Gut‑liver axis in liver disease: From basic science to clinical treatment (Review)
- Authors:
- Published online on: October 23, 2024 https://doi.org/10.3892/mmr.2024.13375
- Article Number: 10
-
Copyright: © Wang et al. This is an open access article distributed under the terms of Creative Commons Attribution License.
Abstract
Introduction
The liver is the organ that interacts most with the digestive system. As such, the liver is exposed to numerous gut microbiota (GM). The GM is a diverse ecosystem that contains bacteria, protozoa, archaea, fungi and viruses. There are >1×1014 species of microorganisms in the human gastrointestinal tract, including ~1×104 species of bacteria (1). The unique microenvironment and physicochemical barriers of each region of the digestive system determine the growth of specific microbiota. It is widely recognized that transgenes serve a role in the physiological and pathological aspects of human health, particularly liver health (2,3).
Previous studies have reported the role of the GM in occurrence and development of a number of liver diseases (such as hepatitis, alcoholic liver disease (ALD), non-alcoholic liver disease (NAFLD), liver fibrosis, cirrhosis, and liver cancer, etc.) (4,5). In recent years, researchers have assessed the gut-liver axis (6–10). For example, Pseudomonas aeruginosa can significantly inhibit NAFLD-HCC progression by secreting acetate salts (7). The present review aimed to assess the previous research to provide a broader understanding of this axis and discuss the mechanism of the gut-liver axis in a number of common types of liver diseases, highlighting the potential drugs and treatment methods targeting GM in clinical treatment of liver disease.
Overview of liver disease
The term ‘liver disease’ covers a range of illnesses, including acute problems caused by harmful agents such as viruses, poisons, alcohol and pharmaceutical agents, as well as chronic liver disease, which that may result in cirrhosis. Any type of cirrhosis increases the risk of developing hepatic cell carcinoma (HCC), a primary liver cancer; this risk is higher if the cirrhosis is caused by hepatitis B (HBV) or hepatitis C (HCV) infection (11,12). Most types of liver diseases (such as hepatitis, ALD, NAFLD, focal liver disease, and some types of liver cancer) (13–18) can be conservatively treated with non-surgical treatment methods), including targeted therapy (19,20), immunotherapy (21), radiotherapy (22), etc. Acute liver failure is associated with rapid and massive injury to hepatocytes, which is rare, but has a high incidence and mortality rate (23). HCV infection causes ~290,000 deaths worldwide each year and is the primary cause of liver cirrhosis and associated complications (i.e. decompensated cirrhosis) (24). Other data indicate that globally, the number of HBV-related deaths is expected to reach 1,109,500 by 2030 (25). ALD is the most common cause of liver cirrhosis worldwide. According to WHO data in 2018, the global prevalence of alcohol use disorders was 5.1% (26). A modeling study suggests that if current drinking trends are not controlled for, the age-specific mortality rate (ASDR) associated with ALD in the United States is expected to increase from 8.2 deaths per 100,000 patients per year in 2019 to 15.2 deaths per 100,000 patients per year in 2040 (27). Other data indicate that globally, the number of HBV-related deaths is expected to reach 1,109,500 by 2030 (25). ALD is the most common cause of liver cirrhosis worldwide. According to WHO data in 2018, the global prevalence of alcohol use disorders was 5.1% (26). Primary liver cancer is the seventh most common cancer in the world and the second most common cause of cancer death. HCC is the main type of liver cancer worldwide, accounting for approximately 75% of the total. It is the primary cause of diagnosis and death in liver cancer cases (28).
Overview of the gut-liver axis
The gut and liver communicate with each other. The liver can regulate gut function through the bile ducts. The intestine can regulate liver function through the portal vein. In addition, these two organs can indirectly affect each other's function through the whole body blood circulation. The liver delivers bile salts and antimicrobial molecules such as immunoglobulin A and angiopoietin to the gut via the biliary tract. This maintains the GM by modulatin microbiota growth (29). Bile acids (BAs) exert direct antibacterial effects by disrupting the cell membranes of intestinal bacteria and causing membrane protein degradation (30). Moreover, BAs indirectly regulate the composition of the GM by activating BA receptors in the intestine, particularly farnesoid X receptor (FXR) encoded by Nuclear Receptor Subfamily 1 Group H Member 4 (31). Liu et al (32) reported that activated FXR is involved in the expression of gut tight junction markers (claudin1 and zonula occludens-1), maintaining BA homeostasis and inhibiting the expression of inflammatory factors, thereby inhibiting bacterial overgrowth and mucosal damage in the ileum. Mice lacking FXR exhibit an increase in harmful bacteria in the ileum and damage to the epithelial barrier. BAs also regulate hepatic BA synthesis, glucose and lipid metabolism and dietary energy use via nuclear receptors such as the FXR and the G protein-coupled BA receptor (TGR5). FXR receptors inhibit the expression of BA synthase by binding to endogenous BAs, which provides negative feedback to regulate BA synthesis (33). For example, cholesterol 7-α hydroxylase 1 and cytochrome P450 Family 27 Subfamily A Member 1 are enzymes required for the synthesis of BAs, with their expression significantly reduced upon FXR stimulation (34). BAs can interact with nuclear transcription factors in the promoter region of gluconeogenesis-associated genes via the FXR-small heterodimer partner-dependent pathway and inhibit their expression (35). TGR5 belongs to the G protein-coupled receptor superfamily. The activated TGR5 receptor is associated with energy expenditure of the body. According to Watanabe's research report, treating brown adipocytes and human skeletal muscle cells with BA can increase the activity of TGR5 in cells, thereby upregulating the expression of cAMP-dependent type 2 deiodinase (DIO2). This enzyme catalyzes the deiodination of prothyroid hormones to triiodothyronine (T3), thereby enhancing oxygen and energy consumption in key thermogenic tissues such as brown adipose tissue and skeletal muscle (36,37). In terms of regulating blood sugar, previous studies have reported that the liver decreases hepatic gluconeogenesis through the BA FXR signal, which induces hepatic glycogen synthesis to regulate blood glucose levels (33). The GM and its metabolites, such as lipopolysaccharides (LPS), short-chain fatty acids (SCFAs), and tryptophan metabolites, are transported to the liver via the portal vein, inducing a local inflammatory response and exacerbating hepatic necrosis (38–40). In addition, metabolites produced by the liver, such as free fatty acids (FFAs), inflammatory factors, choline metabolites, and ethanol metabolites, can enter the systemic circulation, thereby prolonging the gut-liver axis and exerting systemic effects on multiple organs throughout the body, including the gut. For example, butyrate in the blood can enhance gut barrier function and reduce the translocation of gut microbial toxic metabolites to extraintestinal sites (41,42). Ethanol and acetaldehyde can increase intracellular calcium ion (Ca2+) concentration and disrupt the integrity of gut epithelial tight junctions (43).
GM in the gut-liver axis
The human microbiota, which includes bacteria, fungi, viruses, archaea and protozoa, is a collection of microorganisms that live in humans. The term ‘human microbiome’ refers to genes carried by these microbes and the surrounding environment in which they live and interact (44). The human microbiota, especially in the gastrointestinal tract, serves a role in human health. As the gastrointestinal tract makes direct contact with the liver through the portal vein, GM can directly affect liver (45).
The number of GM is in the order of 1×106 times higher than the number of human cells and GM total weight is estimated to be ~2 kg. The GM comprises ~3,000,000 genes, which is 150 times that of the entire human genome (46). GM is composed of >1,000 species, distributed in more than 50 different phyla (47); Bacteroidetes, Firmicutes, Proteobacteria, Fusobacteria, Tenericutes, Actinobacteria, and Verrucomicrobia are the most advantageous phyla, making up to 90% of the total microbial population in humans. Among them, Bacteroidetes and Firmicutes are the most dominant phyla (44).
GM are concentrated within the lumen of the gut and adhere to the mucosal surface. The location and diameter of the gut lumen vary, and the types and abundance of GM present also vary. The bacterial population density in the jejunum and ileum is higher than that in the stomach cavity and duodenum. However, the most densely populated area is the colon, which contains ~1,000 colony-forming units per milliliter/ml and is mainly composed of anaerobic bacteria such as Bacteroides, Porphyromonas, Bifidobacterium, Lactobacillus and Clostridium. In the colon, the ratio of anaerobic to aerobic bacteria is 100:1-1,000:1. This is influenced by changes in optimal growth conditions for these bacteria, which are caused by local colonic lesions (48–50). Microbes are concentrated in the lumen of the gut wall or adhere to the surface of the mucous membrane.
GM induces inflammation and immune stress
The gastrointestinal tract contains a large number of microorganisms, particularly bacteria, which are a source of pathogen-associated molecular patterns (PAMPs) and metabolites (51). Under normal conditions, small amounts of GM and metabolites enter the liver and are rapidly cleared. However, when the normal gut barrier permeability is increased, as in gut ecological dysbiosis, large amounts of GM and GM metabolites enter the liver, leading to activation of the immune cascade in the liver and production of pro-inflammatory cytokines (52,53). Increased gut permeability is attributed to tight junction disruption, potentially from the pathological change in the composition of GM and its metabolites or their induced immune cascade and inflammatory response (54–57). Dendritic cells form an extensive network under the gut epithelium. Dysregulated GM stimulates immature dendritic cells to produce IL-23, which promotes the secretion of cytokines IL-17A and IL-22 by interacting with surface receptors on activated CD4+ T cells, thereby inducing a local gut inflammatory response (58,59). Moreover, dendritic cells and macrophages produce cytokines including IL-1β, IL-6, IL-18 and TNF to exacerbate the inflammatory response (60). Large amounts of pro-inflammatory cytokines affect tight junctions between gut epithelial cells, in addition to increasing the inflammatory burden. IL-1β recruits granulocytes to infiltrate foci of infection and directly disrupt the junctions and tightness of gut epithelial cells (61). TNF-α promotes myosin light chain kinase (MLCK) protein expression level in the gut epithelium (62). Previous studies have shown that MLCK triggers perijunction actinosin ring (PAMR) contraction, leading to increased permeability of tight junctions adjacent to gut epithelial cells (63). Moreover, TNF-α and IL-1β induce endoplasmic reticulum stress, which affects gut epithelial cells and alters proteins in the apical and basal lateral membranes, including E-calmodulin. This further disrupts the tight junctions of the gut epithelium (64). Release of these cytokines can activate natural killer cells, which bind to epithelial cells, releasing toxic particles (such as perforin, and granzymes) (65,66) and inducing apoptosis in epithelial cells. Furthermore, dendritic cells phagocytose antigens, which activate T cells and promote the differentiation of T helper 0 (Th0) cells into Th1, Th2 and Th17 cells. Th1 cells induce cytotoxic T cells to activate, proliferate and attack infected gut epithelial cells. IL-18 is a pro-inflammatory cytokine that promote the secretion of significant amounts of interferon-γ (IFN-γ) by Th1 cells. IFN-γ induces apoptosis in gut epithelial cells, thereby compromising the integrity of the gut epithelium (67–69). Th2 cells activate B cells, causing B cell proliferation and differentiation into plasma cells that secrete IL-4, IL-5 and IL-13 (70). These factors are believed to be associated with local eosinophil and mononuclear infiltration, increased mucus production, and epithelial cell proliferation and hypertrophy in the gastrointestinal tract (71). In addition, IL-13 activates STAT6 in epithelial cells and affects tight junctions in gut epithelium (72). IL-13 can also induce apoptosis of gut epithelial cells and further increase gut permeability (73,74). Th17 cells secrete IL-17A to mediate inflammatory responses (75). Large amounts of pro-inflammatory cytokines affect tight junctions between gut epithelial cells, in addition to increasing the inflammatory burden.
Increased gut permeability results in a heightened passage of GM and metabolites, such as LPS and endotoxins, into the portal circulation (76). Metabolites produced by GM, including trimethylamine and alcohol, exert direct toxic effects on the liver, while PAMPs- the distinctive molecular structures of GM-induce liver injury through the activation of the innate immune system (77). GM is detected by pathogen recognition receptors in the liver, encompassing toll-like receptors (TLRs) and inflammasomes. TLRs are present on hepatic sinusoidal cells, such as Kupffer cells and hepatic stellate cells, and they identify PAMPs located on cell membranes (78,79). TLR-mediated signaling pathways lead to sustained production of inflammatory cytokines, which cause or exacerbate liver injury (80) (Fig. 1).
Circadian regulation of GM promotes liver metabolism
The ‘biological clock’ is an intrinsic rhythm formed by organisms to adapt to changes in the surrounding environment. The ability of the circadian clock to persist in the absence of environmental cues provides internal temporal organization, allowing rhythmic activities to occur at characteristic times during the circadian cycle (81). The mammalian biological clock is composed of a number of core transcriptional regulators, including Brain and muscle arnt-like protein 1, Clock Circadian Regulator (CLOCK), period and cryptochrome. Disturbances in the biological clock are associated with the progression of a number of diseases, including fatty liver disease, heart disease, diabetes and cancer (82–85). However, the specific pathological mechanisms have not been fully identified. Previous studies have suggested that the GM may serve a link between circadian rhythm disorders and disease progression, which may be associated with the role of GM on host immune system function and metabolism (54,86,87).
