Next Article in Journal
MicroRNAs as Endocrine Modulators of Breast Cancer
Previous Article in Journal
Genome-Wide Identification and Expression Analysis of Thionin Family in Rice (Oryza sativa) and Functional Characterization of OsTHION15 in Drought Stress and ABA Stress
Previous Article in Special Issue
The Microbiome and Metabolic Dysfunction-Associated Steatotic Liver Disease
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Hidden in the Fat: Unpacking the Metabolic Tango Between Metabolic Dysfunction-Associated Steatotic Liver Disease and Metabolic Syndrome

1
Internal Medicine Department, Galilee Medical Centre, Nahariya 221001, Israel
2
The Azrieli Faculty of Medicine, Bar-Ilan University, Safed 1311502, Israel
*
Author to whom correspondence should be addressed.
Int. J. Mol. Sci. 2025, 26(7), 3448; https://doi.org/10.3390/ijms26073448
Submission received: 7 February 2025 / Revised: 25 March 2025 / Accepted: 30 March 2025 / Published: 7 April 2025

Abstract

:
Metabolic syndrome (MetS) and metabolic dysfunction-associated steatotic liver disease (MASLD) are closely related, with rapidly increasing prevalence globally, driving significant public health concerns. Both conditions share common pathophysiological mechanisms such as insulin resistance (IR), adipose tissue dysfunction, oxidative stress, and gut microbiota dysbiosis, which contribute to their co-occurrence and progression. While the clinical implications of this overlap, including increased cardiovascular, renal, and hepatic risk, are well recognized, current diagnostic and therapeutic approaches remain insufficient due to the clinical and individuals’ heterogeneity and complexity of these diseases. This review aims to provide an in-depth exploration of the molecular mechanisms linking MetS and MASLD, identify critical gaps in our understanding, and highlight existing challenges in early detection and treatment. Despite advancements in biomarkers and therapeutic interventions, the need for a comprehensive, integrated approach remains. The review also discusses emerging therapies targeting specific pathways, the potential of precision medicine, and the growing role of artificial intelligence in enhancing research and clinical management. Future research is urgently needed to combine multi-omics data, precision medicine, and novel biomarkers to better understand the complex interactions between MetS and MASLD. Collaborative, multidisciplinary efforts are essential to develop more effective diagnostic tools and therapies to address these diseases on a global scale.

1. Introduction

One of the most important global health issues facing modern medicine is the changing landscape of MASLD and MetS [1,2]. Several studies suggest differences in the prevalence and severity of MASLD by race or ethnicity [3,4,5,6]. According to recent epidemiological data, prevalence varies significantly by geography, with figures ranging from 29.6 percent to 32.2 percent in the Asia–Pacific region, 37.3 percent in North America, and 32.8 percent in Europe, and the highest prevalence of 44.37% was documented among Latin America [7,8]. This distribution pattern illustrates the intricate relationships that exist between lifestyle choices, socioeconomic determinants, and genetic predisposition [9]. According to recent meta-analyses, the global prevalence of the disease is expected to rise by 47% by 2030, with annual incidence rates of 2–8% in Asian populations and 6–9% in Western nations [10,11].
Pro-inflammatory mediators, altered adipokine profiles, and disruption of metabolic homeostasis are all examples of the complex bidirectional pathways that make up the pathophysiological relationship between MetS and MASLD [12,13]. The oxidative stress pathway and IR are two mechanistic paradigms that best illustrate this relationship [14]. Adipose tissue dysfunction sets off a series of events that result in hepatic lipid accumulation and inflammatory activation. It is characterized by dysregulated adipokine secretion and increased lipolysis [15,16]. Furthermore, hepatic steatosis creates a self-reinforcing cycle of metabolic degradation by sustaining systemic metabolic abnormalities via a variety of mechanisms [17,18].
Genetic factors also play an important role in the development of MASLD and can explain the variability in the natural history of MASLD. In studies involving family clusters, one fifth of metabolic dysfunction-associated steatohepatitis (MASH) patients had a similar first-degree relative [19]. Another study reported that seven of the eight families had MASH and cryptogenic cirrhosis [20]. These hypotheses about genetic factors that contribute to MASLD may explain the development and progression of MASLD in lean patients, although the mechanism is still poorly understood in this population. One study reported a higher prevalence of IR in thin Asian men compared to Hispanic, black, and white men [21]. The Genome-Wide-Association Study (GWAS) identified 17 MASLD-associated genes, such as torsin family 1 member B (TOR1B), fat mass and obesity-associated (FTO) gene, growth factor receptor-bound protein 14 (GRB14), insulin receptor (INSR), cordon-bleu WH2 repeat protein like 1 (COBLL1), sterol regulatory element-binding transcription factor 1 (SREBF1), and patatin-like phospholipase domain-containing protein 2 (PNPLA2) [22].
Existing diagnostic approaches often detect these diseases in an advanced stage, which highlights the need for preventive strategies [23]. With new developments in metabolomics, proteomics, and imaging techniques offering promising routes to early detection, the development of new biomarkers and more sensitive diagnostic tools is a major unmet need [24]. Integrating artificial intelligence with traditional diagnostic methods has opened up new possibilities for stratifying risk and diagnosing rare diseases [25].
In view of the rapidly evolving understanding of these interrelated conditions and their significant impact on global health, this review aims to examine in greater detail the molecular mechanisms behind the two-way relationship between METS and MASLD, with a focus on new pathophysiological pathways and clinical implications. It will also explore possible therapeutic targets based on our growing understanding of the common pathophysiology of these conditions. This compilation of the latest data will provide researchers and clinicians with a modern framework for understanding how diseases progress and will guide the development of more effective treatment options.

2. Change of Nomenclature

There has been a substantial change in the way the medical community views these conditions with the changing of the term “NAFLD” to “metabolic-dysfunction associated liver disease” (MASLD). In 2020, a global consensus panel suggested changing the name of “NAFLD” to “MASLD” to more accurately represent the disease’s intricate metabolic foundations [26]. By recognizing the importance of IR, obesity, type 2 diabetes, and other metabolic abnormalities in the pathophysiology of fatty liver disease, the new nomenclature highlights the connection between liver disease and metabolic dysfunction [1,27]. The change was prompted by the realization that the term “non-alcoholic” is restrictive because it fails to encompass the wider range of metabolic elements that contribute to the disease. MASLD enables a broader definition that takes into consideration the metabolic environment in which liver steatosis arises and advances.
The reclassification of non-alcoholic steatohepatitis (NASH) to metabolic dysfunction-associated steatohepatitis (MASH) followed, together with the change to MASLD. The switch to MASH highlights the growing understanding that hepatocyte damage and inflammation in steatosis reflect complex interactions between metabolic disorders such as oxidative stress, IR, and adipose tissue dysfunction rather than being the sole consequence of fat accumulation. The purpose of coining the term MASH was to identify metabolic factors that contribute to the progression from simple steatosis to more severe liver diseases such as fibrosis and cirrhosis, and to move away from the restrictive label of alcoholics only. With the official adoption of these changes to the nomenclature in 2020, our understanding of liver disease is changing.

3. Underlying Mechanisms

3.1. Insulin Resistance: The Central Pathway

IR serves as a cornerstone of both MetS and MASLD, linking these two conditions through multiple interconnected metabolic pathways. In MetS, IR results in a diminished cellular response to insulin, particularly in adipose tissue, muscle, and the liver. This impaired response drives a cascade of metabolic disruptions that promote the development and progression to MASLD [28].
At the hepatic level, IR is a key driver of hepatic steatosis, which is the hallmark of MASLD. Normally, insulin regulates hepatic glucose production and promotes lipid storage through pathways such as de novo lipogenesis. In the insulin-resistant state, these processes become dysregulated, resulting in several pathological changes in the liver. One of the key processes affected is de novo lipogenesis, where chronic hyperinsulinemia stimulates the liver to convert excess carbohydrates into fatty acids. This process is mediated by the upregulation of the sterol regulatory element-binding protein 1c (SREBP-1c) pathway, a transcription factor that promotes fatty acid synthesis [29]. The overproduction of triglycerides, driven by this mechanism, leads to their accumulation in hepatocytes, contributing to hepatic steatosis.
Additionally, IR impairs the normal suppression of lipolysis in adipose tissue. Under normal conditions, insulin inhibits the release of free fatty acids (FFAs) from adipose stores. However, in the context of IR, this suppression is impaired, leading to an increased flux of FFAs into the circulation. These excess FFAs are taken up by the liver, where they further contribute to triglyceride accumulation and exacerbate hepatic steatosis [30]. Furthermore, IR reduces fatty acid β-oxidation in hepatocytes, a process critical for breaking down fatty acids to energy production. This reduction is partly due to the dysregulation of peroxisome proliferator-activated receptor-α (PPAR-α), a nuclear receptor responsible for promoting fatty acid oxidation [31]. The diminished capacity of oxidation leads to the accumulation of lipotoxic intermediates, which further drive hepatocyte injury and inflammation.
In addition to promoting hepatic fat accumulation, IR contributes to a pro-inflammatory state in the liver. This is compounded by the secretion of hepatokines, liver-derived proteins such as fetuin-A, which impair insulin signaling in peripheral tissues and promote systemic inflammation [32]. Fetuin-A, for instance, disrupts insulin receptor signaling in muscle and adipose tissue, creating a feedback loop of worsening IR and metabolic dysfunction.
Moreover, IR is associated with endoplasmic reticulum (ER) stress and oxidative stress, both of which play key roles in the progression to MASLD. The chronic overnutrition and lipid overload, both hallmark features of MetS, compromise the liver’s metabolic capacity, resulting to ER dysfunction and the accumulation of reactive oxygen species (ROS) [33].
These processes contribute to hepatocyte injury and promote progression from simple steatosis to more severe stages of liver disease, such as MASH and cirrhosis. IR is therefore the primary mechanism by which MetS induces MASLD development and progression.

3.2. Adipose Tissue Dysfunction and Pro-Inflammatory State

Adipose tissue dysfunction is a hallmark of MetS and plays a critical role in the development of MASLD by promoting both IR and systemic inflammation. In healthy metabolic states, adipose tissue serves as an energy reservoir and a dynamic endocrine organ, releasing a range of adipokines that regulate insulin sensitivity, lipid metabolism, and inflammatory responses. However, in the context of MetS, excess adiposity leads to significant alterations in the secretion of these adipokines, creating a pro-inflammatory and insulin-resistant environment that directly impacts the liver.
One of the key alterations in adipose tissue dysfunction is the imbalance between anti-inflammatory and pro-inflammatory adipokines. In MetS, there is typically a reduction in adiponectin, an adipokine that possesses insulin-sensitizing, anti-inflammatory, and antifibrotic properties. Reduced adiponectin levels are strongly associated with increased hepatic fat accumulation and inflammation, contributing to the development of MASLD. In contrast, pro-inflammatory adipokines such as leptin, tumor necrosis factor-α (TNF-α), and interleukin-6 (IL-6) are elevated in individuals with MetS. These cytokines are known to promote systemic and hepatic IR, as well as hepatic inflammation, both of which are key drivers to MASLD onset and progression [34,35].
Leptin, although primarily involved in regulating appetite and energy homeostasis, becomes dysregulated in the context of obesity and MetS. Elevated leptin levels, coupled with the development of leptin resistance, impair the normal metabolic functions of this hormone. Leptin resistance contributes to ectopic lipid accumulation in organs such as the liver, thereby exacerbating hepatic steatosis and inflammation. This leptin dysfunction further feeds into the systemic inflammatory milieu that characterizes MetS, contributing to the worsening of IR [36].
Adipose tissue dysfunction also plays a role in the infiltration of immune cells, particularly macrophages, into adipose depots. These immune cells contribute to the secretion of pro-inflammatory cytokines that exacerbate systemic inflammation and liver injury. In obesity and MetS, macrophages undergo a phenotypic switch to a more pro-inflammatory, M1-like state, characterized by the production of elevated levels of TNF-α and IL-6. These cytokines not only impair insulin signaling but also promote the activation of inflammatory pathways in the liver, leading to hepatocyte injury, steatosis, and the progression to MASH [32,33].
Moreover, excess adipose tissue, particularly visceral adiposity, contributes to increased FFA release into the circulation due to impaired suppression of lipolysis by insulin. The elevated levels of circulating FFAs are taken up by the liver, where they serve as substrates for triglyceride synthesis, leading to hepatic steatosis. This excess FFA influx also exacerbates hepatic IR, further driving fat accumulation and inflammation in the liver. The combination of these factors—altered adipokine secretion, immune cell infiltration, and increased FFA release—creates a chronic pro-inflammatory state that fosters the progression of MASLD [31].
In summary, the dysfunction of adipose tissue in MetS plays a pivotal role in the pathogenesis of MASLD by promoting systemic inflammation, IR, and driving hepatic fat accumulation. This pro-inflammatory environment not only contributes to hepatic steatosis but also facilitates the progression of liver disease toward more severe forms, including MASH and fibrosis.

3.3. ER Stress and Oxidative Stress: Amplifying Liver Injury

In the context of MetS and MASLD, ER stress and oxidative stress play key roles in amplifying liver injury and promoting the progression from simple steatosis to MASH. Both processes are tightly linked to the metabolic dysregulation seen in MetS, contributing to hepatocyte dysfunction, inflammation, and fibrosis.
When ER’s folding capacity is overwhelmed due to excessive nutrient load, lipid accumulation, ER stress occurs which leads to ER lumen accumulation of misfolded or unfolded proteins, triggering the unfolded protein response (UPR). While the latter is protective initially with the aim to restore normal ER function, prolonged or unresolved ER stress can lead to apoptosis and inflammation. In MetS, lipid overload and chronic overnutrition overwhelm the liver’s ability to maintain protein homeostasis leading to subsequent hepatocyte injury [32].
In parallel with ER stress, oxidative stress plays a significant role in the pathogenesis of MASLD. Oxidative stress occurs when the production of ROS exceeds the liver’s antioxidant defenses, leading to cellular damage. In MetS, the increased flux of free FFAs to the liver, coupled with impaired fatty acid oxidation, results in excessive β-oxidation in mitochondria. This overloads the mitochondrial electron transport chain, leading to the excessive production of ROS, particularly superoxide anions [37].
These ROS not only directly damage cellular components, including lipids, proteins, and Deoxyribonucleic acid (DNA), but also activate inflammatory pathways. Oxidative stress has been shown to activate the NACHT, LRR, and PYD domain-containing protein 3 (NLRP3) inflammasome, a multiprotein complex that promotes the production of pro-inflammatory cytokines such as interleukin-1β (IL-1β) and interleukin 18 (IL-18) [33]. The activation of the NLRP3 inflammasome contributes to the progression of simple steatosis to MASH, as it fosters a pro-inflammatory environment that perpetuates hepatocyte injury and fibrosis.
Moreover, oxidative stress and ER stress are mutually reinforcing processes. The accumulation of misfolded proteins in the ER can trigger oxidative stress by disturbing calcium homeostasis and increasing ROS production. Conversely, oxidative damage to proteins and lipids can further impair ER function, exacerbating the unfolded protein response and driving further hepatocyte injury [38]. This interplay between ER stress and oxidative stress represents a critical mechanism underlying the progression of MASLD, as it leads to both metabolic dysfunction and chronic liver inflammation.
In summary, both ER stress and oxidative stress are key amplifiers of liver injury in MetS and MASLD. These processes not only exacerbate IR and lipid accumulation but also drive inflammation and hepatocyte apoptosis, contributing to the progression from simple steatosis to MASH and fibrosis. Addressing these stress responses offers potential therapeutic avenues for mitigating the liver damage associated with MASLD.

3.4. Gut Microbiota Dysbiosis and Metabolic-Endotoxemia

The gut microbiota has emerged as a key player in the pathogenesis of both MetS and MASLD. Dysbiosis, or the imbalance in gut microbial composition, is linked to metabolic disturbances that characterize MetS, including IR, obesity, and chronic inflammation. In recent years, evidence has increasingly supported the notion that alterations in the gut microbiota contribute to the development of MASLD by promoting hepatic steatosis, inflammation, and fibrosis.
One of the primary mechanisms by which gut dysbiosis drives liver pathology is through increased intestinal permeability, often referred to as “leaky gut”. In healthy individuals, the intestinal barrier limits the translocation of bacterial products into the circulation. However, in individuals with MetS, alterations in the gut microbiota compromise the integrity of the intestinal barrier, allowing bacterial endotoxins such as lipopolysaccharides (LPS) to enter the portal circulation. This process, known as metabolic endotoxemia, plays a central role in the development of MASLD [39].
LPS, a component of the outer membrane of Gram-negative bacteria, is a potent activator of the innate immune system. Once translocated into the liver, LPS binds to toll-like receptor 4 (TLR4) on Kupffer cells (KCs) and hepatocytes, triggering the production of pro-inflammatory cytokines such as TNF-α and IL-6. This inflammatory response promotes hepatic IR and contributes to hepatocyte injury, steatosis, and fibrosis [40]. Additionally, metabolic endotoxemia perpetuates a cycle of chronic, low-grade inflammation that exacerbates the metabolic disturbances associated with MetS.
Beyond LPS, gut dysbiosis also impacts the metabolism of BAs, which are crucial regulators of lipid and glucose homeostasis. The gut microbiota plays a key role in transformation and deconjugation of BAs, by affecting the signaling through receptors such as the Farnesoid X receptor (FXR) and G protein-coupled bile acid receptor 1 (TGR5). Dysbiosis causing alterations in the BA pool might lead to impaired lipid metabolism and increased hepatic fat accumulation. In the setting of MASLD, disrupted BA signaling contributes to IR, hepatic steatosis, and inflammation [41].
Moreover, the gut microbiota produces a variety of microbial metabolites that can influence host metabolism. Short-chain fatty acids (SCFAs), which are produced by the fermentation of dietary fiber, have been shown to enhance insulin sensitivity and exert anti-inflammatory effects. However, in the context of dysbiosis, the production of SCFAs may be reduced, contributing to metabolic dysfunction and inflammation. Conversely, other microbial metabolites, such as imidazole propionate, have been shown to impair insulin signaling and promote IR, further linking gut dysbiosis to the pathogenesis of MetS and MASLD [32,42].
In summary, gut microbiota dysbiosis plays a vital role in the development and progression of MASLD through mechanisms involving metabolic endotoxemia, BA dysregulation, and altered microbial metabolite production. These factors contribute to hepatic steatosis, IR, and inflammation, highlighting the gut–liver axis as a potential therapeutic target in the management of MASLD.

3.5. Innate Immune Response and Hepatic Inflammation

The innate immune response plays a significant role in the pathogenesis of MASLD, particularly in driving hepatic inflammation and promoting the progression from simple steatosis to MASH. In individuals with MetS, the liver becomes a key site of immune cell activation, leading to a chronic inflammatory state that exacerbates IR, hepatocyte injury, and fibrosis.
At the forefront of the innate immune response in the liver are Kupffer cells, the liver’s resident macrophages. In a healthy liver, KCs maintain tissue homeostasis by clearing debris and apoptotic cells while preventing excessive immune activation. However, in the context of MetS and MASLD, KCs become activated in response to metabolic stressors, such as FFAs and microbial products translocated from the gut. This activation leads to the production of pro-inflammatory cytokines, such as TNF-α, IL-1β, and IL-6, that promote hepatic inflammation and IR [33,42].
The activation of KCs is also influenced by LPS, bacterial endotoxins that enter the liver via the portal circulation during a metabolic endotoxemia state. LPS binds to TLR4 on KCs and other hepatic immune cells, triggering downstream signaling pathways that enhance the production of inflammatory cytokines. This LPS-mediated activation of TLR4 contributes to the amplification of hepatic inflammation in MASLD and fosters a microenvironment conducive for the progression of steatosis to MASH [39].
In addition to Kupffer cells, infiltrating monocytes and macrophages play a crucial role in the innate immune response in the liver. In response to signals from damaged hepatocytes and adipose tissue, monocytes are recruited to the liver, where they differentiate into pro-inflammatory macrophages. These macrophages, often referred to as M1-like macrophages, produce elevated levels of pro-inflammatory cytokines and contribute to the ongoing cycle of inflammation and liver damage. The accumulation of M1-like macrophages in the liver is a hallmark of MASH and is closely linked to the progression of liver fibrosis [32,40].
In summary, the innate immune response, driven by Kupffer cells, macrophages, and the NLRP3 inflammasome, plays a pivotal role in the pathogenesis of MASLD. Through the production of pro-inflammatory cytokines and the activation of fibrotic pathways, innate immune cells contribute to the progression of liver disease from simple steatosis to MASH and fibrosis. Targeting these immune pathways represents a potential therapeutic strategy for halting the progression of MASLD.

3.6. Fibrosis Pathways and Liver Disease Progression

Fibrosis is a hallmark of the progression of MASLD from simple steatosis to more advanced stages of liver disease, MASH, advanced fibrosis, and cirrhosis. The development to fibrosis is driven by chronic inflammation and metabolic dysregulation, both of which are characteristic features of MetS. In MASLD, fibrosis occurs because of the excessive deposition of extracellular matrix (ECM) components, including collagen, which leads to scarring and architectural distortion of the liver. This process is orchestrated by hepatic stellate cells (HSCs), which become activated in response to injury signals from damaged hepatocytes, immune cells, and the metabolic environment.
In healthy liver tissue, HSCs remain in a quiescent state and function primarily in vitamin A storage. However, in the setting of MASLD, persistent metabolic stress, lipid accumulation, and chronic inflammation lead to the activation of HSCs. Once activated, HSCs transform into myofibroblast-like cells that secrete substantial amounts of ECM proteins, including collagen types I and III, which contribute to the development of fibrotic tissue. This activation process is mediated by a variety of signaling molecules, including transforming growth factor-β (TGF-β), which is the most potent profibrogenic cytokine in the liver [33]. TGF-β is secreted by Kupffer cells, activated HSCs, and infiltrating immune cells in response to hepatic injury. Through the TGF-β/Smad signaling pathway, HSCs are stimulated to proliferate and synthesize ECM components, leading to progressive fibrosis.
In addition to TGF-β, other growth factors and cytokines contribute to the activation of HSCs and the fibrotic process. Connective tissue growth factor (CTGF), also known as Cellular communication network 2 (CCN2), plays a significant role in amplifying the fibrogenic response in the liver. CTGF is upregulated in response to TGF-β and other pro-fibrotic stimuli and enhances the production of collagen and other ECM proteins by HSCs [38]. Furthermore, platelet-derived growth factor (PDGF), which is released by platelets, Kupffer cells, and damaged hepatocytes, acts as a potent mitogen for HSCs, stimulating their proliferation and migration within the liver. Together, these signaling pathways create a pro-fibrogenic environment that promotes the accumulation of scar tissue and the progression of MASLD to cirrhosis.
The progression of fibrosis in MASLD is a major determinant of patient outcomes, as advanced fibrosis is associated with an increased risk of liver-related complications, including hepatocellular carcinoma (HCC), portal hypertension, and liver failure [43]. Studies have shown that patients with advanced fibrosis, particularly those with bridging fibrosis or cirrhosis, present significantly higher mortality rates compared to those with mild or no fibrosis [44]. As such, targeting the fibrotic pathways involved in HSC activation and ECM deposition represents a key therapeutic strategy for preventing the progression of MASLD.
To summarize, fibrosis in MASLD is driven by a complex interplay of metabolic dysregulation, chronic inflammation, and the activation of pro-fibrogenic signaling pathways. TGF-β and CTGF are central mediators of HSC activation and ECM deposition, while chronic inflammation perpetuates the fibrotic response. Understanding these mechanisms is crucial for the development of effective therapies aimed at halting or reversing fibrosis in MASLD.
In summary, MASLD and MetS are tightly linked by IR, which disrupts hepatic glucose/lipid metabolism, leading to fat accumulation and a pro-inflammatory state amplified by adipose tissue dysfunction, ER/oxidative stress, and gut dysbiosis. These factors activate the innate immune response in the liver, resulting in fibrosis, the key process in MASLD progression to severe liver disease (Figure 1).

4. Extrahepatic Co-Morbidities

Beyond hepatic symptoms, MetS and MASLD represent a substantial health burden with extensive systemic ramifications. The combinations of these disorders form a complex and vicious environment that impacts the cardiovascular, renal, endocrine, and immune systems, among other organ systems. Comprehending these systemic effects is essential for both optimal clinical outcomes and comprehensive patient care [45,46].

4.1. Cardiovascular Morbidity and Mortality

Cardiovascular disease (CVD) is still the world’s leading cause of death, with metabolic syndrome being strongly associated with an increased risk. Cardiovascular outcomes are doubled in patients with MetS, and all-cause mortality is increased by one to five times [47].
This risk is increased further when MASLD is present. The Targher et al. meta-analysis reported that regardless of conventional cardiovascular risk factors or the elements of MetS, MASLD was linked to a 64% higher risk of fatal and/or non-fatal CVD events (2016) [47,48]. Even after controlling for changes in Body Mass Index (BMI), progression of cardiovascular risk factors was linked to rising hepatic fat over a 6-year period in a longitudinal cohort [49]. Additionally, in a prospective research study, Basheer M et al. concluded that a strong independent risk factor for coronary artery disease (CAD) and carotid atherosclerosis is liver fat storage more than visceral fat storage [50]. Nevertheless, epicardial adipose tissue (EAT) was found to be significantly higher in MASLD patients than in controls in a meta-analysis involving 13 case–control studies (n = 2260 patients). Moreover, EAT was associated with the degree of atherosclerotic CVD and liver steatosis and fibrosis [51]. Nonetheless, it is the visceral fat depot of the heart that produces both pro-inflammatory and anti-inflammatory mediators [52]. According to one study involving 868 participants, an increased incidence of extra-cardiac plaques was associated with hepatic steatosis and epicardial fat thickness [53].
MASLD increases the risk of CVD through a number of mechanisms, including systemic low-grade inflammation, adhesion molecules, hepatic IR, and a prothrombotic state [54]. A notable reduction in brachial artery endothelial flow-mediated vasodilatation is another indication of endothelial dysfunction in people with MASLD [55]. Additionally, through the liver’s secreted proteins, including IL-6, CRP, fibrinogen, Monocyte chemoattractant protein-1 (MCP-1), TNF-α, β-trophin, and fetuin-A, MASH may contribute to systemic low-grade inflammation and cardio-metabolic disease [56].
In conclusion, metabolic disorders like obesity, dyslipidemia, hypertension, and diabetes are strongly associated with MASLD and MASH. An increased risk of CVD is linked to the existence and severity of MASLD, specifically MASH and fibrosis. However, the coexistence of type 2 diabetes (T2D) or a metabolic syndrome is necessary for this association to exist. Significantly, MASLD raises the risk of arrhythmia, cardiomyopathy, and atherosclerosis on its own, which may lead to cardiovascular morbidity and death [57]. Given the strong correlation between obesity and cardio-metabolic disorders, diet and exercise-based lifestyle interventions to promote weight loss are currently the gold standard for treating these conditions.

