- 1Gastroenterology, Changchun University of Traditional Chinese Medicine, Changchun, Jilin, China
- 2Department of Liver, Spleen and Gastroenterology, Affiliated Hospital of Changchun University of Traditional Chinese Medicine, Changchun, Jilin, China
Metabolic-associated fatty liver disease (MAFLD) and type 2 diabetes mellitus (T2DM) frequently co-exist on the pathological basis of dysregulated glucose and lipid metabolism, forming a bidirectional causal relationship. The upstream mechanisms underlying this association require further elucidation. Recent studies suggest that the interactive network comprising the “gut–adipose axis” and “gut–pancreatic axis” represents a core component of the comorbidity mechanism. This network initiates with gut microbiota dysbiosis, which alters short-chain fatty acids (SCFAs), lipopolysaccharide (LPS), branched-chain amino acids (BCAAs), secondary bile acids(SBAs), and other microbial metabolites, as well as endocrine signals such as the endocannabinoid system (ECS) and incretin hormones. This network simultaneously influences adipose tissue and the pancreas to coordinate glucose and lipid homeostasis. Therefore, this paper proposes the “Common Messengers, Dual-Axis Convergence” model to systematically elucidate how the gut microbiota, through a shared set of messenger molecules, simultaneously and independently drives lipid and glucose metabolic dysregulation via the gut–adipose and gut–pancreatic axes, ultimately leading to the comorbidity of MAFLD and T2DM.
1 Introduction
In recent years, the network of interactions among the gut and other organs has become a research hotspot across different areas of physiology. These interactions exert extensive regulatory effects on host behavior, energy metabolism, appetite control, development, reproduction, and immune homeostasis. A growing body of research focuses on the regulation of glucose and lipid metabolism in the guy, aiming to explore new intervention targets (1, 2). Within this network, adipose tissue and the pancreas play pivotal roles in gut–organ crosstalk. Specifically, the “gut–adipose axis” and “gut–pancreas axis” establish bidirectional communication between the gut and distal metabolic tissues, thereby mediating the fine-tuned regulation of insulin resistance (IR), lipid homeostasis, and energy partitioning. This multilevel dialogue not only maintains glucose and lipid metabolic balance but also plays a key role in the pathological processes of both Metabolic-associated fatty liver disease(MAFLD) and type 2 diabetes mellitus (T2DM). The aim of this article is to reveal that the comorbidity of MAFLD and T2DM stems not only from the classic gut–liver axis cascade but also, more critically, from a “dual-axis branching” network originating from the gut as a common starting point. This means that gut dysbiosis generates a core set of messenger molecules. One branch of these messengers primarily drives lipid metabolic disorders via the “gut–adipose axis,” while the other branch primarily drives glucose metabolic disorders via the “gut–pancreatic islet axis.” These two signaling pathways act synergistically, jointly establishing the pathological foundation for comorbidity.
2 Strong association between MAFLD and T2DM under the influence of glucose and lipid metabolism
MAFLD is a metabolic disorder influenced by etiological factors such as abnormal lipid metabolism or IR. This disorder is characterized mainly by hepatic lipid droplet accumulation, hepatocyte steatosis, and histological abnormalities. MAFLD is currently among the most common chronic liver diseases. In fact, recent studies have indicated that its global prevalence has reached 39.22% (3, 4). The concept of MAFLD was proposed in 2020 by the Asia-Pacific Working Party on Non-Alcoholic Fatty Liver Disease (5). To highlight its alignment with complex systemic metabolic changes, the “multiple-hit pathogenesis” hypothesis (6–8) has been proposed, encompassing factors such as gut microbiota dysbiosis, IR, oxidative stress, inflammatory responses, hormonal imbalances, and dysglycaemia.
As the hepatic manifestation of metabolic disease, MAFLD often co-exists with other metabolism-related extrahepatic conditions. Cross-sectional data revealed a 65.33% global coprevalence of T2DM and MAFLD (9) and 96.82% of T2DM patients have metabolic dysfunction-associated steatohepatitis (MASH) (10). The co-morbidity of MAFLD and T2DM is believed to stem from disturbances in glucose and lipid metabolism. Furthermore (11), these two conditions may influence each other’s progression through direct or indirect pathways, ultimately forming the “triangular association” illustrated in Table 1.
However, controversies remain regarding the specific mechanisms involved in this series of reactions, suggesting the existence of a common upstream pathway driving this process. Mounting evidence points to gut as the origin of this driving force.
3 The gut: the upstream hub of glucose and lipid metabolism abnormalities
Recent shifts in dietary patterns have propelled research on the gut and metabolic diseases into a new stage (12). Mechanistic studies have indicated that the gut influences energy metabolism homeostasis and glucose/lipid metabolism by regulating the gut microbiota, inflammatory responses, oxidative stress, and the neuroendocrine system (13). These mechanisms are closely related to the pathological progression of both MAFLD and T2DM. Consequently, the gut is regarded as the upstream hub of systemic metabolic disorders (14).
Daniel (15) proposed the hypothesis that disruption of the intestinal barrier and its associated dysbiosis could lead to IR and hepatic steatosis and accelerate the progression of metabolic syndrome, a view subsequently validated by further research. Metabolomic studies of body fluids (16) have revealed that distinct categories of metabolites are altered in metabolic disorders associated with gut microbiota dysbiosis. This dysbiosis is not a generalized change but manifests as a systematic imbalance of specific bacterial taxa with crucial metabolic functions, directly regulating the levels and activity of messenger molecules.
This is primarily reflected in a reduction in protective bacteria: core probiotics such as Akkermansia muciniphila and Faecalibacterium prausnitzii (17, 18) which enhance the gut barrier and ferment dietary fibers to produce SCFAs, are often significantly reduced in patients with MAFLD and T2DM. Their depletion directly leads to insufficient SCFAs production, thereby weakening their multiple protective effects, including nourishing intestinal epithelial cells, exerting anti-inflammatory effects, and stimulating glucagon-like peptide-1(GLP-1) secretion. Another key aspect is the expansion of conditional pathogens: some LPS-rich gram-negative bacteria, such as Escherichia coli (19) from the phylum Proteobacteria, may overgrow. The increase in such bacteria serves as an intrinsic source for persistently elevated circulating LPS levels, providing a continuous trigger for driving chronic inflammation and IR via the TLR4/NF-κB pathway.
Furthermore, the imbalance of metabolically functional bacteria is equally critical: an abnormal abundance of bacteria responsible for converting primary to SBAs (secondary bile acids), such as Clostridium scindens (20), directly disrupts the composition and dynamics of the entire bile acid pool. This not only affects lipid digestion but also, more importantly, interferes with the normal activation of metabolic receptors such as FXR and TGR5 by SBAs, thereby disrupting glucose and energy metabolism homeostasis (Table 2). Additionally, at the mechanistic level, studies further indicate that bacteria such as Prevotella and certain Bacteroides can participate in the regulation of IR by influencing imidazole propionate concentrations (21–25).
Table 2. Metabolites produced by probiotics alone or through interactions with the gut microbiota and their effects.
The dysbiosis-driven disruption of various metabolites not only establishes the molecular basis for MAFLD and T2DM comorbidity but also triggers a cascade of metabolic abnormalities. This cascade, depicted in Figure 1, originates in the gut and extends through a network of key organs, including the liver, pancreas, and adipose tissue.
Figure 1. The gut–adipose–pancreas crosstalk cascade. (The schematic illustrates a self-perpetuating vicious cycle. Solid arrows indicate direct promoting actions or metabolic fluxes, while dashed arrows represent reinforcing feedback loops. Key pathophysiological steps include: 1) Gut dysbiosis and increased intestinal permeability; 2) Hepatic influx of bacterial LPS and FFAs via the portal vein; 3) Activation of pro-inflammatory pathways (e.g., TLR4/NF-κB) and induction of HIR; 4) Compensatory pancreatic β-cell hypersecretion progressing to exhaustion under glucolipotoxicity; 5) Systemic IR promoting adipose tissue lipolysis; and 6) Release of inflammatory factors from liver and adipose tissue, fueling systemic inflammation. This interconnected network, centered on IR, collectively propels disease progression from steatosis to steatohepatitis and overt diabetes. LPS, lipopolysaccharide; FFAs, free fatty acids; TLR4, Toll-like receptor 4; NF-κB, nuclear factor kappa B; HIR, hepatic insulin resistance; IR, insulin resistance.).
Initiated by dysbiosis, harmful gut-derived products including LPS and FFAs translocate into the liver via the portal circulation. Within the liver, such substances trigger lipid deposition and activate inflammatory pathways like TLR4/NF-κB, leading to hepatic insulin resistance and MASH development. Concurrently, pancreatic β-cells compensate through insulin hypersecretion, causing hyperinsulinaemia. Combined with HIR-driven uncontrolled gluconeogenesis, a state of persistent hyperglycaemia emerges. Chronic glucolipotoxicity then promotes β-cell functional exhaustion and overt T2DM onset (26, 27). The disorder amplifies systemically as insulin-resistant adipose tissue increases lipolysis, flooding the liver with additional FFAs and further exacerbating hepatic steatosis and inflammation. Ultimately, compromised liver and adipose tissues secrete copious inflammatory factors, generating a systemic inflammatory state which intensifies insulin resistance and β-cell dysfunction across all tissues. A self-perpetuating, vicious cycle results, centered on the “gut-liver-pancreas-adipose tissue” axis, interconnected by worsening propelling disease progression (28).
Throughout this process, hormones, inflammatory factors, and metabolites interact through a complex network, leading to a cascading failure of the metabolic network and jointly driving metabolic imbalance into a cyclical cascade reaction of “glucose and lipid accumulation → metabolic dysregulation → organ injury” (29, 30).
However, this classical cascade view is insufficient to fully explain the high frequency of MAFLD and T2DM comorbidity. Unger et al. introduced the concept of the “gut–pancreas axis” in 1969 (31); and scholars have subsequently proposed the “gut–adipose axis” in recent years (32). Building on these advances, we recognize that the influence of the gut on comorbidities involves a parallel and crucial core mechanism beyond the classic cascade of the gut–liver–pancreas–adipose axis.
