Food Bioscience, volume 62, pages 105379

Alleviation of Alcoholic Liver Injury through Composite Postbiotics Regulation of Intestinal Flora and Promotion of Bile Acid Metabolism

Qi Peng
Kaiming Yang
Qifan Hou
Huajun Zheng
Xiaomin Zheng
Guangfa Xie
Publication typeJournal Article
Publication date2024-12-01
Journal: Food Bioscience
scimago Q1
SJR0.959
CiteScore6.4
Impact factor4.8
ISSN22124292, 22124306
Wang B., Han D., Hu X., Chen J., Liu Y., Wu J.
Microbiological Research scimago Q1 wos Q1
2024-10-01 citations by CoLab: 6 Abstract  
The gut microbiota, mainly resides in the colon, possesses a remarkable ability to metabolize different substrates to create bioactive substances, including short-chain fatty acids, indole-3-propionic acid, and secondary bile acids. In the liver, bile acids are synthesized from cholesterol and then undergo modification by the gut microbiota. Beyond those reclaimed by the enterohepatic circulation, small percentage of bile acids escaped reabsorption, entering the systemic circulation to bind to several receptors, such as farnesoid X receptor (FXR), thereby exert their biological effects. Gut microbiota interplays with bile acids by affecting their synthesis and determining the production of secondary bile acids. Reciprocally, bile acids shape out the structure of gut microbiota. The interplay of bile acids and FXR is involved in the development of multisystemic conditions, encompassing metabolic diseases, hepatobiliary diseases, immune associated disorders. In the review, we aim to provide a thorough review of the intricate crosstalk between the gut microbiota and bile acids, the physiological roles of bile acids and FXR in mammals' health and disease, and the clinical translational considerations of gut microbiota-bile acids-FXR in the treatment of the diseases.
Fang H., Rodrigues e-Lacerda R., Barra N.G., Kukje Zada D., Robin N., Mehra A., Schertzer J.D.
Endocrine Reviews scimago Q1 wos Q1
2024-09-05 citations by CoLab: 3 Abstract  
Abstract The gut microbiota influences aspects of metabolic disease, including tissue inflammation, adiposity, blood glucose, insulin, and endocrine control of metabolism. Prebiotics or probiotics are often sought to combat metabolic disease. However, prebiotics lack specificity and can have deleterious bacterial community effects. Probiotics require live bacteria to find a colonization niche sufficient to influence host immunity or metabolism. Postbiotics encompass bacterial-derived components and molecules, which are well-positioned to alter host immunometabolism without relying on colonization efficiency or causing widespread effects on the existing microbiota. Here, we summarize the potential for beneficial and detrimental effects of specific postbiotics related to metabolic disease and the underlying mechanisms of action. Bacterial cell wall components, such as lipopolysaccharides, muropeptides, lipoteichoic acids and flagellin, have context-dependent effects on host metabolism by engaging specific immune responses. Specific types of postbiotics within broad classes of compounds, such as lipopolysaccharides and muropeptides, can have opposing effects on endocrine control of host metabolism, where certain postbiotics are insulin sensitizers and others promote insulin resistance. Bacterial metabolites, such as short-chain fatty acids, bile acids, lactate, glycerol, succinate, ethanolamine, and ethanol, can be substrates for host metabolism. Postbiotics can fuel host metabolic pathways directly or influence endocrine control of metabolism through immunomodulation or mimicking host-derived hormones. The interaction of postbiotics in the host-microbe relationship should be considered during metabolic inflammation and metabolic disease.
Song L., Wang D., Zhai Y., Zhang X., Zhang Y., Yu Y., Sun L., Zhou K.
Journal of Ethnopharmacology scimago Q1 wos Q1
2024-07-01 citations by CoLab: 4 Abstract  
