Location via proxy:   [ UP ]  
[Report a bug]   [Manage cookies]                
 
 
Sign in to use this feature.

Years

Between: -

Subjects

remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline

Journals

Article Types

Countries / Regions

Search Results (312)

Search Parameters:
Keywords = gut–liver axis

Order results
Result details
Results per page
Select all
Export citation of selected articles as:
16 pages, 2088 KiB  
Article
Mesenchymal Stem Cell-Derived Exosomes Attenuate Hepatic Steatosis and Insulin Resistance in Diet-Induced Obese Mice by Activating the FGF21-Adiponectin Axis
by Bobae Kim, Rwubuzizi Ronaldo, Beet-Na Kweon, Solhee Yoon, Yein Park, Jea-Hyun Baek, Jung Min Lee and Chang-Kee Hyun
Int. J. Mol. Sci. 2024, 25(19), 10447; https://doi.org/10.3390/ijms251910447 - 27 Sep 2024
Viewed by 365
Abstract
Exosomes derived from mesenchymal stem cells have shown promise in treating metabolic disorders, yet their specific mechanisms remain largely unclear. This study investigates the protective effects of exosomes from human umbilical cord Wharton’s jelly mesenchymal stem cells (hWJMSCs) against adiposity and insulin resistance [...] Read more.
Exosomes derived from mesenchymal stem cells have shown promise in treating metabolic disorders, yet their specific mechanisms remain largely unclear. This study investigates the protective effects of exosomes from human umbilical cord Wharton’s jelly mesenchymal stem cells (hWJMSCs) against adiposity and insulin resistance in high-fat diet (HFD)-induced obese mice. HFD-fed mice treated with hWJMSC-derived exosomes demonstrated improved gut barrier integrity, which restored immune balance in the liver and adipose tissues by reducing macrophage infiltration and pro-inflammatory cytokine expression. Furthermore, these exosomes normalized lipid metabolism including lipid oxidation and lipogenesis, which alleviate lipotoxicity-induced endoplasmic reticulum (ER) stress, thereby decreasing fat accumulation and chronic tissue inflammation in hepatic and adipose tissues. Notably, hWJMSC-derived exosomes also promoted browning and thermogenic capacity of adipose tissues, which was linked to reduced fibroblast growth factor 21 (FGF21) resistance and increased adiponectin production. This process activated the AMPK-SIRT1-PGC-1α pathway, highlighting the role of the FGF21–adiponectin axis. Our findings elucidate the molecular mechanisms through which hWJMSC-derived exosomes counteract HFD-induced metabolic dysfunctions, supporting their potential as therapeutic agents for metabolic disorders. Full article
Show Figures

Figure 1

16 pages, 15464 KiB  
Article
Sinisan Alleviates Stress-Induced Intestinal Dysfunction and Depressive-like Behaviors in Mice with Irritable Bowel Syndrome by Enhancing the Intestinal Barrier and Modulating Central 5-Hydroxytryptamine
by Haizhou Zeng, Yupeng Jiang, Qiuxiong Yin, Xinran Li, Yanli Xiong, Boyi Li, Xiaoying Xu, Huimei Hu and Guoqiang Qian
Int. J. Mol. Sci. 2024, 25(19), 10262; https://doi.org/10.3390/ijms251910262 - 24 Sep 2024
Viewed by 432
Abstract
Irritable bowel syndrome (IBS) is a common chronic functional bowel disorder and is strongly associated with an increased risk of depression and anxiety. The brain–gut axis plays an important role in the pathophysiologic changes in IBS, yet effective treatments for IBS are still [...] Read more.
Irritable bowel syndrome (IBS) is a common chronic functional bowel disorder and is strongly associated with an increased risk of depression and anxiety. The brain–gut axis plays an important role in the pathophysiologic changes in IBS, yet effective treatments for IBS are still lacking. Sinisan, originating from the Treatise on Typhoid Fever by the medical sage Zhang Zhongjing, is a classic formula in the Eight Methods of Traditional Chinese Medicine (TCM) that focuses on dispersing the liver and regulating the spleen, relieving depression and transmitting evils, and has been widely used in the treatment of liver-depression and spleen-deficiency, diarrhea, and related liver and stomach disorders. However, the therapeutic effect of sinisan in IBS has not been clarified. The aim of this study was to investigate the effects of sinisan on stress-induced intestinal dysfunction and depressive behavior in IBS mice. We established a diarrhea-predominant irritable bowel syndrome (IBS-D) mouse model using a 4% acetic acid enema combined with restraint stress, and analyzed the results using behavioral tests, relevant test kits, hematoxylin-eosin (HE) staining, immunofluorescence (IF), Western blot (WB), and quantitative fluorescence polymerase chain reaction (qRT-PCR). The results showed that sinisan administration significantly alleviated intestinal dysfunction and depressive-like behaviors in IBS-D mice, improved mild colonic inflammation and intestinal mucosal permeability, up-regulated the expression of tight junction proteins ZO-1 and occludin. Sinisan significantly alleviated intestinal dysfunction and depressive-like behaviors in IBS-D mice by decreasing the expression of TNF-α, promoting the expression of tight junction proteins (occludin, ZO-1) expression, and inhibiting the Tlr4/Myd88 signaling pathway, thereby attenuating the inflammatory response, protecting the intestinal barrier, and alleviating symptoms in the IBS-D mouse model. Taken together, sinisan may ameliorate intestinal inflammation and the intestinal barrier by regulating 5-HT expression and the Tlr4/Myd88 pathway, thereby alleviating stress-induced intestinal dysfunction and depressive behaviors in IBS-D mice. Full article
Show Figures