Although GM is not directly affected by external environmental, self-regulation of the biological clock, host activity, and metabolic patterns, particularly changes in eating patterns, can induce rhythmic oscillations in the abundance of GM and GM metabolite. For example, Thaiss et al (88) reported rhythmic changes in GM composition over a 24-h period by analyzing fecal microbiota of mice. The abundance of Lactobacillus reuteri in the mouse gut increased during the light and decreased during the dark period. By contrast Per1/2−/− mice, which lack a functional host biological clock, exhibit almost complete loss of this GM abundance variation. GMs with different compositions secrete different metabolites. GM affects host metabolism and energy homeostasis by metabolite signaling. Previous studies have reported an increase in body fat percentage and insulin resistance in mice fed without transgenic genes under the same feeding conditions compared to normal mice (89,90). Further research reports indicate that differences exist in the composition of GM between obese and normal mice (91,92). For example, compared with the normal feed group, the high-fat feed group had higher abundance of Lachnospiraceae and Blautia in the gut of mice, while the abundance of Lactobacillus, Faecalibaculum, Lachnoclostridium, Bacteroides and Desulfovibrio was lower (92). The aforementioned research indicates that GM is an important environmental factor affecting energy collection and storage in the host. Turnbaugh et al (93) reported that the increased ability of the GM of obese mice to obtain energy from the diet is associated with a reduced abundance of Bacteroidetes and an increased abundance of Firmicutes in the GM. This change has also been confirmed in humans (94). Akkermansia muciniphila, Bifidobacterium longum, Clostridium leptum group, Faecalibacterium prausnitzii and Faecalibacterium and Dorea were suggested to serve a role in the regulation of blood glucose; changes in the abundance of these genera can lead to dysregulation of glucose metabolism and the progression of type 2 diabetes in humans (95).
Role of the gut-liver axis in liver disease
Association between GM and viral hepatitis
Viral hepatitis is the most common type of hepatitis worldwide. Hepatitis is defined as inflammation of the liver tissue. More than 300 million people worldwide are affected by viral hepatitis infections, which has a notable negative impact on public health and the economy and leads to high mortality (96). Hepatitis A, B, C, D and E are the five most common types of viral hepatitis. HBV and HCV often lead to chronic infections, and in severe cases, may lead to cirrhosis and liver cancer, affecting 257 million and 71 million people worldwide, respectively (97,98). Chou et al (99) reported that GM serves a role in age-dependent immunity of mice against HBV infection. After 6 weeks of infection, normal adult mice with mature GM completely eliminate HBV, but young mice without GM remain positive for HBV. Following clearance of the GM of adult mice, their resistance to HBV decreased. Another study reported that after treatment with fecal microbiota transplantation (FMT), patients with hepatitis B e-antigen (HBeAg) positivity showed a significant decrease in their blood HBeAg levels, indicating a weakened viral replication activity (100).
Previous studies have reported that the GM composition of patients with hepatitis changes compared with healthy individuals (101–103). For example, the levels of Bacteroides in the GM of patients with HBV-related cirrhosis is low, at 4 vs. 53% in healthy patients and the level of Proteus is high at 43 vs. 4% for healthy patients (104). Furthermore, Bajaj et al (105) reported the unique composition of GM in patients with HCV. GM of these patients predominantly comprises Enterobacteriaceae, Clostridium and Ruminococcaceae genera. A previous study compared the GM of stage 4 HCV patients with that of healthy individuals, revealing that the relative abundance of Bacteroides in the GM of HCV patients increased, whereas the abundance of Firmicutes decreased (106). Using high-throughput 16S rRNA gene sequencing, Aly et al (106) reported that the GM of patients with HCV exhibits high level of Proctor and levels of Acinetobacter, Vibrio and Lactobacillus were also increased compared with healthy patients. However, the probiotic genus, Bifidobacterium were found exclusively in the GM of HCV patients, while no Bifidobacterium were detected in the GM of healthy individuals. In conclusion, because viral hepatitis is associated with GM composition, GM could be targeted in the development of novel hepatitis treatment strategies in future.
Association between GM and ALD/NAFLD
ALD is the most prevalent form of chronic liver disease, affecting 150 million people worldwide (107). Alcoholic steatohepatitis (ASH), which is characterized by hepatic inflammation, may develop from alcoholic fatty liver. ASH is characterized by acute inflammatory response with neutrophils and hepatocellular damage, whereas cirrhosis involves chronic architectural remodeling with fibrosis and regenerative nodules (108). Chronic ASH can lead to fibrosis and cirrhosis. In 2019, ~371,964 people exhibited alcoholic cirrhosis-related mortality, accounting for 25% of all liver cirrhosis-associated mortalities. There were 90,741 mortalities due to alcohol-related HCC (109). Moreover, ASH can directly lead to liver failure, and severe ASH may lead to high mortality rates (110).
Chronic alcohol consumption directly or indirectly alters composition of GM and leads to changes in human and animal gut development (111). Chronic ethanol feeding leads to a decrease in abundance of Bacteroidetes and Firmicutes phyla in the GM and an increase in the proportion of Gram-negative Proteobacteria and Gram-positive Actinobacteria phyla (112). Alcohol can directly and indirectly increase the permeability of the gut wall, as dysfunctional GM and its metabolites enter the liver and activate Kupffer cells by binding with TLR4 or TLR9, inducing Kupffer cells to secrete pro-inflammatory cytokines. The GM of healthy individuals produces SCFAs by breaking down dietary fiber and resistant starch in the gastrointestinal tract. SCFA plays a role in gastrointestinal physiology, immune function, and host metabolism (113). Alcohol use is associated with lower levels of gut SCFA, suggesting that GM may serve a role in development and progression of liver disease (114–116) (Fig. 2). SCFA in the mouse gut helps alleviate the progression of ALD, which may be related to the regulatory effect of SCFA on the liver immune microenvironment (114).
Furthermore, prevalence of NAFLD increases with obesity and has replaced alcoholic hepatitis as the most common type of chronic liver disease worldwide (117). Obesity is associated with dysbiosis of the GM and GM modulation has potential for prevention and treatment of NAFLD (118). According to mouse studies and fecal transplantation trials, GM serves a key role in the development of NAFLD (119,120). Ecological dysbiosis of GM and its metabolites promotes the signal cascade reaction in the liver during translocation (such as activation of TLR and NLRP3 signaling pathway) promotes the secretion of cytokines such as TNF-α and IL-1β, and leads to steatosis and inflammation in the liver of susceptible mice (121). Regardless of the amount of alcohol consumed, GM produce endogenous ethanol, particularly when sugar-rich foods are consumed (122). Ethanol synthesized by GM activates TLRs in the liver, promoting cytokine synthesis and secretion and altering BA profile in obese humans and mice (123–125).
In conclusion, as GM contributes to the development of ALD and NAFLD, the GM may be a potential therapeutic target for ALD and NAFLD. Using MIYAIRI 588, a butyrate-producing probiotic, to treat NAFLD in rats significantly improves liver lipid deposition, insulin resistance, serum endotoxin levels, and the liver inflammation index (126). Additionally, a meta-analysis indicated that probiotic therapy significantly reduces blood ALT, AST, total cholesterol (T-chol), high-density lipoprotein (HDL), and TNF-α levels in NAFLD patients, while also improving insulin resistance (127). Kirpich et al (128) showed that short-term oral supplementation with Bifidobacterium and Lactobacillus plantarum 8PA3 can improve the dysbiosis of GM in patients with ALD, and reduce serum levels of ALT, AST, gamma-glutamyl transpeptidase (GGT), lactate dehydrogenase, and total bilirubin.
Association between GM and cirrhosis
Cirrhosis is histological development of regenerative nodules surrounded by fibrous bands, which arises from chronic liver injury and leads to portal hypertension and end-stage liver disease (129). The mechanism underlying cirrhosis has been extensively studied (130–132). Cirrhosis is caused by abnormal accumulation of the extracellular matrix (ECM) (mainly composed of fibrous collagen, elastin, and matrix proteins) under chronic (133). Excessive deposition of ECM can lead to the replacement of the normal structure of liver lobules with fibrous tissue, resulting in the destruction of the normal liver architecture and the formation of fibrous septa and nodules (133). It is hypothesized that the first step of cirrhosis involves the production of oxygen-free radicals and inflammatory substances that damage liver cells and recruitment of Kupffer and inflammatory cells. Additionally, elevated levels of oxygen free radicals and inflammatory substances in liver tissue can cause hepatic stellate cells (HSC) to differentiate into myofibroblasts, the primary source of ECM (134). The most common causes of cirrhosis are viral hepatitis, NASH and ALD (135). According to a 2023 survey, cirrhosis is frequently attributed to alcohol use disorders (~45% of cases), HCV (41%), and NAFLD (26%) (136). The 2019 Global Burden of Disease Study estimated global deaths related to cirrhosis as follows: 395,000 from HCV-associated cirrhosis, 331,000 from HBV-related cirrhosis, 372,000 from alcohol-related cirrhosis, and 134,000 from NASH-related cirrhosis (137). Cirrhosis is the 11th most common cause of mortality and the third most common cause of mortality among people aged 45–64 years. Together with liver cancer, cirrhosis accounts for 3.5% of all deaths worldwide (138). To date, there is no consensus regarding the treatment of cirrhosis, with current methods limited to controlling symptoms and complications, as well slowing the progression of cirrhosis. If the liver is severely damaged, liver transplantation may be the only treatment option (139,140).
The gut-liver axis serves a role in progression of cirrhosis. The gut epithelium is a single-cell layer that serves as a selective permeation barrier, facilitating the absorption of nutrients, electrolytes, and water, while effectively defending against intracavitary toxins, antigens, and GM (141). Previous studies have demonstrated that gut permeability in patients with cirrhosis is increased, which has been reported to be related to the degree of endotoxemia (142,143). A prospective study demonstrated higher serum endotoxin levels in patients with cirrhosis compared to healthy individuals (144). Subsequent studies have indicated that increased gut permeability facilitates the translocation of intestinal-derived endotoxins (LPS) into the bloodstream (145). In addition, the increase in gut permeability promotes pathological translocation of GM and its metabolites into liver, leading to the activation of numerous inflammatory cytokine signaling pathways in the liver, driving immune dysfunction associated with inflammation and cirrhosis (146). Moreover, cytokines secreted by immune cells can both reduce (i.e. TNFα and IFNγ) gut barrier function and enhance (i.e. TGFβ and IL-10) gut barrier function. The immune response may lead to ecological imbalance or microbial changes in feces, intestinal mucosa, ascites, liver, serum and saliva (147). Ecological imbalance is related to gut barrier dysfunction as GM and its products regulate the barrier function by affecting epithelial inflammatory reaction and mucosal repair functions (148). Further research has shown that gut permeability can be directly regulated by GM through the release of soluble peptides or toxins, which in turn regulate the expression of gut tight junction proteins, including integrated membrane proteins, junction complex proteins, and cytoskeletal structural proteins (149). Additionally, other metabolites of GM, such as SCFAs, BA metabolites, conjugated FAs, indole derivatives, and polyamines, can regulate the expression of gut tight junction proteins by binding to receptors such as FXR, G protein-coupled bile acid receptor (TGR), and aromatic hydrogen receptor (AHR) on the surface of gut epithelial cells (150).
Small intestinal bacterial overgrowth (SIBO) is frequently observed in patients with liver cirrhosis and is more prevalent in those with advanced cirrhosis. Spontaneous bacterial peritonitis (SBP) and hepatic encephalopathy (HE) are two frequent complications of liver cirrhosis, both closely associated with increased patient mortality (136). Increasing evidence suggests that the imbalance of intestinal ecology in patients with liver cirrhosis is closely related to disease progression (143,151). Chang et al (152) reported that 70% of patients with SBP cirrhosis exhibit SIBO, whereas only 20% of patients with non-SBP cirrhosis had SIBO. In patients with a history of SBP, intestinal peristalsis is impaired and may contribute to development of SIBO. Corradi et al demonstrated that the control of SIBO with antibiotic treatment may mitigate the progression of spontaneous bacterial peritonitis in cirrhotic rats (153). HE is associated with the GM (154). A randomized controlled trial demonstrated that FMT improved the dysbiosis of GM in patients with liver cirrhosis and delayed the progression of HE (155). Bajaj et al (156) suggested that the sigmoid microbiota of patients with cirrhosis exhibits a low abundance of autochthonous genera including Dorea, Subdoligranulum, and Incertae Sedis other and a high abundance of potentially pathogenic bacteria, including Enterococcus, Burkholderia, Proteus and Clostridium. Other studies have reported that the abundance of Veillonella, Megasphaera, Dialister, Atopobium and Prevotella increased in the GM of patients with cirrhosis (143,157). Changes in fungi in GM have also been reported in alcohol-related cirrhosis (158). Compared with healthy individuals, the diversity of gut fungi in patients with ALD is decreased, demonstrated by the overgrowth of Candida (74). Candida hemolysin is a peptide toxin secreted by Candida that can transfer from the gut to the bloodstream and translocate to the liver, causing direct damage to liver cells. β-glucan is a cell wall component of many symbiotic fungi. Serum levels of β-glucan were significantly elevated in alcohol-fed mice. Concentration of serum β-glucan is closely related to gut integrity, inflammation, and the severity of liver disease (159). β-glucan can stimulate immune cells to produce a strong immune response, which is an important cause of disease exacerbation (160). By analyzing the composition and abundance of GM, the etiology of cirrhosis can be effectively determined, guiding treatment (161). Controlling the abnormal growth of GM can effectively manage the progression of cirrhosis. Oral probiotic lactobacilli can regulate the decrease in Enterococcal abundance, which is associated with reducing the severity of liver injury (162).