4.2. Progression of Liver Disease

The development of hepatic steatosis into MASH is strongly associated with the level of metabolic dysfunction. Hepatocellular ballooning, inflammatory infiltration, and progressive fibrosis are some of the changes that have occurred [44]. Clinical evidence indicates that patients with metabolic syndrome exhibit an accelerated disease trajectory, with type 2 diabetes mellitus associated with a 1.5–2.0-fold increase in fibrosis progression rate compared to non-diabetic individuals [58]. The combined impact of several metabolic risk factors also shows synergistic potential in the progression of the disease, increasing the risk of liver-related mortality, cirrhosis, and hepatocellular carcinoma [59].
Multiple pathogenic mechanisms beyond simple fat accumulation are involved in the progression of liver disease in MASLD [60]. In hepatocytes, lipotoxicity, which is caused by the buildup of toxic lipid species, sets off cellular stress reactions and death pathways [61]. Chronic inflammation and tissue damage result from this process activation of inflammatory cascades and encouragement of immune cell recruitment [62]. In addition to causing significant alterations in the intestinal barrier function and the gut microbiota’s composition, MASLD also raises endotoxemia and bacterial translocation, both of which exacerbate hepatic inflammation and fibrosis [63].
The progression of MASLD to advanced liver disease is characterized by molecular signatures and distinct histopathological changes [64]. Hepatocyte ballooning, progressive fibrosis, and inflammatory cell infiltration become noticeable characteristics as the disease progresses, although simple steatosis is present in the initial stages. The progression from MASLD to MASH and ultimately HCC is influenced by a number of factors, such as oxidative stress, mitochondrial dysfunction, cellular senescence, and genetic predisposition (PNPLA3 and Transmembrane 6 superfamily member 2 (TM6SF2) variants) [65]. Hepatocyte death and the recruitment of inflammatory cells are also influenced by altered hepatic insulin signaling and chronic endoplasmic reticulum stress [60]. The accumulation of genetic changes in an inflammatory milieu, along with the aberrant activation of oncogenic pathways, specifically Phosphatidylinositol-4,5-bisphosphate 3-kinase (PI3K)/protein kinase B (Akt) PI3K/AKT and mammalian target of rapamycin mTOR signaling, further promotes the progression to HCC [66]. Influenced by both environmental and genetic factors, the rate of progression varies significantly among individuals [67]. About 20% of MASLD patients, according to recent data, progress to MASH, and of these, up to 20% may develop advanced fibrosis in a comparatively short period of time—two to three years—with an annual incidence of 2–3 percent for HCC in patients with cirrhosis related to MASH [9,68].
In summary, a harmful cycle of increasing severity is established by the reciprocal relationship between metabolic syndrome and the progression of hepatic disease. Hepatic injury is accelerated by systemic IR and metabolic disturbances that are made worse by progressive liver dysfunction.

4.3. Type 2 Diabetes Mellitus

New definitions were developed through international consensus to better characterize the complex interrelationships between MetS, MASLD, and their combined impact on T2D. This presents a significant challenge in modern medicine [45]. These conditions have been shown to have a synergistic effect that significantly affects glycemic control, the course of the disease, and the results of treatment for diabetic patients in meta-analyses involving over 500,000 adults [69].
Extensive clinical studies have shown that the primary manifestation of the impact on glycemic control is impaired hepatic glucose regulation [70]. Recent studies have demonstrated that the liver’s reduced response to insulin causes increased hepatic glucose output despite hyperglycemia, impaired glycogen storage, and altered glucose homeostasis. MASLD significantly impairs hepatic insulin sensitivity, resulting in dysregulated glucose production and altered metabolic pathways [71]. MetS compounds these effects through adipose tissue dysfunction, increased inflammatory cytokine production, and altered adipokine profiles, as documented in systematic reviews [72]. Clinical research has shown that a more severe form of IR that impacts several tissues and metabolic pathways at once results from the coexistence of both conditions [73].
The disease progression mechanisms show that the combination of MASLD and MetS accelerates beta-cell dysfunction through several pathways [74,75]. Beta-cell function deterioration is caused by oxidative stress, endoplasmic reticulum stress, chronic inflammation, and lipotoxicity from elevated free fatty acids, according to current research [76]. This creates a vicious cycle where declining beta-cell function necessitates increasingly aggressive therapy [74].
Vascular complications are a major concern as well. Recent research indicates that both conditions activate the polyol pathway, produce advanced glycation end products, generate reactive oxygen species and oxidative stress, activate the hexosamine pathway, cause endothelial dysfunction, and cause IR, among other mechanisms that accelerate vascular damage. Further studies have shown that these illnesses can contribute to diabetic patients’ chronic kidney disease on their own [77]. The clinical outcomes present both short-term and long-term challenges, with systematic reviews demonstrating that patients experience greater difficulty achieving glycemic targets and more frequent complications [78]. Reduced quality of life, elevated cardiovascular risk, higher mortality rates, and accelerated progression of complications have all been documented in long-term studies [79].

4.4. Extrahepatic Complications

Beyond its effects on cardiovascular health and diabetes risk, MASLD is associated with a range of extrahepatic complications, many of which are also components or consequences of MetS, some of which are described below.

4.4.1. Chronic Kidney Disease (CKD)

The elevated incidence and prevalence of CKD are independently linked to MASLD. Patients with more severe forms of MASLD are especially at risk of CKD [80]. Chen et al. published a fascinating study in Hepatology International that included 337,783 participants from the United Kingdom (UK) Biobank who were tracked for a median of 8–12 years. The study found that MASLD with both CKD and non-CKD participants had a twofold increased risk of end-stage kidney disease (ESKD) compared to those without the condition. As liver fibrosis scores increase, the association increases. The polygenic risk alleles PNPLA3 rs738409, TM6SF2 rs58542926, Glucokinase regulatory protein (GCKR) rs1260326, and Membrane-bound O-acyltransferase 7 (MBOAT7) rs641738 were also shown to enhance the MASLD effect on ESKD [81].
It is not entirely clear how MASLD and ESKD are related. It is reasonable to assume, nevertheless, that CKD and MASLD are two sides of the same coin. The etiology of both MASLD and CKD is typically similar, and the majority of those affected have metabolic comorbidities like atherogenic dyslipidemia, T2D, obesity, or hypertension [82]. These disorders have remarkably similar molecular mediators, underlying metabolic pathways, and molecular pathophysiologic mechanisms. These include ectopic fat deposition, IR, inflammation, macrophage activation, oxidative stress, and gut dysbiosis [83].

4.4.2. Obstructive Sleep Apnea (OSA)

There is a strong association between MASLD and OSA, with each condition exacerbating the other. OSA-suffering MASLD patients have more severe liver disease. Intermediary mechanisms between OSA and MASLD seem to be similar. OSA is associated with obesity, dyslipidemia, and metabolic syndrome, a well-known comorbidity of MASLD. In addition, OSA results in IR, an oxidative stress state, metabolic lipid disturbances, and gut barrier dysfunction [84]. Therefore, it has been proposed that the pathophysiology and aggravation of the severity of MASLD are related to the presence of OSA, but particularly to its consequence, severe chronic intermittent hypoxia (CIH) [85]. Most research shows a connection between these two long-term illnesses. Türkayet al. demonstrated that, independent of BMI, the presence of OSA increased the severity of steatosis as assessed by ultrasound [86]. According to a recent meta-analysis by Sookoian and Piorola, there were 404 control subjects and 668 OSA patients in 11 studies, consistently using a non-invasive surrogate of MASLD; they showed that OSA was linked to a significant increase in liver enzymes, specifically alanine transaminase (ALT), indicating that OSA is linked to liver deterioration regardless of BMI and diabetic status [87]. According to other recent research, having OSA, with or without CIH which is indicative of OSA severity, increased the severity of MASLD and promoted the development of MASH [88,89,90]. Morbidly obese patients with liver fibrosis had more severe oxygen desaturations and a higher apnea–hypopnea index (AHI) than those without the condition according to a recent study [88]. An interesting finding is that MASLD was identified using a non-invasive systemic algorithm in an OSA population with a broad range of BMIs [from lean to morbidly obese] (i.e., E. With the severity of OSA, the prevalence of MASLD rose (Fibrotest®, MASHtest®, and Steatotest®) [91]. Crucially, steatosis and the degree of nocturnal hypoxia were linked independently [92,93]. The pathophysiology of this interaction is multifactorial, with CIH inducing metabolic abnormalities such as uncontrolled glucose level by increasing the IR [94]. CIH induces gene expression involved in lipogenesis [95], CIH induces Lysyl oxidase (LOX) expression which is involved in extra-cellular matrix rigidity [96], it also induces oxidative stress and lipid peroxidation [97], mitochondrial dysfunction [98], and finally, CIH induces intestinal permeability and disrupts the gut–liver axis [99], which were all discussed earlier as causative mechanisms of MASLD.

4.4.3. Osteoporosis

The relationship between MASLD and osteoporosis is complex and multifactorial. Several recent studies support the association between a decreased bone mineral density and MASLD [100,101,102].
Several mechanisms are suggested to contribute to this association. One potential mechanism is the disruption of vitamin D metabolism. Vitamin D plays a crucial role in calcium absorption and bone health. Studies have shown that individuals with MASLD often have vitamin D deficiency, which can lead to decreased bone mineral density and increased risk of fractures [103,104]. Another potential mechanism is the impact of MASLD on IR and glucose metabolism. IR is a common feature of MASLD and is associated with increased bone resorption and decreased bone formation [105,106]. Additionally, elevated levels of inflammatory markers, often seen in MASLD, can also contribute to bone loss.
Furthermore, the presence of advanced liver disease, such as cirrhosis, may further increase the risk of osteoporosis [107]. Cirrhosis can lead to hormonal imbalances, such as estrogen deficiency in women, which can significantly impact bone health [108].
In summary, mounting data points to a close connection between MASLD and osteoporosis development. The underlying mechanisms are intricate and could include hormone imbalances, IR, inflammation, and vitamin D deficiency. It is important for people with MASLD to be aware of this potential risk and discuss with their doctors’ ways to maintain healthy bones, such as getting enough exercise, consuming adequate calcium and vitamin D, and taking the appropriate medication when necessary.

4.4.4. Endocrine Disorders

Different studies have suggested that MASLD is associated with different endocrine axes. Accumulating clinical and experimental studies have reported that MASLD is associated with polycystic ovarian syndrome (PCOS) and other endocrine disorders. These associations are primarily mediated through shared pathophysiological mechanisms such as IR, hormonal imbalances, and metabolic dysregulation.

Polycystic Ovary Syndrome

This syndrome is characterized by ovulatory dysfunction, hyperandrogenism, and polycystic ovarian morphology [109]. The relationship between MASLD and PCOS is bidirectional, with IR serving as a central link. In PCOS, ovarian androgen production is enhanced by hyperinsulinemia, which results in hyperandrogenism. Hepatic steatosis is worsened by this hormonal imbalance by promoting lipid accumulation in the liver. The other way around, MASLD exacerbates IR as discussed above, further aggravating PCOS symptoms [110]. A randomization analysis reported genetic evidence of a causal association between MASLD and PCOS, emphasizing the closely-connected nature of these conditions [111].

Thyroid Dysfunction

The thyroid axis has extreme effects on energy metabolism, fatty acid oxidation, and hepatic lipogenesis. The liver not only receives signals from thyroid hormones but also has receptors for thyroid-stimulating hormone (TSH), an agonist which induces hepatic steatosis. In addition, thyroid hormones are also crucial in regulating metabolism, and their imbalance can significantly impact liver function. Hypothyroidism, characterized by reduced thyroid hormone levels, results in a decreased basal metabolic rate, lipid accumulation, and weight gain [112], therefore increasing the risk of MASLD. Conversely, MASLD-associated metabolic disturbances and an inflammatory state can impair thyroid function, suggesting a bidirectional relationship. Recent studies have explored the potential of thyroid hormone receptor-beta agonists as therapeutic agents for MASLD, aiming to harness the metabolic regulatory functions of thyroid hormones to ameliorate liver steatosis and inflammation. Thyroid hormone receptor (THR)-β is responsible for regulating metabolic pathways in the liver and is frequently impaired in MASH. On 14 March 2024, the United States Food and Drug Administration (FDA) approved the first treatment for MASH [106]; MAESTRO-NASH reported that resmetirom was superior to a placebo in MASH resolution and improvement in liver fibrosis by at least one stage [107].
In summary, MASLD’s interplay with endocrine disorders such as PCOS and thyroid dysfunction highlights the necessity for a multidisciplinary approach in managing affected individuals. Addressing these interconnected conditions holistically can lead to improved metabolic health and better overall patient outcomes.

4.4.5. Implications for Clinical Management

The diverse clinical effects of METS and MASLD underscore the need for a comprehensive, multidisciplinary approach to patient management. Given the potential for disease progression and development of serious complications, early detection and intervention are critical [113]. Regular screening for METs components in MASLD patients and vice versa is essential. In addition, given the increased cardiovascular risk, aggressive management of cardiovascular risk factors in all patients with MetS and MASLD is warranted. These include lifestyle changes such as weight loss and increased physical activity, as well as pharmacological interventions where appropriate [114]. The complex interplay of these disorders also underscores the importance of a personalized medical approach. Factors such as genetic predisposition, environmental influences, and the presence of comorbidities should be considered when developing treatment strategies for individual patients [115].
In conclusion, the clinical implications of MetS and MASLD are far-reaching, affecting multiple organ systems and significantly impacting patient morbidity, mortality, and quality of life. A thorough understanding of these implications is crucial for healthcare providers to deliver optimal care and improve patient outcomes [116].

5. Therapeutic Strategies and Future Directions

The treatment of MetS and MASLD requires a multi-layered approach that targets various aspects of these complex disorders. Recent advances in our understanding of its pathophysiology have led to the development of novel therapeutic strategies and potential future treatments. In this section, the mechanisms of action of the individual treatment strategies and their specific significance for both MetS and MASLD are explained in more detail.

5.1. Lifestyle Modifications

Lifestyle interventions remain the cornerstone of treatment for MetS and MASLD. Weight loss through calorie reduction and increased physical activity has been shown to improve all components of METs and reduce liver steatosis [117].
Weight loss improves insulin sensitivity, reduces adipose tissue inflammation, and decreases the flow of free fatty acids to the liver. In the context of MetS, this results in improvements in blood pressure, lipid profiles, and glucose metabolism. In MASLD, weight loss reduces liver fat levels, improves liver enzyme levels and, in many cases, can lead to the abatement of MASH [118]. Recent studies have focused on the type of diet that may be most beneficial. The Mediterranean diet, characterized by a high consumption of olive oil, nuts, vegetables, fruits, and fish, has shown promise for improving METs and MASLD. The Mediterranean diet is rich in mono-unsaturated fatty acids, omega-3 polyunsaturated FA, and antioxidants. These components have an anti-inflammatory effect, improve insulin sensitivity, and promote a favorable composition of the intestinal microbiome. In MetS, this diet improves lipid profiles and insulin sensitivity. In MASLD, it reduces liver fat content and improves liver enzyme levels [119,120,121].
Intermittent fasting has also gained attention. Some studies suggest benefits for insulin sensitivity and liver steatosis. Intermittent fasting can improve insulin sensitivity by promoting metabolic changes between glucose and fatty acid oxidation [122]. It may also activate cellular stress response pathways that improve overall metabolic health. For MASLD, intermittent fasting may reduce liver fat levels by promoting fat oxidation during fasting [123].
Physical activity has a variety of effects on MASLD. It increases insulin sensitivity and enables tissue to absorb glucose more efficiently. This reduces glucose production in the liver and decreases the substrate for de novo lipogenesis, an important metabolic pathway in fat accumulation in the liver [124]. Regular physical activity stimulates the breakdown of FAs for energy consumption, a process known as fatty acid oxidation that directly reduces liver fat levels [125]. Exercise also has powerful anti-inflammatory effects and alleviates systemic and liver inflammation, which play an important role in MASLD pathogenesis [126]. In addition, physical activity promotes weight loss or weight maintenance, which is critical for improving MASLD. Even modest weight loss can significantly reduce liver fat levels and improve liver function [127].
Both aerobic and resistance training have demonstrated beneficial effects on MASLD. Aerobic exercise, such as brisk walking, jogging, swimming, and cycling, enhances cardiovascular fitness, improves insulin sensitivity, and promotes fat oxidation [128]. Resistance training, including weightlifting and bodyweight exercises, increases muscle mass, boosts metabolism, and improves insulin sensitivity [129]. A combination of both aerobic and resistance training is often recommended for optimal outcomes [130].

5.2. Pharmacological Interventions

5.2.1. Insulin Sensitizers

Metformin, a first-line treatment for T2D, increases peripheral (including hepatic) insulin sensitivity, reduces hepatic glycogenesis, and increases glucose uptake and utilization of insulin-stimulated peripheral tissues in MASLD patients [131]. A meta-analysis suggests that low-dose metformin (500–3000 mg/d) over a short-term period (up to 6 months) can reduce liver enzymes (ALT, aspartate aminotransferase (AST)), triglycerides (TG), and total cholesterol (TC), while improving IR (HOMA-IR) [132]. The improvement in liver function and lipid metabolism appears to be independent of its glucose-lowering effects. Both ALT and AST levels decrease with higher metformin doses and prolonged use, indicating reduced liver damage.
Metformin is also linked to a reduction in TG levels in patients with MASLD [133]. The liver’s ability to balance fat storage and breakdown can be disrupted in metabolic disorders, leading to fat accumulation in the liver and the development of fatty liver. MASLD is associated with dyslipidemia, high blood sugar, and IR. Elevated TG levels can worsen IR and promote a cycle of increasing liver fat [134]. TC, which includes both esterified cholesterol and free (unesterified) cholesterol “cholesteryl ester”, reflects lipid metabolism and can indicate liver damage when elevated in MASLD patients [135]. It has been reported that metformin significantly improves blood lipid levels, including TC and TG, in MASLD patients. Moreover, metformin helps reduce IR and improve liver function, highlighting its potential role in treating MASLD and its associated metabolic issues [136].
The difference between preclinical and clinical outcomes could be due to different disease severity in human patients or differences in metformin absorption between species. Further research, particularly with larger sample sizes, is needed to clarify these effects.
While metformin improves liver parameters in diabetic MASLD patients, newer antidiabetic drugs have shown stronger effects: thiazolidinediones (TZD), Glucagon-like peptide-1 (GLP-1) receptor agonists, and Sodium-Glucose Linked Transporter (SGLT2) inhibitors have shown greater effectiveness in reducing liver fat and improving liver biochemistry than metformin alone; dipeptidyl peptidase 4 (DPP-4) inhibitors (such as sitagliptin) were less effective at reducing liver enzymes as metformin [132] and Sulfonylureas (e.g., gliclazide) also reduced liver fat but to a lesser extent than metformin [137]. In addition, the combination of metformin with other antidiabetic agents has shown additional advantages over monotherapies: TZDs, GLP-1 receptor agonists, DPP-4 inhibitors, and SGLT2 inhibitors, when used together with metformin, improved liver parameters more effectively than metformin alone [138,139].
Thiazolidinediones, nuclear receptors, known as PPARs, are found in adipose tissue, liver, muscle, heart, and kidneys, among others, where genes associated with fatty acid oxidation, lipid transport, and gluconeogenesis are regulated. α, β/β, and ω are the three PPAR subtypes [140,141,142]. Some PPAR agonists are used to treat T2DM, such as pioglitazone and rosiglitazone [143,144].
Pioglitazone, an agonist for PPAR-α, modulates key genes in lipid metabolism [145]. Through increased expression of Apo AI and AII, it lowers triglyceride levels in lipoproteins, increases fatty acid oxidation, promotes fatty acid transporters, decreases ketogenesis, and increases high-density lipoprotein (HDL-C) synthesis [146]. In the pioglitazone versus Vitamin E versus Placebo for the Treatment of Non-Diabetic Patients with Nonalcoholic Steatohepatitis (PIVENS) trial, despite the fact that pioglitazone did not have any benefit in the primary outcome over the placebo, it was associated with a highly significant inflammation reduction, hepatocellular ballooning, and reduction in steatosis, and lower IR and liver enzyme levels [147]. Various studies, including Belfort et al. and Aithal, showed that pioglitazone significantly improved liver histology in MASH patients, particularly obese patients with glucose intolerance [147,148].
As for rosiglitazone, studies have shown that it can reduce hepatic fat accumulation by enhancing adipocyte differentiation and lipid storage in peripheral fat depots, thereby reducing ectopic lipid deposition in the liver. Additionally, rosiglitazone has anti-inflammatory properties, which may help mitigate liver inflammation and fibrosis associated with MASH [149,150]. Several clinical trials have assessed rosiglitazone in MASLD/MASH, with some demonstrating histological improvements in steatosis and inflammation. The Fatty Liver Improvement with Rosiglitazone Therapy (FLIRT) trial reported improvement in transaminase levels and steatosis but also weight gain with no improvement of other liver injury parameters [151]. The continuous FLIRT-2 study reported a substantial steatosis effect within one year though maintaining transaminase levels and an IR effect [152]. Rosiglitazone use was limited due to concerns about weight gain and fluid retention especially among comorbid cardiovascular patients [153].
In summary, metformin is promising at improving liver function, lipid metabolism, and insulin sensitivity in MASLD patients, particularly those with T2D. Its positive effects are independent of glucose control and work primarily through activated protein kinase (AMPK) activation, reduced lipogenesis, increased fatty acid oxidation, and anti-inflammatory mechanisms. However, clinical results remain mixed. Some studies report improvements in liver enzymes and fat accumulation, while others show minimal or even negative effects. Compared to newer antidiabetic agents such as TZD, GLP-1 receptor agonists, and SGLT2 inhibitors, metformin alone has a weaker effect on liver histology. Pioglitazone showed significant improvements in MASH histology, whereas rosiglitazone showed mixed results and is limited due to concerns about cardiovascular risks. Given the variability of clinical responses, further large-scale studies are needed to determine the optimal role of metformin in MASLD treatment, particularly when combined with other metabolic therapies.

5.2.2. GLP-1 Receptor Agonists

GLP-1, a hormone produced by the L cells of the small intestine and proximal large intestine, plays a critical role in regulating plasma glucose levels. It increases insulin secretion in a glucose-dependent manner and inhibits glucagon secretion, which helps lower blood sugar [154]. In addition, GLP-1 has positive effects on metabolism, including improving IR, promoting weight loss through delayed gastric emptying, and reducing appetite [155].
However, GLP-1 is rapidly degraded by the enzyme DPP-4, which is abundantly expressed in the liver [156]. Elevated circulating DPP-4 levels are associated with more severe liver disease, particularly in individuals with MASLD [157]. This has led to the hypothesis that inhibiting DPP-4 could potentially improve the histological characteristics of MASLD and MASH (non-alcoholic steatohepatitis) by preserving GLP-1’s beneficial actions [158]. The positive effects of GLP-1 on MASLD can be both indirect and direct. Indirect effects occur by reducing appetite and delaying gastric emptying, GLP-1 may help to reduce body weight and insulin sensitivity, which in this case alleviates metabolic disorders commonly associated with MASLD. In addition, it acts directly as GLP-1 can act directly on hepatocytes (liver cells) to reduce triglyceride accumulation, improving liver fat levels and reducing inflammation [159].
One promising therapeutic approach involves tirzepatide, an agonist targeting both the GLP-1 receptor and the glucose-dependent insulinotropic polypeptide (GIP) receptor [160]. In a randomized phase 2 trial of patients with T2D, tirzepatide showed a significant reduction in MASH-related biomarkers including AST/ALT and procollagen III (a marker for liver fibrosis) [161]. In addition, tirzepatide increased levels of adiponectin, a hormone with antifibrotic and anti-steatogenic properties, which helps reduce liver fat and fibrosis. These results suggest that tirzepatide may have a dual benefit in treating MASLD and MASH, as it both improves metabolic factors such as IR and directly affects the liver.
In addition, liraglutide, a long-acting GLP-1 analog, has been approved by the FDA for the treatment of type 2 diabetes and has shown promise for treating MASH. In the Liraglutide efficacy and action in NASH (LEAN) study, liraglutide resolved MASH in 39% of patients compared with 9% in the placebo group (p = 0.02), with only minimal side effects, primarily gastrointestinal (e.g., diarrhea). Importantly, liraglutide was also associated with weight loss; nonetheless, it is not clear whether this improves liver histology or liraglutide itself had an independent effect on MASH. The study also suggested that liraglutide could reduce the fibrosis stage in patients [162].

