Fatty Liver Disease. Int J Mol Sci. 2020 Oct 30;21(21):8138. doi: 10.3390/ijms21218138.
View at Publisher | View at Google Scholar -
Duseja A, Singh SP, De A, Madan K, Rao PN, Shukla A, et al. Indian National Association for Study of the Liver (INASL) Guidance Paper on Nomenclature, Diagnosis and Treatment of Nonalcoholic Fatty Liver Disease (NAFLD). J Clin Exp Hepatol. 2023 Mar-Apr;13(2):273-302. doi: 10.1016/j.jceh.2022.11.014.
View at Publisher | View at Google Scholar -
Ferraioli G, Soares Monteiro LB. Ultrasound-based techniques for the diagnosis of liver steatosis. World J Gastroenterol. 2019 Oct 28;25(40):6053-6062. doi: 10.3748/wjg.v25.i40.6053.
View at Publisher | View at Google Scholar -
Kechagias S, Ekstedt M, Simonsson C, Nasr P. Non-invasive diagnosis and staging of non-alcoholic fatty liver disease. Hormones (Athens). 2022 Sep;21(3):349-368. doi: 10.1007/s42000-022-00377-8. Epub 2022 Jun 4.
View at Publisher | View at Google Scholar -
van Beek JH, de Moor MH, de Geus EJ, Lubke GH, Vink JM, Willemsen G, Boomsma DI. The genetic architecture of liver enzyme levels: GGT, ALT and AST. Behav Genet. 2013 Jul;43(4):329-39. doi: 10.1007/s10519-013-9593-y.
View at Publisher | View at Google Scholar -
Qiu J, Kuang M, He S, Yu C, Wang C, Huang X, et al. Gender perspective on the association between liver enzyme markers and non-alcoholic fatty liver disease: insights from the general population. Front Endocrinol (Lausanne). 2023 Dec 6;14:1302322. doi: 10.3389/fendo.2023.1302322.
View at Publisher | View at Google Scholar -
Park H, Yoon EL, Kim M, Lee J, Kim HL, Cho S, et al. Diagnostic performance of the fibrosis-4 index and the NAFLD fibrosis score for screening at-risk individuals in a health check-up setting. Hepatol Commun. 2023 Sep 22;7(10):e0249. doi: 10.1097/HC9.0000000000000249.
View at Publisher | View at Google Scholar -
Zhou JH, Cai JJ, She ZG, Li HL. Noninvasive evaluation of nonalcoholic fatty liver disease: Current evidence and practice. World J Gastroenterol. 2019 Mar 21;25(11):1307-1326. doi: 10.3748/wjg.v25.i11.1307.
View at Publisher | View at Google Scholar -
Park CC, Nguyen P, Hernandez C, Bettencourt R, Ramirez K, Fortney L, et al. Magnetic Resonance Elastography vs Transient Elastography in Detection of Fibrosis and Noninvasive Measurement of Steatosis in Patients With Biopsy-Proven Nonalcoholic Fatty Liver Disease. Gastroenterology. 2017 Feb;152(3):598-607.e2. doi: 10.1053/j.gastro.2016.10.026.
View at Publisher | View at Google Scholar -
Caussy C, Reeder SB, Sirlin CB, Loomba R. Noninvasive, Quantitative Assessment of Liver Fat by MRI-PDFF as an Endpoint in NASH Trials. Hepatology. 2018 Aug;68(2):763-772. doi: 10.1002/hep.29797.
View at Publisher | View at Google Scholar -
Wang JL, Jiang SW, Hu AR, Zhou AW, Hu T, Li HS, et al. Non-invasive diagnosis of non-alcoholic fatty liver disease: Current status and future perspective. Heliyon. 2024 Feb 29;10(5):e27325. doi: 10.1016/j.heliyon.2024.e27325.
View at Publisher | View at Google Scholar -
Di Sessa A, Riccio S, Pirozzi E, Verde M, Passaro AP, Umano GR, et al. Advances in paediatric nonalcoholic fatty liver disease: Role of lipidomics. World J Gastroenterol. 2021 Jul 7;27(25):3815-3824. doi: 10.3748/wjg.v27.i25.3815.
View at Publisher | View at Google Scholar -
Ajmera V, Kim BK, Yang K, Majzoub AM, Nayfeh T, Tamaki N, et al. Liver Stiffness on Magnetic Resonance Elastography and the MEFIB Index and Liver-Related Outcomes in Nonalcoholic Fatty Liver Disease: A Systematic Review and Meta-Analysis of Individual Participants. Gastroenterology. 2022 Oct;163(4):1079-1089.e5. doi: 10.1053/j.gastro.2022.06.073.
View at Publisher | View at Google Scholar -
Pepin KM, Welle CL, Guglielmo FF, Dillman JR, Venkatesh SK. Magnetic resonance elastography of the liver: everything you need to know to get started. Abdom Radiol (NY). 2022 Jan;47(1):94-114. doi: 10.1007/s00261-021-03324-0.
View at Publisher | View at Google Scholar -
Sumida Y, Nakajima A, Itoh Y. Limitations of liver biopsy and non-invasive diagnostic tests for the diagnosis of nonalcoholic fatty liver disease/nonalcoholic steatohepatitis. World J Gastroenterol. 2014 Jan 14;20(2):475-85. doi: 10.3748/wjg.v20.i2.475.