Association between GM and liver cancer
Primary liver cancer is the sixth most common malignancy and the fourth highest cause of cancer-associated mortality worldwide. The most common type of primary liver cancer is HCC, according to the World Health Organization (163). A number of risk factors are associated with liver cancer, notably HBV, HCV, alcohol abuse and aflatoxins in the diet (164).
Based on studies in human and animal models, GM may contribute to development of HCC (91,101,120,143,157,165–190)(Table I). Patients with HCC and mice treated with diethylnitrosamine exhibit dysbiosis of the GM. The reduction of Lactobacillus, Bifidobacterium and Enterococcus in the gut of rats with liver cancer, as well as proliferation of Escherichia coli, suggests an imbalance in composition of GM in liver cancer (191). Lipopolysaccharide (LPS), a metabolite of the GM, binds to TLR4 on HSCs, triggering the activation of HSCs, which leads to the development of liver fibrosis and cirrhosis (Fig. 3). Dapito et al (192) reported that activation of TLR4 accelerates HCC progression by promoting cell proliferation and inhibiting apoptosis. Furthermore, Mou et al (193) reported the role of the LPS/TLR4 signaling pathway in regulating liver fibrosis progression in rats. Neomycin is an antibiotic that inhibit the overgrowth of GM. Yu et al found that the accumulation of LPS and the expression of TLR4 were reduced in the liver of liver cancer mice treated with neomycin (194). Further studies using antibiotics in mouse models are needed to understand the disease progression in the absence of GM. Dapito et al (192) demonstrated that removal of the GM by antibiotics protected mice from liver fibrosis and HCC. This may be due to antibiotics reducing the quantity of GM, decreasing the secretion of toxic metabolites such as LPS, thereby lessening the degree of liver damage.
Yoshimoto et al (195) reported that deoxycholic acid (DCA), a metabolite of the GM known to induce DNA damage, is more frequently produced in the gut of obese mice. As a result of elevated levels of DCA in the gut-liver axis, HSCs exhibit a senescence-related secretory phenotype and release inflammatory factors and pro-tumor chemicals (196). Further research has shown that DCA can activate the NF-κB signaling pathway, TNF signaling pathway, and NLRP3 signaling pathway in HSC cells, triggering a large secretion of downstream factors such as IL-1 β, IL-6, TNF, ROS, etc. that promote damage. Exposure of the liver to these damaging factors can easily lead to HCC. These findings suggest that GM metabolites contribute to development of obesity-induced HCC in animals.
Clinical treatment of liver disease based on the liver-gut axis
FMT
FMT is an increasingly popular method of altering GM composition during disease. FMT involves transplantation of GM obtained from the stool of a healthy donor into the gastrointestinal tract of a patient (197–199). In most cases, this therapy is used to treat gastrointestinal diseases caused by activity of pathogenic or conditionally pathogenic microorganisms (200). Recent studies have reported that this approach has potential for clinical application in treatment of liver disease (201,202).
FMT can be administered through three channels: Oral, through the upper gastric portion; nasal, via nasogastric tubes; and rectal through colonoscopy or enema (203). Fecal suspension perfusion in the rectum using colonoscopy is considered to be the best method for FMT (204). By contrast, FMT using the upper gastric route, including nasogastric tube, nasogastric or upper endoscopy exposes the entire gastrointestinal tract to donor stool and can lead to pulmonary or gastrointestinal complications due to the presence of large numbers of pathogenic bacteria in the upper GI and respiratory tracts (205). The use of fecal microbiota in oral capsules has also been reported (206). In a randomized controlled study of 22 obese patients (206), FMT capsules exhibited no significant side effects and significantly improved composition of the GM of patients and decreased metabolic levels of taurocholic, which cause damage to the liver (207,208) and BAs.
Phage therapy
Phages are viruses that specifically infect bacteria. In the early days of phage therapy application, infectious phage agents were commonly used to treat diseases caused by bacterial infections including Staphylococcus, Streptococcus, Vibrio, Klebsiella, Enterobacter, Shigella, Escherichia, Pseudomonas and Providencia, which have the advantage over antibiotics of targeting specific bacterial species or strains while self-replicating and spreading to infect other target bacterial cells (209). Notably, phage therapy has the ability to edit the GM. In two randomized placebo-controlled trials (study nos. NCT03269617 and NCT04511221), phages improved the GM profile by targeting specific bacterial genera, modulated the overall metabolism and reduced the incidence and severity of gastrointestinal discomfort (210–212).
Specific GM serve a role in the pathogenesis of a number of types of liver diseases; therefore, phage therapy capable of eliminating specific GM has potential value in treatment of liver disease. In humanized mice colonized with bacteria from the feces of patients with alcoholic hepatitis, phage therapy targeting lysogenic Enterococcus faecalis decreases mortality as well as ethanol-induced liver injury, steatosis, inflammation and fibrosis (213). However, to the best of our knowledge, there are no clinical trials to validate the safety and efficacy of phage therapy in treatment of human liver disease. Despite this, phage therapy has been suggested as potential novel therapy for the treatment of liver disease (214).
Engineered bacterial therapy
Bacteria can acquire the ability to transcribe and translate various genes through gene editing technology. Through oral administration and other delivery methods, these genetically modified bacteria can reach the human intestine and colonize it. These genetically engineered bacteria can express specific enzymes, thereby promoting the conversion of toxic metabolites in the intestine to non-toxic products (215). Hyperammonemia is associated with liver disease, and the intestine is the primary source of systemic ammonia (NH3) (216). Kurtz et al (217) developed an engineered bacterium called SYNB1020 that can colonize the intestine via oral administration. This bacterium can convert NH3 in the intestine into l-arginine, thereby reducing blood ammonia levels. In a mouse model of hyperammonemia, SYNB1020 treatment increases survival rate. Moreover, SYNB1020 has good tolerability in a phase I clinical trial of hyperammonemia disease (217). Thus, SYNB1020 warrants further clinical development.
E. coli Nissle 1917 is a traditional probiotic with a well-established safety record, which has been widely used in the production of therapeutic agents, delivery carriers, and microbial platforms in industrial production (218). Lynch et al (219) genetically engineered E. coli Nissle 1917 to overexpress catalase and superoxide dismutase; inflammatory response of colonic tissue in mice with inflammatory bowel disease model significantly reduced following oral administration and adjusted the composition of GM and repaired the gut epithelial barrier.
Duodenal mucosal surface replacement (DMR)
As the most proximal gut segment, the duodenal mucosa has a unique chemosensory capacity to detect luminal contents and rapidly release bioactive mediators and hormones with local and systemic effects. These bioactive compounds include GM and GM metabolites, which are recognized by metabolite-sensing receptors (220). Intestinal luminal chemosensing involves the regulation of gut function and the systemic regulation of metabolism, energy balance, and food intake (221). Pharmacological modulation targeting the duodenum can maintain metabolic homeostasis in obesity, diabetes and NAFLD (222,223). Duodenal mucosal surface reconstruction (DMR) is a novel surgical procedure that, under endoscopic guidance, involves the introduction of a catheter with a balloon into the duodenum. The balloon is then expanded to segment the duodenum, and the duodenal mucosa is separated by injecting saline into the submucosal layer, followed by mucosal ablation using circulating hot and cold water. The ablation range covers all duodenal mucosa from 1 cm distal to the main papilla to the ligament of Treitz. After mucosal regeneration, the formation of new gut cells and the re-establishment of a healthy neuroendocrine axis can restore gut function and provide a healthy gut environment (224–226). In a randomized controlled clinical trial (227), DMR reported safety and efficacy in glycemic control and liver fat content in type 2 diabetes. However, in another clinical trial, DMR did not improve NASH (228).
Conclusion
The gut-liver axis underscores the connection between gut health and liver function. Numerous types of liver diseases, including NAFLD, ALD, HE and HCC, are influenced by changes in GM. This axis is a target for clinical applications, aiding in diagnosis, prognosis and the development of treatment. GM analysis offers insights into the mechanisms and phenotypes of liver disease. Modulating BA signaling and fecal transplantation or probiotics from human sources show potential as treatments. However, safety evaluation data for these methods are still insufficient. Further research is needed to ensure their efficacy and safety, bridging the gap between animal models and clinical practice to prevent progression of early liver disease.
In summary, the correlation between the gut and liver serves as a pathway for exploring the clinical treatment of liver disease. The function of a complete gut barrier, GM, and their associated metabolites communicates complex host-microbial interactions that can maintain health or promote disease. However, more research is needed to elucidate the changes in gut microbiota abundance in patients with different types of liver diseases, as well as the specific signaling pathways involved, and how to regulate GM in a way that minimizes side effects, to develop better clinical treatments for liver diseases. However, the present review has limitations. The impact of environmental exposure and lifestyle factors on the occurrence and development of liver disease was not discussed. Finally, GM could serve as a diagnostic tool and therapeutic target in patients with liver disease.
Acknowledgements
Not applicable.
Funding
The present study was supported by the National University Student Innovation Training Program of the Ministry of Education of the People's Republic of China (grant no. 202110366008) and and Basic and Clinical Cooperative Research Promotion Program of Anhui Medical University (grant no. 2023×kiT024).
Availability of data and materials
Not applicable.
Authors' contributions
JW and SC conceived the study and drafted the manuscript. XW constructed the figures and table and revised the manuscript. EZ and BC revised the manuscript. All authors have read and approved the final manuscript. Data authentication is not applicable.
Ethics approval and consent to participate
Not applicable.
Patient consent for publication
Not applicable.
Competing interests
The authors declare that they have no competing interests.
