Preprint
Review

This version is not peer-reviewed.

Sirtuins and Gut Microbiota: Dynamics in Health and a Journey from Metabolic Dysfunction to Hepatocellular Carcinoma

A peer-reviewed article of this preprint also exists.

Submitted:

31 January 2025

Posted:

31 January 2025

You are already at the latest version

Abstract

Metabolic dysfunction leading to non-alcoholic fatty liver disease (NAFLD) exhibits distinct molecular and immune signatures that are influenced by factors like gut microbiota. The gut microbiome interacts with the liver via a bidirectional relationship the gut-liver axis. The microbial metabolites, sirtuins and immune responses play a pivotal role in different metabolic diseases. This extensive review explores the complex and multifaceted interrelationship between sirtuins and gut microbiota, highlighting their importance in both health and disease, particularly in relation to metabolic dysfunction and hepatocellular carcinoma (HCC). Sirtuins (SIRTs), classified as a group of NAD+-dependent deacetylases, serve as crucial modulators of a wide spectrum of cellular functions, including metabolic pathways, the inflammatory response, and the process of senescence. Their subcellular localization and diverse functions link them to various health conditions, including NAFLD and cancer. Concurrently, the gut microbiota, comprising diverse microorganisms, significantly influences host metabolism and immune responses. Recent findings indicate that sirtuins modulate gut microbiota composition and function, while the microbiota can affect sirtuin activity. This bidirectional relationship is particularly relevant in metabolic disorders, where dysbiosis contributes to disease progression. The review highlights recent findings on the roles of specific sirtuins in maintaining gut health and their implications in metabolic dysfunction and HCC development. Understanding these interactions offers potential therapeutic avenues for managing diseases linked to metabolic dysregulation and liver pathology.

Keywords: 
;  ;  ;  ;  

1. Background

Sirtuins (SIRTs), categorized as class III histone deacetylases (HDACs), have emerged as a crucial regulators in numerous cellular mechanisms, particularly those related to aging, metabolism, and disease [1,2]. These NAD+-dependent enzymes serve a crucial function in the preservation of cellular homeostasis by deacetylating histones, transcription factors, and other proteins, thus influencing gene expression and metabolic pathways. Research has increasingly highlighted the significance of sirtuins in health conditions ranging from metabolic dysfunction to cancer, particularly hepatocellular carcinoma (HCC) [3].
The gut microbiota, which harbors an intricate consortium of microorganisms that inhabit the intestinal tract, has also been recognized for its profound impact on host health [4]. Emerging evidence indicates that sirtuins and gut microbiota significantly influence metabolic health and disease outcomes [4]. Sirtuins affect gut microbiota by enhancing gut barrier integrity and immune modulation. Conversely, gut microbiota-derived metabolites can impact sirtuin activity, creating a dynamic feedback loop that affects overall health [5].
The intricate dynamics between sirtuins and gut microbiota are pivotal in the study of NAFLD and its development into HCC [3]. As the prevalence of NAFLD increases, it is imperative to elucidate its molecular pathogenesis for the advancement of effective therapeutic strategies [6]. This review explores the correlation between sirtuins and gut microbiota in the context of health and disease, elucidating their roles in metabolic dysfunction and the progression to liver cancer. By integrating insights from recent research, we aim to highlight potential therapeutic interventions targeting these pathways to improve health outcomes. Additionally, we propose future research directions to investigate sirtuin-microbiota interactions, paving the way for innovative strategies in preventing and treating metabolic disorders and liver cancer.

2. Overview of Sirtuins and Their Biological Functions

Sirtuins Classification and Subcellular Localization

Sirtuins (SIRTs), a group of proteins known as class III HDACs, play a pivotal in various cellular processes and have a significant impact in aging research and related diseases. [1,2]. They catalyse NAD+ dependent deacetylation reaction linking them with the metabolic regulation. Structurally, sirtuins conatin a N-terminal, C-terminal and Zinc binding domain. SIRT1-7 have a structurally conserved catalytic core bearing two bi-lobed globular domains comprising of 275 amino acid residues harbouring NAD+ as a cofactor. Isoform specific N and C-terminal domains are variable in their length, chemical composition and susceptibility to post-translational modifications (PTM) subsequently regulating its localization and regulation [7].
Phylogenetic analysis based on the amino acid sequence revealed four different classes of sirtuins. SIRT1-3 belong to Class I, SIRT-4 to Class II, SIRT 5 to Class III, SIRT6-7 to Class IV [7]. Each protein exhibits distinct subcellular compartmentalization [8]. SIRT1, 6, and 7 predominantly reside within the nucleus. However, SIRT1 can translocate to the cytoplasm under physiological or pathological stimuli [9]. SIRT2, on the other hand, is primarily situated within the cytoplasm but is capable of translocating to the nucleus under certain conditions to influence cellular processes [10]. Additionally, SIRT3 can also undergo relocation between the mitochondria and the nucleus, enabling it, to regulate a wide range of cellular processes [11]. while SIRT 4, and 5 are typically located in the mitochondrial compartment [7,12]. These dynamic subcellular distributions and trafficking abilities allow the sirtuins to coordinate their regulatory functions across different compartments of the cell [13]. SIRTs play a key regulatory role in metabolism, health-span, and longevity by deacetylating histones, transcriptional regulators, and other proteins, in addition to their ADP-ribosyltransferase and deacytlase activities [2,14].
Table 1. SIRT1, SIRT2, and SIRT3 function as deacetylases, primarily localized in the nucleus, cytoplasm, and mitochondria, respectively. SIRT4 acts as an ADP-ribosyltransferase and is also localized to the mitochondria. SIRT5 possesses both deacetylase and deacylase activities within the mitochondrial compartment. SIRT6 operates as both a deacetylase and an ADP-ribosyltransferase, predominantly found in the nucleolus, while SIRT7 is identified as a nuclear deacetylase.
Figure 1. Subcellular Localizations and Catalytic Functions of Human Sirtuins.
Figure 1. Subcellular Localizations and Catalytic Functions of Human Sirtuins.
Preprints 147863 g001

1.2. Role of Sirtuins in Cellular Homeostasis and Metabolism

SIRTs are involved in post-translational modifications, particularly protein deacetylation, and contribute to gene expression regulation, DNA repair, and cellular senescence [15]. These enzymes enhance genomic stability through chromatin structural modulation and involvement in various DNA repair mechanisms, including base excision repair, nucleotide excision repair, and double-strand break repair [16,17]. SIRTs also serve a crucial role in the modification of histones and proteins, influencing cellular metabolism, mitochondrial function, and stem cell maintenance [18,19]. SIRT1, the most widely studied sirtuin, is recognized for its role in transcriptional regulation, genomic silencing, and epigenetic elements within the nucleus, while also contributing to metabolism and nutrient perception in the cytosol [20]. The involvement of SIRTs in aging and cancer has been extensively studied, with different sirtuin family members showing diverse effects on stem cells and cancer cells [21,22].
SIRTs play vital roles in various physiological and pathological conditions, such as neurodegenerative diseases [23], kidney disorders [24], and cardiovascular diseases [8]. They are important for maintaining cellular integrity [25] and regulating metabolic balance, including glucose and lipid metabolism [26]. Additionally, they modulate mitochondrial activity and have been associated with various metabolic disorders [8]. SIRTs also contribute to the regulation of female reproductive processes [27] and influence women's health, particularly in ovarian function and cancer development [28]. In the immune system, SIRTs modulate T cell metabolism and function, making them promising therapeutic targets for immune-related diseases [29]. Furthermore, SIRTs have been implicated in counteracting several hallmarks of aging, potentially contributing to healthy longevity [30].
Different investigations underscore the crucial and multifaceted functions of sirtuins in neoplastic advancement, acting as tumor suppressors as well as activators of tumorigenesis, depending on the specific cellular environment.[31,32]. SIRTs regulate various cancer-related processes, including cell viability, apoptosis, metastasis, and metabolism [31,33]. Among all SIRTs, SIRT1 has been extensively researched for its dual role in cancer, contributing to tumor suppression and promotion [34,35]. SIRT2's role remains controversial, with evidence suggesting it can act as either an oncogene or a tumor suppressor across multiple malignancies [36]. The roles of other sirtuins (SIRT3-7) vary across different cancer types, influencing processes such as proliferation, invasion, and chemoresistance [37,38,39]. Furthermore, SIRTs play crucial roles in hepatocellular carcinoma (HCC) development and progression [3,40]. SIRT1 is overexpressed in HCC, promoting oncogenesis and multidrug resistance [41]. Collectively, these findings emphasize sirtuins' vital role in cellular homeostasis and disease management, particularly in cancer, necessitating further investigation into their specific functions and therapeutic implications.
Table 1. Classification and Functional Roles of Sirtuins in Metabolism and Cellular Processes.
Table 1. Classification and Functional Roles of Sirtuins in Metabolism and Cellular Processes.
Sirtuin Class Type of Activity Acyl Substrates Cellular Function Target substrates Metabolic role Biological role
SIRT1 I Strong deacytylase activity Remove acetyl and long chain fatty acyl group from Lysine Formation of facultative chromatin, Mitochondrial biogenesis, p53, FOXO1/3, NF-κB, CRTC2, PGAM-1, PGC1α, SREBP, LXR, FXR, LKB1
Fatty acid oxidation, Regulation of cholesterol and bile acid homeostasis Cell survival and lipid metabolism
SIRT2 I Both deacetylase and mono-ADP-ribosyl transferase activity
Remove of acetyl, long-chain fatty acyl, 4-oxononanoyl, and benzoyl groups
Cell cycle regulation, Tumor suppression/promotion
Neurodegeneration
 
α-Tubulin, FOXO1, FOXO3, p300
Promotion of lipolysis in adipocytes
Regulation of cell cycle and cell motility
 
SIRT3 I Both deacetylase and mono-ADP-ribosyl transferase activity
Remove
acetyl and long-chain fatty acyl groups from lysine
Regulation of mitochondrial activity
Protection against oxidative stress
Tumor suppression
LCAD, ACS2, SOD2, IDH2, HMGCS, OTC, SOD2, subunits of the electron transport chain and ATP synthase
Metabolism and thermogenesis
SIRT4 Class II Mono-ADP-ribosyl transferase activity
Remove
lipoyl, biotinyl, methylglutaryl, hydroxymethylglutaryl, and 3-methylglutaconyl groups
Tumor suppression
IDE, ANT2, ANT3, GDH, MCD, PDH
Glucose metabolism
Amino acid catabolism

Glucose metabolism and Insulin secretion
SIRT5 Class III Weak deacetylase activity
Removes charged malonyl, succinyl, and glutaryl groups
CPS1, UOX
Urea cycle
Fatty acid metabolism
Amino acid metabolism
Cellular energy Metabolism
SIRT6 Class IV Mono-ADP-ribosyl transferase activity
Remove acetyl and long-chain fatty acyl groups
Genomic stability/DNA repair
HIF1α, PARP1, TNFα, GCN5
Glucose and lipid metabolism
Inflammation
DNA repair/Glucose homeostasis
SIRT7 Class IV Mono-ADP-ribosyl transferase activity
Remove acetyl groups
Ribosome biogenesis
Tumor promotion
RNA polymerase 1

Metabolism, rDNA transcription

3. The Role of Gut Microbiota in Host Health and Disease

The gut microbiota includes a variety of microorganisms, such as bacteria, archaea, fungi, viruses, and parasites [42,43,44]. These microorganisms, inhabit within the gastrointestinal tract—particularly in the intestinal—are commonly designated as gut flora or gut microbiota [45]. While bacteria dominate the gut microbiome, the significance of other microbes—often referred to as the "dark matter" of microbiomes—has become increasingly recognized [43]. The predominant bacterial phyla present within the human gut microbiome include Firmicutes, Bacteroidetes, Proteobacteria, Actinobacteria, and Fusobacteria [46]. Furthermore, this complex community plays essential roles in host physiology, metabolism, and immune function [47,48]. The gut microbiota helps maintain intestinal integrity through the production of microbial metabolites and anti-microbial peptides modulating the immunity [49]. Additionally, it serves as a potential source of novel antimicrobials, which could help address antimicrobial resistance [50]. Moreover, the gut microbiota contributes significantly to digestion by breaking down indigestible dietary components and producing essential nutrients like vitamins and enzymes [51]. Furthermore, the microbiota plays a critical role in modulating inflammation, cancer-related processes, and oxidative stress through the production of metabolites, especially short-chain fatty acids (SCFAs) and tryptophan catabolites [52,53,54,55].
Gut microbiota are essential for the fermentation of non-digestible carbohydrates, yielding SCFAs such as acetate, propionate, and butyrate [56,57]. These SCFAs serve as energy sources for colonocytes and maintain gut barrier integrity [58,59]. Moreover, SCFAs influence host metabolic processes and immune responses via diverse mechanisms, such as the stimulation of G-protein-coupled receptors and the suppression of histone deacetylases [60]. The gut microbiota is also participates in bile acid metabolism, which is essential for lipid digestion and absorption [61]. Consequently, microbial SCFA production and bile acid metabolism significantly impact host health via intricate interactions with the gut epithelium, immune response, and metabolic pathways [62,63]. Grasping these mechanisms is essential for developing approaches to enhance gut health and avoid metabolic diseases.
Figure 2. The Role of Gut Microbiota in SCFAs Production and Metabolic Health.
Figure 2. The Role of Gut Microbiota in SCFAs Production and Metabolic Health.
Preprints 147863 g002
Gut microbiota produce SCFAs via the fermentation of indigestible carbohydrates. These organic acids, typically containing fewer than six carbon atoms, play essential roles in energy metabolism and the synthesis of vital molecules, significantly contributing to energy homeostasis and overall metabolic health.
The composition of gut microbiota is modulated by factors including diet, genetics, and environmental conditions [64,65,66]. Diet significantly influences the microbiome, alongside seasonal and geographical factors impacting dietary habits and microbial composition [67]. Environmental determinants, such as communal residences, exert a more significant influence on microbiome composition than host genetics [68]. Nevertheless, host genetics continue to exert an influence in ascertaining microbiome composition, especially via immune system-related genes [69]. Early life factors, including birth mode, feeding methods, and antibiotic use, also contribute to microbiota development [70]. The gut microbiome is dynamic, changing throughout an individual's lifetime due to factors like age, BMI, exercise, and lifestyle [66]. Recognizing these influences is important for maintaining a healthy microbiome and preventing dysbiosis, which has been linked to several diseases [65,71].
Dysbiosis, characterized by a perturbation in the assemblage of gastrointestinal microbiota, has been associated with numerous pathological states, encompassing inflammatory bowel disease (IBD), cardiovascular diseases, diabetes mellitus, obesity, and various neurological disorders [72,73,74,75,76]. This dysbiosis frequently presents as a diminution in microbial variety alongside an elevation in particular bacterial taxa [75]. Factors contributing to dysbiosis include antibiotic use, diet, and environmental stressors [74,75]. Dysbiosis can lead to altered microbial metabolite production, immune dysregulation, and chronic inflammation [77]. Therapeutic modalities aimed at addressing dysbiosis encompass fecal microbiota transplantation, the administration of probiotics, the utilization of prebiotics, as well as various dietary interventions [78,79].
Figure 3. Determinants of Gut Microbiome Composition.
Figure 3. Determinants of Gut Microbiome Composition.
Preprints 147863 g003
The gut microbiota composition is affected by a wide range of factors. Some elements, such as genetic background, delivery method, early infant feeding practices, and maternal health during pregnancy, are established early in life and tend to remain stable (A). In contrast, factors like diet, medication use, exercise habits, age, and stress levels are more variable and can be modified throughout life (B). Furthermore, environmental influences, including chronic viral infections, living conditions, and geographical factors, also contribute to microbiome composition, although these may be more challenging to alter (C).