View at Publisher | View at Google Scholar -
Chow KW, Futela P, Saharan A, Saab S. Comparison of Guidelines for the Screening, Diagnosis, and Noninvasive Assessment of Nonalcoholic Fatty Liver Disease. J Clin Exp Hepatol. 2023 Sep-Oct;13(5):783-793. doi: 10.1016/j.jceh.2023.01.016.
View at Publisher | View at Google Scholar -
Cernea S, Onișor D. Screening and interventions to prevent nonalcoholic fatty liver disease/nonalcoholic steatohepatitis-associated hepatocellular carcinoma. World J Gastroenterol. 2023 Jan 14;29(2):286-309. doi: 10.3748/wjg.v29.i2.286.
View at Publisher | View at Google Scholar -
Riazi K, Swain MG, Congly SE, Kaplan GG, Shaheen AA. Race and Ethnicity in Non-Alcoholic Fatty Liver Disease (NAFLD): A Narrative Review. Nutrients. 2022 Oct 28;14(21):4556. doi: 10.3390/nu14214556.
View at Publisher | View at Google Scholar -
Rahman MM, Kibria MG, Begum H, Haque M, Sultana N, Akhter M, et al. Prevalence, risk factors and metabolic profile of the non-obese and obese non-alcoholic fatty liver disease in a rural community of South Asia. BMJ Open Gastroenterol. 2020 Dec;7(1):e000535. doi: 10.1136/bmjgast-2020-000535.
View at Publisher | View at Google Scholar -
Duseja A, Das A, Dhiman RK, Chawla YK, Das R, Bhadada S, et al. Indian patients with nonalcoholic fatty liver disease presenting with raised transaminases are different at presentation. World J Gastroenterol. 2007 Jan 28;13(4):649-50. doi: 10.3748/wjg.v13.i4.649.
View at Publisher | View at Google Scholar -
Asadullah M, Shivashankar R, Shalimar, Kandasamy D, Kondal D, Rautela G, et al. Rural-Urban differentials in prevalence, spectrum and determinants of Non-alcoholic Fatty Liver Disease in North Indian population. PLoS One. 2022 Feb 10;17(2):e0263768. doi: 10.1371/journal.pone.0263768.
View at Publisher | View at Google Scholar -
Orkin S, Brokamp C, Yodoshi T, Trout AT, Liu C, Meryum S, et al. Community Socioeconomic Deprivation and Nonalcoholic Fatty Liver Disease Severity. J Pediatr Gastroenterol Nutr. 2020 Mar;70(3):364-370. doi: 10.1097/MPG.0000000000002527.
View at Publisher | View at Google Scholar -
Nurwanti E, Hadi H, Chang JS, Chao JC, Paramashanti BA, Gittelsohn J, Bai CH. Rural-Urban Differences in Dietary Behavior and Obesity: Results of the Riskesdas Study in 10-18-Year-Old Indonesian Children and Adolescents. Nutrients. 2019 Nov 18;11(11):2813. doi: 10.3390/nu11112813.
View at Publisher | View at Google Scholar -
Sharma M, Kishore A, Roy D, Joshi K. A comparison of the Indian diet with the EAT-Lancet reference diet. BMC Public Health. 2020 May 29;20(1):812. doi: 10.1186/s12889-020-08951-8.
View at Publisher | View at Google Scholar -
Rasmussen L, Knorr S, Antoniussen CS, Bruun JM, Ovesen PG, Fuglsang J, Kampmann U. The Impact of Lifestyle, Diet and Physical Activity on Epigenetic Changes in the Offspring-A Systematic Review. Nutrients. 2021 Aug 17;13(8):2821. doi: 10.3390/nu13082821.
View at Publisher | View at Google Scholar -
Abraham MJ, El Sherbini A, El-Diasty M, Askari S, Szewczuk MR. Restoring Epigenetic Reprogramming with Diet and Exercise to Improve Health-Related Metabolic Diseases. Biomolecules. 2023 Feb 7;13(2):318. doi: 10.3390/biom13020318.
View at Publisher | View at Google Scholar -
Premkumar M, Anand AC. Lean Fatty Liver Disease: Through Thick and Thin. J Clin Exp Hepatol. 2021 Sep-Oct;11(5):523-527. doi: 10.1016/j.jceh.2021.06.011.
View at Publisher | View at Google Scholar -
Chrysavgis L, Ztriva E, Protopapas A, Tziomalos K, Cholongitas E. Nonalcoholic fatty liver disease in lean subjects: Prognosis, outcomes and management. World J Gastroenterol. 2020 Nov 14;26(42):6514-6528. doi: 10.3748/wjg.v26.i42.6514.
View at Publisher | View at Google Scholar -
Curcic IB, Berkovic MC, Kuna L, Roguljic H, Smolic R, Varzic SC, et al. Obesity Paradox in Chronic Liver Diseases: Product of Bias or a Real Thing? J Clin Transl Hepatol. 2019 Sep 28;7(3):275-279. doi: 10.14218/JCTH.2019.00029.
View at Publisher | View at Google Scholar -
Wu Y, Li D, Vermund SH. Advantages and Limitations of the Body Mass Index (BMI) to Assess Adult Obesity. Int J Environ Res Public Health. 2024 Jun 10;21(6):757. doi: 10.3390/ijerph21060757.
View at Publisher | View at Google Scholar -
Holmes CJ, Racette SB. The Utility of Body Composition Assessment in Nutrition and Clinical Practice: An Overview of Current Methodology. Nutrients. 2021 Jul 22;13(8):2493. doi: 10.3390/nu13082493.