Glossary
Abbreviations
Abbreviations:
GM |
gut microbiota |
HBV |
hepatitis B |
HCV |
hepatitis C |
HCC |
hepatic cell carcinoma |
ALD |
alcoholic liver disease |
NAFLD |
non-alcoholic fatty liver disease |
FXR |
farnesoid X receptor |
PAMP |
pathogen-associated molecular pattern |
FMT |
fecal microbiota transplantation |
BA |
bile acid |
HSC |
hepatic stellate cell |
SBP |
spontaneous bacterial peritonitis |
HE |
hepatic encephalopathy |
SIBO |
small intestinal bacterial overgrowth |
LPS |
lipopolysaccharide |
DCA |
deoxycholic acid |
NASH |
non-alcoholic steatohepatitis |
DMR |
duodenal mucosal surface replacement |
References
Adak A and Khan MR: An insight into gut microbiota and its functionalities. Cell Mol Life Sci. 76:473–493. 2019. View Article : Google Scholar : PubMed/NCBI | |
Woodruff AW, Salih SY, de Savigny D, Baya EI, Shah AI and Dafalla AA: Toxocariasis in the Sudan. Ann Trop Med Parasitol. 75:559–561. 1981. View Article : Google Scholar : PubMed/NCBI | |
Fan Y and Pedersen O: Gut microbiota in human metabolic health and disease. Nat Rev Microbiol. 19:55–71. 2021. View Article : Google Scholar : PubMed/NCBI | |
Wiest R, Albillos A, Trauner M, Bajaj JS and Jalan R: Targeting the gut-liver axis in liver disease. J Hepatol. 67:1084–1103. 2017. View Article : Google Scholar : PubMed/NCBI | |
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. 20:3952019. View Article : Google Scholar : PubMed/NCBI | |
Kim ER, Park JS, Kim JH, Oh JY, Oh IJ, Choi DH, Lee YS, Park IS, Kim S, Lee DH, et al: A GLP-1/GLP-2 receptor dual agonist to treat NASH: Targeting the gut-liver axis and microbiome. Hepatology. 75:1523–1538. 2022. View Article : Google Scholar : PubMed/NCBI | |
Song Q, Zhang X, Liu W, Wei H, Liang W, Zhou Y, Ding Y, Ji F, Ho-Kwan Cheung A, Wong N and Yu J: Bifidobacterium pseudolongum-generated acetate suppresses non-alcoholic fatty liver disease-associated hepatocellular carcinoma. J Hepatol. 79:1352–1365. 2023. View Article : Google Scholar : PubMed/NCBI | |
Bertocchi A, Carloni S, Ravenda PS, Bertalot G, Spadoni I, Lo Cascio A, Gandini S, Lizier M, Braga D, Asnicar F, et al: Gut vascular barrier impairment leads to intestinal bacteria dissemination and colorectal cancer metastasis to liver. Cancer Cell. 39:708–724.e11. 2021. View Article : Google Scholar : PubMed/NCBI | |
Hu X, Chen F, Jia L, Long A, Peng Y, Li X, Huang J, Wei X, Fang X, Gao Z, et al: A gut-derived hormone regulates cholesterol metabolism. Cell. 187:1685–700.e18. 2024. View Article : Google Scholar : PubMed/NCBI | |
Kuang J, Wang J, Li Y, Li M, Zhao M, Ge K, Zheng D, Cheung KCP, Liao B, Wang S, et al: Hyodeoxycholic acid alleviates non-alcoholic fatty liver disease through modulating the gut-liver axis. Cell Metab. 35:1752–1766.e8. 2023. View Article : Google Scholar : PubMed/NCBI | |
Sheng Z, Xu J, Li F, Yuan Y, Peng X, Chen S, Zhou R and Huang W: The RING-domain E3 ubiquitin ligase RNF146 promotes cardiac hypertrophy by suppressing the LKB1/AMPK signaling pathway. Exp Cell Res. 410:1129542022. View Article : Google Scholar : PubMed/NCBI | |
Goto J, Otaki Y, Watanabe T, Kobayashi Y, Aono T, Watanabe K, Wanezaki M, Kutsuzawa D, Kato S, Tamura H, et al: HECT (Homologous to the E6-AP Carboxyl Terminus)-Type ubiquitin E3 ligase ITCH attenuates cardiac hypertrophy by suppressing the Wnt/β-catenin signaling pathway. Hypertension. 76:1868–1878. 2020. View Article : Google Scholar : PubMed/NCBI | |
Broquetas T and Carrion JA: Past, present, and future of long-term treatment for hepatitis B virus. World J Gastroenterol. 29:3964–3983. 2023. View Article : Google Scholar : PubMed/NCBI | |
Frenette C, Mendiratta-Lala M, Salgia R, Wong RJ, Sauer BG and Pillai A: ACG clinical guideline: Focal liver lesions. Am J Gastroenterol. 119:1235–1271. 2024. View Article : Google Scholar : PubMed/NCBI | |
European Association for the Study of the Liver (EASL); European Association for the Study of Diabetes (EASD); European Association for the Study of Obesity (EASO) and the European Association for the Study of the Liver (EASL), . EASL-EASD-EASO Clinical Practice Guidelines on the management of metabolic dysfunction-associated steatotic liver disease (MASLD). J Hepatol. 81:492–542. 2024. View Article : Google Scholar : PubMed/NCBI | |
Forrest EH, Atkinson SR, Richardson P, Masson S, Ryder S, Thursz MR and Allison M: ACG clinical guideline for alcoholic liver disease: The MELD threshold for corticosteroid treatment has yet to be established. Am J Gastroenterol. 114:175–176. 2019. View Article : Google Scholar : PubMed/NCBI | |
Dai JJ, Zhang YF and Zhang ZH: Global trends and hotspots of treatment for nonalcoholic fatty liver disease: A bibliometric and visualization analysis (2010–2023). World J Gastroenterol. 29:5339–5360. 2023. View Article : Google Scholar : PubMed/NCBI | |
Suddle A, Reeves H, Hubner R, Marshall A, Rowe I, Tiniakos D, Hubscher S, Callaway M, Sharma D, See TC, et al: British Society of Gastroenterology guidelines for the management of hepatocellular carcinoma in adults. Gut. 73:1235–1268. 2024. View Article : Google Scholar : PubMed/NCBI | |
Zhou Q, Li B and Li J: DLL4-Notch signalling in acute-on-chronic liver failure: State of the art and perspectives. Life Sci. 317:1214382023. View Article : Google Scholar : PubMed/NCBI | |
Conde de la Rosa L, Garcia-Ruiz C, Vallejo C, Baulies A, Nuñez S, Monte MJ, Marin JJG, Baila-Rueda L, Cenarro A, Civeira F, et al: STARD1 promotes NASH-driven HCC by sustaining the generation of bile acids through the alternative mitochondrial pathway. J Hepatol. 74:1429–1441. 2021. View Article : Google Scholar : PubMed/NCBI | |
Bernsmeier C, Singanayagam A, Patel VC, Wendon J and Antoniades CG: Immunotherapy in the treatment and prevention of infection in acute-on-chronic liver failure. Immunotherapy. 7:641–654. 2015. View Article : Google Scholar : PubMed/NCBI | |
Wulf J, Guckenberger M, Haedinger U, Oppitz U, Mueller G, Baier K and Flentje M: Stereotactic radiotherapy of primary liver cancer and hepatic metastases. Acta Oncol. 45:838–847. 2006. View Article : Google Scholar : PubMed/NCBI | |
Devarbhavi H, Asrani SK, Arab JP, Nartey YA, Pose E and Kamath PS: Global burden of liver disease: 2023 update. J Hepatol. 79:516–537. 2023. View Article : Google Scholar : PubMed/NCBI | |
Taha G, Ezra L and Abu-Freha N: Hepatitis C elimination: Opportunities and challenges in 2023. Viruses. 15:14132023. View Article : Google Scholar : PubMed/NCBI | |
Hsu YC, Huang DQ and Nguyen MH: Global burden of hepatitis B virus: Current status, missed opportunities and a call for action. Nat Rev Gastroenterol Hepatol. 20:524–537. 2023. View Article : Google Scholar : PubMed/NCBI | |
Hernandez-Evole H, Jimenez-Esquivel N, Pose E and Bataller R: Alcohol-associated liver disease: Epidemiology and management. Ann Hepatol. 29:1011622024. View Article : Google Scholar : PubMed/NCBI | |
Julien J, Ayer T, Bethea ED, Tapper EB and Chhatwal J: Projected prevalence and mortality associated with alcohol-related liver disease in the USA, 2019–40: A modelling study. Lancet Public Health. 5:e316–e23. 2020. View Article : Google Scholar : PubMed/NCBI | |
McGlynn KA, Petrick JL and El-Serag HB: Epidemiology of hepatocellular carcinoma. Hepatology. 73 (Suppl 1):S4–S13. 2021. View Article : Google Scholar | |
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. 21:236–247. 2023. View Article : Google Scholar : PubMed/NCBI | |
Taranto MP, Perez-Martinez G and Font de Valdez G: Effect of bile acid on the cell membrane functionality of lactic acid bacteria for oral administration. Res Microbiol. 157:720–725. 2006. View Article : Google Scholar : PubMed/NCBI | |
Han B, Lv X, Liu G, Li S, Fan J, Chen L, Huang Z, Lin G, Xu X, Huang Z, et al: Gut microbiota-related bile acid metabolism-FXR/TGR5 axis impacts the response to anti-α4β7-integrin therapy in humanized mice with colitis. Gut Microbes. 15:22321432023. View Article : Google Scholar : PubMed/NCBI | |
Liu HM, Chang ZY, Yang CW, Chang HH and Lee TY: Farnesoid X receptor agonist GW4064 protects lipopolysaccharide-induced intestinal epithelial barrier function and colorectal tumorigenesis signaling through the αKlotho/βKlotho/FGFs pathways in mice. Int J Mol Sci. 24:169322023. View Article : Google Scholar : PubMed/NCBI | |
Ploton M, Mazuy C, Gheeraert C, Dubois V, Berthier A, Dubois-Chevalier J, Maréchal X, Bantubungi K, Diemer H, Cianférani S, et al: The nuclear bile acid receptor FXR is a PKA- and FOXA2-sensitive activator of fasting hepatic gluconeogenesis. J Hepatol. 69:1099–1109. 2018. View Article : Google Scholar : PubMed/NCBI | |
Cao Y, Xiao Y, Zhou K, Yan J, Wang P, Yan W and Cai W: FXR agonist GW4064 improves liver and intestinal pathology and alters bile acid metabolism in rats undergoing small intestinal resection. Am J Physiol Gastrointest Liver Physiol. 317:G108–G115. 2019. View Article : Google Scholar : PubMed/NCBI | |
Yan Y, Sha Y, Huang X, Yuan W, Wu F, Hong J, Fang S, Huang B, Hu C, Wang B and Zhang X: Roux-en-Y gastric bypass improves metabolic conditions in association with increased serum bile acids level and hepatic Farnesoid X receptor expression in a T2DM rat model. Obes Surg. 29:2912–2922. 2019. View Article : Google Scholar : PubMed/NCBI | |
Watanabe M, Houten SM, Mataki C, Christoffolete MA, Kim BW, Sato H, Messaddeq N, Harney JW, Ezaki O, Kodama T, et al: Bile acids induce energy expenditure by promoting intracellular thyroid hormone activation. Nature. 439:484–489. 2006. View Article : Google Scholar : PubMed/NCBI | |
Zietak M and Kozak LP: Bile acids induce uncoupling protein 1-dependent thermogenesis and stimulate energy expenditure at thermoneutrality in mice. Am J Physiol Endocrinol Metab. 310:E346–E354. 2016. View Article : Google Scholar : PubMed/NCBI | |
Van Treuren W and Dodd D: Microbial contribution to the human metabolome: Implications for health and disease. Annu Rev Pathol. 15:345–369. 2020. View Article : Google Scholar : PubMed/NCBI | |
Cornell RP: Restriction of gut-derived endotoxin impairs DNA synthesis for liver regeneration. Am J Physiol. 249:R563–R569. 1985.PubMed/NCBI | |
Nolan JP: The role of intestinal endotoxin in liver injury: A long and evolving history. Hepatology. 52:1829–1835. 2010. View Article : Google Scholar : PubMed/NCBI | |
Liu H, Xi Q, Tan S, Qu Y, Meng Q, Zhang Y, Cheng Y and Wu G: The metabolite butyrate produced by gut microbiota inhibits cachexia-associated skeletal muscle atrophy by regulating intestinal barrier function and macrophage polarization. Int Immunopharmacol. 124:1110012023. View Article : Google Scholar : PubMed/NCBI | |
Tang G, Du Y, Guan H, Jia J, Zhu N, Shi Y, Rong S and Yuan W: Butyrate ameliorates skeletal muscle atrophy in diabetic nephropathy by enhancing gut barrier function and FFA2-mediated PI3K/Akt/mTOR signals. Br J Pharmacol. 179:159–178. 2022. View Article : Google Scholar : PubMed/NCBI | |
Meena AS, Shukla PK, Bell B, Giorgianni F, Caires R, Fernández-Peña C, Beranova S, Aihara E, Montrose MH, Chaib M, et al: TRPV6 channel mediates alcohol-induced gut barrier dysfunction and systemic response. Cell Rep. 39:1109372022. View Article : Google Scholar : PubMed/NCBI | |
Dominguez-Bello MG, Godoy-Vitorino F, Knight R and Blaser MJ: Role of the microbiome in human development. Gut. 68:1108–1114. 2019. View Article : Google Scholar : PubMed/NCBI | |