4. Interrelationship Between Sirtuins and Gut Microbiota: A Bidirectional Perspective

4.1. Influence of Sirtuins on Gut Microbiota Composition

Sirtuins are essential in gut function regulation, particularly in preserving the intestinal barrier and mucosal immune mechanism, which are vital for regulating intestinal microbiota composition [80].
SIRT1
Intestinal epithelial SIRT1 regulates gut microbial composition consequently preventing age related intestinal inflammation[81]. The gut microbiome remotely regulates the expression of miR-204 subsequently impairing the endothelial function by targeting Sirt1[82]. SIRT1 deficiency in the intestinal epithelium leads to increased fecal bile acid levels, reduced Lactobacillus abundance, and heightened susceptibility to intestinal inflammation and colitis [83]. Moreover, the gut microbiome modulates systemic hydrogen sulphide levels impacting SIRT1 activation followed by regulation of neurinflammation [84].
SIRT2
Similarly, SIRT2 deficiency enhances the progression of NAFLD by altering gut microbiota composition and inducing metabolic disorders [6]. whereas inhibiting SIRT2 may improve gut barrier integrity and protect against colitis [85]. SIRT2 knockout mouse displayed increased susceptibility to obesity, liver injury and metabolic dysfunction when placed on high fat/high sucrose/high cholesterol diet [6].
SIRT3
SIRT3 and the gut microbiota interaction leads to modulation of key cellular processes like mitochondrial function, inflammation and energy metabolism[86]. SIRT3 deficiency promotes NAFLD progression through gut microbial dysbiosis and impaired intestinal permeability [87]. Moreover, the gut microbiota modulates hydrogen sulphide levels consequently affecting SIRT3 activation [84].
SIRT4
Mitochondrial SIRT4 regulates intestinal metabolism and homeostasis. It regulates lysozyme expression in the gut, influencing microbiota composition [88]. Loss of SIRT4 can lead to dysregulated glutamine and nucleotide metabolism in intestinal adenomas [89].
SIRT5 -7
While the roles of SIRT5, SIRT6, and SIRT7 in gut microbiota regulation are not directly established, it is notable that downregulation of SIRT5 has been linked to increased bile acid production, which may contribute to an immunosuppressive tumor microenvironment and facilitate hepatocellular carcinoma (HCC) development [90]. Moreover, SIRT6 knockout mice exhibit premature aging associated with gut dysbiosis, a condition reversible through fecal microbiota transplantation or a high-fat diet [91]. It is noteworthy that the presence of an inflammatory response of the colorectal mucosa is associated with higher concentrations of SIRT7 and lower concentrations of SIRT1 [92].
Together, these roles underscore the importance of sirtuins in gut health and their potential influence on microbiota dynamics, highlighting areas for future research.
Table 2. Roles of Sirtuins in Gut Health: Impact on Barrier Integrity, Inflammation, and Microbial Diversity.
Table 2. Roles of Sirtuins in Gut Health: Impact on Barrier Integrity, Inflammation, and Microbial Diversity.
Sirtuin Roles of Sirtuins in Gut Health Ref.
SIRT1 Maintains intestinal epithelial barrier integrity, regulates inflammation, and modulates autophagy, potentially influencing gut microbiota composition and diversity. [81,93,94,95]
SIRT2 Regulates intestinal epithelial cell proliferation and differentiation, impacting the gut environment and reducing inflammation, facilitating better host-microbiota interactions. [85,96,97]
SIRT3 Enhances mitochondrial function in intestinal cells, regulates oxidative stress, and maintains gut barrier homeostasis; deficiency leads to microbial dysbiosis and impaired permeability. [84,98,99]
SIRT4 Modulates amino acid metabolism in intestinal cells, potentially influencing nutrient availability for gut microbiota. [89,100]
SIRT5 Regulates cellular homeostasis and various metabolic pathways in intestinal cells, potentially influencing nutrient availability for gut microbiota. [101,102]
SIRT6 Maintains intestinal epithelial barrier integrity, mitigates inflammation, and enhances favourable immune responses; may affect gut microbiota composition and diversity. [103,104,105]
SIRT7 Maintains intestinal homeostasis and modulates inflammation; potentially affecting gut microbiota composition. [92,106]

4.2. Impact of Gut Microbiota on Sirtuin Activity

Conversely, gut microbiota also influences sirtuin activity and expression. Studies indicate that gut microbiota can regulate the expression of sirtuins, along with senescence-regulating miRNAs and mitochondrial DNA, all associated with overall well-being [107]. Moreover, gut microbiota interacts with sirtuin-activating compounds to influence molecular pathways that counteract aging and inflammation, while enhancing specific gut microbiota groups to improve immune function [108].
Studies have elucidated that gastrointestinal microbiota may influence critical transcriptional co-activators, transcription factors, and enzymatic pathways pertinent to mitochondrial biogenesis, encompassing genes such as PGC-1α, SIRT1, and AMPK [109]. Additionally, the gut microbiome regulates vascular microRNA-204, which targets SIRT1 and affects endothelial function [82]. Furthermore, metabolites produced by gut microbiota from fermenting indigestible food components can impact sirtuin function [110]. For instance, gut microbiota-derived metabolites have anti-inflammatory and antioxidative properties that influence sirtuin activity [111]. Particularly, Urolithin A (UroA), a microbial metabolite derived from polyphenolics, exemplifies this by exhibiting anti-inflammatory and antioxidative effects [111]. These properties of microbial metabolites can directly affect sirtuin function, as sirtuins are involved in regulating oxidative stress and inflammation [112]. By modulating these pathways, gut microbiota-derived metabolites can indirectly affect sirtuin activity and contribute to overall host health.
Additionally, the gut microbiome possesses the capacity to affect sirtuin-associated pathways; for example, Saccharomyces boulardii has been demonstrated to alter necrotizing enterocolitis through the modulation of the SIRT1/NF-κB signaling pathway and the intestinal microbiota [113]. The overall outcomes shed light on the intricate relationship between sirtuins and gut microbiota, stressing the requirement for more extensive investigation to clarify current shortcomings in understanding their roles and interconnections.

5. Role of Sirtuins and Gut Microbiota in Non-Alcoholic Fatty Liver Disease (NAFLD)

NAFLD represents the most widespread form of chronic hepatic disorder globally and is projected to emerge as the primary etiology for liver transplantation by the year 2030 [114]. Furthermore, NAFLD is distinguished by an atypical aggregation of hepatic lipids., concomitant with insulin resistance, and demonstrable steatosis, while systematically ruling out secondary etiologies of hepatic steatosis, such as alcohol consumption [115]. Additionally, NAFLD comprises a spectrum of disorders extending from hepatic steatosis to steatohepatitis, leading to inflammation, hepatocirrhosis, hepatocellular carcinoma, and mortality [116]. Also, is closely associated with insulin resistance and may function as both a cause and a result of metabolic syndrome [117].
Formerly, the "two-hit" hypothesis previously explained NAFLD pathogenesis, identifying lipid accumulation as the "first hit." This initial accumulation of lipid predisposes the liver to further injury, termed the "second hit," resulting in inflammation and fibrosis [118]. Recent research proposes the multiple-hit hypothesis, which more comprehensively describes the molecular and metabolic alterations in NAFLD. This updated framework includes interconnected mechanisms such as insulin resistance, lipotoxicity, innate immune activation, and gut microbiome effects, influenced by genetic (PNPLA3) and dietary elements (saturated fat and fructose) [118].
The interplay between sirtuins and intestinal microbiota is a complex relationship. The gut microbiota plays a crucial role in host metabolism by breaking down nutrients and producing metabolites that influence metabolic processes and modulate immunity [119]. Sirtuins are recognized for their ability to govern the intestinal microbiome, thereby suggesting their participation in the pathophysiological mechanisms underlying a variety of diseases. They are integral to numerous physiological processes, encompassing glucose and lipid metabolism, insulin resistance, and mitochondrial function, thereby rendering them pivotal factors in the etiology of conditions such as type 2 diabetes, obesity, and NAFLD [120,121,122]. Furthermore, sirtuins influence essential metabolic hormones such as leptin, ghrelin, melatonin, and serotonin, which perform a crucial function in the regulation of gastrointestinal homeostasis [123].
Recent studies indicate a significant reduction of sirtuin levels in NAFLD patients. Wu et al. found decreased expression of SIRT1, SIRT3, SIRT5, and SIRT6 in NAFLD patients, alongside increased lipogenic gene expression and SIRT4 [124]. Furthermore, Bruce et al. demonstrated that excess dietary fat exposure during early and postnatal periods elevates NASH risk in adulthood, particularly affecting sirtuin levels. Offspring on a high-fat diet exhibited NAFLD, with those from high-fat diet mothers developing NASH, characterized by decreased NAD+/NADH and lower SIRT1 and SIRT3 levels, coupled with increased lipid metabolism gene expression [125].
The gut microbiota is essential for NAFLD development through dietary metabolism, yielding vital nutrients and energy [126,127,128]. Its composition is diet-dependent, with high-fat/high-cholesterol (HFC) and high-fat/high-sucrose (HFS) diets causing dysbiosis linked to NAFLD [129,130,131]. This dysbiosis increases intestinal permeability, exposing the liver to bacterial products through the portal vein and inducing metabolic endotoxemia, thereby disrupting the gut-liver axis [132,133]. NAFLD microbiota differs from that of healthy individuals and is influenced by genetic factors related to metabolic syndrome [134,135]. Additionally, gastrointestinal microbiomes can synthesize lipopolysaccharides (LPS), which may infiltrate the circulatory system and impair the hepatic tissue when the intestinal barrier is compromised (Figure 4) [131,136]. In contrast, SCFAs such as butyrate are crucial for maintaining gut barrier integrity. Zhou et al. demonstrated that sodium butyrate enhances gut microbiota and fortifies the intestinal barrier, mitigating LPS translocation and reducing steatohepatitis in mice [137]. This underscores the significance of a robust gut barrier.
Chen et al. demonstrated that SIRT3 deficiency worsens NAFLD from a high-fat diet through impaired intestinal permeability linked to gut microbial dysbiosis in SIRT3 knockout mice. The SIRT3KO mice exhibited increased Oscillibacter, Parabacteroides, and Mucispirillum, while Alloprevotella decreased [98]. Prior studies have shown a negative correlation between Oscillibacter levels and the mRNA expression of zonula occludens-1, a protein relevant to gut permeability [138,139,140]. Furthermore, Everard et al. noted increased Mucispirillum and Parabacteroides in humans and mice on a high-fat diet [141]. The elevation of Oscillibacter, Mucispirillum, and Parabacteroides correlates with exacerbated HFD-induced NAFLD in SIRT3KO mice, characterized by diminished tight junction protein expression like ZO-1 and claudins [98]. Thus, SIRT3-mediated intestinal barrier dysfunction, coupled with LPS release from gut microbiome alterations, facilitates NAFLD progression.
This figure depicts events following gut dysbiosis, leading to LPS release. These LPS molecules reach the liver via the portal vein, activating macrophages through Toll-like receptor 4 (TLR4). This activation induces proinflammatory cytokine release, including TNFα, IL-1β, and IL-6, resulting in heightened inflammation and hepatic insulin resistance. Additionally, activated macrophages infiltrate adipose tissue, worsening inflammation and insulin resistance, thereby contributing to metabolic disorders, including obesity, diabetes, and NAFLD.