View at Publisher | View at Google Scholar -
Jiang N, Zhang S, Chu J, Yang N, Lu M. Association between body roundness index and non-alcoholic fatty liver disease detected by Fibroscan in America. J Clin Lab Anal. 2023 Oct;37(19-20):e24973. doi: 10.1002/jcla.24973.
View at Publisher | View at Google Scholar -
Zhang F, Shi W, Wen J, Cao H, Xu W, Lan T, et al. Elevated body roundness index increases the risk of cardiovascular disease in Chinese patients with circadian syndrome. Front Endocrinol (Lausanne). 2025 Feb 25;16:1532344. doi: 10.3389/fendo.2025.1532344.
View at Publisher | View at Google Scholar -
Hernández-Conde M, Llop E, Carrillo CF, Tormo B, Abad J, Rodriguez L, et al. Estimation of visceral fat is useful for the diagnosis of significant fibrosis in patients with non-alcoholic fatty liver disease. World J Gastroenterol. 2020 Nov 14;26(42):6658-6668. doi: 10.3748/wjg.v26.i42.6658.
View at Publisher | View at Google Scholar -
Radmehr M, Homayounfar R, Djazayery A. The relationship between anthropometric indices and non-alcoholic fatty liver disease in adults: a cross-sectional study. Front Nutr. 2025 Jan 7;11:1494497. doi: 10.3389/fnut.2024.1494497.
View at Publisher | View at Google Scholar -
Fatima H, Sohail Rangwala H, Mustafa MS, Shafique MA, Abbas SR, Sohail Rangwala B. Analyzing and evaluating the prevalence and metabolic profile of lean NAFLD compared to obese NAFLD: a systemic review and meta-analysis. Ther Adv Endocrinol Metab. 2024 Sep 3;15:20420188241274310. doi: 10.1177/20420188241274310.
View at Publisher | View at Google Scholar -
Friedman SL, Neuschwander-Tetri BA, Rinella M, Sanyal AJ. Mechanisms of NAFLD development and therapeutic strategies. Nat Med. 2018 Jul;24(7):908-922. doi: 10.1038/s41591-018-0104-9.
View at Publisher | View at Google Scholar -
Fernandez CJ, Nagendra L, Pappachan JM. Metabolic Dysfunction-associated Fatty Liver Disease: An Urgent Call for Global Action. touchREV Endocrinol. 2024 Apr;20(1):5-9. doi: 10.17925/EE.2023.20.1.1.
View at Publisher | View at Google Scholar -
Syed-Abdul MM. Lipid Metabolism in Metabolic-Associated Steatotic Liver Disease (MASLD). Metabolites. 2023 Dec 23;14(1):12. doi: 10.3390/metabo14010012.
View at Publisher | View at Google Scholar -
Harlan TS, Gow RV, Kornstädt A, Alderson PW, Lustig RH. The Metabolic Matrix: Re-engineering ultraprocessed foods to feed the gut, protect the liver, and support the brain. Front Nutr. 2023 Mar 30;10:1098453. doi: 10.3389/fnut.2023.1098453.
View at Publisher | View at Google Scholar -
Clemente-Suárez VJ, Beltrán-Velasco AI, Redondo-Flórez L, Martín-Rodríguez A, Tornero-Aguilera JF. Global Impacts of Western Diet and Its Effects on Metabolism and Health: A Narrative Review. Nutrients. 2023 Jun 14;15(12):2749. doi: 10.3390/nu15122749.
View at Publisher | View at Google Scholar -
Singh SP, Singh A, Misra D, Misra B, Pati GK, Panigrahi MK, et al. Risk Factors Associated With Non-Alcoholic Fatty Liver Disease in Indians: A Case-Control Study. J Clin Exp Hepatol. 2015 Dec;5(4):295-302. doi: 10.1016/j.jceh.2015.09.001.
View at Publisher | View at Google Scholar -
Zelber-Sagi S, Ratziu V, Oren R. Nutrition and physical activity in NAFLD: an overview of the epidemiological evidence. World J Gastroenterol. 2011 Aug 7;17(29):3377-89. doi: 10.3748/wjg.v17.i29.3377.
View at Publisher | View at Google Scholar -
Hallsworth K, Adams LA. Lifestyle modification in NAFLD/NASH: Facts and figures. JHEP Rep. 2019 Nov 5;1(6):468-479. doi: 10.1016/j.jhepr.2019.10.008.
View at Publisher | View at Google Scholar -
Dong H, Sun Y, Nie L, Cui A, Zhao P, Leung WK, Wang Q. Metabolic memory: mechanisms and diseases. Signal Transduct Target Ther. 2024 Feb 28;9(1):38. doi: 10.1038/s41392-024-01755-x.
View at Publisher | View at Google Scholar -
Samaddar A, Cuevas RP, Custodio MC, Ynion J, Ray Chakravarti A, Mohanty SK, Demont M. Capturing diversity and cultural drivers of food choice in eastern India. Int J Gastron Food Sci. 2020 Dec;22:100249. doi: 10.1016/j.ijgfs.2020.100249.
View at Publisher | View at Google Scholar -
Mahmoud AM. An Overview of Epigenetics in Obesity: The Role of Lifestyle and Therapeutic Interventions. Int J Mol Sci. 2022 Jan 25;23(3):1341. doi: 10.3390/ijms23031341.