Therefore, we have distilled the “Common Messengers, Dual-Axis Branching” model of MAFLD and T2DM comorbidity. The core premise of this model is that gut microbiota dysbiosis triggers not an isolated change in a single molecule but rather a collective dysregulation of a core set of messenger molecules. These “common messengers” are simultaneously and precisely directed via two relatively independent communication pathways—the “gut-adipose axis” and the “gut-pancreatic islet axis”—to targets beyond the reach of the aforementioned cascade reaction. Specifically, they directly disrupt lipid metabolic homeostasis in adipose tissue and glucose metabolic regulation in the pancreas, respectively. In this process, the gut acts as the source, while adipose tissue and the pancreas serve as the key effector organs.
4 Key messengers linking the gut–adipose axis and the gut–pancreatic islet axis
In this model, the “gut-pancreatic axis (33, 34)” is a bidirectional pathway between the intestine and pancreatic islets that coordinates insulin secretion, glucose homeostasis, and β-cell protection. It integrates neuroendocrine signals (e.g., vagal reflexes, GLP-1/GIP hormones), immune cues (e.g., cytokines), and microbial metabolites (e.g., SCFAs) to regulate pancreatic function. These mechanisms collectively enhance insulin release, suppress glucagon, mitigate inflammation, and maintain β-cell integrity, ensuring precise systemic glucose control.
The “gut-adipose axis (35, 36)” refers to a bidirectional communication network between the gut and adipose tissue. It finely regulates systemic lipid storage, breakdown, and energy expenditure through the following key messenger mechanisms: (1) gut hormones (e.g., GLP-1, PYY) directly regulate adipocyte differentiation and lipolysis; (2) sympathetic nerve activation promotes lipolysis; (3) microbial metabolites (e.g., SBAs) modulate systemic energy expenditure and lipid metabolism by activating host receptors such as FXR and TGR5; and (4) inflammatory responses, among others. By coordinating intestinal nutrient absorption and adipokine secretion, these integrated mechanisms collectively maintain adipose tissue homeostasis.
We propose that the co-morbidity of MAFLD and T2DM can be traced to a core set of gut-derived signaling molecules. These messengers diverge to engage the “gut-adipose axis” and the “gut-pancreatic axis”, perturbing the metabolic homeostasis of adipose tissue and the pancreas, respectively. The ensuing organ-specific dysfunctions then converge to establish a systemic comorbid network. Subsequent sections will delineate how specific messengers—such as SCFAs, LPS, BCAAs, and SBAs—orchestrate these divergent effects within the two axes.
4.1 Microbial metabolites: key pathways regulating glucose and lipid metabolism
4.1.1 Lipopolysaccharide
4.1.1.1 Effects on the gut–pancreatic islet axis
When gut microbiota dysbiosis and impaired barrier function occur, LPS derived from gram-negative bacteria is translocated into the systemic circulation, primarily affecting glucose metabolism by inducing chronic low-grade inflammation (37). It disrupts pancreatic endocrine function and glucose homeostasis through three mechanisms: “inflammatory toxicity,” “signal interference,” and “hormone suppression.”
In vivo experiments in mice have demonstrated that LPS activates immune cells via the TLR4/NF-κB pathway, leading to the production of pro-inflammatory cytokines (e.g., TNF-α, IL-1β) and the induction of inducible nitric oxide synthase (iNOS). iNOS catalyzes the massive production of nitric oxide (•NO), which rapidly reacts with superoxide (O2•−) to form potent biological oxidants such as peroxynitrite (ONOO−). Peroxynitrite directly damages mitochondrial function in β-cells, leading to apoptosis (38). The aforementioned inflammatory cytokines can activate kinases such as JNK and IKKβ, interfere with insulin receptor substrate-1 (IRS-1) function, and downregulate GLUT2 expression, thereby impeding insulin signal transduction and reducing insulin sensitivity (39, 40). Furthermore, LPS can modulate glucose homeostasis by affecting the LPS/GLP-1 pathway. Another animal study demonstrated that LPS reduces L-cell viability, increases TNF-α levels, induces apoptosis, and decreases both the mRNA and protein levels of GLP-1, hindering insulin release (41, 42).
4.1.1.2 Effects on the gut–adipose axis
LPS enters the circulation via two principal routes: the paracellular pathway under impaired tight junctions, and the transcellular pathway during fat absorption after a high-fat diet. The latter utilizes lipid rafts and CD36, allowing LPS to bind to chylomicrons and access the lympho−hematogenous circulation (43), thereby promoting fat storage, accelerating lipolysis, and worsening lipotoxicity.
LPS activates the TLR4/NF-κB pathway in adipose tissue, inducing the expression of factors such as TNF-α. These factors, in turn, inhibit IRS-1 via JNK/IKKβ signaling, block the antilipolytic effect of insulin, and promote the phosphorylation of hormone-sensitive lipase (HSL), accelerating triglyceride (TG) hydrolysis and FFA release (44–46). Additionally, LPS can stimulate the sympathetic nervous system, increasing catecholamine release, which activates the cAMP/PKA pathway via β-adrenergic receptors (47), further promoting HSL phosphorylation and lipolysis. Additionally, LPS inhibits PPARγ activity in adipocytes within mice, reducing adiponectin secretion, which further exacerbates lipolysis and IR (48). Intestinal LPS can also activate the TLR/MyD88 signaling axis, upregulating the expression of Rev-Erbα in small intestinal epithelial cells, directly increasing dietary lipid absorption and exacerbating the lipid load from the source (49). Consequently, LPS promotes adipose tissue dysfunction, elevated systemic FFA levels, and ectopic lipid deposition in the liver through both direct and indirect pathways.
Comorbidity mechanism: LPS translocation under gut microbiota dysbiosis simultaneously activates inflammation in both adipose and pancreatic tissues. On the one hand, it impairs β-cell function, suppresses GLP-1, and exacerbates peripheral IR; on the other hand, it promotes adipose tissue inflammation, lipolysis, and lipid absorption. These two major pathways collectively lead to the deterioration of the “glucolipotoxic” environment, ultimately synergistically driving intrahepatic lipid deposition in MAFLD and the loss of glycaemic control in T2DM, thereby establishing comorbidity.
4.1.2 Short-chain fatty acids
SCFAs (50), primarily comprising acetate, propionate, and butyrate, are key metabolites produced by the gut microbiota through the fermentation of dietary fiber. They play a central role in maintaining glucose and lipid metabolic homeostasis.
4.1.2.1 Effects on the gut–pancreatic islet axis
SCFAs preserve pancreatic function and glucose homeostasis via multiple mechanisms, particularly butyrate. First, butyrate and others activate the GPR43 receptor on intestinal L cells, stimulating the release of GLP-1, thereby enhancing glucose-dependent insulin secretion in mice (51). Second, in vivo animal studies have demonstrated that SCFAs, acting as a histone deacetylase (HDAC) inhibitor, increases histone acetylation levels to promote the expression of anti-inflammatory genes such as the NF-κB inhibitor protein (IκBα). This subsequently inhibits NF-κB signaling and reduces the production of inflammatory cytokines such as IL-1β. Concurrently, SCFAs can competitively inhibit the binding of LPS to TLR4, synergistically decreasing the synthesis of TNF-α and IL-6, thereby alleviating local islet inflammation and protecting mice β-cells from inflammation-induced apoptosis (52–55). Furthermore, butyrate activates the nuclear factor erythroid 2-related factor 2 (Nrf2) pathway, promoting its nuclear translocation and upregulating the expression of antioxidant enzymes such as superoxide dismutase (SOD) and glutathione peroxidase (GPx), effectively mitigating oxidative stress and mitochondrial damage in mice β-cells (56).
4.1.2.2 Effects on the gut–adipose axis
Regarding lipid metabolism, SCFAs alleviate lipotoxicity by coordinating the functions of adipose tissue and the liver. An in vivo animal experiment has confirmed that, in adipose tissue, SCFAs activate GPR43 and GPR41 receptors. Through pathways involving PPARγ, they upregulate lipogenesis and promote fatty acid oxidation, ultimately inhibiting lipolysis, promoting normal adipocyte differentiation, helping to reduce circulating FFA levels, and improving ectopic lipid deposition and insulin sensitivity (57, 58). Butyrate can also serve as a preferential energy substrate, promoting the expression of peroxisome proliferator-activated receptor gamma coactivator 1-alpha (PGC-1α) and mitochondrial uncoupling protein 1 (UCP1) in brown adipose tissue, thereby accelerating fat metabolism and thermogenesis (59). Their immunomodulatory role is equally crucial: by inhibiting TLR4/NF-κB signaling, SCFAs drive the polarization of adipose tissue macrophages from the pro-inflammatory M1 phenotype to the anti-inflammatory M2 phenotype, thus improving IR and indirectly regulating fat accumulation (60). Additionally, upon reaching the liver, propionate can directly inhibit the expression of fatty acid synthase (FAS) and sterol regulatory element-binding protein 1c (SREBP-1c), effectively reducing de novo lipogenesis in the liver and intervening at the source in the progression of MAFLD (61).
Comorbidity mechanism: A reduction in the abundance of SCFAs weakens their dual protective effects on both the gut-pancreatic islet axis and the gut-adipose axis. In the pancreas, diminished stimulation of GLP-1, anti-inflammatory activity, and antioxidant activity collectively exacerbate β-cell dysfunction and insulin resistance. In adipose tissue, a decrease in the ability to promote healthy fat storage, thermogenesis, and anti-inflammatory effects leads to increased lipolysis, ectopic deposition, and hepatic de novo lipogenesis. This collective loss of protective signaling disrupts the balance of glucose and lipid metabolism, ultimately synergistically driving the comorbid progression of MAFLD and T2DM.