6. Role of Sirtuins and Gut Microbiota in Hepatocellular Carcinoma (HCC)

Liver cancer represents a considerable global health issue, with rising rates primarily attributed to hepatocellular carcinoma (HCC), which constitutes about 90% of cases [142,143]. Significant risk factors for HCC development include chronic hepatitis B and C virus infections. Non-viral factors involve environmental carcinogens such as aflatoxin B1, alcohol misuse, and genetic conditions like hemochromatosis and Wilson disease [144]. Importantly, non-alcoholic steatohepatitis (NASH) has emerged as a critical criterion for liver transplantation among HCC patients in the United States [145]. The transition from NAFLD to HCC entails a multifaceted interaction of metabolic variables, inflammation, gut microbiota imbalance, oxidative stress, and aberrant lipid metabolism [146,147,148]. Dysbiosis, has been implicated in the pathogenesis of HCC and its related chronic diseases, which encompass chronic hepatitis B and C, alcoholic liver disease, NAFLD, and NASH [149].
SIRT1 has a critical role in HCC by virtue of promoting tumorigenicity, metastasis, chemoresistance, and heralding a poor prognosis [150,151,152]. The interplay between SIRT1 and intestinal microbiota is complex. Butyrate, a short-chain fatty acid from gut bacteria, is crucial in this relationship. A decrease in butyrate-producing bacteria may harm the intestinal mucosa, possibly leading to HCC development [153]. Pant et al. illustrated that butyrate induces microRNA-22 (miR-22), which downregulates SIRT1 in a concentration-dependent manner, increasing reactive oxygen species (ROS) and apoptosis in hepatic cells exposed to sodium butyrate [154]. MiR-22's downregulation in human liver cancer cells correlates with enhanced tumorigenicity and cellular proliferation [155]. It is implicated in HCC development via the miR-29a-SIRT1-WNT/β-catenin pathway [156]. Moreover, overexpression of SIRT1 promotes HCC proliferation and resistance to chemotherapy by promoting autophagy [157,158]. SIRT1 protects mitochondrial function of HCC cells by suppressing the expression of hypoxia-induced factor-1 alpha expression and also promotes stem-cell like features in HCC cells[159]. SIRT1 also promotes HCC metastasis by enhancing PGC-1α-mediated mitochondrial biogenesis[160]. Conversely, Herranz et al. reported that SIRT1 overexpression can protect transgenic animals from diethylnitrosamine/high-fat diet-induced liver cancer by mitigating NF-kB-mediated inflammation and averting malignant transformation [161]. This observation contrasts with earlier assertions of SIRT1's role in promoting HCC and other studies indicating elevated SIRT1 in human HCC samples [162,163]. Malignant cells may utilize survival strategies typically reserved for non-malignant cells [164]. Portmann et al. have demonstrated that inhibiting SIRT1 leads to impaired tumor growth both in vivo and in vitro and this supports that notion that SIRT1 activity in healthy hepatocytes protects against cancer, but after transformation, SIRT1 becomes a protective force for the tumor cells as a survival advantage[165]. However, additional research is essential to elucidate the intricate relationship between butyrate and SIRT1 in the etiology of HCC.
SIRT2 mediates the deacetylation and activation of protein kinase B, impacting the glycogen synthase kinase-3β/β-catenin signaling pathway, which is implicated in epithelial-mesenchymal transition (EMT) [166]. Huang et al. support the tumor-promoting role of SIRT2, revealing that its downregulation hinders energy metabolism and invasion in HCC cells [167]. Further investigation is warranted to elucidate SIRT2's specific functions in HCC.
SIRT7 plays a multifaceted role in HCC, with its upregulation noted in numerous patients [168]. It facilitates HCC cell proliferation through ERK1/2 phosphorylation and activation of the RAF/MEK/ERK signaling cascade, promoting tumor growth [169]. Moreover, SIRT7 boosts HCC cell proliferation by inhibiting MST1 and modulating the Hippo/YAP pathway, resulting in enhanced YAP activation [170].
In summary, the interplay between sirtuins and gut microbiota is essential for elucidating the pathogenesis of NAFLD and HCC. Table (3) highlights the microbial changes linked to these diseases, indicating opportunities for specific therapeutic interventions. Subsequent investigations should aim to utilize these findings for the formulation of microbiota-targeted therapies to improve disease management and patient prognosis.
Table 3. Microbiota Composition Changes in NAFLD and HCC: A Summary of Human Studies.
Table 3. Microbiota Composition Changes in NAFLD and HCC: A Summary of Human Studies.
Disease Composition Change References
Increase Decrease
NAFLD Streptococcus, Megasphaera,
Enterobacteriaceae,
Streptococcus, Gallibacterium
Bacillus and Lactococcus, Pseudomonas, Faecalibacterium prausnitzii, Catenibacterium, Rikenellaceae, Mogibacterium, Peptostreptococcaceae [171]
Firmicutes (Streptococcus mitis and Roseburia inulinivorans) and Bacteroidetes (Barnesiella intestinihominis and Bacteroides uniformis) Bacteroidetes (Prevotella sp.CAG 520, Prevotella sp. AM42 24, Butyricimonas
virosa, and Odoribacter
splanchnicus), Proteobacteria
(Escherichia coli),
Lentisphaerae (Victivallis
vadensis), and Firmicutes
(Holdemanella biformis,
Dorea longicatena, Allisonella
histaminiformans, and
Blautia obeum)
[172]
Bacteroidetes, Proteobacteria, Bacteroides, Alistipes, Verrucomicrobia, Faecalibaculum, Helicobacter, Epsilonbacteraeota Muribaculaceae, Lactobacillus [173]
HCC Escherichia coli [174]
Proteobacteria, Desulfococcus, Enterobacter, Prevotella, Veillonella Cetobacterium [175]
Bacteroides Akkermansia, Bifidobacterium [176]
Neisseria, Enterobacteriaceae, Veillonella, Limnobacter Enterococcus, Phyllobacterium, lostridium, Ruminococcus, Coprococcus [177]
Proteobacteria, Enterobacteriaceae, Bacteroides xylanisolvens, B. caecimuris, Ruminococcus gnavus, Clostridium bolteae, Veillonella parvula Erysipelotrichaceae, Oscillospiraceae [178]

Klebsiella, Haemophilus
Alistipes, Phascolarctobacterium, Ruminococcus [179]

7. Interventions Targeting Sirtuins and Gut Microbiota

SIRTs are pivotal in linking health and disease, highlighting their potential for targeting interventions [180]. The healthcare sector increasingly recognizes the therapeutic value of manipulating the sirtuin pathway and gut microbiome in disease management [181]. SIRT1 and SIRT2, have been identified as potential targets for therapeutic interventions in various gut dysbiosis-mediated diseases, including cancer, neurodegenerative diseases, and metabolic disorders [182,183]. Studies have been limited in demonstrating the pharmacological utility of modifying other sirtuins in the context of disease where gut dysbiosis contributes to the underlying mechanisms.

a. Sirtuin Activators

Resveratrol

One prominent example of therapeutic intervention is the targeted activation of the SIRT1 pathway [184]. Resveratrol, a polyphenolic compound, has demonstrated efficacy in mitigating the progression of NAFLD in both preclinical and clinical contexts [185]. Resveratrol enhances gut microbiota by promoting the proliferation of beneficial bacteria, thereby improving gut health. This effect is characterized by a decrease in harmful bacteria and an increase in short-chain fatty acid (SCFA)-producing bacteria, which contributes to overall gut microbiota balance and metabolic health [186]. Furthermore, resveratrol alleviates NAFLD by strengthening gut barrier integrity, reducing inflammation, and increasing the production of short-chain fatty acids (SCFAs) [187,188]. Additionally, it exhibits potential protective effects against hepatocellular carcinoma through the modulation of inflammatory, angiogenic, and oxidative stress pathways [189]. Resveratrol's proposed mechanism of action involves the modulation of hepatic lipid metabolism and the reduction of oxidative stress, primarily through the activation of the AMPKα/SIRT1 signalling pathway. This activation effectively suppresses the nuclear factor kappa B (NF-κB) inflammatory pathway, resulting in decreased inflammation and reduced hepatic steatosis [190,191]. Figure 5.
This figure illustrates the multifaceted mechanisms through which resveratrol mitigates the progression of non-alcoholic fatty liver disease (NAFLD) and improves liver health. Key processes include the enhancement of gut microbiota by promoting beneficial bacterial proliferation, which strengthens gut barrier integrity and overall gut health. Additionally, resveratrol activates the AMPKα/SIRT1 signaling pathway, suppressing the NF-κB inflammatory pathway and leading to decreased inflammation and hepatic steatosis. Furthermore, resveratrol influences hepatic lipid metabolism and reduces lipid accumulation by increasing the production of short-chain fatty acids (SCFAs), which enhances AMPK activation and contributes to decreased lipid accumulation.

Pterostilbene

Pterostilbene, a dimethyl ether variant of resveratrol, exhibits encouraging effects against NAFLD and obesity. It diminishes liver fat accumulation by influencing the miR-34a/Sirt1/SREBP-1 pathway in rats fed a fructose diet [192]. Compared to resveratrol, pterostilbene has enhanced bioavailability and metabolic stability[193]. In rats consuming a high-calorie diet, pterostilbene lowers adipose tissue volume, inhibits lipogenesis in fat tissue, and promotes fatty acid oxidation in the liver [194].From a metabolic perspective, pterostilbene displays greater stability than resveratrol and often shows more potent pharmacological effects [195]. Taken together, these studies indicate that pterostilbene is a promising candidate for addressing NAFLD and obesity, with potential benefits over resveratrol due to its superior bioavailability and metabolic characteristics.

E1231

E1231, treatment activates SIRT1alleviating NAFLD by regulating lipid metabolism. Moreover, E1231 prevented lipid accumulation and improved mitochondrial function in free fatty acid challenged hepatocytes. E1231 prevented liver injury via regulation of SIRT1 and AMPK-α pathway [196].

Quercetin

Quercetin, a natural flavonoid present in plants, has demonstrated encouraging effects in the treatment of NAFLD. Laboratory studies indicate that quercetin diminishes lipid buildup, lowers inflammatory cytokines, and boosts antioxidant activity in liver cells [197]. Animal studies show that quercetin helps improve NAFLD by triggering AMPK-mediated mitophagy, reducing lipid storage, and alleviating oxidative stress [198] Supplementing the diet with quercetin in gerbils suffering from high-fat diet-induced NASH led to better lipid profiles, a decline in inflammatory markers, and regulation of Sirt1 and NF-κB p65 expression[199]. Thus, the hepatoprotective properties of quercetin are linked to enhanced fatty acid metabolism, anti-inflammatory and antioxidant effects, and modulation of gut microbiota as well as bile acids [200].These results emphasize the potential of quercetin as a therapeutic option for NAFLD.

Nicotinamide Riboside (NR)

Nicotinamide riboside (NR), an NAD+ precursor, shows promise in addressing NAFLD and its progression to HCC. NR supplementation reduces hepatic lipid accumulation, inflammation, and fibrosis in various NAFLD models [201,202,203]. It activates SIRT1/AMPK-mediated browning of white adipose tissue and modulates gut microbiota, potentially improving lipid metabolism [204].

Berberine

Berberine, a natural plant alkaloid, has emerged as a promising therapeutic agent for NAFLD and its progression to HCC [205]. It exerts beneficial effects on gut microbiota by promoting the growth of beneficial bacterial populations while reducing pathogenic strains [206,207]. Additionally, berberine enhances lipid metabolism, improves insulin sensitivity, and diminishes inflammation [205,208]. Berberine has beneficial effects on NAFLD through various molecular pathways, including the activation of SIRT3, SIRT1, AMPK, and PPAR-γ, as well as the suppression of the NLRP3 pathway [208]. Additionally, berberine activates intestinal farnesoid X receptor (FXR), resulting in increased expression of fibroblast growth factor 15 (FGF15), which in turn inhibits lipogenesis and the activation of NF-κB pathway in the liver [207]. Furthermore, berberine suppresses the p38 MAPK/ERK-COX2 signaling pathways, thereby reducing inflammation and angiogenesis associated with non-alcoholic steatohepatitis (NASH) and HCC [209].

Yinchen Linggui Zhugan Decoction (YLZD)

Yinchen Linggui Zhugan decoction (YLZD), a traditional Chinese medicine, has demonstrated promise in treating NAFLD by modulating the SIRT1/Nrf2 pathway and gut microbiota. In rat models, YLZD treatment has been shown to reduce NAFLD induced by a high-fat diet, increasing serum and fecal butyric acid levels and total SCFAs, while promoting a favorable shift in gut microbiota composition towards SCFA-producing bacteria [210].

The Tangshen Formula (TSF)

The Tangshen formula (TSF), an herbal medicine from China, has exhibited encouraging effects in the management of NAFLD. Research indicates that TSF reduces hepatic steatosis and enhances lipid metabolism in several animal models[211,212,213]). Its mechanism of action involves various pathways, such as the modulation of gut microbiota and metabolic profiles [211], the activation of autophagy via the AMPK/SIRT1 pathway [212] and the regulation of macrophage activation and their phenotypic changes [213]. TSF treatment has been found to decrease lipid accumulation, lessen inflammation, and enhance insulin resistance and the integrity of the intestinal barrier [211,212]. Collectively, these findings suggest that TSF is a promising therapeutic option for NAFLD, functioning as a modulator of gut microbiota and metabolic profiles while also affecting hepatic cellular processes to reduce steatosis and related metabolic issues.

Curcumin

Curcumin, a natural polyphenol, appears to be a promising candidate for the treatment of NAFLD. Research indicates that curcumin supplementation can lower liver fat levels, enhance lipid profiles, and reduce insulin resistance in patients with NAFLD[214]. The beneficial effects of curcumin are linked to its properties as an antioxidant, anti-inflammatory agent, and its ability to prevent fat accumulation [215]. From a mechanistic standpoint, curcumin inhibits the O-GlcNAcylation pathway, promoting antioxidant responses in NASH mice [216]. Furthermore, curcumin improves mitochondrial function via the SIRT3 pathway, reducing liver fat accumulation in postnatal overfed rats and fatty L02 cells[217]. These results imply that curcumin may represent an effective therapeutic option for NAFLD, targeting various components of the disease's underlying mechanisms. Nevertheless, despite substantial experimental support, clinical evidence is still scarce, highlighting the necessity for more human studies to comprehensively determine curcumin's effectiveness in treating NAFLD.

Dihydromyricetin

Dihydromyricetin (DHM) has demonstrated encouraging effects in the treatment of NAFLD. Research suggests that DHM improves NAFLD by managing lipid and glucose metabolism, diminishing inflammation, and restoring the balance of gut microbiota [218,219]. DHM inhibits inflammatory signaling pathways, specifically TLR4/NF-κB, while promoting the growth of beneficial gut bacteria[219]. Additionally, it boosts mitochondrial function and redox equilibrium through SIRT3-dependent pathways, enhancing both mitochondrial respiratory capacity and antioxidant functions [219,220]. Laboratory studies indicate that DHM shields hepatocytes from lipid buildup and oxidative stress induced by oleic acid by inhibiting lipogenesis and modulating the PPARγ, AMPK, and AKT signaling pathways [221].These results imply that DHM has the potential to serve as a therapeutic option for NAFLD, targeting various factors involved in the disease's development, such as lipid metabolism, inflammation, oxidative stress, and gut microbiota imbalance.

b. Sirtuin Inhibitors

AK-7

In the realm of hepatocellular carcinoma, the pharmaceutical sector has explored the potential of sirtuin-targeting compounds, such as the SIRT2 inhibitor AK-7, which has shown anti-tumor effects in preclinical models of HCC [222]. However, the therapeutic potential of SIRT2 inhibition remains contentious, with some studies suggesting both tumor-promoting and tumor-suppressing effects [223,224].

8. Gut Microbiota-Based Interventions: Probiotics, Prebiotics, and Synbiotics

Alongside sirtuin-targeted therapies, the pharmaceutical field is exploring gut microbiota-based interventions for disease management. Strategies addressing dysbiosis encompass FMT, probiotics, prebiotics, and dietary modifications. [225]. Probiotics are beneficial live microorganisms, whereas prebiotics are selective substrates for host microorganisms [226]. These components can influence gut microbiota, improve immune response, and alleviate various health conditions, including gastrointestinal disorders, allergies, and infections [227]. The combined application of probiotics and prebiotics, referred to as synbiotics, demonstrates positive effects on the maintenance of a healthy gut microbiome [228]. Probiotics, prebiotics, and synbiotics have exhibited effectiveness in modulating immune responses, treating infections, managing inflammatory bowel diseases, and augmenting cancer treatment modalities [229].
In NAFLD, FMT is a novel strategy to modify gut microbiome and achieve metabolic balance [77]. FMT entails transferring a healthy microbiome to individuals with dysbiosis. The main aim is to replenish the recipient's gut with beneficial microbes and restore a balanced microbial population [230]. FMT has shown promise in improving intestinal structure and function, enhancing lipid metabolism, reducing insulin resistance, suppressing inflammation, and alleviating NAFLD symptoms [231,232]. Figure 6.
By diversifying sirtuin-targeting and gut microbiome-modulating interventions, the pharmaceutical industry seeks to offer enhanced and individualized treatment modalities for various diseases, capitalizing on the intricate interactions between these biological pathways and their health ramifications.
In FMT, a healthy donor's stool is processed and given to a NAFLD patient. Delivery can be via oral or endoscopic methods. NAFLD is linked to gut dysbiosis and heightened intestinal permeability, facilitating the transfer of gut-derived factors to the liver, exacerbating the condition. FMT seeks to rectify dysbiosis and enhance the gut barrier, aiming to improve liver health in NAFLD individuals.