View at Publisher | View at Google Scholar -
Asif S, Morrow NM, Mulvihill EE, Kim KH. Understanding Dietary Intervention-Mediated Epigenetic Modifications in Metabolic Diseases. Front Genet. 2020 Oct 15;11:590369. doi: 10.3389/fgene.2020.590369.
View at Publisher | View at Google Scholar -
Herranz JM, López-Pascual A, Clavería-Cabello A, Uriarte I, Latasa MU, Irigaray-Miramon A, et al. Comprehensive analysis of epigenetic and epitranscriptomic genes' expression in human NAFLD. J Physiol Biochem. 2023 Nov;79(4):901-924. doi: 10.1007/s13105-023-00976-y.
View at Publisher | View at Google Scholar -
Meroni M, Longo M, Rustichelli A, Dongiovanni P. Nutrition and Genetics in NAFLD: The Perfect Binomium. Int J Mol Sci. 2020 Apr 23;21(8):2986. doi: 10.3390/ijms21082986.
View at Publisher | View at Google Scholar -
Banik A, Kandilya D, Ramya S, Stünkel W, Chong YS, Dheen ST. Maternal Factors that Induce Epigenetic Changes Contribute to Neurological Disorders in Offspring. Genes (Basel). 2017 May 24;8(6):150. doi: 10.3390/genes8060150.
View at Publisher | View at Google Scholar -
Rodrigo N, Saad S, Pollock C, Glastras SJ. Diet Modification before or during Pregnancy on Maternal and Foetal Outcomes in Rodent Models of Maternal Obesity. Nutrients. 2022 May 21;14(10):2154. doi: 10.3390/nu14102154.
View at Publisher | View at Google Scholar -
Zhang Q, Xiao X, Zheng J, Li M, Yu M, Ping F, et al. A Maternal High-Fat Diet Induces DNA Methylation Changes That Contribute to Glucose Intolerance in Offspring. Front Endocrinol (Lausanne). 2019 Dec 13;10:871. doi: 10.3389/fendo.2019.00871.
View at Publisher | View at Google Scholar -
Lee HS. Impact of Maternal Diet on the Epigenome during In Utero Life and the Developmental Programming of Diseases in Childhood and Adulthood. Nutrients. 2015 Nov 17;7(11):9492-507. doi: 10.3390/nu7115467.
View at Publisher | View at Google Scholar -
Nguyen P, Leray V, Diez M, Serisier S, Le Bloc'h J, Siliart B, Dumon H. Liver lipid metabolism. J Anim Physiol Anim Nutr (Berl). 2008 Jun;92(3):272-83. doi: 10.1111/j.1439-0396.2007.00752.x.
View at Publisher | View at Google Scholar -
Pei K, Gui T, Kan D, Feng H, Jin Y, Yang Y, et al. An Overview of Lipid Metabolism and Nonalcoholic Fatty Liver Disease. Biomed Res Int. 2020 Jul 18;2020:4020249. doi: 10.1155/2020/4020249.
View at Publisher | View at Google Scholar -
Hodson L. Hepatic fatty acid synthesis and partitioning: the effect of metabolic and nutritional state. Proc Nutr Soc. 2019 Feb;78(1):126-134. doi: 10.1017/S0029665118002653.
View at Publisher | View at Google Scholar -
Alves-Bezerra M, Cohen DE. Triglyceride Metabolism in the Liver. Compr Physiol. 2017 Dec 12;8(1):1-8. doi: 10.1002/cphy.c170012.
View at Publisher | View at Google Scholar -
Rao G, Peng X, Li X, An K, He H, Fu X, et al. Unmasking the enigma of lipid metabolism in metabolic dysfunction-associated steatotic liver disease: from mechanism to the clinic. Front Med (Lausanne). 2023 Nov 27;10:1294267. doi: 10.3389/fmed.2023.1294267.
View at Publisher | View at Google Scholar -
Softic S, Cohen DE, Kahn CR. Role of Dietary Fructose and Hepatic De Novo Lipogenesis in Fatty Liver Disease. Dig Dis Sci. 2016 May;61(5):1282-93. doi: 10.1007/s10620-016-4054-0.
View at Publisher | View at Google Scholar -
Solinas G, Borén J, Dulloo AG. De novo lipogenesis in metabolic homeostasis: More friend than foe? Mol Metab. 2015 Mar 20;4(5):367-77. doi: 10.1016/j.molmet.2015.03.004.
View at Publisher | View at Google Scholar -
Tamas C, Tamas F, Kovecsi A, Cehan A, Balasa A. Metabolic Contrasts: Fatty Acid Oxidation and Ketone Bodies in Healthy Brains vs. Glioblastoma Multiforme. Int J Mol Sci. 2024 May 17;25(10):5482. doi: 10.3390/ijms25105482.
View at Publisher | View at Google Scholar -
Choi SH, Ginsberg HN. Increased very low density lipoprotein (VLDL) secretion, hepatic steatosis, and insulin resistance. Trends Endocrinol Metab. 2011 Sep;22(9):353-63. doi: 10.1016/j.tem.2011.04.007.
View at Publisher | View at Google Scholar -
Ruiz R, Jideonwo V, Ahn M, Surendran S, Tagliabracci VS, Hou Y, et al. Sterol regulatory element-binding protein-1 (SREBP-1) is required to regulate glycogen synthesis and gluconeogenic gene expression in mouse liver. J Biol Chem. 2014 Feb 28;289(9):5510-7. doi: 10.1074/jbc.M113.541110.