Graphical abstract

17 pages, 12342 KiB  
Article
Ameliorative Effects of Camel Milk and Fermented Camel Milk on Acute Alcoholic Liver Injury
by Chunxia Zhu, Wancheng Sun and Yihao Luo
Fermentation 2024, 10(10), 493; https://doi.org/10.3390/fermentation10100493 - 24 Sep 2024
Viewed by 543
Abstract
Probiotics, which are prevalent in camel milk (CM) and naturally fermented camel milk (FCM), can regulate the intestinal ecological structure to alleviate alcoholic liver disease (ALD) through the “gut–liver” axis. The protective effects and mechanisms of CM and FCM interventions on alcohol-induced acute [...] Read more.
Probiotics, which are prevalent in camel milk (CM) and naturally fermented camel milk (FCM), can regulate the intestinal ecological structure to alleviate alcoholic liver disease (ALD) through the “gut–liver” axis. The protective effects and mechanisms of CM and FCM interventions on alcohol-induced acute liver injury were investigated by combining the behavior observed in rats following alcohol exposure. The results revealed that CM and FCM effectively controlled the increased levels of alcohol-induced ALT, AST, TG, MDA, and proinflammatory cytokines. Alcohol-induced oxidative depletion of hepatic CAT, GPX, GSH, and ALDH was reversed, diminishing lipid accumulation, ameliorating severe pathological damage, increasing antioxidant capabilities, and postponing oxidative stress. Additionally, the abundance of the phylum Bacteroidota (which reduces the F/B ratio); the family Prevotellaceae; the genera Clostridia_vadinBB60_group, parabacteroides, Alloprevotella, and Prevotellaceae_UC_G001; the gastrointestinal barrier; and the microbiological environment was increased. The steroid hormone biosynthesis pathway was altered to reduce alcohol-induced predominant steroid metabolites such as 17-hydroxyprogesterone, cortisol, and dehydroepiandrosterone, preventing alcoholic liver impairment. Taken together, CM could be a therapeutic dietary supplement for preventing alcoholic liver injury by ameliorating the intestinal ecology and hepatic metabolism. Full article
(This article belongs to the Section Probiotic Strains and Fermentation)
Show Figures