Conclusions

The intricate interplay between sirtuins and gut microbiota represents a critical nexus in the regulation of metabolic health and disease. As pivotal regulators of cellular mechanisms, sirtuins significantly affect the composition and functionality of gut microbiota, while concurrently serving an essential role in preserving the integrity of the gut barrier and modulating immune responses. Conversely, metabolites derived from gut microbiota can impact sirtuin activity, establishing a dynamic feedback loop that influences overall health outcomes. Given the rising prevalence of NAFLD and its progression to HCC, understanding the molecular mechanisms underlying these interactions plays a key role in creating targeted therapeutic interventions. This review underscores the potential for novel interventions that modulate sirtuin and microbiota pathways to improve health outcomes. Future research should focus on elucidating the detailed mechanisms of sirtuin-microbiota interactions and exploring their implications for innovative approaches to preventing and treating metabolic disorders and liver cancer.

Author Contributions

Conceptualization, A.T. (Aamira Tariq) and M.Z (Mahmoud Zhra); writing—original draft preparation, M.A.E. (Muhammad Affan Elahi), A.Z. (AbuZaid), A.Y. (Ahmed Yaqinuddin); writing—review and editing, A.T. (Aamira Tariq), M. Z (Mahmoud Zhra); All authors have read and agreed to the published version of the manuscript.

Funding

This work is not supported by any research grant.

Institutional Review Board Statement

Not applicable as the manuscript entails literature review of existing data.

Informed Consent Statement

Not applicable as the manuscript harbors literature review of already published research articles.

Data Availability Statement

Not applicable.

Acknowledgments

All the images have been drawn using BioRender Web based tool.

Conflicts of Interest

The authors declare no conflicts of interest.