View at Publisher | View at Google Scholar -
Ferré P, Foufelle F. Hepatic steatosis: a role for de novo lipogenesis and the transcription factor SREBP-1c. Diabetes Obes Metab. 2010 Oct;12 Suppl 2:83-92. doi: 10.1111/j.1463-1326.2010.01275.x.
View at Publisher | View at Google Scholar -
Heeren J, Scheja L. Metabolic-associated fatty liver disease and lipoprotein metabolism. Mol Metab. 2021 Aug;50:101238. doi: 10.1016/j.molmet.2021.101238.
View at Publisher | View at Google Scholar -
Berlanga A, Guiu-Jurado E, Porras JA, Auguet T. Molecular pathways in non-alcoholic fatty liver disease. Clin Exp Gastroenterol. 2014 Jul 5;7:221-39. doi: 10.2147/CEG.S62831.
View at Publisher | View at Google Scholar -
Geng Y, Faber KN, de Meijer VE, Blokzijl H, Moshage H. How does hepatic lipid accumulation lead to lipotoxicity in non-alcoholic fatty liver disease? Hepatol Int. 2021 Feb;15(1):21-35. doi: 10.1007/s12072-020-10121-2.
View at Publisher | View at Google Scholar -
Mota M, Banini BA, Cazanave SC, Sanyal AJ. Molecular mechanisms of lipotoxicity and glucotoxicity in nonalcoholic fatty liver disease. Metabolism. 2016 Aug;65(8):1049-61. doi: 10.1016/j.metabol.2016.02.014.
View at Publisher | View at Google Scholar -
Berardo C, Di Pasqua LG, Cagna M, Richelmi P, Vairetti M, Ferrigno A. Nonalcoholic Fatty Liver Disease and Non-Alcoholic Steatohepatitis: Current Issues and Future Perspectives in Preclinical and Clinical Research. Int J Mol Sci. 2020 Dec 17;21(24):9646. doi: 10.3390/ijms21249646.
View at Publisher | View at Google Scholar -
Fielding CM, Angulo P. Hepatic steatosis and steatohepatitis: Are they really two distinct entities? Curr Hepatol Rep. 2014 Jun 1;13(2):151-158. doi: 10.1007/s11901-014-0227-5.
View at Publisher | View at Google Scholar -
Baldelli S, Aiello G, Mansilla Di Martino E, Campaci D, Muthanna FMS, Lombardo M. The Role of Adipose Tissue and Nutrition in the Regulation of Adiponectin. Nutrients. 2024 Jul 26;16(15):2436. doi: 10.3390/nu16152436.
View at Publisher | View at Google Scholar -
Fajkić A, Jahić R, Ejubović M, Đešević M, Ejubović AJ, Lepara O. The Trend of Changes in Adiponectin, Resistin, and Adiponectin-Resistin Index Values in Type 2 Diabetic Patients with the Development of Metabolic Syndrome. Medicina (Kaunas). 2024 Nov 1;60(11):1795. doi: 10.3390/medicina60111795.
View at Publisher | View at Google Scholar -
Kern L, Mittenbühler MJ, Vesting AJ, Ostermann AL, Wunderlich CM, Wunderlich FT. Obesity-Induced TNFα and IL-6 Signaling: The Missing Link between Obesity and Inflammation-Driven Liver and Colorectal Cancers. Cancers (Basel). 2018 Dec 27;11(1):24. doi: 10.3390/cancers11010024.
View at Publisher | View at Google Scholar -
Popko K, Gorska E, Stelmaszczyk-Emmel A, Plywaczewski R, Stoklosa A, Gorecka D, et al. Proinflammatory cytokines Il-6 and TNF-α and the development of inflammation in obese subjects. Eur J Med Res. 2010 Nov 4;15 Suppl 2(Suppl 2):120-2. doi: 10.1186/2047-783x-15-s2-120.
View at Publisher | View at Google Scholar -
Gupta H, Min BH, Ganesan R, Gebru YA, Sharma SP, Park E, et al. Gut Microbiome in Non-Alcoholic Fatty Liver Disease: From Mechanisms to Therapeutic Role. Biomedicines. 2022 Feb 25;10(3):550. doi: 10.3390/biomedicines10030550.
View at Publisher | View at Google Scholar -
Tripathi A, Debelius J, Brenner DA, Karin M, Loomba R, Schnabl B, Knight R. The gut-liver axis and the intersection with the microbiome. Nat Rev Gastroenterol Hepatol. 2018 Jul;15(7):397-411. doi: 10.1038/s41575-018-0011-z. Erratum in: Nat Rev Gastroenterol Hepatol. 2018 Dec;15(12):785. doi: 10.1038/s41575-018-0031-8.
View at Publisher | View at Google Scholar -
Plaza-Díaz J, Solís-Urra P, Rodríguez-Rodríguez F, Olivares-Arancibia J, Navarro-Oliveros M, Abadía-Molina F, Álvarez-Mercado AI. The Gut Barrier, Intestinal Microbiota, and Liver Disease: Molecular Mechanisms and Strategies to Manage. Int J Mol Sci. 2020 Nov 7;21(21):8351. doi: 10.3390/ijms21218351.