5.2.3. SGLT2 Inhibitors

Normally, the renal filtration threshold for glucose equals to 180 mg/dL, meanwhile when plasma glucose exceeds that number, glucose is eliminated in the urine [163].
SGLT consists of two transporters: SGLT1, which is present in both the intestines and kidneys, particularly in the thick part of the proximal tubule, and SGLT2, which is instead exclusively present in the renal tubule. Approximately 97% of glucose is absorbed upstream of the proximal tubule by SGLT2, while the remaining glucose is absorbed downstream by SGLT1 [164].
In hyperglycemic conditions, the kidneys increase their renal absorption capacity to a maximum of 600 g/day to prevent renal glucose loss. This effect, which is mediated by the SGLT cotransporter, not only results in an increase in the reabsorption of glucose but also of sodium and fluids, so that inhibiting this transporter would not only have a positive effect on blood sugar control but also on sodium homeostasis and water retention [165,166].
SGLT2 inhibitors have been developed to treat T2D. They inhibit the reabsorption of glucose in the kidneys and lower blood sugar levels [167]. For these pathophysiological reasons, SGLT2 inhibitors are promising and important therapeutic agents for patients with MASLD [168,169].
Numerous studies have shown that SGLT2 inhibitors reduce the risk of cardiovascular and kidney diseases, reduce body weight, and improve ALT plasma levels [24,170]. In addition, by reducing fat mass, SGLT2 inhibitors prevent the adipocyte release of inflammatory cytokines, thus reducing the inflammatory effects, which are a major cause of MASH progression [171].
Key studies include the e-LIFT study (2018), whereby Empagliflozin (10 mg) significantly reduced liver fat by 16.2% compared to 11.3% in the placebo group, thus improving liver enzymes (ALT) [172]; the study carried out by Kahl et al., which reported that Empagliflozin (25 mg) reduced liver fat content in T2D patients with MASLD, confirmed by magnetic resonance spectroscopy [173]; Shimizu et al.’s study, which reported dapagliflozin (5 mg/day) to result in significant reductions in liver fat and liver stiffness, which was investigated by transient elastography [174]. In addition, the study carried out by Ito et al. reported that ipragliflozin (50 mg) also improves liver steatosis, reduced liver enzymes, and lowers body weight and visceral fat [144], and finally, according to another study, over a period of six to twelve months, canagliflozin (100 mg/day) improved liver fat and histology, decreased IR, and decreased liver enzymes [175].
These studies show that SGLT2 inhibitors not only help control blood sugar but also improve liver function, reduce fat mass, and lower liver inflammation, which contributes to a reduced risk of MASH progression and other complications associated with T2DM and MASLD. This makes this drug class one of the most promising future therapies for the specific indication of MASLD.

5.2.4. Statins

Statins are commonly used to treat lipid disorders, particularly those associated with elevated cholesterol levels, and are essential for treating CVD. They act as 3-hydroxymethyl-3-methylglutaryl-coenzyme A lyase (HMG-CoA) reductase inhibitors, reduce cholesterol synthesis, and lower serum cholesterol levels, which significantly reduces the morbidity and mortality of cardiovascular diseases [176]. Statins increase lipoprotein clearance primarily by reducing low-density lipoprotein (LDL) cholesterol and increasing LDL receptor expression [177]. They also have wider metabolic effects, including lowering TG, moderating post-meal TG increases, and increasing HDL cholesterol [178,179,180].
Statins not only lower cholesterol levels but also have pleiotropic effects. Lower levels of highly sensitive C-reactive protein (CRP) suggest that vascular inflammation has declined, and an increase in nitric oxide (NO) availability improves endothelial function [181]. Statins not only lower pro-inflammatory cytokines and increase platelet reactivity, but also support the healing process after myocardial ischemia—a reperfusion injury [182,183,184]. Overall, statins offer broad cardiovascular benefits, making them a cornerstone of contemporary cardiovascular treatment.
Statins are beneficial for patients with MASLD, especially those with concurrent CVD [185,186]. MASLD and CVD share common risk factors, and although liver-related mortality in MASLD is primarily due to complications such as cirrhosis and liver cancer, patients with MASLD are at higher risk of cardiovascular morbidity and mortality [187]. By blocking HMG-CoA reductase, statins lower cholesterol biosynthesis and effectively treat dyslipidemia, which is commonly seen in patients with non-alcoholic fatty liver disease. In particular, atorvastatin was found to improve liver enzyme levels and MASLD ultrasound markers while drastically reducing cardiovascular disease morbidity and mortality [185]. Statin therapy is particularly beneficial for statin-naïve MASLD patients with advanced fibrosis and increased cardiovascular risk [188,189,190]. Overall, statins are efficient in treating both MASLD and its associated cardiovascular risks, potentially saving lives as the disease progresses.
As for the safety part, statins are generally safe for treating MASLD and MASH despite concerns over potential liver toxicity [185,191,192]. Although statins can have side effects like myopathy, kidney, and liver dysfunction [193], these are rare, and recent studies suggest statins do not significantly elevate liver enzymes in MASLD patients. Large studies, including those focused on elderly and cirrhotic patients, indicate that statins are safe even in high-risk groups, reducing the risk of liver complications like HCC [194,195,196]. The National Lipid Association and recent guidelines advocate for statin use in MASLD/MASH patients with hypercholesterolemia, given their low hepatotoxic risk [197,198].
In addition, statins have shown promising effects in reducing inflammation and improving liver conditions in patients with MASLD and MASH. They exhibit anti-inflammatory properties by inhibiting pathways that lead to the activation of pro-inflammatory cytokines, such as IL-1β, IL-18, and TNF-α, which are linked to MASLD progression [199,200]. For example, atorvastatin reduces inflammation by blocking the NLRP3 inflammasome pathway, and long-term use can alleviate liver inflammation, steatosis, and hepatocellular damage [201].
Statins also have potential antifibrotic effects, which are critical to prevent liver disease progression. Studies suggest that statins, including simvastatin and fluvastatin, may reduce liver fibrosis by modulating enzymes such as nitric oxide synthase and inhibiting HSC activation [202]. Clinical evidence supports the assumption that taking statins correlates with a lower likelihood of advanced liver fibrosis and may reduce the risk of HCC in patients with MASH and advanced fibrosis [195,203]. Additionally, these drugs can help lower cholesterol levels and inflammation state by preventing cholesterol buildup in liver cells, reducing inflammation by inhibiting the activation of KCs and HSCs, both key players in fibrosis development. They also restore the health of liver sinusoidal endothelial cells (LSECs), reducing portal pressure and improving liver prognosis. Through Human serum paraoxonase-1 (PON1); statins restore serum PON1, an antioxidant enzyme, which reduces oxidative stress and helps reverse MASLD progression [189,204,205]. MASH is primarily caused by lipid peroxidation (LPO), which is reduced by increased PON1 activity. In addition, statins have anti-fibrotic and anti-inflammatory effects by blocking isoprenylation of small Guanosine-5′-triphosphate (GTPases) (such as Ras homolog family member A (RhoA) and renin–angiotensin system Ras) and reducing inflammation and fibrosis via signaling pathways such as Ras/extracellular signal-regulated kinase (ERK)1/2 and RhoA/RhoA kinase [206]. Statins also alter PPARs, particularly PPARα, which improves fatty acid metabolism and decreases fibrosis, IR, and liver steatosis. It improves liver health by inhibiting lipid synthesis and activating AMPK, a key metabolic regulator that helps reduce fat accumulation in the liver [207,208]. More research is needed to determine whether these drugs can help treat liver fibrosis by inducing ferroptosis, a type of iron-dependent cell death. According to recent research, changing the gut microbiota to a healthier profile may also help treat MASH and MASLD associated with obesity by reducing systemic inflammation [209].
In conclusion, and despite the growing evidence of the beneficial effects of statins on MASH and cardiovascular health, more clinical trials are needed to fully understand their role, particularly in relation to microbiota.

6. Emerging Therapies

6.1. FXR Agonists

Maintaining BA metabolic homeostasis is critical for preventing steatosis and liver damage, which may result from excessive cholesterol accumulation in the liver due to impaired BA conversion or cholestasis. In MASLD, BA metabolism is disrupted, resulting in elevated BA levels and an abnormal profile [210]. FXR plays a key role in regulating BA metabolism by controlling BA synthesis, excretion, and reabsorption, and significantly influencing MASLD pathogenesis. FXR is a nuclear receptor that is primarily activated by bile acids. Activating FXR suppresses the new synthesis of BAs from cholesterol, limits the circulating BA pool, and promotes bile acid transport from hepatocytes [211,212]. This receptor activation has several beneficial effects: improved insulin sensitivity, reduced hepatic gluconeogenesis, and increased expression of hepatic scavenger receptors, which accelerates reverse cholesterol transport [213,214,215]. Active FXR also causes small heterodimer partner (SHP) expression in the liver. Cholesterol 7 alpha-hydroxylase (CYP7A1) and cytochrome P450, family 8, and subfamily B, polypeptide 1 (CYP8B1), enzymes involved in BA synthesis, are then suppressed by SHP through its interaction with liver receptor homolog 1 (LRH-1) [216]. Additionally, activated FXR inhibits Multitasking Na+/Taurocholate Cotransporting Polypeptide (NTCP) and Organic anion transporting polypeptides (OATP1) to stop BA reuptake and increases the expression of bile salt export pump (BSEP) to facilitate BA secretion into bile ducts [217,218,219]. To further aid in the excretion of BAs and liver protection, FXR also activates enzymes involved in BA modification, including Cytochrome P450 3A4 (CYP3A4) and Uridine diphosphate (UDP)-glucuronosyltransferase [220]. Through the induction of fibroblast growth factor 15/19 (FGF15/19), intestinal FXR activation also aids in the restoration of BA homeostasis in MASLD and MASH [221,222]. The FGFR4 is bound by this protein, which decreases BA secretion by the liver. This activation of the ERK and Jun N-terminal kinase (JNK) signaling pathways prevents the synthesis of CYP7A1 and BA [223,224]. In addition, FXR activation helps reduce liver lipids by lowering the levels of mono- and polyunsaturated fatty acids (MUFA and PUFA) [214]. FXR also stimulates the expression of silent information regulator sirtuin 1 (SIRT1), which inhibits fatty acid synthesis and promotes oxidation; loss of SIRT1 results in MASLD. FXR and SIRT1 interact to regulate each other, with SIRT1 increasing FXR stability [225].
Since FXR activation has shown a protective effect in the treatment of MASLD, efforts are being made to develop various FXR agonists. However, most of these agonists, such as obeticholic acid (OCA), vonafexor, and tropifexor, cause side effects such as itching and increased LDL cholesterol levels that hinder their approval and use [226,227]. Research indicates that pruritus is linked to increased serum IL-31 levels caused by FXR activation in the liver [228].
OCA, an FXR agonist derived from chenodeoxycholic acid, has shown promise in the treatment of non-alcoholic steatohepatitis (MASH) and has improved the histological characteristics of MASH, particularly with regard to the NAFLD Activity Score (NAS) [229,230]. In phase 2a studies, OCA resulted in weight loss, increased insulin sensitivity, and reduced liver inflammation and fibrosis in patients with diabetes and MASLD. The phase 2b Farnesoid X Receptor Ligand Obeticholic Acid in Nonalcoholic Steatohepatitis Treatment (FLINT) trial involving 283 MASH patients showed that 45% of patients treated with 25 mg OCA daily had a significant reduction in NAS without the fibrosis worsening, compared with 21% in the placebo group. OCA also reduced fibrosis (35% versus 19%), steatosis, lobular inflammation, and hepatocyte balloon formation, leading to improvements in body weight and ALT levels [231]. OCA is currently being investigated in a phase 3 trial (REGENERATE) for its effectiveness in MASH for fibrosis and compensated cirrhosis due to MASH [232]. Nonetheless, one of the phase 2 studies was interrupted as the primary endpoints were not met by 18 months [233].
Alternative strategies include intestinal FXR-targeting FXR antagonists like Tauro-α-muricholic acid (T-α-MCA) and Gly-MCA, which have been shown to improve MASLD. Other substances that selectively block intestinal FXR and provide a better therapeutic profile than current treatments, like F6 and V023-9340, are presently under development [234,235]. In addition, drugs and natural extracts that indirectly target FXR are being researched. Plant compounds such as emodin and caffeic acid phenethyl ester (CAPE) activate FXR signaling pathways and have been shown to be effective in improving liver steatosis and MASLD [236,237]. Reused drugs such as disulfiram (DSF) and hydrogen sulphide (H2S) are also being investigated for their potential to increase FXR activity and improve lipid metabolism in MASLD/MASH patients [238,239]. Probiotics such as Akkermansia muciniphila and Bifidobacterium bifidum are also promising to regulate FXR and alleviate MASLD [240].

6.2. Acetyl-CoA Carboxylase (ACC) Inhibitors

De novo lipogenesis (DNL) plays a critical role in the development of MASH, in which excessive triglyceride accumulation leads to steatosis, lipid toxicity, inflammation, and fibrosis [241]. ACC inhibitors, which target DNL and promote mitochondrial fatty acid oxidation, are being explored as potential treatments for MASH [242]. These carboxylases have two isoforms, ACC1 and ACC2, which are different. The ACC2 of mitochondrial membrane controls fatty acid oxidation, whereas ACC1 primarily promotes fatty acid synthesis in the cytosol [243].
In healthy lean individuals, DNL contributes approximately 10% to liver lipids, but in obese individuals, this figure rises to 10–20%, and in patients with MASLD, to 25–40% [244]. DNL is primarily regulated at the transcriptional level, with insulin activating the SREBP1c transcription factor, which upregulates genes involved in fatty acid synthesis. Similarly, glucose uptake promotes carbohydrate-responsive element-binding protein (ChREBP), another transcription factor that enhances the biosynthetic gene expression of FA. Studies show that important DNL genes such as those for SREBP1c and DNL enzymes are upregulated in MASLD patients [245].
While there are limited data on the regulation of ACC isoforms in MASLD, studies indicate that ACC1 expression is increased in these patients [246]. MASLD may be exacerbated by high-glucose diets and sugar-sweetened beverages (SSBs), particularly those that contain fructose, which are associated with elevated DNL. Fatty acid synthase and acetyl-CoA carboxylase alpha (ACACA) (which codes for ACC1) are two genes in the DNL pathway that have been demonstrated to be upregulated by fructose consumption [247]. This suggests that the rise in fructose consumption, particularly through SSBs, may be a significant factor in the growing prevalence of MASLD in western countries [248].
Several ACC inhibitors, including Pfizer’s PF-05175157 [249], (MSD’s MK-4074 [250], Gilead’s GS-0976 (Firsocostat) [246,251], and Pfizer’s PF-05221304 [243,252], have shown promising results in reducing hepatic DNL, but also raised concerns about side effects, particularly hypertriglyceridemia (elevated TGs). For example, PF-05175157 was discontinued due to a reduction in platelet counts [253], while GS-0976 and PF-05221304 showed improvements in liver fat but also led to increased serum triglycerides in some patients [249,254]. This unexpected side effect may result from a deficiency of PUFAs in the liver, triggering a cascade of metabolic changes [246]. To address these issues, combination therapies are being tested. For instance, combining PF-05221304 with PF-06865571 (a diacylglycerol acyltransferase (DGAT) 2 inhibitor) or GS-0976 with Fenofibrate has shown potential in reducing hypertriglyceridemia and improving liver health in MASLD patients [254,255].

6.3. Thyroid Hormone Receptor (THR)-β Agonists

Thyroid hormones, which are generated by the thyroid and hypothalamus, control lipid metabolism. Thyroxin 3 (T3) binds to THR-α in the heart and bones and THR-β in the liver to affect gene transcription [256]. Studies show that hypothyroidism and elevated thyroid-stimulating hormone (TSH) levels are associated with an increased risk of MASLD [257]. Thyroid hormones affect liver function by promoting β-oxidation of fatty acids and reducing lipid accumulation in the liver through mechanisms such as autophagy, lipophagy, and mitochondrial biogenesis [258,259].
Studies on MASH in mice suggest that intrahepatic hypothyroidism plays a role in the development of the disease, with low thyroid hormone levels associated with increased inflammation and liver fat [260,261]. The conversion of T4 into the active hormone T3 is facilitated by the liver enzyme (Type 1 Iodothyronine Deiodinase) DIO1, but with advanced MASH, its activity decreases, which further impairs lipid metabolism. Since THR-β controls important metabolic pathways and is highly expressed in hepatocytes, it is a crucial target for the development of therapies for lipid-associated liver diseases such as MASLD and MASH [262].
Resmetirom is an experimental THR-β agonist that is highly selective for THR-β over THR-α and is 28 times more selective [262]. This agonist uses the OATP1B1 receptor for hepatocyte uptake and strongly activates THR-β [263,264]. It also supports DIO1’s function in converting T4 to T3 by upregulating it. This mechanism is further supported by the low serum T4 levels in patients treated with resmetirom [265]. Due to its specificity and effects on lipophagy, mitophagy, mitogenesis, and β-oxidation in hepatocytes, several studies have been conducted with the drug. The results were promising and reduced LDL-C, TGs, and apoloprotein B without affecting TSH and free T3 levels. In addition, another phase 2 study with this agonist showed a significant reduction in liver fat compared to a placebo and an improvement in liver enzyme levels, lipid profiles, and inflammatory markers. Thus, resmetirom was approved by the US FDA on 14 March 2024 for the treatment of MASH. Safety profiles were favorable, there were no signs of adverse effects from THR-α activity, and thyroid function remained stable [266,267,268].

6.4. Antibiotics

The gut–liver axis plays a significant role in the mechanism of MASLD. Dysbiosis as discussed above has been associated with fat accumulation, liver inflammation, and fibrosis. Since it has the potential effect on gut microbiome, the use of antibiotics has gained a lot of attention in recent years. It may help restore microbial balance by modifying gut microbiome. Recent studies suggest specific gut-targeting antibiotics could reduce harmful substance translocation from the gut to liver. One of the investigated antibiotics is Rifaximin, a derivative of Rifamycin but gut-selective antibiotic with minor systemic effect. It has FDA approval for treating irritable bowel syndrome (IBS) [269,270,271,272], hepatic encephalopathy for reducing recurrent episodes [273,274,275,276], and traveler’s diarrhea [277,278,279,280]. A recent study suggested that rifaximin affects liver inflammation and intestinal microbiome, and reported efficacy and safety of the drug through a reduction of serum endotoxin, cytokine18 CK18, proinflammatory cytokine (PROC 3,4,8), and fat scoring of MASLD [281]. We advise the investigation of this drug since it is gut selective with almost no systemic bioavailability (less than 0.04% in blood stream) and could avoid possible antibiotics resistance in the long-term effects [278,282,283]. Nonetheless, the therapeutic use of antibiotics in MASLD remains controversial, with some experts cautioning against their use due to concerns over long-term effects on the microbiome.
In conclusion, while antibiotics, especially those like rifaximin, show promise as a therapeutic tool in modulating the gut–liver axis in MASLD, further research is needed to better understand their efficacy and safety in this context. The development of targeted microbiome-based therapies integrated with multi-omics profiling harnessing the benefits of antibiotics without disrupting the microbiome balance may provide a new avenue for MASLD treatment in the future.

6.5. Novel Anti-Inflammatory and Anti-Fibrotic Agents

Since inflammation and fibrosis are key components in the progression from MASLD to MASH and cirrhosis, new agents are currently being investigated that target these processes. These agents target specific inflammatory pathways (e.g., C-C chemokine receptor type (CCR) (CCR2/CCR5 antagonists) or fibrosis-promoting processes (e.g., galectin 3 inhibitors, anti-Lysyl oxidase Homolog (LOXL2 antibodies). These agents reduce inflammation and fibrosis and aim to stop or reverse the progression of MASLD. Although reducing systemic inflammation is primarily aimed at liver diseases, it could also have a positive effect on METS components [284].
In addition to the various current therapeutic targets, one approach includes FGF21. It has been reported to have anti-fibrotic effects and improve metabolic status. In the BALANCED study, the use of efruxifermine effectively and safely reduced liver fat in patients with MASH cirrhosis [285]. In addition, FGF-21 (efruxifermine) replaced MASH in the HARMONY study, a double-blind randomized controlled phase 2 trial, and improved liver fibrosis in patients with F2 or F3 fibrosis [286]. In addition, in another phase 2 trial, FGF-21 agonist in combination with GLP-1 significantly improved noninvasive markers of MASH-related disease [287]. To add more, A current ongoing clinical trial in a phase 2 phase is investigating the role of FGF19 analogue as a reversal drug in MASH and liver fibrosis; further trials are currently underway [288].
In summary, the FGF-21 agonist, the FGF-19 analog, and the CCR2/CCR5 antagonist have great therapeutic potential for the treatment of MASH and MASLD. However, thorough phase 3 clinical trials are essential to determine their place in clinical practice and to develop long-term safety and efficacy profiles in a larger patient population.

7. Future Directions

7.1. Precision Medicine Approaches

A one-size-fits-all strategy might not be the best option due to the heterogeneity of MetS and MASLD. The onset and progression of MetS and MASLD are influenced by both genetic and environmental factors. The goal of precision medicine techniques is to customize care according to each patient’s unique genetic profile, environmental exposures, and disease characteristics [289].
Omics approaches are recent evolving approaches in the field of clinical research. The integration of multi-omics modalities could provide more thorough understanding of the underlying mechanisms of liver disease, especially MASH. Multi-omics involving research recently investigated MASH and alcoholic liver disease and cirrhosis. The use of genome, epigenome, transcriptome proteome metabolome, and microbiome profiling in combination with a clinical scoring system can shed light on complex relationships and individual therapeutic potentials. A multi-omics study of MASLD published in nature genetics revealed 18 sequence variants associated with MASLD, 4 variants associated with cirrhosis, and surprisingly two protective variants. Nonetheless, the phenotype was not similar between these shared variants which suggests more than one biochemical pathway that leads to MASLD [290]. Another notable study, featured in Frontiers in Microbiology, employed multi-omics techniques to investigate the biological mechanisms of MASLD. By analyzing genomic, transcriptomic, and metabolomic data, the researchers identified specific microbial signatures and metabolic alterations associated with the disease [291].
These advancements in multi-omics approaches have significantly enhanced our understanding of non-alcoholic fatty liver disease (MASLD), hopefully facilitating the identification of novel biomarkers and therapeutic targets. Furthermore, using multi-omics techniques in combination with already-proven therapy in context with the resolution of fibrosis or normalization of hepatocellular enzyme, which have not been done before, may at last discover the needed metabolite or protein than can be used as a primary or secondary preventive screening tool. Thus, we encourage revolutionary approaches in the field of hepatology research by combining different types of studies.

7.2. Microbiome Modulation

Novel therapeutic approaches that target the microbiome have surfaced as promising tools for managing MASLD and enhancing liver function as its prevalence rises worldwide.
Probiotic use is among the most researched methods for modifying the gut microbiota. These are live microorganisms that provide the host with health benefits when given in sufficient amounts [292,293]. In order to stop harmful endotoxins and bacterial products from leaking into the bloodstream, probiotics improve the intestinal barrier [294], encourage the growth of good bacteria [295], and restore the balance of the gut microbiota [296]. In individuals with MASLD, probiotics can help reduce inflammation, improve insulin sensitivity, and reduce liver fat accumulation. Several studies have demonstrated that specific strains of probiotics, such as Lactobacillus and Bifidobacterium, can modulate metabolic parameters and improve liver enzymes in patients with MASLD [297,298]. For instance, a randomized clinical trial reported that probiotic supplementation improved liver function and reduced inflammation in patients with MASLD [299].
In addition to probiotics, prebiotics—non-digestible food components that selectively promote the growth of beneficial bacteria—have been investigated as a therapeutic intervention for MASLD [300]. Prebiotics, particularly dietary fibers and oligosaccharides, influence the composition of gut microbiota by stimulating the growth of beneficial bacteria such as Bifidobacteria and Lactobacilli This in turn leads to the production of short-chain fatty acids (SCFAs) such as butyrate, acetate, and propionate, which have been shown to have anti-inflammatory and anti-fibrotic effects [301,302]. Another systematic review emphasized the beneficial effects of prebiotics in regulating liver fat accumulation and improving liver function [303]. Furthermore, a high-fiber diet enriched with prebiotics led to a reduction in liver fat content and improved insulin sensitivity in mice with MASLD [304].
Synbiotics, which combine both probiotics and prebiotics, are another therapeutic approach that has gained traction. The rationale behind using synbiotics is that they can provide both live beneficial bacteria and substrates that enhance their growth and activity, thereby offering a more comprehensive approach to restoring gut microbiome balance [305,306]. Furthermore synbiotic supplementation improved metabolic parameters, liver enzymes, and inflammatory markers in MASLD patients [307]. The combined action of probiotics and prebiotics in synbiotics may offer synergistic effects in restoring gut microbiota homeostasis, reducing hepatic fat content, and preventing liver damage [308].
Fecal microbiota transplantation (FMT) has emerged as a promising therapeutic approach for MASLD. This treatment involves introducing donor stool into the patient’s gut with minimal manipulation to restore the patient’s disrupted gut microbiome [309]. The onset and progression of MASLD and MASH have been linked to dysbiosis, or changes in the composition of the gut microbiota. Increased intestinal permeability brought on by dysbiosis may enable toxic substances to enter the liver through the portal circulation, causing inflammation and liver damage. By restoring a balanced gut microbiota, FMT may lessen intestinal permeability and lessen inflammation in the liver [310].
Recent studies have explored its potential to restore gut microbiota balance and improve liver function. A randomized controlled trial by Craven et al. (2020) investigated the effects of allogenic FMT in MASLD patients. The study found that while FMT did not significantly alter IR or hepatic fat content, it notably reduced small intestinal permeability, suggesting a potential benefit in restoring gut barrier integrity [311].
Furthermore, research by Xue et al. (2017) demonstrated that FMT could decrease hepatic fat accumulation by improving gut microbiota dysbiosis, thereby attenuating fatty liver disease. The study highlighted significant differences in clinical features and gut microbiota between lean and obese MASLD patients, with FMT showing more pronounced effects in lean individuals [296]. Additionally, a comprehensive review by Del Barrio et al. (2023) emphasized the therapeutic potential of FMT in MASLD. The authors noted that FMT could correct intestinal bacterial imbalances, thereby preventing the development and progression of MASLD [312]. FMT represents a novel approach to managing MASLD and MASH by targeting gut microbiota dysbiosis. Although preliminary studies are promising, comprehensive clinical trials are necessary to fully establish their role in the treatment of these liver diseases.