References

  1. Wu, Q.; Zhang, T.-N.; Chen, H.-h.; Yu, X.-F.; Lv, J.; Liu, Y.-y.; Liu, Y.; Zheng, G.; Zhao, J.; Wei, Y.-F.; Guo, J.-y.; Liu, F.-H.; Chang, Q.; Zhang, Y.-X.; Liu, C.; Zhao, Y., The sirtuin family in health and disease. Signal Transduction and Targeted Therapy 2022, 7.
  2. Bhatt, V.; Tiwari, A.K. Sirtuins, a key regulator of ageing and age-related neurodegenerative diseases. Int. J. Neurosci. 2022, 133, 1167–1192. [CrossRef]
  3. Ceballos, M.P.; Quiroga, A.D.; Palma, N.F. Role of sirtuins in hepatocellular carcinoma progression and multidrug resistance: Mechanistical and pharmacological perspectives. Biochem. Pharmacol. 2023, 212, 115573. [CrossRef]
  4. Afzaal, M.; Saeed, F.; Shah, Y.A.; Hussain, M.; Rabail, R.; Socol, C.T.; Hassoun, A.; Pateiro, M.; Lorenzo, J.M.; Rusu, A.V.; et al. Human gut microbiota in health and disease: Unveiling the relationship. Front. Microbiol. 2022, 13. [CrossRef]
  5. Kim, S.; Seo, S.-U.; Kweon, M.-N. Gut microbiota-derived metabolites tune host homeostasis fate. Semin. Immunopathol. 2024, 46, 1–15. [CrossRef]
  6. Li, X.; Du, Y.; Xue, C.; Kang, X.; Sun, C.; Peng, H.; Fang, L.; Han, Y.; Xu, X.; Zhao, C. SIRT2 Deficiency Aggravates Diet-Induced Nonalcoholic Fatty Liver Disease through Modulating Gut Microbiota and Metabolites. Int. J. Mol. Sci. 2023, 24, 8970. [CrossRef]
  7. Sharma, A.; Mahur, P.; Muthukumaran, J.; Singh, A.K.; Jain, M. Shedding light on structure, function and regulation of human sirtuins: a comprehensive review. 3 Biotech 2022, 13, 1–15. [CrossRef]
  8. Afzaal, A.; Rehman, K.; Kamal, S.; Akash, M.S.H. Versatile role of sirtuins in metabolic disorders: From modulation of mitochondrial function to therapeutic interventions. J. Biochem. Mol. Toxicol. 2022, 36, e23047. [CrossRef]
  9. D’Angelo, S.; Mele, E.; Di Filippo, F.; Viggiano, A.; Meccariello, R. Sirt1 Activity in the Brain: Simultaneous Effects on Energy Homeostasis and Reproduction. Int. J. Environ. Res. Public Health 2021, 18, 1243. [CrossRef]
  10. Zhang, Y.; Long, X.; Ruan, X.; Wei, Q.; Zhang, L.; Wo, L.; Huang, D.; Lin, L.; Wang, D.; Xia, L.; et al. SIRT2-mediated deacetylation and deubiquitination of C/EBPβ prevents ethanol-induced liver injury. Cell Discov. 2021, 7, 1–17. [CrossRef]
  11. Lambona, C.; Zwergel, C.; Valente, S.; Mai, A. SIRT3 Activation a Promise in Drug Development? New Insights into SIRT3 Biology and Its Implications on the Drug Discovery Process. J. Med. Chem. 2024, 67, 1662–1689. [CrossRef]
  12. Tao, Z.; Jin, Z.; Wu, J.; Cai, G.; Yu, X. Sirtuin family in autoimmune diseases. Front. Immunol. 2023, 14, 1186231. [CrossRef]
  13. Su, S.; Ndiaye, M.A.; Singh, C.K.; Ahmad, N. Mitochondrial Sirtuins in Skin and Skin Cancers. Photochem. Photobiol. 2020, 96, 973–980. [CrossRef]
  14. Poljšak, B.; Kovač, V.; Špalj, S.; Milisav, I. The Central Role of the NAD+ Molecule in the Development of Aging and the Prevention of Chronic Age-Related Diseases: Strategies for NAD+ Modulation. Int. J. Mol. Sci. 2023, 24, 2959. [CrossRef]
  15. Kosciuk, T.; Wang, M.; Hong, J.Y.; Lin, H. Updates on the epigenetic roles of sirtuins. Curr. Opin. Chem. Biol. 2019, 51, 18–29. [CrossRef]
  16. Lagunas-Rangel, F.A. Current role of mammalian sirtuins in DNA repair. 2019, 80, 85–92. [CrossRef]
  17. Roos, W.P.; Krumm, A. The multifaceted influence of histone deacetylases on DNA damage signalling and DNA repair. Nucleic Acids Res. 2016, 44, 10017–10030. [CrossRef]
  18. Chang, A.R.; Ferrer, C.M.; Mostoslavsky, R. SIRT6, a Mammalian Deacylase with Multitasking Abilities. Physiol. Rev. 2020, 100, 145–169. [CrossRef]
  19. Carter, R.J.; Parsons, J.L. Base Excision Repair, a Pathway Regulated by Posttranslational Modifications. Mol. Cell. Biol. 2016, 36, 1426–1437. [CrossRef]
  20. Alves-Fernandes, D.K.; Jasiulionis, M.G. The Role of SIRT1 on DNA Damage Response and Epigenetic Alterations in Cancer. Int. J. Mol. Sci. 2019, 20, 3153. [CrossRef]
  21. Wu, X.; Cao, N.; Fenech, M.; Wang, X. Role of Sirtuins in Maintenance of Genomic Stability: Relevance to Cancer and Healthy Aging. DNA Cell Biol. 2016, 35, 542–575. [CrossRef]
  22. O'Callaghan, C.; Vassilopoulos, A. Sirtuins at the crossroads of stemness, aging, and cancer. Aging Cell 2017, 16, 1208–1218. [CrossRef]
  23. Chojdak-Łukasiewicz, J.; Bizoń, A.; Waliszewska-Prosół, M.; Piwowar, A.; Budrewicz, S.; Pokryszko-Dragan, A. Role of Sirtuins in Physiology and Diseases of the Central Nervous System. Biomedicines 2022, 10, 2434. [CrossRef]
  24. Hong, Y. A.; Kim, J. E.; Jo, M. J.; Ko, G. J., The Role of Sirtuins in Kidney Diseases. International Journal of Molecular Sciences 2020, 21.
  25. Kumari, P.; Tarighi, S.; Braun, T.; Ianni, A. SIRT7 Acts as a Guardian of Cellular Integrity by Controlling Nucleolar and Extra-Nucleolar Functions. Genes 2021, 12, 1361. [CrossRef]
  26. Maissan, P.; Mooij, E.J.; Barberis, M. Sirtuins-Mediated System-Level Regulation of Mammalian Tissues at the Interface between Metabolism and Cell Cycle: A Systematic Review. Biology 2021, 10, 194. [CrossRef]
  27. Vazquez, B.N.; Vaquero, A.; Schindler, K. Sirtuins in female meiosis and in reproductive longevity. Mol. Reprod. Dev. 2020, 87, 1175–1187. [CrossRef]
  28. Kratz, E.M.; Kokot, I.; Dymicka-Piekarska, V.; Piwowar, A. Sirtuins—The New Important Players in Women’s Gynecological Health. Antioxidants 2021, 10, 84. [CrossRef]
  29. Hamaidi, I.; Kim, S. Sirtuins are crucial regulators of T cell metabolism and functions. Exp. Mol. Med. 2022, 54, 207–215. [CrossRef]
  30. Watroba, M.; Szukiewicz, D. Sirtuins at the Service of Healthy Longevity. Front. Physiol. 2021, 12. [CrossRef]
  31. Zhao, E.; Hou, J.; Ke, X.; Abbas, M.N.; Kausar, S.; Zhang, L.; Cui, H. The Roles of Sirtuin Family Proteins in Cancer Progression. Cancers 2019, 11, 1949. [CrossRef]
  32. Carafa, V.; Altucci, L.; Nebbioso, A. Dual Tumor Suppressor and Tumor Promoter Action of Sirtuins in Determining Malignant Phenotype. Front. Pharmacol. 2019, 10, 38. [CrossRef]
  33. Zhu, S.; Dong, Z.; Ke, X.; Hou, J.; Zhao, E.; Zhang, K.; Wang, F.; Yang, L.; Xiang, Z.; Cui, H. The roles of sirtuins family in cell metabolism during tumor development. Semin. Cancer Biol. 2018, 57, 59–71. [CrossRef]
  34. Alves-Fernandes, D.K.; Jasiulionis, M.G. The Role of SIRT1 on DNA Damage Response and Epigenetic Alterations in Cancer. Int. J. Mol. Sci. 2019, 20, 3153. [CrossRef]
  35. Yin, J.-Y.; Lu, X.-T.; Hou, M.-L.; Cao, T.; Tian, Z. Sirtuin1-p53: A potential axis for cancer therapy. Biochem. Pharmacol. 2023, 212, 115543. [CrossRef]
  36. Chen, G.; Huang, P.; Hu, C. The role of SIRT2 in cancer: A novel therapeutic target. Int. J. Cancer 2020, 147, 3297–3304. [CrossRef]
  37. Onyiba, C.I.; Scarlett, C.J.; Weidenhofer, J. The Mechanistic Roles of Sirtuins in Breast and Prostate Cancer. Cancers 2022, 14, 5118. [CrossRef]
  38. George, J.; Ahmad, N. Mitochondrial Sirtuins in Cancer: Emerging Roles and Therapeutic Potential. Cancer Res. 2016, 76, 2500–2506. [CrossRef]
  39. Fiorentino, F.; Carafa, V.; Favale, G.; Altucci, L.; Mai, A.; Rotili, D. The Two-Faced Role of SIRT6 in Cancer. Cancers 2021, 13, 1156. [CrossRef]
  40. Karbasforooshan, H.; Hayes, A.W.; Mohammadzadeh, N.; Zirak, M.R.; Karimi, G. The possible role of Sirtuins and microRNAs in hepatocellular carcinoma therapy. Cell Cycle 2020, 19, 3209–3221. [CrossRef]
  41. Farcas, M.; Gavrea, A.-A.; Gulei, D.; Ionescu, C.; Irimie, A.; Catana, C.S.; Berindan-Neagoe, I. SIRT1 in the Development and Treatment of Hepatocellular Carcinoma. Front. Nutr. 2019, 6, 148. [CrossRef]
  42. Matijašić, M.; Meštrović, T.; Čipčić Paljetak, H.; Perić, M.; Barešić, A.; Verbanac, D., Gut Microbiota beyond Bacteria—Mycobiome, Virome, Archaeome, and Eukaryotic Parasites in IBD. International Journal of Molecular Sciences 2020, 21.
  43. Vemuri, R.; Shankar, E.M.; Chieppa, M.; Eri, R.; Kavanagh, K. Beyond Just Bacteria: Functional Biomes in the Gut Ecosystem Including Virome, Mycobiome, Archaeome and Helminths. Microorganisms 2020, 8, 483. [CrossRef]
  44. Garcia-Bonete, M.J.; Rajan, A.; Suriano, F.; Layunta, E. The Underrated Gut Microbiota Helminths, Bacteriophages, Fungi, and Archaea. Life 2023, 13, 1765. [CrossRef]
  45. Sasso, J.M.; Ammar, R.M.; Tenchov, R.; Lemmel, S.; Kelber, O.; Grieswelle, M.; Zhou, Q.A. Gut Microbiome–Brain Alliance: A Landscape View into Mental and Gastrointestinal Health and Disorders. ACS Chem. Neurosci. 2023, 14, 1717–1763. [CrossRef]
  46. Abenavoli, L.; Giubilei, L.; Procopio, A.C.; Spagnuolo, R.; Luzza, F.; Boccuto, L.; Scarpellini, E. Gut Microbiota in Non-Alcoholic Fatty Liver Disease Patients with Inflammatory Bowel Diseases: A Complex Interplay. Nutrients 2022, 14, 5323. [CrossRef]
  47. Barko, P. C.; McMichael, M. A.; Swanson, K. S.; Williams, D. A., The Gastrointestinal Microbiome: A Review. Journal of Veterinary Internal Medicine 2017, 32, 9 - 25.
  48. Pickard, J.M.; Zeng, M.Y.; Caruso, R.; Núñez, G. Gut microbiota: Role in pathogen colonization, immune responses, and inflammatory disease. Immunol. Rev. 2017, 279, 70–89. [CrossRef]
  49. Srivastava, A.; Prabhakar, M.R.; Mohanty, A.; Meena, S.S. Influence of gut microbiome on the human physiology. Syst. Microbiol. Biomanufacturing 2021, 2, 217–231. [CrossRef]
  50. Garcia-Gutierrez, E.; Mayer, M.J.; Cotter, P.D.; Narbad, A. Gut microbiota as a source of novel antimicrobials. Gut Microbes 2018, 10, 1–21. [CrossRef]
  51. I, R.; G, G.; A, H.; K, S.; J, S.; I, T.; K, T., Gut microbiota functions: metabolism of nutrients and other food compo nents. European Journal of Nutrition.
  52. Levy, M.; Blacher, E.; Elinav, E., Microbiome, metabolites and host immunity. Current opinion in microbiology 2017, 35, 8-15.
  53. Fung, T.C.; Olson, C.A.; Hsiao, E.Y. Interactions between the microbiota, immune and nervous systems in health and disease. Nat. Neurosci. 2017, 20, 145–155. [CrossRef]
  54. Gasaly, N.; de Vos, P.; Hermoso, M.A. Impact of Bacterial Metabolites on Gut Barrier Function and Host Immunity: A Focus on Bacterial Metabolism and Its Relevance for Intestinal Inflammation. Front. Immunol. 2021, 12, 658354. [CrossRef]
  55. Postler, T.S.; Ghosh, S. Understanding the Holobiont: How Microbial Metabolites Affect Human Health and Shape the Immune System. Cell Metab. 2017, 26, 110–130. [CrossRef]
  56. Louis, P.; Flint, H.J. Formation of propionate and butyrate by the human colonic microbiota. Environ. Microbiol. 2017, 19, 29–41. [CrossRef]
  57. Blaak, E.E.; Canfora, E.E.; Theis, S.; Frost, G.; Groen, A.K.; Mithieux, G.; Nauta, A.; Scott, K.; Stahl, B.; Van Harsselaar, J.; et al. Short chain fatty acids in human gut and metabolic health. Benef. Microbes 2020, 11, 411–455. [CrossRef]
  58. Parada Venegas, D.; De La Fuente, M.K.; Landskron, G.; González, M.J.; Quera, R.; Dijkstra, G.; Harmsen, H.J.M.; Faber, K.N.; Hermoso, M.A. Short Chain Fatty Acids (SCFAs)-Mediated Gut Epithelial and Immune Regulation and Its Relevance for Inflammatory Bowel Diseases. Front. Immunol. 2019, 10, 277. [CrossRef]
  59. Martin-Gallausiaux, C.; Marinelli, L.; Blottière, H.M.; Larraufie, P.; Lapaque, N. SCFA: Mechanisms and functional importance in the gut. Proc. Nutr. Soc. 2021, 80, 37–49. [CrossRef]
  60. He, J.; Zhang, P.; Shen, L.; Niu, L.; Tan, Y.; Chen, L.; Zhao, Y.; Bai, L.; Hao, X.; Li, X.; et al. Short-Chain Fatty Acids and Their Association with Signalling Pathways in Inflammation, Glucose and Lipid Metabolism. Int. J. Mol. Sci. 2020, 21, 6356. [CrossRef]
  61. Ramirez-Perez, O. L.; Cruz-Ramon, V. C.; Chinchilla-López, P.; Méndez-Sánchez, N., The Role of the Gut Microbiota in Bile Acid Metabolism. Annals of hepatology 2017, 16 Suppl. 1: s3-105., s15-s20.
  62. 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. [CrossRef]
  63. Oliphant, K.; Allen-Vercoe, E. Macronutrient metabolism by the human gut microbiome: major fermentation by-products and their impact on host health. Microbiome 2019, 7, 91. [CrossRef]
  64. Moszak, M.; Szulińska, M.; Bogdański, P. You Are What You Eat—The Relationship between Diet, Microbiota, and Metabolic Disorders—A Review. Nutrients 2020, 12, 1096. [CrossRef]
  65. Anwar, H.; Iftikhar, A.; Muzaffar, H.; Almatroudi, A.; Allemailem, K.S.; Navaid, S.; Saleem, S.; Khurshid, M. Biodiversity of Gut Microbiota: Impact of Various Host and Environmental Factors. BioMed Res. Int. 2021, 2021. [CrossRef]
  66. Rinninella, E.; Raoul, P.; Cintoni, M.; Franceschi, F.; Miggiano, G.A.D.; Gasbarrini, A.; Mele, M.C. What Is the Healthy Gut Microbiota Composition? A Changing Ecosystem across Age, Environment, Diet, and Diseases. Microorganisms 2019, 7, 14. [CrossRef]
  67. Wang, Y.; Xu, B.; Chen, H.; Yang, F.; Huang, J.; Jiao, X.; Zhang, Y. Environmental factors and gut microbiota: Toward better conservation of deer species. Front. Microbiol. 2023, 14, 1136413. [CrossRef]
  68. Rothschild, D.; Weissbrod, O.; Barkan, E.; Kurilshikov, A.; Korem, T.; Zeevi, D.; Costea, P.I.; Godneva, A.; Kalka, I.N.; Bar, N.; et al. Environment dominates over host genetics in shaping human gut microbiota. Nature 2018, 555, 210–215. [CrossRef]
  69. Kurilshikov, A.; Wijmenga, C.; Fu, J.; Zhernakova, A. Host Genetics and Gut Microbiome: Challenges and Perspectives. Trends Immunol. 2017, 38, 633–647. [CrossRef]
  70. Jeong, S. Factors influencing development of the infant microbiota: from prenatal period to early infancy. Clin. Exp. Pediatr. 2021, 65, 438–447. [CrossRef]