View at Publisher | View at Google Scholar -
Singh R, Zogg H, Wei L, Bartlett A, Ghoshal UC, Rajender S, Ro S. Gut Microbial Dysbiosis in the Pathogenesis of Gastrointestinal Dysmotility and Metabolic Disorders. J Neurogastroenterol Motil. 2021 Jan 30;27(1):19-34. doi: 10.5056/jnm20149.
View at Publisher | View at Google Scholar -
Paradis T, Bègue H, Basmaciyan L, Dalle F, Bon F. Tight Junctions as a Key for Pathogens Invasion in Intestinal Epithelial Cells. Int J Mol Sci. 2021 Mar 2;22(5):2506. doi: 10.3390/ijms22052506.
View at Publisher | View at Google Scholar -
Stolfi C, Maresca C, Monteleone G, Laudisi F. Implication of Intestinal Barrier Dysfunction in Gut Dysbiosis and Diseases. Biomedicines. 2022 Jan 27;10(2):289. doi: 10.3390/biomedicines10020289.
View at Publisher | View at Google Scholar -
Bahitham W, Alghamdi S, Omer I, Alsudais A, Hakeem I, Alghamdi A, et al. Double Trouble: How Microbiome Dysbiosis and Mitochondrial Dysfunction Drive Non-Alcoholic Fatty Liver Disease and Non-Alcoholic Steatohepatitis. Biomedicines. 2024 Feb 29;12(3):550. doi: 10.3390/biomedicines12030550.
View at Publisher | View at Google Scholar -
Long C, Zhou X, Xia F, Zhou B. Intestinal Barrier Dysfunction and Gut Microbiota in Non-Alcoholic Fatty Liver Disease: Assessment, Mechanisms, and Therapeutic Considerations. Biology (Basel). 2024 Apr 6;13(4):243. doi: 10.3390/biology13040243.
View at Publisher | View at Google Scholar -
Wu J, Wang K, Wang X, Pang Y, Jiang C. The role of the gut microbiome and its metabolites in metabolic diseases. Protein Cell. 2021 May;12(5):360-373. doi: 10.1007/s13238-020-00814-7.
View at Publisher | View at Google Scholar -
Fujisaka S, Watanabe Y, Tobe K. The gut microbiome: a core regulator of metabolism. J Endocrinol. 2023 Jan 19;256(3):e220111. doi: 10.1530/JOE-22-0111.
View at Publisher | View at Google Scholar -
Wang Y, Xu H, Zhou X, Chen W, Zhou H. Dysregulated bile acid homeostasis: unveiling its role in metabolic diseases. Med Rev (2021). 2024 Jun 4;4(4):262-283. doi: 10.1515/mr-2024-0020.
View at Publisher | View at Google Scholar -
Larabi AB, Masson HLP, Bäumler AJ. Bile acids as modulators of gut microbiota composition and function. Gut Microbes. 2023 Jan-Dec;15(1):2172671. doi: 10.1080/19490976.2023.2172671.
View at Publisher | View at Google Scholar -
Staley C, Weingarden AR, Khoruts A, Sadowsky MJ. Interaction of gut microbiota with bile acid metabolism and its influence on disease states. Appl Microbiol Biotechnol. 2017 Jan;101(1):47-64. doi: 10.1007/s00253-016-8006-6.
View at Publisher | View at Google Scholar -
McMillan AS, Theriot CM. Bile acids impact the microbiota, host, and C. difficile dynamics providing insight into mechanisms of efficacy of FMTs and microbiota-focused therapeutics. Gut Microbes. 2024 Jan-Dec;16(1):2393766. doi: 10.1080/19490976.2024.2393766.
View at Publisher | View at Google Scholar -
McMillan AS, Foley MH, Perkins CE, Theriot CM. Loss of Bacteroides thetaiotaomicron bile acid-altering enzymes impacts bacterial fitness and the global metabolic transcriptome. Microbiol Spectr. 2024 Jan 11;12(1):e0357623. doi: 10.1128/spectrum.03576-23.
View at Publisher | View at Google Scholar -
Pathak P, Liu H, Boehme S, Xie C, Krausz KW, Gonzalez F, Chiang JYL. Farnesoid X receptor induces Takeda G-protein receptor 5 cross-talk to regulate bile acid synthesis and hepatic metabolism. J Biol Chem. 2017 Jun 30;292(26):11055-11069. doi: 10.1074/jbc.M117.784322.
View at Publisher | View at Google Scholar -
Chiang JYL, Ferrell JM. Bile acid receptors FXR and TGR5 signaling in fatty liver diseases and therapy. Am J Physiol Gastrointest Liver Physiol. 2020 Mar 1;318(3):G554-G573. doi: 10.1152/ajpgi.00223.2019.
View at Publisher | View at Google Scholar -
Iracheta-Vellve A, Calenda CD, Petrasek J, Ambade A, Kodys K, Adorini L, Szabo G. FXR and TGR5 Agonists Ameliorate Liver Injury, Steatosis, and Inflammation After Binge or Prolonged Alcohol Feeding in Mice. Hepatol Commun. 2018 Oct 15;2(11):1379-1391. doi: 10.1002/hep4.1256.
View at Publisher | View at Google Scholar -
Li Y, Jadhav K, Zhang Y. Bile acid receptors in non-alcoholic fatty liver disease. Biochem Pharmacol. 2013 Dec 1;86(11):1517-24. doi: 10.1016/j.bcp.2013.08.015.