Epimedium sagittatum (Sieb. et Zucc.) Maxim. has been used traditionally in Asia. It can dispel wind and cold, tonify the kidney, and strengthen bones and tendons. However, adverse effects of E. sagittatum have been reported, and the underlying mechanisms remain unclear. This study aimed to investigate liver injury caused by an aqueous extract of E. sagittatum in Institute of Cancer Research (ICR) mice and explore its potential mechanisms. Dried E. sagittatum leaves were decocted in water to prepare aqueous extracts for ultra-high performance liquid chromatography analysis. Mice were administered an aqueous extract of E. sagittatum equivalent to either 3 g raw E. sagittatum/kg or 10 g raw E. sagittatum/kg once daily via intragastric injection for three months. The liver weights and levels of the serum biochemical parameters including alanine transaminase (ALT), aspartate aminotransferase (AST), lactate dehydrogenase (LDH), total bilirubin (TBIL), and alkaline phosphatase were measured. Hematoxylin-eosin staining was performed for histopathology. Apoptosis was detected using the TUNEL apoptosis assay kit. IL-1β was detected using ELISA kits. Proteomics was used to identify the differentially expressed proteins. Western blot analysis was performed to determine the levels of proteins significantly affected by the aqueous extract of E. sagittatum. E. sagittatum treatment increased the liver weights and liver coefficients, and ALT and AST levels significantly increased (p < 0.05). A high dose of E. sagittatum significantly increased LDH and TBIL levels (p < 0.05). Ruptured cell membranes and multiple sites of inflammatory cell infiltration were also observed. No evidence of apoptosis was observed. IL-1β levels were significantly increased (p < 0.05). The expressions of PIK3R1, p-MAP2K4, p-Jun N-terminal kinase (JNK)/JNK, p-c-Jun, VDAC2, Bax, and CYC were upregulated, whereas that of Bcl-2 was inhibited by E. sagittatum. The expression of cleaved caspase-1 was significantly increased; however, its effects on GSDMD and GSDMD-N were significantly decreased. The expression levels of cleaved caspase-3 and its effector proteins GSDME and GSDME-N significantly increased. Our results suggest that the aqueous extract of E. sagittatum induces liver injury in ICR mice after three months of intragastric injection via inflammatory pyroptosis.
Tao W., Fan Q., Wei J.
2024-02-01 citations by CoLab: 3 PDF Abstract  
Drug-induced liver injury (DILI) is a liver disease that remains difficult to predict and diagnose, and the underlying mechanisms are yet to be fully clarified. The gut–liver axis refers to the reciprocal interactions between the gut and the liver, and its homeostasis plays a prominent role in maintaining liver health. It has been recently reported that patients and animals with DILI have a disrupted gut–liver axis, involving altered gut microbiota composition, increased intestinal permeability and lipopolysaccharide translocation, decreased short-chain fatty acids production, and impaired bile acid metabolism homeostasis. The present review will summarize the evidence from both clinical and preclinical studies about the role of the gut–liver axis in the pathogenesis of DILI. Moreover, we will focus attention on the potential therapeutic strategies for DILI based on improving gut–liver axis function, including herbs and phytochemicals, probiotics, fecal microbial transplantation, postbiotics, bile acids, and Farnesoid X receptor agonists.
Kalambokis G.N., Chouliara N., Tsiakas I., Filippas-Ntekuan S., Christaki M., Despotis G., Milionis H.
2024-01-01 citations by CoLab: 4 Abstract  
Background and aim The prevalence of alcohol-associated cirrhosis is increasing. In this respect, we investigated the long-term impact of non-abstinence on the clinical course of alcohol-associated cirrhosis. Methods We retrospectively evaluated 440 patients with alcohol-associated cirrhosis (compensated cirrhosis: n = 190; decompensated cirrhosis: n = 250) diagnosed between January 2000 and July 2017 who consumed alcohol until diagnosis of cirrhosis. We assessed liver-related outcomes including first and further decompensating events (ascites, variceal bleeding, and hepatic encephalopathy), and death in relation to continued alcohol use. Results Overall, 53.6% of patients remained abstinent (compensated cirrhosis: 57.9%; decompensated cirrhosis: 50.4%). Non-abstinent versus abstinent patients with compensated cirrhosis and decompensated cirrhosis showed significantly higher 5-year probability of first decompensation (80.2% vs. 36.8%; P < 0.001) and further decompensation (87.9% vs. 20.6%; P < 0.001), respectively. Five-year survival was substantially lower among non-abstinent patients with compensated cirrhosis (45.9% vs. 90.7%; P < 0.001) and decompensated cirrhosis (22.9% vs. 73.8%; P < 0.001) compared to abstinent. Non-abstinent versus abstinent patients of the total cohort showed an exceedingly lower 5-year survival (32.2% vs. 82.4%; P < 0.001). Prolonged abstinence (≥2 years) was required to influence outcomes. Non-abstinence independently predicted mortality in the total cohort (hazard ratio [HR] 3.371; confidence interval [CI]: 2.388–4.882; P < 0.001) along with the Child-Pugh class (HR: 4.453; CI: 2.907–6.823; P < 0.001) and higher age (HR: 1.023; CI: 1.007–1.039; P = 0.005). Conclusion Liver-related outcomes are worse among non-abstinent patients with alcohol- associated cirrhosis prompting urgent interventions ensuring abstinence.