  71. Hasan, N.; Yang, H. Factors affecting the composition of the gut microbiota, and its modulation. PeerJ 2019, 7, e7502. [CrossRef]
  72. Kriss, M.; Hazleton, K.Z.; Nusbacher, N.M.; Martin, C.G.; A Lozupone, C. Low diversity gut microbiota dysbiosis: drivers, functional implications and recovery. 2018, 44, 34–40. [CrossRef]
  73. Heravi, F.S. Gut Microbiota and Autoimmune Diseases: Mechanisms, Treatment, Challenges, and Future Recommendations. Curr. Clin. Microbiol. Rep. 2024, 11, 18–33. [CrossRef]
  74. Weiss, G.A.; Hennet, T. Mechanisms and consequences of intestinal dysbiosis. Cell. Mol. Life Sci. 2017, 74, 2959–2977. [CrossRef]
  75. Willighagen, E., Defining Dysbiosis for a Cluster of Chronic Diseases. Scientific Reports 2019, 9.
  76. Jalandra, R.; Dhar, R.; Pethusamy, K.; Sharma, M.; Karmakar, S. Dysbiosis: Gut feeling. F1000Research 2022, 11, 911. [CrossRef]
  77. Hou, K.; Wu, Z.; Chen, X.-Y.; Wang, J.-Q.; Zhang, D.; Xiao, C.; Zhu, D.; Koya, J. B.; Wei, L.; Li, J.; Chen, Z. S., Microbiota in health and diseases. Signal Transduction and Targeted Therapy 2022, 7.
  78. Kho, Z.Y.; Lal, S.K. The Human Gut Microbiome – A Potential Controller of Wellness and Disease. Front. Microbiol. 2018, 9, 1835. [CrossRef]
  79. Meng, X.; Zhang, G.; Cao, H.; Yu, D.; Fang, X.; Vos, W.M.; Wu, H. Gut dysbacteriosis and intestinal disease: mechanism and treatment. J. Appl. Microbiol. 2020, 129, 787–805. [CrossRef]
  80. Zhang, Y.; Wang, X.-L.; Zhou, M.; Kang, C.; Lang, H.-D.; Chen, M.-T.; Hui, S.-C.; Wang, B.; Mi, M.-T. Crosstalk between gut microbiota and Sirtuin-3 in colonic inflammation and tumorigenesis. Exp. Mol. Med. 2018, 50, 1–11. [CrossRef]
  81. Caron, A.Z.; He, X.; Mottawea, W.; Seifert, E.L.; Jardine, K.; Dewar-Darch, D.; Cron, G.O.; Harper, M.; Stintzi, A.; McBurney, M.W. The SIRT1 deacetylase protects mice against the symptoms of metabolic syndrome. FASEB J. 2013, 28, 1306–1316. [CrossRef]
  82. Vikram, A.; Kim, Y.-R.; Kumar, S.; Li, Q.; Kassan, M.; Jacobs, J.S.; Irani, K. Vascular microRNA-204 is remotely governed by the microbiome and impairs endothelium-dependent vasorelaxation by downregulating Sirtuin1. Nat. Commun. 2016, 7, 12565. [CrossRef]
  83. Wellman, A.S.; Metukuri, M.R.; Kazgan, N.; Xu, X.; Xu, Q.; Ren, N.S.; Czopik, A.; Shanahan, M.T.; Kang, A.; Chen, W.; et al. Intestinal Epithelial Sirtuin 1 Regulates Intestinal Inflammation During Aging in Mice by Altering the Intestinal Microbiota. Gastroenterology 2017, 153, 772–786. [CrossRef]
  84. Munteanu, C.; Onose, G.; Poștaru, M.; Turnea, M.; Rotariu, M.; Galaction, A.I. Hydrogen Sulfide and Gut Microbiota: Their Synergistic Role in Modulating Sirtuin Activity and Potential Therapeutic Implications for Neurodegenerative Diseases. Pharmaceuticals 2024, 17, 1480. [CrossRef]
  85. Hou, D.; Yu, T.; Lu, X.; Hong, J.Y.; Yang, M.; Zi, Y.; Ho, T.T.; Lin, H. Sirt2 inhibition improves gut epithelial barrier integrity and protects mice from colitis. Proc. Natl. Acad. Sci. 2024, 121. [CrossRef]
  86. Liu, L.; Xie, Y.; Li, G.; Zhang, T.; Sui, Y.; Zhao, Z.; Zhang, Y.; Yang, W.; Geng, X.; Xue, D.; et al. Gut microbiota-derived nicotinamide mononucleotide alleviates acute pancreatitis by activating pancreatic SIRT3 signalling. Br. J. Pharmacol. 2022, 180, 647–666. [CrossRef]
  87. Chen, M.; Hui, S.; Lang, H.; Zhou, M.; Zhang, Y.; Kang, C.; Zeng, X.; Zhang, Q.; Yi, L.; Mi, M. SIRT3 Deficiency Promotes High-Fat Diet-Induced Nonalcoholic Fatty Liver Disease in Correlation with Impaired Intestinal Permeability through Gut Microbial Dysbiosis. Mol. Nutr. Food Res. 2018, 63, e1800612. [CrossRef]
  88. Knop, M.; Treitz, C.; Bettendorf, S.; Bossen, J.; von Frieling, J.; Doms, S.; Bruchhaus, I.; Kühnlein, R. P.; Baines, J. F.; Tholey, A.; Roeder, T., A mitochondrial sirtuin shapes the intestinal microbiota by controlling lysozyme expression. bioRxiv 2023.
  89. Tucker, S.A.; Hu, S.-H.; Vyas, S.; Park, A.; Joshi, S.; Inal, A.; Lam, T.; Tan, E.; Haigis, K.M.; Haigis, M.C. SIRT4 loss reprograms intestinal nucleotide metabolism to support proliferation following perturbation of homeostasis. Cell Rep. 2024, 43, 113975. [CrossRef]
  90. Sun, R.; Zhang, Z.; Bao, R.; Guo, X.; Gu, Y.; Yang, W.; Wei, J.; Chen, X.; Tong, L.; Meng, J.; et al. Loss of SIRT5 promotes bile acid-induced immunosuppressive microenvironment and hepatocarcinogenesis. J. Hepatol. 2022, 77, 453–466. [CrossRef]
  91. Xu, K.; Guo, Y.; Wang, Y.; Ren, Y.; Low, V.; Cho, S.; Ping, L.; Peng, K.; Li, X.; Qiu, Y.; et al. Decreased Enterobacteriaceae translocation due to gut microbiota remodeling mediates the alleviation of premature aging by a high-fat diet. Aging Cell 2022, 22. [CrossRef]
  92. Kim, S.; Byun, J.; Jung, S.; Kim, B.; Lee, K.; Jeon, H.; Lee, J.; Choi, H.; Kim, E.; Jeen, Y.; et al. Sirtuin 7 Inhibitor Attenuates Colonic Mucosal Immune Activation in Mice—Potential Therapeutic Target in Inflammatory Bowel Disease. Biomedicines 2022, 10, 2693. [CrossRef]
  93. Caruso, R.; Marafini, I.; Franzè, E.; Stolfi, C.; Zorzi, F.; Monteleone, I.; Caprioli, F.; Colantoni, A.; Sarra, M.; Sedda, S.; et al. Defective expression of SIRT1 contributes to sustain inflammatory pathways in the gut. Mucosal Immunol. 2014, 7, 1467–1479. [CrossRef]
  94. Lytvynenko, A.; Voznesenskaya, T.; Janchij, R. SIRT1 IS A REGULATOR OF AUTOPHAGY IN INTESTINAL CELLS. Fiziolohichnyĭ zhurnal 2020, 66, 97–103. [CrossRef]
  95. Ma, Y.; Xu, C.; Wang, W.; Sun, L.; Yang, S.; Lu, D.; Liu, Y.; Yang, H. [Role of SIRT1 in the protection of intestinal epithelial barrier under hypoxia and its mechanism]. Zhonghua wei chang wai ke za zhi = Chinese journal of gastrointestinal surgery 2014, 17, 602–6.
  96. Li, C.; Zhou, Y.; Rychahou, P.; Weiss, H.L.; Lee, E.Y.; Perry, C.L.; Barrett, T.A.; Wang, Q.; Evers, B.M. SIRT2 Contributes to the Regulation of Intestinal Cell Proliferation and Differentiation. Cell. Mol. Gastroenterol. Hepatol. 2020, 10, 43–57. [CrossRef]
  97. Wu, Q.-J.; Zhang, T.-N.; Chen, H.-H.; Yu, X.-F.; Lv, J.-L.; Liu, Y.-Y.; Liu, Y.-S.; Zheng, G.; Zhao, J.-Q.; Wei, Y.-F.; Guo, J.-Y.; Liu, F.-H.; Chang, Q.; Zhang, Y.-X.; Liu, C.-G.; Zhao, Y.-H., The sirtuin family in health and disease. Signal Transduction and Targeted Therapy 2022, 7, (1), 402.
  98. Chen, M.; Hui, S.; Lang, H.; Zhou, M.; Zhang, Y.; Kang, C.; Zeng, X.; Zhang, Q.; Yi, L.; Mi, M. SIRT3 Deficiency Promotes High-Fat Diet-Induced Nonalcoholic Fatty Liver Disease in Correlation with Impaired Intestinal Permeability through Gut Microbial Dysbiosis. Mol. Nutr. Food Res. 2018, 63, e1800612. [CrossRef]
  99. Wang, T.; Cao, Y.; Zheng, Q.; Tu, J.; Zhou, W.; He, J.; Zhong, J.; Chen, Y.; Wang, J.; Cai, R.; et al. SENP1-Sirt3 Signaling Controls Mitochondrial Protein Acetylation and Metabolism. Mol. Cell 2019, 75, 823–834.e5. [CrossRef]
  100. Nasrin, N.; Wu, X.; Fortier, E.; Feng, Y.; Bare', O.C.; Chen, S.; Ren, X.; Wu, Z.; Streeper, R.S.; Bordone, L. SIRT4 Regulates Fatty Acid Oxidation and Mitochondrial Gene Expression in Liver and Muscle Cells. J. Biol. Chem. 2010, 285, 31995–32002. [CrossRef]
  101. Fabbrizi, E.; Fiorentino, F.; Carafa, V.; Altucci, L.; Mai, A.; Rotili, D. Emerging Roles of SIRT5 in Metabolism, Cancer, and SARS-CoV-2 Infection. Cells 2023, 12, 852. [CrossRef]
  102. Bringman-Rodenbarger, L.R.; Guo, A.H.; Lyssiotis, C.A.; Lombard, D.B. Emerging Roles for SIRT5 in Metabolism and Cancer. Antioxidants Redox Signal. 2018, 28, 677–690. [CrossRef]
  103. Xiong, X.; Yang, C.; He, W.-Q.; Yu, J.; Xin, Y.; Zhang, X.; Huang, R.; Ma, H.; Xu, S.; Li, Z.; et al. Sirtuin 6 maintains epithelial STAT6 activity to support intestinal tuft cell development and type 2 immunity. Nat. Commun. 2022, 13, 1–16. [CrossRef]
  104. Xu, K.; Guo, Y.; Ping, L.; Qiu, Y.; Liu, Q.; Li, Z.; Wang, Z. Protective Effects of SIRT6 Overexpression against DSS-Induced Colitis in Mice. Cells 2020, 9, 1513. [CrossRef]
  105. Lerrer, B.; Gertler, A.A.; Cohen, H.Y. The complex role of SIRT6 in carcinogenesis. Carcinog. 2015, 37, 108–118. [CrossRef]
  106. Liu, X.; Li, C.; Li, Q.; Chang, H.-C.; Tang, Y.-C. SIRT7 Facilitates CENP-A Nucleosome Assembly and Suppresses Intestinal Tumorigenesis. iScience 2020, 23, 101461. [CrossRef]
  107. Haslberger, A.; Lilja, S.; Bäck, H.; Stoll, C.; Mayer, A.; Pointner, A.; Hippe, B.; Krammer, U. Increased Sirtuin expression, senescence regulating miRNAs, mtDNA, and Bifidobateria correlate with wellbeing and skin appearance after Sirtuin- activating drink. Bioact. Compd. Heal. Dis. 2021, 4, 45. [CrossRef]
  108. Salazar, J.; Durán, P.; Díaz, M.P.; Chacín, M.; Santeliz, R.; Mengual, E.; Gutiérrez, E.; León, X.; Díaz, A.; Bernal, M.; et al. Exploring the Relationship between the Gut Microbiota and Ageing: A Possible Age Modulator. Int. J. Environ. Res. Public Heal. 2023, 20, 5845. [CrossRef]
  109. Clark, A.; Mach, N. The Crosstalk between the Gut Microbiota and Mitochondria during Exercise. Front. Physiol. 2017, 8, 319. [CrossRef]
  110. Gong, S.; Feng, Y.; Zeng, Y.; Zhang, H.; Pan, M.; He, F.; Wu, R.; Chen, J.; Lu, J.; Zhang, S.; et al. Gut microbiota accelerates cisplatin-induced acute liver injury associated with robust inflammation and oxidative stress in mice. J. Transl. Med. 2021, 19, 1–13. [CrossRef]
  111. Singh, R.; Chandrashekharappa, S.; Bodduluri, S.R.; Baby, B.V.; Hegde, B.; Kotla, N.G.; Hiwale, A.A.; Saiyed, T.; Patel, P.; Vijay-Kumar, M.; et al. Enhancement of the gut barrier integrity by a microbial metabolite through the Nrf2 pathway. Nat. Commun. 2019, 10, 1–18. [CrossRef]
  112. Yang, M.; Massad, K.; Kimchi, E.T.; Staveley-O’carroll, K.F.; Li, G. Gut microbiota and metabolite interface-mediated hepatic inflammation. Immunometabolism 2024, 6, e00037–e00037. [CrossRef]
  113. Zhang, K.; Zhang, X.; Lv, A.; Fan, S.; Zhang, J., Saccharomyces Boulardii Modulates Necrotizing Enterocolitis in Neonatal Mice by Regulating the Sirtuin 1/NF-κB Pathway and the Intestinal Microbiota. Molecular Medicine Reports 2020, 22, (2), 671-680.
  114. Estes, C.; Razavi, H.; Loomba, R.; Younossi, Z.; Sanyal, A.J. Modeling the epidemic of nonalcoholic fatty liver disease demonstrates an exponential increase in burden of disease. Hepatology 2018, 67, 123–133. [CrossRef]
  115. EASL-EASD-EASO Clinical Practice Guidelines for the management of non-alcoholic fatty liver disease. J Hepatol 2016, 64, (6), 1388-402.
  116. Tiniakos, D.G.; Vos, M.B.; Brunt, E.M. Nonalcoholic Fatty Liver Disease: Pathology and Pathogenesis. Annu. Rev. Pathol. Mech. Dis. 2010, 5, 145–171. [CrossRef]
  117. Grander, C.; Grabherr, F.; Moschen, A.R.; Tilg, H. Non-Alcoholic Fatty Liver Disease: Cause or Effect of Metabolic Syndrome. Visc. Med. 2016, 32, 329–334. [CrossRef]
  118. Paschos, P.; Paletas, K. Non alcoholic fatty liver disease two-hit process: multifactorial character of the second hit. Hippokratia 2009, 13, (2), 128.
  119. Jang, H.R.; Lee, H.-Y. Mechanisms linking gut microbial metabolites to insulin resistance. World J. Diabetes 2021, 12, 730–744. [CrossRef]
  120. Zhou, S.; Tang, X.; Chen, H.-Z. Sirtuins and Insulin Resistance. Front. Endocrinol. 2018, 9, 748. [CrossRef]
  121. Caron, A.Z.; He, X.; Mottawea, W.; Seifert, E.L.; Jardine, K.; Dewar-Darch, D.; Cron, G.O.; Harper, M.; Stintzi, A.; McBurney, M.W. The SIRT1 deacetylase protects mice against the symptoms of metabolic syndrome. FASEB J. 2013, 28, 1306–1316. [CrossRef]
  122. Wang, Y.-J.; Paneni, F.; Stein, S.; Matter, C.M. Modulating Sirtuin Biology and Nicotinamide Adenine Diphosphate Metabolism in Cardiovascular Disease—From Bench to Bedside. Front. Physiol. 2021, 12. [CrossRef]
  123. Chandramowlishwaran, P.; Vijay, A.; Abraham, D.; Li, G.; Mwangi, S.M.; Srinivasan, S. Role of Sirtuins in Modulating Neurodegeneration of the Enteric Nervous System and Central Nervous System. Front. Neurosci. 2020, 14. [CrossRef]
  124. Wu, T.; Liu, Y.-H.; Fu, Y.-C.; Liu, X.-M.; Zhou, X.-H. Direct evidence of sirtuin downregulation in the liver of non-alcoholic fatty liver disease patients. Ann. Clin. Lab. Sci. 2014, 44, 410–418.
  125. Bruce, K.D.; Szczepankiewicz, D.; Sihota, K.K.; Ravindraanandan, M.; Thomas, H.; Lillycrop, K.A.; Burdge, G.C.; Hanson, M.A.; Byrne, C.D.; Cagampang, F.R. Altered cellular redox status, sirtuin abundance and clock gene expression in a mouse model of developmentally primed NASH. Biochim. et Biophys. Acta (BBA) - Mol. Cell Biol. Lipids 2016, 1861, 584–593. [CrossRef]
  126. Canfora, E.E.; Meex, R.C.R.; Venema, K.; Blaak, E.E. Gut microbial metabolites in obesity, NAFLD and T2DM. Nat. Rev. Endocrinol. 2019, 15, 261–273. [CrossRef]
  127. Aron-Wisnewsky, J.; Vigliotti, C.; Witjes, J.; Le, P.; Holleboom, A.G.; Verheij, J.; Nieuwdorp, M.; Clément, K. Gut microbiota and human NAFLD: disentangling microbial signatures from metabolic disorders. Nat. Rev. Gastroenterol. Hepatol. 2020, 17, 279–297. [CrossRef]
  128. Loo, T. M.; Kamachi, F.; Watanabe, Y.; Yoshimoto, S.; Kanda, H.; Arai, Y.; Nakajima-Takagi, Y.; Iwama, A.; Koga, T.; Sugimoto, Y., Gut microbiota promotes obesity-associated liver cancer through PGE2-mediated suppression of antitumor immunity. Cancer discovery 2017, 7, (5), 522-538.
  129. Bo, T.; Shao, S.; Wu, D.; Niu, S.; Zhao, J.; Gao, L. Relative variations of gut microbiota in disordered cholesterol metabolism caused by high-cholesterol diet and host genetics. Microbiologyopen 2017, 6. [CrossRef]