7.3. The Role of Artificial Intelligence in Prediction, Diagnosis, and Treatment

Artificial intelligence (AI) techniques are enhancing disease prediction for MASLD, and MetS. Machine learning models trained on clinical and laboratory features can identify patients at risk with high accuracy [313,314,315]. For example, a machine learning (ML)-based model using routine lab markers achieved an area under the receiver operating characteristic curve (AUROC) ~0.81–0.84 in validation for MASLD detection [314,316]. In MetS, AI algorithms (e.g., gradient boosting, support vector machine (SVM)) have been applied to large datasets to predict syndrome onset, enabling early lifestyle interventions [317]. These predictive models can flag high-risk individuals before clinical diagnosis, supporting preventive care planning.
Deep learning applied to medical imaging has improved MASLD diagnosis. AI-assisted ultrasound, for instance, can noninvasively detect hepatic steatosis with sensitivity and specificity ~0.97 and 0.98, markedly outperforming conventional clinical scoring methods [318]. Advanced models analyze ultrasound, computed topography (CT), or magnetic resonance imaging (MRI) scans to quantify liver fat and fibrosis, reducing the need for biopsy. In one study, a deep-learning index derived from ultrasound images distinguished moderate-to-severe MASLD with AUROC ≈ 0.966. Such image-based AI tools can stratify disease severity and even predict complications (like cirrhosis or HCC risk) from routine scans, aiding radiologists in faster, more accurate MASLD diagnosis.
AI is transforming histological assessment of liver biopsies in MASLD. Digital pathology algorithms can evaluate biopsy slides for steatosis, inflammation, ballooning, and fibrosis with high concordance to expert pathologists. In a recent study, a deep neural network trained on hundreds of NASH biopsy images matched pathologist scores for key features and fibrosis stage. Similarly, machine learning classifiers have achieved ~89% accuracy in identifying histologic fat and architectural features on whole-slide images [319]. These tools reduce inter-observer variability and offer rapid, standardized MASH activity and fibrosis scoring. AI-based digital pathology is already being used in clinical trials to quantify treatment response (e.g., fibrosis regression) more sensitively than traditional pathology [319]. Overall, automated histopathology analysis promises to streamline MASLD diagnosis and staging in both research and practice.
AI-driven approaches are accelerating drug discovery for MASLD/MetS. By mining big data and using predictive modeling, AI can identify novel therapeutic targets and drug candidates. For example, an AI-guided ensemble learning strategy was used to discover new nonsteroidal FXR agonists for MASH treatment [319,320]. An integrated machine learning which poses filters with virtual screening to overcome protein-flexibility issues, has successfully uncovered a potent FXR agonist that traditional methods missed [321]. AI is also being applied to optimize clinical trial design and personalize therapy. In MetS, AI models can predict which patients will respond to interventions (diet, exercise, or medications), supporting more tailored treatment plans [317]. Additionally, “digital twin” simulations and AI-powered organ-on-a-chip systems are emerging to test drug effects in silico. Early studies show AI-enhanced liver micro-physiological models can detect drug-induced liver changes and toxicity faster and more reliably [322,323,324]. Such innovations accelerate the development of effective pharmacological therapies for MASLD, a field that until recently had limited approved drugs.
AI integration in clinical workflows supports personalized medicine for patients with MASLD and MetS. By analyzing electronic health records (EHR), lab results, imaging, and even genomic data, AI can provide clinicians with decision support tailored to the individual. For instance, algorithms combining EHR variables with imaging data have been shown to accurately predict liver fibrosis and MASH, potentially obviating some biopsies [315,325,326,327]. AI-based clinical decision support systems can risk-stratify MASLD patients (e.g., identifying those likely to progress to cirrhosis) and suggest optimal management plans. In metabolic syndrome, ML models have been used to scan large healthcare databases to identify patients with undiagnosed MASH or diabetes risk, alerting providers to intervene earlier. One such study in a Veterans cohort which flagged 12% of at-risk patients as likely undiagnosed MASH, enabling targeted outreach [328]. This kind of AI-driven decision support can improve outcomes by ensuring timely interventions (e.g., weight loss programs or pharmacotherapy) for those who need them most. As these systems evolve, they incorporate more data (wearables, lifestyle, multi-omics) to further personalize care, aligning with precision medicine initiatives in metabolic diseases.
In summary, the pathophysiology of MASH and MASLD has been better understood. These diseases are now a major health problem worldwide, and their rising incidence is linked to rising rates of metabolic syndrome. The understanding of disease mechanisms has improved thanks to developments in imaging techniques, the discovery of genetic markers, and the investigation of inflammatory and metabolic pathways. Yet, there are still significant gaps, particularly in the areas of early intervention, disease stratification, and noninvasive diagnosis. Invasive liver biopsies are still required to accurately differentiate between simple liver steatosis and more severe forms of the disease, such as MASH or advanced fibrosis, which cannot be reliably differentiated using the current biomarkers. There is also a significant unmet need in clinical treatment, as existing therapies are not precise enough to target specific disease mechanisms.
The variability of disease progression in MASLD and MASH research is one of the biggest obstacles and could be a crucial part of future treatment. Simple steatosis can remain stable in some people, but in others, it can quickly progress to more serious forms, such as cirrhosis and hepatocellular carcinoma. This variation in disease progression and our incomplete knowledge of the underlying molecular causes underscore the need for more individualized therapeutic strategies. This trajectory is heavily influenced by genetic, epigenetic, and environmental factors, but little is known about how these factors work together. In addition, the dynamics of the gut–liver axis and its role in aggravating diseases, particularly through mechanisms such as metabolic endotoxemia and microbiota dysbiosis, add further complexity that has not yet been thoroughly investigated.
A hitherto unprecedented opportunity to close these gaps has arisen with the development of multi-omics technologies. Understanding the molecular mechanisms underlying MASLD and MASH can be achieved by integrating data from the fields of genomics, transcriptomics, proteomics, metabolomics, and microbiomes. These technologies shed light on the complex networks of protein interactions, metabolic pathways, gene expression, and microbial influences that lead to liver dysfunction and disease progression. These cutting-edge techniques enable researchers to find new therapeutic targets and biomarkers, which can lead to early detection and more individualized, successful treatments. In addition, by identifying novel drug pathways, multi-omics techniques can help develop targeted therapies that target the underlying causes of liver damage and not just its symptoms.
In parallel with these technological developments, it is crucial to conduct cooperative, multidisciplinary research. Clinical, molecular and bioinformatics expertise must be integrated into future research to fully exploit the potential of multi-omics. These types of partnerships will be critical to gain a better understanding of the various mechanisms underlying MASLD and MASH, including their response to treatment. More effective and economical healthcare solutions could result from identifying patient subgroups who would benefit most from specific therapies through a combined clinical and omics-based approach. The transition from research to clinical practice can also be accelerated by integrating artificial intelligence and machine learning tools to improve predictive modeling of disease progression and treatment outcomes.
Finally, research into MASLD and MASH could result in significant progress in our knowledge of disease mechanisms and the development of efficient therapies. We will gain important new information about the pathophysiology and evolution of these disorders as we continue to explore their complex, multifactorial nature using multi-omics platforms. With these developments, the prospect of precision medicine and more specialized treatment approaches is becoming ever more realistic. Realizing this potential, however, requires consistent, collaborative work to fill current knowledge gaps and promote creative, integrated strategies to address the global burden of MASLD and MASH.

Author Contributions

Conceptualization: M.B., R.M., N.J., E.S., K.A., B.B. and N.A.; methodology: M.B., R.S.M., N.J., E.S., K.A. and B.B.; software: M.B., R.S.M., N.J., E.S., K.A. and B.B.; validation, M.B. and N.A.; formal analysis, M.B.; investigation, M.B., R.S.M., N.J., E.S., K.A., B.B. and N.A.; resources, M.B., R.S.M., N.J., E.S., K.A. and B.B.; data curation, M.B. and N.A.; writing—original draft preparation, M.B., R.S.M., N.J., E.S., K.A. and B.B.; writing—review and editing, M.B., R.S.M. and N.A.; visualization, M.B. and R.S.M.; supervision, M.B., R.S.M. and N.A.; project administration, M.B. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Acknowledgments

The references cited in this manuscript were selected through a comprehensive search strategy across several databases, including PubMed, Scopus, and Google Scholar, focusing on studies published in the last 10 years. In a letter to the editor of the Journal of Hepatology, 99.7% of MASLD patients met the criteria of NAFLD [329]. Keywords such as ‘metabolic dysfunction-associated steatotic liver disease’, ‘metabolic syndrome’, ‘non-alcoholic fatty liver disease’, and ‘therapeutic interventions in NAFLD/MASLD’ were utilized to gather relevant articles. Additionally, studies exploring molecular mechanisms, biomarkers, and emerging therapies for MASLD/NAFLD and MetS were prioritized. In addition, artificial intelligence platforms were utilized into the search strategy, such as ChatGPT-4o and Elicit-Basic to provide published articles within 5 years in specific high-impact factor journals (Hepatology, Journal of Hepatology, The Lancet, Jama, Circulation, Nature, and New England Journal of Medicine (NEJM).

Conflicts of Interest

The authors declare no conflicts of interest.

Abbreviations

ACCAcetyl-CoA carboxylase
ACC1Acetyl-CoA carboxylase 1 (Cytosolic Isoform)
ACC2Acetyl-CoA carboxylase 2 (Mitochondrial Isoform)
AHIApnea–hypopnea index
AKTProtein kinase B
ALDAlcoholic liver disease
ALTAlanine aminotransferase
AMPKAMP-activated protein kinase
Apo AIApolipoprotein A-I
ASTAspartate aminotransferase
BABile acid
BALANCEDA study name related to FGF21
BMDBone mineral density
BMIBody mass index
BSEPBile salt export pump
CADCoronary artery disease
CAFLDCombined causes of fatty liver disease
CAPECaffeic acid phenethyl ester
CCN2Cellular communication network 2 (another name for CTGF)
CCR2/CCR5C-C chemokine receptors 2 and 5
Ch-REBPCarbohydrate response element-binding protein
CIHChronic Intermittent Hypoxia
CK18Cytokeratin 18
CKDChronic kidney disease
CRPC-reactive Protein
CTGFConnective tissue growth factor
CVDCardiovascular disease
CYP7A1Cholesterol 7 Alpha-Hydroxylase
CYP8B1Cytochrome P450 Family 8 Subfamily B Member 1
DGAT2Diacylglycerol O-acyltransferase 2
DIO1Deiodinase Type 1
DNLDe novo lipogenesis
DPP-4Dipeptidyl Peptidase-4
DSFDisulfiram
EATEpicardial adipose tissue
ECMExtracellular matrix
EpigenomeThe chemical changes to DNA that regulate gene expression
EREndoplasmic reticulum
ERKExtracellular signal-regulated kinase
ERK1/2Extracellular signal-regulated kinase ½
ESFLDEnd-stage fatty liver disease
ESKDEnd-stage kidney disease
FAFLDAdvanced stage fibrotic fatty liver disease
FDAFood and Drug Administration
FFA sFree fatty acids
FGF15Fibroblast growth factor 15
FGF19Fibroblast growth factor 19
FGF21Fibroblast growth factor 21
FGFR4Fibroblast growth factor receptor 4
FLDFatty Liver Disease
FMTFecal Microbiota Transplantation
FXRFarnesoid X receptor
GAFLDGenetics-associated fatty liver disease
GCKRGlucokinase regulatory protein (Gene)
GenomeThe complete set of genes or genetic material in an organism
GIPGlucose-Dependent Insulinotropic Polypeptide
GLP-1Glucagon-Like Peptide-1
GS-0976Firsocostat (Gilead’s ACC inhibitor)
H2SHydrogen sulphide
HARMONYA phase 2 trial name
HCCHepatocellular carcinoma
HDL-CHigh-density lipoprotein cholesterol
HMG-CoA3-Hydroxy-3-Methylglutaryl-Coenzyme A
HOMA-IRHomeostasis Model Assessment of Insulin Resistance
HSCHepatic stellate cells
Hs-CRPHighly Sensitive C-Reactive Protein
HSCsHepatic stellate cells
IBSIrritable bowel syndrome
IL-18Interleukin-18
IL-1βInterleukin-1β
IL-31Interleukin 31
IL-6Interleukin-6
IRInsulin resistance
JNKc-Jun N-terminal Kinase
LAFLDLipodystrophy-associated fatty liver disease
LDL-CLow-density lipoprotein cholesterol
LDL-RLow-density lipoprotein receptor
LOXLysyl oxidase
LOXL2Lysyl oxidase Like 2
LPOLipid peroxidation
LPSLipopolysaccharide
LSECsLiver sinusoidal endothelial cells
M1Macrophage Type 1 (Pro-inflammatory)
MASLDMetabolic dysfunction-associated fatty liver disease
MASHMetabolic dysfunction-associated steatohepatitis
lMASLDMetabolic dysfunction-associated steatotic liver disease
MBOAT7Membrane Bound O-acyltransferase Domain-Containing 7 (Gene)
MCP-1Monocyte Chemoattractant Protein-1
MetabolomeThe complete set of small-molecule metabolites in an organism
MetSMetabolic syndrome
MicrobiomeThe community of microorganisms living in and on the body
MK-4074An MSD drug (ACC inhibitor)
mTORMechanistic Target of Rapamycin
MUFAMono-unsaturated fatty acids
NAFLDNon-alcoholic fatty liver disease
NASNon-Alcoholic Fatty Liver Disease Activity Score
NASHNon-alcoholic steatohepatitis
NLRP3NACHT, LRR and PYD domains-containing protein 3 (inflammasome)
NONitric oxide
NTCPSodium-Taurocholate Cotransporting Polypeptide
OAFLDObesity-associated fatty liver disease
OATP1Organic Anion Transporter Polypeptide 1
OCAObeticholic acid
OSAObstructive Sleep Apnea
PDGFPlatelet-derived growth factor
PF-05175157A Pfizer drug (ACC1 inhibitor)
PF-05221304A Pfizer drug (ACC2 inhibitor)
PI3KPhosphoinositide 3-Kinase
PNPLA3Patatin-Like Phospholipase Domain-Containing 3 (Gene)
PON1Paraoxonase 1
PPAR-αPeroxisome Proliferator-Activated Receptor-α
PROC3,4,8Proinflammatory Cytokine 3, 4, and 8
ProteomeThe entire set of proteins produced by an organism
PUFAPolyunsaturated fatty acids
RasRat Sarcoma (a family of proteins)
RhoARas Homolog Family Member A
ROSReactive oxygen species
SAFLDSarcopenia-associated fatty liver disease
SCFA sShort-chain fatty acids
SGLTSodium-Glucose Cotransporter
SGLT1Sodium-Glucose Cotransporter 1
SGLT2Sodium-Glucose Cotransporter 2
SHPSmall heterodimer partner
SIRT1Sirtuin 1
SmadSuppressor of Mothers against Decapentaplegic
SNPsSingle Nucleotide Polymorphisms (genetic variations)
SREBP-1cSterol regulatory element-binding protein 1c
SSB sSugar-sweetened beverages
T2DType 2 diabetes
T3Triiodothyronine
T4Thyroxine
TCTotal cholesterol
TGTriglycerides
TGF-βTransforming growth factor beta
TGR5G Protein-Coupled Bile Acid Receptor 1
THR-αThyroid hormone receptor alpha
THR-βThyroid hormone receptor beta
TLR4Toll-Like Receptor 4
TM6SF2Transmembrane 6 Superfamily Member 2 (Gene)
TNF-αTumor necrosis factor alpha
TranscriptomeThe full range of messenger RNA molecules expressed by an organism
TSBTotal Stool Bacteria
TSHThyroid-stimulating hormone
TZDThiazolidinediones
UPRUnfolded protein response
Vitamin DVitamin D (commonly referred in discussions about bone health)
XAFLDFatty liver disease of multiple/unknown causes
β-oxidationBeta-Oxidation
β-trophinBeta-Trophin