4.1.3 Branched-chain amino acids
Blood concentrations of BCAAs, which are essential amino acids, are influenced not only by dietary intake but also by the metabolic activity of the gut microbiota (e.g., Prevotella and Bacteroides). Gut dysbiosis leads to reduced BCAAs breakdown, causing abnormally elevated circulating BCAAs levels. This exerts profound effects on glucose and lipid metabolism through excessive activation of mechanistic target of rapamycin complex 1 (mTORC1) signaling.
4.1.3.1 Effects on the gut–pancreatic islet axis
Elevated BCAAs levels have multiple adverse effects on pancreatic function. In pancreatic β cells, chronic high concentrations of BCAAs induce sustained activation of mTORC1 signaling. On the one hand, this inhibits the key autophagy kinase ULK1, impairing the clearance of damaged mitochondria and leading to mitophagy defects and dysfunction. On the other hand, it causes excessive protein accumulation in the endoplasmic reticulum, inducing endoplasmic reticulum stress (ERS). Together, these ultimately lead to β-apoptosis and inhibit insulin synthesis and secretion (62, 63). Extensive in vitro and in vivo studies have demonstrated that in peripheral tissues, excessive mTORC1 activation inhibits PI3K/Akt insulin signaling pathway transduction, reduces Akt phosphorylation, and consequently inhibits the Rab protein-mediated translocation of GLUT4 to the cell membrane, reducing glucose uptake and directly causing IR (64–66).
4.1.3.2 Effects on the gut–adipose axis
BCAAs alter fat metabolism in a way that promotes storage and inhibits expenditure. After entering adipocytes, the catabolic product of BCAAs, Ac-CoA, can acetylate the brown fat transcription factor PRDM16, enhancing its binding to PPARγ. This process promotes white adipogenesis while inhibiting the transformation of white to brown/beige fat, thereby reducing energy expenditure and promoting lipid droplet formation and fat accumulation (67). Furthermore, Furthermore, extensive in vivo animal studies demonstrate that leucine can bind to and relieve the inhibition of the mTORC1 inhibitory protein Sestrin2, thereby activating SREBP-1c. This promotes the conversion of acetyl-CoA to FFAs and the subsequent esterification of these fatty acids into TG, resulting in the formation of lipid deposits in adipocytes and hepatocytes and directly exacerbating the pathological process of MAFLD (68–70).
Comorbidity mechanism: Abnormally elevated circulating BCAAs levels constitute a common pathological signal linking metabolic disorders in the pancreas and adipose tissue. In the pancreas, they impair β-cell function and survival via mTORC1 and exacerbate peripheral insulin resistance. In adipose tissue, they alter the metabolic program of fat cells, promoting lipid storage and inhibiting energy expenditure. This synchronous occurrence of “impaired islet function” and “lipid metabolism shifted toward storage” provides the core driving force for intrahepatic lipid deposition in MAFLD and loss of glycaemic control in T2DM, forming a unique comorbidity pathway.
4.1.4 Secondary bile acids
SBAs are key signaling molecules produced by the gut microbiota (e.g., Clostridium species) from primary bile acids. They primarily regulate the host’s glucose and lipid metabolic balance by activating FXR and TGR5. In vivo studies have shown that disrupting the gut microbiota perturbs bile acid metabolism, inhibits FXR signaling, and consequently triggers glucose and lipid metabolic disorders, whereas supplementation with FXR agonists can significantly ameliorate these abnormalities (71).
4.1.4.1 Effects on the gut–pancreatic islet axis
SBAs exert important regulatory effects on pancreatic function and glucose homeostasis through receptor signaling, as demonstrated in in vivo animal studies. First, TGR5 receptor activation upregulates the expression of IκBα, anchoring NF-κB in the cytoplasm and preventing its nuclear translocation, thereby inhibiting the transcription of pro-inflammatory genes. This anti-inflammatory mechanism helps protect β cells from inflammatory damage (72). More importantly, SBAs can improve glucose homeostasis by activating the intestinal FXR/TGR5-GLP-1 axis, stimulating intestinal L cells to secrete GLP-1, which subsequently promotes the closure of ATP-sensitive potassium channels in pancreatic β cells, leading to membrane depolarization and calcium influx and ultimately enhancing glucose-stimulated insulin secretion (73).
4.1.4.2 Effects on the gut–adipose axis
In terms of lipid metabolism, SBAs co-ordinately regulate lipid storage and consumption through FXR in different organs. An in vivo animal study revealed that in adipose tissue, SBAs activate FXR and, through pathways such as upregulating ApoC2, significantly increase the expression of mitochondrial UCP1 and PGC1α in beige adipocytes, promoting the browning of WAT and thereby increasing energy expenditure (74). In the liver, FXR activation upregulates PPARα, enhances mitochondrial β-oxidation capacity in hepatocytes, accelerates FFA clearance, and reduces the hepatic lipid burden (75, 76). Furthermore, the activation of intestinal FXR can directly regulate the intestinal absorption of lipids, modulating lipid metabolism from the source and showing great potential for preventing and treating MAFLD (77, 78).
Comorbidity mechanism: Dysregulation of secondary bile acid metabolism essentially represents the disruption of an important signaling network connecting the gut, pancreas, and adipose tissue. When their production is abnormal, the protective effects on the gut-pancreatic islet axis are weakened, exacerbating hyperglycaemia and β-cell damage. Moreover, the ability to promote adipose thermogenesis and enhance hepatic lipolysis via the gut–adipose axis is also reduced, leading to ectopic lipid deposition. Therefore, the imbalance in the secondary bile acid signaling network simultaneously impairs the fine regulation of both glucose metabolism and lipid metabolism, promoting the synergistic progression of MAFLD and T2DM.
4.2 Hormones: effectors regulating glucose and lipid metabolism
4.2.1 Endocannabinoid system
The endocannabinoid system (ECS) is a complex signaling network composed of endocannabinoids, their synthesis and degradation enzymes, and cannabinoid receptors (e.g., CB1R and CB2R). It is widely distributed in the brain, gastrointestinal tract, liver, adipose tissue, pancreas, and other tissues and is among the core systems that regulate energy balance, glucose/lipid metabolism, and immune inflammation (79). Gut microbiota dysbiosis can alter ECS tone, whereas dietary interventions and probiotics can modulate receptor expression, thereby improving metabolic syndrome (80).
4.2.1.1 Effects on the gut–pancreatic islet axis
The ECS bidirectionally regulates pancreatic secretory function, with the effect depending on the activated receptor subtype. CB2R activation has protective effects on glucose metabolism; in pancreatic β-cells, CB2R activation enhances glucose-induced Ca2+ oscillations, thereby promoting insulin secretion (81). Furthermore, in immune cells (e.g., ILC2s), CB2R agonism can improve established IR and restore glucose tolerance by activating the AKT, ERK1/2, and CREB pathways (82). In contrast, excessive activation of CB1R is typically associated with metabolic deterioration.
4.2.1.2 Effects on the gut–adipose axis
The ECS, particularly CB1R, plays a key role in regulating lipid metabolism in adipose tissue and the liver. CB1R activation is a core pathway that promotes lipid storage and exacerbates MAFLD; experiments conducted at both the cellular and animal levels have demonstrated that, in adipose tissue, CB1R activation stimulates p38 MAPK phosphorylation, reduces adiponectin production, and inhibits the expression of the thermogenic gene UCP1, thereby reducing energy expenditure and promoting fat accumulation (83, 84). In the liver, CB1R activation promotes SREBP-1c expression, driving de novo lipogenesis (DNL), while simultaneously inhibiting fatty acid β-oxidation and reducing FFA clearance, directly leading to intrahepatic lipid deposition (85, 86). Therefore, CB1R inhibitors such as rimonabant can effectively improve systemic lipid metabolic homeostasis by reversing these processes.
Comorbidity mechanism: An imbalance in ECS tone is an important bridge connecting pancreatic dysfunction with adipose/liver metabolic disorders. When CB1R signaling is overactive but protective CB2R signaling is insufficient, it simultaneously leads to the following: in the pancreas, dysregulated insulin secretion control and exacerbated insulin resistance; in adipose tissue and the liver, increased lipid storage, decreased energy expenditure, and enhanced hepatic de novo lipogenesis. This synergistic action of signals promoting lipid deposition and impairing glucose-lowering function drives the comorbidity of MAFLD and T2DM, making the ECS a highly promising therapeutic target.
4.2.2 Incretins
Incretins, primarily GLP-1 and GIP, are core hormones secreted by the gut in response to nutrient intake. They play a pivotal role in communication between the gut and distant metabolic organs.
4.2.2.1 Glucose-dependent insulinotropic polypeptide
GIP is secreted by K cells in the proximal small intestine, and its release is triggered primarily by glucose and lipids. With respect to the gut–pancreatic islet axis, GIP binds to the GIPR on β cells, activating the cAMP-PKA/EPAC2 signaling pathway and significantly enhancing glucose-stimulated insulin secretion. Furthermore, a study combining in vitro cellular models with human and mice pancreatic islet tissue confirmed that under hypoglycaemic conditions, GIP can act on pancreatic α cells to stimulate glucagon release and, via activation of the ERK/CREB pathway, exert protective and regenerative effects on β cells similar to the effects of GLP-1, collectively maintaining glucose homeostasis (87–89).
Regarding the gut–adipose axis, GIP activates GIPR in adipocytes and the cAMP/PKA/CREB pathway, upregulating the expression and activity of lipoprotein lipase (LPL). This promotes the storage of TGs in adipose tissue, reduces ectopic lipid deposition at sites such as the liver and muscle, and helps alleviate the severity of MAFLD (90–93).