  130. Zhang, X.; Coker, O.O.; Chu, E.S.; Fu, K.; Lau, H.C.H.; Wang, Y.-X.; Chan, A.W.H.; Wei, H.; Yang, X.; Sung, J.J.Y.; et al. Dietary cholesterol drives fatty liver-associated liver cancer by modulating gut microbiota and metabolites. Gut 2020, 70, 761–774. [CrossRef]
  131. Takaki, A.; Kawai, D.; Yamamoto, K. Multiple Hits, Including Oxidative Stress, as Pathogenesis and Treatment Target in Non-Alcoholic Steatohepatitis (NASH). Int. J. Mol. Sci. 2013, 14, 20704–20728. [CrossRef]
  132. Henao-Mejia, J.; Elinav, E.; Jin, C.; Hao, L.; Mehal, W.Z.; Strowig, T.; Thaiss, C.A.; Kau, A.L.; Eisenbarth, S.C.; Jurczak, M.J.; et al. Inflammasome-mediated dysbiosis regulates progression of NAFLD and obesity. Nature 2012, 482, 179–185. [CrossRef]
  133. Wiest, R.; Albillos, A.; Trauner, M.; Bajaj, J. S.; Jalan, R., Targeting the gut-liver axis in liver disease. J Hepatol 2017, 67, (5), 1084-1103.
  134. Schnabl, B.; Brenner, D.A. Interactions Between the Intestinal Microbiome and Liver Diseases. Gastroenterology 2014, 146, 1513–1524. [CrossRef]
  135. Goodrich, J.K.; Waters, J.L.; Poole, A.C.; Sutter, J.L.; Koren, O.; Blekhman, R.; Beaumont, M.; Van Treuren, W.; Knight, R.; Bell, J.T.; et al. Human Genetics Shape the Gut Microbiome. Cell 2014, 159, 789–799. [CrossRef]
  136. Buzzetti, E.; Pinzani, M.; Tsochatzis, E.A. The multiple-hit pathogenesis of non-alcoholic fatty liver disease (NAFLD). Metabolism 2016, 65, 1038–1048. [CrossRef]
  137. Zhou, D.; Pan, Q.; Xin, F.-Z.; Zhang, R.-N.; He, C.-X.; Chen, G.-Y.; Liu, C.; Chen, Y.-W.; Fan, J.-G. Sodium butyrate attenuates high-fat diet-induced steatohepatitis in mice by improving gut microbiota and gastrointestinal barrier. World J. Gastroenterol. 2017, 23, 60–75. [CrossRef]
  138. Kang, C.; Wang, B.; Kaliannan, K.; Wang, X.; Lang, H.; Hui, S.; Huang, L.; Zhang, Y.; Zhou, M.; Chen, M.; et al. Gut Microbiota Mediates the Protective Effects of Dietary Capsaicin against Chronic Low-Grade Inflammation and Associated Obesity Induced by High-Fat Diet. mBio 2017, 8, e00470-17. [CrossRef]
  139. Muccioli, G.G.; Naslain, D.; Bäckhed, F.; Reigstad, C.S.; Lambert, D.M.; Delzenne, N.M.; Cani, P.D. The endocannabinoid system links gut microbiota to adipogenesis. Mol. Syst. Biol. 2010, 6, 392. [CrossRef]
  140. Lam, Y.Y.; Ha, C.W.Y.; Campbell, C.R.; Mitchell, A.J.; Dinudom, A.; Oscarsson, J.; Cook, D.I.; Hunt, N.H.; Caterson, I.D.; Holmes, A.J.; et al. Increased Gut Permeability and Microbiota Change Associate with Mesenteric Fat Inflammation and Metabolic Dysfunction in Diet-Induced Obese Mice. PLoS ONE 2012, 7, e34233. [CrossRef]
  141. Everard, A.; Lazarevic, V.; Gaïa, N.; Johansson, M.; Ståhlman, M.; Bäckhed, F.; Delzenne, N.M.; Schrenzel, J.; Francois, P.; Cani, P.D. Microbiome of prebiotic-treated mice reveals novel targets involved in host response during obesity. ISME J. 2014, 8, 2116–2130. [CrossRef]
  142. Villanueva, A., Hepatocellular Carcinoma. N Engl J Med 2019, 380, (15), 1450-1462.
  143. Akinyemiju, T.; Abera, S.; Ahmed, M.; Alam, N.; Alemayohu, M.A.; Allen, C.; Al-Raddadi, R.; Alvis-Guzman, N.; Amoako, Y.; Artaman, A.; et al. The Burden of Primary Liver Cancer and Underlying Etiologies from 1990 to 2015 at the Global, Regional, and National Level: Results from the Global Burden of Disease Study 2015. JAMA Oncol. 2017, 3, 1683–1691.
  144. Jindal, A.; Thadi, A.; Shailubhai, K. Hepatocellular Carcinoma: Etiology and Current and Future Drugs. 2019, 9, 221–232. [CrossRef]
  145. Wong, R.J.; Cheung, R.; Ahmed, A. Nonalcoholic steatohepatitis is the most rapidly growing indication for liver transplantation in patients with hepatocellular carcinoma in the U.S. Hepatology 2014, 59, 2188–2195. [CrossRef]
  146. An, L.; Wirth, U.; Koch, D.; Schirren, M.; Drefs, M.; Koliogiannis, D.; Niess, H.; Andrassy, J.; Guba, M.; Bazhin, A.V.; et al. Metabolic Role of Autophagy in the Pathogenesis and Development of NAFLD. Metabolites 2023, 13, 101. [CrossRef]
  147. Itoh, M.; Suganami, T.; Nakagawa, N.; Tanaka, M.; Yamamoto, Y.; Kamei, Y.; Terai, S.; Sakaida, I.; Ogawa, Y. Melanocortin 4 Receptor–Deficient Mice as a Novel Mouse Model of Nonalcoholic Steatohepatitis. American Journal of Pathology 2011, 179, 2454–2463. [CrossRef]
  148. Wang, J.; Xie, G., Bile Acid–microbiota Crosstalk in Gastrointestinal Inflammation and Carcinogenesis. Nature Reviews Gastroenterology & Hepatology 2017, 15, (2), 111-128.
  149. Milosevic, I.; Vujovic, A.; Barac, A.; Djelic, M.; Korac, M.; Radovanovic Spurnic, A.; Gmizic, I.; Stevanovic, O.; Djordjevic, V.; Lekic, N.; et al. Gut-Liver Axis, Gut Microbiota, and Its Modulation in the Management of Liver Diseases: A Review of the Literature. Int. J. Mol. Sci. 2019, 20, 395. [CrossRef]
  150. Farcas, M.; Gavrea, A.-A.; Gulei, D.; Ionescu, C.; Irimie, A.; Catana, C.S.; Berindan-Neagoe, I. SIRT1 in the Development and Treatment of Hepatocellular Carcinoma. Front. Nutr. 2019, 6, 148. [CrossRef]
  151. Ling, S.; Li, J.; Shan, Q.; Dai, H.; Lu, D.; Wen, X.; Song, P.; Xie, H.; Zhou, L.; Liu, J.; et al. USP22 mediates the multidrug resistance of hepatocellular carcinoma via the SIRT1/AKT/MRP1 signaling pathway. Mol. Oncol. 2017, 11, 682–695. [CrossRef]
  152. Hao, C.; Zhu, P.-X.; Yang, X.; Han, Z.-P.; Jiang, J.-H.; Zong, C.; Zhang, X.-G.; Liu, W.-T.; Zhao, Q.-D.; Fan, T.-T.; et al. Overexpression of SIRT1 promotes metastasis through epithelial-mesenchymal transition in hepatocellular carcinoma. BMC Cancer 2014, 14, 978. [CrossRef]
  153. Ren, Z.; Li, A.; Jiang, J.; Zhou, L.; Yu, Z.; Lu, H.; Xie, H.; Chen, X.; Shao, L.; Zhang, R.; et al. Gut microbiome analysis as a tool towards targeted non-invasive biomarkers for early hepatocellular carcinoma. Gut 2018, 68, 1014–1023. [CrossRef]
  154. Pant, K.; Yadav, A. K.; Gupta, P.; Islam, R.; Saraya, A.; Venugopal, S. K., Butyrate induces ROS-mediated apoptosis by modulating miR-22/SIRT-1 pathway in hepatic cancer cells. Redox Biol 2017, 12, 340-349.
  155. Zhang, J.; Yang, Y.; Yang, T.; Liu, Y.; Li, A.; Fu, S.; Wu, M.; Pan, Z.; Zhou, W. microRNA-22, downregulated in hepatocellular carcinoma and correlated with prognosis, suppresses cell proliferation and tumourigenicity. Br. J. Cancer 2010, 103, 1215–1220. [CrossRef]
  156. Qian, L.; Zhang, Y.; Wang, G.; Li, B.; Zhou, H.; Qiu, J.; Qin, L., MicroRNA-29 Regulates Tumor Progression and Survival Through miR-29a-SIRT1-Wnt/β-catenin Pathway in Hepatocellular Carcinoma. 2023.
  157. Wang, X.; Ling, S.; Wu, W.; Shan, Q.; Liu, P.; Wang, C.; Wei, X.; Ding, W.; Teng, X.; Xu, X., Ubiquitin-Specific Protease 22/Silent Information Regulator 1 Axis Plays a Pivotal Role in the Prognosis and 5-Fluorouracil Resistance in Hepatocellular Carcinoma. Digestive Diseases and Sciences 2019, 65, (4), 1064-1073.
  158. Xiong, H.; Ni, Z.; He, J.; Jiang, S.; Li, X.; Gong, W.; Zheng, L.; Chen, S.; Li, B.; Zhang, N.; et al. LncRNA HULC triggers autophagy via stabilizing Sirt1 and attenuates the chemosensitivity of HCC cells. Oncogene 2017, 36, 3528–3540. [CrossRef]
  159. Zhou, Z.-Q.; Guan, J.; Chen, S.; Sun, J.; Zhang, Z., Sirtuin 1 Protects the Mitochondria in Hepatocellular Carcinoma Cells via Suppressing Hypoxia-Induced Factor-1 Alpha Expression. 2022.
  160. Li, Y.; Xu, S.; Li, J.; Zheng, L.; Feng, M.; Wang, X.; Han, K.; Pi, H.; Li, M.; Huang, X.; et al. SIRT1 facilitates hepatocellular carcinoma metastasis by promoting PGC-1α-mediated mitochondrial biogenesis. Oncotarget 2016, 7, 29255–29274. [CrossRef]
  161. Herranz, D.; Muñoz-Martin, M.; Cañamero, M.; Mulero, F.; Martinez-Pastor, B.; Fernandez-Capetillo, O.; Serrano, M. Sirt1 improves healthy ageing and protects from metabolic syndrome-associated cancer. Nat. Commun. 2010, 1, 1–8. [CrossRef]
  162. Chen, H.-C.; Jeng, Y.-M.; Yuan, R.-H.; Hsu, H.-C.; Chen, Y.-L. SIRT1 Promotes Tumorigenesis and Resistance to Chemotherapy in Hepatocellular Carcinoma and its Expression Predicts Poor Prognosis. Ann. Surg. Oncol. 2011, 19, 2011–2019. [CrossRef]
  163. Chen, J.; Zhang, B.; Wong, N.; Lo, A. W.; To, K. F.; Chan, A. W.; Ng, M. H.; Ho, C. Y.; Cheng, S. H.; Lai, P. B.; Yu, J.; Ng, H. K.; Ling, M. T.; Huang, A. L.; Cai, X. F.; Ko, B. C., Sirtuin 1 is upregulated in a subset of hepatocellular carcinomas where it is essential for telomere maintenance and tumor cell growth. Cancer Res 2011, 71, (12), 4138-49.
  164. Tanos, B.; Rodriguez-Boulan, E. The epithelial polarity program: machineries involved and their hijacking by cancer. Oncogene 2008, 27, 6939–6957. [CrossRef]
  165. Portmann, S.; Fahrner, R.; Lechleiter, A.; Keogh, A.; Overney, S.; Laemmle, A.; Mikami, K.; Montani, M.; Tschan, M.P.; Candinas, D.; et al. Antitumor Effect of SIRT1 Inhibition in Human HCC Tumor Models In Vitro and In Vivo. Mol. Cancer Ther. 2013, 12, 499–508. [CrossRef]
  166. Chen, J.; Chan, A.W.; To, K.-F.; Chen, W.; Zhang, Z.; Ren, J.; Song, C.; Cheung, Y.-S.; Lai, P.B.; Cheng, S.-H.; et al. SIRT2 overexpression in hepatocellular carcinoma mediates epithelial to mesenchymal transition by protein kinase B/glycogen synthase kinase-3β/β-catenin signaling. Hepatology 2013, 57, 2287–2298. [CrossRef]
  167. Huang, S.; Zhao, Z.; Tang, D.; Zhou, Q.; Li, Y.; Zhou, L.; Yin, Y.; Wang, Y.; Pan, Y.; Dorfman, R.G.; et al. Downregulation of SIRT2 Inhibits Invasion of Hepatocellular Carcinoma by Inhibiting Energy Metabolism. Transl. Oncol. 2017, 10, 917–927. [CrossRef]
  168. Kim, J.K.; Noh, J.H.; Jung, K.H.; Eun, J.W.; Bae, H.J.; Kim, M.G.; Chang, Y.G.; Shen, Q.; Park, W.S.; Lee, J.Y.; et al. Sirtuin7 oncogenic potential in human hepatocellular carcinoma and its regulation by the tumor suppressors MiR-125a-5p and MiR-125b. Hepatology 2013, 57, 1055–1067. [CrossRef]
  169. Kim, Y.; Jung, K.-Y.; Kim, Y. H.; Xu, P.; Kang, B. E.; Jo, Y.; Pandit, N.; Kwon, J.; Gariani, K.; Gariani, J.; Lee, J.; Verbeek, J.; Nam, S.; Bae, S.-J.; Ha, K.-T.; Yi, H.-S.; Shong, M.; Kim, K.-H.; Kim, D.; Jung, H. J.; Lee, C.-W.; Kim, K. R.; Schoonjans, K.; Auwerx, J.; Ryu, D., Inhibition of SIRT7 Overcomes Sorafenib Acquired Resistance by Suppressing ERK1/2 Phosphorylation via the DDX3X-mediated NLRP3 Inflammasome in Hepatocellular Carcinoma. Drug Resistance Updates 2024, 73, 101054.
  170. Gu, Y.; Ding, C.; Yu, T.; Liu, B.; Tang, W.; Wang, Z.; Tang, X.; Liang, G.; Peng, J.; Zhang, X.; Li, Z., SIRT7 Promotes Hippo/YAP Activation and Cancer Cell Proliferation in Hepatocellular Carcinoma via Suppressing MST1. Cancer Science 2024, 115, (4), 1209-1223.
  171. Caussy, C.; Hsu, C.; Lo, M. T.; Liu, A.; Bettencourt, R.; Ajmera, V. H.; Bassirian, S.; Hooker, J.; Sy, E.; Richards, L.; Schork, N.; Schnabl, B.; Brenner, D. A.; Sirlin, C. B.; Chen, C. H.; Loomba, R., Link between gut-microbiome derived metabolite and shared gene-effects with hepatic steatosis and fibrosis in NAFLD. Hepatology 2018, 68, (3), 918-932.
  172. Zeybel, M.; Arif, M.; Li, X.; Altay, O.; Yang, H.; Shi, M.; Akyildiz, M.; Saglam, B.; Gonenli, M.G.; Yigit, B.; et al. Multiomics Analysis Reveals the Impact of Microbiota on Host Metabolism in Hepatic Steatosis. Adv. Sci. 2022, 9, 2104373. [CrossRef]
  173. Liu, L.; Fu, Q.; Li, T.; Shao, K.; Zhu, X.; Cong, Y.; Zhao, X. Gut microbiota and butyrate contribute to nonalcoholic fatty liver disease in premenopause due to estrogen deficiency. PLOS ONE 2022, 17, e0262855. [CrossRef]
  174. Grąt, M.; Wronka, K.; Krasnodębski, M.; Masior, Ł.; Lewandowski, Z.; Kosińska, I.; Grąt, K.; Stypułkowski, J.; Rejowski, S.; Wasilewicz, M.; et al. Profile of Gut Microbiota Associated With the Presence of Hepatocellular Cancer in Patients With Liver Cirrhosis. Transplant. Proc. 2016, 48, 1687–1691. [CrossRef]
  175. Ni, J.; Huang, R.; Zhou, H.; Xu, X.; Li, Y.; Cao, P.; Zhong, K.; Ge, M.; Chen, X.; Hou, B.; et al. Analysis of the Relationship Between the Degree of Dysbiosis in Gut Microbiota and Prognosis at Different Stages of Primary Hepatocellular Carcinoma. Front. Microbiol. 2019, 10, 1458. [CrossRef]
  176. Ponziani, F.R.; Bhoori, S.; Castelli, C.; Putignani, L.; Rivoltini, L.; Del Chierico, F.; Sanguinetti, M.; Morelli, D.; Sterbini, F.P.; Petito, V.; et al. Hepatocellular Carcinoma Is Associated With Gut Microbiota Profile and Inflammation in Nonalcoholic Fatty Liver Disease. Hepatology 2019, 69, 107–120. [CrossRef]
  177. Zheng, R.; Wang, G.; Pang, Z.; Ran, N.; Gu, Y.; Guan, X.; Yuan, Y.; Zuo, X.; Pan, H.; Zheng, J.; et al. Liver cirrhosis contributes to the disorder of gut microbiota in patients with hepatocellular carcinoma. Cancer Med. 2020, 9, 4232–4250. [CrossRef]
  178. Behary, J.; Amorim, N.; Jiang, X.-T.; Raposo, A.; Gong, L.; McGovern, E.; Ibrahim, R.; Chu, F.; Stephens, C.; Jebeili, H.; et al. Gut microbiota impact on the peripheral immune response in non-alcoholic fatty liver disease related hepatocellular carcinoma. Nat. Commun. 2021, 12, 1–14. [CrossRef]
  179. Ren, Z.; Li, A.; Jiang, J.; Zhou, L.; Yu, Z.; Lu, H.; Xie, H.; Chen, X.; Shao, L.; Zhang, R.; et al. Gut microbiome analysis as a tool towards targeted non-invasive biomarkers for early hepatocellular carcinoma. Gut 2018, 68, 1014–1023. [CrossRef]
  180. Dai, H.; Sinclair, D.A.; Ellis, J.L.; Steegborn, C. Sirtuin activators and inhibitors: Promises, achievements, and challenges. Pharmacol. Ther. 2018, 188, 140–154. [CrossRef]
  181. Chandramowlishwaran, P.; Vijay, A.; Abraham, D.; Li, G.; Mwangi, S.M.; Srinivasan, S. Role of Sirtuins in Modulating Neurodegeneration of the Enteric Nervous System and Central Nervous System. Front. Neurosci. 2020, 14. [CrossRef]