References

  1. Eslam, M.; Sanyal, A.J.; George, J.; on behalf of the International Consensus Panel. MAFLD: A Consensus-Driven Proposed Nomenclature for Metabolic Associated Fatty Liver Disease. Gastroenterology 2020, 158, 1999–2014.e1. [Google Scholar] [CrossRef] [PubMed]
  2. Saklayen, M.G. The Global Epidemic of the Metabolic Syndrome. Curr. Hypertens. Rep. 2018, 20, 12. [Google Scholar] [CrossRef] [PubMed]
  3. Rich, N.E.; Oji, S.; Mufti, A.R.; Browning, J.D.; Parikh, N.D.; Odewole, M.; Mayo, H.; Singal, A.G. Racial and Ethnic Disparities in Nonalcoholic Fatty Liver Disease Prevalence, Severity, and Outcomes in the United States: A Systematic Review and Meta-analysis. Clin. Gastroenterol. Hepatol. Off. Clin. Pract. J. Am. Gastroenterol. Assoc. 2018, 16, 198–210.e2. [Google Scholar] [CrossRef] [PubMed]
  4. Lazo, M.; Hernaez, R.; Eberhardt, M.S.; Bonekamp, S.; Kamel, I.; Guallar, E.; Koteish, A.; Brancati, F.L.; Clark, J.M. Prevalence of nonalcoholic fatty liver disease in the United States: The Third National Health and Nutrition Examination Survey, 1988-1994. Am. J. Epidemiol. 2013, 178, 38–45. [Google Scholar] [CrossRef]
  5. Huang, Q.; Zou, X.; Wen, X.; Zhou, X.; Ji, L. NAFLD or MAFLD: Which Has Closer Association With All-Cause and Cause-Specific Mortality?-Results From NHANES III. Front. Med. 2021, 8, 693507. [Google Scholar] [CrossRef]
  6. Zou, B.; Yeo, Y.H.; Nguyen, V.H.; Cheung, R.; Ingelsson, E.; Nguyen, M.H. Prevalence, characteristics and mortality outcomes of obese, nonobese and lean NAFLD in the United States, 1999–2016. J. Intern. Med. 2020, 288, 139–151. [Google Scholar] [CrossRef]
  7. Li, J.; Zou, B.; Yeo, Y.H.; Feng, Y.; Xie, X.; Lee, D.H.; Fujii, H.; Wu, Y.; Kam, L.Y.; Ji, F.; et al. Prevalence, incidence, and outcome of non-alcoholic fatty liver disease in Asia, 1999-2019: A systematic review and meta-analysis. Lancet Gastroenterol. Hepatol. 2019, 4, 389–398. [Google Scholar] [CrossRef]
  8. Estes, C.; Anstee, Q.M.; Arias-Loste, M.T.; Bantel, H.; Bellentani, S.; Caballeria, J.; Colombo, M.; Craxi, A.; Crespo, J.; Day, C.P.; et al. Modeling NAFLD disease burden in China, France, Germany, Italy, Japan, Spain, United Kingdom, and United States for the period 2016–2030. J. Hepatol. 2018, 69, 896–904. [Google Scholar] [CrossRef]
  9. Huang, D.Q.; El-Serag, H.B.; Loomba, R. Global epidemiology of NAFLD-related HCC: Trends, predictions, risk factors and prevention. Nat. Rev. Gastroenterol. Hepatol. 2021, 18, 223–238. [Google Scholar] [CrossRef]
  10. Byrne, C.D.; Olufadi, R.; Bruce, K.D.; Cagampang, F.R.; Ahmed, M.H. Metabolic disturbances in non-alcoholic fatty liver disease. Clin. Sci. Lond. Engl. 1979 2009, 116, 539–564. [Google Scholar] [CrossRef]
  11. Gastaldelli, A.; Cusi, K. From NASH to diabetes and from diabetes to NASH: Mechanisms and treatment options. JHEP Rep. Innov. Hepatol. 2019, 1, 312–328. [Google Scholar] [CrossRef]
  12. Hotamisligil, G.S. Inflammation and metabolic disorders. Nature 2006, 444, 860–867. [Google Scholar] [CrossRef] [PubMed]
  13. Shulman, G.I. Cellular mechanisms of insulin resistance. J. Clin. Investig. 2000, 106, 171–176. [Google Scholar] [CrossRef] [PubMed]
  14. Czech, M.P. Mechanisms of insulin resistance related to white, beige, and brown adipocytes. Mol. Metab. 2020, 34, 27–42. [Google Scholar] [CrossRef]
  15. Ye, J. Mechanisms of insulin resistance in obesity. Front. Med. 2013, 7, 14–24. [Google Scholar] [CrossRef]
  16. Saltiel, A.R.; Olefsky, J.M. Inflammatory mechanisms linking obesity and metabolic disease. J. Clin. Investig. 2017, 127, 1–4. [Google Scholar] [CrossRef]
  17. Haas, J.T.; Francque, S.; Staels, B. Pathophysiology and Mechanisms of Nonalcoholic Fatty Liver Disease. Annu. Rev. Physiol. 2016, 78, 181–205. [Google Scholar] [CrossRef]
  18. Stefan, N.; Häring, H.-U.; Cusi, K. Non-alcoholic fatty liver disease: Causes, diagnosis, cardiometabolic consequences, and treatment strategies. Lancet Diabetes Endocrinol. 2019, 7, 313–324. [Google Scholar] [CrossRef]
  19. Willner, I.R.; Waters, B.; Patil, S.R.; Reuben, A.; Morelli, J.; Riely, C.A. Ninety patients with nonalcoholic steatohepatitis: Insulin resistance, familial tendency, and severity of disease. Am. J. Gastroenterol. 2001, 96, 2957–2961. [Google Scholar] [CrossRef]
  20. Struben, V.M.D.; Hespenheide, E.E.; Caldwell, S.H. Nonalcoholic steatohepatitis and cryptogenic cirrhosis within kindreds. Am. J. Med. 2000, 108, 9–13. [Google Scholar] [CrossRef]
  21. Zeng, J.; Yang, R.-X.; Sun, C.; Pan, Q.; Zhang, R.-N.; Chen, G.-Y.; Hu, Y.; Fan, J.-G. Prevalence, clinical characteristics, risk factors, and indicators for lean Chinese adults with nonalcoholic fatty liver disease. World J. Gastroenterol. 2020, 26, 1792–1804. [Google Scholar] [CrossRef] [PubMed]
  22. Chen, Y.; Du, X.; Kuppa, A.; Feitosa, M.F.; Bielak, L.F.; O’Connell, J.R.; Musani, S.K.; Guo, X.; Kahali, B.; Chen, V.L.; et al. Genome-wide association meta-analysis identifies 17 loci associated with nonalcoholic fatty liver disease. Nat. Genet. 2023, 55, 1640–1650. [Google Scholar] [CrossRef] [PubMed]
  23. Wong, V.W.-S.; Adams, L.A.; de Lédinghen, V.; Wong, G.L.-H.; Sookoian, S. Noninvasive biomarkers in NAFLD and NASH — current progress and future promise. Nat. Rev. Gastroenterol. Hepatol. 2018, 15, 461–478. [Google Scholar] [CrossRef] [PubMed]
  24. Vilar-Gomez, E.; Yasells-Garcia, A.; Martinez-Perez, Y.; Calzadilla-Bertot, L.; Torres-Gonzalez, A.; Gra-Oramas, B.; Gonzalez-Fabian, L.; Villa-Jimenez, O.; Friedman, S.L.; Diago, M.; et al. Development and validation of a noninvasive prediction model for nonalcoholic steatohepatitis resolution after lifestyle intervention. Hepatology 2016, 63, 1875–1887. [Google Scholar] [CrossRef]
  25. Nam, D.; Chapiro, J.; Paradis, V.; Seraphin, T.P.; Kather, J.N. Artificial intelligence in liver diseases: Improving diagnostics, prognostics and response prediction. JHEP Rep. 2022, 4, 100443. [Google Scholar] [CrossRef]
  26. Lim, S.; Kim, J.-W.; Targher, G. Links between metabolic syndrome and metabolic dysfunction-associated fatty liver disease. Trends Endocrinol. Metab. 2021, 32, 500–514. [Google Scholar] [CrossRef]
  27. Ludwig, J.; Viggiano, T.R.; McGILL, D.B.; Ott, B.J. Nonalcoholic Steatohepatitis Mayo Clinic Experiences With a Hitherto Unnamed Disease. Mayo Clin. Proc. 1980, 55, 434–438. [Google Scholar] [CrossRef]
  28. Alberti, K.G.M.M.; Eckel, R.H.; Grundy, S.M.; Zimmet, P.Z.; Cleeman, J.I.; Donato, K.A.; Fruchart, J.-C.; James, W.P.T.; Loria, C.M.; Smith, S.C.; et al. Harmonizing the metabolic syndrome: A joint interim statement of the International Diabetes Federation Task Force on Epidemiology and Prevention; National Heart, Lung, and Blood Institute; American Heart Association; World Heart Federation; International Atherosclerosis Society; and International Association for the Study of Obesity. Circulation 2009, 120, 1640–1645. [Google Scholar] [CrossRef]
  29. Younossi, Z.M.; Rinella, M.E.; Sanyal, A.J.; Harrison, S.A.; Brunt, E.M.; Goodman, Z.; Cohen, D.E.; Loomba, R. From NAFLD to MAFLD: Implications of a Premature Change in Terminology. Hepatology 2021, 73, 1194–1198. [Google Scholar] [CrossRef]
  30. Stefan, N.; Häring, H.-U. The role of hepatokines in metabolism. Nat. Rev. Endocrinol. 2013, 9, 144–152. [Google Scholar] [CrossRef]
  31. Meex, R.C.R.; Watt, M.J. Hepatokines: Linking nonalcoholic fatty liver disease and insulin resistance. Nat. Rev. Endocrinol. 2017, 13, 509–520. [Google Scholar] [CrossRef] [PubMed]
  32. Spahis, S.; Delvin, E.; Borys, J.-M.; Levy, E. Oxidative Stress as a Critical Factor in Nonalcoholic Fatty Liver Disease Pathogenesis. Antioxid. Redox Signal. 2017, 26, 519–541. [Google Scholar] [CrossRef] [PubMed]
  33. Mridha, A.R.; Wree, A.; Robertson, A.A.B.; Yeh, M.M.; Johnson, C.D.; Van Rooyen, D.M.; Haczeyni, F.; Teoh, N.C.-H.; Savard, C.; Ioannou, G.N.; et al. NLRP3 inflammasome blockade reduces liver inflammation and fibrosis in experimental NASH in mice. J. Hepatol. 2017, 66, 1037–1046. [Google Scholar] [CrossRef] [PubMed]
  34. Polyzos, S.A.; Kountouras, J.; Mantzoros, C.S. Adipokines in nonalcoholic fatty liver disease. Metabolism 2016, 65, 1062–1079. [Google Scholar] [CrossRef]
  35. Lonardo, A.; Nascimbeni, F.; Mantovani, A.; Targher, G. Hypertension, diabetes, atherosclerosis and NASH: Cause or consequence? J. Hepatol. 2018, 68, 335–352. [Google Scholar] [CrossRef]
  36. Longo, M.; Zatterale, F.; Naderi, J.; Parrillo, L.; Formisano, P.; Raciti, G.A.; Beguinot, F.; Miele, C. Adipose Tissue Dysfunction as Determinant of Obesity-Associated Metabolic Complications. Int. J. Mol. Sci. 2019, 20, 2358. [Google Scholar] [CrossRef]
  37. Simões, I.C.M.; Fontes, A.; Pinton, P.; Zischka, H.; Wieckowski, M.R. Mitochondria in non-alcoholic fatty liver disease. Int. J. Biochem. Cell Biol. 2018, 95, 93–99. [Google Scholar] [CrossRef]
  38. Grattagliano, I.; de Bari, O.; Bernardo, T.C.; Oliveira, P.J.; Wang, D.Q.-H.; Portincasa, P. Role of mitochondria in nonalcoholic fatty liver disease--from origin to propagation. Clin. Biochem. 2012, 45, 610–618. [Google Scholar] [CrossRef]
  39. Leung, C.; Rivera, L.; Furness, J.B.; Angus, P.W. The role of the gut microbiota in NAFLD. Nat. Rev. Gastroenterol. Hepatol. 2016, 13, 412–425. [Google Scholar] [CrossRef]
  40. Arab, J.P.; Karpen, S.J.; Dawson, P.A.; Arrese, M.; Trauner, M. Bile acids and nonalcoholic fatty liver disease: Molecular insights and therapeutic perspectives. Hepatology 2017, 65, 350–362. [Google Scholar] [CrossRef]
  41. Koh, A.; Molinaro, A.; Ståhlman, M.; Khan, M.T.; Schmidt, C.; Mannerås-Holm, L.; Wu, H.; Carreras, A.; Jeong, H.; Olofsson, L.E.; et al. Microbially Produced Imidazole Propionate Impairs Insulin Signaling through mTORC1. Cell 2018, 175, 947–961.e17. [Google Scholar] [CrossRef]
  42. Albillos, A.; de Gottardi, A.; Rescigno, M. The gut-liver axis in liver disease: Pathophysiological basis for therapy. J. Hepatol. 2020, 72, 558–577. [Google Scholar] [CrossRef] [PubMed]
  43. Ekstedt, M.; Hagström, H.; Nasr, P.; Fredrikson, M.; Stål, P.; Kechagias, S.; Hultcrantz, R. Fibrosis stage is the strongest predictor for disease-specific mortality in NAFLD after up to 33 years of follow-up. Hepatology 2015, 61, 1547–1554. [Google Scholar] [CrossRef] [PubMed]
  44. Younossi, Z.M.; Golabi, P.; Paik, J.M.; Henry, A.; Van Dongen, C.; Henry, L. The global epidemiology of nonalcoholic fatty liver disease (NAFLD) and nonalcoholic steatohepatitis (NASH): A systematic review. Hepatology 2023, 77, 1335–1347. [Google Scholar] [CrossRef] [PubMed]
  45. Eslam, M.; Newsome, P.N.; Sarin, S.K.; Anstee, Q.M.; Targher, G.; Romero-Gomez, M.; Zelber-Sagi, S.; Wai-Sun Wong, V.; Dufour, J.-F.; Schattenberg, J.M.; et al. A new definition for metabolic dysfunction-associated fatty liver disease: An international expert consensus statement. J. Hepatol. 2020, 73, 202–209. [Google Scholar] [CrossRef]
  46. Mantovani, A.; Petracca, G.; Beatrice, G.; Csermely, A.; Tilg, H.; Byrne, C.D.; Targher, G. Non-alcoholic fatty liver disease and increased risk of incident extrahepatic cancers: A meta-analysis of observational cohort studies. Gut 2022, 71, 778–788. [Google Scholar] [CrossRef]
  47. Mottillo, S.; Filion, K.B.; Genest, J.; Joseph, L.; Pilote, L.; Poirier, P.; Rinfret, S.; Schiffrin, E.L.; Eisenberg, M.J. The metabolic syndrome and cardiovascular risk a systematic review and meta-analysis. J. Am. Coll. Cardiol. 2010, 56, 1113–1132. [Google Scholar] [CrossRef]
  48. Targher, G.; Byrne, C.D.; Lonardo, A.; Zoppini, G.; Barbui, C. Non-alcoholic fatty liver disease and risk of incident cardiovascular disease: A meta-analysis. J. Hepatol. 2016, 65, 589–600. [Google Scholar] [CrossRef]
  49. Brunner, K.T.; Pedley, A.; Massaro, J.M.; Hoffmann, U.; Benjamin, E.J.; Long, M.T. Increasing liver fat is associated with progression of cardiovascular risk factors. Liver Int. Off. J. Int. Assoc. Study Liver 2020, 40, 1339–1343. [Google Scholar] [CrossRef]
  50. Basheer, M.; Saad, E.; Jeries, H.; Assy, N. Liver Fat Storage Is a Better Predictor of Coronary Artery Disease than Visceral Fat. Metabolites 2023, 13, 896. [Google Scholar] [CrossRef]
  51. Eslam, M.; George, J. Refining the role of epicardial adipose tissue in non-alcoholic fatty liver disease. Hepatol. Int. 2019, 13, 662–664. [Google Scholar] [CrossRef]
  52. Gutiérrez-Cuevas, J.; Santos, A.; Armendariz-Borunda, J. Pathophysiological Molecular Mechanisms of Obesity: A Link between MAFLD and NASH with Cardiovascular Diseases. Int. J. Mol. Sci. 2021, 22, 11629. [Google Scholar] [CrossRef] [PubMed]
  53. Adler, M.; Schaffner, F. Fatty liver hepatitis and cirrhosis in obese patients. Am. J. Med. 1979, 67, 811–816. [Google Scholar] [CrossRef]
  54. Ballestri, S.; Lonardo, A.; Bonapace, S.; Byrne, C.D.; Loria, P.; Targher, G. Risk of cardiovascular, cardiac and arrhythmic complications in patients with non-alcoholic fatty liver disease. World J. Gastroenterol. 2014, 20, 1724–1745. [Google Scholar] [CrossRef]
  55. Gaggini, M.; Morelli, M.; Buzzigoli, E.; DeFronzo, R.A.; Bugianesi, E.; Gastaldelli, A. Non-alcoholic fatty liver disease (NAFLD) and its connection with insulin resistance, dyslipidemia, atherosclerosis and coronary heart disease. Nutrients 2013, 5, 1544–1560. [Google Scholar] [CrossRef]
  56. Santos, R.D.; Valenti, L.; Romeo, S. Does nonalcoholic fatty liver disease cause cardiovascular disease? Current knowledge and gaps. Atherosclerosis 2019, 282, 110–120. [Google Scholar] [CrossRef]
  57. Anstee, Q.M.; Mantovani, A.; Tilg, H.; Targher, G. Risk of cardiomyopathy and cardiac arrhythmias in patients with nonalcoholic fatty liver disease. Nat. Rev. Gastroenterol. Hepatol. 2018, 15, 425–439. [Google Scholar] [CrossRef]
  58. Taylor, R.S.; Taylor, R.J.; Bayliss, S.; Hagström, H.; Nasr, P.; Schattenberg, J.M.; Ishigami, M.; Toyoda, H.; Wai-Sun Wong, V.; Peleg, N.; et al. Association Between Fibrosis Stage and Outcomes of Patients With Nonalcoholic Fatty Liver Disease: A Systematic Review and Meta-Analysis. Gastroenterology 2020, 158, 1611–1625.e12. [Google Scholar] [CrossRef]
  59. Vilar-Gomez, E.; Athinarayanan, S.J.; Adams, R.N.; Hallberg, S.J.; Bhanpuri, N.H.; McKenzie, A.L.; Campbell, W.W.; McCarter, J.P.; Phinney, S.D.; Volek, J.S.; et al. Post hoc analyses of surrogate markers of non-alcoholic fatty liver disease (NAFLD) and liver fibrosis in patients with type 2 diabetes in a digitally supported continuous care intervention: An open-label, non-randomised controlled study. BMJ Open 2019, 9, e023597. [Google Scholar] [CrossRef]
  60. Friedman, S.L.; Neuschwander-Tetri, B.A.; Rinella, M.; Sanyal, A.J. Mechanisms of NAFLD development and therapeutic strategies. Nat. Med. 2018, 24, 908–922. [Google Scholar] [CrossRef]
  61. Marra, F.; Svegliati-Baroni, G. Lipotoxicity and the gut-liver axis in NASH pathogenesis. J. Hepatol. 2018, 68, 280–295. [Google Scholar] [CrossRef] [PubMed]
  62. Schwabe, R.F.; Tabas, I.; Pajvani, U.B. Mechanisms of Fibrosis Development in Nonalcoholic Steatohepatitis. Gastroenterology 2020, 158, 1913–1928. [Google Scholar] [CrossRef] [PubMed]
  63. Arab, J.P.; Izzy, M.; Leggio, L.; Bataller, R.; Shah, V.H. Management of alcohol use disorder in patients with cirrhosis in the setting of liver transplantation. Nat. Rev. Gastroenterol. Hepatol. 2022, 19, 45–59. [Google Scholar] [CrossRef] [PubMed]
  64. Kleiner, D.E.; Brunt, E.M.; Wilson, L.A.; Behling, C.; Guy, C.; Contos, M.; Cummings, O.; Yeh, M.; Gill, R.; Chalasani, N.; et al. Association of Histologic Disease Activity With Progression of Nonalcoholic Fatty Liver Disease. JAMA Netw. Open 2019, 2, e1912565. [Google Scholar] [CrossRef]
  65. Zhao, Y.; Zhao, W.; Ma, J.; Toshiyoshi, M.; Zhao, Y. Patatin-like phospholipase domain-containing 3 gene (PNPLA3) polymorphic (rs738409) single nucleotide polymorphisms and susceptibility to nonalcoholic fatty liver disease: A meta-analysis of twenty studies. Medicine 2023, 102, e33110. [Google Scholar] [CrossRef]
  66. Tian, L.-Y.; Smit, D.J.; Jücker, M. The Role of PI3K/AKT/mTOR Signaling in Hepatocellular Carcinoma Metabolism. Int. J. Mol. Sci. 2023, 24, 2652. [Google Scholar] [CrossRef]
  67. Anstee, Q.M.; Darlay, R.; Cockell, S.; Meroni, M.; Govaere, O.; Tiniakos, D.; Burt, A.D.; Bedossa, P.; Palmer, J.; Liu, Y.-L.; et al. Genome-wide association study of non-alcoholic fatty liver and steatohepatitis in a histologically characterised cohort☆. J. Hepatol. 2020, 73, 505–515. [Google Scholar] [CrossRef]
  68. Wong, V.W.-S.; Wong, G.L.-H.; Woo, J.; Abrigo, J.M.; Chan, C.K.-M.; Shu, S.S.-T.; Leung, J.K.-Y.; Chim, A.M.-L.; Kong, A.P.-S.; Lui, G.C.-Y.; et al. Impact of the New Definition of Metabolic Associated Fatty Liver Disease on the Epidemiology of the Disease. Clin. Gastroenterol. Hepatol. Off. Clin. Pract. J. Am. Gastroenterol. Assoc. 2021, 19, 2161–2171.e5. [Google Scholar] [CrossRef]
  69. Mantovani, A.; Byrne, C.D.; Bonora, E.; Targher, G. Nonalcoholic Fatty Liver Disease and Risk of Incident Type 2 Diabetes: A Meta-analysis. Diabetes Care 2018, 41, 372–382. [Google Scholar] [CrossRef]
  70. Bhat, N.; Mani, A. Dysregulation of Lipid and Glucose Metabolism in Nonalcoholic Fatty Liver Disease. Nutrients 2023, 15, 2323. [Google Scholar] [CrossRef]
  71. Sakurai, Y.; Kubota, N.; Yamauchi, T.; Kadowaki, T. Role of Insulin Resistance in MAFLD. Int. J. Mol. Sci. 2021, 22, 4156. [Google Scholar] [CrossRef] [PubMed]
  72. Zhou, X.-D.; Cai, J.; Targher, G.; Byrne, C.D.; Shapiro, M.D.; Sung, K.-C.; Somers, V.K.; Chahal, C.A.A.; George, J.; Chen, L.-L.; et al. Metabolic dysfunction-associated fatty liver disease and implications for cardiovascular risk and disease prevention. Cardiovasc. Diabetol. 2022, 21, 270. [Google Scholar] [CrossRef] [PubMed]
  73. Kim, H.; Lee, D.S.; An, T.H.; Park, H.-J.; Kim, W.K.; Bae, K.-H.; Oh, K.-J. Metabolic Spectrum of Liver Failure in Type 2 Diabetes and Obesity: From NAFLD to NASH to HCC. Int. J. Mol. Sci. 2021, 22, 4495. [Google Scholar] [CrossRef] [PubMed]
  74. Hudish, L.I.; Reusch, J.E.; Sussel, L. β Cell dysfunction during progression of metabolic syndrome to type 2 diabetes. J. Clin. Investig. 2019, 129, 4001–4008. [Google Scholar] [CrossRef]
  75. Heindel, J.J.; Blumberg, B.; Cave, M.; Machtinger, R.; Mantovani, A.; Mendez, M.A.; Nadal, A.; Palanza, P.; Panzica, G.; Sargis, R.; et al. Metabolism disrupting chemicals and metabolic disorders. Reprod. Toxicol. 2017, 68, 3–33. [Google Scholar] [CrossRef]
  76. Vilas-Boas, E.A.; Almeida, D.C.; Roma, L.P.; Ortis, F.; Carpinelli, A.R. Lipotoxicity and β-Cell Failure in Type 2 Diabetes: Oxidative Stress Linked to NADPH Oxidase and ER Stress. Cells 2021, 10, 3328. [Google Scholar] [CrossRef]
  77. Lu, Y.; Wang, W.; Liu, J.; Xie, M.; Liu, Q.; Li, S. Vascular complications of diabetes: A narrative review. Medicine 2023, 102, e35285. [Google Scholar] [CrossRef]
  78. Zhou, Z.; Sun, B.; Huang, S.; Zhu, C.; Bian, M. Glycemic variability: Adverse clinical outcomes and how to improve it? Cardiovasc. Diabetol. 2020, 19, 102. [Google Scholar] [CrossRef]
  79. Ferdous, S.-E.; Ferrell, J.M. Pathophysiological Relationship between Type 2 Diabetes Mellitus and Metabolic Dysfunction-Associated Steatotic Liver Disease: Novel Therapeutic Approaches. Int. J. Mol. Sci. 2024, 25, 8731. [Google Scholar] [CrossRef]
  80. Musso, G.; Gambino, R.; Tabibian, J.H.; Ekstedt, M.; Kechagias, S.; Hamaguchi, M.; Hultcrantz, R.; Hagström, H.; Yoon, S.K.; Charatcharoenwitthaya, P.; et al. Association of non-alcoholic fatty liver disease with chronic kidney disease: A systematic review and meta-analysis. PLoS Med. 2014, 11, e1001680. [Google Scholar] [CrossRef]
  81. Chen, S.; Pang, J.; Huang, R.; Xue, H.; Chen, X. Association of MAFLD with end-stage kidney disease: A prospective study of 337,783 UK Biobank participants. Hepatol. Int. 2023, 17, 595–605. [Google Scholar] [CrossRef] [PubMed]
  82. Zhang, H.-J.; Wang, Y.-Y.; Chen, C.; Lu, Y.-L.; Wang, N.-J. Cardiovascular and renal burdens of metabolic associated fatty liver disease from serial US national surveys, 1999–2016. Chin. Med. J. 2021, 134, 1593–1601. [Google Scholar] [CrossRef] [PubMed]
  83. Byrne, C.D.; Targher, G. NAFLD as a driver of chronic kidney disease. J. Hepatol. 2020, 72, 785–801. [Google Scholar] [CrossRef]
  84. Tang, H.; Lv, F.; Zhang, P.; Liu, J.; Mao, J. The impact of obstructive sleep apnea on nonalcoholic fatty liver disease. Front. Endocrinol. 2023, 14, 1254459. [Google Scholar] [CrossRef]
  85. Aron-Wisnewsky, J.; Clement, K.; Pépin, J.-L. Nonalcoholic fatty liver disease and obstructive sleep apnea. Metabolism 2016, 65, 1124–1135. [Google Scholar] [CrossRef]
  86. Influence of Obstructive Sleep Apnea on Fatty Liver Disease: Role of Chronic Intermittent Hypoxia—PubMed. Available online: https://pubmed.ncbi.nlm.nih.gov/21762556/ (accessed on 6 February 2025).
  87. Sookoian, S.; Pirola, C.J. Obstructive sleep apnea is associated with fatty liver and abnormal liver enzymes: A meta-analysis. Obes. Surg. 2013, 23, 1815–1825. [Google Scholar] [CrossRef]
  88. Aron-Wisnewsky, J.; Minville, C.; Tordjman, J.; Lévy, P.; Bouillot, J.-L.; Basdevant, A.; Bedossa, P.; Clément, K.; Pépin, J.-L. Chronic intermittent hypoxia is a major trigger for non-alcoholic fatty liver disease in morbid obese. J. Hepatol. 2012, 56, 225–233. [Google Scholar] [CrossRef]
  89. Wang, D.; Si, D.; Li, G.; Ding, Z.; Yang, X.; Gao, C. Dysregulated autophagic activity induced in response to chronic intermittent hypoxia contributes to the pathogenesis of NAFLD. Front. Physiol. 2022, 13, 941706. [Google Scholar] [CrossRef]
  90. Global ETD Search. Available online: http://search.ndltd.org/show.php?id=oai%3Aunion.ndltd.org%3Abu.edu%2Foai%3Aopen.bu.edu%3A2144%2F31230&back=http%3A%2F%2Fsearch.ndltd.org%2Fsearch.php%3Fq%3Dsubject%253A%2522OSA%2522%26start%3D10 (accessed on 6 February 2025).