View at Publisher | View at Google Scholar -
Khan A, Ding Z, Ishaq M, Bacha AS, Khan I, Hanif A, et al. Understanding the Effects of Gut Microbiota Dysbiosis on Nonalcoholic Fatty Liver Disease and the Possible Probiotics Role: Recent Updates. Int J Biol Sci. 2021 Feb 8;17(3):818-833. doi: 10.7150/ijbs.56214.
View at Publisher | View at Google Scholar -
Gao R, Meng X, Xue Y, Mao M, Liu Y, Tian X, et al. Bile acids-gut microbiota crosstalk contributes to the improvement of type 2 diabetes mellitus. Front Pharmacol. 2022 Oct 25;13:1027212. doi: 10.3389/fphar.2022.1027212.
View at Publisher | View at Google Scholar -
Radun R, Trauner M. Role of FXR in Bile Acid and Metabolic Homeostasis in NASH: Pathogenetic Concepts and Therapeutic Opportunities. Semin Liver Dis. 2021 Nov;41(4):461-475. doi: 10.1055/s-0041-1731707.
View at Publisher | View at Google Scholar -
Adorini L, Trauner M. FXR agonists in NASH treatment. J Hepatol. 2023 Nov;79(5):1317-1331. doi: 10.1016/j.jhep.2023.07.034.
View at Publisher | View at Google Scholar -
Safari Z, Gérard P. The links between the gut microbiome and non-alcoholic fatty liver disease (NAFLD). Cell Mol Life Sci. 2019 Apr;76(8):1541-1558. doi: 10.1007/s00018-019-03011-w.
View at Publisher | View at Google Scholar -
Kolodziejczyk AA, Zheng D, Shibolet O, Elinav E. The role of the microbiome in NAFLD and NASH. EMBO Mol Med. 2019 Feb;11(2):e9302. doi: 10.15252/emmm.201809302.
View at Publisher | View at Google Scholar -
Rosendo-Silva D, Viana S, Carvalho E, Reis F, Matafome P. Are gut dysbiosis, barrier disruption, and endotoxemia related to adipose tissue dysfunction in metabolic disorders? Overview of the mechanisms involved. Intern Emerg Med. 2023 Aug;18(5):1287-1302. doi: 10.1007/s11739-023-03262-3.
View at Publisher | View at Google Scholar -
Di Vincenzo F, Del Gaudio A, Petito V, Lopetuso LR, Scaldaferri F. Gut microbiota, intestinal permeability, and systemic inflammation: a narrative review. Intern Emerg Med. 2024 Mar;19(2):275-293. doi: 10.1007/s11739-023-03374-w.
View at Publisher | View at Google Scholar -
An L, Wirth U, Koch D, Schirren M, Drefs M, Koliogiannis D, et al. The Role of Gut-Derived Lipopolysaccharides and the Intestinal Barrier in Fatty Liver Diseases. J Gastrointest Surg. 2022 Mar;26(3):671-683. doi: 10.1007/s11605-021-05188-7.
View at Publisher | View at Google Scholar -
Ghosh S, Whitley CS, Haribabu B, Jala VR. Regulation of Intestinal Barrier Function by Microbial Metabolites. Cell Mol Gastroenterol Hepatol. 2021;11(5):1463-1482. doi: 10.1016/j.jcmgh.2021.02.007.
View at Publisher | View at Google Scholar -
Silva YP, Bernardi A, Frozza RL. The Role of Short-Chain Fatty Acids From Gut Microbiota in Gut-Brain Communication. Front Endocrinol (Lausanne). 2020 Jan 31;11:25. doi: 10.3389/fendo.2020.00025.
View at Publisher | View at Google Scholar -
Fusco W, Lorenzo MB, Cintoni M, Porcari S, Rinninella E, Kaitsas F, et al. Short-Chain Fatty-Acid-Producing Bacteria: Key Components of the Human Gut Microbiota. Nutrients. 2023 May 6;15(9):2211. doi: 10.3390/nu15092211.
View at Publisher | View at Google Scholar -
Nogal A, Valdes AM, Menni C. The role of short-chain fatty acids in the interplay between gut microbiota and diet in cardio-metabolic health. Gut Microbes. 2021 Jan-Dec;13(1):1-24. doi: 10.1080/19490976.2021.1897212.
View at Publisher | View at Google Scholar -
Vinelli V, Biscotti P, Martini D, Del Bo' C, Marino M, Meroño T, et al. Effects of Dietary Fibers on Short-Chain Fatty Acids and Gut Microbiota Composition in Healthy Adults: A Systematic Review. Nutrients. 2022 Jun 21;14(13):2559. doi: 10.3390/nu14132559.
View at Publisher | View at Google Scholar -
Chen J, Vitetta L. The Role of Butyrate in Attenuating Pathobiont-Induced Hyperinflammation. Immune Netw. 2020 Feb 4;20(2):e15. doi: 10.4110/in.2020.20.e15.
View at Publisher | View at Google Scholar -
Coccia C, Bonomi F, Lo Cricchio A, Russo E, Peretti S, Bandini G, et al. The Potential Role of Butyrate in the Pathogenesis and Treatment of Autoimmune Rheumatic Diseases. Biomedicines. 2024 Aug 5;12(8):1760. doi: 10.3390/biomedicines12081760.