  182. Manjula, R.; Anuja, K.; Alcain, F.J. SIRT1 and SIRT2 Activity Control in Neurodegenerative Diseases. Front. Pharmacol. 2021, 11. [CrossRef]
  183. Li, X.; Du, Y.; Xue, C.; Kang, X.; Sun, C.; Peng, H.; Fang, L.; Han, Y.; Xu, X.; Zhao, C. SIRT2 Deficiency Aggravates Diet-Induced Nonalcoholic Fatty Liver Disease through Modulating Gut Microbiota and Metabolites. Int. J. Mol. Sci. 2023, 24, 8970. [CrossRef]
  184. E Lakhan, S.; Kirchgessner, A. Gut microbiota and sirtuins in obesity-related inflammation and bowel dysfunction. J. Transl. Med. 2011, 9, 202–202. [CrossRef]
  185. Bayram, H.M.; Majoo, F.M.; Ozturkcan, A. Polyphenols in the prevention and treatment of non-alcoholic fatty liver disease: An update of preclinical and clinical studies. Clin. Nutr. ESPEN 2021, 44, 1–14. [CrossRef]
  186. Wang, P.; Wang, J.; Li, D.; Ke, W.; Chen, F.; Hu, X. Targeting the gut microbiota with resveratrol: a demonstration of novel evidence for the management of hepatic steatosis. J. Nutr. Biochem. 2020, 81, 108363. [CrossRef]
  187. Du, F.; Huang, R.; Lin, D.; Wang, Y.; Yang, X.; Huang, X.; Zheng, B.; Chen, Z.; Huang, Y.; Wang, X.; et al. Resveratrol Improves Liver Steatosis and Insulin Resistance in Non-alcoholic Fatty Liver Disease in Association With the Gut Microbiota. Front. Microbiol. 2021, 12. [CrossRef]
  188. Prakash, V.; Bose, C.; Sunilkumar, D.; Cherian, R.M.; Thomas, S.S.; Nair, B.G. Resveratrol as a Promising Nutraceutical: Implications in Gut Microbiota Modulation, Inflammatory Disorders, and Colorectal Cancer. Int. J. Mol. Sci. 2024, 25, 3370. [CrossRef]
  189. Karabekir, S.C.; Ozgorgulu, A. Possible protective effects of resveratrol in hepatocellular carcinoma. 2020, 23, 71–78. [CrossRef]
  190. He, X.; Li, Y.; Deng, X.; Xiao, X.; Zeng, J. Integrative evidence construction for resveratrol treatment of nonalcoholic fatty liver disease: preclinical and clinical meta-analyses. Front. Pharmacol. 2023, 14, 1230783. [CrossRef]
  191. Tian, Y.; Ma, J.; Wang, W.; Zhang, L.; Xu, J.; Wang, K.; Li, D., Resveratrol supplement inhibited the NF-κB inflammation pathway through activating AMPKα-SIRT1 pathway in mice with fatty liver. Molecular and cellular biochemistry 2016, 422, (1-2), 75-84.
  192. Wu, W.-Y.; Ding, X.-Q.; Gu, T.-T.; Guo, W.-J.; Jiao, R.-Q.; Song, L.; Sun, Y.; Pan, Y.; Kong, L.-D., Pterostilbene improves hepatic lipid accumulation via the MiR-34a/Sirt1/SREBP-1 pathway in fructose-fed rats. Journal of agricultural and food chemistry 2020, 68, (5), 1436-1446.
  193. Estrela, J. M.; Ortega, A.; Mena, S.; Rodriguez, M. L.; Asensi, M., Pterostilbene: biomedical applications. Critical reviews in clinical laboratory sciences 2013, 50, (3), 65-78.
  194. Gómez-Zorita, S.; Fernández-Quintela, A.; Lasa, A.; Aguirre, L.; Rimando, A.M.; Portillo, M.P. Pterostilbene, a Dimethyl Ether Derivative of Resveratrol, Reduces Fat Accumulation in Rats Fed an Obesogenic Diet. J. Agric. Food Chem. 2014, 62, 8371–8378. [CrossRef]
  195. Wang, P.; Sang, S. Metabolism and pharmacokinetics of resveratrol and pterostilbene. BioFactors 2018, 44, 16–25. [CrossRef]
  196. Han, J.; Li, S.; Wang, W.; Jiang, X.; Liu, C.; Lei, L.; Li, Y.; Sheng, R.; Zhang, Y.; Wu, Y.; et al. SIRT1 Activator E1231 Alleviates Nonalcoholic Fatty Liver Disease by Regulating Lipid Metabolism. Curr. Issues Mol. Biol. 2023, 45, 5052–5070. [CrossRef]
  197. Vidyashankar, S.; Varma, R.S.; Patki, P.S. Quercetin ameliorate insulin resistance and up-regulates cellular antioxidants during oleic acid induced hepatic steatosis in HepG2 cells. Toxicol. Vitr. 2013, 27, 945–953. [CrossRef]
  198. Cao, P.; Wang, Y.; Zhang, C.; Sullivan, M.A.; Chen, W.; Jing, X.; Yu, H.; Li, F.; Wang, Q.; Zhou, Z.; et al. Quercetin ameliorates nonalcoholic fatty liver disease (NAFLD) via the promotion of AMPK-mediated hepatic mitophagy. J. Nutr. Biochem. 2023, 120, 109414. [CrossRef]
  199. Ying, H.-Z.; Liu, Y.-H.; Yu, B.; Wang, Z.-Y.; Zang, J.-N.; Yu, C.-H. Dietary quercetin ameliorates nonalcoholic steatohepatitis induced by a high-fat diet in gerbils. Food Chem. Toxicol. 2013, 52, 53–60. [CrossRef]
  200. Chen, L.; Liu, J.; Mei, G.; Chen, H.; Peng, S.; Zhao, Y.; Yao, P.; Tang, Y. Quercetin and non-alcoholic fatty liver disease: A review based on experimental data and bioinformatic analysis. Food Chem. Toxicol. 2021, 154, 112314. [CrossRef]
  201. Pham, T.X.; Bae, M.; Kim, M.-B.; Lee, Y.; Hu, S.; Kang, H.; Park, Y.-K.; Lee, J.-Y. Nicotinamide riboside, an NAD+ precursor, attenuates the development of liver fibrosis in a diet-induced mouse model of liver fibrosis. Biochim. et Biophys. Acta (BBA) - Mol. Basis Dis. 2019, 1865, 2451–2463. [CrossRef]
  202. Longo, L.; de Castro, J.M.; Keingeski, M.B.; Rampelotto, P.H.; Stein, D.J.; Guerreiro, G.T.S.; de Souza, V.E.G.; Cerski, C.T.S.; Uribe-Cruz, C.; Torres, I.L.; et al. Nicotinamide riboside and dietary restriction effects on gut microbiota and liver inflammatory and morphologic markers in cafeteria diet–induced obesity in rats. Nutrition 2023, 110, 112019. [CrossRef]
  203. Han, X.; Bao, X.; Lou, Q.; Xie, X.; Zhang, M.; Zhou, S.; Guo, H.; Jiang, G.; Shi, Q. Nicotinamide riboside exerts protective effect against aging-induced NAFLD-like hepatic dysfunction in mice. PeerJ 2019, 7, e7568. [CrossRef]
  204. Zhang, H.; Zhao, X.; Zhang, L.; Sun, D.; Ma, Y.; Bai, Y.; Bai, X.; Liang, X.; Liang, H. Nicotinamide Riboside Ameliorates Fructose-Induced Lipid Metabolism Disorders in Mice by Activating Browning of WAT, and May Be Also Related to the Regulation of Gut Microbiota. Nutrients 2024, 16, 3920. [CrossRef]
  205. Lin, X.; Zhang, J.; Chu, Y.; Nie, Q.; Zhang, J. Berberine prevents NAFLD and HCC by modulating metabolic disorders. Pharmacol. Ther. 2024, 254, 108593. [CrossRef]
  206. Dai, Y.; Zhu, W.; Zhou, J.; Shen, T. The combination of berberine and evodiamine ameliorates high-fat diet-induced non-alcoholic fatty liver disease associated with modulation of gut microbiota in rats. Braz. J. Med Biol. Res. 2022, 55, e12096. [CrossRef]
  207. Shu, X.; Li, M.; Cao, Y.; Li, C.; Zhou, W.; Ji, G.; Zhang, L. Berberine Alleviates Non-alcoholic Steatohepatitis Through Modulating Gut Microbiota Mediated Intestinal FXR Activation. Front. Pharmacol. 2021, 12. [CrossRef]
  208. Ionita-Radu, F.; Patoni, C.; Nancoff, A.S.; Marin, F.-S.; Gaman, L.; Bucurica, A.; Socol, C.; Jinga, M.; Dutu, M.; Bucurica, S. Berberine Effects in Pre-Fibrotic Stages of Non-Alcoholic Fatty Liver Disease—Clinical and Pre-Clinical Overview and Systematic Review of the Literature. Int. J. Mol. Sci. 2024, 25, 4201. [CrossRef]
  209. Luo, Y.; Tian, G.; Zhuang, Z.; Chen, J.; You, N.; Zhuo, L.; Liang, B.; Song, Y.; Zang, S.; Liu, J.; et al. Berberine prevents non-alcoholic steatohepatitis-derived hepatocellular carcinoma by inhibiting inflammation and angiogenesis in mice. American journal of translational research 2019, 11, 2668–2682.
  210. Jiang, H.; Mao, T.; Sun, Z.; Shi, L.; Han, X.; Zhang, Y.; Zhang, X.; Wang, J.; Hu, J.; Zhang, L.; Li, J.; Han, H.-X., Yinchen Linggui Zhugan decoction ameliorates high fat diet-induced nonalcoholic fatty liver disease by modulation of SIRT1/Nrf2 signaling pathway and gut microbiota. Frontiers in Microbiology 2022, 13.
  211. Wang, S.; Li, X.; Zhang, B.; Li, Y.; Chen, K.; Qi, H.; Gao, M.; Rong, J.; Liu, L.; Wan, Y.; et al. Tangshen formula targets the gut microbiota to treat non-alcoholic fatty liver disease in HFD mice: A 16S rRNA and non-targeted metabolomics analyses. Biomed. Pharmacother. 2024, 173, 116405. [CrossRef]
  212. Wang, Y.; Zhao, H.; Li, X.; Li, N.; Wang, Q.; Liu, Y.; Liang, Q.; Shao, Z.; Zhang, N.; Zhao, T.; et al. Tangshen Formula Alleviates Hepatic Steatosis by Inducing Autophagy Through the AMPK/SIRT1 Pathway. Front. Physiol. 2019, 10, 494. [CrossRef]
  213. Kong, Q.; Zhang, B.-X.; Zhang, H.-J.; Yan, M.-H.; Li, P. Experimental study of Tangshen formulain improved lipid metabolism and phenotypic switch of macrophage in db/db mice. China J. Chin. Mater. Medica 2014, 41, 1693–1698. [CrossRef]
  214. Rahmani, S.; Asgary, S.; Askari, G.; Keshvari, M.; Hatamipour, M.; Feizi, A.; Sahebkar, A. Treatment of Non-alcoholic Fatty Liver Disease with Curcumin: A Randomized Placebo-controlled Trial. Phytotherapy Res. 2016, 30, 1540–1548. [CrossRef]
  215. Amel Zabihi, N.; Pirro, M.; P Johnston, T.; Sahebkar, A., Is there a role for curcumin supplementation in the treatment of non-alcoholic fatty liver disease? The data suggest yes. Current Pharmaceutical Design 2017, 23, (7), 969-982.
  216. Lee, D. E.; Lee, S. J.; Kim, S. J.; Lee, H.-S.; Kwon, O.-S., Curcumin ameliorates nonalcoholic fatty liver disease through inhibition of O-GlcNAcylation. Nutrients 2019, 11, (11), 2702.
  217. Du, S.; Zhu, X.; Zhou, N.; Zheng, W.; Zhou, W.; Li, X. Curcumin alleviates hepatic steatosis by improving mitochondrial function in postnatal overfed rats and fatty L02 cells through the SIRT3 pathway. Food Funct. 2022, 13, 2155–2171. [CrossRef]
  218. Gong, H.; Xu, H.; Li, M.; Zhang, D. Molecular mechanism and therapeutic significance of dihydromyricetin in nonalcoholic fatty liver disease. Eur. J. Pharmacol. 2022, 935, 175325. [CrossRef]
  219. Kang, L.; Ma, X.; Yu, F.; Xu, L.; Lang, L. Dihydromyricetin Alleviates Non-Alcoholic Fatty Liver Disease by Modulating Gut Microbiota and Inflammatory Signaling Pathways. J. Microbiol. Biotechnol. 2024, 34, 2637–2647. [CrossRef]
  220. Zeng, X.; Yang, J.; Hu, O.; Huang, J.; Ran, L.; Chen, M.; Zhang, Y.; Zhou, X.; Zhu, J.; Zhang, Q.; et al. Dihydromyricetin Ameliorates Nonalcoholic Fatty Liver Disease by Improving Mitochondrial Respiratory Capacity and Redox Homeostasis Through Modulation of SIRT3 Signaling. Antioxidants Redox Signal. 2019, 30, 163–183. [CrossRef]
  221. Xie, C.; Chen, Z.; Zhang, C.; Xu, X.; Jin, J.; Zhan, W.; Han, T.; Wang, J. Dihydromyricetin ameliorates oleic acid-induced lipid accumulation in L02 and HepG2 cells by inhibiting lipogenesis and oxidative stress. Life Sci. 2016, 157, 131–139. [CrossRef]
  222. Huang, S.; Zhao, Z.; Tang, D.; Zhou, Q.; Li, Y.; Zhou, L.; Yin, Y.; Wang, Y.; Pan, Y.; Dorfman, R.G.; et al. Downregulation of SIRT2 Inhibits Invasion of Hepatocellular Carcinoma by Inhibiting Energy Metabolism. Transl. Oncol. 2017, 10, 917–927. [CrossRef]
  223. Wang, T.; Xu, Z.; Lu, Y.; Shi, J.; Liu, W.; Zhang, C.; Jiang, Z.; Qi, B.; Bai, L. Recent Progress on the Discovery of Sirt2 Inhibitors for the Treatment of Various Cancers. Curr. Top. Med. Chem. 2019, 19, 1051–1058. [CrossRef]
  224. Kaya, S.G.; Eren, G. Selective inhibition of SIRT2: A disputable therapeutic approach in cancer therapy. Bioorganic Chem. 2023, 143, 107038. [CrossRef]
  225. Holmes, A.; Finger, C.; Morales-Scheihing, D.; Lee, J.; McCullough, L.D. Gut dysbiosis and age-related neurological diseases; an innovative approach for therapeutic interventions. Transl. Res. journal of laboratory and clinical medicine 2020, 226, 39–56. [CrossRef]
  226. Yamashiro, Y. Probiotics to Prebiotics and Their Clinical Use. Reference Module in Biomedical Sciences 2021.
  227. Manzoor, S.; Wani, S.M.; Mir, S.A.; Rizwan, D. Role of probiotics and prebiotics in mitigation of different diseases. Nutrition 2022, 96, 111602. [CrossRef]
  228. Upasana, Application of Probiotics, Prebiotics and Synbiotics in Maintaining Gut Health. The Indian Journal of Nutrition and Dietetics 2022.
  229. Yadav, M.K.; Kumari, I.; Singh, B.; Sharma, K.K.; Tiwari, S.K. Probiotics, prebiotics and synbiotics: Safe options for next-generation therapeutics. Appl. Microbiol. Biotechnol. 2022, 106, 505–521. [CrossRef]
  230. Shao, T.; Hsu, R.; Hacein-Bey, C.; Zhang, W.; Gao, L.; Kurth, M.J.; Zhao, H.; Shuai, Z.; Leung, P.S.C. The Evolving Landscape of Fecal Microbial Transplantation. Clin. Rev. Allergy Immunol. 2023, 65, 101–120. [CrossRef]
  231. Abenavoli, L.; Maurizi, V.; Rinninella, E.; Tack, J.; Di Berardino, A.; Santori, P.; Rasetti, C.; Procopio, A.C.; Boccuto, L.; Scarpellini, E. Fecal Microbiota Transplantation in NAFLD Treatment. Medicina 2022, 58, 1559. [CrossRef]
  232. Qiu, X.-X.; Cheng, S.-L.; Liu, Y.-H.; Li, Y.; Zhang, R.; Li, N.-N.; Li, Z. Fecal microbiota transplantation for treatment of non-alcoholic fatty liver disease: Mechanism, clinical evidence, and prospect. World J. Gastroenterol. 2024, 30, 833–842. [CrossRef]
Figure 4. Impact of Gut Dysbiosis on Inflammation and Metabolic Dysfunction.
Figure 4. Impact of Gut Dysbiosis on Inflammation and Metabolic Dysfunction.
Preprints 147863 g004
Figure 5. Impact of Resveratrol on Gut Health and Liver Disease.
Figure 5. Impact of Resveratrol on Gut Health and Liver Disease.
Preprints 147863 g005
Figure 6. The Use of Fecal Microbiota Transplantation (FMT) in Treating NAFLD.
Figure 6. The Use of Fecal Microbiota Transplantation (FMT) in Treating NAFLD.
Preprints 147863 g006
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.
Copyright: This open access article is published under a Creative Commons CC BY 4.0 license, which permit the free download, distribution, and reuse, provided that the author and preprint are cited in any reuse.
Prerpints.org logo

Preprints.org is a free preprint server supported by MDPI in Basel, Switzerland.

Subscribe

Disclaimer

Terms of Use

Privacy Policy

Privacy Settings

© 2025 MDPI (Basel, Switzerland) unless otherwise stated