  91. NASH in Lean Individuals—PubMed. Available online: https://pubmed.ncbi.nlm.nih.gov/30654392/ (accessed on 6 February 2025).
  92. Mesarwi, O.A.; Shin, M.-K.; Drager, L.F.; Bevans-Fonti, S.; Jun, J.C.; Putcha, N.; Torbenson, M.S.; Pedrosa, R.P.; Lorenzi-Filho, G.; Steele, K.E.; et al. Lysyl Oxidase as a Serum Biomarker of Liver Fibrosis in Patients with Severe Obesity and Obstructive Sleep Apnea. Sleep 2015, 38, 1583–1591. [Google Scholar] [CrossRef]
  93. Ji, Y.; Liang, Y.; Mak, J.C.W.; Ip, M.S.M. Obstructive sleep apnea, intermittent hypoxia and non-alcoholic fatty liver disease. Sleep Med. 2022, 95, 16–28. [Google Scholar] [CrossRef]
  94. Briançon-Marjollet, A.; Weiszenstein, M.; Henri, M.; Thomas, A.; Godin-Ribuot, D.; Polak, J. The impact of sleep disorders on glucose metabolism: Endocrine and molecular mechanisms. Diabetol. Metab. Syndr. 2015, 7, 25. [Google Scholar] [CrossRef] [PubMed]
  95. Li, J.; Grigoryev, D.N.; Ye, S.Q.; Thorne, L.; Schwartz, A.R.; Smith, P.L.; O’Donnell, C.P.; Polotsky, V.Y. Chronic intermittent hypoxia upregulates genes of lipid biosynthesis in obese mice. J. Appl. Physiol. Bethesda Md 1985 2005, 99, 1643–1648. [Google Scholar] [CrossRef]
  96. Liu, J.; Li, W.; Zhu, W.; He, W.; Zhao, H.; Xiang, Y.; Liu, C.; Wu, W. Chronic intermittent hypoxia promotes the development of experimental non-alcoholic steatohepatitis by modulating Treg/Th17 differentiation. Acta Biochim. Biophys. Sin. 2018, 50, 1200–1210. [Google Scholar] [CrossRef] [PubMed]
  97. Liu, J.; Zhang, J.; Lu, G.; Qiu, Y.; Yang, D.; Yin, G.; Zhang, X. The effect of oxidative stress in myocardial cell injury in mice exposed to chronic intermittent hypoxia. Chin. Med. J. 2010, 123, 74–78. [Google Scholar]
  98. Yan, Y.R.; Zhang, L.; Lin, Y.N.; Sun, X.W.; Ding, Y.J.; Li, N.; Li, H.P.; Li, S.Q.; Zhou, J.P.; Li, Q.Y. Chronic intermittent hypoxia-induced mitochondrial dysfunction mediates endothelial injury via the TXNIP/NLRP3/IL-1β signaling pathway. Free Radic. Biol. Med. 2021, 165, 401–410. [Google Scholar] [CrossRef]
  99. Aron-Wisnewsky, J.; Pepin, J.-L. New insights in the pathophysiology of chronic intermittent hypoxia-induced NASH: The role of gut-liver axis impairment. Thorax 2015, 70, 713–715. [Google Scholar] [CrossRef]
  100. Pardee, P.E.; Dunn, W.; Schwimmer, J.B. Non-alcoholic fatty liver disease is associated with low bone mineral density in obese children. Aliment. Pharmacol. Ther. 2012, 35, 248–254. [Google Scholar] [CrossRef]
  101. Moon, S.-S.; Lee, Y.-S.; Kim, S.W. Association of nonalcoholic fatty liver disease with low bone mass in postmenopausal women. Endocrine 2012, 42, 423–429. [Google Scholar] [CrossRef]
  102. Cui, R.; Sheng, H.; Rui, X.-F.; Cheng, X.-Y.; Sheng, C.-J.; Wang, J.-Y.; Qu, S. Low bone mineral density in chinese adults with nonalcoholic Fatty liver disease. Int. J. Endocrinol. 2013, 2013, 396545. [Google Scholar] [CrossRef]
  103. Barchetta, I.; Cimini, F.A.; Cavallo, M.G. Vitamin D and Metabolic Dysfunction-Associated Fatty Liver Disease (MAFLD): An Update. Nutrients 2020, 12, 3302. [Google Scholar] [CrossRef]
  104. Kwok, R.M.; Torres, D.M.; Harrison, S.A. Vitamin D and nonalcoholic fatty liver disease (NAFLD): Is it more than just an association? Hepatology 2013, 58, 1166–1174. [Google Scholar] [CrossRef] [PubMed]
  105. Drapkina, O.M.; Elkina, A.Y.; Sheptulina, A.F.; Kiselev, A.R. Non-Alcoholic Fatty Liver Disease and Bone Tissue Metabolism: Current Findings and Future Perspectives. Int. J. Mol. Sci. 2023, 24, 8445. [Google Scholar] [CrossRef] [PubMed]
  106. Filip, R.; Radzki, R.P.; Bieńko, M. Novel insights into the relationship between nonalcoholic fatty liver disease and osteoporosis. Clin. Interv. Aging 2018, 13, 1879–1891. [Google Scholar] [CrossRef] [PubMed]
  107. Santos, L.A.A.; Romeiro, F.G. Diagnosis and Management of Cirrhosis-Related Osteoporosis. BioMed Res. Int. 2016, 2016, 1423462. [Google Scholar] [CrossRef]
  108. Nakchbandi, I.A. Osteoporosis and fractures in liver disease: Relevance, pathogenesis and therapeutic implications. World J. Gastroenterol. 2014, 20, 9427–9438. [Google Scholar] [CrossRef]
  109. Azziz, R.; Carmina, E.; Chen, Z.; Dunaif, A.; Laven, J.S.E.; Legro, R.S.; Lizneva, D.; Natterson-Horowtiz, B.; Teede, H.J.; Yildiz, B.O. Polycystic ovary syndrome. Nat. Rev. Dis. Primers 2016, 2, 16057. [Google Scholar] [CrossRef]
  110. Hutchison, A.L.; Tavaglione, F.; Romeo, S.; Charlton, M. Endocrine aspects of metabolic dysfunction-associated steatotic liver disease (MASLD): Beyond insulin resistance. J. Hepatol. 2023, 79, 1524–1541. [Google Scholar] [CrossRef]
  111. Liu, D.; Gao, X.; Pan, X.-F.; Zhou, T.; Zhu, C.; Li, F.; Fan, J.-G.; Targher, G.; Zhao, J. The hepato-ovarian axis: Genetic evidence for a causal association between non-alcoholic fatty liver disease and polycystic ovary syndrome. BMC Med. 2023, 21, 62. [Google Scholar] [CrossRef]
  112. Mullur, R.; Liu, Y.-Y.; Brent, G.A. Thyroid Hormone Regulation of Metabolism. Physiol. Rev. 2014, 94, 355–382. [Google Scholar] [CrossRef]
  113. Zhang, J.-Z.; Cai, J.-J.; Yu, Y.; She, Z.-G.; Li, H. Nonalcoholic Fatty Liver Disease: An Update on the Diagnosis. Gene Expr. 2019, 19, 187–198. [Google Scholar] [CrossRef]
  114. Josloff, K.; Beiriger, J.; Khan, A.; Gawel, R.J.; Kirby, R.S.; Kendrick, A.D.; Rao, A.K.; Wang, R.X.; Schafer, M.M.; Pearce, M.E.; et al. Comprehensive Review of Cardiovascular Disease Risk in Nonalcoholic Fatty Liver Disease. J. Cardiovasc. Dev. Dis. 2022, 9, 419. [Google Scholar] [CrossRef] [PubMed]
  115. Pirola, C.J.; Sookoian, S. Personalized medicine in nonalcoholic fatty liver disease. Clin. Mol. Hepatol. 2022, 28, 935–938. [Google Scholar] [CrossRef] [PubMed]
  116. Rosato, V.; Masarone, M.; Dallio, M.; Federico, A.; Aglitti, A.; Persico, M. NAFLD and Extra-Hepatic Comorbidities: Current Evidence on a Multi-Organ Metabolic Syndrome. Int. J. Environ. Res. Public. Health 2019, 16, 3415. [Google Scholar] [CrossRef]
  117. Vilar-Gomez, E.; Martinez-Perez, Y.; Calzadilla-Bertot, L.; Torres-Gonzalez, A.; Gra-Oramas, B.; Gonzalez-Fabian, L.; Friedman, S.L.; Diago, M.; Romero-Gomez, M. Weight Loss Through Lifestyle Modification Significantly Reduces Features of Nonalcoholic Steatohepatitis. Gastroenterology 2015, 149, 367–378.e5; quiz e14–e15. [Google Scholar] [CrossRef]
  118. European Association for the Study of the Liver (EASL); European Association for the Study of Diabetes (EASD); European Association for the Study of Obesity (EASO) EASL-EASD-EASO Clinical Practice Guidelines for the management of non-alcoholic fatty liver disease. J. Hepatol. 2016, 64, 1388–1402. [CrossRef]
  119. Zelber-Sagi, S.; Salomone, F.; Mlynarsky, L. The Mediterranean dietary pattern as the diet of choice for non-alcoholic fatty liver disease: Evidence and plausible mechanisms. Liver Int. Off. J. Int. Assoc. Study Liver 2017, 37, 936–949. [Google Scholar] [CrossRef]
  120. Montemayor, S.; Mascaró, C.M.; Ugarriza, L.; Casares, M.; Llompart, I.; Abete, I.; Zulet, M.Á.; Martínez, J.A.; Tur, J.A.; Bouzas, C. Adherence to Mediterranean Diet and NAFLD in Patients with Metabolic Syndrome: The FLIPAN Study. Nutrients 2022, 14, 3186. [Google Scholar] [CrossRef]
  121. Plaz Torres, M.C.; Aghemo, A.; Lleo, A.; Bodini, G.; Furnari, M.; Marabotto, E.; Miele, L.; Giannini, E.G. Mediterranean Diet and NAFLD: What We Know and Questions That Still Need to Be Answered. Nutrients 2019, 11, 2971. [Google Scholar] [CrossRef]
  122. Anton, S.D.; Moehl, K.; Donahoo, W.T.; Marosi, K.; Lee, S.; Mainous, A.G.; Leeuwenburgh, C.; Mattson, M.P. Flipping the Metabolic Switch: Understanding and Applying Health Benefits of Fasting. Obes. Silver Spring Md. 2018, 26, 254–268. [Google Scholar] [CrossRef]
  123. Diab, R.; Dimachkie, L.; Zein, O.; Dakroub, A.; Eid, A.H. Intermittent Fasting Regulates Metabolic Homeostasis and Improves Cardiovascular Health. Cell Biochem. Biophys. 2024, 82, 1583–1597. [Google Scholar] [CrossRef]
  124. von Loeffelholz, C.; Roth, J.; Coldewey, S.M.; Birkenfeld, A.L. The Role of Physical Activity in Nonalcoholic and Metabolic Dysfunction Associated Fatty Liver Disease. Biomedicines 2021, 9, 1853. [Google Scholar] [CrossRef] [PubMed]
  125. Mika, A.; Macaluso, F.; Barone, R.; Di Felice, V.; Sledzinski, T. Effect of Exercise on Fatty Acid Metabolism and Adipokine Secretion in Adipose Tissue. Front. Physiol. 2019, 10, 26. [Google Scholar] [CrossRef]
  126. Gleeson, M.; Bishop, N.C.; Stensel, D.J.; Lindley, M.R.; Mastana, S.S.; Nimmo, M.A. The anti-inflammatory effects of exercise: Mechanisms and implications for the prevention and treatment of disease. Nat. Rev. Immunol. 2011, 11, 607–615. [Google Scholar] [CrossRef] [PubMed]
  127. Hickman, I.J.; Jonsson, J.R.; Prins, J.B.; Ash, S.; Purdie, D.M.; Clouston, A.D.; Powell, E.E. Modest weight loss and physical activity in overweight patients with chronic liver disease results in sustained improvements in alanine aminotransferase, fasting insulin, and quality of life. Gut 2004, 53, 413–419. [Google Scholar] [CrossRef]
  128. Kirwan, J.P.; Sacks, J.; Nieuwoudt, S. The essential role of exercise in the management of type 2 diabetes. Cleve. Clin. J. Med. 2017, 84, S15–S21. [Google Scholar] [CrossRef]
  129. Van Der Heijden, G.-J.; Wang, Z.J.; Chu, Z.; Toffolo, G.; Manesso, E.; Sauer, P.J.J.; Sunehag, A.L. Strength exercise improves muscle mass and hepatic insulin sensitivity in obese youth. Med. Sci. Sports Exerc. 2010, 42, 1973–1980. [Google Scholar] [CrossRef]
  130. Eckard, C.; Cole, R.; Lockwood, J.; Torres, D.M.; Williams, C.D.; Shaw, J.C.; Harrison, S.A. Prospective histopathologic evaluation of lifestyle modification in nonalcoholic fatty liver disease: A randomized trial. Ther. Adv. Gastroenterol. 2013, 6, 249–259. [Google Scholar] [CrossRef]
  131. Mazza, A.; Fruci, B.; Garinis, G.A.; Giuliano, S.; Malaguarnera, R.; Belfiore, A. The role of metformin in the management of NAFLD. Exp. Diabetes Res. 2012, 2012, 716404. [Google Scholar] [CrossRef]
  132. Huang, Y.; Wang, X.; Yan, C.; Li, C.; Zhang, L.; Zhang, L.; Liang, E.; Liu, T.; Mao, J. Effect of metformin on nonalcoholic fatty liver based on meta-analysis and network pharmacology. Medicine 2022, 101, e31437. [Google Scholar] [CrossRef]
  133. Haukeland, J.W.; Konopski, Z.; Eggesbø, H.B.; von Volkmann, H.L.; Raschpichler, G.; Bjøro, K.; Haaland, T.; Løberg, E.M.; Birkeland, K. Metformin in patients with non-alcoholic fatty liver disease: A randomized, controlled trial. Scand. J. Gastroenterol. 2009, 44, 853–860. [Google Scholar] [CrossRef]
  134. Bugianesi, E.; McCullough, A.J.; Marchesini, G. Insulin resistance: A metabolic pathway to chronic liver disease. Hepatology 2005, 42, 987–1000. [Google Scholar] [CrossRef] [PubMed]
  135. Horn, C.L.; Morales, A.L.; Savard, C.; Farrell, G.C.; Ioannou, G.N. Role of Cholesterol-Associated Steatohepatitis in the Development of NASH. Hepatol. Commun. 2022, 6, 12–35. [Google Scholar] [CrossRef] [PubMed]
  136. Li, M.; Chi, X.; Wang, Y.; Setrerrahmane, S.; Xie, W.; Xu, H. Trends in insulin resistance: Insights into mechanisms and therapeutic strategy. Signal Transduct. Target. Ther. 2022, 7, 1–25. [Google Scholar] [CrossRef] [PubMed]
  137. Feng, W.; Gao, C.; Bi, Y.; Wu, M.; Li, P.; Shen, S.; Chen, W.; Yin, T.; Zhu, D. Randomized trial comparing the effects of gliclazide, liraglutide, and metformin on diabetes with non-alcoholic fatty liver disease. J. Diabetes 2017, 9, 800–809. [Google Scholar] [CrossRef]
  138. Yan, J.; Yao, B.; Kuang, H.; Yang, X.; Huang, Q.; Hong, T.; Li, Y.; Dou, J.; Yang, W.; Qin, G.; et al. Liraglutide, Sitagliptin, and Insulin Glargine Added to Metformin: The Effect on Body Weight and Intrahepatic Lipid in Patients With Type 2 Diabetes Mellitus and Nonalcoholic Fatty Liver Disease. Hepatology 2019, 69, 2414–2426. [Google Scholar] [CrossRef]
  139. Koureta, E.; Cholongitas, E. Combination therapies in nonalcoholic fatty liver disease using antidiabetic and disease-specific drugs. Ann. Gastroenterol. 2023, 36, 378–391. [Google Scholar] [CrossRef]
  140. Kersten, S. Integrated physiology and systems biology of PPARα. Mol. Metab. 2014, 3, 354–371. [Google Scholar] [CrossRef]
  141. Molecular Actions of PPARα in Lipid Metabolism and Inflammation|Endocrine Reviews|Oxford Academic. Available online: https://academic.oup.com/edrv/article/39/5/760/5055100 (accessed on 7 February 2025).
  142. Nakagawa, Y.; Satoh, A.; Tezuka, H.; Han, S.; Takei, K.; Iwasaki, H.; Yatoh, S.; Yahagi, N.; Suzuki, H.; Iwasaki, Y.; et al. CREB3L3 controls fatty acid oxidation and ketogenesis in synergy with PPARα. Sci. Rep. 2016, 6, 39182. [Google Scholar] [CrossRef]
  143. Orasanu, G.; Ziouzenkova, O.; Devchand, P.R.; Nehra, V.; Hamdy, O.; Horton, E.S.; Plutzky, J. The PPARγ Agonist Pioglitazone Represses Inflammation In A PPARα-Dependent Manner In Vitro and In Vivo In Mice. J. Am. Coll. Cardiol. 2008, 52, 869–881. [Google Scholar] [CrossRef]
  144. Ito, D.; Shimizu, S.; Inoue, K.; Saito, D.; Yanagisawa, M.; Inukai, K.; Akiyama, Y.; Morimoto, Y.; Noda, M.; Shimada, A. Comparison of Ipragliflozin and Pioglitazone Effects on Nonalcoholic Fatty Liver Disease in Patients With Type 2 Diabetes: A Randomized, 24-Week, Open-Label, Active-Controlled Trial. Diabetes Care 2017, 40, 1364–1372. [Google Scholar] [CrossRef]
  145. Nagashima, K.; Lopez, C.; Donovan, D.; Ngai, C.; Fontanez, N.; Bensadoun, A.; Fruchart-Najib, J.; Holleran, S.; Cohn, J.S.; Ramakrishnan, R.; et al. Effects of the PPARγ agonist pioglitazone on lipoprotein metabolism in patients with type 2 diabetes mellitus. J. Clin. Investig. 2005, 115, 1323–1332. [Google Scholar] [CrossRef] [PubMed]
  146. Sanyal, A.J.; Chalasani, N.; Kowdley, K.V.; McCullough, A.; Diehl, A.M.; Bass, N.M.; Neuschwander-Tetri, B.A.; Lavine, J.E.; Tonascia, J.; Unalp, A.; et al. Pioglitazone, Vitamin E, or Placebo for Nonalcoholic Steatohepatitis. N. Engl. J. Med. 2010, 362, 1675–1685. [Google Scholar] [CrossRef] [PubMed]
  147. Aithal, G.P.; Thomas, J.A.; Kaye, P.V.; Lawson, A.; Ryder, S.D.; Spendlove, I.; Austin, A.S.; Freeman, J.G.; Morgan, L.; Webber, J. Randomized, Placebo-Controlled Trial of Pioglitazone in Nondiabetic Subjects With Nonalcoholic Steatohepatitis. Gastroenterology 2008, 135, 1176–1184. [Google Scholar] [CrossRef] [PubMed]
  148. Belfort, R.; Harrison, S.A.; Brown, K.; Darland, C.; Finch, J.; Hardies, J.; Balas, B.; Gastaldelli, A.; Tio, F.; Pulcini, J.; et al. A Placebo-Controlled Trial of Pioglitazone in Subjects with Nonalcoholic Steatohepatitis. N. Engl. J. Med. 2006, 355, 2297–2307. [Google Scholar] [CrossRef]
  149. Mohanty, P.; Aljada, A.; Ghanim, H.; Hofmeyer, D.; Tripathy, D.; Syed, T.; Al-Haddad, W.; Dhindsa, S.; Dandona, P. Evidence for a potent antiinflammatory effect of rosiglitazone. J. Clin. Endocrinol. Metab. 2004, 89, 2728–2735. [Google Scholar] [CrossRef]
  150. Crossno, J.T.; Majka, S.M.; Grazia, T.; Gill, R.G.; Klemm, D.J. Rosiglitazone promotes development of a novel adipocyte population from bone marrow–derived circulating progenitor cells. J. Clin. Investig. 2006, 116, 3220–3228. [Google Scholar] [CrossRef]
  151. Ratziu, V.; Giral, P.; Jacqueminet, S.; Charlotte, F.; Hartemann-Heurtier, A.; Serfaty, L.; Podevin, P.; Lacorte, J.-M.; Bernhardt, C.; Bruckert, E.; et al. Rosiglitazone for nonalcoholic steatohepatitis: One-year results of the randomized placebo-controlled Fatty Liver Improvement with Rosiglitazone Therapy (FLIRT) Trial. Gastroenterology 2008, 135, 100–110. [Google Scholar] [CrossRef]
  152. Ratziu, V.; Charlotte, F.; Bernhardt, C.; Giral, P.; Halbron, M.; Lenaour, G.; Hartmann-Heurtier, A.; Bruckert, E.; Poynard, T.; LIDO Study Group. Long-term efficacy of rosiglitazone in nonalcoholic steatohepatitis: Results of the fatty liver improvement by rosiglitazone therapy (FLIRT 2) extension trial. Hepatology 2010, 51, 445–453. [Google Scholar] [CrossRef]
  153. Nesto, R.W.; Bell, D.; Bonow, R.O.; Fonseca, V.; Grundy, S.M.; Horton, E.S.; Le Winter, M.; Porte, D.; Semenkovich, C.F.; Smith, S.; et al. Thiazolidinedione Use, Fluid Retention, and Congestive Heart Failure. Circulation 2003, 108, 2941–2948. [Google Scholar] [CrossRef]
  154. Müller, T.D.; Finan, B.; Bloom, S.R.; D’Alessio, D.; Drucker, D.J.; Flatt, P.R.; Fritsche, A.; Gribble, F.; Grill, H.J.; Habener, J.F.; et al. Glucagon-like peptide 1 (GLP-1). Mol. Metab. 2019, 30, 72–130. [Google Scholar] [CrossRef]
  155. Bednarz, K.; Kowalczyk, K.; Cwynar, M.; Czapla, D.; Czarkowski, W.; Kmita, D.; Nowak, A.; Madej, P. The Role of Glp-1 Receptor Agonists in Insulin Resistance with Concomitant Obesity Treatment in Polycystic Ovary Syndrome. Int. J. Mol. Sci. 2022, 23, 4334. [Google Scholar] [CrossRef] [PubMed]
  156. Deacon, C.F. Physiology and Pharmacology of DPP-4 in Glucose Homeostasis and the Treatment of Type 2 Diabetes. Front. Endocrinol. 2019, 10, 80. [Google Scholar] [CrossRef]
  157. Barchetta, I.; Ceccarelli, V.; Cimini, F.A.; Barone, E.; Sentinelli, F.; Coluzzi, M.; Chiappetta, C.; Bertoccini, L.; Tramutola, A.; Labbadia, G.; et al. Circulating dipeptidyl peptidase-4 is independently associated with the presence and severity of NAFLD/NASH in individuals with and without obesity and metabolic disease. J. Endocrinol. Investig. 2021, 44, 979–988. [Google Scholar] [CrossRef]
  158. Bae, J.C. DPP-4 Inhibitor in Type 2 Diabetes Mellitus Patient with Non-Alcoholic Fatty Liver Disease: Achieving Two Goals at Once? Endocrinol. Metab. 2022, 37, 858–860. [Google Scholar] [CrossRef]
  159. Seghieri, M.; Christensen, A.S.; Andersen, A.; Solini, A.; Knop, F.K.; Vilsbøll, T. Future Perspectives on GLP-1 Receptor Agonists and GLP-1/glucagon Receptor Co-agonists in the Treatment of NAFLD. Front. Endocrinol. 2018, 9, 649. [Google Scholar] [CrossRef]
  160. Hartman, M.L.; Sanyal, A.J.; Loomba, R.; Wilson, J.M.; Nikooienejad, A.; Bray, R.; Karanikas, C.A.; Duffin, K.L.; Robins, D.A.; Haupt, A. Effects of Novel Dual GIP and GLP-1 Receptor Agonist Tirzepatide on Biomarkers of Nonalcoholic Steatohepatitis in Patients With Type 2 Diabetes. Diabetes Care 2020, 43, 1352–1355. [Google Scholar] [CrossRef]
  161. Loomba, R.; Hartman, M.L.; Lawitz, E.J.; Vuppalanchi, R.; Boursier, J.; Bugianesi, E.; Yoneda, M.; Behling, C.; Cummings, O.W.; Tang, Y.; et al. Tirzepatide for Metabolic Dysfunction-Associated Steatohepatitis with Liver Fibrosis. N. Engl. J. Med. 2024, 391, 299–310. [Google Scholar] [CrossRef]
  162. Armstrong, M.J.; Gaunt, P.; Aithal, G.P.; Barton, D.; Hull, D.; Parker, R.; Hazlehurst, J.M.; Guo, K.; LEAN Trial Team; Abouda, G.; et al. Liraglutide safety and efficacy in patients with non-alcoholic steatohepatitis (LEAN): A multicentre, double-blind, randomised, placebo-controlled phase 2 study. Lancet Lond. Engl. 2016, 387, 679–690. [Google Scholar] [CrossRef]
  163. DeFronzo, R.A.; Davidson, J.A.; Del Prato, S. The role of the kidneys in glucose homeostasis: A new path towards normalizing glycaemia. Diabetes Obes. Metab. 2012, 14, 5–14. [Google Scholar] [CrossRef]
  164. Ghezzi, C.; Loo, D.D.F.; Wright, E.M. Physiology of renal glucose handling via SGLT1, SGLT2 and GLUT2. Diabetologia 2018, 61, 2087–2097. [Google Scholar] [CrossRef]
  165. Tahrani, A.A.; Barnett, A.H.; Bailey, C.J. SGLT inhibitors in management of diabetes. Lancet Diabetes Endocrinol. 2013, 1, 140–151. [Google Scholar] [CrossRef] [PubMed]
  166. Tang, J.; Ye, L.; Yan, Q.; Zhang, X.; Wang, L. Effects of Sodium-Glucose Cotransporter 2 Inhibitors on Water and Sodium Metabolism. Front. Pharmacol. 2022, 13, 800490. [Google Scholar] [CrossRef]
  167. Renal Sodium-Glucose Cotransporter Inhibition in the Management of Type 2 Diabetes Mellitus—PubMed. Available online: https://pubmed.ncbi.nlm.nih.gov/26354881/ (accessed on 7 February 2025).
  168. Pradhan, R.; Yin, H.; Yu, O.; Azoulay, L. Glucagon-Like Peptide 1 Receptor Agonists and Sodium-Glucose Cotransporter 2 Inhibitors and Risk of Nonalcoholic Fatty Liver Disease Among Patients with Type 2 Diabetes. Diabetes Care 2022, 45, 819–829. [Google Scholar] [CrossRef] [PubMed]
  169. Ding, C.; Tang, Y.; Zhu, W.; Huang, P.; Lian, P.; Ran, J.; Huang, X. Sodium-glucose cotransporter protein-2 inhibitors and glucagon-like peptide-1 receptor agonists versus thiazolidinediones for non-alcoholic fatty liver disease: A network meta-analysis. Acta Diabetol. 2022, 59, 519–533. [Google Scholar] [CrossRef]
  170. Hamaguchi, E.; Takamura, T.; Sakurai, M.; Mizukoshi, E.; Zen, Y.; Takeshita, Y.; Kurita, S.; Arai, K.; Yamashita, T.; Sasaki, M.; et al. Histological Course of Nonalcoholic Fatty Liver Disease in Japanese Patients. Diabetes Care 2010, 33, 284–286. [Google Scholar] [CrossRef]
  171. Garvey, W.T.; Van Gaal, L.; Leiter, L.A.; Vijapurkar, U.; List, J.; Cuddihy, R.; Ren, J.; Davies, M.J. Effects of canagliflozin versus glimepiride on adipokines and inflammatory biomarkers in type 2 diabetes. Metabolism 2018, 85, 32–37. [Google Scholar] [CrossRef]
  172. Kuchay, M.S.; Krishan, S.; Mishra, S.K.; Farooqui, K.J.; Singh, M.K.; Wasir, J.S.; Bansal, B.; Kaur, P.; Jevalikar, G.; Gill, H.K.; et al. Effect of Empagliflozin on Liver Fat in Patients With Type 2 Diabetes and Nonalcoholic Fatty Liver Disease: A Randomized Controlled Trial (E-LIFT Trial). Diabetes Care 2018, 41, 1801–1808. [Google Scholar] [CrossRef]
  173. Kahl, S.; Gancheva, S.; Straßburger, K.; Herder, C.; Machann, J.; Katsuyama, H.; Kabisch, S.; Henkel, E.; Kopf, S.; Lagerpusch, M.; et al. Empagliflozin Effectively Lowers Liver Fat Content in Well-Controlled Type 2 Diabetes: A Randomized, Double-Blind, Phase 4, Placebo-Controlled Trial. Diabetes Care 2020, 43, 298–305. [Google Scholar] [CrossRef]
  174. Shimizu, M.; Suzuki, K.; Kato, K.; Jojima, T.; Iijima, T.; Murohisa, T.; Iijima, M.; Takekawa, H.; Usui, I.; Hiraishi, H.; et al. Evaluation of the effects of dapagliflozin, a sodium-glucose co-transporter-2 inhibitor, on hepatic steatosis and fibrosis using transient elastography in patients with type 2 diabetes and non-alcoholic fatty liver disease. Diabetes Obes. Metab. 2019, 21, 285–292. [Google Scholar] [CrossRef]