View at Publisher | View at Google Scholar -
Morrison DJ, Preston T. Formation of short chain fatty acids by the gut microbiota and their impact on human metabolism. Gut Microbes. 2016 May 3;7(3):189-200. doi: 10.1080/19490976.2015.1134082.
View at Publisher | View at Google Scholar -
Lange O, Proczko-Stepaniak M, Mika A. Short-Chain Fatty Acids-A Product of the Microbiome and Its Participation in Two-Way Communication on the Microbiome-Host Mammal Line. Curr Obes Rep. 2023 Jun;12(2):108-126. doi: 10.1007/s13679-023-00503-6.
View at Publisher | View at Google Scholar -
Nakadate K, Saitoh H, Sakaguchi M, Miruno F, Muramatsu N, Ito N, et al. Advances in Understanding Lipopolysaccharide-Mediated Hepatitis: Mechanisms and Pathological Features. Curr Issues Mol Biol. 2025 Jan 27;47(2):79. doi: 10.3390/cimb47020079.
View at Publisher | View at Google Scholar -
Xie D, Ouyang S. The role and mechanisms of macrophage polarization and hepatocyte pyroptosis in acute liver failure. Front Immunol. 2023 Oct 26;14:1279264. doi: 10.3389/fimmu.2023.1279264.
View at Publisher | View at Google Scholar -
Khanmohammadi S, Kuchay MS. Toll-like receptors and metabolic (dysfunction)-associated fatty liver disease. Pharmacol Res. 2022 Nov;185:106507. doi: 10.1016/j.phrs.2022.106507.
View at Publisher | View at Google Scholar -
Markowiak P, Śliżewska K. Effects of Probiotics, Prebiotics, and Synbiotics on Human Health. Nutrients. 2017 Sep 15;9(9):1021. doi: 10.3390/nu9091021.
View at Publisher | View at Google Scholar -
Zhou P, Chen C, Patil S, Dong S. Unveiling the therapeutic symphony of probiotics, prebiotics, and postbiotics in gut-immune harmony. Front Nutr. 2024 Feb 8;11:1355542. doi: 10.3389/fnut.2024.1355542.
View at Publisher | View at Google Scholar -
Lonardo A, Arab JP, Arrese M. Perspectives on Precision Medicine Approaches to NAFLD Diagnosis and Management. Adv Ther. 2021 May;38(5):2130-2158. doi: 10.1007/s12325-021-01690-1.
View at Publisher | View at Google Scholar -
Valenzuela-Vallejo L, Sanoudou D, Mantzoros CS. Precision Medicine in Fatty Liver Disease/Non-Alcoholic Fatty Liver Disease. J Pers Med. 2023 May 14;13(5):830. doi: 10.3390/jpm13050830.
View at Publisher | View at Google Scholar -
Valenzuela-Vallejo L, Sanoudou D, Mantzoros CS. Precision Medicine in Fatty Liver Disease/Non-Alcoholic Fatty Liver Disease. J Pers Med. 2023 May 14;13(5):830. doi: 10.3390/jpm13050830.
View at Publisher | View at Google Scholar -
Arechederra M, Recalde M, Gárate-Rascón M, Fernández-Barrena MG, Ávila MA, Berasain C. Epigenetic Biomarkers for the Diagnosis and Treatment of Liver Disease. Cancers (Basel). 2021 Mar 12;13(6):1265. doi: 10.3390/cancers13061265.
View at Publisher | View at Google Scholar -
Gîlcă-Blanariu GE, Budur DS, Mitrică DE, Gologan E, Timofte O, Bălan GG, et al. Advances in Noninvasive Biomarkers for Nonalcoholic Fatty Liver Disease. Metabolites. 2023 Oct 29;13(11):1115. doi: 10.3390/metabo13111115.
View at Publisher | View at Google Scholar -
Decharatanachart P, Chaiteerakij R, Tiyarattanachai T, Treeprasertsuk S. Application of artificial intelligence in non-alcoholic fatty liver disease and liver fibrosis: a systematic review and meta-analysis. Therap Adv Gastroenterol. 2021 Dec 21;14:17562848211062807. doi: 10.1177/17562848211062807.
View at Publisher | View at Google Scholar -
Alshagathrh FM, Househ MS. Artificial Intelligence for Detecting and Quantifying Fatty Liver in Ultrasound Images: A Systematic Review. Bioengineering (Basel). 2022 Dec 1;9(12):748. doi: 10.3390/bioengineering9120748.
View at Publisher | View at Google Scholar -
Kaushal K. Non-alcoholic fatty liver disease (NAFLD) integration into India's NCD program - Obstacles and solutions for the implementation of guidelines at the national level. J Family Med Prim Care. 2024 Sep;13(9):3536-3539. doi: 10.4103/jfmpc.jfmpc_458_24.
View at Publisher | View at Google Scholar -
Lazarus JV, Mark HE, Anstee QM, Arab JP, Batterham RL, Castera L, et al. Advancing the global public health agenda for NAFLD: a consensus statement. Nat Rev Gastroenterol Hepatol. 2022 Jan;19(1):60-78. doi: 10.1038/s41575-021-00523-4.
View at Publisher | View at Google Scholar -
Schattenberg JM, Allen AM, Jarvis H, Zelber-Sagi S, Cusi K, Dillon JF, et al. A multistakeholder approach to innovations in NAFLD care. Commun Med (Lond). 2023 Jan 3;3(1):1. doi: 10.1038/s43856-022-00228-y.
View at Publisher | View at Google Scholar