4.2.2.2 Glucagon-like peptide-1
GLP-1 is secreted by L cells in the distal ileum and colon. Its role in the gut–pancreatic islet axis, as mentioned earlier, centers on its potent stimulation of insulin secretion. Its effects on the gut–adipose axis are manifested through multiple pathways that improve lipid metabolism: GLP-1 can activate the GLP-1 receptor (GLP-1R) in adipocytes, increase AMPK-PGC1α-UCP1 signaling activity, and induce adipose thermogenesis and lipolysis (94). Moreover, it can enhance intestinal barrier function by upregulating the activity of the PI3K/AKT/HIF-1α pathway, reducing circulating LPS levels, and indirectly alleviating WAT inflammation (95). Its receptor agonists, such as liraglutide, can also upregulate adiponectin by inhibiting the MKK4/JNK signaling pathway, improving adipose tissue-liver axis function, and reducing hepatic fat deposition (96).
Comorbidity mechanism and therapeutic prospects: Although GLP-1 and GIP stimulate insulin secretion, they exhibit a “division of labor” in fat metabolism: GLP-1 (Figure 2) promotes energy expenditure and lipid breakdown, whereas GIP promotes the safe storage of lipids in adipose tissue. In pathological states, the responsiveness of both may be diminished; i.e., the “incretin effect” is reduced, collectively leading to islet functional decompensation and abnormal lipid metabolism distribution. This explains why dual GLP-1/GIP receptor agonists such as cotadutide, by synergistically activating both pathways, can not only significantly lower blood glucose but also more effectively reduce body weight and improve hepatic steatosis and inflammation, making them promising therapies for the comorbidity of T2DM, obesity, and MAFLD (97).
Figure 2. Mechanism of inulin in the dual-axis system. (GLP-1 secreted by intestinal L cells and GIP secreted by intestinal K cells bind to receptors on target cells, activating the Gs protein-adenylate cyclase-cAMP-PKA signaling pathway, thereby promoting insulin secretion from pancreatic β cells. Within adipocytes, GIP-1 enhances LPL-mediated triglyceride storage via this pathway. Furthermore, activation of GLP-1R in adipocytes triggers the TAK1-MKK4-JNK pathway to regulate adiponectin expression, while AMPK-mediated phosphorylation of PGC1α promotes PPARγ-dependent UCP1 expression and thermogenesis. AC, adenylate cyclase; AMPK, AMP-activated protein kinase; cAMP, cyclic adenosine monophosphate; Gs, G-protein stimulatory; JNK, c-Jun N-terminal kinase; LPL, lipoprotein lipase; MKK4, mitogen-activated protein kinase kinase 4; PGC1α, PPARγ coactivator-1α; PKA, protein kinase A; PPARγ, peroxisome proliferator-activated receptor gamma; TAK1, TGF-β-activated kinase 1; TG, triglyceride; UCP1, uncoupling protein 1.).
5 Therapeutic prospects
5.1 FMT and probiotics
Current strategies for managing metabolic syndrome focus primarily on weight loss, such as through increased physical activity and dietary changes. However, maintaining these habits in the long term is challenging. Thus, bariatric surgery has proven to be the most effective method for controlling obesity and thereby improving obesity-related comorbidities. Common surgical techniques include sleeve gastrectomy (SG) and Roux-en-Y gastric bypass (RYGB). SG is a restrictive procedure that removes most of the stomach. RYGB creates a small gastric pouch connected to the distal jejunum, resulting in both restrictive and malabsorptive effects. This approach can influence GLP-1 secretion, further improving glucose and lipid metabolism. Given the irreversible nature of these surgeries, fecal microbiota transplantation (FMT) has been explored in recent years (98). FMT involves transplanting microbiota from healthy donors via colonoscopy, enema, or specialized oral preparations to restore gut microbiota diversity and health. This microbiota-targeted approach is more acceptable for patients with metabolic syndrome and shows great potential for treating metabolic diseases.
However, FMT remains at an experimental stage for treating metabolic syndrome, similarly constrained by small sample sizes and limited intervention duration. Consequently, precision modulation of the gut microbiota through clearly defined probiotics—moving beyond broad-spectrum approaches—has emerged as a highly promising strategy. The selection criteria for such probiotics increasingly emphasize restoring specific functions impaired in metabolic syndrome. Examples include supplementing strains that produce SCFAs – such as certain Lactobacillus and Bifidobacterium species – to enhance intestinal barrier function and systemic metabolism. In a rat model, Liang et al. (99) confirmed that oral probiotics can modulate the composition of the gut microbiota, alter SCFAs levels, reduce body lipid deposition, and promote insulin release, demonstrating significant effects on reducing hepatic lipid deposition and blood glucose levels. In vivo animal studies revealed (100) that oral polysaccharide administration activates the SCFA/GPR43/GLP-1 pathway, thereby normalizing blood glucose levels in mice exhibiting glucose and lipid metabolism abnormalities induced by tacrolimus-induced disruption of gut butyrate-producing microbiota. Furthermore, mechanistic studies confirmed that administering specific Bifidobacterium strains can alleviate metabolic syndrome, and Bifidobacterium animalis ssp. lactis GCL2505 can further reduce visceral fat and improve glucose tolerance (101). A recent Japanese cross-sectional study (102) revealed Weizmannia (formerly Bacillus) as a genus highly associated with metabolic syndrome. Oral administration of this bacterium was shown to affect mice liver fat accumulation and glucose tolerance by altering amino acid metabolism. Although related experiments are ongoing and targeted intestinally derived pharmaceuticals have not yet been developed, their potential role cannot be ignored. These approaches focusing on gut regulation—such as restoring microbial balance, blocking LPS leakage, and reshaping systemic metabolic homeostasis by breaking the vicious cycle—may become novel noninvasive strategies for intervening in MAFLD and T2DM in the future.
5.2 Receptor-targeted therapies
Rimonabant, initially approved in the EU in 2007 for improving lipid metabolism and insulin sensitivity, was later withdrawn from the market due to severe central nervous system side effects, including the worsening of pre-existing depression and suicidal tendencies, resulting from CB1 receptor blockade (103, 104). This case underscores the importance of precise receptor targeting in metabolic disease therapy. Research on CB1R, however, continues. For instance, INV-202, a peripherally restricted CB1R inhibitor currently in clinical trials, has not shown CNS-related side effects to date, indicating its potential as a novel therapeutic agent (105).
Owing to the effector role of incretins in the gut–adipose and gut–pancreatic axes, the safe and well-tolerated agents GLP-1 and GIP, beyond their use as glucose-lowering drugs, are now widely investigated for the treatment of MAFLD. Liraglutide (a long-acting GLP-1 analogue) and semaglutide (a GLP-1 receptor agonist) have been confirmed by clinical controlled trials to slow hepatic steatosis and non-alcoholic steatohepatitis, in addition to affecting insulin release (106, 107). Furthermore, a randomized controlled trial this year (108) demonstrated that treatment with the dual GLP-1/GIP receptor agonist pemvidutide significantly improved blood glucose levels, reduced liver fat content, improved liver inflammatory activity, and reduced body weight. These findings indicate that incretin-related agents may be effective for treating metabolic dysfunction-associated steatohepatitis and obesity.
5.3 Dietary intervention
Dietary intervention is a cornerstone strategy for managing MAFLD and T2DM and targeting the “gut–adipose/pancreatic axis”. Its core lies in reshaping the gut microbiota ecosystem by optimizing dietary structure, thereby positively regulating downstream metabolic signals. The Mediterranean diet (MD), which is rich in dietary fiber, polyunsaturated fatty acids, and polyphenols, is a prime example. Adherence to the MD is associated with reduced blood sugar and liver fat deposition rates in patients (109, 110). Mitsou et al. (111) reported that individuals with high MD scores had lower Escherichia coli counts, higher Bifidobacterium/Escherichia coli ratios, and higher acetate molar ratios than subjects with low adherence. De Filippis et al. (112) reported that among subjects consuming a seemingly Western diet, high consumption of plant-based foods consistent with the MD was associated with a beneficial microbiome-related metabolomic profile (higher abundance of Prevotella and certain fiber-degrading Firmicutes and higher SCFAs production). The elevated intestinal SCFAs levels in MD adherents are determined by high consumption of vegetables, fruits, and legumes, which are rich sources of complex and insoluble fibers, the primary substrates for microbial SCFAs production. Therefore, remodeling the gut microenvironment through diet can send beneficial metabolic regulation instructions simultaneously to both the gut–adipose axis and the gut–pancreatic axis, representing an effective nonpharmacological means to break the vicious cycle of glucose and lipid metabolism.
6 Discussion
In recent years, extensive research has investigated the gut–organ axes, which encompass various functional categories and exert broad influences on host behavior, metabolism, appetite, growth, reproduction, and immunity. Currently, numerous studies focus on revealing the role and mechanisms of gut-related dysfunction in metabolic diseases to explore new treatment strategies. The roles of adipose tissue and the pancreas in gut–organ axis crosstalk have gained widespread recognition and significant focus. In summary, this review moves beyond traditional cascade models to propose the new paradigm of “Common Messengers, Dual-Axis Branching” for MAFLD and T2DM comorbidity. This paradigm emphasizes that a limited set of core gut-derived messengers, by activating different downstream axes—namely, the gut–adipose axis and the gut–pancreatic islet axis—simultaneously yet distinctly disrupt lipid metabolism and glucose metabolism, respectively. This model not only explains the high frequency of comorbidity but also provides a revolutionary theoretical basis for developing therapies that simultaneously target multiple pathways. Although microbiome research is still in the development stage, numerous studies have confirmed associations between hormones and the gut microbiota. The perspective of centring on the gut and formulating preventive strategies aimed at reducing metabolic complications via the gut–systemic axes is gradually gaining prominence. Future work needs to further explore the mechanisms of cross-talk between other organ axes in metabolism-related diseases.
Author contributions
JL: Writing – original draft. XC: Writing – review & editing. YC: Writing – review & editing. YL: Writing – review & editing.
Funding
The author(s) declare financial support was received for the research and/or publication of this article. Jilin Provincial Science and Technology Development Program Project (No.: 20240304068SF); Jilin Provincial Department of Education Science and Technology Research Project (No.: JJKH20250641KJ).
Conflict of interest
The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest.