  175. Leiter, L.A.; Forst, T.; Polidori, D.; Balis, D.A.; Xie, J.; Sha, S. Effect of canagliflozin on liver function tests in patients with type 2 diabetes. Diabetes Metab. 2016, 42, 25–32. [Google Scholar] [CrossRef]
  176. Lefer, A.M.; Scalia, R.; Lefer, D.J. Vascular effects of HMG CoA-reductase inhibitors (statins) unrelated to cholesterol lowering: New concepts for cardiovascular disease. Cardiovasc. Res. 2001, 49, 281–287. [Google Scholar] [CrossRef] [PubMed]
  177. Ali, A.H.; Younis, N.; Abdallah, R.; Shaer, F.; Dakroub, A.; Ayoub, M.A.; Iratni, R.; Yassine, H.M.; Zibara, K.; Orekhov, A.; et al. Lipid-Lowering Therapies for Atherosclerosis: Statins, Fibrates, Ezetimibe and PCSK9 Monoclonal Antibodies. Curr. Med. Chem. 2021, 28, 7427–7445. [Google Scholar] [CrossRef] [PubMed]
  178. Stein, E.A.; Lane, M.; Laskarzewski, P. Comparison of statins in hypertriglyceridemia. Am. J. Cardiol. 1998, 81, 66B–69B. [Google Scholar] [CrossRef]
  179. Karlson, B.W.; Palmer, M.K.; Nicholls, S.J.; Lundman, P.; Barter, P.J. A VOYAGER Meta-Analysis of the Impact of Statin Therapy on Low-Density Lipoprotein Cholesterol and Triglyceride Levels in Patients With Hypertriglyceridemia. Am. J. Cardiol. 2016, 117, 1444–1448. [Google Scholar] [CrossRef]
  180. Yamashita, S.; Tsubakio-Yamamoto, K.; Ohama, T.; Nakagawa-Toyama, Y.; Nishida, M. Molecular mechanisms of HDL-cholesterol elevation by statins and its effects on HDL functions. J. Atheroscler. Thromb. 2010, 17, 436–451. [Google Scholar] [CrossRef]
  181. Blum, A.; Shamburek, R. The pleiotropic effects of statins on endothelial function, vascular inflammation, immunomodulation and thrombogenesis. Atherosclerosis 2009, 203, 325–330. [Google Scholar] [CrossRef]
  182. Blanco-Colio, L.M.; Tuñón, J.; Martín-Ventura, J.L.; Egido, J. Anti-inflammatory and immunomodulatory effects of statins. Kidney Int. 2003, 63, 12–23. [Google Scholar] [CrossRef]
  183. Statin Therapy and Myocardial No-Reflow—Calvert—2006—British JOURNAL of Pharmacology—Wiley Online Library. Available online: https://bpspubs.onlinelibrary.wiley.com/doi/10.1038/sj.bjp.0706863 (accessed on 5 February 2025).
  184. Horwich, T.B.; MacLellan, W.R.; Fonarow, G.C. Statin therapy is associated with improved survival in ischemic and non-ischemic heart failure. J. Am. Coll. Cardiol. 2004, 43, 642–648. [Google Scholar] [CrossRef]
  185. Athyros, V.G.; Boutari, C.; Stavropoulos, K.; Anagnostis, P.; Imprialos, K.P.; Doumas, M.; Karagiannis, A. Statins: An Under-Appreciated Asset for the Prevention and the Treatment of NAFLD or NASH and the Related Cardiovascular Risk. Curr. Vasc. Pharmacol. 2018, 16, 246–253. [Google Scholar] [CrossRef]
  186. Lee, J.I.; Lee, H.W.; Lee, K.S.; Lee, H.S.; Park, J.-Y. Effects of Statin Use on the Development and Progression of Nonalcoholic Fatty Liver Disease: A Nationwide Nested Case-Control Study. Am. J. Gastroenterol. 2021, 116, 116–124. [Google Scholar] [CrossRef]
  187. Targher, G.; Byrne, C.D.; Tilg, H. NAFLD and increased risk of cardiovascular disease: Clinical associations, pathophysiological mechanisms and pharmacological implications. Gut 2020, 69, 1691–1705. [Google Scholar] [CrossRef] [PubMed]
  188. Kaplan, D.E.; Serper, M.A.; Mehta, R.; Fox, R.; John, B.; Aytaman, A.; Baytarian, M.; Hunt, K.; Albrecht, J.; Njei, B.; et al. Effects of Hypercholesterolemia and Statin Exposure on Survival in a Large National Cohort of Patients With Cirrhosis. Gastroenterology 2019, 156, 1693–1706.e12. [Google Scholar] [CrossRef] [PubMed]
  189. Doumas, M.; Imprialos, K.; Dimakopoulou, A.; Stavropoulos, K.; Binas, A.; Athyros, V.G. The Role of Statins in the Management of Nonalcoholic Fatty Liver Disease. Curr. Pharm. Des. 2018, 24, 4587–4592. [Google Scholar] [CrossRef] [PubMed]
  190. van den Berg, E.H.; Wolters, A.A.B.; Dullaart, R.P.F.; Moshage, H.; Zurakowski, D.; de Meijer, V.E.; Blokzijl, H. Prescription of statins in suspected non-alcoholic fatty liver disease and high cardiovascular risk, a population-based study. Liver Int. Off. J. Int. Assoc. Study Liver 2019, 39, 1343–1354. [Google Scholar] [CrossRef]
  191. Pastori, D.; Polimeni, L.; Baratta, F.; Pani, A.; Del Ben, M.; Angelico, F. The efficacy and safety of statins for the treatment of non-alcoholic fatty liver disease. Dig. Liver Dis. Off. J. Ital. Soc. Gastroenterol. Ital. Assoc. Study Liver 2015, 47, 4–11. [Google Scholar] [CrossRef]
  192. Abdallah, M.; Brown, L.; Provenza, J.; Tariq, R.; Gowda, S.; Singal, A.K. Safety and efficacy of dyslipidemia treatment in NAFLD patients: A meta-analysis of randomized controlled trials. Ann. Hepatol. 2022, 27, 100738. [Google Scholar] [CrossRef]
  193. Golomb, B.A.; Evans, M.A. Statin adverse effects: A review of the literature and evidence for a mitochondrial mechanism. Am. J. Cardiovasc. Drugs Drugs Devices Interv. 2008, 8, 373–418. [Google Scholar] [CrossRef]
  194. Simon, T.G.; Bonilla, H.; Yan, P.; Chung, R.T.; Butt, A.A. Atorvastatin and fluvastatin are associated with dose-dependent reductions in cirrhosis and hepatocellular carcinoma, among patients with hepatitis C virus: Results from ERCHIVES. Hepatology 2016, 64, 47–57. [Google Scholar] [CrossRef]
  195. Zou, B.; Odden, M.C.; Nguyen, M.H. Statin Use and Reduced Hepatocellular Carcinoma Risk in Patients with Nonalcoholic Fatty Liver Disease. Clin. Gastroenterol. Hepatol. Off. Clin. Pract. J. Am. Gastroenterol. Assoc. 2023, 21, 435–444.e6. [Google Scholar] [CrossRef]
  196. Kim, G.; Jang, S.-Y.; Nam, C.M.; Kang, E.S. Statin use and the risk of hepatocellular carcinoma in patients at high risk: A nationwide nested case-control study. J. Hepatol. 2018, 68, 476–484. [Google Scholar] [CrossRef]
  197. Sebastiani, G.; Patel, K.; Ratziu, V.; Feld, J.J.; Neuschwander-Tetri, B.A.; Pinzani, M.; Petta, S.; Berzigotti, A.; Metrakos, P.; Shoukry, N.; et al. Current considerations for clinical management and care of non-alcoholic fatty liver disease: Insights from the 1st International Workshop of the Canadian NASH Network (CanNASH). Can. Liver J. 2022, 5, 61–90. [Google Scholar] [CrossRef] [PubMed]
  198. Mancini, G.B.J.; Baker, S.; Bergeron, J.; Fitchett, D.; Frohlich, J.; Genest, J.; Gupta, M.; Hegele, R.A.; Ng, D.; Pearson, G.J.; et al. Diagnosis, Prevention, and Management of Statin Adverse Effects and Intolerance: Canadian Consensus Working Group Update (2016). Can. J. Cardiol. 2016, 32, S35–S65. [Google Scholar] [CrossRef] [PubMed]
  199. Arnaud, C.; Burger, F.; Steffens, S.; Veillard, N.R.; Nguyen, T.H.; Trono, D.; Mach, F. Statins reduce interleukin-6-induced C-reactive protein in human hepatocytes: New evidence for direct antiinflammatory effects of statins. Arterioscler. Thromb. Vasc. Biol. 2005, 25, 1231–1236. [Google Scholar] [CrossRef] [PubMed]
  200. Zhang, S.; Ren, X.; Zhang, B.; Lan, T.; Liu, B. A Systematic Review of Statins for the Treatment of Nonalcoholic Steatohepatitis: Safety, Efficacy, and Mechanism of Action. Molecules 2024, 29, 1859. [Google Scholar] [CrossRef]
  201. Chen, D.; Sui, L.; Chen, C.; Liu, S.; Sun, X.; Guan, J. Atorvastatin suppresses NLRP3 inflammasome activation in intracerebral hemorrhage via TLR4- and MyD88-dependent pathways. Aging 2022, 14, 462–476. [Google Scholar] [CrossRef]
  202. Vargas, J.I.; Arrese, M.; Shah, V.H.; Arab, J.P. Use of Statins in Patients with Chronic Liver Disease and Cirrhosis: Current Views and Prospects. Curr. Gastroenterol. Rep. 2017, 19, 43. [Google Scholar] [CrossRef]
  203. Islam, M.M.; Poly, T.N.; Walther, B.A.; Yang, H.-C.; Jack Li, Y.-C. Statin Use and the Risk of Hepatocellular Carcinoma: A Meta-Analysis of Observational Studies. Cancers 2020, 12, 671. [Google Scholar] [CrossRef]
  204. McConnell, M.J.; Kostallari, E.; Ibrahim, S.H.; Iwakiri, Y. The evolving role of liver sinusoidal endothelial cells in liver health and disease. Hepatology 2023, 78, 649–669. [Google Scholar] [CrossRef]
  205. Fujita, T.; Narumiya, S. Roles of hepatic stellate cells in liver inflammation: A new perspective. Inflamm. Regen. 2016, 36, 1. [Google Scholar] [CrossRef]
  206. Torres-Peña, J.D.; Martín-Piedra, L.; Fuentes-Jiménez, F. Statins in Non-alcoholic Steatohepatitis. Front. Cardiovasc. Med. 2021, 8, 777131. [Google Scholar] [CrossRef]
  207. da Silva Pereira, E.N.G.; Franco, R.L.C.; Santos, R.D.C.D.; Daliry, A. Statins and non-alcoholic fatty liver disease: A concise review. Biomed. Pharmacother. 2025, 183, 117805. [Google Scholar] [CrossRef]
  208. Yano, M.; Matsumura, T.; Senokuchi, T.; Ishii, N.; Murata, Y.; Taketa, K.; Motoshima, H.; Taguchi, T.; Sonoda, K.; Kukidome, D.; et al. Statins activate peroxisome proliferator-activated receptor gamma through extracellular signal-regulated kinase 1/2 and p38 mitogen-activated protein kinase-dependent cyclooxygenase-2 expression in macrophages. Circ. Res. 2007, 100, 1442–1451. [Google Scholar] [CrossRef] [PubMed]
  209. Vieira-Silva, S.; Falony, G.; Belda, E.; Nielsen, T.; Aron-Wisnewsky, J.; Chakaroun, R.; Forslund, S.K.; Assmann, K.; Valles-Colomer, M.; Nguyen, T.T.D.; et al. Statin therapy is associated with lower prevalence of gut microbiota dysbiosis. Nature 2020, 581, 310–315. [Google Scholar] [CrossRef] [PubMed]
  210. Wang, Y.; Xu, H.; Zhou, X.; Chen, W.; Zhou, H. Dysregulated bile acid homeostasis: Unveiling its role in metabolic diseases. Med. Rev. 2024, 4, 262–283. [Google Scholar] [CrossRef]
  211. Xiang, D.; Yang, J.; Liu, L.; Yu, H.; Gong, X.; Liu, D. The regulation of tissue-specific farnesoid X receptor on genes and diseases involved in bile acid homeostasis. Biomed. Pharmacother. 2023, 168, 115606. [Google Scholar] [CrossRef]
  212. Farnesoid X Receptor Activation by Bile Acids Suppresses Lipid Peroxidation and Ferroptosis|Nature Communications. Available online: https://www.nature.com/articles/s41467-023-42702-8 (accessed on 7 February 2025).
  213. Han, S.Y.; Song, H.K.; Cha, J.J.; Han, J.Y.; Kang, Y.S.; Cha, D.R. Farnesoid X receptor (FXR) agonist ameliorates systemic insulin resistance, dysregulation of lipid metabolism, and alterations of various organs in a type 2 diabetic kidney animal model. Acta Diabetol. 2021, 58, 495–503. [Google Scholar] [CrossRef]
  214. Clifford, B.L.; Sedgeman, L.R.; Williams, K.J.; Morand, P.; Cheng, A.; Jarrett, K.E.; Chan, A.P.; Brearley-Sholto, M.C.; Wahlström, A.; Ashby, J.W.; et al. FXR activation protects against NAFLD via bile-acid-dependent reductions in lipid absorption. Cell Metab. 2021, 33, 1671–1684.e4. [Google Scholar] [CrossRef]
  215. Pelton, P.D.; Patel, M.; Demarest, K.T. Nuclear receptors as potential targets for modulating reverse cholesterol transport. Curr. Top. Med. Chem. 2005, 5, 265–282. [Google Scholar] [CrossRef]
  216. Goodwin, B.; Jones, S.A.; Price, R.R.; Watson, M.A.; McKee, D.D.; Moore, L.B.; Galardi, C.; Wilson, J.G.; Lewis, M.C.; Roth, M.E.; et al. A regulatory cascade of the nuclear receptors FXR, SHP-1, and LRH-1 represses bile acid biosynthesis. Mol. Cell 2000, 6, 517–526. [Google Scholar] [CrossRef]
  217. Aguilar-Olivos, N.E.; Carrillo-Córdova, D.; Oria-Hernández, J.; Sánchez-Valle, V.; Ponciano-Rodríguez, G.; Ramírez-Jaramillo, M.; Chablé-Montero, F.; Chávez-Tapia, N.C.; Uribe, M.; Méndez-Sánchez, N. The nuclear receptor FXR, but not LXR, up-regulates bile acid transporter expression in non-alcoholic fatty liver disease. Ann. Hepatol. 2015, 14, 487–493. [Google Scholar]
  218. Malagnino, V.; Hussner, J.; Issa, A.; Midzic, A.; Meyer Zu Schwabedissen, H.E. OATP1B3-1B7, a novel organic anion transporting polypeptide, is modulated by FXR ligands and transports bile acids. Am. J. Physiol. Gastrointest. Liver Physiol. 2019, 317, G751–G762. [Google Scholar] [CrossRef] [PubMed]
  219. Ren, T.; Pang, L.; Dai, W.; Wu, S.; Kong, J. Regulatory mechanisms of the bile salt export pump (BSEP/ABCB11) and its role in related diseases. Clin. Res. Hepatol. Gastroenterol. 2021, 45, 101641. [Google Scholar] [CrossRef] [PubMed]
  220. Tang, Y.; Fan, Y.; Wang, Y.; Wang, D.; Huang, Q.; Chen, T.; Cao, X.; Wen, C.; Shen, X.; Li, J.; et al. A Current Understanding of FXR in NAFLD: The multifaceted regulatory role of FXR and novel lead discovery for drug development. Biomed. Pharmacother. 2024, 175, 116658. [Google Scholar] [CrossRef]
  221. Wei, M.; Cao, W.-B.; Zhao, R.-D.; Sun, D.-P.; Liang, Y.-Z.; Huang, Y.-D.; Cheng, Z.-W.; Ouyang, J.; Yang, W.-S.; Yu, W.-B. Fibroblast growth factor 15, induced by elevated bile acids, mediates the improvement of hepatic glucose metabolism after sleeve gastrectomy. World J. Gastroenterol. 2023, 29, 3280–3291. [Google Scholar] [CrossRef]
  222. Kliewer, S.A.; Mangelsdorf, D.J. Bile Acids as Hormones: The FXR-FGF15/19 Pathway. Dig. Dis. 2015, 33, 327–331. [Google Scholar] [CrossRef]
  223. Song, L.; Wang, L.; Hou, Y.; Zhou, J.; Chen, C.; Ye, X.; Dong, W.; Gao, H.; Liu, Y.; Qiao, G.; et al. FGF4 protects the liver from nonalcoholic fatty liver disease by activating the AMP-activated protein kinase-Caspase 6 signal axis. Hepatology 2022, 76, 1105–1120. [Google Scholar] [CrossRef]
  224. Song, L.; Hou, Y.; Xu, D.; Dai, X.; Luo, J.; Liu, Y.; Huang, Z.; Yang, M.; Chen, J.; Hu, Y.; et al. Hepatic FXR-FGF4 is required for bile acid homeostasis via an FGFR4-LRH-1 signal node under cholestatic stress. Cell Metab. 2025, 37, 104–120.e9. [Google Scholar] [CrossRef]
  225. Meng, D.; Zhang, F.; Yu, W.; Zhang, X.; Yin, G.; Liang, P.; Feng, Y.; Chen, S.; Liu, H. Biological Role and Related Natural Products of SIRT1 in Nonalcoholic Fatty Liver. Diabetes Metab. Syndr. Obes. 2023, 16, 4043–4064. [Google Scholar] [CrossRef]
  226. Schramm, C.; Wedemeyer, H.; Mason, A.; Hirschfield, G.M.; Levy, C.; Kowdley, K.V.; Milkiewicz, P.; Janczewska, E.; Malova, E.S.; Sanni, J.; et al. Farnesoid X receptor agonist tropifexor attenuates cholestasis in a randomised trial in patients with primary biliary cholangitis. JHEP Rep. Innov. Hepatol. 2022, 4, 100544. [Google Scholar] [CrossRef]
  227. Ratziu, V.; Harrison, S.A.; Loustaud-Ratti, V.; Bureau, C.; Lawitz, E.; Abdelmalek, M.; Alkhouri, N.; Francque, S.; Girma, H.; Darteil, R.; et al. Hepatic and renal improvements with FXR agonist vonafexor in individuals with suspected fibrotic NASH. J. Hepatol. 2023, 78, 479–492. [Google Scholar] [CrossRef]
  228. Xu, J.; Wang, Y.; Khoshdeli, M.; Peach, M.; Chuang, J.-C.; Lin, J.; Tsai, W.-W.; Mahadevan, S.; Minto, W.; Diehl, L.; et al. IL-31 levels correlate with pruritus in patients with cholestatic and metabolic liver diseases and is farnesoid X receptor responsive in NASH. Hepatology 2023, 77, 20–32. [Google Scholar] [CrossRef] [PubMed]
  229. Abenavoli, L.; Falalyeyeva, T.; Boccuto, L.; Tsyryuk, O.; Kobyliak, N. Obeticholic Acid: A New Era in the Treatment of Nonalcoholic Fatty Liver Disease. Pharmacy 2018, 11, 104. [Google Scholar] [CrossRef]
  230. Makri, E.; Cholongitas, E.; Tziomalos, K. Emerging role of obeticholic acid in the management of nonalcoholic fatty liver disease. World J. Gastroenterol. 2016, 22, 9039–9043. [Google Scholar] [CrossRef] [PubMed]
  231. Neuschwander-Tetri, B.A.; Loomba, R.; Sanyal, A.J.; Lavine, J.E.; Natta, M.L.V.; Abdelmalek, M.F.; Chalasani, N.; Dasarathy, S.; Diehl, A.M.; Hameed, B.; et al. Farnesoid X nuclear receptor ligand obeticholic acid for non-cirrhotic, non-alcoholic steatohepatitis (FLINT): A multicentre, randomised, placebo-controlled trial. Lancet 2015, 385, 956–965. [Google Scholar] [CrossRef]
  232. Younossi, Z.M.; Ratziu, V.; Loomba, R.; Rinella, M.; Anstee, Q.M.; Goodman, Z.; Bedossa, P.; Geier, A.; Beckebaum, S.; Newsome, P.N.; et al. Obeticholic acid for the treatment of non-alcoholic steatohepatitis: Interim analysis from a multicentre, randomised, placebo-controlled phase 3 trial. Lancet Lond. Engl. 2019, 394, 2184–2196. [Google Scholar] [CrossRef]
  233. Nathani, R.R.; Bansal, M.B. Update on Clinical Trials for Nonalcoholic Steatohepatitis. Gastroenterol. Hepatol. 2023, 19, 371–381. [Google Scholar]
  234. Jiang, J.; Ma, Y.; Liu, Y.; Lu, D.; Gao, X.; Krausz, K.W.; Desai, D.; Amin, S.G.; Patterson, A.D.; Gonzalez, F.J.; et al. Glycine-β-muricholic acid antagonizes the intestinal farnesoid X receptor-ceramide axis and ameliorates NASH in mice. Hepatol. Commun. 2022, 6, 3363–3378. [Google Scholar] [CrossRef]
  235. Fleishman, J.S.; Kumar, S. Bile acid metabolism and signaling in health and disease: Molecular mechanisms and therapeutic targets. Signal Transduct. Target. Ther. 2024, 9, 97. [Google Scholar] [CrossRef]
  236. Bezerra, R.M.N.; Veiga, L.F.; Caetano, A.C.; Rosalen, P.L.; Amaral, M.E.C.; Palanch, A.C.; de Alencar, S.M. Caffeic acid phenethyl ester reduces the activation of the nuclear factor κB pathway by high-fat diet-induced obesity in mice. Metabolism 2012, 61, 1606–1614. [Google Scholar] [CrossRef]
  237. Shen, C.; Pan, Z.; Wu, S.; Zheng, M.; Zhong, C.; Xin, X.; Lan, S.; Zhu, Z.; Liu, M.; Wu, H.; et al. Emodin palliates high-fat diet-induced nonalcoholic fatty liver disease in mice via activating the farnesoid X receptor pathway. J. Ethnopharmacol. 2021, 279, 114340. [Google Scholar] [CrossRef]
  238. Lei, Y.; Tang, L.; Chen, Q.; Wu, L.; He, W.; Tu, D.; Wang, S.; Chen, Y.; Liu, S.; Xie, Z.; et al. Disulfiram ameliorates nonalcoholic steatohepatitis by modulating the gut microbiota and bile acid metabolism. Nat. Commun. 2022, 13, 6862. [Google Scholar] [CrossRef] [PubMed]
  239. Renga, B.; Mencarelli, A.; Migliorati, M.; Distrutti, E.; Fiorucci, S. Bile-acid-activated farnesoid X receptor regulates hydrogen sulfide production and hepatic microcirculation. World J. Gastroenterol. WJG 2009, 15, 2097–2108. [Google Scholar] [CrossRef] [PubMed]
  240. Nian, F.; Wu, L.; Xia, Q.; Tian, P.; Ding, C.; Lu, X. Akkermansia muciniphila and Bifidobacterium bifidum Prevent NAFLD by Regulating FXR Expression and Gut Microbiota. J. Clin. Transl. Hepatol. 2023, 11, 763–776. [Google Scholar] [CrossRef] [PubMed]
  241. Paglialunga, S.; Dehn, C.A. Clinical assessment of hepatic de novo lipogenesis in non-alcoholic fatty liver disease. Lipids Health Dis. 2016, 15, 159. [Google Scholar] [CrossRef]
  242. Lambrecht, J.; Tacke, F. Acetyl-CoA Carboxylase Inhibition as a Therapeutic Tool in the Battle Against NASH: Hitting More Than Just One Mechanism? Cell. Mol. Gastroenterol. Hepatol. 2020, 10, 859–861. [Google Scholar] [CrossRef]
  243. Ross, T.T.; Crowley, C.; Kelly, K.L.; Rinaldi, A.; Beebe, D.A.; Lech, M.P.; Martinez, R.V.; Carvajal-Gonzalez, S.; Boucher, M.; Hirenallur-Shanthappa, D.; et al. Acetyl-CoA Carboxylase Inhibition Improves Multiple Dimensions of NASH Pathogenesis in Model Systems. Cell. Mol. Gastroenterol. Hepatol. 2020, 10, 829–851. [Google Scholar] [CrossRef]
  244. Mateo-Marín, M.A.; Alves-Bezerra, M. Targeting acetyl-CoA carboxylases for the treatment of MASLD. J. Lipid Res. 2024, 65, 100676. [Google Scholar] [CrossRef]
  245. Wang, Y.; Viscarra, J.; Kim, S.-J.; Sul, H.S. Transcriptional regulation of hepatic lipogenesis. Nat. Rev. Mol. Cell Biol. 2015, 16, 678–689. [Google Scholar] [CrossRef]
  246. Goedeke, L.; Bates, J.; Vatner, D.F.; Perry, R.J.; Wang, T.; Ramirez, R.; Li, L.; Ellis, M.W.; Zhang, D.; Wong, K.E.; et al. Acetyl-CoA Carboxylase Inhibition Reverses NAFLD and Hepatic Insulin Resistance but Promotes Hypertriglyceridemia in Rodents. Hepatology 2018, 68, 2197–2211. [Google Scholar] [CrossRef]
  247. Zhao, S.; Jang, C.; Liu, J.; Uehara, K.; Gilbert, M.; Izzo, L.; Zeng, X.; Trefely, S.; Fernandez, S.; Carrer, A.; et al. Dietary fructose feeds hepatic lipogenesis via microbiota-derived acetate. Nature 2020, 579, 586–591. [Google Scholar] [CrossRef]
  248. DiNicolantonio, J.J.; Subramonian, A.M.; O’Keefe, J.H. Added fructose as a principal driver of non-alcoholic fatty liver disease: A public health crisis. Open Heart 2017, 4, e000631. [Google Scholar] [CrossRef] [PubMed]
  249. Bergman, A.; Carvajal-Gonzalez, S.; Tarabar, S.; Saxena, A.R.; Esler, W.P.; Amin, N.B. Safety, Tolerability, Pharmacokinetics, and Pharmacodynamics of a Liver-Targeting Acetyl-CoA Carboxylase Inhibitor (PF-05221304): A Three-Part Randomized Phase 1 Study. Clin. Pharmacol. Drug Dev. 2020, 9, 514–526. [Google Scholar] [CrossRef] [PubMed]
  250. Kim, C.-W.; Addy, C.; Kusunoki, J.; Anderson, N.N.; Deja, S.; Fu, X.; Burgess, S.C.; Li, C.; Ruddy, M.; Chakravarthy, M.; et al. Acetyl CoA Carboxylase Inhibition Reduces Hepatic Steatosis but Elevates Plasma Triglycerides in Mice and Humans: A Bedside to Bench Investigation. Cell Metab. 2017, 26, 394–406.e6. [Google Scholar] [CrossRef] [PubMed]
  251. Matsumoto, M.; Yashiro, H.; Ogino, H.; Aoyama, K.; Nambu, T.; Nakamura, S.; Nishida, M.; Wang, X.; Erion, D.M.; Kaneko, M. Acetyl-CoA carboxylase 1 and 2 inhibition ameliorates steatosis and hepatic fibrosis in a MC4R knockout murine model of nonalcoholic steatohepatitis. PLoS ONE 2020, 15, e0228212. [Google Scholar] [CrossRef]
  252. Stiede, K.; Miao, W.; Blanchette, H.S.; Beysen, C.; Harriman, G.; Harwood, H.J.; Kelley, H.; Kapeller, R.; Schmalbach, T.; Westlin, W.F. Acetyl-coenzyme A carboxylase inhibition reduces de novo lipogenesis in overweight male subjects: A randomized, double-blind, crossover study. Hepatology 2017, 66, 324–334. [Google Scholar] [CrossRef]
  253. Kelly, K.L.; Reagan, W.J.; Sonnenberg, G.E.; Clasquin, M.; Hales, K.; Asano, S.; Amor, P.A.; Carvajal-Gonzalez, S.; Shirai, N.; Matthews, M.D.; et al. De novo lipogenesis is essential for platelet production in humans. Nat. Metab. 2020, 2, 1163–1178. [Google Scholar] [CrossRef]
  254. Alkhouri, N.; Lawitz, E.; Noureddin, M.; DeFronzo, R.; Shulman, G.I. GS-0976 (Firsocostat): An investigational liver-directed acetyl-CoA carboxylase (ACC) inhibitor for the treatment of non-alcoholic steatohepatitis (NASH). Expert Opin. Investig. Drugs 2020, 29, 135–141. [Google Scholar] [CrossRef]