Han YH, Onufer EJ, Huang LH, Sprung RW, Davidson WS, Czepielewski RS, Wohltmann M, Sorci-Thomas MG, Warner BW and Randolph GJ: Enterically derived high-density lipoprotein restrains liver injury through the portal vein. Science. 373:eabe67292021. View Article : Google Scholar : PubMed/NCBI | |
Gomaa EZ: Human gut microbiota/microbiome in health and diseases: A review. Antonie Van Leeuwenhoek. 113:2019–2040. 2020. View Article : Google Scholar : PubMed/NCBI | |
Robles-Alonso V and Guarner F: Progress in the knowledge of the intestinal human microbiota. Nutr Hosp. 28:553–557. 2013.(In Spanish). PubMed/NCBI | |
Charlet R, Bortolus C, Barbet M, Sendid B and Jawhara S: A decrease in anaerobic bacteria promotes Candida glabrata overgrowth while β-glucan treatment restores the gut microbiota and attenuates colitis. Gut Pathog. 10:502018. View Article : Google Scholar : PubMed/NCBI | |
Charlet R, Pruvost Y, Tumba G, Istel F, Poulain D, Kuchler K, Sendid B and Jawhara S: Remodeling of the Candida glabrata cell wall in the gastrointestinal tract affects the gut microbiota and the immune response. Sci Rep. 8:33162018. View Article : Google Scholar : PubMed/NCBI | |
Bertin Y, Girardeau JP, Chaucheyras-Durand F, Lyan B, Pujos-Guillot E, Harel J and Martin C: Enterohaemorrhagic Escherichia coli gains a competitive advantage by using ethanolamine as a nitrogen source in the bovine intestinal content. Environ Microbiol. 13:365–377. 2011. View Article : Google Scholar : PubMed/NCBI | |
Ma HD, Zhao ZB, Ma WT, Liu QZ, Gao CY, Li L, Wang J, Tsuneyama K, Liu B, Zhang W, et al: Gut microbiota translocation promotes autoimmune cholangitis. J Autoimmun. 95:47–57. 2018. View Article : Google Scholar : PubMed/NCBI | |
Shao T, Zhao C, Li F, Gu Z, Liu L, Zhang L, Wang Y, He L, Liu Y, Liu Q, et al: Intestinal HIF-1α deletion exacerbates alcoholic liver disease by inducing intestinal dysbiosis and barrier dysfunction. J Hepatol. 69:886–895. 2018. View Article : Google Scholar : PubMed/NCBI | |
Giuffre M, Campigotto M, Campisciano G, Comar M and Croce LS: A story of liver and gut microbes: How does the intestinal flora affect liver disease? A review of the literature. Am J Physiol Gastrointest Liver Physiol. 318:G889–G906. 2020. View Article : Google Scholar : PubMed/NCBI | |
Bellot P, Frances R and Such J: Pathological bacterial translocation in cirrhosis: Pathophysiology, diagnosis and clinical implications. Liver Int. 33:31–39. 2013. View Article : Google Scholar : PubMed/NCBI | |
Giorgio V, Miele L, Principessa L, Ferretti F, Villa MP, Negro V, Grieco A, Alisi A and Nobili V: Intestinal permeability is increased in children with non-alcoholic fatty liver disease, and correlates with liver disease severity. Dig Liver Dis. 46:556–560. 2014. View Article : Google Scholar : PubMed/NCBI | |
Miele L, Valenza V, La Torre G, Montalto M, Cammarota G, Ricci R, Mascianà R, Forgione A, Gabrieli ML, Perotti G, et al: Increased intestinal permeability and tight junction alterations in nonalcoholic fatty liver disease. Hepatology. 49:1877–1887. 2009. View Article : Google Scholar : PubMed/NCBI | |
Rao R: Endotoxemia and gut barrier dysfunction in alcoholic liver disease. Hepatology. 50:638–644. 2009. View Article : Google Scholar : PubMed/NCBI | |
Sewell GW and Kaser A: Interleukin-23 in the pathogenesis of inflammatory bowel disease and implications for therapeutic intervention. J Crohns Colitis. 16 (Suppl 2):ii3–ii19. 2022. View Article : Google Scholar : PubMed/NCBI | |
Sokol H, Leducq V, Aschard H, Pham HP, Jegou S, Landman C, Cohen D, Liguori G, Bourrier A, Nion-Larmurier I, et al: Fungal microbiota dysbiosis in IBD. Gut. 66:1039–1048. 2017. View Article : Google Scholar : PubMed/NCBI | |
Ng SC, Benjamin JL, McCarthy NE, Hedin CR, Koutsoumpas A, Plamondon S, Price CL, Hart AL, Kamm MA, Forbes A, et al: Relationship between human intestinal dendritic cells, gut microbiota, and disease activity in Crohn's disease. Inflamm Bowel Dis. 17:2027–2037. 2011. View Article : Google Scholar : PubMed/NCBI | |
Coccia M, Harrison OJ, Schiering C, Asquith MJ, Becher B, Powrie F and Maloy KJ: IL-1β mediates chronic intestinal inflammation by promoting the accumulation of IL-17A secreting innate lymphoid cells and CD4(+) Th17 cells. J Exp Med. 209:1595–1609. 2012. View Article : Google Scholar : PubMed/NCBI | |
Ma TY, Boivin MA, Ye D, Pedram A and Said HM: Mechanism of TNF-{alpha} modulation of Caco-2 intestinal epithelial tight junction barrier: Role of myosin light-chain kinase protein expression. Am J Physiol Gastrointest Liver Physiol. 288:G422–G430. 2005. View Article : Google Scholar : PubMed/NCBI | |
He WQ, Wang J, Sheng JY, Zha JM, Graham WV and Turner JR: Contributions of myosin light chain kinase to regulation of epithelial paracellular permeability and mucosal homeostasis. Int J Mol Sci. 21:9932020. View Article : Google Scholar : PubMed/NCBI | |
Chotikatum S, Naim HY and El-Najjar N: Inflammation induced ER stress affects absorptive intestinal epithelial cells function and integrity. Int Immunopharmacol. 55:336–344. 2018. View Article : Google Scholar : PubMed/NCBI | |
Kinoshita N, Hiroi T, Ohta N, Fukuyama S, Park EJ and Kiyono H: Autocrine IL-15 mediates intestinal epithelial cell death via the activation of neighboring intraepithelial NK cells. J Immunol. 169:6187–6192. 2002. View Article : Google Scholar : PubMed/NCBI | |
Rohr M, Narasimhulu CA, Keewan E, Hamid S and Parthasarathy S: The dietary peroxidized lipid, 13-HPODE, promotes intestinal inflammation by mediating granzyme B secretion from natural killer cells. Food Funct. 11:9526–9534. 2020. View Article : Google Scholar : PubMed/NCBI | |
Yasuda K, Nakanishi K and Tsutsui H: Interleukin-18 in health and disease. Int J Mol Sci. 19:6492019. View Article : Google Scholar | |
Woznicki JA, Saini N, Flood P, Rajaram S, Lee CM, Stamou P, Skowyra A, Bustamante-Garrido M, Regazzoni K, Crawford N, et al: TNF-α synergises with IFN-γ to induce caspase-8-JAK1/2-STAT1-dependent death of intestinal epithelial cells. Cell Death Dis. 12:8642021. View Article : Google Scholar : PubMed/NCBI | |
Chang JT: Pathophysiology of inflammatory bowel diseases. N Engl J Med. 383:2652–2664. 2020. View Article : Google Scholar : PubMed/NCBI | |
Romagnani S: Lymphokine production by human T cells in disease states. Annu Rev Immunol. 12:227–257. 1994. View Article : Google Scholar : PubMed/NCBI | |
Fort MM, Cheung J, Yen D, Li J, Zurawski SM, Lo S, Menon S, Clifford T, Hunte B, Lesley R, et al: IL-25 induces IL-4, IL-5, and IL-13 and Th2-associated pathologies in vivo. Immunity. 15:985–995. 2001. View Article : Google Scholar : PubMed/NCBI | |
Lin Y, Li B, Yang X, Liu T, Shi T, Deng B, Zhang Y, Jia L, Jiang Z and He R: Non-hematopoietic STAT6 induces epithelial tight junction dysfunction and promotes intestinal inflammation and tumorigenesis. Mucosal Immunol. 12:1304–1315. 2019. View Article : Google Scholar : PubMed/NCBI | |
Ceponis PJ, Botelho F, Richards CD and McKay DM: Interleukins 4 and 13 increase intestinal epithelial permeability by a phosphatidylinositol 3-kinase pathway. Lack of evidence for STAT 6 involvement. J Biol Chem. 275:29132–29137. 2000. View Article : Google Scholar : PubMed/NCBI | |
He L, Liu T, Shi Y, Tian F, Hu H, Deb DK, Bissonnette M and Li YC: Gut epithelial Vitamin D receptor regulates Microbiota-dependent mucosal inflammation by suppressing intestinal epithelial cell apoptosis. Endocrinology. 159:967–979. 2018. View Article : Google Scholar : PubMed/NCBI | |
Lee SH, Kwon JE and Cho ML: Immunological pathogenesis of inflammatory bowel disease. Intest Res. 16:26–42. 2018. View Article : Google Scholar : PubMed/NCBI | |
Wang X, Ni J, You Y, Feng G, Zhang S, Bao W, Hou H, Li H, Liu L, Zheng M, et al: SNX10-mediated LPS sensing causes intestinal barrier dysfunction via a caspase-5-dependent signaling cascade. EMBO J. 40:e1080802021. View Article : Google Scholar : PubMed/NCBI | |
Li Q, Rempel JD, Yang J and Minuk GY: The effects of Pathogen-associated molecular patterns on peripheral blood monocytes in patients with Non-alcoholic fatty liver disease. J Clin Exp Hepatol. 12:808–817. 2022. View Article : Google Scholar : PubMed/NCBI | |
Nakamoto N and Kanai T: Role of toll-like receptors in immune activation and tolerance in the liver. Front Immunol. 5:2212014. View Article : Google Scholar : PubMed/NCBI | |
Szabo G, Dolganiuc A and Mandrekar P: Pattern recognition receptors: A contemporary view on liver diseases. Hepatology. 44:287–298. 2006. View Article : Google Scholar : PubMed/NCBI | |
Kesar V and Odin JA: Toll-like receptors and liver disease. Liver Int. 34:184–196. 2014. View Article : Google Scholar : PubMed/NCBI | |
Hardin PE: From biological clock to biological rhythms. Genome Biol. 1:REVIEWS10232000. View Article : Google Scholar : PubMed/NCBI | |
Jouffe C, Weger BD, Martin E, Atger F, Weger M, Gobet C, Ramnath D, Charpagne A, Morin-Rivron D, Powell EE, et al: Disruption of the circadian clock component BMAL1 elicits an endocrine adaption impacting on insulin sensitivity and liver disease. Proc Natl Acad Sci USA. 119:e22000831192022. View Article : Google Scholar : PubMed/NCBI | |
Kinouchi K and Sassone-Corsi P: Metabolic rivalry: Circadian homeostasis and tumorigenesis. Nat Rev Cancer. 20:645–661. 2020. View Article : Google Scholar : PubMed/NCBI | |
Nassan M and Videnovic A: Circadian rhythms in neurodegenerative disorders. Nat Rev Neurol. 18:7–24. 2022. View Article : Google Scholar : PubMed/NCBI | |
Song S, Tien CL, Cui H, Basil P, Zhu N, Gong Y, Li W, Li H, Fan Q, Min Choi J, et al: Myocardial Rev-erb-mediated diurnal metabolic rhythm and obesity paradox. Circulation. 145:448–464. 2022. View Article : Google Scholar : PubMed/NCBI | |
Choi H, Rao MC and Chang EB: Gut microbiota as a transducer of dietary cues to regulate host circadian rhythms and metabolism. Nat Rev Gastroenterol Hepatol. 18:679–689. 2021. View Article : Google Scholar : PubMed/NCBI | |
Heddes M, Altaha B, Niu Y, Reitmeier S, Kleigrewe K, Haller D and Kiessling S: The intestinal clock drives the microbiome to maintain gastrointestinal homeostasis. Nat Commun. 13:60682022. View Article : Google Scholar : PubMed/NCBI | |
Thaiss CA, Zeevi D, Levy M, Zilberman-Schapira G, Suez J, Tengeler AC, Abramson L, Katz MN, Korem T, Zmora N, et al: Transkingdom control of microbiota diurnal oscillations promotes metabolic homeostasis. Cell. 159:514–529. 2014. View Article : Google Scholar : PubMed/NCBI | |
Backhed F, Ding H, Wang T, Hooper LV, Koh GY, Nagy A, Semenkovich CF and Gordon JI: The gut microbiota as an environmental factor that regulates fat storage. Proc Natl Acad Sci USA. 101:15718–15723. 2004. View Article : Google Scholar : PubMed/NCBI | |
Rabot S, Membrez M, Bruneau A, Gerard P, Harach T, Moser M, Raymond F, Mansourian R and Chou CJ: Germ-free C57BL/6J mice are resistant to high-fat-diet-induced insulin resistance and have altered cholesterol metabolism. FASEB J. 24:4948–4959. 2010. View Article : Google Scholar : PubMed/NCBI | |
Zhang X, Coker OO, Chu ES, Fu K, Lau HCH, Wang YX, Chan AWH, Wei H, Yang X, Sung JJY and Yu J: Dietary cholesterol drives fatty liver-associated liver cancer by modulating gut microbiota and metabolites. Gut. 70:761–774. 2021. View Article : Google Scholar : PubMed/NCBI | |
Wang B, Kong Q, Li X, Zhao J, Zhang H, Chen W and Wang G: A High-fat diet increases gut microbiota biodiversity and energy expenditure due to nutrient difference. Nutrients. 12:31972020. View Article : Google Scholar : PubMed/NCBI | |
Turnbaugh PJ, Ley RE, Mahowald MA, Magrini V, Mardis ER and Gordon JI: An Obesity-associated gut microbiome with increased capacity for energy harvest. Nature. 444:1027–1031. 2006. View Article : Google Scholar : PubMed/NCBI | |
Riva A, Borgo F, Lassandro C, Verduci E, Morace G, Borghi E and Berry D: Pediatric obesity is associated with an altered gut microbiota and discordant shifts in Firmicutes populations. Environ Microbiol. 19:95–105. 2017. View Article : Google Scholar : PubMed/NCBI | |
Palmnas-Bedard MSA, Costabile G, Vetrani C, Aberg S, Hjalmarsson Y, Dicksved J, Riccardi G and Landberg R: The human gut microbiota and glucose metabolism: A scoping review of key bacteria and the potential role of SCFAs. Am J Clin Nutr. 116:862–874. 2022. View Article : Google Scholar : PubMed/NCBI | |
Dunn R, Wetten A, McPherson S and Donnelly MC: Viral hepatitis in 2021: The challenges remaining and how we should tackle them. World J Gastroenterol. 28:76–95. 2022. View Article : Google Scholar : PubMed/NCBI | |
Zhao X and Guo S: Methods for visualizing intracellular organelles. J Vis Exp. Mar 3–2023.doi: 10.3791/64966. | |
Zhao W, Ma L, Cai C and Gong X: Caffeine Inhibits NLRP3 inflammasome activation by suppressing MAPK/NF-κB and A2aR signaling in LPS-induced THP-1 macrophages. Int J Biol Sci. 15:1571–1581. 2019. View Article : Google Scholar : PubMed/NCBI | |
Chou HH, Chien WH, Wu LL, Cheng CH, Chung CH, Horng JH, Ni YH, Tseng HT, Wu D, Lu X, et al: Age-related immune clearance of hepatitis B virus infection requires the establishment of gut microbiota. Proc Natl Acad Sci USA. 112:2175–2180. 2015. View Article : Google Scholar : PubMed/NCBI | |
Chauhan A, Kumar R, Sharma S, Mahanta M, Vayuuru SK, Nayak B and Kumar S: Shalimar: Fecal microbiota transplantation in Hepatitis B e antigen-positive chronic Hepatitis B patients: A pilot study. Dig Dis Sci. 66:873–880. 2021. View Article : Google Scholar : PubMed/NCBI | |
Huang H, Ren Z, Gao X, Hu X, Zhou Y, Jiang J, Lu H, Yin S, Ji J, Zhou L and Zheng S: Integrated analysis of microbiome and host transcriptome reveals correlations between gut microbiota and clinical outcomes in HBV-related hepatocellular carcinoma. Genome Med. 12:1022020. View Article : Google Scholar : PubMed/NCBI | |
Preveden T, Scarpellini E, Milic N, Luzza F and Abenavoli L: Gut microbiota changes and chronic hepatitis C virus infection. Expert Rev Gastroenterol Hepatol. 11:813–819. 2017. View Article : Google Scholar : PubMed/NCBI | |
Shen Y, Wu SD, Chen Y, Li XY, Zhu Q, Nakayama K, Zhang WQ, Weng CZ, Zhang J, Wang HK, et al: Alterations in gut microbiome and metabolomics in chronic hepatitis B infection-associated liver disease and their impact on peripheral immune response. Gut Microbes. 15:21550182023. View Article : Google Scholar : PubMed/NCBI | |
Wei X, Yan X, Zou D, Yang Z, Wang X, Liu W, Wang S, Li X, Han J, Huang L and Yuan J: Abnormal fecal microbiota community and functions in patients with hepatitis B liver cirrhosis as revealed by a metagenomic approach. BMC Gastroenterol. 13:1752013. View Article : Google Scholar : PubMed/NCBI | |
Bajaj JS, Liu EJ, Kheradman R, Fagan A, Heuman DM, White M, Gavis EA, Hylemon P, Sikaroodi M and Gillevet PM: Fungal dysbiosis in cirrhosis. Gut. 67:1146–1154. 2018. View Article : Google Scholar : PubMed/NCBI | |
Aly AM, Adel A, El-Gendy AO, Essam TM and Aziz RK: Gut microbiome alterations in patients with stage 4 hepatitis C. Gut Pathog. 8:422016. View Article : Google Scholar : PubMed/NCBI | |
Luther J, Khan S, Gala MK, Kedrin D, Sridharan G, Goodman RP, Garber JJ, Masia R, Diagacomo E, Adams D, et al: Hepatic gap junctions amplify alcohol liver injury by propagating cGAS-mediated IRF3 activation. Proc Natl Acad Sci USA. 117:11667–11673. 2020. View Article : Google Scholar : PubMed/NCBI | |
Jophlin LL, Singal AK, Bataller R, Wong RJ, Sauer BG, Terrault NA and Shah VH: ACG clinical guideline: Alcohol-associated liver disease. Am J Gastroenterol. 119:30–54. 2024. View Article : Google Scholar : PubMed/NCBI | |
Huang DQ, Mathurin P, Cortez-Pinto H and Loomba R: Global epidemiology of alcohol-associated cirrhosis and HCC: Trends, projections and risk factors. Nat Rev Gastroenterol Hepatol. 20:37–49. 2023. View Article : Google Scholar : PubMed/NCBI | |
Singal AK, Bataller R, Ahn J, Kamath PS and Shah VH: ACG clinical guideline: Alcoholic liver disease. Am J Gastroenterol. 113:175–194. 2018. View Article : Google Scholar : PubMed/NCBI | |
Acharya C and Bajaj JS: Gut Microbiota and complications of liver disease. Gastroenterol Clin North Am. 46:155–169. 2017. View Article : Google Scholar : PubMed/NCBI | |
Dubinkina VB, Tyakht AV, Odintsova VY, Yarygin KS, Kovarsky BA, Pavlenko AV, Ischenko DS, Popenko AS, Alexeev DG, Taraskina AY, et al: Links of gut microbiota composition with alcohol dependence syndrome and alcoholic liver disease. Microbiome. 5:1412017. View Article : Google Scholar : PubMed/NCBI | |
Blaak EE, Canfora EE, Theis S, Frost G, Groen AK, 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. 11:411–455. 2020. View Article : Google Scholar : PubMed/NCBI | |
Wang Z, Zhang X, Zhu L, Yang X, He F, Wang T, Bao T, Lu H, Wang H and Yang S: Inulin alleviates inflammation of alcoholic liver disease via SCFAs-inducing suppression of M1 and facilitation of M2 macrophages in mice. Int Immunopharmacol. 78:1060622020. View Article : Google Scholar : PubMed/NCBI | |
Yang X, He F, Zhang Y, Xue J, Li K, Zhang X, Zhu L, Wang Z, Wang H and Yang S: Inulin ameliorates alcoholic liver disease via suppressing LPS-TLR4-mpsi axis and modulating gut microbiota in mice. Alcohol Clin Exp Res. 43:411–424. 2019. View Article : Google Scholar : PubMed/NCBI | |
Deng M, Qu F, Chen L, Liu C, Zhang M, Ren F, Guo H, Zhang H, Ge S, Wu C and Zhao L: SCFAs alleviated steatosis and inflammation in mice with NASH induced by MCD. J Endocrinol. 245:425–437. 2020. View Article : Google Scholar : PubMed/NCBI | |
Mundi MS, Velapati S, Patel J, Kellogg TA, Abu Dayyeh BK and Hurt RT: Evolution of NAFLD and its management. Nutr Clin Pract. 35:72–84. 2020. View Article : Google Scholar : PubMed/NCBI | |
Aron-Wisnewsky J, Warmbrunn MV, Nieuwdorp M and Clement K: Metabolism and metabolic disorders and the microbiome: The intestinal microbiota associated with obesity, lipid metabolism, and metabolic health-pathophysiology and therapeutic strategies. Gastroenterology. 160:573–599. 2021. View Article : Google Scholar : PubMed/NCBI | |
Canfora EE, Meex RCR, Venema K and Blaak EE: Gut microbial metabolites in obesity, NAFLD and T2DM. Nat Rev Endocrinol. 15:261–273. 2019. View Article : Google Scholar : PubMed/NCBI | |
Kolodziejczyk AA, Zheng D, Shibolet O and Elinav E: The role of the microbiome in NAFLD and NASH. EMBO Mol Med. 11:e93022019. View Article : Google Scholar : PubMed/NCBI | |
Henao-Mejia J, Elinav E, Jin C, Hao L, Mehal WZ, Strowig T, Thaiss CA, Kau AL, Eisenbarth SC, Jurczak MJ, et al: Inflammasome-mediated dysbiosis regulates progression of NAFLD and obesity. Nature. 482:179–185. 2012. View Article : Google Scholar : PubMed/NCBI | |
Zhong S, Li L, Liang N, Zhang L, Xu X, Chen S and Yin H: Acetaldehyde dehydrogenase 2 regulates HMG-CoA reductase stability and cholesterol synthesis in the liver. Redox Biol. 41:1019192021. View Article : Google Scholar : PubMed/NCBI | |
Inokuchi S, Tsukamoto H, Park E, Liu ZX, Brenner DA and Seki E: Toll-like receptor 4 mediates alcohol-induced steatohepatitis through bone marrow-derived and endogenous liver cells in mice. Alcohol Clin Exp Res. 35:1509–1518. 2011.PubMed/NCBI | |
Bogatyrev SR, Rolando JC and Ismagilov RF: Self-reinoculation with fecal flora changes microbiota density and composition leading to an altered bile-acid profile in the mouse small intestine. Microbiome. 8:192020. View Article : Google Scholar : PubMed/NCBI | |
Ma C, Han M, Heinrich B, Fu Q, Zhang Q, Sandhu M, Agdashian D, Terabe M, Berzofsky JA, Fako V, et al: Gut microbiome-mediated bile acid metabolism regulates liver cancer via NKT cells. Science. 360:eaan59312018. View Article : Google Scholar : PubMed/NCBI | |
Endo H, Niioka M, Kobayashi N, Tanaka M and Watanabe T: Butyrate-producing probiotics reduce nonalcoholic fatty liver disease progression in rats: New insight into the probiotics for the gut-liver axis. PLoS One. 8:e633882013. View Article : Google Scholar : PubMed/NCBI | |
Ma YY, Li L, Yu CH, Shen Z, Chen LH and Li YM: Effects of probiotics on nonalcoholic fatty liver disease: A meta-analysis. World J Gastroenterol. 19:6911–6918. 2013. View Article : Google Scholar : PubMed/NCBI | |
Kirpich IA, Solovieva NV, Leikhter SN, Shidakova NA, Lebedeva OV, Sidorov PI, Bazhukova TA, Soloviev AG, Barve SS, McClain CJ and Cave M: Probiotics restore bowel flora and improve liver enzymes in human alcohol-induced liver injury: A pilot study. Alcohol. 42:675–682. 2008. View Article : Google Scholar : PubMed/NCBI | |
Schuppan D and Afdhal NH: Liver cirrhosis. Lancet. 371:838–851. 2008. View Article : Google Scholar : PubMed/NCBI | |
Quiroz-Aldave JE, Gamarra-Osorio ER, Durand-Vasquez MDC, Rafael-Robles LDP, Gonzales-Yovera JG, Quispe-Flores MA, Concepción-Urteaga LA, Román-González A, Paz-Ibarra J and Concepción-Zavaleta MJ: From liver to hormones: The endocrine consequences of cirrhosis. World J Gastroenterol. 30:1073–1095. 2024. View Article : Google Scholar : PubMed/NCBI | |
Horn P and Tacke F: Metabolic reprogramming in liver fibrosis. Cell Metab. 36:1439–1455. 2024. View Article : Google Scholar : PubMed/NCBI | |
Yang X, Li Q, Liu W, Zong C, Wei L, Shi Y and Han Z: Mesenchymal stromal cells in hepatic fibrosis/cirrhosis: From pathogenesis to treatment. Cell Mol Immunol. 20:583–599. 2023. View Article : Google Scholar : PubMed/NCBI | |
Iredale JP, Thompson A and Henderson NC: Extracellular matrix degradation in liver fibrosis: Biochemistry and regulation. Biochim Biophys Acta. 1832:876–883. 2013. View Article : Google Scholar : PubMed/NCBI | |
Bataller R and Brenner DA: Liver fibrosis. J Clin Invest. 115:209–218. 2005. View Article : Google Scholar : PubMed/NCBI | |
Smith A, Baumgartner K and Bositis C: Cirrhosis: Diagnosis and management. Am Fam Physician. 100:759–770. 2019.PubMed/NCBI | |
Tapper EB and Parikh ND: Diagnosis and management of cirrhosis and its complications: A review. JAMA. 329:1589–1602. 2023. View Article : Google Scholar : PubMed/NCBI | |
GBD 2019 Diseases and Injuries Collaborators: Global burden of 369 diseases and injuries in 204 countries and territories, 1990–2019: A systematic analysis for the Global Burden of Disease Study 2019. Lancet. 396:1204–1222. 2020. View Article : Google Scholar | |
Asrani SK, Devarbhavi H, Eaton J and Kamath PS: Burden of liver diseases in the world. J Hepatol. 70:151–171. 2019. View Article : Google Scholar : PubMed/NCBI | |
European Association for the Study of the Liver. Electronic address, . simpleeasloffice@easloffice.eu and European Association for the Study of the Liver: EASL clinical practice guidelines on Acute-on-chronic liver failure. J Hepatol. 79:461–491. 2023. View Article : Google Scholar : PubMed/NCBI | |
Gines P, Krag A, Abraldes JG, Sola E, Fabrellas N and Kamath PS: Liver cirrhosis. Lancet. 398:1359–1376. 2021. View Article : Google Scholar : PubMed/NCBI | |
Groschwitz KR and Hogan SP: Intestinal barrier function: Molecular regulation and disease pathogenesis. J Allergy Clin Immunol. 124:3–22. 2009. View Article : Google Scholar : PubMed/NCBI | |
Nishimura N, Kaji K, Kitagawa K, Sawada Y, Furukawa M, Ozutsumi T, Fujinaga Y, Tsuji Y, Takaya H, Kawaratani H, et al: Intestinal permeability is a mechanical rheostat in the pathogenesis of liver cirrhosis. Int J Mol Sci. 22:69212021. View Article : Google Scholar : PubMed/NCBI | |
Trebicka J, Macnaughtan J, Schnabl B, Shawcross DL and Bajaj JS: The microbiota in cirrhosis and its role in hepatic decompensation. J Hepatol. 75 (Suppl 1):S67–S81. 2021. View Article : Google Scholar : PubMed/NCBI | |
Benjamin J, Singla V, Arora I, Sood S and Joshi YK: Intestinal permeability and complications in liver cirrhosis: A prospective cohort study. Hepatol Res. 43:200–207. 2013. View Article : Google Scholar : PubMed/NCBI | |
Shibamoto A, Kaji K, Nishimura N, Kubo T, Iwai S, Tomooka F, Suzuki J, Tsuji Y, Fujinaga Y, Kawaratani H, et al: Vitamin D deficiency exacerbates alcohol-related liver injury via gut barrier disruption and hepatic overload of endotoxin. J Nutr Biochem. 122:1094502023. View Article : Google Scholar : PubMed/NCBI | |
Suk KT and Kim DJ: Gut microbiota: Novel therapeutic target for nonalcoholic fatty liver disease. Expert Rev Gastroenterol Hepatol. 13:193–204. 2019. View Article : Google Scholar : PubMed/NCBI | |
Albillos A, Lario M and Alvarez-Mon M: Cirrhosis-associated immune dysfunction: Distinctive features and clinical relevance. J Hepatol. 61:1385–1396. 2014. View Article : Google Scholar : PubMed/NCBI | |
Fukui H: Role of gut dysbiosis in liver diseases: What have we learned so far? Diseases. 7:582019. View Article : Google Scholar : PubMed/NCBI | |
Camilleri M, Madsen K, Spiller R, Greenwood-Van Meerveld B and Verne GN: Intestinal barrier function in health and gastrointestinal disease. Neurogastroenterol Motil. 24:503–512. 2012. View Article : Google Scholar : PubMed/NCBI | |
Ghosh S, Whitley CS, Haribabu B and Jala VR: Regulation of intestinal barrier function by microbial metabolites. Cell Mol Gastroenterol Hepatol. 11:1463–1482. 2021. View Article : Google Scholar : PubMed/NCBI | |
Oh TG, Kim SM, Caussy C, Fu T, Guo J, Bassirian S, Singh S, Madamba EV, Bettencourt R, Richards L, et al: A universal Gut-microbiome-derived signature predicts cirrhosis. Cell Metab. 32:878–888.e6. 2020. View Article : Google Scholar : PubMed/NCBI | |
Chang CS, Chen GH, Lien HC and Yeh HZ: Small intestine dysmotility and bacterial overgrowth in cirrhotic patients with spontaneous bacterial peritonitis. Hepatology. 28:1187–1190. 1998. View Article : Google Scholar : PubMed/NCBI | |
Corradi F, Brusasco C, Fernandez J, Vila J, Ramirez MJ, Seva-Pereira T, Fernández-Varo G, Mosbah IB, Acevedo J, Silva A, et al: Effects of pentoxifylline on intestinal bacterial overgrowth, bacterial translocation and spontaneous bacterial peritonitis in cirrhotic rats with ascites. Dig Liver Dis. 44:239–244. 2012. View Article : Google Scholar : PubMed/NCBI | |
Bajaj JS, Heuman DM, Hylemon PB, Sanyal AJ, Puri P, Sterling RK, Luketic V, Stravitz RT, Siddiqui MS, Fuchs M, et al: Randomised clinical trial: Lactobacillus GG modulates gut microbiome, metabolome and endotoxemia in patients with cirrhosis. Aliment Pharmacol Ther. 39:1113–1125. 2014. View Article : Google Scholar : PubMed/NCBI | |
Bajaj JS, Kassam Z, Fagan A, Gavis EA, Liu E, Cox IJ, Kheradman R, Heuman D, Wang J, Gurry T, et al: Fecal microbiota transplant from a rational stool donor improves hepatic encephalopathy: A randomized clinical trial. Hepatology. 66:1727–1738. 2017. View Article : Google Scholar : PubMed/NCBI | |
Bajaj JS, Hylemon PB, Ridlon JM, Heuman DM, Daita K, White MB, Monteith P, Noble NA, Sikaroodi M and Gillevet PM: Colonic mucosal microbiome differs from stool microbiome in cirrhosis and hepatic encephalopathy and is linked to cognition and inflammation. Am J Physiol Gastrointest Liver Physiol. 303:G675–G685. 2012. View Article : Google Scholar : PubMed/NCBI | |
Chen Y, Yang F, Lu H, Wang B, Chen Y, Lei D, Wang Y, Zhu B and Li L: Characterization of fecal microbial communities in patients with liver cirrhosis. Hepatology. 54:562–572. 2011. View Article : Google Scholar : PubMed/NCBI | |
Shen TD, Daniel SG, Patel S, Kaplan E, Phung L, Lemelle-Thomas K, Chau L, Herman L, Trisolini C, Stonelake A, et al: The Mucosally-adherent rectal microbiota contains features unique to alcohol-related cirrhosis. Gut Microbes. 13:19877812021. View Article : Google Scholar : PubMed/NCBI | |
Egger M, Horvath A, Pruller F, Fickert P, Finkelman M, Kriegl L, Grønbaek H, Møller HJ, Prattes J, Krause R, et al: Fungal translocation measured by serum 1,3-β-D-glucan correlates with severity and outcome of liver cirrhosis-A pilot study. Liver Int. 43:1975–1983. 2023. View Article : Google Scholar : PubMed/NCBI | |
Saaoud F, Liu L, Xu K, Cueto R, Shao Y, Lu Y, Sun Y, Snyder NW, Wu S, Yang L, et al: Aorta- and liver-generated TMAO enhances trained immunity for increased inflammation via ER stress/mitochondrial ROS/glycolysis pathways. JCI Insight. 8:e1581832023. View Article : Google Scholar : PubMed/NCBI | |
Pleguezuelo M, Benitez JM, Jurado J, Montero JL and De la Mata M: Diagnosis and management of bacterial infections in decompensated cirrhosis. World J Hepatol. 5:16–25. 2013. View Article : Google Scholar : PubMed/NCBI | |
Kim J, Ahn SW, Kim JY, Whon TW, Lim SK, Ryu BH, Han NS, Choi HJ, Roh SW and Lee SH: Probiotic Lactobacilli ameliorate alcohol-induced hepatic damage via gut microbial alteration. Front Microbiol. 13:8692502022. View Article : Google Scholar : PubMed/NCBI | |
Llovet JM, Pinyol R, Yarchoan M, Singal AG, Marron TU, Schwartz M, Pikarsky E, Kudo M and Finn RS: Adjuvant and neoadjuvant immunotherapies in hepatocellular carcinoma. Nat Rev Clin Oncol. 21:294–311. 2024. View Article : Google Scholar : PubMed/NCBI | |
Ding JH, Jin Z, Yang XX, Lou J, Shan WX, Hu YX, Du Q, Liao QS, Xie R and Xu JY: Role of gut microbiota via the gut-liver-brain axis in digestive diseases. World J Gastroenterol. 26:6141–6162. 2020. View Article : Google Scholar : PubMed/NCBI | |
Queipo-Ortuño MI, Boto-Ordóñez M, Murri M, Gomez-Zumaquero JM, Clemente-Postigo M, Estruch R, Cardona Diaz F, Andrés-Lacueva C and Tinahones FJ: Influence of red wine polyphenols and ethanol on the gut microbiota ecology and biochemical biomarkers. Am J Clin Nutr. 95:1323–1334. 2012. View Article : Google Scholar : PubMed/NCBI | |
Mutlu E, Keshavarzian A, Engen P, Forsyth CB, Sikaroodi M and Gillevet P: Intestinal dysbiosis: A possible mechanism of alcohol-induced endotoxemia and alcoholic steatohepatitis in rats. Alcohol Clin Exp Res. 33:1836–1846. 2009. View Article : Google Scholar : PubMed/NCBI | |
Yan AW, Fouts DE, Brandl J, Starkel P, Torralba M, Schott E, Tsukamoto H, Nelson KE, Brenner DA and Schnabl B: Enteric dysbiosis associated with a mouse model of alcoholic liver disease. Hepatology. 53:96–105. 2011. View Article : Google Scholar : PubMed/NCBI | |
Mutlu EA, Gillevet PM, Rangwala H, Sikaroodi M, Naqvi A, Engen PA, Kwasny M, Lau CK and Keshavarzian A: Colonic microbiome is altered in alcoholism. Am J Physiol Gastrointest Liver Physiol. 302:G966–G978. 2012. View Article : Google Scholar : PubMed/NCBI | |
Boursier J, Mueller O, Barret M, Machado M, Fizanne L, Araujo-Perez F, Guy CD, Seed PC, Rawls JF, David LA, et al: The severity of nonalcoholic fatty liver disease is associated with gut dysbiosis and shift in the metabolic function of the gut microbiota. Hepatology. 63:764–775. 2016. View Article : Google Scholar : PubMed/NCBI | |
Wang B, Jiang X, Cao M, Ge J, Bao Q, Tang L, Chen Y and Li L: Altered fecal microbiota correlates with liver biochemistry in nonobese patients with Non-alcoholic fatty liver disease. Sci Rep. 6:320022016. View Article : Google Scholar : PubMed/NCBI | |
Singh DP, Khare P, Bijalwan V, Baboota RK, Singh J, Kondepudi KK, Chopra K and Bishnoi M: Coadministration of isomalto-oligosaccharides augments metabolic health benefits of cinnamaldehyde in high fat diet fed mice. Biofactors. 43:821–835. 2017. View Article : Google Scholar : PubMed/NCBI | |
Ponziani FR, Bhoori S, Castelli C, Putignani L, Rivoltini L, Del Chierico F, Sanguinetti M, Morelli D, Paroni Sterbini F, Petito V, et al: Hepatocellular carcinoma is associated with gut microbiota profile and inflammation in nonalcoholic fatty liver disease. Hepatology. 69:107–120. 2019. View Article : Google Scholar : PubMed/NCBI | |
Sarangi AN, Goel A, Singh A, Sasi A and Aggarwal R: Faecal bacterial microbiota in patients with cirrhosis and the effect of lactulose administration. BMC Gastroenterol. 17:1252017. View Article : Google Scholar : PubMed/NCBI | |
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. 68:1014–1023. 2019. View Article : Google Scholar : PubMed/NCBI | |
Schneider KM, Mohs A, Gui W, Galvez EJC, Candels LS, Hoenicke L, Muthukumarasamy U, Holland CH, Elfers C, Kilic K, et al: Imbalanced gut microbiota fuels hepatocellular carcinoma development by shaping the hepatic inflammatory microenvironment. Nat Commun. 13:39642022. View Article : Google Scholar : PubMed/NCBI | |
Li Z, Zhang Y, Hong W, Wang B, Chen Y, Yang P, Zhou J, Fan J, Zeng Z and Du S: Gut microbiota modulate radiotherapy-associated antitumor immune responses against hepatocellular carcinoma Via STING signaling. Gut Microbes. 14:21190552022. View Article : Google Scholar : PubMed/NCBI | |
Thoen RU, Longo L, Leonhardt LC, Pereira MHM, Rampelotto PH, Cerski CTS and Álvares-da-Silva MR: Alcoholic liver disease and intestinal microbiota in an experimental model: Biochemical, inflammatory, and histologic parameters. Nutrition. 106:1118882023. View Article : Google Scholar : PubMed/NCBI | |
McMahan RH, Hulsebus HJ, Najarro KM, Giesy LE, Frank DN and Kovacs EJ: Changes in gut microbiome correlate with intestinal barrier dysfunction and inflammation following a 3-day ethanol exposure in aged mice. Alcohol. 107:136–143. 2023. View Article : Google Scholar : PubMed/NCBI | |
Sangineto M, Grander C, Grabherr F, Mayr L, Enrich B, Schwärzler J, Dallio M, Bukke VN, Moola A, Moschetta A, et al: Recovery of Bacteroides thetaiotaomicron ameliorates hepatic steatosis in experimental alcohol-related liver disease. Gut Microbes. 14:20890062022. View Article : Google Scholar : PubMed/NCBI | |
Day AW and Kumamoto CA: Gut microbiome dysbiosis in alcoholism: Consequences for health and recovery. Front Cell Infect Microbiol. 12:8401642022. View Article : Google Scholar : PubMed/NCBI | |
Wang W, Li Q, Chai W, Sun C, Zhang T, Zhao C, Yuan Y, Wang X, Liu H and Ye H: Lactobacillus paracasei Jlus66 extenuate oxidative stress and inflammation via regulation of intestinal flora in rats with non alcoholic fatty liver disease. Food Sci Nutr. 7:2636–2646. 2019. View Article : Google Scholar : PubMed/NCBI | |
Safari Z and Gerard P: The links between the gut microbiome and non-alcoholic fatty liver disease (NAFLD). Cell Mol Life Sci. 76:1541–1558. 2019. View Article : Google Scholar : PubMed/NCBI | |
Ji Y, Yin Y, Li Z and Zhang W: Gut Microbiota-derived components and metabolites in the progression of non-alcoholic fatty liver disease (NAFLD). Nutrients. 11:17122019. View Article : Google Scholar : PubMed/NCBI | |
Fang J, Yu CH, Li XJ, Yao JM, Fang ZY, Yoon SH and Yu WY: Gut dysbiosis in nonalcoholic fatty liver disease: Pathogenesis, diagnosis, and therapeutic implications. Front Cell Infect Microbiol. 12:9970182022. View Article : Google Scholar : PubMed/NCBI | |
Liu S and Yang X: Intestinal flora plays a role in the progression of hepatitis-cirrhosis-liver cancer. Front Cell Infect Microbiol. 13:11401262023. View Article : Google Scholar : PubMed/NCBI | |
Lee NY and Suk KT: The role of the gut microbiome in liver cirrhosis treatment. Int J Mol Sci. 22:1992020. View Article : Google Scholar : PubMed/NCBI | |
Qin N, Yang F, Li A, Prifti E, Chen Y, Shao L, Guo J, Le Chatelier E, Yao J, Wu L, et al: Alterations of the human gut microbiome in liver cirrhosis. Nature. 513:59–64. 2014. View Article : Google Scholar : PubMed/NCBI | |
Akkiz H: The gut microbiome and hepatocellular carcinoma. J Gastrointest Cancer. 52:1314–1319. 2021. View Article : Google Scholar : PubMed/NCBI | |
Schwabe RF and Greten TF: Gut microbiome in HCC-mechanisms, diagnosis and therapy. J Hepatol. 72:230–238. 2020. View Article : Google Scholar : PubMed/NCBI | |
Zhang S, Hou L and Sun Q: Correlation analysis of intestinal flora and immune function in patients with primary hepatocellular carcinoma. J Gastrointest Oncol. 13:1308–1316. 2022. View Article : Google Scholar : PubMed/NCBI | |
Zhang HL, Yu LX, Yang W, Tang L, Lin Y, Wu H, Zhai B, Tan YX, Shan L, Liu Q, et al: Profound impact of gut homeostasis on chemically-induced pro-tumorigenic inflammation and hepatocarcinogenesis in rats. J Hepatol. 57:803–812. 2012. View Article : Google Scholar : PubMed/NCBI | |
Dapito DH, Mencin A, Gwak GY, Pradere JP, Jang MK, Mederacke I, Caviglia JM, Khiabanian H, Adeyemi A, Bataller R, et al: Promotion of hepatocellular carcinoma by the intestinal microbiota and TLR4. Cancer Cell. 21:504–516. 2012. View Article : Google Scholar : PubMed/NCBI | |
Mou WL, Chen SR, Wu ZT, Hu LH, Zhang JY, Chang HJ, Zhou H and Liu Y: LPS-TLR4/MD-2-TNF-α signaling mediates alcohol-induced liver fibrosis in rats. J Toxicol Pathol. 35:193–203. 2022. View Article : Google Scholar : PubMed/NCBI | |
Yu LX, Yan HX, Liu Q, Yang W, Wu HP, Dong W, Tang L, Lin Y, He YQ, Zou SS, et al: Endotoxin accumulation prevents carcinogen-induced apoptosis and promotes liver tumorigenesis in rodents. Hepatology. 52:1322–1333. 2010. View Article : Google Scholar : PubMed/NCBI | |
Yoshimoto S, Loo TM, Atarashi K, Kanda H, Sato S, Oyadomari S, Iwakura Y, Oshima K, Morita H, Hattori M, et al: Obesity-induced gut microbial metabolite promotes liver cancer through senescence secretome. Nature. 499:97–101. 2013. View Article : Google Scholar : PubMed/NCBI | |
Nguyen PT, Kanno K, Pham QT, Kikuchi Y, Kakimoto M, Kobayashi T, Otani Y, Kishikawa N, Miyauchi M, Arihiro K, et al: Senescent hepatic stellate cells caused by deoxycholic acid modulates malignant behavior of hepatocellular carcinoma. J Cancer Res Clin Oncol. 146:3255–3268. 2020. View Article : Google Scholar : PubMed/NCBI | |
Vaughn BP, Rank KM and Khoruts A: Fecal microbiota transplantation: Current status in treatment of gi and liver disease. Clin Gastroenterol Hepatol. 17:353–361. 2019. View Article : Google Scholar : PubMed/NCBI | |
Routy B, Lenehan JG, Miller WH Jr, Jamal R, Messaoudene M, Daisley BA, Hes C, Al KF, Martinez-Gili L, Punčochář M, et al: Fecal microbiota transplantation plus anti-PD-1 immunotherapy in advanced melanoma: A phase I trial. Nat Med. 29:2121–2132. 2023. View Article : Google Scholar : PubMed/NCBI | |
Belvoncikova P, Maronek M and Gardlik R: Gut dysbiosis and fecal microbiota transplantation in autoimmune diseases. Int J Mol Sci. 23:107292022. View Article : Google Scholar : PubMed/NCBI | |
Borody TJ, Eslick GD and Clancy RL: Fecal microbiota transplantation as a new therapy: From Clostridioides difficile infection to inflammatory bowel disease, irritable bowel syndrome, and colon cancer. Curr Opin Pharmacol. 49:43–51. 2019. View Article : Google Scholar : PubMed/NCBI | |
Burz SD, Monnoye M, Philippe C, Farin W, Ratziu V, Strozzi F, Paillarse JM, Chêne L, Blottière HM and Gérard P: Fecal microbiota transplant from human to mice gives insights into the role of the gut microbiota in non-alcoholic fatty liver disease (NAFLD). Microorganisms. 9:1992021. View Article : Google Scholar : PubMed/NCBI | |
Purohit A, Alam MJ, Kandiyal B, Shalimar Das B and Banerjee SK: Gut microbiome and non-alcoholic fatty liver disease. Prog Mol Biol Transl Sci. 191:187–206. 2022. View Article : Google Scholar : PubMed/NCBI | |
Brandt LJ and Aroniadis OC: An overview of fecal microbiota transplantation: Techniques, indications, and outcomes. Gastrointest Endosc. 78:240–249. 2013. View Article : Google Scholar : PubMed/NCBI | |
Persky SE and Brandt LJ: Treatment of recurrent Clostridium difficile-associated diarrhea by administration of donated stool directly through a colonoscope. Am J Gastroenterol. 95:3283–3285. 2000. View Article : Google Scholar : PubMed/NCBI | |
Michailidis L, Currier AC, Le M and Flomenhoft DR: Adverse events of fecal microbiota transplantation: A meta-analysis of high-quality studies. Ann Gastroenterol. 34:802–814. 2021.PubMed/NCBI | |
Allegretti JR, Kassam Z, Mullish BH, Chiang A, Carrellas M, Hurtado J, Marchesi JR, McDonald JAK, Pechlivanis A, Barker GF, et al: Effects of fecal microbiota transplantation with oral capsules in obese patients. Clin Gastroenterol Hepatol. 18:855–863.e2. 2020. View Article : Google Scholar : PubMed/NCBI | |
Yang J, Tang X, Liang Z, Chen M and Sun L: Taurocholic acid promotes hepatic stellate cell activation via S1PR2/p38 MAPK/YAP signaling under cholestatic conditions. Clin Mol Hepatol. 29:465–481. 2023. View Article : Google Scholar : PubMed/NCBI | |
Mancinelli R, Ceci L, Kennedy L, Francis H, Meadows V, Chen L, Carpino G, Kyritsi K, Wu N, Zhou T, et al: The effects of Taurocholic acid on biliary damage and liver fibrosis are mediated by calcitonin-gene-related peptide signaling. Cells. 11:15912022. View Article : Google Scholar : PubMed/NCBI | |
Uyttebroek S, Chen B, Onsea J, Ruythooren F, Debaveye Y, Devolder D, Spriet I, Depypere M, Wagemans J, Lavigne R, et al: Safety and efficacy of phage therapy in difficult-to-treat infections: A systematic review. Lancet Infect Dis. 22:e208–e220. 2022. View Article : Google Scholar : PubMed/NCBI | |
Federici S, Kredo-Russo S, Valdes-Mas R, Kviatcovsky D, Weinstock E, Matiuhin Y, Silberberg Y, Atarashi K, Furuichi M, Oka A, et al: Targeted suppression of human IBD-associated gut microbiota commensals by phage consortia for treatment of intestinal inflammation. Cell. 185:2879–2898.e24. 2022. View Article : Google Scholar : PubMed/NCBI | |
Duan Y, Young R and Schnabl B: Bacteriophages and their potential for treatment of gastrointestinal diseases. Nat Rev Gastroenterol Hepatol. 19:135–144. 2022. View Article : Google Scholar : PubMed/NCBI | |
Shuwen H and Kefeng D: Intestinal phages interact with bacteria and are involved in human diseases. Gut Microbes. 14:21137172022. View Article : Google Scholar : PubMed/NCBI | |
Duan Y, Llorente C, Lang S, Brandl K, Chu H, Jiang L, White RC, Clarke TH, Nguyen K, Torralba M, et al: Bacteriophage targeting of gut bacterium attenuates alcoholic liver disease. Nature. 575:505–511. 2019. View Article : Google Scholar : PubMed/NCBI | |
Fujiki J and Schnabl B: Phage therapy: Targeting intestinal bacterial microbiota for the treatment of liver diseases. JHEP Rep. 5:1009092023. View Article : Google Scholar : PubMed/NCBI | |
Gong X, Geng H, Yang Y, Zhang S, He Z, Fan Y, Yin F, Zhang Z and Chen GQ: Metabolic engineering of commensal bacteria for gut butyrate delivery and dissection of host-microbe interaction. Metab Eng. 80:94–106. 2023. View Article : Google Scholar : PubMed/NCBI | |
Anand AC and Acharya SK: The story of ammonia in liver disease: An unraveling continuum. J Clin Exp Hepatol. 14:1013612024. View Article : Google Scholar : PubMed/NCBI | |
Kurtz CB, Millet YA, Puurunen MK, Perreault M, Charbonneau MR, Isabella VM, Kotula JW, Antipov E, Dagon Y, Denney WS, et al: An engineered E. coli Nissle improves hyperammonemia and survival in mice and shows dose-dependent exposure in healthy humans. Sci Transl Med. 11:eaau79752019. View Article : Google Scholar : PubMed/NCBI | |
Yu M, Hu S, Tang B, Yang H and Sun D: Engineering Escherichia coli Nissle 1917 as a microbial chassis for therapeutic and industrial applications. Biotechnol Adv. 67:1082022023. View Article : Google Scholar : PubMed/NCBI | |
Lynch JP, Goers L and Lesser CF: Emerging strategies for engineering Escherichia coli Nissle 1917-based therapeutics. Trends Pharmacol Sci. 43:772–786. 2022. View Article : Google Scholar : PubMed/NCBI | |
Husted AS, Trauelsen M, Rudenko O, Hjorth SA and Schwartz TW: GPCR-mediated signaling of metabolites. Cell Metab. 25:777–796. 2017. View Article : Google Scholar : PubMed/NCBI | |
Akiba Y and Kaunitz JD: Duodenal luminal chemosensing; acid, ATP, and nutrients. Curr Pharm Des. 20:2760–2765. 2014. View Article : Google Scholar : PubMed/NCBI | |
Kokorovic A, Cheung GW, Breen DM, Chari M, Lam CK and Lam TK: Duodenal mucosal protein kinase C-δ regulates glucose production in rats. Gastroenterology. 141:1720–1727. 2011. View Article : Google Scholar : PubMed/NCBI | |
van Baar ACG, Beuers U, Wong K, Haidry R, Costamagna G, Hafedi A, Deviere J, Ghosh SS, Lopez-Talavera JC, Rodriguez L, et al: Endoscopic duodenal mucosal resurfacing improves glycaemic and hepatic indices in type 2 diabetes: 6-month multicentre results. JHEP Rep. 1:429–437. 2019. View Article : Google Scholar : PubMed/NCBI | |
de Oliveira GHP, de Moura DTH, Funari MP, McCarty TR, Ribeiro IB, Bernardo WM, Sagae VMT, Freitas JR Jr, Souza GMV and de Moura EGH: Metabolic effects of endoscopic duodenal mucosal resurfacing: A systematic review and Meta-analysis. Obes Surg. 31:1304–1312. 2021. View Article : Google Scholar : PubMed/NCBI | |
Shamseddeen H, Vuppalanchi R and Gromski MA: Duodenal mucosal resurfacing for nonalcoholic fatty liver disease. Clin Liver Dis (Hoboken). 20:166–169. 2022. View Article : Google Scholar : PubMed/NCBI | |
Condello G and Chen CY: Minireview: Current status of endoscopic duodenal mucosal resurfacing. Obes Res Clin Pract. 14:504–507. 2020. View Article : Google Scholar : PubMed/NCBI | |
Mingrone G, van Baar AC, Deviere J, Hopkins D, Moura E, Cercato C, Rajagopalan H, Lopez-Talavera JC, White K, Bhambhani V, et al: Safety and efficacy of hydrothermal duodenal mucosal resurfacing in patients with type 2 diabetes: The randomised, double-blind, sham-controlled, multicentre REVITA-2 feasibility trial. Gut. 71:254–264. 2022. View Article : Google Scholar : PubMed/NCBI | |
Hadefi A, Verset L, Pezzullo M, Rosewick N, Degre D, Gustot T, Moreno C, Devière J and Trépo E: Endoscopic duodenal mucosal resurfacing for nonalcoholic steatohepatitis (NASH): A pilot study. Endosc Int Open. 9:E1792–E1800. 2021. View Article : Google Scholar : PubMed/NCBI |