Li F., Zhang Z., Bai Y., Che Q., Cao H., Guo J., Su Z.
Molecules scimago Q1 wos Q2 Open Access
2023-10-03 citations by CoLab: 10 PDF Abstract  
Non-alcoholic fatty liver disease (NAFLD) is a liver disease syndrome. The prevalence of NAFLD has continued to increase globally, and NAFLD has become a worldwide public health problem. Glucosamine (GLC) is an amino monosaccharide derivative of glucose. GLC has been proven to not only be effective in anti-inflammation applications, but also to modulate the gut microbiota effectively. Therefore, in this study, the therapeutic effect of GLC in the NAFLD context and the mechanisms underlying these effects were explored. Specifically, an NAFLD model was established by feeding mice a high-fat and high-sugar diet (HFHSD), and the HFHSD-fed NAFLD mice were treated with GLC. First, we investigated the effect of treating NAFLD mice with GLC by analyzing serum- and liver-related indicator levels. We found that GLC attenuated insulin resistance and inflammation, increased antioxidant function, and attenuated serum and liver lipid metabolism in the mice. Then, we investigated the mechanism underlying liver lipid metabolism, inflammation, and intestinal barrier function in these mice. We found that GLC can improve liver lipid metabolism and relieve insulin resistance and oxidative stress levels. In addition, GLC treatment increased intestinal barrier function, reduced LPS translocation, and reduced liver inflammation by inhibiting the activation of the LPS/TLR4/NF-κB pathway, thereby effectively ameliorating liver lesions in NAFLD mice.
Cheng L., Shi J., Peng H., Tong R., Hu Y., Yu D.
Journal of Functional Foods scimago Q1 wos Q2 Open Access
2023-10-01 citations by CoLab: 3 Abstract  
Liver fibrosis is one of the most common chronic liver diseases in the world, which can gradually develop into cirrhosis or even liver cancer, posing a serious threat to human health. The causes of liver fibrosis are multiple, including cholestasis, inflammation, oxidative stress, and other factors that may disrupt hepatic homeostasis and induce liver fibrosis. Until now, the primary treatment for liver fibrosis has been to inhibit the activation of hepatic stellate cells and suppress extracellular matrix production. The enterohepatic circulation establishes a strong bond between the liver and the intestine. Alterations of the intestinal flora may cause intestinal disorders and damage the liver through the gut-liver axis. Recent studies have shown that the regulation of the gut microbiota and the gut-liver axis play an important role in the treatment of liver fibrosis. Studies have shown that probiotics can improve the pathological condition of liver inflammation and oxidative stress and stop the progression of liver fibrosis by modulating the intestinal microbiome. This review summarizes the effects of probiotics in the subsequent treatment of liver fibrosis by altering bile acid metabolism, preventing oxidative stress and inflammation during liver fibrosis development, and providing new options for the adjuvant treatment of liver fibrosis.
Brahadeeswaran S., Dasgupta T., Manickam V., Saraswathi V., Tamizhselvi R.
Frontiers in Immunology scimago Q1 wos Q1 Open Access
2023-07-13 citations by CoLab: 22 PDF Abstract  
The liver is in charge of a wide range of critical physiological processes and it plays an important role in activating the innate immune system which elicits the inflammatory events. Chronic ethanol exposure disrupts hepatic inflammatory mechanism and leads to the release of proinflammatory mediators such as chemokines, cytokines and activation of inflammasomes. The mechanism of liver fibrosis/cirrhosis involve activation of NLRP3 inflammasome, leading to the destruction of hepatocytes and subsequent metabolic dysregulation in humans. In addition, increasing evidence suggests that alcohol intake significantly modifies liver epigenetics, promoting the development of alcoholic liver disease (ALD). Epigenetic changes including histone modification, microRNA-induced genetic modulation, and DNA methylation are crucial in alcohol-evoked cell signaling that affects gene expression in the hepatic system. Though we are at the beginning stage without having the entire print of epigenetic signature, it is time to focus more on NLRP3 inflammasome and epigenetic modifications. Here we review the novel aspect of ALD pathology linking to inflammation and highlighting the role of epigenetic modification associated with NLRP3 inflammasome and how it could be a therapeutic target in ALD.
Mishra G., Singh P., Molla M., Yimer Y.S., Dinda S.C., Chandra P., Singh B.K., Dagnew S.B., Assefa A.N., Ewunetie A.
Frontiers in Pharmacology scimago Q1 wos Q1 Open Access
2023-06-09 citations by CoLab: 15 PDF Abstract  
In the current scenario, prolonged consumption of alcohol across the globe is upsurging an appreciable number of patients with the risk of alcohol-associated liver diseases. According to the recent report, the gut-liver axis is crucial in the progression of alcohol-induced liver diseases, including steatosis, steatohepatitis, fibrosis, cirrhosis, and hepatocellular carcinoma. Despite several factors associated with alcoholic liver diseases, the complexity of the gut microflora and its great interaction with the liver have become a fascinating area for researchers due to the high exposure of the liver to free radicals, bacterial endotoxins, lipopolysaccharides, inflammatory markers, etc. Undoubtedly, alcohol-induced gut microbiota imbalance stimulates dysbiosis, disrupts the intestinal barrier function, and trigger immune as well as inflammatory responses which further aggravate hepatic injury. Since currently available drugs to mitigate liver disorders have significant side effects, hence, probiotics have been widely researched to alleviate alcohol-associated liver diseases and to improve liver health. A broad range of probiotic bacteria like Lactobacillus, Bifidobacteria, Escherichia coli, Sacchromyces, and Lactococcus are used to reduce or halt the progression of alcohol-associated liver diseases. Several underlying mechanisms, including alteration of the gut microbiome, modulation of intestinal barrier function and immune response, reduction in the level of endotoxins, and bacterial translocation, have been implicated through which probiotics can effectively suppress the occurrence of alcohol-induced liver disorders. This review addresses the therapeutic applications of probiotics in the treatment of alcohol-associated liver diseases. Novel insights into the mechanisms by which probiotics prevent alcohol-associated liver diseases have also been elaborated.
Zou L., Tian Y., Wang Y., Chen D., Lu X., Zeng Z., Chen Z., Lin C., Liang Y.
Journal of Affective Disorders scimago Q1 wos Q1
2023-04-01 citations by CoLab: 41 Abstract  
Neuropsychiatric disorders, including anxiety and depression, are one of the most common mental illnesses worldwide. A growing body of evidence shows that there is a complex relationship between dietary patterns and mental health. In our study, C57BL/6J mice were divided into three groups: control diet group (CON, 10 % kcal fat), high-cholesterol diet model group (HCD, 42.0 % kcal fat + 1.25 % kcal Cholesterol), and chronic restraint stress group (CRS, 10 % kcal fat) which as a positive control group for the depression model. Six weeks later, depressive- and anxiety-like behavior were evaluated for using the OFT, SPT and TST. Glucose intolerance and liver fat were detected by IGTT and liver lipid kit. The expression of peripheral and central inflammation was detected by LEGEND plex kits. 5-HT (also named 5-hydroxytryptamine, 5-HT) and related receptors expression were monitored by ELISA, RT-PCR and Western blot. Meantime, gut microbe of stool samples was performed by 16S rRNA gene sequencing. Similar to CRS model, short-term HCD intervention induced anxiety and depression-like behavior behavioral abnormalities in mice. HCD consumption resulted in significantly increased body weight, liver fat (LDL-C, TC, TG), peripheral inflammation (IL-1β, MCP-1, IL-17A) and neuroinflammation (MCP-1). The concentration of 5-HT increased in the hippocampus, meanwhile, the expression of 5-HT receptor HTR2A was distinct in different regions of the brain tissue. More importantly, we found that compared with the CON diet, HCD induced the decrease of intestinal flora diversity, especially the decrease the relative abundance of Akkermansia_muciniphila, which was statistically significant. Further, Pearson correlation analysis showed that Akkermansia_muciniphila was significantly negatively correlated with the concentration of MCP-1, IL-17A in serum and 5-HT in hippocampus. Therefore, we speculated that the disorder of neuroinflammation induced by HCD consumption promotes depression- and anxiety-like behaviors in mice through the gut microbe.