  255. Calle, R.A.; Amin, N.B.; Carvajal-Gonzalez, S.; Ross, T.T.; Bergman, A.; Aggarwal, S.; Crowley, C.; Rinaldi, A.; Mancuso, J.; Aggarwal, N.; et al. ACC inhibitor alone or co-administered with a DGAT2 inhibitor in patients with non-alcoholic fatty liver disease: Two parallel, placebo-controlled, randomized phase 2a trials. Nat. Med. 2021, 27, 1836–1848. [Google Scholar] [CrossRef]
  256. Sinha, R.A.; Singh, B.K.; Yen, P.M. Direct effects of thyroid hormones on hepatic lipid metabolism. Nat. Rev. Endocrinol. 2018, 14, 259–269. [Google Scholar] [CrossRef]
  257. Chung, G.E.; Kim, D.; Kim, W.; Yim, J.Y.; Park, M.J.; Kim, Y.J.; Yoon, J.-H.; Lee, H.-S. Non-alcoholic fatty liver disease across the spectrum of hypothyroidism. J. Hepatol. 2012, 57, 150–156. [Google Scholar] [CrossRef]
  258. Ritter, M.J.; Amano, I.; Hollenberg, A.N. Thyroid Hormone Signaling and the Liver. Hepatology 2020, 72, 742–752. [Google Scholar] [CrossRef] [PubMed]
  259. The Roles of Autophagy and Thyroid Hormone in the Pathogenesis and Treatment of NAFLD. Available online: https://www.oaepublish.com/articles/2394-5079.2021.82 (accessed on 7 February 2025).
  260. Araki, O.; Ying, H.; Zhu, X.G.; Willingham, M.C.; Cheng, S.Y. Distinct Dysregulation of Lipid Metabolism by Unliganded Thyroid Hormone Receptor Isoforms. Mol. Endocrinol. 2009, 23, 308–315. [Google Scholar] [CrossRef] [PubMed]
  261. Jornayvaz, F.R.; Lee, H.-Y.; Jurczak, M.J.; Alves, T.C.; Guebre-Egziabher, F.; Guigni, B.A.; Zhang, D.; Samuel, V.T.; Silva, J.E.; Shulman, G.I. Thyroid Hormone Receptor-α Gene Knockout Mice Are Protected from Diet-Induced Hepatic Insulin Resistance. Endocrinology 2012, 153, 583–591. [Google Scholar] [CrossRef] [PubMed]
  262. Karim, G.; Bansal, M.B. Resmetirom: An Orally Administered, Smallmolecule, Liver-directed, β-selective THR Agonist for the Treatment of Non-alcoholic Fatty Liver Disease and Non-alcoholic Steatohepatitis. TouchREVIEWS Endocrinol. 2023, 19, 60–70. [Google Scholar] [CrossRef]
  263. Hönes, G.S.; Sivakumar, R.G.; Hoppe, C.; König, J.; Führer, D.; Moeller, L.C. Cell-Specific Transport and Thyroid Hormone Receptor Isoform Selectivity Account for Hepatocyte-Targeted Thyromimetic Action of MGL-3196. Int. J. Mol. Sci. 2022, 23, 13714. [Google Scholar] [CrossRef]
  264. Luong, X.G.; Stevens, S.K.; Jekle, A.; Lin, T.-I.; Gupta, K.; Misner, D.; Chanda, S.; Mukherjee, S.; Williams, C.; Stoycheva, A.; et al. Regulation of gene transcription by thyroid hormone receptor β agonists in clinical development for the treatment of non-alcoholic steatohepatitis (NASH). PLoS ONE 2020, 15, e0240338. [Google Scholar] [CrossRef]
  265. Bianco, A.C.; Kim, B.W. Deiodinases: Implications of the local control of thyroid hormone action. J. Clin. Investig. 2006, 116, 2571–2579. [Google Scholar] [CrossRef]
  266. FDA. FDA Approves First Treatment for Patients with Liver Scarring Due to Fatty Liver Disease. 2024. Available online: https://www.fda.gov/news-events/press-announcements/fda-approves-first-treatment-patients-liver-scarring-due-fatty-liver-disease (accessed on 7 February 2025).
  267. Harrison, S.A.; Bedossa, P.; Guy, C.D.; Schattenberg, J.M.; Loomba, R.; Taub, R.; Labriola, D.; Moussa, S.E.; Neff, G.W.; Rinella, M.E.; et al. A Phase 3, Randomized, Controlled Trial of Resmetirom in NASH with Liver Fibrosis. N. Engl. J. Med. 2024, 390, 497–509. [Google Scholar] [CrossRef]
  268. Cusi, K. Selective Agonists of Thyroid Hormone Receptor Beta for the Treatment of NASH. N. Engl. J. Med. 2024, 390, 559–561. [Google Scholar] [CrossRef]
  269. FDA Grants Tentative Approval to Generic Version of Rifaximin for IBS-D. Available online: https://www.ajmc.com/view/fda-grants-tentative-approval-to-generic-version-of-rifaximin-for-ibs-d (accessed on 7 February 2025).
  270. Lacy, B.E.; Chang, L.; Rao, S.S.C.; Heimanson, Z.; Sayuk, G.S. Rifaximin Treatment for Individual and Multiple Symptoms of Irritable Bowel Syndrome With Diarrhea: An Analysis Using New End Points. Clin. Ther. 2023, 45, 198–209. [Google Scholar] [CrossRef]
  271. Lembo, A.; Pimentel, M.; Rao, S.S.; Schoenfeld, P.; Cash, B.; Weinstock, L.B.; Paterson, C.; Bortey, E.; Forbes, W.P. Repeat Treatment With Rifaximin Is Safe and Effective in Patients With Diarrhea-Predominant Irritable Bowel Syndrome. Gastroenterology 2016, 151, 1113–1121. [Google Scholar] [CrossRef] [PubMed]
  272. Pimentel, M.; Lembo, A.; Chey, W.D.; Zakko, S.; Ringel, Y.; Yu, J.; Mareya, S.M.; Shaw, A.L.; Bortey, E.; Forbes, W.P. Rifaximin Therapy for Patients with Irritable Bowel Syndrome without Constipation. N. Engl. J. Med. 2011, 364, 22–32. [Google Scholar] [CrossRef] [PubMed]
  273. Kimer, N.; Krag, A.; Møller, S.; Bendtsen, F.; Gluud, L.L. Systematic review with meta-analysis: The effects of rifaximin in hepatic encephalopathy. Aliment. Pharmacol. Ther. 2014, 40, 123–132. [Google Scholar] [CrossRef] [PubMed]
  274. Scott, L.J. Rifaximin: A Review of Its Use in Reducing Recurrence of Overt Hepatic Encephalopathy Episodes. Drugs 2014, 74, 2153–2160. [Google Scholar] [CrossRef]
  275. Gastrointestinal Drugs Advisory Committee; Notice of Meeting. Available online: https://www.federalregister.gov/documents/2010/12/02/2010-30274/gastrointestinal-drugs-advisory-committee-notice-of-meeting (accessed on 7 February 2025).
  276. Patidar, K.R.; Bajaj, J.S. Antibiotics for the treatment of hepatic encephalopathy. Metab. Brain Dis. 2013, 28, 307–312. [Google Scholar] [CrossRef]
  277. Commissioner, O. of the FDA Approves New Drug to Treat Travelers’ Diarrhea. Available online: https://www.fda.gov/news-events/press-announcements/fda-approves-new-drug-treat-travelers-diarrhea (accessed on 7 February 2025).
  278. Koo, H.L.; DuPont, H.L. Rifaximin: A unique gastrointestinal-selective antibiotic for enteric diseases. Curr. Opin. Gastroenterol. 2010, 26, 17. [Google Scholar] [CrossRef]
  279. Hu, Y.; Ren, J.; Zhan, M.; Li, W.; Dai, H. Efficacy of Rifaximin in Prevention of Travelers’ Diarrhea: A Meta-Analysis of Randomized, Double-Blind, Placebo-Controlled Trials. J. Travel Med. 2012, 19, 352–356. [Google Scholar] [CrossRef]
  280. Dupont, H.L.; Jiang, Z.-D.; Belkind–Gerson, J.; Okhuysen, P.C.; Ericsson, C.D.; Ke, S.; Huang, D.B.; Dupont, M.W.; Adachi, J.A.; Cabada, F.J.D.L.; et al. Treatment of Travelers’ Diarrhea: Randomized Trial Comparing Rifaximin, Rifaximin Plus Loperamide, and Loperamide Alone. Clin. Gastroenterol. Hepatol. 2007, 5, 451–456. [Google Scholar] [CrossRef]
  281. Abdel-Razik, A.; Mousa, N.; Shabana, W.; Refaey, M.; Elzehery, R.; Elhelaly, R.; Zalata, K.; Abdelsalam, M.; Eldeeb, A.A.; Awad, M.; et al. Rifaximin in nonalcoholic fatty liver disease: Hit multiple targets with a single shot. Eur. J. Gastroenterol. Hepatol. 2018, 30, 1237. [Google Scholar] [CrossRef]
  282. Zhang, L.; Huang, Y.; Zhou, Y.; Buckley, T.; Wang, H.H. Antibiotic Administration Routes Significantly Influence the Levels of Antibiotic Resistance in Gut Microbiota. Antimicrob. Agents Chemother. 2013, 57, 3659–3666. [Google Scholar] [CrossRef]
  283. Iorio, N.; Malik, Z.; Schey, R. Profile of rifaximin and its potential in the treatment of irritable bowel syndrome. Clin. Exp. Gastroenterol. 2015, 8, 159–167. [Google Scholar] [CrossRef] [PubMed]
  284. Combined Therapy with a CCR2/CCR5 Antagonist and FGF21 Analogue Synergizes in Ameliorating Steatohepatitis and Fibrosis. Available online: https://www.mdpi.com/1422-0067/23/12/6696 (accessed on 7 February 2025).
  285. Harrison, S.A.; Ruane, P.J.; Freilich, B.; Neff, G.; Patil, R.; Behling, C.; Hu, C.; Shringarpure, R.; de Temple, B.; Fong, E.; et al. A randomized, double-blind, placebo-controlled phase IIa trial of efruxifermin for patients with compensated NASH cirrhosis. JHEP Rep. 2023, 5, 100563. [Google Scholar] [CrossRef] [PubMed]
  286. Harrison, S.A.; Frias, J.P.; Neff, G.; Abrams, G.A.; Lucas, K.J.; Sanchez, W.; Gogia, S.; Sheikh, M.Y.; Behling, C.; Bedossa, P.; et al. Safety and efficacy of once-weekly efruxifermin versus placebo in non-alcoholic steatohepatitis (HARMONY): A multicentre, randomised, double-blind, placebo-controlled, phase 2b trial. Lancet Gastroenterol. Hepatol. 2023, 8, 1080–1093. [Google Scholar] [CrossRef] [PubMed]
  287. Akero Therapeutics’ Phase 2b SYMMETRY Cohort D Study Met Safety & Tolerability Endpoints and Showed Adding EFX to GLP-1 Therapy Significantly Improved Non-Invasive Markers of NASH-Related Disease—Akero Therapeutics, Inc. Available online: https://ir.akerotx.com/news-releases/news-release-details/akero-therapeutics-phase-2b-symmetry-cohort-d-study-met-safety/ (accessed on 7 February 2025).
  288. Rinella, M.E.; Lieu, H.D.; Kowdley, K.V.; Goodman, Z.D.; Alkhouri, N.; Lawitz, E.; Ratziu, V.; Abdelmalek, M.F.; Wong, V.W.-S.; Younes, Z.H.; et al. A randomized, double-blind, placebo-controlled trial of aldafermin in patients with NASH and compensated cirrhosis. Hepatology 2024, 79, 674–689. [Google Scholar] [CrossRef]
  289. Lonardo, A.; Arab, J.P.; Arrese, M. Perspectives on Precision Medicine Approaches to NAFLD Diagnosis and Management. Adv. Ther. 2021, 38, 2130–2158. [Google Scholar] [CrossRef]
  290. Sveinbjornsson, G.; Ulfarsson, M.O.; Thorolfsdottir, R.B.; Jonsson, B.A.; Einarsson, E.; Gunnlaugsson, G.; Rognvaldsson, S.; Arnar, D.O.; Baldvinsson, M.; Bjarnason, R.G.; et al. Multiomics study of nonalcoholic fatty liver disease. Nat. Genet. 2022, 54, 1652–1663. [Google Scholar] [CrossRef]
  291. Lin, J.; Zhang, R.; Liu, H.; Zhu, Y.; Dong, N.; Qu, Q.; Bi, H.; Zhang, L.; Luo, O.; Sun, L.; et al. Multi-omics analysis of the biological mechanism of the pathogenesis of non-alcoholic fatty liver disease. Front. Microbiol. 2024, 15, 1379064. [Google Scholar] [CrossRef]
  292. Azad, M.A.K.; Sarker, M.; Li, T.; Yin, J. Probiotic Species in the Modulation of Gut Microbiota: An Overview. BioMed Res. Int. 2018, 2018, 9478630. [Google Scholar] [CrossRef]
  293. Cheng, D.; Song, J.; Xie, M.; Song, D. The bidirectional relationship between host physiology and microbiota and health benefits of probiotics: A review. Trends Food Sci. Technol. 2019, 91, 426–435. [Google Scholar] [CrossRef]
  294. Mennigen, R.; Bruewer, M. Effect of probiotics on intestinal barrier function. Ann. N. Y. Acad. Sci. 2009, 1165, 183–189. [Google Scholar] [CrossRef]
  295. Yousefi, B.; Eslami, M.; Ghasemian, A.; Kokhaei, P.; Salek Farrokhi, A.; Darabi, N. Probiotics importance and their immunomodulatory properties. J. Cell. Physiol. 2019, 234, 8008–8018. [Google Scholar] [CrossRef] [PubMed]
  296. Xue, L.; He, J.; Gao, N.; Lu, X.; Li, M.; Wu, X.; Liu, Z.; Jin, Y.; Liu, J.; Xu, J.; et al. Probiotics may delay the progression of nonalcoholic fatty liver disease by restoring the gut microbiota structure and improving intestinal endotoxemia. Sci. Rep. 2017, 7, 45176. [Google Scholar] [CrossRef]
  297. Kobyliak, N.; Abenavoli, L.; Mykhalchyshyn, G.; Kononenko, L.; Boccuto, L.; Kyriienko, D.; Dynnyk, O. A Multi-strain Probiotic Reduces the Fatty Liver Index, Cytokines and Aminotransferase levels in NAFLD Patients: Evidence from a Randomized Clinical Trial. J. Gastrointest. Liver Dis. JGLD 2018, 27, 41–49. [Google Scholar] [CrossRef]
  298. Iacono, A.; Raso, G.M.; Canani, R.B.; Calignano, A.; Meli, R. Probiotics as an emerging therapeutic strategy to treat NAFLD: Focus on molecular and biochemical mechanisms. J. Nutr. Biochem. 2011, 22, 699–711. [Google Scholar] [CrossRef]
  299. Behrouz, V.; Aryaeian, N.; Zahedi, M.J.; Jazayeri, S. Effects of probiotic and prebiotic supplementation on metabolic parameters, liver aminotransferases, and systemic inflammation in nonalcoholic fatty liver disease: A randomized clinical trial. J. Food Sci. 2020, 85, 3611–3617. [Google Scholar] [CrossRef]
  300. Parnell, J.A.; Raman, M.; Rioux, K.P.; Reimer, R.A. The potential role of prebiotic fibre for treatment and management of non-alcoholic fatty liver disease and associated obesity and insulin resistance. Liver Int. Off. J. Int. Assoc. Study Liver 2012, 32, 701–711. [Google Scholar] [CrossRef]
  301. Slavin, J. Fiber and prebiotics: Mechanisms and health benefits. Nutrients 2013, 5, 1417–1435. [Google Scholar] [CrossRef]
  302. Holscher, H.D. Dietary fiber and prebiotics and the gastrointestinal microbiota. Gut Microbes 2017, 8, 172–184. [Google Scholar] [CrossRef]
  303. Guo, Q.; Li, Y.; Dai, X.; Wang, B.; Zhang, J.; Cao, H. Polysaccharides: The Potential Prebiotics for Metabolic Associated Fatty Liver Disease (MAFLD). Nutrients 2023, 15, 3722. [Google Scholar] [CrossRef]
  304. Wang, S.; Sun, W.; Swallah, M.S.; Amin, K.; Lyu, B.; Fan, H.; Zhang, Z.; Yu, H. Preparation and characterization of soybean insoluble dietary fiber and its prebiotic effect on dyslipidemia and hepatic steatosis in high fat-fed C57BL/6J mice. Food Funct. 2021, 12, 8760–8773. [Google Scholar] [CrossRef]
  305. Hadi, A.; Mohammadi, H.; Miraghajani, M.; Ghaedi, E. Efficacy of synbiotic supplementation in patients with nonalcoholic fatty liver disease: A systematic review and meta-analysis of clinical trials: Synbiotic supplementation and NAFLD. Crit. Rev. Food Sci. Nutr. 2019, 59, 2494–2505. [Google Scholar] [CrossRef] [PubMed]
  306. Zhang, C.; Fang, T.; Shi, L.; Wang, Y.; Deng, X.; Wang, J.; Zhou, Y. The synbiotic combination of probiotics and inulin improves NAFLD though modulating gut microbiota. J. Nutr. Biochem. 2024, 125, 109546. [Google Scholar] [CrossRef]
  307. Khan, M.Y.; Mihali, A.B.; Rawala, M.S.; Aslam, A.; Siddiqui, W.J. The promising role of probiotic and synbiotic therapy in aminotransferase levels and inflammatory markers in patients with nonalcoholic fatty liver disease—A systematic review and meta-analysis. Eur. J. Gastroenterol. Hepatol. 2019, 31, 703–715. [Google Scholar] [CrossRef] [PubMed]
  308. Vyas, U.; Ranganathan, N. Probiotics, prebiotics, and synbiotics: Gut and beyond. Gastroenterol. Res. Pract. 2012, 2012, 872716. [Google Scholar] [CrossRef]
  309. Allegretti, J.R.; Mullish, B.H.; Kelly, C.; Fischer, M. The evolution of the use of faecal microbiota transplantation and emerging therapeutic indications. Lancet 2019, 394, 420–431. [Google Scholar] [CrossRef]
  310. Non-Alcoholic Fatty Liver Disease and Gut Microbial Dysbiosis—Underlying Mechanisms and Gut Microbiota Mediated Treatment Strategies. Available online: http://ouci.dntb.gov.ua/en/works/4bwZWedl/ (accessed on 7 February 2025).
  311. Craven, L.; Rahman, A.; Nair Parvathy, S.; Beaton, M.; Silverman, J.; Qumosani, K.; Hramiak, I.; Hegele, R.; Joy, T.; Meddings, J.; et al. Allogenic Fecal Microbiota Transplantation in Patients With Nonalcoholic Fatty Liver Disease Improves Abnormal Small Intestinal Permeability: A Randomized Control Trial. Am. J. Gastroenterol. 2020, 115, 1055–1065. [Google Scholar] [CrossRef]
  312. Del Barrio, M.; Lavín, L.; Santos-Laso, Á.; Arias-Loste, M.T.; Odriozola, A.; Rodriguez-Duque, J.C.; Rivas, C.; Iruzubieta, P.; Crespo, J. Faecal Microbiota Transplantation, Paving the Way to Treat Non-Alcoholic Fatty Liver Disease. Int. J. Mol. Sci. 2023, 24, 6123. [Google Scholar] [CrossRef]
  313. Popa, S.L.; Ismaiel, A.; Cristina, P.; Cristina, M.; Chiarioni, G.; David, L.; Dumitrascu, D.L. Non-Alcoholic Fatty Liver Disease: Implementing Complete Automated Diagnosis and Staging. A Systematic Review. Diagnostics 2021, 11, 1078. [Google Scholar] [CrossRef]
  314. Yang, B.; Lu, H.; Ran, Y. Advancing non-alcoholic fatty liver disease prediction: A comprehensive machine learning approach integrating SHAP interpretability and multi-cohort validation. Front. Endocrinol. 2024, 15, 1450317. [Google Scholar] [CrossRef]
  315. 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. Ther. Adv. Gastroenterol. 2021, 14, 17562848211062807. [Google Scholar] [CrossRef]
  316. Ma, X.; Yang, C.; Liang, K.; Sun, B.; Jin, W.; Chen, L.; Dong, M.; Liu, S.; Xin, Y.; Zhuang, L. A predictive model for the diagnosis of non-alcoholic fatty liver disease based on an integrated machine learning method. Am. J. Transl. Res. 2021, 13, 12704–12713. [Google Scholar] [PubMed]
  317. Sghaireen, M.G.; Al-Smadi, Y.; Al-Qerem, A.; Srivastava, K.C.; Ganji, K.K.; Alam, M.K.; Nashwan, S.; Khader, Y. Machine Learning Approach for Metabolic Syndrome Diagnosis Using Explainable Data-Augmentation-Based Classification. Diagnostics 2022, 12, 3117. [Google Scholar] [CrossRef] [PubMed]
  318. Cao, W.; An, X.; Cong, L.; Lyu, C.; Zhou, Q.; Guo, R. Application of Deep Learning in Quantitative Analysis of 2-Dimensional Ultrasound Imaging of Nonalcoholic Fatty Liver Disease. J. Ultrasound Med. Off. J. Am. Inst. Ultrasound Med. 2020, 39, 51–59. [Google Scholar] [CrossRef] [PubMed]
  319. Pugliese, N.; Bertazzoni, A.; Hassan, C.; Schattenberg, J.M.; Aghemo, A. Revolutionizing MASLD: How Artificial Intelligence Is Shaping the Future of Liver Care. Cancers 2025, 17, 722. [Google Scholar] [CrossRef]
  320. Aggarwal, P.; Alkhouri, N. Artificial Intelligence in Nonalcoholic Fatty Liver Disease: A New Frontier in Diagnosis and Treatment. Clin. Liver Dis. 2021, 17, 392–397. [Google Scholar] [CrossRef]
  321. Xia, J.; Wang, Z.; Huan, Y.; Xue, W.; Wang, X.; Wang, Y.; Liu, Z.; Hsieh, J.-H.; Zhang, L.; Wu, S.; et al. Pose Filter-Based Ensemble Learning Enables Discovery of Orally Active, Nonsteroidal Farnesoid X Receptor Agonists. J. Chem. Inf. Model. 2020, 60, 1202–1214. [Google Scholar] [CrossRef]
  322. Velliou, R.-I.; Giannousi, E.; Ralliou, C.; Kassi, E.; Chatzigeorgiou, A. Ex Vivo Tools and Models in MASLD Research. Cells 2024, 13, 1827. [Google Scholar] [CrossRef]
  323. De Chiara, F.; Ferret-Miñana, A.; Ramón-Azcón, J. The Synergy between Organ-on-a-Chip and Artificial Intelligence for the Study of NAFLD: From Basic Science to Clinical Research. Biomedicines 2021, 9, 248. [Google Scholar] [CrossRef]
  324. Chu, A.; Nguyen, D.; Talathi, S.S.; Wilson, A.C.; Ye, C.; Smith, W.L.; Kaplan, A.D.; Duoss, E.B.; Stolaroff, J.K.; Giera, B. Automated detection and sorting of microencapsulation via machine learning. Lab. Chip 2019, 19, 1808–1817. [Google Scholar] [CrossRef]
  325. Kalapala, R.; Rughwani, H.; Reddy, D.N. Artificial Intelligence in Hepatology—Ready for the Primetime. J. Clin. Exp. Hepatol. 2023, 13, 149–161. [Google Scholar] [CrossRef]
  326. Reiniš, J.; Petrenko, O.; Simbrunner, B.; Hofer, B.S.; Schepis, F.; Scoppettuolo, M.; Saltini, D.; Indulti, F.; Guasconi, T.; Albillos, A.; et al. Assessment of portal hypertension severity using machine learning models in patients with compensated cirrhosis. J. Hepatol. 2023, 78, 390–400. [Google Scholar] [CrossRef] [PubMed]
  327. Li, Y.; Wang, X.; Zhang, J.; Zhang, S.; Jiao, J. Applications of artificial intelligence (AI) in researches on non-alcoholic fatty liver disease(NAFLD): A systematic review. Rev. Endocr. Metab. Disord. 2022, 23, 387–400. [Google Scholar] [CrossRef] [PubMed]
  328. Baser, O.; Samayoa, G.; Yapar, N.; Baser, E. Artificial Intelligence in Identifying Patients With Undiagnosed Nonalcoholic Steatohepatitis. J. Health Econ. Outcomes Res. 2024, 11, 86–94. [Google Scholar] [CrossRef]
  329. Hagström, H.; Vessby, J.; Ekstedt, M.; Shang, Y. 99% of patients with NAFLD meet MASLD criteria and natural history is therefore identical. J. Hepatol. 2024, 80, e76–e77. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Mechanistic contributors to metabolic-associated fatty liver disease (MASLD). The figure highlights six interconnected mechanisms driving the pathogenesis of MASLD, including IR, adipose tissue dysfunction, lipo-toxicity and oxidative stress, gut dysbiosis, genetic polymorphisms, and chronic inflammation. These pathways collectively exacerbate hepatic fat accumulation, inflammation, and fibrosis, illustrating the multifactorial nature of MASLD and its systemic impacts. Understanding these mechanisms is crucial for developing targeted therapeutic strategies.
Figure 1. Mechanistic contributors to metabolic-associated fatty liver disease (MASLD). The figure highlights six interconnected mechanisms driving the pathogenesis of MASLD, including IR, adipose tissue dysfunction, lipo-toxicity and oxidative stress, gut dysbiosis, genetic polymorphisms, and chronic inflammation. These pathways collectively exacerbate hepatic fat accumulation, inflammation, and fibrosis, illustrating the multifactorial nature of MASLD and its systemic impacts. Understanding these mechanisms is crucial for developing targeted therapeutic strategies.
Ijms 26 03448 g001
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Boulos, M.; Mousa, R.S.; Jeries, N.; Simaan, E.; Alam, K.; Bulus, B.; Assy, N. Hidden in the Fat: Unpacking the Metabolic Tango Between Metabolic Dysfunction-Associated Steatotic Liver Disease and Metabolic Syndrome. Int. J. Mol. Sci. 2025, 26, 3448. https://doi.org/10.3390/ijms26073448

AMA Style

Boulos M, Mousa RS, Jeries N, Simaan E, Alam K, Bulus B, Assy N. Hidden in the Fat: Unpacking the Metabolic Tango Between Metabolic Dysfunction-Associated Steatotic Liver Disease and Metabolic Syndrome. International Journal of Molecular Sciences. 2025; 26(7):3448. https://doi.org/10.3390/ijms26073448

Chicago/Turabian Style

Boulos, Mariana, Rabia S. Mousa, Nizar Jeries, Elias Simaan, Klode Alam, Bulus Bulus, and Nimer Assy. 2025. "Hidden in the Fat: Unpacking the Metabolic Tango Between Metabolic Dysfunction-Associated Steatotic Liver Disease and Metabolic Syndrome" International Journal of Molecular Sciences 26, no. 7: 3448. https://doi.org/10.3390/ijms26073448

APA Style

Boulos, M., Mousa, R. S., Jeries, N., Simaan, E., Alam, K., Bulus, B., & Assy, N. (2025). Hidden in the Fat: Unpacking the Metabolic Tango Between Metabolic Dysfunction-Associated Steatotic Liver Disease and Metabolic Syndrome. International Journal of Molecular Sciences, 26(7), 3448. https://doi.org/10.3390/ijms26073448

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop