Next Article in Journal
Comparison of Differentially Expressed Genes in Human Versus in Chimeric Mouse Livers Following HBV Infection
Previous Article in Journal
Comparative Analysis of the Human Proteome Profile in Visceral Adipose and Liver Tissue in Individuals with Obesity with and Without MASLD and MASH
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

The Intestinal Thread of Fate: How the Microbiota Shapes the Story of Liver Disease

1
Department of Infectious Diseases, San Carlo Hospital, 85100 Potenza, Italy
2
Liver Unit, AORN S. G. Moscati, “A. Landolfi” Hospital, 83029 Solofra, Italy
3
Department of Medicine and Health Science “Vincenzo Tiberio”, Università degli Studi del Molise, 86100 Campobasso, Italy
4
Department of Advanced Medical and Surgical Sciences, University of Campania “Luigi Vanvitelli”, 80138 Napoli, Italy
5
Department of Human Sciences and Promotion of the Quality of Life, San Raffaele Roma Open University, 00166 Rome, Italy
*
Author to whom correspondence should be addressed.
Livers 2025, 5(2), 17; https://doi.org/10.3390/livers5020017
Submission received: 28 January 2025 / Revised: 25 March 2025 / Accepted: 8 April 2025 / Published: 10 April 2025

Abstract

:
Metabolic dysfunction–associated steatotic liver disease (MASLD) is a multifactorial condition linked to liver injury, insulin resistance, and disrupted gut–liver interactions. A key aspect of MASLD pathogenesis is the dysfunction of intestinal barriers, including mechanical, immunological, and microbial alterations that amplify liver damage. The disruption of tight junctions and increased intestinal permeability allow microbial products, such as lipopolysaccharides, to enter the bloodstream, triggering liver inflammation via Kupffer cell activation. In MASLD, the gut vascular barrier is also compromised, marked by increased expression of PV-1. Additionally, dysbiosis, driven by high-fat, high-sugar diets, shifts the gut microbiota toward pro-inflammatory species, exacerbating systemic inflammation and intestinal permeability. This imbalance activates Toll-like receptor signaling, which promotes endotoxin-induced liver injury. Gut dysbiosis further impairs lipid metabolism, contributing to hepatic steatosis and MASLD progression. The gut–liver axis plays a critical role, with factors like altered bile acid metabolism and toxic metabolites such as hydrogen sulfide worsening intestinal barrier function and fueling chronic inflammation. This review aims to explore the complex role of the gut–liver axis in MASLD progression, highlighting the mechanisms of intestinal barrier dysfunction, dysbiosis, and microbial contributions to liver injury. It also discusses therapeutic strategies targeting intestinal barriers, including dietary and microbiota-based interventions, while acknowledging the challenges of personalized treatment approaches. Future research should focus on multi-omics technologies and the safety and efficacy of microbiota-targeted therapies in MASLD management.

1. Introduction

Metabolic dysfunction–associated steatotic liver disease (MASLD) represents a recent nosological redefinition in the landscape of liver disorders, introduced to replace the term non-alcoholic fatty liver disease (NAFLD). This semantic update emphasizes the primary role of metabolic disorders in the pathogenesis of the condition [1]. MASLD is defined by the excessive accumulation of triglycerides in the liver, leading to hepatic steatosis without significant alcohol consumption, in the presence of at least one cardiometabolic risk factor, including obesity, insulin resistance (IR), and type 2 diabetes mellitus (T2DM) [1]. The global increase in the prevalence of these conditions has made MASLD one of the leading causes of chronic liver disease, closely linked to the rising incidence of obesity and metabolic syndrome [2]. This condition affects approximately 30% of the global population, with its prevalence exceeding 50% in high-risk groups, such as individuals with T2DM. In Latin America, the prevalence of this condition exceeds 40%, while in Europe and the United States, it ranges between 25% and 31% [3,4,5].
Several risk factors contribute to the onset and progression of MASLD, including genetic predisposition (e.g., PNPLA3 polymorphisms), dietary habits, and sedentary lifestyle [3]. The clinical spectrum of MASLD varies from simple steatosis to metabolic dysfunction–associated steatohepatitis (MASH), which is characterized by hepatocyte injury and inflammation, potentially progressing to fibrosis, cirrhosis, and hepatocellular carcinoma (HCC) [3]. Beyond liver-related outcomes, MASLD is a strong predictor of cardiovascular disease (CVD), which remains the leading cause of mortality in affected patients. Additionally, MASLD has been associated with an increased risk of chronic kidney disease (CKD), extrahepatic cancers, and all-cause mortality [6,7,8,9].
The diagnosis of MASLD relies on the presence of hepatic steatosis, which can be detected using imaging modalities such as ultrasound, magnetic resonance imaging (MRI), or controlled attenuation parameter (CAP) in transient elastography [3]. Liver biopsy remains the gold standard for assessing fibrosis and inflammation in advanced cases, although non-invasive biomarkers and fibrosis scores, such as the FIB-4 index and NAFLD fibrosis score, are increasingly utilized in clinical practice [3].
From a pathogenetic perspective, MASLD is a multifactorial condition in which IR, lipotoxicity, oxidative stress, and the activation of innate and adaptive immune responses play crucial roles. These mechanisms contribute to progressive liver damage [10,11,12]. In recent years, the gut–liver axis has taken center stage in studying the pathogenesis and progression of MASLD, attracting growing scientific interest [13]. This axis is characterized by a bidirectional interaction between the gastrointestinal tract and the liver, mediated by complex biological signals, including gut-microbiota-derived metabolites, immune molecules, and neurotransmitters [14,15]. Portal circulation serves as the primary connection point, transporting nutrients, bacterial endotoxins, bile acids, and other bioactive molecules, influencing liver function and contributing to liver disease pathogenesis [16].
Under physiological conditions, the gut microbiota maintains hepatic homeostasis by facilitating digestion, producing beneficial metabolites such as short-chain fatty acids (SCFAs), and modulating intestinal and systemic immune responses [17]. Pathological alterations in gut microbiota composition, known as dysbiosis, can impair intestinal permeability, promoting the translocation of bacteria, lipopolysaccharides (LPS), and other endotoxins into the portal circulation. This phenomenon, referred to as “leaky gut”, triggers hepatic inflammatory responses [18,19,20]. This process contributes to the progression of the disease toward metabolic dysfunction–associated steatohepatitis (MASH) and fibrosis [20,21]. Emerging data underscore the pivotal role of the gut–liver axis in MASLD pathogenesis. Alterations in gut microbiota composition and impaired intestinal permeability stimulate the activation of Kupffer cells and hepatic stellate cells, promoting fibrogenesis and the secretion of pro-inflammatory cytokines [20,21,22]. Additionally, gut-microbiota-derived metabolites, such as trimethylamine N-oxide (TMAO), modulate hepatic lipid metabolism and contribute to the progression of cardiovascular and metabolic diseases, including MASLD [23,24]. Accordingly, this review aims to provide a comprehensive overview of the molecular and cellular mechanisms that underpin the gut–liver interaction in MASLD. We focus on the contributions of intestinal dysbiosis, altered permeability, and bacterial metabolites, and we discuss emerging therapeutic strategies, including probiotics, prebiotics, and novel pharmacologic agents, that target this axis to improve clinical outcomes in MASLD patients.

2. Literature Research Methodology

A comprehensive literature search was conducted to capture a wide spectrum of studies relevant to MASLD and its intricate relationship with the gut microbiota. The search strategy involved querying major databases PubMed/MEDLINE, EMBASE, Scopus, and Web of Science using a combination of keywords and Medical Subject Headings (MeSH) terms. These search terms included, but were not limited to, “MASLD”, “Metabolic Dysfunction-Associated Steatotic Liver Disease”, “gut microbiota”, “intestinal barriers”, “liver disease”, “pathogenesis”, “risk factors”, “dysbiosis”, and “Fecal Microbiota Transplantation”. No restrictions were placed on the publication date to ensure the inclusion of both seminal and contemporary literature, while only studies published in English were considered. In addition to database searches, the reference lists of relevant articles were manually screened to identify further studies that might have been missed initially, driven by the specific themes of the review. Studies were selected based on their relevance to these topics and their methodological rigor.

3. Risk Factors and Predisposition for MASLD

3.1. Genetic and Epigenetic Factors

Genetic and epigenetic predisposition plays a critical role in the development and progression of MASLD and HCC. Advanced genomic studies, such as genome-wide association studies (GWAS) and exome-wide association studies (EWAS), have identified key single nucleotide polymorphisms (SNPs) in genes including PNPLA3, TM6SF2, MBOAT7, GCKR, and HSD17B13. These genetic variants modulate crucial processes such as lipid metabolism, oxidative stress, and hepatic inflammation, influencing disease susceptibility and progression (Table 1) [25,26].

3.1.1. PNPLA3

The PNPLA3 (patatin-like phospholipase domain-containing protein 3) gene encodes a multifunctional enzyme that regulates hepatic lipid metabolism, exhibiting both triglyceride hydrolase and acyltransferase activity on lipid droplets [27]. The I148M variant (rs738409 C>G), resulting from an isoleucine-to-methionine substitution at position 148, was the first genetic variant identified as a major risk factor for NAFLD and has since been strongly associated with MASLD and HCC risk [28]. This mutation impairs PNPLA3 lipase function, leading to triglyceride accumulation in hepatocytes and contributing to steatosis, steatohepatitis, fibrosis, and hepatocellular carcinoma [29]. Experimental studies in murine models indicate that PNPLA3 interacts with CGI-58, a protein essential for lipid droplet degradation. The absence of hepatic CGI-58 exacerbates the effects of the I148M variant, further increasing lipid deposition [30]. Notably, another PNPLA3 polymorphism, rs6006460 (G>T), appears to have a protective role, potentially altering enzymatic activity in a way that mitigates hepatic lipid accumulation and HCC risk [31,32].

3.1.2. TM6SF2

The TM6SF2 (transmembrane 6 superfamily member 2) gene regulates the hepatic lipid metabolism by influencing triglyceride secretion via very-low-density lipoprotein (VLDL) formation [33]. The E167K variant (rs58542926), involving a glutamate-to-lysine amino acid substitution, reduces VLDL secretion efficiency, leading to hepatic lipid accumulation and promoting the progression of steatosis and fibrosis [34]. Although this polymorphism is associated with lower total cholesterol and LDL levels, it paradoxically increases the risk of advanced fibrosis and MASLD-HCC, highlighting the dual impact of genetic alterations on lipid metabolism and liver disease progression [35,36]. Another variant, rs5842926 (C>T), has also been implicated in modifying lipid metabolism and liver disease susceptibility [35,36].

3.1.3. GCKR

The GCKR (glucokinase regulatory protein) gene encodes a key regulator of glucokinase, an enzyme involved in glucose metabolism. The rs1260326 (C>T) polymorphism enhances glycolysis and de novo lipogenesis, thereby increasing the risk of hepatic lipid accumulation and the progression to MASH and fibrosis [37,38,39].

3.1.4. HSD17B13

The HSD17B13 (hydroxysteroid 17-beta dehydrogenase 13) gene encodes an enzyme associated with hepatic lipid droplets. The rs72613567 (T>TA) variant has been identified as a protective factor against MASLD, particularly in individuals carrying the PNPLA3 I148M risk allele. This variant reduces hepatic inflammation and fibrosis and is associated with a lower risk of HCC development [40,41].

3.1.5. Epigenetic and Epitranscriptomic Modifications

Beyond genetics, epigenetic mechanisms, including DNA methylation and histone modifications, influence gene expression and the hepatic microenvironment, further modulating disease susceptibility and progression [42]. Emerging evidence highlights the role of epitranscriptomic modifications, such as N6-methyladenine (m6A), in regulating MASLD-HCC progression [42,43]. Enzymes such as METTL3 (a “writer” of m6A modifications) and YTHDF1 (a “reader” of m6A marks) modulate the expression of key genes and activate oncogenic pathways. Notably, the EZH2/interleukin-6 (IL-6) signaling axis has been implicated in immune dysfunction and tumor progression [44].
The complex interplay of genetic, epigenetic, and epitranscriptomic factors significantly contributes to the pathogenesis of MASLD and its progression to HCC. These mechanisms represent crucial targets for innovative diagnostic and therapeutic strategies aimed at improving clinical outcomes in high-risk patients.

3.2. Metabolic Consequences and Other Contributing Factors

MASLD is closely linked with metabolic syndrome, a condition defined by the presence of at least three of the following five criteria—central obesity (increased waist circumference), hypertriglyceridemia, low levels of high-density lipoprotein cholesterol (HDL-C), impaired fasting glucose or T2DM, and arterial hypertension—which collectively serve as primary risk determinants for liver pathologies [45,46]. In this context, each component contributes through distinct yet interrelated mechanisms that not only initiate hepatic steatosis but also drive its progression to more advanced states, such as MASH and fibrosis (Table 1).
Central obesity plays a pivotal role in MASLD pathogenesis. The hypertrophy of visceral adipocytes leads to reduced insulin sensitivity and altered adipocytokine secretion, resulting in an increased release of free fatty acids (FFAs) into the circulation [47]. Once in the bloodstream, these FFAs are transported via the portal vein to the liver, where they accumulate in hepatocytes through processes that include de novo lipogenesis, diminished hepatic lipid metabolism, and augmented exogenous lipid supply. This lipid overload triggers mitochondrial dysfunction and enhances reactive oxygen species (ROS) production, while toxic lipid metabolites—such as ceramides and diacylglycerols—induce endoplasmic reticulum (ER) stress, activate inflammatory cascades, and lead to hepatocyte cell death [48,49].
Insulin resistance (IR), a hallmark of metabolic syndrome, further exacerbates hepatic lipid deposition by pro de novo lipogenesis and impairing fatty acid oxidation [50,51]. This metabolic imbalance sets off a vicious cycle characterized by chronic hyperglycemia and hyperinsulinemia, which in turn amplify pro-inflammatory and pro-fibrotic pathways, thereby escalating liver damage. Moreover, hyperinsulinemia impedes lipolysis in adipocytes, resulting in a further increase in circulating FFAs that continue to fuel hepatic steatosis [52].
The metabolic dysregulation is compounded by hypertriglyceridemia, which contributes to the production of toxic lipid species that intensify oxidative stress and inflammation. Elevated triglyceride levels are also associated with the formation of small, dense VLDL and LDL particles with high atherogenic potential, linking these lipid abnormalities to both liver and cardiovascular disease [53,54]. Concurrently, reduced levels of HDL-C compromise reverse cholesterol transport and, in the presence of systemic inflammation and IR, lead to a transformation of HDL into pro-inflammatory particles. Alterations in HDL composition, including diminished levels of apolipoproteins ApoAI and ApoAII, further exacerbate systemic inflammation and promote hepatic lipid accumulation [55,56,57]. Additional metabolic disturbances such as impaired fasting glucose and T2DM are closely tied to IR and contribute to a pro-inflammatory state that worsens liver injury and perpetuates the cycle of hyperinsulinemia and intrahepatic lipid accumulation [58,59,60]. Likewise, arterial hypertension—mediated by the chronic activation of the renin–angiotensin–aldosterone system and increased expression of angiotensin type 1 receptors—leads to vasoconstriction, vascular fibrosis, and reduced nitric oxide synthesis. These vascular changes not only disrupt hepatic microcirculation but also exacerbate the intricate interplay between metabolic, cardiovascular, and hepatic dysfunctions [61,62].

3.3. Prevalence of Lean/Non-Obese MASLD and Medication-Induced MASH

Recent studies suggest that approximately 10–15% of patients with MASLD are lean or non-obese, demonstrating that liver disease can develop even in the absence of overt obesity [12] (Table 1). Although these individuals typically exhibit a normal body mass index, they may harbor distinct metabolic abnormalities, such as increased visceral adiposity, subtle insulin resistance, or dyslipidemia, that predispose them to hepatic fat accumulation and progression to steatohepatitis [12]. This lean/non-obese phenotype of MASLD may involve unique pathophysiological mechanisms, including specific genetic and epigenetic factors, that differentiate it from the classic presentation of MASLD associated with metabolic syndrome [8,12]. As such, these cases require tailored diagnostic approaches, such as advanced imaging modalities and metabolic profiling, to accurately assess liver fat content and associated risks. In turn, therapeutic strategies may also need to be customized, focusing on both lifestyle interventions and targeted pharmacological treatments that address these specific metabolic derangements [8,12].
In parallel, medication-induced MASH represents another important, though less common, etiology of liver injury [46]. Certain drugs, including tamoxifen, corticosteroids, and various antipsychotic agents, have been implicated in the development of hepatic steatosis and the progression to MASH [46]. The underlying mechanisms for medication-induced MASH may involve direct hepatotoxicity, alterations in lipid metabolism, or disruptions in insulin signaling pathways, which can mimic the metabolic disturbances observed in MASLD associated with metabolic syndrome [46]. Therefore, a comprehensive clinical evaluation should include a detailed medication history to identify potential drug-related contributions to liver disease. Recognizing medication-induced MASH is crucial, as it may necessitate a modification of the therapeutic regimen, such as dose adjustment or switching to an alternative medication, along with supportive measures to protect liver function [46].
In summary, the close relationship between metabolic syndrome and MASLD underscores the need for integrated approaches to mitigate metabolic risk factors. Targeted interventions, including lifestyle modifications, weight reduction, and pharmacological therapies to improve insulin sensitivity and reduce systemic inflammation, are essential to prevent disease progression and improve clinical outcomes.
Table 1. Risk factors and predisposition for MASLD.
Table 1. Risk factors and predisposition for MASLD.
CategorySpecific FactorMain Pathogenetic MechanismsClinical Effects/RisksRef.
GeneticPNPLA3 (I148M, rs738409, G allele)Impaired lipase activity; accumulation of triglycerides in hepatocytes; lipid droplet remodeling dysfunctionSteatosis, NASH, Fibrosis, Increased risk of HCC[25,26,27,28,29,30,31,32]
PNPLA3 (rs6006460, G>T variant)Modulates risk in protective directionPotential protective role against hepatic lipid accumulation[25,26,27,28,29,30,31,32]
TM6SF2 (E167K, rs5842926, C>T variant)Reduced VLDL secretion; hepatic lipid accumulation; lower total cholesterol and LDL levelsProgression to advanced hepatic fibrosis, MASLD-HCC[33,34,35,36]
GCKR (rs1260326, C>T variant)Enhanced glycolysis and de novo lipogenesis; increased hepatic lipid accumulationProgression to MASH and fibrosis[37,38,39]
MBOAT7 polymorphismsAlters phospholipid remodeling; increases hepatic fat accumulation and inflammationAssociated with fibrosis and progression of MASLD[25,26]
HSD17B13 (rs72613567, T>TA variant)Loss-of-function variant; reduced hepatic inflammation and fibrosis, especially in PNPLA3 I148M carriersProtective against MASLD progression and HCC[40,41,42]
EpigeneticDNA methylationAlters gene expression; promotes pro-inflammatory and pro-fibrotic pathwaysProgression from MASLD to MASH, fibrosis, HCC[43]
Histone modificationsModifies chromatin structure; impacts transcriptional activity of genes related to inflammation and fibrosisIncreased susceptibility to MASLD and progression to advanced liver disease[43]
Epitranscriptomicm6A RNA modificationRegulates RNA stability and translation; affects lipid metabolism and oncogenic pathwaysPromotes MASLD progression and HCC development[44]
METTL3 (m6A writer) and YTHDF1 (m6A reader)Modulate gene expression; activate oncogenic signals (e.g., EZH2, IL-6)Immune dysfunction, enhanced tumorigenesis, risk of HCC[43,44,45]
Metabolic SyndromeCentral obesity (increased waist circumference)Visceral adipocyte hypertrophy; increased FFAs; portal delivery of lipids to the liverSteatosis, mitochondrial dysfunction, ROS production, hepatocyte apoptosis[46,47,48,49,50]
Insulin resistance (IR)Promotes de novo lipogenesis; impairs fatty acid oxidation; perpetuates hyperglycemia and hyperinsulinemiaInflammatory and fibrotic liver damage; progression to MASH[51,52,53,54]
HypertriglyceridemiaIncreases toxic lipid species; generates small dense VLDL and LDL particles with high atherogenic potentialOxidative stress, inflammation, liver and cardiovascular disease[55,56,57,58]
Low HDL-CImpaired reverse cholesterol transport; pro-inflammatory HDL phenotype with reduced ApoAI and ApoAII levelsSystemic inflammation; hepatic lipid accumulation[56,57,58]
Impaired fasting glucose/Type 2 Diabetes Mellitus (T2DM)Chronic hyperinsulinemia; promotes pro-inflammatory state; exacerbates hepatic lipid depositionAccelerated liver injury, steatosis, fibrosis[63,64,65]
Arterial hypertensionActivation of RAAS; vascular fibrosis; decreased NO synthesis; impaired hepatic microcirculationWorsens metabolic, cardiovascular, and hepatic dysfunction[66,67,68,69,70]
Lean/Non-obese MASLDIncreased visceral adiposityExcess intra-abdominal fat despite normal BMI; metabolic abnormalities (IR, dyslipidemia)Hepatic fat accumulation, progression to steatohepatitis and fibrosis[8,12]
Subtle insulin resistanceMild impairment in insulin action, undetected by conventional markersContributes to hepatic lipid accumulation[50,51]
DyslipidemiaElevated triglycerides, low HDL-CPromotes hepatic steatosis[53,54,55,56,57]
Specific genetic and epigenetic factors unique to lean MASLDGenetic variants distinct from classic MASLDDifferent pathogenetic pathways; tailored diagnostic and therapeutic approaches[8,12]
Drug-induced MASHTamoxifenInduces hepatic steatosis via altered lipid metabolism and mitochondrial dysfunctionDrug-induced MASLD/MASH[46]
CorticosteroidsIncreases gluconeogenesis and lipogenesis; promotes IRHepatic steatosis, progression to MASH[46]
Antipsychotics (e.g., olanzapine)Promotes weight gain, IR, and dyslipidemia; direct hepatotoxic effectsMASLD/MASH secondary to pharmacologic agents[46]
Other Contributing FactorsHigh-calorie dietExcess caloric intake; stimulates de novo lipogenesisHepatic steatosis, progression to MASLD[45,46,47,48,49,50,51,52,53,54]
Physical inactivityReduced energy expenditure; promotes weight gain and IRIncreased risk of MASLD[45,46,47,48,49,50,51,52,53,54]
Intestinal dysbiosisAlters gut–liver axis; increases intestinal permeability and endotoxemiaInflammation, hepatic injury, progression to MASLD[15]

4. Pathogenesis of MASLD

The pathogenesis of MASLD was initially described by the “two-hit hypothesis”, which proposed hepatic lipid accumulation (first hit) followed by oxidative stress and inflammation (second hit) [63]. However, this model has been recently surpassed by the “multiple-hit hypothesis”, recognizing the simultaneous and complex interplay of metabolic, genetic, immunological, and environmental factors [64].
Systemic inflammation is a cornerstone in the progression of MASLD, bridging metabolic dysfunction and chronic liver damage. While hepatic lipid accumulation is the initial event, progression to MASH and fibrosis depends heavily on immune system activation and chronic inflammatory states [65]. In individuals with MASLD, systemic inflammation not only contributes to liver damage but also exacerbates IR and endothelial dysfunction [66]. Elevated levels of cytokines such as tumor necrosis factor-α (TNF-α), IL-6, and IL-1β sustain low-grade chronic inflammation, amplified by the secretion of pro-inflammatory chemokines like monocyte chemoattractant protein-1 (MCP-1) from hypertrophic adipocytes [66,67].
At cellular level, IR promotes increased adipocyte lipolysis, resulting in a heightened influx of FFAs to the liver. The subsequent accumulation of FFAs in hepatic mitochondria generates ROS, fostering lipotoxicity and triggering both inflammatory and fibrotic processes [69,70]. Mitochondrial overload and ROS production contribute to hepatocyte necrosis and activation of stellate cells, which deposit collagen and drive fibrotic progression [71]. Oxidative stress, primarily from mitochondrial fatty acid oxidation, peroxisomal activity, and endoplasmic reticulum (ER) stress, inflicts damage on lipids, proteins, and DNA, thereby disrupting cellular functions and perpetuating innate immune responses [70,71,72,73]. Toxic lipid peroxidation produces toxic molecules such as MDA and 4-HNE, further damaging cellular membranes and amplifying inflammatory responses [74]. Oxidative stress activates hepatic stellate cells (HSCs), leading to the production of collagen and other extracellular matrix components, driving fibrotic progression [71]. In addition, mitochondrial dysfunction—marked by impaired oxidative phosphorylation and the induction of apoptotic processes—exacerbates both lipid accumulation and fibrogenesis [70,75]. Strategies aimed at enhancing mitochondrial biogenesis and employing selective antioxidants have thus emerged as promising therapeutic avenues [76].
Oxidative stress and redox dysregulation in the ER impair proper protein folding, activating the unfolded protein response (UPR). When UPR becomes maladaptive, it results in hepatocyte apoptosis, further exacerbating fibrosis [77,78]. Compounding these events, impaired lysosomal function, often a consequence of lipid accumulation that destabilizes lysosomes and results in the release of proteolytic enzymes such as cathepsin B, promotes inflammation and cell death [79]. Defective autophagy and reduced lysosomal acid lipase activity exacerbate intrahepatic lipid accumulation, self-perpetuating the cycle of liver damage [80,81].
Apoptosis, regulated by Bcl-2 family proteins, is a crucial process in MASLD progression. Pro-apoptotic proteins like Bax and Bak induce mitochondrial membrane permeabilization, causing cytochrome c release and caspase activation [82,83]. Lipid accumulation activates “BH3-only” proteins that interact with Bcl-2, intensifying hepatocyte apoptosis and accelerating progression to MASH and fibrosis [84,85]. Additionally, ferroptosis, a form of cell death mediated by iron-dependent lipid peroxidation, has emerged as a key mechanism in MASLD. Reduced activity of glutathione peroxidase 4 (GPX4) in the liver impairs ROS neutralization, exacerbating lipid accumulation and fibrosis [86,87]. The use of ferroptosis inhibitors, such as liproxstatin-1, has shown promising effects in preventing liver damage and improving lipid homeostasis, suggesting a potential therapeutic target [88,89].
The immune system plays a central role in MASLD progression. Kupffer cell activation by danger signals, such as LPS and lipid peroxidation products, induces the secretion of pro-inflammatory cytokines (TNF-α, IL-6) and amplifies hepatic oxidative stress [90,91]. Neutrophils also contribute to liver damage by releasing proteolytic enzymes and ROS. The formation of neutrophil extracellular traps (NETs) is associated with fibrosis and hepatocarcinogenesis [92]. T cells and NK cells play immunomodulatory roles: CD8+ T cells promote chronic liver damage, while NK cells limit fibrosis through direct cytotoxicity. However, their functionality can be compromised in chronic inflammation settings, facilitating disease progression [93].
Collectively, these complex cellular and molecular events drive the progression of MASLD from simple steatosis to MASH and advanced fibrosis. Notably, emerging evidence highlights the gut–liver axis as a critical link between hepatic injury and intestinal barrier dysfunction. Disruptions in the intestinal barrier, increasing permeability and allow microbial products such as LPS to enter the portal circulation, further exacerbate hepatic inflammation and fibrogenesis. By facilitating the translocation of pro-inflammatory molecules from the compromised gut barrier to the liver, the gut–liver axis plays a central role in triggering and sustaining inflammatory and fibrotic responses [15]. A deeper understanding of the pathogenic mechanisms underlying MASLD offers new opportunities for targeted therapeutic interventions. Strategies that modulate oxidative stress, improve mitochondrial function, inhibit ferroptosis, and regulate immune responses could represent innovative approaches to prevent disease progression and improve clinical outcomes.

5. Intestinal Barriers

The intestine is one of the largest and most dynamic mucosal surfaces in the human body, serving as a crucial barrier against pathogenic microorganisms and toxic substances. In addition to its role in digestion and nutrient absorption, the intestine acts as the first line of defense against external threats, playing a key role in the interaction between the external environment and the host’s internal microenvironment [94]. The gut–liver axis, mediated by the portal circulation, represents a critical anatomical and functional connection for regulating metabolic and immune flows. This bidirectional relationship allows for dynamic interaction between the intestine and liver, modulating systemic metabolism and immunity [95].
The human intestine is divided into two main sections: the small intestine (comprising the duodenum, jejunum, and ileum) and the large intestine (including the colon, sigmoid colon, and rectum). The morphological and functional structure of these two sections is highly specialized. The small intestine, with its villi and crypts, offers a vast absorptive surface supported by specialized epithelial cells such as enterocytes and enteroendocrine cells, essential for selective molecular transport [96,97]. In contrast, the large intestine, characterized by a flatter surface, maintains barrier integrity through the continuous renewal of epithelial cells generated in the crypts of Lieberkühn, including goblet cells, Paneth cells, and stem cells [98,99]. The intestinal barrier, composed of epithelial cells tightly connected by tight junctions, a mucus layer, and immune components, regulates the interaction between the gut microbiota and the mucosal immune system, playing an integrated role in defending against pathogens and toxins [100]. The selective permeability of this barrier enables the absorption of essential nutrients while limiting the passage of toxins and pathogenic microorganisms. However, environmental factors, gut microbiota dysbiosis, and inflammatory conditions can impair this function, leading to increased intestinal permeability, known as “leaky gut” [94]. This phenomenon facilitates the translocation of pro-inflammatory molecules, such as LPS, into the portal circulation. LPS, a component of Gram-negative bacterial membranes, activates hepatic immune cells, such as Kupffer cells, inducing local and systemic inflammation that contributes to the progression of MASLD into MASH and liver fibrosis [101]. Recent evidence indicates a correlation between plasma LPS levels and the severity of liver disease in patients with MASLD, underscoring the pathogenic role of altered intestinal permeability [102,103]. The liver, receiving 70% of its blood supply from the portal vein, is exposed to the metabolic products of the gut microbiota, such as SCFAs, secondary bile acids, and other microbial metabolites. These compounds modulate hepatic metabolism and immune responses, directly influencing the pathophysiology of liver diseases [95]. The integrated and interdependent system characterizing the gut–liver axis provides a fundamental framework for understanding liver disease pathogenesis, opening new perspectives for developing targeted therapeutic strategies.
The intestine employs a complex system of barriers divided into three main levels: mechanical, immune, and microbial. These barriers work synergistically to preserve intestinal homeostasis, prevent pathogenic invasion, and ensure the selective absorption of nutrients [104]. The mechanical barrier includes the intestinal epithelium, mucus-producing goblet cells, Paneth cells that secrete antimicrobial peptides, and enteric glial cells [105,106,107]. Intestinal epithelial cells (IECs), arranged in a monolayer connected by junctional complexes such as tight junctions, adherens junctions, and desmosomes, ensure the barrier’s selectivity [108]. The mucus layer, particularly developed in the colon, acts as a chemical filter preventing direct contact between microorganisms and the intestinal epithelium. Gel-forming mucins, such as MUC2, are the main structural components of mucus, creating a protective network that harbors the commensal microbiota [109]. The immune barrier consists of innate and adaptive immune cells as well as IECs with immune functions. IECs release cytokines such as IL-10 and antimicrobial peptides to modulate immune homeostasis and prevent excessive bacterial proliferation [110]. Dendritic cells, regulatory T cells (Tregs), and plasma cells in the lamina propria contribute to immune tolerance, partly through the production of secretory immunoglobulin A (SIgA) [111,112].
The gut microbiota, comprising trillions of microorganisms, serves as a biological barrier by competing with pathogens for resources and producing beneficial metabolites, such as SCFAs and secondary bile acids [113,114]. Alterations in microbiota balance, referred to as dysbiosis, are associated with inflammatory, metabolic, and autoimmune diseases, emphasizing the importance of maintaining a healthy microbiota to preserve intestinal barrier integrity [115].

6. Gut Microbiota and Microbiome

Life on Earth has evolved from a common unicellular ancestor, giving rise to the remarkable biological complexity observed today, with sophisticated cellular and systemic organization. Within this context, humans emerge as a superorganism, integrated with trillions of symbiotic bacteria that coexist synergistically with the host’s eukaryotic cells. This dynamic entity, termed the “holobiont”, encompasses both the human genome and the microbial genome, collectively referred to as the “hologenome”. Variation within this system can occur through changes in either the host or microbial genome, maintaining a balance between stability and adaptability. In this context, humans function as a “superorganism”, coexisting with trillions of symbiotic microorganisms that form a dynamic entity known as the “holobiont”. This holobiont integrates both the human genome and the microbial genome, collectively referred to as the “hologenome”. Variations within this system can occur due to changes in either the host or the microbial genome, balancing stability with adaptability [116,117].
Following the completion of the Human Genome Project in 2001, it became evident clear that a comprehensive deeper understanding of human biology requires exploring interactions between the human host organism and its microbiota [118,119,120]. Subsequent initiatives, such as the Human Microbiome Project (HMP) and the Metagenomics of the Human Intestinal Tract (MetaHIT), revolutionized research by focusing on four main microbial colonization sites: the mouth, gut, vagina, and skin. Among these, the gut microbiota has garnered particular interest due to its clinical relevance [121]. The human gut, with a mucosal surface area of approximately 200–300 m2, harbors a diverse community ten trillion symbionts belonging to about 50 bacterial phyla and 100–1000 species of symbiotic microorganisms, encompassing around 50 bacterial phyla and hundreds of species. The term “microbiota” refers to the collection of microorganisms colonizing a specific habitat, such as the gut, and includes bacteria, viruses, fungi, and archaea. The “microbiome”, on the other hand, represents the entire refers to the complete genetic repertoire of these microbial communities and their interactions with the host environment [122]. While interconnected, the microbiota reflects the physical entities of the microorganisms, whereas the microbiome expresses their functional potential. While the microbiota reflects the physical organisms, the microbiome captures their functional potential [123,124].
The analysis study of gut microbiota employs culture-dependent and culture-independent methods. Traditional culture-dependent techniques, first developed by Robert Koch in 1881, involve isolating and characterizing microorganisms but are limited in scope, capturing only 30–50% of the intestinal bacteria due to growth challenges. Advances in enriched culture media and pre-incubation methods have expanded the ability to isolate previously uncultivable species [125]. Furthermore, culturomics, combining extensive cultivation with mass spectrometry and 16S rRNA sequencing, has led to the identification of over 1000 prokaryotic species, including novel ones [126]. Culture-independent methods, initially based on Sanger sequencing and 16S rRNA analysis, have evolved with metagenomic sequencing and next-generation sequencing (NGS) technologies, improving efficiency and reducing costs [127]. More recently, third-generation technologies, such as PacBio and MinION, have enabled rapid detection of epigenetic modifications and pathogens [128].
The human gastrointestinal tract presents distinct microenvironments that exert a profound influence on the composition and distribution of the resident microbiota [129,130,131,132]. These ecological niches vary significantly along the gastrointestinal axis, primarily due to differences in pH, oxygen tension, nutrient availability, and immune surveillance mechanisms. The stomach represents the first critical barrier to microbial colonization. Its highly acidic environment, with a pH typically ranging between 1 and 3, creates an inhospitable setting for most microorganisms. Nevertheless, a limited number of acid-resistant species are capable of colonizing this region. Among them, Streptococcus, Neisseria, and Lactobacillus are the predominant genera identified, belonging mainly to the phyla Firmicutes, Bacteroidetes, and Proteobacteria (Table 2) [129]. Despite the low bacterial density and diversity in the stomach, these microorganisms play pivotal roles in maintaining mucosal homeostasis and contributing to the early phases of digestion. Progressing distally, the small intestine offers a more favorable environment for microbial growth, though the density and diversity of microorganisms vary considerably along its length. In the duodenum, bacterial concentrations are relatively low, ranging from 103 to 104 colony-forming units per milliliter (CFU/mL), primarily composed of Gram-positive aerobic species [130]. As the luminal content moves towards the ileum, bacterial density markedly increases, reaching up to 109 CFU/mL. This transition is accompanied by a compositional shift favoring anaerobic and Gram-negative bacteria, which thrive in the progressively less oxygenated environment [130,131]. The small intestine microbiota plays essential roles in nutrient absorption, the metabolism of bile acids, and the modulation of the mucosal immune system. The large intestine, or colon, constitutes the most densely populated microbial habitat within the human body, harboring an estimated 1012 CFU/mL [132]. It serves as the principal site for the fermentation of indigestible carbohydrates and the synthesis of SCFAs, such as acetate, propionate, and butyrate, which are critical for energy homeostasis and epithelial integrity [132,133]. The colonic microbiota is dominated by species within the Firmicutes and Bacteroidetes phyla, with these taxa playing vital roles in regulating local immune responses and maintaining intestinal barrier function.
The overall composition of the gut microbiota is shaped by a complex interplay of genetic, environmental, and dietary factors. Among the dominant phyla consistently observed in healthy individuals are Firmicutes, Bacteroidetes, Actinobacteria, Proteobacteria, and Verrucomicrobia [134]. These microbial communities contribute to key physiological processes, including energy harvest, immune modulation, and protection against pathogenic invasion. Notably, specific genera such as Clostridium, Lactobacillus, Bacteroides, and Akkermansia muciniphila have been identified as crucial players in maintaining metabolic balance and immune homeostasis [134,135,136].
The gut microbiota represents a highly complex and dynamic ecosystem that performs several critical functions essential for maintaining host health (Table 3). Among its primary roles is the metabolism of nutrients. The microbial community actively ferments dietary fibers, resulting in the production of SCFAs, such as acetate, butyrate, and propionate. These metabolites are fundamental for sustaining energy homeostasis and modulating inflammatory responses at both the intestinal and systemic levels [137]. In addition, the gut microbiota contributes to the biosynthesis of essential vitamins, notably vitamin K and members of the B-group vitamin family, which play pivotal roles in coagulation, cellular metabolism, and neurological function [114]. Another vital function of the gut microbiota is the maintenance of intestinal barrier integrity. By modulating the expression and organization of tight junction proteins and promoting mucin production, the microbiota fortifies the mucosal barrier, thereby preventing the translocation of pathogenic microorganisms and harmful antigens into the systemic circulation [138]. This barrier function is crucial for preserving intestinal homeostasis and preventing inflammatory disorders. Furthermore, the gut microbiota plays an indispensable role in the regulation of immune responses. It orchestrates the development and maturation of the host’s immune system, fostering tolerance to commensal organisms while maintaining vigilance against potential pathogens. Interactions between the microbiota and immune components such as dendritic cells, regulatory T cells, and the production of secretory immunoglobulin A (SIgA) are fundamental to this modulatory activity [136,139]. Beyond its metabolic and immunological functions, the gut microbiota is also responsible for producing a wide range of bioactive metabolites. In addition to SCFAs, microbial metabolism gives rise to compounds such as TMAO and various phenolic derivatives. These metabolites exert significant effects on systemic metabolism, influencing processes that include lipid metabolism, glucose homeostasis, and liver function [140].
The diversity and functional complexity of the gut microbiota underscore its integral role in sustaining both intestinal and systemic health. The intricate interplay between host physiology and microbial activity not only supports gut homeostasis but also has far-reaching implications for overall metabolic regulation. This highlights the gut microbiota as a promising target for therapeutic strategies aimed at preventing and managing a broad spectrum of diseases.

7. Alterations in Intestinal Barriers in MASLD Pathogenesis

MASLD is closely associated with intestinal barrier dysfunction, including tight junction (TJ) alterations, increased intestinal permeability, and dysbiosis. These factors interact in a complex pathological cycle, amplifying inflammation and liver damage.
The intestinal mechanical barrier serves as the first line of defense against pathogens, and its impairment is a critical element in MASLD pathogenesis. Disruption of the mucus layer and protective chemical compounds compromise intestinal homeostasis, favoring bacterial overgrowth and increasing intestinal permeability [141]. Recent studies have demonstrated that epithelial barrier dysfunction in patients with MASH and animal models is linked to chronic mucosal inflammation. Under these conditions, tight junction proteins undergo contraction and cytoplasmic dislocation, which heightens mucosal permeability, allowing for the translocation of bacteria and microbial products, such as LPS [141,142]. Once in the portal circulation, LPS reaches the liver and stimulates Kupffer cells, triggering the release of pro-inflammatory cytokines that drive liver injury [143].
The gut vascular barrier (GVB) plays a crucial role in preventing bacterial dissemination. In individuals with MASLD, increased expression of PV-1, a GVB permeability marker, reflects compromised integrity. Studies have shown that during MASH progression, enteric pathogens interfere with the WNT/β-catenin pathway in endothelial cells, exacerbating liver damage [144,145]. The intestinal immunological barrier, primarily composed of SIgA, lymphocytes, and plasma cells, plays a key role in modulating mucosal immunity. SIgA helps neutralize pathogens by specifically binding to Gram-negative bacteria. However, in patients with MASLD, immunological barrier functionality is compromised, promoting bacterial translocation and inflammation [146,147].
The gut–liver axis is central to MASLD pathogenesis. Damage-associated molecular patterns (DAMPs) and microbe-associated molecular patterns (MAMPs) activate sterile inflammatory responses in the liver, contributing to hepatic damage [148]. Elevated serum IgA levels have been observed in patients with MASLD, correlating with advanced fibrosis and increased activation of plasma cells in secondary lymphoid organs [149,150]. In murine models, hepatic B lymphocytes are stimulated by intestinal microbial factors which exacerbate inflammation and liver fibrosis [151]. Similarly, patients with MASLD exhibit reduced regulatory T cells (Tregs) expressing FOXP3 and increased Th1 and CD8+ T cells in the intestinal lamina propria [15]. Mast cells (MCs), which are present in the intestinal barrier, also contribute to dysfunction. These cells release cytokines, histamine, and proteases, including trypsin and chymase, that degrade ZO-1 protein, reduce JAM-A expression, and compromise epithelial integrity [152,153]. Histamine, in particular, has been shown to increase intestinal permeability and bacterial translocation, aggravating inflammation [153,154].
The intestinal microbial barrier is essential for maintaining microbiota homeostasis and preventing pathogenic colonization. However, a Western diet (WD), rich in saturated fats, cholesterol, and refined carbohydrates, is associated with dysbiosis and reduced populations of commensal bacteria, favoring the proliferation of pro-inflammatory species such as Gram-negative bacteria [155,156]. These changes increase intestinal permeability and activate Toll-like receptor (TLR) family signaling, promoting LPS-mediated inflammation and endotoxin activity [157,158]. Obesity and hypercaloric diets also induce dysbiosis and metabolic alterations, characterized by reduced microbial diversity and a lower Firmicutes/Bacteroidetes ratio in individuals with obesity compared to lean controls [159,160]. In patients with MASLD, the intestinal microbiome exhibits an altered “signature”, characterized by increased Proteobacteria, Enterobacteria, Escherichia, and Bacteroides species, alongside reduced Firmicutes species [160,161]. These microbial shifts not only compromise the intestinal barrier but also exacerbate the gut–liver inflammatory cycle.
The alterations in intestinal barriers (Table 4)—mechanical, immunological, and microbial—play a pivotal role in MASLD pathogenesis, contributing to chronic inflammation and liver damage. While the underlying mechanisms are complex and interdependent, understanding these dynamics is essential for developing targeted therapeutic interventions aimed at restoring intestinal homeostasis and modulating inflammatory responses associated with MASLD.

8. Gut Dysbiosis in the Pathogenesis of MASLD

Gut dysbiosis is a pivotal factor in the pathogenesis of MASLD, influencing key processes such as hepatic metabolism, intestinal barrier integrity, and the activation of systemic inflammatory signaling. The gut microbiota plays a crucial role in maintaining metabolic homeostasis, but alterations in its composition and function can increase intestinal permeability. This allows bacteria and microbial products, such as LPS, to enter the portal circulation, activating Kupffer cells via TLRs. This cascade triggers an inflammatory response that exacerbates liver damage [162,163].
Diet is one of the main drivers of gut microbial dysbiosis. A Western diet (WD), rich in refined sugars and saturated fats, reduces microbial diversity and promotes the overgrowth of Gram-negative bacteria such as Bacteroidetes, while decreasing beneficial bacteria such as Prevotella [151,164]. This microbial imbalance disrupts the production of SCFAs, which play vital roles in regulating gut barrier integrity and inflammation. While butyrate supports intestinal health and reduces inflammation, acetate, the most abundant SCFAs, has obesogenic effects and contributes to hepatic lipid accumulation [165]. In addition, diets high in saturated fats and taurine-conjugated bile salts promote the production of hydrogen sulfide (H₂S), a toxic molecule that further damages the intestinal barrier and exacerbates inflammation, accelerating MASLD progression [166].
The gut microbiota also influences lipid metabolism through the activation of lipoprotein lipase (LPL) and modulation of AMP-activated protein kinase (AMPK), which regulates lipid oxidation and metabolic balance. Furthermore, microbial communities chemically modify bile acids, affecting hepatic glucose and lipid metabolism [167].
As gut dysbiosis compromises the intestinal barrier, bacterial translocation is facilitated, allowing bacterial components and endotoxins to enter the portal circulation. These elements reach the liver, where they activate Kupffer cells via TLRs, amplifying the inflammatory response and promoting liver damage [168]. Upon arrival in the liver, gut-derived endotoxins, primarily LPS, bind to TLR4 on Kupffer cells, initiating a MyD88-dependent signaling cascade [167,168]. This cascade activates nuclear factor-κB, leading to the transcription of numerous pro-inflammatory cytokines such as TNF-α and pro-IL-1β [167,168]. Importantly, TLR activation also primes the assembly of the NLRP3 inflammasome, a multiprotein complex that serves as a critical mediator of inflammation [167,168]. A secondary signal, often in the form of ROS or other metabolic stressors, is then required to fully activate the inflammasome [167,168]. Once activated, caspase-1 cleaves pro-IL-1β and pro-IL-18 into their active forms, IL-1β and IL-18, which further amplify hepatic inflammation and contribute to the progression of liver injury in MASLD [167,168]. This TLR–inflammasome axis thus provides a crucial mechanistic link between gut barrier dysfunction, microbial dysbiosis, and the chronic inflammatory state observed in MASLD. Moreover, microbial fermentation of carbohydrates generates endogenous ethanol, which is transported to the liver via the portal vein. This ethanol exacerbates oxidative stress and mitochondrial dysfunction, worsening hepatic steatosis. Elevated levels of alcohol-producing bacteria have been found in individuals with MASLD, highlighting the significance of this pathogenic pathway [169,170].
Patients with MASLD exhibit reduced microbial diversity, with a marked increase in Gram-negative bacteria and a decrease in beneficial species such as Prevotella. Recent studies have suggested that targeted dietary interventions, such as supplementation with oat beta-glucan and alanyl-glutamine dipeptides, can restore microbial balance and improve the pathological features of MASLD [171]. Gut dysbiosis also impacts immune responses, compromising the intestinal barrier and facilitating bacterial translocation. This contributes to chronic systemic inflammation, driven by cytokines like TNF-α, which further exacerbates IR and liver damage [105,171] Additionally, reduced secretion of IgA worsens intestinal permeability and mucosal dysfunction [172].
The gut–liver axis—characterized by interactions between the intestinal epithelial, vascular, and immune barriers and the hepatic system—plays a central role in MASLD. Dysregulation of this axis, driven by microbial dysbiosis and increased intestinal permeability, promotes TLR activation and chronic hepatic inflammation [173,174]. Changes in the Firmicutes/Bacteroidetes ratio, reductions in beneficial species like Akkermansia muciniphila and Faecalibacterium prausnitzii, and the accumulation of pro-inflammatory metabolites such as TMAO and H2S are strongly associated with MASLD progression [39,175,176]. Gut dysbiosis is thus a critical contributor to MASLD pathogenesis, influencing inflammatory, metabolic, and immune responses (Figure 1). Therapeutic strategies aimed at restoring microbial balance—such as probiotics, prebiotics, and dietary interventions—hold promise for mitigating disease progression. However, further research is needed to develop personalized interventions that fully leverage the potential of the gut microbiota in managing MASLD.

9. Dietary Approach and Gut Microbiota Modulation in the Management of MASLD: Evidence and Perspectives

9.1. Dietary Interventions in MASLD

A weight loss of 7–10% from the initial body weight is recommended to achieve a negative energy balance and improve hepatic outcomes. This objective can be attained through a hypocaloric diet, with an energy deficit of 500–1000 kcal per day, combined with a structured exercise program [177,178]. Although the role of ultra-processed foods and alcohol in MASLD remains debated, emerging evidence favors the Mediterranean diet (MD). Its nutritional profile—rich in dietary fiber, monounsaturated and polyunsaturated fatty acids, and bioactive compounds with anti-inflammatory and antioxidant properties—has demonstrated efficacy in reducing hepatic steatosis and improving metabolic parameters, even in patients with type 2 diabetes mellitus (T2DM) [3,179,180]. Dietary modifications have a profound impact on the gut microbiota. Chronic consumption of a high-fat diet in murine models has been shown to increase the Firmicutes/Bacteroidetes ratio, a feature associated with enhanced metabolic efficiency and hepatic lipid accumulation [181]. Conversely, a fiber-rich diet exerts protective effects by attenuating hepatic inflammation and promoting the proliferation of beneficial bacterial species such as Akkermansia muciniphila [182]. Moderate coffee intake has also been associated with a protective role against MASLD and its progression to MASH and fibrosis. The proposed mechanisms include enhanced glutathione synthesis, reduced oxidative stress via the scavenging of reactive oxygen species (ROS), and modulation of gut microbiota composition, specifically, the restoration of the Firmicutes/Bacteroidetes ratio and promotion of probiotic species such as Bifidobacterium [183,184,185].

9.2. Role of Exercise in Modulating the Gut Microbiota

Exercise is a well-established non-pharmacological intervention that exerts beneficial effects on metabolic outcomes and modulates the gut microbiota. Studies conducted in athletes and healthy individuals have demonstrated that regular physical activity increases microbial diversity, with a higher relative abundance of Akkermansia muciniphila observed in lean individuals compared to those with elevated body mass index (BMI) [186]. In animal models, exercise has been shown to reduce certain microbial taxa (e.g., Lactobacillaceae, Proteobacteria, Bacteroidetes, Flavobacterium, Alkaliphilus), decrease the Firmicutes/Bacteroidetes ratio, and increase populations within the phylum Verrucomicrobia and the family Turicibacteraceae [187]. When combined with a high-fat diet, exercise provides benefits beyond caloric restriction alone, improving insulin sensitivity, lowering LDL cholesterol levels, and reducing hepatic fat accumulation and serum triglycerides [188].

9.3. Antibiotics and Gut Microbiota Modulation in MASLD

Antibiotics have been explored as a therapeutic option to modulate the gut microbiota in MASLD, although the evidence remains inconclusive. Preclinical studies have demonstrated that norfloxacin and neomycin can improve hepatic function by altering microbiota composition and reducing bacterial translocation; however, a randomized controlled trial (RCT) in obese patients with MASLD did not demonstrate significant metabolic benefits [189]. In contrast, the combination of metronidazole and inulin has been associated with significant reductions in serum alanine aminotransferase (ALT) levels compared to placebo [190]. Rifaximin, one of the most extensively studied non-absorbable antibiotics, has been shown to have modest effects on fecal bile acid profiles in patients with cirrhosis [191]. More recently, an RCT demonstrated that six months of rifaximin therapy (1100 mg/day) resulted in significant reductions in serum ALT (from 64.6 ± 34.2 to 38.2 ± 29.2; p = 0.01) and aspartate aminotransferase (AST) levels (from 66.5 ± 42.5 to 41.8 ± 30.4; p = 0.02), as well as improvements in insulin resistance, pro-inflammatory cytokines, endotoxemia, cytokeratin-18 (CK-18), and the NAFLD steatosis score [192]. Nonetheless, the effects of rifaximin in MASLD remain variable: while some studies have reported improvements in ALT levels, reductions in endotoxins, and lower pro-inflammatory cytokine concentrations [193], others have not observed significant benefits [194].

9.4. Probiotics, Prebiotics, and Synbiotics in MASLD Management

Probiotics, prebiotics, and synbiotics are emerging as promising adjunctive therapies in MASLD. Probiotics are live microorganisms that confer health benefits by reducing hepatic inflammation, oxidative stress, and fat accumulation. Prebiotics, which are non-digestible food components, selectively stimulate the growth and/or activity of beneficial gut bacteria. Synbiotics combine both approaches, enhancing probiotic survival and efficacy. Although not curative, these interventions may complement standard MASLD management strategies [195]. For instance, a randomized controlled trial demonstrated that six months of probiotic supplementation significantly improved the AST-to-platelet ratio index in patients with non-alcoholic steatohepatitis (NASH) [195]. Another study found that 12 weeks of probiotic treatment reduced hepatic inflammation, although histological evidence remains limited [196]. Conversely, an RCT showed that 24 weeks of probiotic supplementation did not yield additional cardiovascular benefits over placebo in patients with MASH [197]. Studies evaluating prebiotics have shown mixed results. Oligofructose supplementation (16 g/day for eight weeks) significantly reduced AST levels but had no effect on serum triglycerides [23]. A systematic review highlighted methodological limitations across studies, reducing the strength of evidence supporting prebiotic use in MASLD management [198]. A recent systematic review suggests that probiotics and prebiotics may improve metabolic and inflammatory parameters, including BMI, cholesterol, triglycerides, HOMA-IR, transaminases, gamma-glutamyl transferase (GGT), and pro-inflammatory cytokines [199]. Synbiotics, in particular, have demonstrated beneficial effects on BMI, AST, ALT, GGT, and the NAFLD fibrosis score, as confirmed by a network meta-analysis of 26 RCTs [200]. Additional research has reported improvements in inflammatory markers (e.g., TNF-α and C-reactive protein) and liver histological parameters following synbiotic treatment. Specifically, 24 weeks of synbiotic supplementation with Bifidobacterium longum and fructo-oligosaccharides led to significant reductions in hepatic inflammation and histological activity in patients with NASH [201]. Another study observed reduced steatosis on ultrasound after eight weeks of synbiotic therapy, although ALT, AST, and CRP levels remained unchanged [202]. Moreover, a study in lean MASLD demonstrated that 28 weeks of synbiotic supplementation significantly reduced hepatic fibrosis, steatosis, and inflammatory markers [203].

9.5. Fecal Microbiota Transplantation in MASLD

Fecal microbiota transplantation (FMT) is an emerging therapeutic strategy that involves transferring fecal material from a healthy donor to a recipient, with the goal of restoring intestinal eubiosis. FMT specifically targets the gut–liver axis by improving intestinal barrier integrity and reducing systemic inflammation [204,205,206]. In clinical settings, a randomized controlled trial (RCT) demonstrated that a single allogenic FMT infusion improved small intestinal permeability—as assessed by the lactulose/mannitol test—in patients with MASLD, although it did not significantly impact hepatic steatosis or insulin resistance [207]. Conversely, a pilot study administering FMT over three consecutive days reported a modest but statistically significant reduction in hepatic steatosis [208]. Long-term follow-up studies in patients with cirrhosis have shown sustained clinical and cognitive improvements, including reductions in hepatic encephalopathy episodes and hospitalizations [209]. Donor characteristics are critical determinants of FMT efficacy. Evidence suggests that FMT from lean donors improves insulin sensitivity in individuals with metabolic syndrome, whereas FMT from obese donors may transiently impair insulin sensitivity [210,211]. FMT can be administered via oral capsules, nasogastric tubes, enemas, or colonoscopy, with the latter often considered the most effective method for ensuring microbiota engraftment [212,213]. Notably, oral-capsule-based FMT has demonstrated safety and efficacy in modulating gut microbiota composition and reducing circulating levels of taurocholate, a bile acid implicated in hepatic injury and fibrosis [214,215,216].
Targeting gut microbiota through dietary interventions, exercise, antibiotics, probiotics, synbiotics, and fecal microbiota transplantation represents a promising avenue for MASLD management (Table 5). While emerging evidence supports their metabolic and hepatic benefits, further research is needed to define optimal therapeutic strategies and long-term outcomes.

10. Conclusions

In recent years, the gut–liver axis has emerged as a central player in MASLD pathogenesis, where alterations in intestinal barriers increase permeability, promote bacterial translocation, and trigger hepatic inflammation and fibrosis. Dysbiosis, characterized by a rise in pro-inflammatory bacteria and harmful metabolites (e.g., LPS, TMAO), exacerbates metabolic dysfunction and liver injury through TLR activation and NLRP3 inflammasome assembly.
Lifestyle modifications, particularly a Mediterranean diet, physical activity, and weight loss, remain the cornerstone of MASLD management. Targeted microbiota-based interventions (probiotics, prebiotics, synbiotics, FMT) show promise, but further large-scale RCTs are needed to confirm their efficacy.
Advances in multi-omics technologies (metagenomics, metabolomics, proteomics) offer new insights into microbiota–host interactions, facilitating biomarker discovery and personalized therapies [217,218]. Integrating clinical, genetic, and microbiome data may revolutionize MASLD management, paving the way for precision medicine. The gut microbiota is an intricate determinant of liver health, and its modulation represents both a scientific challenge and a therapeutic opportunity in MASLD.

Author Contributions

Conceptualization, C.A.; writing—original draft preparation, C.A.; writing—review and editing, R.N., L.R., F.C.S., L.E.A. and A.C.; supervision A.C. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Institutional Review Board Statement

The authors have reviewed the literature data and have reported results coming from studies approved by local ethics committee.

Informed Consent Statement

Not applicable.

Data Availability Statement

No new data were created or analyzed in this study. Data sharing is not applicable to this article.

Conflicts of Interest

The authors declare no conflicts of interest.

References

  1. 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] [PubMed]
  2. Fahed, G.; Aoun, L.; Bou Zerdan, M.; Allam, S.; Bou Zerdan, M.; Bouferraa, Y.; Assi, H.I. Metabolic Syndrome: Updates on Pathophysiology and Management in 2021. Int. J. Mol. Sci. 2022, 23, 786. [Google Scholar] [CrossRef] [PubMed]
  3. Chalasani, N.; Younossi, Z.; Lavine, J.E.; Charlton, M.; Cusi, K.; Rinella, M.; Harrison, S.A.; Brunt, E.M.; Sanyal, A.J. The Diagnosis and Management of Nonalcoholic Fatty Liver Disease: Practice Guidance from the American Association for the Study of Liver Diseases. Hepatology 2018, 67, 328–357. [Google Scholar] [CrossRef] [PubMed]
  4. 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]
  5. 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]
  6. Targher, G.; Corey, K.E.; Byrne, C.D. NAFLD, and Cardiovascular and Cardiac Diseases: Factors Influencing Risk, Prediction and Treatment. Diabetes Metab. 2021, 47, 101215. [Google Scholar] [CrossRef]
  7. Adams, L.A.; Anstee, Q.M.; Tilg, H.; Targher, G. Non-Alcoholic Fatty Liver Disease and Its Relationship with Cardiovascular Disease and Other Extrahepatic Diseases. Gut 2017, 66, 1138–1153. [Google Scholar] [CrossRef]
  8. Li, M.; Chen, W.; Deng, Y.; Xie, W. Impacts of Cardiometabolic Risk Factors and Alcohol Consumption on All-Cause Mortality among MASLD and Its Subgroups. Nutr. Metab. Cardiovasc. Dis. 2024, 34, 2085–2094. [Google Scholar] [CrossRef]
  9. Vetrano, E.; Rinaldi, L.; Mormone, A.; Giorgione, C.; Galiero, R.; Caturano, A.; Nevola, R.; Marfella, R.; Sasso, F.C. Non-Alcoholic Fatty Liver Disease (NAFLD), Type 2 Diabetes, and Non-Viral Hepatocarcinoma: Pathophysiological Mechanisms and New Therapeutic Strategies. Biomedicines 2023, 11, 468. [Google Scholar] [CrossRef]
  10. 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]
  11. Byrne, C.D.; Targher, G. NAFLD: A Multisystem Disease. J. Hepatol. 2015, 62, S47–S64. [Google Scholar] [CrossRef] [PubMed]
  12. Salvatore, T.; Galiero, R.; Caturano, A.; Rinaldi, L.; Criscuolo, L.; Di Martino, A.; Albanese, G.; Vetrano, E.; Catalini, C.; Sardu, C.; et al. Current Knowledge on the Pathophysiology of Lean/Normal-Weight Type 2 Diabetes. Int. J. Mol. Sci. 2022, 24, 658. [Google Scholar] [CrossRef] [PubMed]
  13. Tilg, H.; Adolph, T.E.; Dudek, M.; Knolle, P. Non-Alcoholic Fatty Liver Disease: The Interplay Between Metabolism, Microbes and Immunity. Nat. Metab. 2021, 3, 1596–1607. [Google Scholar] [CrossRef] [PubMed]
  14. Schnabl, B.; Brenner, D.A. Interactions Between the Intestinal Microbiome and Liver Diseases. Gastroenterology 2014, 146, 1513–1524. [Google Scholar] [CrossRef]
  15. 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]
  16. Sharpton, S.R.; Schnabl, B.; Knight, R.; Loomba, R. Current Concepts, Opportunities, and Challenges of Gut Microbiome-Based Personalized Medicine in Nonalcoholic Fatty Liver Disease. Cell Metab. 2021, 33, 21–32. [Google Scholar] [CrossRef]
  17. Miele, L.; Marrone, G.; Lauritano, C.; Cefalo, C.; Gasbarrini, A.; Day, C.; Grieco, A. Gut-Liver Axis and Microbiota in NAFLD: Insight Pathophysiology for Novel Therapeutic Target. Curr. Pharm. Des. 2013, 19, 5314–5324. [Google Scholar] [CrossRef]
  18. Aron-Wisnewsky, J.; Gaborit, B.; Dutour, A.; Clement, K. Gut Microbiota and Non-Alcoholic Fatty Liver Disease: New Insights. Clin. Microbiol. Infect. 2013, 19, 338–348. [Google Scholar] [CrossRef]
  19. Miele, L.; Valenza, V.; La Torre, G.; Montalto, M.; Cammarota, G.; Ricci, R.; Mascianà, R.; Forgione, A.; Gabrieli, M.L.; Perotti, G.; et al. Increased Intestinal Permeability and Tight Junction Alterations in Nonalcoholic Fatty Liver Disease. Hepatology 2009, 49, 1877–1887. [Google Scholar] [CrossRef]
  20. Zampino, R.; Marrone, A.; Rinaldi, L.; Guerrera, B.; Nevola, R.; Boemio, A.; Iuliano, N.; Giordano, M.; Passariello, N.; Sasso, F.C.; et al. Endotoxinemia Contributes to Steatosis, Insulin Resistance and Atherosclerosis in Chronic Hepatitis C: The Role of Pro-Inflammatory Cytokines and Oxidative Stress. Infection 2018, 46, 793–799. [Google Scholar] [CrossRef]
  21. Borrelli, A.; Bonelli, P.; Tuccillo, F.M.; Goldfine, I.D.; Evans, J.L.; Buonaguro, F.M.; Mancini, A. Role of Gut Microbiota and Oxidative Stress in the Progression of Non-Alcoholic Fatty Liver Disease to Hepatocarcinoma: Current and Innovative Therapeutic Approaches. Redox Biol. 2018, 15, 467–479. [Google Scholar] [CrossRef] [PubMed]
  22. Lombardi, R.; Petta, S.; Pisano, G.; Dongiovanni, P.; Rinaldi, L.; Adinolfi, L.E.; Acierno, C.; Valenti, L.; Boemi, R.; Spatola, F.; et al. FibroScan Identifies Patients with Nonalcoholic Fatty Liver Disease and Cardiovascular Damage. Clin. Gastroenterol. Hepatol. 2020, 18, 517–519. [Google Scholar] [CrossRef] [PubMed]
  23. Wang, Z.; Roberts, A.B.; Buffa, J.A.; Levison, B.S.; Zhu, W.; Org, E.; Gu, X.; Huang, Y.; Zamanian-Daryoush, M.; Culley, M.K.; et al. Non-Lethal Inhibition of Gut Microbial Trimethylamine Production for the Treatment of Atherosclerosis. Cell 2015, 163, 1585–1595. [Google Scholar] [CrossRef] [PubMed]
  24. Martin-Grau, M.; Monleón, D. The Role of Microbiota-Related Co-Metabolites in MASLD Progression: A Narrative Review. Curr. Issues Mol. Biol. 2024, 46, 6377–6389. [Google Scholar] [CrossRef]
  25. Trépo, E.; Valenti, L. Update on NAFLD Genetics: From New Variants to the Clinic. J. Hepatol. 2020, 72, 1196–1209. [Google Scholar] [CrossRef]
  26. Targher, G.; Corey, K.E.; Byrne, C.D.; Roden, M. The Complex Link between NAFLD and Type 2 Diabetes Mellitus—Mechanisms and Treatments. Nat. Rev. Gastroenterol. Hepatol. 2021, 18, 599–612. [Google Scholar] [CrossRef]
  27. Sookoian, S. PNPLA3, the Triacylglycerol Synthesis/Hydrolysis/Storage Dilemma, and Nonalcoholic Fatty Liver Disease. World J. Gastroenterol. 2012, 18, 6018. [Google Scholar] [CrossRef]
  28. Pingitore, P.; Romeo, S. The Role of PNPLA3 in Health and Disease. Biochim. Biophys. Acta BBA—Mol. Cell Biol. Lipids 2019, 1864, 900–906. [Google Scholar] [CrossRef]
  29. Qadri, S.; Lallukka-Brück, S.; Luukkonen, P.K.; Zhou, Y.; Gastaldelli, A.; Orho-Melander, M.; Sammalkorpi, H.; Juuti, A.; Penttilä, A.K.; Perttilä, J.; et al. The PNPLA3-I148M Variant Increases Polyunsaturated Triglycerides in Human Adipose Tissue. Liver Int. 2020, 40, 2128–2138. [Google Scholar] [CrossRef]
  30. Wang, Y.; Kory, N.; BasuRay, S.; Cohen, J.C.; Hobbs, H.H. PNPLA3, CGI-58, and Inhibition of Hepatic Triglyceride Hydrolysis in Mice. Hepatology 2019, 69, 2427–2441. [Google Scholar] [CrossRef]
  31. Liu, Y.L.; Patman, G.L.; Leathart, J.B.S.; Piguet, A.C.; Burt, A.D.; Dufour, J.F.; Day, C.P.; Daly, A.K.; Reeves, H.L.; Anstee, Q.M. Carriage of the PNPLA3 rs738409 C>G Polymorphism Confers an Increased Risk of Non-Alcoholic Fatty Liver Disease-Associated Hepatocellular Carcinoma. J. Hepatol. 2014, 61, 75–81. [Google Scholar] [CrossRef] [PubMed]
  32. Romeo, S.; Kozlitina, J.; Xing, C.; Pertsemlidis, A.; Cox, D.; Pennacchio, L.A.; Boerwinkle, E.; Cohen, J.C.; Hobbs, H.H. Genetic Variation in PNPLA3 Confers Susceptibility to Nonalcoholic Fatty Liver Disease. Nat. Genet. 2008, 40, 1461–1465. [Google Scholar] [CrossRef] [PubMed]
  33. Sharma, D.; Mandal, P. NAFLD: Genetics and Its Clinical Implications. Clin. Res. Hepatol. Gastroenterol. 2022, 46, 102003. [Google Scholar] [CrossRef] [PubMed]
  34. Newberry, E.P.; Hall, Z.; Xie, Y.; Molitor, E.A.; Bayguinov, P.O.; Strout, G.W.; Fitzpatrick, J.A.J.; Brunt, E.M.; Griffin, J.L.; Davidson, N.O. Liver-Specific Deletion of Mouse Tm6sf2 Promotes Steatosis, Fibrosis, and Hepatocellular Cancer. Hepatology 2021, 74, 1203–1219. [Google Scholar] [CrossRef]
  35. O’Hare, E.A.; Yang, R.; Yerges-Armstrong, L.M.; Sreenivasan, U.; McFarland, R.; Leitch, C.C.; Wilson, M.H.; Narina, S.; Gorden, A.; Ryan, K.A.; et al. TM6SF2 rs58542926 Impacts Lipid Processing in Liver and Small Intestine. Hepatology 2017, 65, 1526–1542. [Google Scholar] [CrossRef]
  36. Liu, Y.L.; Reeves, H.L.; Burt, A.D.; Tiniakos, D.; McPherson, S.; Leathart, J.B.S.; Allison, M.E.; Alexander, G.J.; Piguet, A.C.; Anty, R.; et al. TM6SF2 rs58542926 Influences Hepatic Fibrosis Progression in Patients with Non-Alcoholic Fatty Liver Disease. Nat. Commun. 2014, 5, 4309. [Google Scholar] [CrossRef]
  37. Peter, A.; Stefan, N.; Cegan, A.; Walenta, M.; Wagner, S.; Königsrainer, A.; Machicao, F.; Schick, F.; Häring, H.U.; Schleicher, E. Hepatic Glucokinase Expression Is Associated with Lipogenesis and Fatty Liver in Humans. J. Clin. Endocrinol. Metab. 2011, 96, E1126–E1130. [Google Scholar] [CrossRef]
  38. Meroni, M.; Longo, M.; Tria, G.; Dongiovanni, P. Genetics Is of the Essence to Face NAFLD. Biomedicines 2021, 9, 1359. [Google Scholar] [CrossRef]
  39. Santoro, N.; Zhang, C.K.; Zhao, H.; Pakstis, A.J.; Kim, G.; Kursawe, R.; Dykas, D.J.; Bale, A.E.; Giannini, C.; Pierpont, B.; et al. Variant in the Glucokinase Regulatory Protein (GCKR) Gene Is Associated with Fatty Liver in Obese Children and Adolescents. Hepatology 2012, 55, 781–789. [Google Scholar] [CrossRef]
  40. Ma, Y.; Belyaeva, O.V.; Brown, P.M.; Fujita, K.; Valles, K.; Karki, S.; de Boer, Y.S.; Koh, C.; Chen, Y.; Du, X.; et al. 17-Beta Hydroxysteroid Dehydrogenase 13 Is a Hepatic Retinol Dehydrogenase Associated with Histological Features of Nonalcoholic Fatty Liver Disease. Hepatology 2019, 69, 1504–1519. [Google Scholar] [CrossRef]
  41. Abul-Husn, N.S.; Cheng, X.; Li, A.H.; Xin, Y.; Schurmann, C.; Stevis, P.; Liu, Y.; Kozlitina, J.; Stender, S.; Wood, G.C.; et al. A Protein-Truncating HSD17B13 Variant and Protection from Chronic Liver Disease. N. Engl. J. Med. 2018, 378, 1096–1106. [Google Scholar] [CrossRef] [PubMed]
  42. Berger, S.L.; Kouzarides, T.; Shiekhattar, R.; Shilatifard, A. An Operational Definition of Epigenetics. Genes Dev. 2009, 23, 781–783. [Google Scholar] [CrossRef] [PubMed]
  43. Pan, Y.; Chen, H.; Zhang, X.; Liu, W.; Ding, Y.; Huang, D.; Zhai, J.; Wei, W.; Wen, J.; Chen, D.; et al. METTL3 Drives NAFLD-Related Hepatocellular Carcinoma and Is a Therapeutic Target for Boosting Immunotherapy. Cell Rep. Med. 2023, 4, 101144. [Google Scholar] [CrossRef] [PubMed]
  44. Wang, L.; Zhu, L.; Liang, C.; Huang, X.; Liu, Z.; Huo, J.; Zhang, Y.; Zhang, Y.; Chen, L.; Xu, H.; et al. Targeting N6-Methyladenosine Reader YTHDF1 with siRNA Boosts Antitumor Immunity in NASH-HCC by Inhibiting EZH2-IL-6 Axis. J. Hepatol. 2023, 79, 1185–1200. [Google Scholar] [CrossRef]
  45. Chan, W.K.; Chuah, K.H.; Rajaram, R.B.; Lim, L.L.; Ratnasingam, J.; Vethakkan, S.R. Metabolic Dysfunction-Associated Steatotic Liver Disease (MASLD): A State-of-the-Art Review. J. Obes. Metab. Syndr. 2023, 32, 197–213. [Google Scholar] [CrossRef]
  46. 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. [Google Scholar] [CrossRef]
  47. Castro, A.V.B.; Kolka, C.M.; Kim, S.P.; Bergman, R.N. Obesity, Insulin Resistance and Comorbidities: Mechanisms of Association. Arq. Bras. Endocrinol. Metabol. 2014, 58, 600–609. [Google Scholar] [CrossRef]
  48. Rampanelli, E.; Ochodnicky, P.; Vissers, J.P.; Butter, L.M.; Claessen, N.; Calcagni, A.; Kors, L.; Gethings, L.A.; Bakker, S.J.; de Borst, M.H.; et al. Excessive Dietary Lipid Intake Provokes an Acquired Form of Lysosomal Lipid Storage Disease in the Kidney. J. Pathol. 2018, 246, 470–484. [Google Scholar] [CrossRef]
  49. Ye, J. Mechanisms of Insulin Resistance in Obesity. Front. Med. 2013, 7, 14–24. [Google Scholar] [CrossRef]
  50. 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]
  51. Hamdy, O. The Role of Adipose Tissue as an Endocrine Gland. Curr. Diab. Rep. 2005, 5, 317–319. [Google Scholar] [CrossRef] [PubMed]
  52. Virtue, S.; Vidal-Puig, A. Adipose Tissue Expandability, Lipotoxicity and the Metabolic Syndrome—An Allostatic Perspective. Biochim. Biophys. Acta BBA—Mol. Cell Biol. Lipids 2010, 1801, 338–349. [Google Scholar] [CrossRef] [PubMed]
  53. Yaskolka Meir, A.; Tene, L.; Cohen, N.; Shelef, I.; Schwarzfuchs, D.; Gepner, Y.; Zelicha, H.; Rein, M.; Bril, N.; Serfaty, D.; et al. Intrahepatic Fat, Abdominal Adipose Tissues, and Metabolic State: Magnetic Resonance Imaging Study. Diabetes Metab. Res. Rev. 2017, 33, e2888. [Google Scholar] [CrossRef] [PubMed]
  54. Rayner, J.J.; Banerjee, R.; Holloway, C.J.; Lewis, A.J.M.; Peterzan, M.A.; Francis, J.M.; Neubauer, S.; Rider, O.J. The Relative Contribution of Metabolic and Structural Abnormalities to Diastolic Dysfunction in Obesity. Int. J. Obes. 2018, 42, 441–447. [Google Scholar] [CrossRef]
  55. Murakami, T.; Michelagnoli, S.; Longhi, R.; Gianfranceschi, G.; Pazzucconi, F.; Calabresi, L.; Sirtori, C.R.; Franceschini, G. Triglycerides Are Major Determinants of Cholesterol Esterification/Transfer and HDL Remodeling in Human Plasma. Arterioscler. Thromb. Vasc. Biol. 1995, 15, 1819–1828. [Google Scholar] [CrossRef]
  56. Shao, B.; Heinecke, J.W. Quantifying HDL Proteins by Mass Spectrometry: How Many Proteins Are There and What Are Their Functions? Expert Rev. Proteom. 2018, 15, 31–40. [Google Scholar] [CrossRef]
  57. Cohen, D.; Fisher, E. Lipoprotein Metabolism, Dyslipidemia, and Nonalcoholic Fatty Liver Disease. Semin. Liver Dis. 2013, 33, 380–388. [Google Scholar]
  58. Caturano, A.; Galiero, R.; Loffredo, G.; Vetrano, E.; Medicamento, G.; Acierno, C.; Rinaldi, L.; Marrone, A.; Salvatore, T.; Monda, M.; et al. Effects of a Combination of Empagliflozin Plus Metformin vs. Metformin Monotherapy on NAFLD Progression in Type 2 Diabetes: The IMAGIN Pilot Study. Biomedicines 2023, 11, 322. [Google Scholar] [CrossRef]
  59. Stehouwer, C.D.A.; Henry, R.M.A.; Ferreira, I. Arterial Stiffness in Diabetes and the Metabolic Syndrome: A Pathway to Cardiovascular Disease. Diabetologia 2008, 51, 527–539. [Google Scholar] [CrossRef]
  60. Marušić, M.; Paić, M.; Knobloch, M.; Liberati Pršo, A.M. NAFLD, Insulin Resistance, and Diabetes Mellitus Type 2. Can. J. Gastroenterol. Hepatol. 2021, 2021, 6613827. [Google Scholar] [CrossRef]
  61. Safar, M.E.; Asmar, R.; Benetos, A.; Blacher, J.; Boutouyrie, P.; Lacolley, P.; Laurent, S.; London, G.; Pannier, B.; Protogerou, A.; et al. Interaction Between Hypertension and Arterial Stiffness: An Expert Reappraisal. Hypertension 2018, 72, 796–805. [Google Scholar] [CrossRef] [PubMed]
  62. Rizzoni, D.; Porteri, E.; Guelfi, D.; Muiesan, M.L.; Valentini, U.; Cimino, A.; Girelli, A.; Rodella, L.; Bianchi, R.; Sleiman, I.; et al. Structural Alterations in Subcutaneous Small Arteries of Normotensive and Hypertensive Patients With Non–Insulin-Dependent Diabetes Mellitus. Circulation 2001, 103, 1238–1244. [Google Scholar] [CrossRef] [PubMed]
  63. Day, C.P.; James, O.F.W. Steatohepatitis: A Tale of Two “Hits”? Gastroenterology 1998, 114, 842–845. [Google Scholar] [CrossRef] [PubMed]
  64. Buzzetti, E.; Pinzani, M.; Tsochatzis, E.A. The Multiple-Hit Pathogenesis of Non-Alcoholic Fatty Liver Disease (NAFLD). Metabolism 2016, 65, 1038–1048. [Google Scholar] [CrossRef]
  65. Schwärzler, J.; Grabherr, F.; Grander, C.; Adolph, T.E.; Tilg, H. The Pathophysiology of MASLD: An Immunometabolic Perspective. Expert Rev. Clin. Immunol. 2024, 20, 375–386. [Google Scholar] [CrossRef]
  66. Hotamisligil, G.S. Inflammation and Metabolic Disorders. Nature 2006, 444, 860–867. [Google Scholar] [CrossRef]
  67. Mladenić, K.; Lenartić, M.; Marinović, S.; Polić, B.; Wensveen, F.M. The “Domino Effect” in MASLD: The Inflammatory Cascade of Steatohepatitis. Eur. J. Immunol. 2024, 54, 2149641. [Google Scholar] [CrossRef]
  68. Duan, Y.; Pan, X.; Luo, J.; Xiao, X.; Li, J.; Bestman, P.L.; Luo, M. Association of Inflammatory Cytokines with Non-Alcoholic Fatty Liver Disease. Front. Immunol. 2022, 13, 880298. [Google Scholar] [CrossRef]
  69. Norton, L.; Shannon, C.; Gastaldelli, A.; DeFronzo, R.A. Insulin: The Master Regulator of Glucose Metabolism. Metabolism 2022, 129, 155142. [Google Scholar] [CrossRef]
  70. Begriche, K.; Igoudjil, A.; Pessayre, D.; Fromenty, B. Mitochondrial Dysfunction in NASH: Causes, Consequences and Possible Means to Prevent It. Mitochondrion 2006, 6, 1–28. [Google Scholar] [CrossRef]
  71. Bansal, S.K.; Bansal, M.B. Pathogenesis of MASLD and MASH—Role of Insulin Resistance and Lipotoxicity. Aliment. Pharmacol. Ther. 2024, 59, S10–S22. Available online: https://onlinelibrary.wiley.com/doi/10.1111/apt.17930 (accessed on 3 December 2024). [CrossRef] [PubMed]
  72. Li, Y.; Yang, P.; Ye, J.; Xu, Q.; Wu, J.; Wang, Y. Updated Mechanisms of MASLD Pathogenesis. Lipids Health Dis. 2024, 23, 117. [Google Scholar] [CrossRef] [PubMed]
  73. Polimeni, L. Oxidative Stress: New Insights on the Association of Non-Alcoholic Fatty Liver Disease and Atherosclerosis. World J. Hepatol. 2015, 7, 1325–1335. [Google Scholar] [CrossRef] [PubMed]
  74. Sjøgaard-Frich, L.M.; Henriksen, M.S.; Lam, S.M.; Birkbak, F.J.; Czaplinska, D.; Flinck, M.; Pedersen, S.F. NHE1 Regulation in NAFLD In Vitro Contributes to Hepatocyte Injury and HSC Crosstalk. J. Endocrinol. 2024, 263, e240099. [Google Scholar] [CrossRef]
  75. Sunny, N.E.; Parks, E.J.; Browning, J.D.; Burgess, S.C. Excessive Hepatic Mitochondrial TCA Cycle and Gluconeogenesis in Humans with Nonalcoholic Fatty Liver Disease. Cell Metab. 2011, 14, 804–810. [Google Scholar] [CrossRef]
  76. Radosavljevic, T.; Brankovic, M.; Samardzic, J.; Djuretić, J.; Vukicevic, D.; Vucevic, D.; Jakovljevic, V. Altered Mitochondrial Function in MASLD: Key Features and Promising Therapeutic Approaches. Antioxidants 2024, 13, 906. [Google Scholar] [CrossRef]
  77. Ajoolabady, A.; Kaplowitz, N.; Lebeaupin, C.; Kroemer, G.; Kaufman, R.J.; Malhi, H.; Ren, J. Endoplasmic Reticulum Stress in Liver Diseases. Hepatology 2023, 77, 619–639. [Google Scholar] [CrossRef]
  78. Almanza, A.; Carlesso, A.; Chintha, C.; Creedican, S.; Doultsinos, D.; Leuzzi, B.; Luís, A.; McCarthy, N.; Montibeller, L.; More, S.; et al. Endoplasmic Reticulum Stress Signaling—From Basic Mechanisms to Clinical Applications. FEBS J. 2019, 286, 241–278. [Google Scholar] [CrossRef]
  79. Feldstein, A.E.; Werneburg, N.W.; Canbay, A.; Guicciardi, M.E.; Bronk, S.F.; Rydzewski, R.; Burgart, L.J.; Gores, G.J. Free Fatty Acids Promote Hepatic Lipotoxicity by Stimulating TNF-α Expression via a Lysosomal Pathway. Hepatology 2004, 40, 185–194. [Google Scholar] [CrossRef]
  80. Baratta, F.; Pastori, D.; Del Ben, M.; Polimeni, L.; Labbadia, G.; Di Santo, S.; Piemonte, F.; Tozzi, G.; Violi, F.; Angelico, F. Reduced Lysosomal Acid Lipase Activity in Adult Patients with Non-Alcoholic Fatty Liver Disease. EBioMedicine 2015, 2, 750–754. [Google Scholar] [CrossRef]
  81. Frietze, K.K.; Brown, A.M.; Das, D.; Franks, R.G.; Cunningham, J.L.; Hayward, M.; Nickels, J.T., Jr. Lipotoxicity Reduces DDX58/Rig-1 Expression and Activity Leading to Impaired Autophagy and Cell Death. Autophagy 2022, 18, 142–160. [Google Scholar] [CrossRef] [PubMed]
  82. Kvansakul, M.; Hinds, M.G. The Bcl-2 Family: Structures, Interactions and Targets for Drug Discovery. Apoptosis 2015, 20, 136–150. [Google Scholar] [CrossRef] [PubMed]
  83. Alkhouri, N.; Carter-Kent, C.; Feldstein, A.E. Apoptosis in Nonalcoholic Fatty Liver Disease: Diagnostic and Therapeutic Implications. Expert Rev. Gastroenterol. Hepatol. 2011, 5, 201–212. [Google Scholar] [CrossRef] [PubMed]
  84. Flores-Romero, H.; Hohorst, L.; John, M.; Albert, M.; King, L.E.; Beckmann, L.; Szabo, T.; Hertlein, V.; Luo, X.; Villunger, A.; et al. BCL-2-Family Protein tBID Can Act as a BAX-Like Effector of Apoptosis. EMBO J. 2022, 41, e108690. [Google Scholar] [CrossRef]
  85. Hartman, L.; Czyz, M. Pro-Apoptotic Activity of BH3-Only Proteins and BH3 Mimetics: From Theory to Potential Cancer Therapy. Anticancer Agents Med. Chem. 2012, 12, 966–981. [Google Scholar] [CrossRef]
  86. Peleman, C.; Hellemans, S.; Veeckmans, G.; Arras, W.; Zheng, H.; Koeken, I.; Van San, E.; Hassannia, B.; Walravens, M.; Kayirangwa, E.; et al. Ferroptosis is a Targetable Detrimental Factor in Metabolic Dysfunction-Associated Steatotic Liver Disease. Cell Death Differ. 2024, 31, 1113–1126. [Google Scholar] [CrossRef]
  87. Qi, J.; Kim, J.W.; Zhou, Z.; Lim, C.W.; Kim, B. Ferroptosis Affects the Progression of Nonalcoholic Steatohepatitis via the Modulation of Lipid Peroxidation-Mediated Cell Death in Mice. Am. J. Pathol. 2020, 190, 68–81. [Google Scholar] [CrossRef]
  88. Tsurusaki, S.; Tsuchiya, Y.; Koumura, T.; Nakasone, M.; Sakamoto, T.; Matsuoka, M.; Imai, H.; Yuet-Yin Kok, C.; Okochi, H.; Nakano, H.; et al. Hepatic Ferroptosis Plays an Important Role as the Trigger for Initiating Inflammation in Nonalcoholic Steatohepatitis. Cell Death Dis. 2019, 10, 449. [Google Scholar] [CrossRef]
  89. Tong, J.; Lan, X.T.; Zhang, Z.; Liu, Y.; Sun, D.Y.; Wang, X.J.; Ou-Yang, S.X.; Zhuang, C.L.; Shen, F.M.; Wang, P.; et al. Ferroptosis Inhibitor Liproxstatin-1 Alleviates Metabolic Dysfunction-Associated Fatty Liver Disease in Mice: Potential Involvement of PANoptosis. Acta Pharmacol. Sin. 2023, 44, 1014–1028. [Google Scholar] [CrossRef]
  90. Kubes, P.; Jenne, C. Immune Responses in the Liver. Annu. Rev. Immunol. 2018, 36, 247–277. [Google Scholar] [CrossRef]
  91. Hammerich, L.; Tacke, F. Hepatic Inflammatory Responses in Liver Fibrosis. Nat. Rev. Gastroenterol. Hepatol. 2023, 20, 633–646. [Google Scholar] [CrossRef] [PubMed]
  92. Wang, H.; Zhang, H.; Wang, Y.; Brown, Z.J.; Xia, Y.; Huang, Z.; Shen, C.; Hu, Z.; Beane, J.; Ansa-Addo, E.A.; et al. Regulatory T-Cell and Neutrophil Extracellular Trap Interaction Contributes to Carcinogenesis in Non-Alcoholic Steatohepatitis. J. Hepatol. 2021, 75, 1271–1283. [Google Scholar] [CrossRef] [PubMed]
  93. Dudek, M.; Pfister, D.; Donakonda, S.; Filpe, P.; Schneider, A.; Laschinger, M.; Hartmann, D.; Hüser, N.; Meiser, P.; Bayerl, F.; et al. Author Correction: Auto-Aggressive CXCR6+ CD8 T Cells Cause Liver Immune Pathology in NASH. Nature 2021, 593, E14. [Google Scholar] [CrossRef] [PubMed]
  94. Leigh, S.J.; Morris, M.J. Diet, Inflammation and the Gut Microbiome: Mechanisms for Obesity-Associated Cognitive Impairment. Biochim. Biophys. Acta BBA—Mol. Basis Dis. 2020, 1866, 165767. [Google Scholar] [CrossRef]
  95. Tilg, H.; Adolph, T.E.; Trauner, M. Gut-Liver Axis: Pathophysiological Concepts and Clinical Implications. Cell Metab. 2022, 34, 1700–1718. [Google Scholar] [CrossRef]
  96. Cinti, S. Anatomy and Physiology of the Nutritional System. Mol. Asp. Med. 2019, 68, 101–107. [Google Scholar] [CrossRef]
  97. Kiela, P.R.; Ghishan, F.K. Physiology of Intestinal Absorption and Secretion. Best Pract. Res. Clin. Gastroenterol. 2016, 30, 145–159. [Google Scholar] [CrossRef]
  98. Mowat, A.M.; Agace, W.W. Regional Specialization Within the Intestinal Immune System. Nat. Rev. Immunol. 2014, 14, 667–685. [Google Scholar] [CrossRef]
  99. Shaker, A.; Rubin, D.C. Intestinal Stem Cells and Epithelial–Mesenchymal Interactions in the Crypt and Stem Cell Niche. Transl. Res. 2010, 156, 180–187. [Google Scholar] [CrossRef]
  100. Zheng, Z.; Wang, B. The Gut-Liver Axis in Health and Disease: The Role of Gut Microbiota-Derived Signals in Liver Injury and Regeneration. Front. Immunol. 2021, 12, 775526. [Google Scholar] [CrossRef]
  101. Chopyk, D.M.; Grakoui, A. Contribution of the Intestinal Microbiome and Gut Barrier to Hepatic Disorders. Gastroenterology 2020, 159, 849–863. [Google Scholar] [CrossRef] [PubMed]
  102. Cani, P.D.; Bibiloni, R.; Knauf, C.; Waget, A.; Neyrinck, A.M.; Delzenne, N.M.; Burcelin, R. Changes in Gut Microbiota Control Metabolic Endotoxemia-Induced Inflammation in High-Fat Diet–Induced Obesity and Diabetes in Mice. Diabetes 2008, 57, 1470–1481. [Google Scholar] [CrossRef] [PubMed]
  103. Violi, F.; Cammisotto, V.; Bartimoccia, S.; Pignatelli, P.; Carnevale, R.; Nocella, C. Gut-Derived Low-Grade Endotoxaemia, Atherothrombosis, and Cardiovascular Disease. Nat. Rev. Cardiol. 2023, 20, 24–37. [Google Scholar] [CrossRef] [PubMed]
  104. Vancamelbeke, M.; Vermeire, S. The Intestinal Barrier: A Fundamental Role in Health and Disease. Expert Rev. Gastroenterol. Hepatol. 2017, 11, 821–834. [Google Scholar] [CrossRef]
  105. Hansson, G.C. Mucins and the Microbiome. Annu. Rev. Biochem. 2020, 89, 769–793. [Google Scholar] [CrossRef]
  106. Bansil, R.; Turner, B.S. The Biology of Mucus: Composition, Synthesis, and Organization. Adv. Drug Deliv. Rev. 2018, 124, 3–15. [Google Scholar] [CrossRef]
  107. Cornick, S.; Tawiah, A.; Chadee, K. Roles and Regulation of the Mucus Barrier in the Gut. Tissue Barriers 2015, 3, e982426. [Google Scholar] [CrossRef]
  108. Vanuytsel, T.; Tack, J.; Farre, R. The Role of Intestinal Permeability in Gastrointestinal Disorders and Current Methods of Evaluation. Front. Nutr. 2021, 8, 717925. [Google Scholar] [CrossRef]
  109. González-Mariscal, L.; Betanzos, A.; Nava, P.; Jaramillo, B.E. Tight Junction Proteins. Prog. Biophys. Mol. Biol. 2003, 81, 1–44. [Google Scholar] [CrossRef]
  110. Hyun, J.; Romero, L.; Riveron, R.; Flores, C.; Kanagavelu, S.; Chung, K.D.; Alonso, A.; Sotolongo, J.; Ruiz, J.; Manukyan, A.; et al. Human Intestinal Epithelial Cells Express Interleukin-10 through Toll-Like Receptor 4-Mediated Epithelial-Macrophage Crosstalk. J. Innate Immun. 2015, 7, 87–101. [Google Scholar] [CrossRef]
  111. Buckley, A.; Turner, J.R. Cell Biology of Tight Junction Barrier Regulation and Mucosal Disease. Cold Spring Harb. Perspect. Biol. 2018, 10, a029314. [Google Scholar] [CrossRef] [PubMed]
  112. Pietrzak, B.; Tomela, K.; Olejnik-Schmidt, A.; Mackiewicz, A.; Schmidt, M. Secretory IgA in Intestinal Mucosal Secretions as an Adaptive Barrier Against Microbial Cells. Int. J. Mol. Sci. 2020, 21, 9254. [Google Scholar] [CrossRef] [PubMed]
  113. Yatsunenko, T.; Rey, F.E.; Manary, M.J.; Trehan, I.; Dominguez-Bello, M.G.; Contreras, M.; Magris, M.; Hidalgo, G.; Baldassano, R.N.; Anokhin, A.P.; et al. Human Gut Microbiome Viewed Across Age and Geography. Nature 2012, 486, 222–227. [Google Scholar] [CrossRef] [PubMed]
  114. Rowland, I.; Gibson, G.; Heinken, A.; Scott, K.; Swann, J.; Thiele, I.; Tuohy, K. Gut Microbiota Functions: Metabolism of Nutrients and Other Food Components. Eur. J. Nutr. 2018, 57, 1–24. [Google Scholar] [CrossRef]
  115. Jiang, L.; Fan, J.G. The Role of the Gut Microbiome in Chronic Liver Diseases: Present Insights and Future Outlook. Hepatobiliary Pancreat. Dis. Int. 2023, 22, 441–443. [Google Scholar] [CrossRef]
  116. Woese, C.R.; Fox, G.E. Phylogenetic Structure of the Prokaryotic Domain: The Primary Kingdoms. Proc. Natl. Acad. Sci. USA 1977, 74, 5088–5090. [Google Scholar] [CrossRef]
  117. Rosenberg, E.; Zilber-Rosenberg, I. Microbes Drive Evolution of Animals and Plants: The Hologenome Concept. mBio 2016, 7, e01395-15. [Google Scholar] [CrossRef]
  118. Davies, J. In a Map for Human Life, Count the Microbes, Too. Science 2001, 291, 2316. [Google Scholar] [CrossRef]
  119. The NIH HMP Working Group; Peterson, J.; Garges, S.; Giovanni, M.; McInnes, P.; Wang, L.; Schloss, J.A.; Bonazzi, V.; McEwen, J.E.; Wetterstrand, K.A.; et al. The NIH Human Microbiome Project. Genome Res. 2009, 19, 2317–2323. [Google Scholar]
  120. Relman, D. The Meaning and Impact of the Human Genome Sequence for Microbiology. Trends Microbiol. 2001, 9, 206–208. [Google Scholar] [CrossRef]
  121. El-Sayed, A.; Aleya, L.; Kamel, M. Microbiota’s Role in Health and Diseases. Environ. Sci. Pollut. Res. 2021, 28, 36967–36983. [Google Scholar] [CrossRef] [PubMed]
  122. Manos, J. The Human Microbiome in Disease and Pathology. APMIS 2022, 130, 690–705. [Google Scholar] [CrossRef] [PubMed]
  123. MetaHIT Consortium; Li, J.; Jia, H.; Cai, X.; Zhong, H.; Feng, Q.; Sunagawa, S.; Arumugam, M.; Kultima, J.R.; Prifti, E.; et al. An Integrated Catalog of Reference Genes in the Human Gut Microbiome. Nat. Biotechnol. 2014, 32, 834–841. [Google Scholar]
  124. Bäckhed, F.; Roswall, J.; Peng, Y.; Feng, Q.; Jia, H.; Kovatcheva-Datchary, P.; Li, Y.; Xia, Y.; Xie, H.; Zhong, H.; et al. Dynamics and Stabilization of the Human Gut Microbiome during the First Year of Life. Cell Host Microbe 2015, 17, 690–703. [Google Scholar] [CrossRef]
  125. Lagier, J.C.; Hugon, P.; Khelaifia, S.; Fournier, P.E.; La Scola, B.; Raoult, D. The Rebirth of Culture in Microbiology through the Example of Culturomics to Study Human Gut Microbiota. Clin. Microbiol. Rev. 2015, 28, 237–264. [Google Scholar] [CrossRef]
  126. Browne, H.P.; Forster, S.C.; Anonye, B.O.; Kumar, N.; Neville, B.A.; Stares, M.D.; Goulding, D.; Lawley, T.D. Culturing of ‘Uncultured’ Human Microbiota Reveals Novel Taxa and Extensive Sporulation. Nature 2016, 533, 543–546. [Google Scholar] [CrossRef]
  127. Metzker, M.L. Emerging Technologies in DNA Sequencing. Genome Res. 2005, 15, 1767–1776. [Google Scholar] [CrossRef]
  128. Le Chatelier, E.; Nielsen, T.; Qin, J.; Prifti, E.; Hildebrand, F.; Falony, G.; Almeida, M.; Arumugam, M.; Batto, J.M.; Kennedy, S.; et al. Richness of Human Gut Microbiome Correlates with Metabolic Markers. Nature 2013, 500, 541–546. [Google Scholar] [CrossRef]
  129. Nardone, G.; Compare, D. The Human Gastric Microbiota: Is It Time to Rethink the Pathogenesis of Stomach Diseases? United Eur. Gastroenterol. J. 2015, 3, 255–260. [Google Scholar] [CrossRef]
  130. El Aidy, S.; Dinan, T.G.; Cryan, J.F. Gut Microbiota: The Conductor in the Orchestra of Immune–Neuroendocrine Communication. Clin. Ther. 2015, 37, 954–967. [Google Scholar] [CrossRef]
  131. Eckburg, P.B.; Bik, E.M.; Bernstein, C.N.; Purdom, E.; Dethlefsen, L.; Sargent, M.; Gill, S.R.; Nelson, K.E.; Relman, D.A. Diversity of the Human Intestinal Microbial Flora. Science 2005, 308, 1635–1638. [Google Scholar] [CrossRef] [PubMed]
  132. Mariat, D.; Firmesse, O.; Levenez, F.; Guimarâes, V.; Sokol, H.; Doré, J.; Corthier, G.; Furet, J.P. The Firmicutes/Bacteroidetes Ratio of the Human Microbiota Changes with Age. BMC Microbiol. 2009, 9, 123. [Google Scholar] [CrossRef] [PubMed]
  133. Hollister, E.B.; Gao, C.; Versalovic, J. Compositional and Functional Features of the Gastrointestinal Microbiome and Their Effects on Human Health. Gastroenterology 2014, 146, 1449–1458. [Google Scholar] [CrossRef] [PubMed]
  134. Binda, C.; Lopetuso, L.R.; Rizzatti, G.; Gibiino, G.; Cennamo, V.; Gasbarrini, A. Actinobacteria: A Relevant Minority for the Maintenance of Gut Homeostasis. Dig. Liver Dis. 2018, 50, 421–428. [Google Scholar] [CrossRef]
  135. Shin, N.R.; Whon, T.W.; Bae, J.W. Proteobacteria: Microbial Signature of Dysbiosis in Gut Microbiota. Trends Biotechnol. 2015, 33, 496–503. [Google Scholar] [CrossRef]
  136. Puljiz, Z.; Kumric, M.; Vrdoljak, J.; Martinovic, D.; Ticinovic Kurir, T.; Krnic, M.O.; Urlic, H.; Puljiz, Z.; Zucko, J.; Dumanic, P.; et al. Obesity, Gut Microbiota, and Metabolome: From Pathophysiology to Nutritional Interventions. Nutrients 2023, 15, 2236. [Google Scholar] [CrossRef]
  137. Vallianou, N.; Stratigou, T.; Christodoulatos, G.S.; Dalamaga, M. Understanding the Role of the Gut Microbiome and Microbial Metabolites in Obesity and Obesity-Associated Metabolic Disorders: Current Evidence and Perspectives. Curr. Obes. Rep. 2019, 8, 317–332. [Google Scholar] [CrossRef]
  138. Usuda, H.; Okamoto, T.; Wada, K. Leaky Gut: Effect of Dietary Fiber and Fats on Microbiome and Intestinal Barrier. Int. J. Mol. Sci. 2021, 22, 7613. [Google Scholar] [CrossRef]
  139. Shi, N.; Li, N.; Duan, X.; Niu, H. Interaction between the Gut Microbiome and Mucosal Immune System. Mil. Med. Res. 2017, 4, 14. [Google Scholar] [CrossRef]
  140. Schoeler, M.; Caesar, R. Dietary Lipids, Gut Microbiota, and Lipid Metabolism. Rev. Endocr. Metab. Disord. 2019, 20, 461–472. [Google Scholar] [CrossRef]
  141. Xin, D.; Zong-Shun, L.; Bang-Mao, W.; Lu, Z. Expression of Intestinal Tight Junction Proteins in Patients with Non-Alcoholic Fatty Liver Disease. Hepatogastroenterology 2014, 61, 136–140. [Google Scholar] [PubMed]
  142. Rahman, K.; Desai, C.; Iyer, S.S.; Thorn, N.E.; Kumar, P.; Liu, Y.; Smith, T.; Neish, A.S.; Li, H.; Tan, S.; et al. Loss of Junctional Adhesion Molecule A Promotes Severe Steatohepatitis in Mice on a Diet High in Saturated Fat, Fructose, and Cholesterol. Gastroenterology 2016, 151, 733–746.e12. [Google Scholar] [CrossRef] [PubMed]
  143. Liu, L.; Yin, M.; Gao, J.; Yu, C.; Lin, J.; Wu, A.; Zhu, J.; Xu, C.; Liu, X. Intestinal Barrier Function in the Pathogenesis of Nonalcoholic Fatty Liver Disease. J. Clin. Transl. Hepatol. 2023, 11, 452–458. [Google Scholar] [CrossRef] [PubMed]
  144. Spadoni, I.; Zagato, E.; Bertocchi, A.; Paolinelli, R.; Hot, E.; Di Sabatino, A.; Caprioli, F.; Bottiglieri, L.; Oldani, A.; Viale, G.; et al. A Gut-Vascular Barrier Controls the Systemic Dissemination of Bacteria. Science 2015, 350, 830–834. [Google Scholar] [CrossRef]
  145. Mouries, J.; Brescia, P.; Silvestri, A.; Spadoni, I.; Sorribas, M.; Wiest, R.; Mileti, E.; Galbiati, M.; Invernizzi, P.; Adorini, L.; et al. Microbiota-Driven Gut Vascular Barrier Disruption Is a Prerequisite for Non-Alcoholic Steatohepatitis Development. J. Hepatol. 2019, 71, 1216–1228. [Google Scholar] [CrossRef]
  146. Cui, Y.; Wang, Q.; Chang, R.; Zhou, X.; Xu, C. Intestinal Barrier Function–Non-Alcoholic Fatty Liver Disease Interactions and Possible Role of Gut Microbiota. J. Agric. Food Chem. 2019, 67, 2754–2762. [Google Scholar] [CrossRef]
  147. Pellicciotta, M.; Rigoni, R.; Falcone, E.L.; Holland, S.M.; Villa, A.; Cassani, B. The Microbiome and Immunodeficiencies: Lessons from Rare Diseases. J. Autoimmun. 2019, 98, 132–148. [Google Scholar] [CrossRef]
  148. Kubes, P.; Mehal, W.Z. Sterile Inflammation in the Liver. Gastroenterology 2012, 143, 1158–1172. [Google Scholar] [CrossRef]
  149. McPherson, S.; Henderson, E.; Burt, A.D.; Day, C.P.; Anstee, Q.M. Serum Immunoglobulin Levels Predict Fibrosis in Patients with Non-Alcoholic Fatty Liver Disease. J. Hepatol. 2014, 60, 1055–1062. [Google Scholar] [CrossRef]
  150. Shalapour, S.; Lin, X.J.; Bastian, I.N.; Brain, J.; Burt, A.D.; Aksenov, A.A.; Vrbanac, A.F.; Li, W.; Perkins, A.; Matsutani, T.; et al. Inflammation-Induced IgA+ Cells Dismantle Anti-Liver Cancer Immunity. Nature 2017, 551, 340–345. [Google Scholar] [CrossRef]
  151. Boursier, J.; Mueller, O.; Barret, M.; Machado, M.; Fizanne, L.; Araujo-Perez, F.; Guy, C.D.; Seed, P.C.; Rawls, J.F.; David, L.A.; et al. The Severity of Nonalcoholic Fatty Liver Disease Is Associated with Gut Dysbiosis and Shift in the Metabolic Function of the Gut Microbiota. Hepatology 2016, 63, 764–775. [Google Scholar] [CrossRef] [PubMed]
  152. Wilcz-Villega, E.M.; McClean, S.; O’Sullivan, M.A. Mast Cell Tryptase Reduces Junctional Adhesion Molecule-A (JAM-A) Expression in Intestinal Epithelial Cells: Implications for the Mechanisms of Barrier Dysfunction in Irritable Bowel Syndrome. Am. J. Gastroenterol. 2013, 108, 1140–1151. [Google Scholar] [CrossRef] [PubMed]
  153. Potts, R.A.; Tiffany, C.M.; Pakpour, N.; Lokken, K.L.; Tiffany, C.R.; Cheung, K.; Tsolis, R.M.; Luckhart, S. Mast Cells and Histamine Alter Intestinal Permeability During Malaria Parasite Infection. Immunobiology 2016, 221, 468–474. [Google Scholar] [CrossRef] [PubMed]
  154. Tomita, K.; Teratani, T.; Yokoyama, H.; Suzuki, T.; Irie, R.; Ebinuma, H.; Saito, H.; Hokari, R.; Miura, S.; Hibi, T. Serum Immunoglobulin A Concentration Is an Independent Predictor of Liver Fibrosis in Nonalcoholic Steatohepatitis Before the Cirrhotic Stage. Dig. Dis. Sci. 2011, 56, 3648–3654. [Google Scholar] [CrossRef]
  155. Fang, J.; Yu, C.H.; Li, X.J.; Yao, J.M.; Fang, Z.Y.; Yoon, S.H.; Yu, W.Y. Gut Dysbiosis in Nonalcoholic Fatty Liver Disease: Pathogenesis, Diagnosis, and Therapeutic Implications. Front. Cell Infect. Microbiol. 2022, 12, 997018. [Google Scholar] [CrossRef]
  156. Tokuhara, D. Role of the Gut Microbiota in Regulating Non-Alcoholic Fatty Liver Disease in Children and Adolescents. Front. Nutr. 2021, 8, 700058. [Google Scholar] [CrossRef]
  157. Ghosh, S.; Whitley, C.S.; Haribabu, B.; Jala, V.R. Regulation of Intestinal Barrier Function by Microbial Metabolites. Cell Mol. Gastroenterol. Hepatol. 2021, 11, 1463–1482. [Google Scholar] [CrossRef]
  158. Brown, K.; DeCoffe, D.; Molcan, E.; Gibson, D.L. Diet-Induced Dysbiosis of the Intestinal Microbiota and the Effects on Immunity and Disease. Nutrients 2012, 4, 1095–1119. [Google Scholar] [CrossRef]
  159. Khan, M.J.; Gerasimidis, K.; Edwards, C.A.; Shaikh, M.G. Role of Gut Microbiota in the Aetiology of Obesity: Proposed Mechanisms and Review of the Literature. J. Obes. 2016, 2016, 7353642. [Google Scholar] [CrossRef]
  160. Magne, F.; Gotteland, M.; Gauthier, L.; Zazueta, A.; Pesoa, S.; Navarrete, P.; Balamurugan, R. The Firmicutes/Bacteroidetes Ratio: A Relevant Marker of Gut Dysbiosis in Obese Patients? Nutrients 2020, 12, 1474. [Google Scholar] [CrossRef]
  161. Duarte, S.B.M.; Stefano, J.T.; Oliveira, C.P. Microbiota and Nonalcoholic Fatty Liver Disease/Nonalcoholic Steatohepatitis (NAFLD/NASH). Ann. Hepatol. 2019, 18, 416–421. [Google Scholar] [CrossRef] [PubMed]
  162. Yuan, H.; Wu, X.; Wang, X.; Zhou, J.Y.; Park, S. Microbial Dysbiosis Linked to Metabolic Dysfunction-Associated Fatty Liver Disease in Asians: Prevotella copri Promotes Lipopolysaccharide Biosynthesis and Network Instability in the Prevotella Enterotype. Int. J. Mol. Sci. 2024, 25, 2183. [Google Scholar] [CrossRef] [PubMed]
  163. Kuraji, R.; Ye, C.; Zhao, C.; Gao, L.; Martinez, A.; Miyashita, Y.; Radaic, A.; Kamarajan, P.; Le, C.; Zhan, L.; et al. Nisin Lantibiotic Prevents NAFLD Liver Steatosis and Mitochondrial Oxidative Stress Following Periodontal Disease by Abrogating Oral, Gut and Liver Dysbiosis. NPJ Biofilms Microbiomes 2024, 10, 3. [Google Scholar] [CrossRef] [PubMed]
  164. Kapil, S.; Duseja, A.; Sharma, B.K.; Singla, B.; Chakraborti, A.; Das, A.; Ray, P.; Dhiman, R.K.; Chawla, Y. Small Intestinal Bacterial Overgrowth and Toll-Like Receptor Signaling in Patients with Non-Alcoholic Fatty Liver Disease. J. Gastroenterol. Hepatol. 2016, 31, 213–221. [Google Scholar] [CrossRef]
  165. Ganesan, R.; Gupta, H.; Jeong, J.J.; Sharma, S.P.; Won, S.M.; Oh, K.K.; Yoon, S.J.; Kim, D.J.; Suk, K.T. A Metabolomics Approach to the Validation of Predictive Metabolites and Phenotypic Expression in Non-Alcoholic Fatty Liver Disease. Life Sci. 2023, 322, 121626. [Google Scholar] [CrossRef]
  166. Zeng, F.; Su, X.; Liang, X.; Liao, M.; Zhong, H.; Xu, J.; Gou, W.; Zhang, X.; Shen, L.; Zheng, J.S.; et al. Gut Microbiome Features and Metabolites in Non-Alcoholic Fatty Liver Disease Among Community-Dwelling Middle-Aged and Older Adults. BMC Med. 2024, 22, 104. [Google Scholar] [CrossRef]
  167. Mardinoglu, A.; Ural, D.; Zeybel, M.; Yuksel, H.H.; Uhlén, M.; Borén, J. The Potential Use of Metabolic Cofactors in Treatment of NAFLD. Nutrients 2019, 11, 1578. [Google Scholar] [CrossRef]
  168. Quesada-Vázquez, S.; Bone, C.; Saha, S.; Triguero, I.; Colom-Pellicer, M.; Aragonès, G.; Hildebrand, F.; Del Bas, J.M.; Caimari, A.; Beraza, N.; et al. Microbiota Dysbiosis and Gut Barrier Dysfunction Associated with Non-Alcoholic Fatty Liver Disease Are Modulated by a Specific Metabolic Cofactors’ Combination. Int. J. Mol. Sci. 2022, 23, 13675. [Google Scholar] [CrossRef]
  169. Hayashi, T.; Yamashita, T.; Takahashi, T.; Tabata, T.; Watanabe, H.; Gotoh, Y.; Shinohara, M.; Kami, K.; Tanaka, H.; Matsumoto, K.; et al. Uncovering the Role of Gut Microbiota in Amino Acid Metabolic Disturbances in Heart Failure Through Metagenomic Analysis. Front. Cardiovasc. Med. 2021, 8, 789325. [Google Scholar] [CrossRef]
  170. Niu, Y.C.; Feng, R.N.; Hou, Y.; Li, K.; Kang, Z.; Wang, J.; Sun, C.H.; Li, Y. Histidine and Arginine Are Associated with Inflammation and Oxidative Stress in Obese Women. Br. J. Nutr. 2012, 108, 57–61. [Google Scholar] [CrossRef]
  171. Quesada-Vázquez, S.; Castells-Nobau, A.; Latorre, J.; Oliveras-Cañellas, N.; Puig-Parnau, I.; Tejera, N.; Tobajas, Y.; Baudin, J.; Hildebrand, F.; Beraza, N.; et al. Potential Therapeutic Implications of Histidine Catabolism by the Gut Microbiota in NAFLD Patients with Morbid Obesity. Cell Rep. Med. 2023, 4, 101341. [Google Scholar] [CrossRef] [PubMed]
  172. Huus, K.E.; Petersen, C.; Finlay, B.B. Diversity and Dynamism of IgA−Microbiota Interactions. Nat. Rev. Immunol. 2021, 21, 514–525. [Google Scholar] [CrossRef] [PubMed]
  173. Hsu, C.L.; Schnabl, B. The Gut–Liver Axis and Gut Microbiota in Health and Liver Disease. Nat. Rev. Microbiol. 2023, 21, 719–733. [Google Scholar] [CrossRef] [PubMed]
  174. Ohtani, N.; Kamiya, T.; Kawada, N. Recent Updates on the Role of the Gut-Liver Axis in the Pathogenesis of NAFLD/NASH, HCC, and Beyond. Hepatol. Commun. 2023, 7, 9. Available online: https://journals.lww.com/10.1097/HC9.0000000000000241 (accessed on 3 December 2024). [CrossRef]
  175. Ma, R.; Shi, G.; Li, Y.; Shi, H. Trimethylamine N-Oxide, Choline and Its Metabolites Are Associated with the Risk of Non-Alcoholic Fatty Liver Disease. Br. J. Nutr. 2024, 131, 1915–1923. [Google Scholar] [CrossRef]
  176. Martínez-Montoro, J.I.; Martín-Núñez, G.M.; González-Jiménez, A.; Garrido-Sánchez, L.; Moreno-Indias, I.; Tinahones, F.J. Interactions between the Gut Microbiome and DNA Methylation Patterns in Blood and Visceral Adipose Tissue in Subjects with Different Metabolic Characteristics. J. Transl. Med. 2024, 22, 1089. [Google Scholar] [CrossRef]
  177. Shirai, Y.; Yoshiji, H.; Noguchi, R.; Kaji, K.; Aihara, Y.; Douhara, A.; Moriya, K.; Namisaki, T.; Kawaratani, H.; Fukui, H. Cross Talk between Toll-Like Receptor-4 Signaling and Angiotensin-II in Liver Fibrosis Development in the Rat Model of Non-Alcoholic Steatohepatitis. J. Gastroenterol. Hepatol. 2013, 28, 723–730. [Google Scholar] [CrossRef]
  178. Li, J.; Deng, X.; Bai, T.; Wang, S.; Jiang, Q.; Xu, K. Resolvin D1 Mitigates Non-Alcoholic Steatohepatitis by Suppressing the TLR4-MyD88-Mediated NF-κB and MAPK Pathways and Activating the Nrf2 Pathway in Mice. Int. Immunopharmacol. 2020, 88, 106961. [Google Scholar] [CrossRef]
  179. American Diabetes Association. 5. Lifestyle Management: Standards of Medical Care in Diabetes—2019. Diabetes Care 2019, 42, S46–S60. [Google Scholar] [CrossRef]
  180. Plauth, M.; Bernal, W.; Dasarathy, S.; Merli, M.; Plank, L.D.; Schütz, T.; Bischoff, S.C. ESPEN Guideline on Clinical Nutrition in Liver Disease. Clin. Nutr. 2019, 38, 485–521. [Google Scholar] [CrossRef]
  181. Murphy, E.F.; Cotter, P.D.; Healy, S.; Marques, T.M.; O’Sullivan, O.; Fouhy, F.; Clarke, S.F.; O’Toole, P.W.; Quigley, E.M.; Stanton, C.; et al. Composition and Energy Harvesting Capacity of the Gut Microbiota: Relationship to Diet, Obesity and Time in Mouse Models. Gut 2010, 59, 1635–1642. [Google Scholar] [CrossRef] [PubMed]
  182. Turnbaugh, P.J.; Bäckhed, F.; Fulton, L.; Gordon, J.I. Diet-Induced Obesity Is Linked to Marked but Reversible Alterations in the Mouse Distal Gut Microbiome. Cell Host Microbe 2008, 3, 213–223. [Google Scholar] [CrossRef] [PubMed]
  183. Nakayama, T.; Oishi, K. Influence of Coffee (Coffea arabica) and Galacto-Oligosaccharide Consumption on Intestinal Microbiota and the Host Responses. FEMS Microbiol. Lett. 2013, 343, 161–168. [Google Scholar] [CrossRef] [PubMed]
  184. Molloy, J.W.; Calcagno, C.J.; Williams, C.D.; Jones, F.J.; Torres, D.M.; Harrison, S.A. Association of Coffee and Caffeine Consumption with Fatty Liver Disease, Nonalcoholic Steatohepatitis, and Degree of Hepatic Fibrosis. Hepatology 2012, 55, 429–436. [Google Scholar] [CrossRef]
  185. Shen, L. Letter: Gut Microbiota Modulation Contributes to Coffee’s Benefits for Non-Alcoholic Fatty Liver Disease. Aliment. Pharmacol. Ther. 2014, 39, 1441–1442. [Google Scholar] [CrossRef]
  186. Clarke, S.F.; Murphy, E.F.; O’Sullivan, O.; Lucey, A.J.; Humphreys, M.; Hogan, A.; Hayes, P.; O’Reilly, M.; Jeffery, I.B.; Wood-Martin, R.; et al. Exercise and Associated Dietary Extremes Impact on Gut Microbial Diversity. Gut 2014, 63, 1913–1920. [Google Scholar] [CrossRef]
  187. Monda, V.; Villano, I.; Messina, A.; Valenzano, A.; Esposito, T.; Moscatelli, F.; Viggiano, A.; Cibelli, G.; Chieffi, S.; Monda, M.; et al. Exercise Modifies the Gut Microbiota with Positive Health Effects. Oxid. Med. Cell. Longev. 2017, 2017, 3831972. [Google Scholar] [CrossRef]
  188. Ortiz-Alvarez, L.; Xu, H.; Martinez-Tellez, B. Influence of Exercise on the Human Gut Microbiota of Healthy Adults: A Systematic Review. Clin. Transl. Gastroenterol. 2020, 11, e00126. [Google Scholar] [CrossRef]
  189. Reijnders, D.; Goossens, G.H.; Hermes, G.D.A.; Neis, E.P.J.G.; van der Beek, C.M.; Most, J.; Holst, J.J.; Lenaerts, K.; Kootte, R.S.; Nieuwdorp, M.; et al. Effects of Gut Microbiota Manipulation by Antibiotics on Host Metabolism in Obese Humans: A Randomized Double-Blind Placebo-Controlled Trial. Cell Metab. 2016, 24, 63–74. [Google Scholar] [CrossRef]
  190. Chong, C.Y.L.; Orr, D.; Plank, L.D.; Vatanen, T.; O’Sullivan, J.M.; Murphy, R. Randomized Double-Blind Placebo-Controlled Trial of Inulin with Metronidazole in Non-Alcoholic Fatty Liver Disease (NAFLD). Nutrients 2020, 12, 937. [Google Scholar] [CrossRef]
  191. Kakiyama, G.; Pandak, W.M.; Gillevet, P.M.; Hylemon, P.B.; Heuman, D.M.; Daita, K.; Takei, H.; Muto, A.; Nittono, H.; Ridlon, J.M.; et al. Modulation of the Fecal Bile Acid Profile by Gut Microbiota in Cirrhosis. J. Hepatol. 2013, 58, 949–955. [Google Scholar] [CrossRef] [PubMed]
  192. 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, 1237–1246. [Google Scholar] [CrossRef] [PubMed]
  193. Gangarapu, V.; Ince, A.T.; Baysal, B.; Kayar, Y.; Kılıç, U.; Gök, Ö.; Uysal, Ö.; Şenturk, H. Efficacy of Rifaximin on Circulating Endotoxins and Cytokines in Patients with Nonalcoholic Fatty Liver Disease. Eur. J. Gastroenterol. Hepatol. 2015, 27, 840–845. [Google Scholar] [CrossRef] [PubMed]
  194. Cobbold, J.F.L.; Atkinson, S.; Marchesi, J.R.; Smith, A.; Wai, S.N.; Stove, J.; Shojaee-Moradie, F.; Jackson, N.; Umpleby, A.M.; Fitzpatrick, J.; et al. Rifaximin in Non-Alcoholic Steatohepatitis: An Open-Label Pilot Study. Hepatol. Res. Off. J. Jpn. Soc. Hepatol. 2018, 48, 69–77. [Google Scholar] [CrossRef]
  195. Escouto, G.S.; Port, G.Z.; Tovo, C.V.; Fernandes, S.A.; Peres, A.; Dorneles, G.P.; Houde, V.P.; Varin, T.V.; Pilon, G.; Marette, A.; et al. Probiotic Supplementation, Hepatic Fibrosis, and the Microbiota Profile in Patients with Nonalcoholic Steatohepatitis: A Randomized Controlled Trial. J. Nutr. 2023, 153, 1984–1993. [Google Scholar] [CrossRef]
  196. Manzhalii, E.; Virchenko, O.; Falalyeyeva, T.; Beregova, T.; Stremmel, W. Treatment Efficacy of a Probiotic Preparation for Non-Alcoholic Steatohepatitis: A Pilot Trial. J. Dig. Dis. 2017, 18, 698–703. [Google Scholar] [CrossRef]
  197. Barcelos, S.T.A.; Silva-Sperb, A.S.; Moraes, H.A.; Longo, L.; De Moura, B.C.; Michalczuk, M.T.; Uribe-Cruz, C.; Cerski, C.T.S.; da Silveira, T.R.; Dall’Alba, V.; et al. Oral 24-Week Probiotics Supplementation Did Not Decrease Cardiovascular Risk Markers in Patients with Biopsy Proven NASH: A Double-Blind Placebo-Controlled Randomized Study. Ann. Hepatol. 2023, 28, 100769. [Google Scholar] [CrossRef]
  198. Tarantino, G.; Finelli, C. Systematic Review on Intervention with Prebiotics/Probiotics in Patients with Obesity-Related Nonalcoholic Fatty Liver Disease. Future Microbiol. 2015, 10, 889–902. [Google Scholar] [CrossRef]
  199. Carpi, R.Z.; Barbalho, S.M.; Sloan, K.P.; Laurindo, L.F.; Gonzaga, H.F.; Grippa, P.C.; Zutin, T.L.M.; Girio, R.J.S.; Repetti, C.S.F.; Detregiachi, C.R.P.; et al. The Effects of Probiotics, Prebiotics and Synbiotics in Non-Alcoholic Fat Liver Disease (NAFLD) and Non-Alcoholic Steatohepatitis (NASH): A Systematic Review. Int. J. Mol. Sci. 2022, 23, 8805. [Google Scholar] [CrossRef]
  200. Kanchanasurakit, S.; Kositamongkol, C.; Lanoi, K.; Nunta, M.; Saetuan, T.; Chaiyakunapruk, N.; Saokaew, S.; Phisalprapa, P. Effects of Synbiotics, Probiotics, and Prebiotics on Liver Enzymes of Patients with Non-Alcoholic Fatty Liver Disease: A Systematic Review and Network Meta-Analysis. Front. Nutr. 2022, 9, 880014. [Google Scholar] [CrossRef]
  201. Malaguarnera, M.; Vacante, M.; Antic, T.; Giordano, M.; Chisari, G.; Acquaviva, R.; Mastrojeni, S.; Malaguarnera, G.; Mistretta, A.; Li Volti, G.; et al. Bifidobacterium longum with Fructo-Oligosaccharides in Patients with Non Alcoholic Steatohepatitis. Dig. Dis. Sci. 2012, 57, 545–553. [Google Scholar] [CrossRef] [PubMed]
  202. Asgharian, A.; Askari, G.; Esmailzade, A.; Feizi, A.; Mohammadi, V. The Effect of Symbiotic Supplementation on Liver Enzymes, C-Reactive Protein and Ultrasound Findings in Patients with Non-Alcoholic Fatty Liver Disease: A Clinical Trial. Int. J. Prev. Med. 2016, 7, 59. [Google Scholar] [PubMed]
  203. Mofidi, F.; Poustchi, H.; Yari, Z.; Nourinayyer, B.; Merat, S.; Sharafkhah, M.; Malekzadeh, R.; Hekmatdoost, A. Synbiotic Supplementation in Lean Patients with Non-Alcoholic Fatty Liver Disease: A Pilot, Randomised, Double-Blind, Placebo-Controlled, Clinical Trial. Br. J. Nutr. 2017, 117, 662–668. [Google Scholar] [CrossRef] [PubMed]
  204. Vaughn, B.P.; Rank, K.M.; Khoruts, A. Fecal Microbiota Transplantation: Current Status in Treatment of GI and Liver Disease. Clin. Gastroenterol. Hepatol. Off. Clin. Pract. J. Am. Gastroenterol. Assoc. 2019, 17, 353–361. [Google Scholar] [CrossRef]
  205. Routy, B.; Lenehan, J.G.; Miller, W.H.; Jamal, R.; Messaoudene, M.; Daisley, B.A.; Hes, C.; Al, K.F.; Martinez-Gili, L.; Punčochář, M.; et al. Fecal Microbiota Transplantation Plus Anti-PD-1 Immunotherapy in Advanced Melanoma: A Phase I Trial. Nat. Med. 2023, 29, 2121–2132. [Google Scholar] [CrossRef]
  206. Belvoncikova, P.; Maronek, M.; Gardlik, R. Gut Dysbiosis and Fecal Microbiota Transplantation in Autoimmune Diseases. Int. J. Mol. Sci. 2022, 23, 10729. [Google Scholar] [CrossRef]
  207. 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]
  208. Xue, L.F.; Luo, W.H.; Wu, L.H.; He, X.X.; Xia, H.H.X.; Chen, Y. Fecal Microbiota Transplantation for the Treatment of Nonalcoholic Fatty Liver Disease. Explor. Res. Hypothesis Med. 2019, 4, 12–18. [Google Scholar] [CrossRef]
  209. Bajaj, J.S.; Fagan, A.; Gavis, E.A.; Kassam, Z.; Sikaroodi, M.; Gillevet, P.M. Long-Term Outcomes of Fecal Microbiota Transplantation in Patients with Cirrhosis. Gastroenterology 2019, 156, 1921–1923.e3. [Google Scholar] [CrossRef]
  210. Vrieze, A.; Van Nood, E.; Holleman, F.; Salojärvi, J.; Kootte, R.S.; Bartelsman, J.F.; Dallinga-Thie, G.M.; Ackermans, M.T.; Serlie, M.J.; Oozeer, R.; et al. Transfer of Intestinal Microbiota from Lean Donors Increases Insulin Sensitivity in Individuals with Metabolic Syndrome. Gastroenterology 2012, 143, 913–916.e7. [Google Scholar] [CrossRef]
  211. de Groot, P.; Scheithauer, T.; Bakker, G.J.; Prodan, A.; Levin, E.; Khan, M.T.; Herrema, H.; Ackermans, M.; Serlie, M.J.M.; de Brauw, M.; et al. Donor Metabolic Characteristics Drive Effects of Faecal Microbiota Transplantation on Recipient Insulin Sensitivity, Energy Expenditure and Intestinal Transit Time. Gut 2020, 69, 502–512. [Google Scholar] [CrossRef] [PubMed]
  212. Brandt, L.J.; Aroniadis, O.C. An Overview of Fecal Microbiota Transplantation: Techniques, Indications, and Outcomes. Gastrointest. Endosc. 2013, 78, 240–249. [Google Scholar] [CrossRef] [PubMed]
  213. Persky, S.E.; Brandt, L.J. Treatment of Recurrent Clostridium difficile-Associated Diarrhea by Administration of Donated Stool Directly Through a Colonoscope. Am. J. Gastroenterol. 2000, 95, 3283–3285. [Google Scholar] [PubMed]
  214. Allegretti, J.R.; Kassam, Z.; Mullish, B.H.; Chiang, A.; Carrellas, M.; Hurtado, J.; Marchesi, J.R.; McDonald, J.A.K.; Pechlivanis, A.; Barker, G.F.; et al. Effects of Fecal Microbiota Transplantation with Oral Capsules in Obese Patients. Clin. Gastroenterol. Hepatol. Off. Clin. Pract. J. Am. Gastroenterol. Assoc. 2020, 18, 855–863.e2. [Google Scholar] [CrossRef]
  215. Yang, J.; Tang, X.; Liang, Z.; Chen, M.; Sun, L. Taurocholic Acid Promotes Hepatic Stellate Cell Activation via S1PR2/p38 MAPK/YAP Signaling under Cholestatic Conditions. Clin. Mol. Hepatol. 2023, 29, 465–481. [Google Scholar] [CrossRef]
  216. Mancinelli, R.; Ceci, L.; Kennedy, L.; Francis, H.; Meadows, V.; Chen, L.; Carpino, G.; Kyritsi, K.; Wu, N.; Zhou, T.; et al. The Effects of Taurocholic Acid on Biliary Damage and Liver Fibrosis Are Mediated by Calcitonin-Gene-Related Peptide Signaling. Cells 2022, 11, 1591. [Google Scholar] [CrossRef]
  217. Duan, D.; Wang, M.; Han, J.; Li, M.; Wang, Z.; Zhou, S.; Xin, W.; Li, X. Advances in Multi-Omics Integrated Analysis Methods Based on the Gut Microbiome and Their Applications. Front. Microbiol. 2025, 15, 1509117. [Google Scholar] [CrossRef]
  218. Thakral, N.; Desalegn, H.; Diaz, L.A.; Cabrera, D.; Loomba, R.; Arrese, M.; Arab, J.P. A Precision Medicine Guided Approach to the Utilization of Biomarkers in MASLD. Semin. Liver Dis. 2024, 44, 273–286. [Google Scholar] [CrossRef]
Figure 1. Graphical representation of the interaction between intestinal dysbiosis, altered intestinal permeability, and the gut–liver axis in the pathogenesis of MASLD (metabolic dysfunction–associated steatotic liver disease). The image highlights the complex relationship between the gut microbiota, altered intestinal permeability, and the gut–liver axis in the pathogenesis of MASLD. It demonstrates how intestinal dysbiosis, characterized by an imbalance between beneficial and pathogenic bacteria, can compromise the integrity of the intestinal barrier, leading to the phenomenon of ‘leaky gut’. This process allows the translocation of lipopolysaccharides (LPS) and other bacterial products into the portal circulation, activating hepatic immune cells such as Kupffer cells and triggering inflammatory responses. These events contribute to the progression of hepatic steatosis to more advanced forms, such as metabolic-associated steatohepatitis (MASH) and liver fibrosis. The figure also emphasizes the role of microbiota in fatty acid metabolism and the production of bioactive metabolites, such as short-chain fatty acids (SCFAs), such as acetate, propionate and butyrate, and trimethylamine-N-oxide (TMAO), which modulate systemic inflammation and cardiovascular risk. The gut–liver axis emerges as a critical node for understanding the pathogenic mechanisms of MASLD and as a potential target for innovative therapies, including probiotics, prebiotics, and personalized dietary interventions.
Figure 1. Graphical representation of the interaction between intestinal dysbiosis, altered intestinal permeability, and the gut–liver axis in the pathogenesis of MASLD (metabolic dysfunction–associated steatotic liver disease). The image highlights the complex relationship between the gut microbiota, altered intestinal permeability, and the gut–liver axis in the pathogenesis of MASLD. It demonstrates how intestinal dysbiosis, characterized by an imbalance between beneficial and pathogenic bacteria, can compromise the integrity of the intestinal barrier, leading to the phenomenon of ‘leaky gut’. This process allows the translocation of lipopolysaccharides (LPS) and other bacterial products into the portal circulation, activating hepatic immune cells such as Kupffer cells and triggering inflammatory responses. These events contribute to the progression of hepatic steatosis to more advanced forms, such as metabolic-associated steatohepatitis (MASH) and liver fibrosis. The figure also emphasizes the role of microbiota in fatty acid metabolism and the production of bioactive metabolites, such as short-chain fatty acids (SCFAs), such as acetate, propionate and butyrate, and trimethylamine-N-oxide (TMAO), which modulate systemic inflammation and cardiovascular risk. The gut–liver axis emerges as a critical node for understanding the pathogenic mechanisms of MASLD and as a potential target for innovative therapies, including probiotics, prebiotics, and personalized dietary interventions.
Livers 05 00017 g001
Table 2. Gut microbiota composition across intestinal sections.
Table 2. Gut microbiota composition across intestinal sections.
Intestinal SectionPredominant BacteriaMain FunctionsRef.
StomachStreptococcus, Neisseria, LactobacillusAcid resistance, limited diversity[129]
Small IntestineAerobic Gram-positive (duodenum), anaerobes (ileum)Nutrient absorption, gradual transition[130,131]
Large IntestineFirmicutes, Bacteroidetes.Fermentative role; Fermentation of complex carbohydrates, SCFA synthesis[132,133]
SCFA: short-chain fatty acid.
Table 3. Critical functions of the gut microbiota and their impact on host health.
Table 3. Critical functions of the gut microbiota and their impact on host health.
FunctionDescriptionRef.
Nutrient MetabolismFermentation of dietary fibers, producing SCFAs (acetate, butyrate, propionate) essential for energy homeostasis and inflammatory modulation.
Synthesis of essential vitamins, such as vitamin K and B-group vitamins.
[114,137]
Intestinal Barrier IntegrityModulation of tight junction proteins and increased mucin production to prevent pathogen translocation.[138]
Immune ModulationRegulation of immune development and tolerance through interactions with dendritic cells, regulatory T cells, and SIgA production.[136,139]
Production of Bioactive MetabolitesBeyond SCFAs, the microbiota produces metabolites such as TMAO and phenolic compounds, influencing systemic metabolism, including liver functions.[140]
SCFAs: short-chain fatty acids, TMAO: trimethylamine N-oxide, SIgA: secretory immunoglobulin A.
Table 4. Mechanisms of intestinal barrier dysfunction in MASLD.
Table 4. Mechanisms of intestinal barrier dysfunction in MASLD.
Type of DamageDescriptionConsequences on MASLDRef.
MechanicalAlterations in tight junctions, increased permeabilityBacterial translocation, activation of Kupffer cells[141,142,143,144,145]
ImmunologicalReduced production of SIgA, increased pro-inflammatory cytokinesSystemic inflammation, liver damage[146,147,148,149,150,151,152,153,154]
MicrobialDecreased microbial diversity, increased pathogenic bacteriaDysbiosis, activation of the gut–liver inflammatory cycle[155,156,157,158,159,160,161]
SIgA: secretory immunoglobulin A.
Table 5. Summary of therapeutic approaches targeting gut microbiota in MASLD management.
Table 5. Summary of therapeutic approaches targeting gut microbiota in MASLD management.
Therapeutic ApproachIntervention and Key FindingsMechanism and OutcomesRef.
Dietary InterventionsHypocaloric Diet and Exercise: Achieving 7–10% weight loss via a 500–1000 kcal/day deficit improves hepatic outcomes.
Mediterranean Diet (MD): Rich in fiber, monounsaturated/polyunsaturated fats, and bioactive compounds; reduces hepatic steatosis and improves metabolic parameters.
Moderate Coffee Intake: Associated with reduced oxidative stress and modulation of gut microbiota.
Promotes a negative energy balance and improves metabolic profiles.
MD’s anti-inflammatory and antioxidant properties support hepatic and gut health, including a favorable gut microbiota composition (e.g., increased Akkermansia muciniphila).
[177,178,179,180,181,182,183,184,185]
ExerciseRegular physical activity in both human and animal studies.
Observed increase in microbial diversity, particularly beneficial species (e.g., Akkermansia muciniphila), and modulation of gut taxa.
Enhances insulin sensitivity, lowers LDL cholesterol, and reduces hepatic fat accumulation and serum triglycerides.
Modulates the gut microbiota by decreasing the Firmicutes/Bacteroidetes ratio and influencing specific taxa.
[186,187,188]
AntibioticsUse of norfloxacin and neomycin in preclinical studies improves hepatic function by altering microbiota and reducing bacterial translocation.
Combination of metronidazole and inulin shows significant reductions in serum ALT.
Rifaximin therapy (1100 mg/day for 6 months) improves liver enzymes and inflammatory markers, although results are variable.
Alters gut microbiota composition, reducing harmful bacterial translocation.
Rifaximin modulates fecal bile acid profiles, improves insulin resistance, and reduces pro-inflammatory cytokines, though its efficacy may vary across studies.
[189,190,191,192,193,194]
Probiotics, Prebiotics, and SynbioticsProbiotics: Supplementation improves the AST-to-platelet ratio and may reduce hepatic inflammation (varied durations).
Prebiotics: Oligofructose (16 g/day for 8 weeks) reduces AST levels, though effects on triglycerides are minimal.
Synbiotics: Combined treatments have shown improvements in BMI, liver enzymes (AST, ALT, GGT), NAFLD fibrosis score, and inflammatory markers.
Modulate gut microbiota to reduce hepatic inflammation and oxidative stress.
Enhance growth of beneficial bacteria, thereby improving metabolic parameters and reducing liver fat accumulation.
Some trials report mixed results, underlining the need for further research.
[195,196,197,198,199,200,201,202,203]
Fecal Microbiota Transplantation (FMT)FMT administered via oral capsules, nasogastric tubes, enemas, or colonoscopy.
Single or multiple infusions have shown improvement in small intestinal permeability, modest reductions in hepatic steatosis, and long-term benefits in cirrhosis (e.g., reduced hepatic encephalopathy episodes).
Restores intestinal eubiosis and improves barrier integrity.
Reduces systemic inflammation by modulating the gut–liver axis.
Donor characteristics (lean vs. obese) are crucial for efficacy, influencing outcomes such as insulin sensitivity.
[204,205,206,207,208,209,210,211,212,213,214,215,216]
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

Acierno, C.; Nevola, R.; Rinaldi, L.; Sasso, F.C.; Adinolfi, L.E.; Caturano, A. The Intestinal Thread of Fate: How the Microbiota Shapes the Story of Liver Disease. Livers 2025, 5, 17. https://doi.org/10.3390/livers5020017

AMA Style

Acierno C, Nevola R, Rinaldi L, Sasso FC, Adinolfi LE, Caturano A. The Intestinal Thread of Fate: How the Microbiota Shapes the Story of Liver Disease. Livers. 2025; 5(2):17. https://doi.org/10.3390/livers5020017

Chicago/Turabian Style

Acierno, Carlo, Riccardo Nevola, Luca Rinaldi, Ferdinando Carlo Sasso, Luigi Elio Adinolfi, and Alfredo Caturano. 2025. "The Intestinal Thread of Fate: How the Microbiota Shapes the Story of Liver Disease" Livers 5, no. 2: 17. https://doi.org/10.3390/livers5020017

APA Style

Acierno, C., Nevola, R., Rinaldi, L., Sasso, F. C., Adinolfi, L. E., & Caturano, A. (2025). The Intestinal Thread of Fate: How the Microbiota Shapes the Story of Liver Disease. Livers, 5(2), 17. https://doi.org/10.3390/livers5020017

Article Metrics

Back to TopTop