Liang B., Xing D.
2023-02-10 citations by CoLab: 67 Abstract  
With the emphasis on intestinal health, probiotics have exploded into a vast market potential. However, new scientific evidence points out that the beneficial health benefits of probiotics are not necessarily directly related to viable bacteria. However, the metabolites or bacterial components of the live bacteria are the driving force behind health promotion. Therefore, scientists gradually noticed that the beneficial effects of probiotics are based on bacteria itself, metabolites, or cell lysates, and these factors are officially named “postbiotics” by the ISAPP. Postbiotic components are diverse and outperform live probiotics in terms of technology, safety, and cost due to their good absorption, metabolism, and organismal distribution. Postbiotics have been shown to have bioactivities such as antimicrobial, antioxidant, anti-inflammatory, anti-proliferative, and immunomodulation. Moreover, numerous studies have revealed the significant potential of postbiotics for disease treatment. This paper first presents the production and classification of postbiotics with examples from lactic acid bacteria (LAB), followed by the mechanisms of action with the most recent pre-clinical and clinical studies and the wide range of non-clinical and clinical applications of postbiotics. Furthermore, the current and future prospects of the postbiotic market with commercial available products are discussed. Finally, we comment on the knowledge gaps and future clinical applications with several examples.
Marchianò S., Biagioli M., Morretta E., Di Giorgio C., Roselli R., Bordoni M., Bellini R., Urbani G., Massa C., Monti M.C., Zampella A., Distrutti E., Fiorucci S.
Scientific Reports scimago Q1 wos Q1 Open Access
2023-01-28 citations by CoLab: 20 PDF Abstract  
AbstractNon-alcoholic steatosis (NAFLD) and steatohepatitis (NASH) are two highly prevalent human disorders for which therapy remains suboptimal. Bile acids are signaling molecules acting on two main receptors the Farnesoid-x-receptor (FXR) and G protein coupled receptor GPB AR1. Clinical trials have shown that FXR agonism might result in side effects along with lack of efficacy in restoring liver histopathology. For these reasons a multi-targets therapy combined FXR agonists with agent targeting additional molecular mechanisms might have improved efficacy over selective FXR agonists. In the present study we have compared the effects of BAR502, a dual FXR/GPBAR1 ligand) alone or in combination with ursodeoxycholic acid (UDCA) in a model of NAFLD/NASH induced by feeding mice with a Western diet for 10 weeks. The results demonstrated that while BAR502 and UDCA partially protected against liver damage caused by Western diet, the combination of the two, reversed the pro-atherogenic lipid profile and completely reversed the histopathology damage, attenuating liver steatosis, ballooning, inflammation and fibrosis. Additionally, while both agents increased insulin sensitivity and bile acid signaling, the combination of the two, modulated up top 85 genes in comparison of mice feed a Western diet, strongly reducing expression of inflammatory markers such as chemokines and cytokines. Additionally, the combination of the two agents redirected the bile acid metabolism toward bile acid species that are GPBAR1 agonist while reduced liver bile acid content and increased fecal excretion. Together, these data, highlight the potential role for a combinatorial therapy based on BAR502 and UDCA in treating of NAFLD.

Top-30

Journals

1
1

Publishers

1
1
  • We do not take into account publications without a DOI.
  • Statistics recalculated only for publications connected to researchers, organizations and labs registered on the platform.
  • Statistics recalculated weekly.

Are you a researcher?

Create a profile to get free access to personal recommendations for colleagues and new articles.
Share
Cite this
GOST | RIS | BibTex
Found error?