Generative AI statement
The author(s) declare that no Generative AI was used in the creation of this manuscript.
Any alternative text (alt text) provided alongside figures in this article has been generated by Frontiers with the support of artificial intelligence and reasonable efforts have been made to ensure accuracy, including review by the authors wherever possible. If you identify any issues, please contact us.
Publisher’s note
All claims expressed in this article are solely those of the authors and do not necessarily represent those of their affiliated organizations, or those of the publisher, the editors and the reviewers. Any product that may be evaluated in this article, or claim that may be made by its manufacturer, is not guaranteed or endorsed by the publisher.
References
1. Moreira de Gouveia MI, Bernalier-Donadille A, and Jubelin G. Enterobacteriaceae in the human gut: dynamics and ecological roles in health and disease. Biol (Basel). (2024) 13:142. doi: 10.3390/biology13030142
2. Luo J, Liang S, and Jin F. Gut microbiota and healthy longevity. Sci China Life Sci. (2024) 67:2590–602. doi: 10.1007/s11427-023-2595-5
3. Lim GEH, Tang A, Ng CH, Chin YH, Lim WH, Tan DJH, et al. An observational data meta-analysis on the differences in prevalence and risk factors between MAFLD vs NAFLD. Clin Gastroenterol Hepatol. (2023) 21:619–29.e7. doi: 10.1016/j.cgh.2021.11.038
4. Lei F, Qin JJ, Song X, Liu YM, Chen MM, Sun T, et al. The prevalence of mafld and its association with atrial fibrillation in a nationwide health check-up population in China. Front Endocrinol (Lausanne). (2022) 13:1007171. doi: 10.3389/fendo.2022.1007171
5. Eslam M, Newsome PN, Sarin SK, Anstee QM, Targher G, Romero-Gomez M, et al. A new definition for metabolic dysfunction-associated fatty liver disease: an international expert consensus statement. J Hepatol. (2020) 73:202–9. doi: 10.1016/j.jhep.2020.03.039
6. Roy A. Advances in non-alcoholic fatty liver disease (NAFLD) research and management: A global perspective(2020–2025). J Emerging Pharm Med Res (JEPMR). (2025) 1:1–34.
7. Feng J, Qiu S, Zhou S, Tan Y, Bai Y, Cao H, et al. Mtor: A potential new target in nonalcoholic fatty liver disease. Int J Mol Sci. (2022) 23:9196. doi: 10.3390/ijms23169196
8. Filipovic B, Marjanovic-Haljilji M, Mijac D, Lukic S, Kapor S, Kapor S, et al. Molecular aspects of MAFLD-new insights on pathogenesis and treatment. Curr Issues Mol Biol. (2023) 45:9132–48. doi: 10.3390/cimb45110573
9. Younossi ZM, Golabi P, Price JK, Owrangi S, Gundu-Rao N, Satchi R, et al. The global epidemiology of nonalcoholic fatty liver disease and nonalcoholic steatohepatitis among patients with type 2 diabetes. Clin Gastroenterol Hepatol. (2024) 22:1999–2010.e8. doi: 10.1016/j.cgh.2024.03.006
10. Masarone M, Rosato V, Aglitti A, Bucci T, Caruso R, Salvatore T, et al. Liver biopsy in type 2 diabetes mellitus: steatohepatitis represents the sole feature of liver damage. PloS One. (2017) 12:e0178473. doi: 10.1371/journal.pone.0178473
11. Romeo M, Di Nardo F, Napolitano C, Basile C, Palma C, Vaia P, et al. Exploring the epidemiologic burden, pathogenetic features, and clinical outcomes of primary liver cancer in patients with type 2 diabetes mellitus (T2DM) and metabolic dysfunction-associated steatotic liver disease (MASLD): A scoping review. Diabetology. (2025) 6:79. doi: 10.3390/diabetology6080079
12. Huang Z, Yao Q, Ma S, Zhou J, Wang X, Meng Q, et al. The synergistic role of gut microbiota and RNA in metabolic diseases: mechanisms and therapeutic insights. Front Microbiol. (2025) 16:1504395. doi: 10.3389/fmicb.2025.1504395
13. Cani PD and Van Hul M. Gut microbiota in overweight and obesity: crosstalk with adipose tissue. Nat Rev Gastroenterol Hepatol. (2024) 21:164–83. doi: 10.1038/s41575-023-00867-z
14. Ronen D, Rokach Y, Abedat S, Qadan A, Daana S, Amir O, et al. Human gut microbiota in cardiovascular disease. Compr Physiol. (2024) 14:5449–90. doi: 10.1002/cphy.c230012
15. Konrad D and Wueest S. The gut-adipose-liver axis in the metabolic syndrome. Physiol (Bethesda). (2014) 29:304–13. doi: 10.1152/physiol.00014.2014
16. Pedersen HK, Gudmundsdottir V, Nielsen HB, Hyotylainen T, Nielsen T, Jensen BA, et al. Human gut microbes impact host serum metabolome and insulin sensitivity. Nature. (2016) 535:376–81. doi: 10.1038/nature18646
17. Shaheen N, Khursheed W, Gurung B, and Wang S. Akkermansia muciniphila: A key player in gut microbiota-based disease modulation. Microbiol Res. (2025) 301:128317. doi: 10.1016/j.micres.2025.128317
18. Liu L, Sadaghian Sadabad M, Gabarrini G, Lisotto P, von Martels JZH, Wardill HR, et al. Riboflavin supplementation promotes butyrate production in the absence of gross compositional changes in the gut microbiota. Antioxid Redox Signal. (2023) 38:282–97. doi: 10.1089/ars.2022.0033
19. Yuan M, Sun T, Zhang Y, Guo C, Wang F, Yao Z, et al. Quercetin alleviates insulin resistance and repairs intestinal barrier in db/db mice by modulating gut microbiota. Nutrients. (2024) 16:1870. doi: 10.3390/nu16121870
20. Si ZL, Wang HY, Wang T, Cao YZ, Li QZ, Liu K, et al. Gut bacteroides ovatus ameliorates renal fibrosis by promoting the production of HDCA through upregulation of clostridium scindens. Cell Rep. (2024) 43:114830. doi: 10.1016/j.celrep.2024.114830
21. Liu R, Hong J, Xu X, Feng Q, Zhang D, Gu Y, et al. Gut microbiome and serum metabolome alterations in obesity and after weight-loss intervention. Nat Med. (2017) 23:859–68. doi: 10.1038/nm.4358
22. Fiamoncini J, Rundle M, Gibbons H, Thomas EL, Geillinger-Kästle K, Bunzel D, et al. Plasma metabolome analysis identifies distinct human metabotypes in the postprandial state with different susceptibility to weight loss-mediated metabolic improvements. FASEB J. (2018) 32:5447–58. doi: 10.1096/fj.201800330R
23. Surowiec I, Noordam R, Bennett K, Beekman M, Slagboom PE, Lundstedt T, et al. Metabolomic and lipidomic assessment of the metabolic syndrome in dutch middle-aged individuals reveals novel biological signatures separating health and disease. Metabolomics. (2019) 15:23. doi: 10.1007/s11306-019-1484-7
24. Org E, Blum Y, Kasela S, Mehrabian M, Kuusisto J, Kangas AJ, et al. Relationships between gut microbiota, plasma metabolites, and metabolic syndrome traits in the metsim cohort. Genome Biol. (2017) 18:70. doi: 10.1186/s13059-017-1194-2
25. Zhang C, Yin A, Li H, Wang R, Wu G, Shen J, et al. Dietary modulation of gut microbiota contributes to alleviation of both genetic and simple obesity in children. EBioMedicine. (2015) 2:968–84. doi: 10.1016/j.ebiom.2015.07.007
26. Gao LL, Ma JM, Fan YN, Zhang YN, Ge R, Tao XJ, et al. Lycium barbarum polysaccharide combined with aerobic exercise ameliorated nonalcoholic fatty liver disease through restoring gut microbiota, intestinal barrier and inhibiting hepatic inflammation. Int J Biol Macromol. (2021) 183:1379–92. doi: 10.1016/j.ijbiomac.2021.05.066
27. Barber TM, Kabisch S, Pfeiffer AFH, and Weickert MO. Metabolic-associated fatty liver disease and insulin resistance: A review of complex interlinks. Metabolites. (2023) 13:757. doi: 10.3390/metabo13060757
28. Dua A, Kumari R, Singh M, Kumar R, Pradeep S, Ojesina AI, et al. Metabolic dysfunction-associated steatotic liver disease (MASLD): the interplay of gut microbiome, insulin resistance, and diabetes. Front Med (Lausanne). (2025) 12:1618275. doi: 10.3389/fmed.2025.1618275
29. Chu Z, Hu Z, Luo Y, Zhou Y, Yang F, and Luo F. Targeting gut-liver axis by dietary lignans ameliorate obesity: evidences and mechanisms. Crit Rev Food Sci Nutr. (2025) 65:243–64. doi: 10.1080/10408398.2023.2272269
30. Zhang W, Zhu M, Liu X, Que M, Dekyi K, Zheng L, et al. Edible bird’s nest regulates glucose and lipid metabolic disorders via the gut-liver axis in obese mice. Food Funct. (2024) 15:7577–91. doi: 10.1039/d4fo00563e
31. Unger RH and Eisentraut AM. Entero-insular axis. Arch Intern Med. (1969) 123:261–6. doi: 10.1001/archinte.1969.00300130043007
32. Bany Bakar R, Reimann F, and Gribble FM. The intestine as an endocrine organ and the role of gut hormones in metabolic regulation. Nat Rev Gastroenterol Hepatol. (2023) 20:784–96. doi: 10.1038/s41575-023-00830-y
33. Chen Q, Gao Y, Li F, and Yuan L. The role of gut-islet axis in pancreatic islet function and glucose homeostasis. Diabetes Obes Metab. (2025) 27:1676–92. doi: 10.1111/dom.16225
34. Yang S, Cao J, Sun C, and Yuan L. The regulation role of the gut-islets axis in diabetes. Diabetes Metab syndrome obesity: Targets Ther. (2024) 17:1415–23. doi: 10.2147/dmso.S455026
35. Guo H, Pan L, Wu Q, Wang L, Huang Z, Wang J, et al. Type 2 diabetes and the multifaceted gut-X axes. Nutrients. (2025) 17:2708. doi: 10.3390/nu17162708
36. Li Y, Fang Y, Wang H, and Zhang H. Balancing act: exploring the gut microbiota-brown adipose tissue axis in PCOS pathogenesis and therapeutic frontiers. Front bioscience (Landmark edition). (2024) 29:208. doi: 10.31083/j.fbl2906208
37. Bischoff SC, Barbara G, Buurman W, Ockhuizen T, Schulzke JD, Serino M, et al. Intestinal permeability–a new target for disease prevention and therapy. BMC Gastroenterol. (2014) 14:189. doi: 10.1186/s12876-014-0189-7
38. Hwang JS, Kwon MY, and Kim KH. Lipopolysaccharide (LPS)-Stimulated iNOS Induction Is Increased by Glucosamine under Normal Glucose Conditions but Is Inhibited by Glucosamine under High Glucose Conditions in Macrophage Cells. J Biol Chem. (2017) 292:1724–36. doi: 10.1074/jbc.M116.737940
39. Chen X, Chen C, and Fu X. Dendrobium officinale polysaccharide alleviates type 2 diabetes mellitus by restoring gut microbiota and repairing intestinal barrier via the LPS/TLR4/TRIF/NF-kB axis. J Agric Food Chem. (2023) 71:11929–40. doi: 10.1021/acs.jafc.3c02429
40. Xuguang H, Aofei T, Tao L, Longyan Z, Weijian B, and Jiao G. Hesperidin ameliorates insulin resistance by regulating the IRS1-GLUT2 pathway via TLR4 in HepG2 cells. Phytotherapy research: PTR. (2019) 33:1697–705. doi: 10.1002/ptr.6358
41. Wan L, Zeng H, Peng L, Yang X, Bai Q, Liu L, et al. Theabrownin alleviates type 2 diabetes mellitus in db/db mice via modulating LPS/GLP-1 levels and restoring islet cells: evidence from gut-pancreas axis. J Agric Food Chem. (2025) 73:13455–70. doi: 10.1021/acs.jafc.4c12501
42. Wang J, Wang X, Li ZZ, Guo F, Ding CZ, Zhao YY, et al. The apoptosis and GLP-1 hyposecretion induced by LPS via RIP/ROS/mTOR pathway in GLUTag cells. Biochimie. (2019) 162:229–38. doi: 10.1016/j.biochi.2019.04.001
43. Tomassen MMM, Govers C, Vos AP, and de Wit NJW. Dietary fat induced chylomicron-mediated LPS translocation in a bicameral Caco-2cell model. Lipids Health Dis. (2023) 22:4. doi: 10.1186/s12944-022-01754-3
44. Jia X, Xu W, Zhang L, Li X, Wang R, and Wu S. Impact of gut microbiota and microbiota-related metabolites on hyperlipidemia. Front Cell Infect Microbiol. (2021) 11:634780. doi: 10.3389/fcimb.2021.634780
45. Giby VG and Ajith TA. Role of adipokines and peroxisome proliferator-activated receptors in nonalcoholic fatty liver disease. World J Hepatol. (2014) 6:570–9. doi: 10.4254/wjh.v6.i8.570
46. Daval M, Foufelle F, and Ferré P. Functions of amp-activated protein kinase in adipose tissue. J Physiol. (2006) 574:55–62. doi: 10.1113/jphysiol.2006.111484
47. Hong S, Dimitrov S, Pruitt C, Shaikh F, and Beg N. Benefit of physical fitness against inflammation in obesity: role of beta adrenergic receptors. Brain Behav Immun. (2014) 39:113–20. doi: 10.1016/j.bbi.2013.12.009
48. Neuschwander-Tetri BA. Hepatic lipotoxicity and the pathogenesis of nonalcoholic steatohepatitis: the central role of nontriglyceride fatty acid metabolites. Hepatology. (2010) 52:774–88. doi: 10.1002/hep.23719
49. Wang Y, Kuang Z, Yu X, Ruhn KA, Kubo M, and Hooper LV. The intestinal microbiota regulates body composition through NFIL3 and the circadian clock. Science. (2017) 357:912–6. doi: 10.1126/science.aan0677
50. Gu Y, Chen H, Li X, Li D, Sun Y, Yang L, et al. Lactobacillus paracasei IMC 502 ameliorates type 2 diabetes by mediating gut microbiota-SCFA-hormone/inflammation pathway in mice. J Sci Food Agric. (2023) 103:2949–59. doi: 10.1002/jsfa.12267
51. Xia T, He W, Luo Z, Wang K, and Tan X. Achyranthes bidentata polysaccharide ameliorates type 2 diabetes mellitus by gut microbiota-derived short-chain fatty acids-induced activation of the GLP-1/GLP-1R/cAMP/PKA/CREB/INS pathway. Int J Biol Macromol. (2024) 270:132256. doi: 10.1016/j.ijbiomac.2024.132256
52. Pedersen SS, Prause M, Williams K, Barrès R, and Billestrup N. Butyrate inhibits IL-1β-induced inflammatory gene expression by suppression of NF-κB activity in pancreatic beta cells. J Biol Chem. (2022) 298:102312. doi: 10.1016/j.jbc.2022.102312
53. Roshanravan N, Alamdari NM, Jafarabadi MA, Mohammadi A, Shabestari BR, Nasirzadeh N, et al. Effects of oral butyrate and inulin supplementation on inflammation-induced pyroptosis pathway in type 2 diabetes: A randomized, double-blind, placebo-controlled trial. Cytokine. (2020) 131:155101. doi: 10.1016/j.cyto.2020.155101
54. Masui R, Sasaki M, Funaki Y, Ogasawara N, Mizuno M, Iida A, et al. G protein-coupled receptor 43 moderates gut inflammation through cytokine regulation from mononuclear cells. Inflammation Bowel Dis. (2013) 19:2848–56. doi: 10.1097/01.MIB.0000435444.14860.ea
55. Du Y, He C, An Y, Huang Y, Zhang H, Fu W, et al. The role of short chain fatty acids in inflammation and body health. Int J Mol Sci. (2024) 25:7379. doi: 10.3390/ijms25137379
56. Li Q, Zhang C, Zhu M, Shan J, Qian H, Ma Y, et al. W-GA nanodots restore intestinal barrier functions by regulating flora disturbance and relieving excessive oxidative stress to alleviate colitis. Acta Biomater. (2024) 182:260–74. doi: 10.1016/j.actbio.2024.05.030
57. Wang Z, Sun Y, Han Y, Chen X, Gong P, Zhai P, et al. Eucommia bark/leaf extract improves HFD-induced lipid metabolism disorders via targeting gut microbiota to activate the Fiaf-LPL gut-liver axis and SCFAS-GPR43 gut-fat axis. Phytomedicine. (2023) 110:154652. doi: 10.1016/j.phymed.2023.154652
58. Canfora EE, Jocken JW, and Blaak EE. Short-chain fatty acids in control of body weight and insulin sensitivity. Nat Rev Endocrinol. (2015) 11:577–91. doi: 10.1038/nrendo.2015.128
59. Wang B, Kong Q, Li X, Zhao J, Zhang H, Chen W, et al. A high-fat diet increases gut microbiota biodiversity and energy expenditure due to nutrient difference. Nutrients. (2020) 12:3197. doi: 10.3390/nu12103197
60. Kushwaha V, Rai P, Varshney S, Gupta S, Khandelwal N, Kumar D, et al. Sodium butyrate reduces endoplasmic reticulum stress by modulating chop and empowers favorable anti-inflammatory adipose tissue immune-metabolism in HFD fed mice model of obesity. Food Chem Mol Sci. (2022) 4:100079. doi: 10.1016/j.fochms.2022.100079
61. Guo Q, Li Y, Dai X, Wang B, Zhang J, and Cao H. Polysaccharides: the potential prebiotics for metabolic associated fatty liver disease (MAFLD). Nutrients. (2023) 15:3722. doi: 10.3390/nu15173722
62. Liu S, Li L, Lou P, Zhao M, Wang Y, Tang M, et al. Elevated branched-chain α-keto acids exacerbate macrophage oxidative stress and chronic inflammatory damage in type 2 diabetes mellitus. Free Radic Biol Med. (2021) 175:141–54. doi: 10.1016/j.freeradbiomed.2021.08.240
63. Bröer S. Amino acid transporters as modulators of glucose homeostasis. Trends Endocrinol Metab. (2022) 33:120–35. doi: 10.1016/j.tem.2021.11.004
64. Newgard CB, An J, Bain JR, Muehlbauer MJ, Stevens RD, Lien LF, et al. A branched-chain amino acid-related metabolic signature that differentiates obese and lean humans and contributes to insulin resistance. Cell Metab. (2009) 9:311–26. doi: 10.1016/j.cmet.2009.02.002
65. Wang TJ, Larson MG, Vasan RS, Cheng S, Rhee EP, McCabe E, et al. Metabolite profiles and the risk of developing diabetes. Nat Med. (2011) 17:448–53. doi: 10.1038/nm.2307
66. Zhenyukh O, Civantos E, Ruiz-Ortega M, Sánchez MS, Vázquez C, Peiró C, et al. High concentration of branched-chain amino acids promotes oxidative stress, inflammation and migration of human peripheral blood mononuclear cells via mTORC1 activation. Free Radic Biol Med. (2017) 104:165–77. doi: 10.1016/j.freeradbiomed.2017.01.009
67. Ma QX, Zhu WY, Lu XC, Jiang D, Xu F, Li JT, et al. BCAA-BCKA axis regulates WAT browning through acetylation of PRDM16. Nat Metab. (2022) 4:106–22. doi: 10.1038/s42255-021-00520-6
68. Lu J, Temp U, Müller-Hartmann A, Esser J, Grönke S, and Partridge L. Sestrin is a key regulator of stem cell function and lifespan in response to dietary amino acids. Nat Aging. (2021) 1:60–72. doi: 10.1038/s43587-020-00001-7
69. Hu H, Lu X, He Y, Li J, Wang S, Luo Z, et al. Sestrin2 in pomc neurons modulates energy balance and obesity related metabolic disorders via mtor signaling. J Nutr Biochem. (2024) 133:109703. doi: 10.1016/j.jnutbio.2024.109703
70. Qiao S, Liu C, Sun LZ, Wang T, Dai H, Wang K, et al. Gut parabacteroides merdae protects against cardiovascular damage by enhancing branched-chain amino acid catabolism. Nat Metab. (2022) 4:1271 – 86. doi: 10.1038/s42255-022-00649-y
71. Zhai Z, Niu KM, Liu H, Lin C, Tu Y, Liu Y, et al. Policosanol alleviates hepatic lipid accumulation by regulating bile acids metabolism in C57BL6/mice through AMPK-FXR-TGR5 cross-talk. J Food Sci. (2021) 86:5466–78. doi: 10.1111/1750-3841.15951
72. Pols TW, Nomura M, Harach T, Lo Sasso G, Oosterveer MH, Thomas C, et al. TGR5 activation inhibits atherosclerosis by reducing macrophage inflammation and lipid loading. Cell Metab. (2011) 14:747–57. doi: 10.1016/j.cmet.2011.11.006
73. Xie Z, Jiang H, Liu W, Zhang X, Chen D, Sun S, et al. The triterpenoid sapogenin (2α-OH-protopanoxadiol) ameliorates metabolic syndrome via the intestinal FXR/GLP-1 axis through gut microbiota remodelling. Cell Death Dis. (2020) 11:770. doi: 10.1038/s41419-020-02974-0
74. Kim SH, Park WY, Kim B, Kim JH, Song G, Park JY, et al. FXR-apoC2 pathway activates UCP1-mediated thermogenesis by promoting the browning of white adipose tissues. J Biol Chem. (2025) 301:108181. doi: 10.1016/j.jbc.2025.108181
75. Parlati L, Régnier M, Guillou H, and Postic C. New targets for NAFLD. JHEP reports: Innovation Hepatol. (2021) 3:100346. doi: 10.1016/j.jhepr.2021.100346
76. Younossi ZM, Ratziu V, Loomba R, Rinella M, Anstee QM, Goodman Z, et al. Obeticholic acid for the treatment of non-alcoholic steatohepatitis: interim analysis from a multicentre, randomised, placebo-controlled phase 3 trial. Lancet (London England). (2019) 394:2184–96. doi: 10.1016/s0140-6736(19)33041-7
77. Clifford BL, Sedgeman LR, Williams KJ, Morand P, Cheng A, Jarrett KE, et al. FXR activation protects against NAFLD via bile-acid-dependent reductions in lipid absorption. Cell Metab. (2021) 33:1671–84.e4. doi: 10.1016/j.cmet.2021.06.012
78. Collins SL, Stine JG, Bisanz JE, Okafor CD, and Patterson AD. Bile acids and the gut microbiota: metabolic interactions and impacts on disease. Nat Rev Microbiol. (2023) 21:236–47. doi: 10.1038/s41579-022-00805-x
79. Freund TF, Katona I, and Piomelli D. Role of endogenous cannabinoids in synaptic signaling. Physiol Rev. (2003) 83:1017–66. doi: 10.1152/physrev.00004.2003
80. Muccioli GG, Naslain D, Bäckhed F, Reigstad CS, Lambert DM, Delzenne NM, et al. The endocannabinoid system links gut microbiota to adipogenesis. Mol Syst Biol. (2010) 6:392. doi: 10.1038/msb.2010.46
81. Deveaux V, Cadoudal T, Ichigotani Y, Teixeira-Clerc F, Louvet A, Manin S, et al. Cannabinoid CB2 receptor potentiates obesity-associated inflammation, insulin resistance and hepatic steatosis. PloS One. (2009) 4:e5844. doi: 10.1371/journal.pone.0005844
82. Shafiei-Jahani P, Yan S, Kazemi MH, Li X, Akbari A, Sakano K, et al. Cb2 stimulation of adipose resident ILC2s orchestrates immune balance and ameliorates type 2 diabetes mellitus. Cell Rep. (2024) 43:114434. doi: 10.1016/j.celrep.2024.114434
83. Ge Q, Maury E, Rycken L, Gérard J, Noël L, Detry R, et al. Endocannabinoids regulate adipokine production and the immune balance of omental adipose tissue in human obesity. Int J Obes. (2013) 37:874–80. doi: 10.1038/ijo.2012.123
84. Iyer MS, Paszkiewicz RL, Bergman RN, Richey JM, Woolcott OO, Asare-Bediako I, et al. Activation of NPRs and UCP1-independent pathway following CB1R antagonist treatment is associated with adipose tissue beiging in fat-fed male dogs. Am J Physiol Endocrinol Metab. (2019) 317:E535–e47. doi: 10.1152/ajpendo.00539.2018
85. Eberlé D, Hegarty B, Bossard P, Ferré P, and Foufelle F. SREBP transcription factors: master regulators of lipid homeostasis. Biochimie. (2004) 86:839–48. doi: 10.1016/j.biochi.2004.09.018
86. Osei-Hyiaman D, DePetrillo M, Pacher P, Liu J, Radaeva S, Bátkai S, et al. Endocannabinoid activation at hepatic CB1 receptors stimulates fatty acid synthesis and contributes to diet-induced obesity. J Clin Invest. (2005) 115:1298–305. doi: 10.1172/jci23057
87. Killion EA, Lu SC, Fort M, Yamada Y, Véniant MM, and Lloyd DJ. Glucose-dependent insulinotropic polypeptide receptor therapies for the treatment of obesity, do agonists = Antagonists? Endocrine Rev. (2020) 41:1–21. doi: 10.1210/endrev/bnz002
88. Christensen MB. Glucose-dependent insulinotropic polypeptide: effects on insulin and glucagon secretion in humans. Danish Med J. (2016) 63:B5230.
89. Zaïmia N, Obeid J, Varrault A, Sabatier J, Broca C, Gilon P, et al. GLP-1 and GIP receptors signal through distinct β-arrestin 2-dependent pathways to regulate pancreatic β Cell function. Cell Rep. (2023) 42:113326. doi: 10.1016/j.celrep.2023.113326
90. Li Y, Gao R, Yang Z, Zong H, and Li Y. Liraglutide modulates lipid metabolism via ZBTB20-LPL pathway. Life Sci. (2025) 360:123267. doi: 10.1016/j.lfs.2024.123267
91. Kim SJ, Nian C, and McIntosh CH. GIP increases human adipocyte LPL expression through CREB and TORC2-mediated trans-activation of the LPL gene. J Lipid Res. (2010) 51:3145–57. doi: 10.1194/jlr.M006841
92. Campbell JE, Müller TD, Finan B, DiMarchi RD, Tschöp MH, and D’Alessio DA. GIPR/GLP-1R dual agonist therapies for diabetes and weight loss-chemistry, physiology, and clinical applications. Cell Metab. (2023) 35:1519–29. doi: 10.1016/j.cmet.2023.07.010
93. Nauck MA, Quast DR, Wefers J, and Pfeiffer AFH. The evolving story of incretins (GIP and GLP-1) in metabolic and cardiovascular disease: A pathophysiological update. Diabetes Obes Metab. (2021) 23 Suppl 3:5–29. doi: 10.1111/dom.14496
94. Drucker DJ. GLP-1 physiology informs the pharmacotherapy of obesity. Mol Metab. (2022) 57:101351. doi: 10.1016/j.molmet.2021.101351
95. Wang JX, Chang SY, Jin ZY, Li D, Zhu J, Luo ZB, et al. Lactobacillus reuteri-enriched eicosatrienoic acid regulates glucose homeostasis by promoting glp-1 secretion to protect intestinal barrier integrity. J Agric Food Chem. (2025) 73:393–408. doi: 10.1021/acs.jafc.4c03818
96. Zhao X, Wang M, Wen Z, Lu Z, Cui L, Fu C, et al. GLP-1 receptor agonists: beyond their pancreatic effects. Front Endocrinol (Lausanne). (2021) 12:721135. doi: 10.3389/fendo.2021.721135
97. Marcondes-de-Castro IA, Oliveira TF, Spezani R, Marinho TS, Cardoso LEM, Aguila MB, et al. Cotadutide effect in liver and adipose tissue in obese mice. J Mol Endocrinol. (2023) 70:220168. doi: 10.1530/jme-22-0168
98. Mocanu V, Zhang Z, Deehan EC, Kao DH, Hotte N, Karmali S, et al. Fecal microbial transplantation and fiber supplementation in patients with severe obesity and metabolic syndrome: A randomized double-blind, placebo-controlled phase 2 trial. Nat Med. (2021) 27:1272–9. doi: 10.1038/s41591-021-01399-2
99. Liang Y, Liang S, Zhang Y, Deng Y, He Y, Chen Y, et al. Oral administration of compound probiotics ameliorates HFD-induced gut microbe dysbiosis and chronic metabolic inflammation via the G protein-coupled receptor 43 in non-alcoholic fatty liver disease rats. Probiotics antimicrobial Proteins. (2019) 11:175–85. doi: 10.1007/s12602-017-9378-3
100. Jiao W, Zhang Z, Xu Y, Gong L, Zhang W, Tang H, et al. Butyric acid normalizes hyperglycemia caused by the tacrolimus-induced gut microbiota. Am J Transplant. (2020) 20:2413–24. doi: 10.1111/ajt.15880
101. Aoki R, Kamikado K, Suda W, Takii H, Mikami Y, Suganuma N, et al. A proliferative probiotic bifidobacterium strain in the gut ameliorates progression of metabolic disorders via microbiota modulation and acetate elevation. Sci Rep. (2017) 7:43522. doi: 10.1038/srep43522
102. Hosomi K, Saito M, Park J, Murakami H, Shibata N, Ando M, et al. Oral administration of blautia wexlerae ameliorates obesity and type 2 diabetes via metabolic remodeling of the gut microbiota. Nat Commun. (2022) 13:4477. doi: 10.1038/s41467-022-32015-7
103. Wang S, Liu X, He J, Cui Y, and Jia A. Serum metabolites and gut microbiota mediate the causal link between anxiety and nonalcoholic fatty liver disease: A mendelian randomization analysis. Eur J Gastroenterol Hepatol. (2025). doi: 10.1097/meg.0000000000003043
104. Mitchell PB and Morris MJ. Depression and anxiety with rimonabant. Lancet (London England). (2007) 370:1671–2. doi: 10.1016/s0140-6736(07)61705-x
105. Crater G, Lalonde K, Ravenelle F, Harvey M, and Després J-P. Effects of CB1R inverse agonist, INV-202, in patients with features of metabolic syndrome. A randomized, placebo-controlled, double-blind phase 1b study. Diabetes Obes Metab. (2023) 26:642–9. doi: 10.1111/dom.15353
106. Armstrong MJ, Gaunt P, Aithal GP, Barton D, Hull D, Parker R, et al. Liraglutide safety and efficacy in patients with non-alcoholic steatohepatitis (LEAN): A multicentre, double-blind, randomised, placebo-controlled phase 2 study. Lancet (London England). (2016) 387:679–90. doi: 10.1016/s0140-6736(15)00803-x
107. Newsome PN, Buchholtz K, Cusi K, Linder M, Okanoue T, Ratziu V, et al. A placebo-controlled trial of subcutaneous semaglutide in nonalcoholic steatohepatitis. New Engl J Med. (2021) 384:1113–24. doi: 10.1056/NEJMoa2028395
108. Harrison SA, Browne SK, Suschak JJ, Tomah S, Gutierrez JA, Yang J, et al. Effect of pemvidutide, a GLP-1/glucagon dual receptor agonist, on MASLD: A randomized, double-blind, placebo-controlled study. J Hepatol. (2025) 82:7–17. doi: 10.1016/j.jhep.2024.07.006
109. Martínez-González MA, Gea A, and Ruiz-Canela M. The mediterranean diet and cardiovascular health. Circ Res. (2019) 124:779–98. doi: 10.1161/circresaha.118.313348
110. Castro-Barquero S, Lamuela-Raventós RM, Doménech M, and Estruch R. Relationship between mediterranean dietary polyphenol intake and obesity. Nutrients. (2018) 10:1523. doi: 10.3390/nu10101523
111. Mitsou EK, Kakali A, Antonopoulou S, Mountzouris KC, Yannakoulia M, Panagiotakos DB, et al. Adherence to the mediterranean diet is associated with the gut microbiota pattern and gastrointestinal characteristics in an adult population. Br J Nutr. (2017) 117:1645–55. doi: 10.1017/s0007114517001593
112. De Filippis F, Pellegrini N, Vannini L, Jeffery IB, La Storia A, Laghi L, et al. High-level adherence to a mediterranean diet beneficially impacts the gut microbiota and associated metabolome. Gut. (2016) 65:1812–21. doi: 10.1136/gutjnl-2015-309957
113. Han HS, Kang G, Kim JS, Choi BH, and Koo SH. Regulation of glucose metabolism from a liver-centric perspective. Exp Mol Med. (2016) 48:e218. doi: 10.1038/emm.2015.122
114. Barbhuiya PA, Yoshitomi R, and Pathak MP. Understanding the link between sterol regulatory element binding protein (SREBPS) and metabolic dysfunction associated steatotic liver disease (MASLD). Curr Obes Rep. (2025) 14:36. doi: 10.1007/s13679-025-00626-y
115. Li Y, Li W, Zhu X, Xu N, Meng Q, Jiang W, et al. VEGFB ameliorates insulin resistance in NAFLD via the PI3K/AKT signal pathway. J Transl Med. (2024) 22:976. doi: 10.1186/s12967-024-05621-w
116. Wu D, Zhong P, Wang Y, Zhang Q, Li J, Liu Z, et al. Hydrogen sulfide attenuates high-fat diet-induced non-alcoholic fatty liver disease by inhibiting apoptosis and promoting autophagy via reactive oxygen species/phosphatidylinositol 3-kinase/AKT/mammalian target of rapamycin signaling pathway. Front Pharmacol. (2020) 11:585860. doi: 10.3389/fphar.2020.585860
117. Hernández MAG, Canfora EE, Jocken JWE, and Blaak EE. The short-chain fatty acid acetate in body weight control and insulin sensitivity. Nutrients. (2019) 11:1943. doi: 10.3390/nu11081943
118. Balmer ML, Ma EH, Thompson AJ, Epple R, Unterstab G, Lötscher J, et al. Memory CD8(+) T cells balance pro- and anti-inflammatory activity by reprogramming cellular acetate handling at sites of infection. Cell Metab. (2020) 32:457–67.e5. doi: 10.1016/j.cmet.2020.07.004
119. Gao X, Lin SH, Ren F, Li JT, Chen JJ, Yao CB, et al. Acetate functions as an epigenetic metabolite to promote lipid synthesis under hypoxia. Nat Commun. (2016) 7:11960. doi: 10.1038/ncomms11960
120. Tedelind S, Westberg F, Kjerrulf M, and Vidal A. Anti-inflammatory properties of the short-chain fatty acids acetate and propionate: A study with relevance to inflammatory bowel disease. World J Gastroenterol. (2007) 13:2826–32. doi: 10.3748/wjg.v13.i20.2826
121. Chambers ES, Byrne CS, Morrison DJ, Murphy KG, Preston T, Tedford C, et al. Dietary supplementation with inulin-propionate ester or inulin improves insulin sensitivity in adults with overweight and obesity with distinct effects on the gut microbiota, plasma metabolome and systemic inflammatory responses: A randomised cross-over trial. Gut. (2019) 68:1430–8. doi: 10.1136/gutjnl-2019-318424
122. Yip W, Hughes MR, Li Y, Cait A, Hirst M, Mohn WW, et al. Butyrate shapes immune cell fate and function in allergic asthma. Front Immunol. (2021) 12:628453. doi: 10.3389/fimmu.2021.628453
123. Chen J and Vitetta L. The role of butyrate in attenuating pathobiont-induced hyperinflammation. Immune network. (2020) 20:e15. doi: 10.4110/in.2020.20.e15
124. Li Z, Yi CX, Katiraei S, Kooijman S, Zhou E, Chung CK, et al. Butyrate reduces appetite and activates brown adipose tissue via the gut-brain neural circuit. Gut. (2018) 67:1269–79. doi: 10.1136/gutjnl-2017-314050
125. Candelli M, Franza L, Pignataro G, Ojetti V, Covino M, Piccioni A, et al. Interaction between lipopolysaccharide and gut microbiota in inflammatory bowel diseases. Int J Mol Sci. (2021) 22:6242. doi: 10.3390/ijms22126242
126. Wahlström A, Sayin SI, Marschall HU, and Bäckhed F. Intestinal crosstalk between bile acids and microbiota and its impact on host metabolism. Cell Metab. (2016) 24:41–50. doi: 10.1016/j.cmet.2016.05.005
127. de Aguiar Vallim TQ, Tarling EJ, and Edwards PA. Pleiotropic roles of bile acids in metabolism. Cell Metab. (2013) 17:657–69. doi: 10.1016/j.cmet.2013.03.013
128. Jiang H, Zhang L, Yang M, Li G, Ding C, Xin M, et al. Branched-chain amino acids promote thrombocytopoiesis by activating mTOR signaling. J Thromb Haemost. (2023) 21:3224–35. doi: 10.1016/j.jtha.2023.06.039
129. Matsumoto M, Kurihara S, Kibe R, Ashida H, and Benno Y. Longevity in mice is promoted by probiotic-induced suppression of colonic senescence dependent on upregulation of gut bacterial polyamine production. PloS One. (2011) 6:e23652. doi: 10.1371/journal.pone.0023652
130. Villanueva-Millan MJ, Leite G, Wang J, Morales W, Parodi G, Pimentel ML, et al. Methanogens and hydrogen sulfide producing bacteria guide distinct gut microbe profiles and irritable bowel syndrome subtypes. Am J Gastroenterol. (2022) 117:2055–66. doi: 10.14309/ajg.0000000000001997
Keywords: metabolic-associated fatty liver disease, type 2 diabetes mellitus, gut microbiota, glucose and lipid metabolism, gut–adipose axis, gut–pancreatic axis
Citation: Liang J, Chu Y, Chen X and Leng Y (2025) The gut–adipose/pancreas axis: a novel perspective on glycolipid metabolism dysregulation in MAFLD and T2DM pathogenesis. Front. Endocrinol. 16:1664233. doi: 10.3389/fendo.2025.1664233
Received: 11 July 2025; Accepted: 22 October 2025;
Published: 31 October 2025.
Edited by:
Rangsun Charoensook, Naresuan University, ThailandReviewed by:
George Grant, Independent Researcher, Aberdeen, United KingdomGang Chen, Zhenjiang Stomatological Hospital, China
Copyright © 2025 Liang, Chu, Chen and Leng. This is an open-access article distributed under the terms of the Creative Commons Attribution License (CC BY). The use, distribution or reproduction in other forums is permitted, provided the original author(s) and the copyright owner(s) are credited and that the original publication in this journal is cited, in accordance with accepted academic practice. No use, distribution or reproduction is permitted which does not comply with these terms.
*Correspondence: Yan Leng, Y2N1Y21fbHlAb3V0bG9vay5jb20=
Yunhang Chu1