Figure 1

22 pages, 1486 KiB  
Review
Probiotics and Non-Alcoholic Fatty Liver Disease: Unveiling the Mechanisms of Lactobacillus plantarum and Bifidobacterium bifidum in Modulating Lipid Metabolism, Inflammation, and Intestinal Barrier Integrity
by Jing Lu, Dilireba Shataer, Huizhen Yan, Xiaoxiao Dong, Minwei Zhang, Yanan Qin, Jie Cui and Liang Wang
Foods 2024, 13(18), 2992; https://doi.org/10.3390/foods13182992 - 21 Sep 2024
Viewed by 713
Abstract
In recent years, the prevalence of non-alcoholic fatty liver disease (NAFLD) has risen annually, yet due to the intricacies of its pathogenesis and therapeutic challenges, there remains no definitive medication for this condition. This review explores the intricate relationship between the intestinal microbiome [...] Read more.
In recent years, the prevalence of non-alcoholic fatty liver disease (NAFLD) has risen annually, yet due to the intricacies of its pathogenesis and therapeutic challenges, there remains no definitive medication for this condition. This review explores the intricate relationship between the intestinal microbiome and the pathogenesis of NAFLD, emphasizing the substantial roles played by Lactobacillus plantarum and Bifidobacterium bifidum. These probiotics manipulate lipid synthesis genes and phosphorylated proteins through pathways such as the AMPK/Nrf2, LPS-TLR4-NF-κB, AMPKα/PGC-1α, SREBP-1/FAS, and SREBP-1/ACC signaling pathways to reduce hepatic lipid accumulation and oxidative stress, key components of NAFLD progression. By modifying the intestinal microbial composition and abundance, they combat the overgrowth of harmful bacteria, alleviating the inflammatory response precipitated by dysbiosis and bolstering the intestinal mucosal barrier. Furthermore, they participate in cellular immune regulation, including CD4+ T cells and Treg cells, to suppress systemic inflammation. L. plantarum and B. bifidum also modulate lipid metabolism and immune reactions by adjusting gut metabolites, including propionic and butyric acids, which inhibit liver inflammation and fat deposition. The capacity of probiotics to modulate lipid metabolism, immune responses, and gut microbiota presents an innovative therapeutic strategy. With a global increase in NAFLD prevalence, these insights propose a promising natural method to decelerate disease progression, avert liver damage, and tackle associated metabolic issues, significantly advancing microbiome-focused treatments for NAFLD. Full article
(This article belongs to the Section Food Nutrition)
Show Figures

Figure 1

20 pages, 2164 KiB  
Review
Deciphering the Gut–Liver Axis: A Comprehensive Scientific Review of Non-Alcoholic Fatty Liver Disease
by Samradhi Singh, Mona Kriti, Roberto Catanzaro, Francesco Marotta, Mustafa Malvi, Ajay Jain, Vinod Verma, Ravinder Nagpal, Rajnarayan Tiwari and Manoj Kumar
Livers 2024, 4(3), 435-454; https://doi.org/10.3390/livers4030032 - 12 Sep 2024
Viewed by 657
Abstract
Non-alcoholic fatty liver disease (NAFLD) has emerged as a significant global health issue. The condition is closely linked to metabolic dysfunctions such as obesity and type 2 diabetes. The gut–liver axis, a bidirectional communication pathway between the liver and the gut, plays a [...] Read more.
Non-alcoholic fatty liver disease (NAFLD) has emerged as a significant global health issue. The condition is closely linked to metabolic dysfunctions such as obesity and type 2 diabetes. The gut–liver axis, a bidirectional communication pathway between the liver and the gut, plays a crucial role in the pathogenesis of NAFLD. This review delves into the mechanisms underlying the gut–liver axis, exploring the influence of gut microbiota, intestinal permeability, and inflammatory pathways. This review also explores the potential therapeutic strategies centered on modulating gut microbiota such as fecal microbiota transplantation; phage therapy; and the use of specific probiotics, prebiotics, and postbiotics in managing NAFLD. By understanding these interactions, we can better comprehend the development and advancement of NAFLD and identify potential therapeutic targets. Full article
Show Figures

Figure 1

14 pages, 2783 KiB  
Article
The Effects of Rice Bran on Neuroinflammation and Gut Microbiota in Ovariectomized Mice Fed a Drink with Fructose
by Yu-Wen Chao, Yu-Tang Tung, Suh-Ching Yang, Hitoshi Shirakawa, Li-Han Su, Pei-Yu Loe and Wan-Chun Chiu
Nutrients 2024, 16(17), 2980; https://doi.org/10.3390/nu16172980 - 4 Sep 2024
Viewed by 1068
Abstract
Rice bran, which is abundant in dietary fiber and phytochemicals, provides multiple health benefits. Nonetheless, its effects on neuroinflammation and gut microbiota in postmenopausal conditions are still not well understood. This study investigated the effects of rice bran and/or tea seed oil supplementation [...] Read more.
Rice bran, which is abundant in dietary fiber and phytochemicals, provides multiple health benefits. Nonetheless, its effects on neuroinflammation and gut microbiota in postmenopausal conditions are still not well understood. This study investigated the effects of rice bran and/or tea seed oil supplementation in d-galactose-injected ovariectomized (OVX) old mice fed a fructose drink. The combination of d-galactose injection, ovariectomy, and fructose drink administration creates a comprehensive model that simulates aging in females under multiple metabolic stressors, including oxidative stress, estrogen deficiency, and high-sugar diets, and allows the study of their combined impact on metabolic disorders and related diseases. Eight-week-old and 6–8-month-old female C57BL/6 mice were used. The mice were divided into six groups: a sham + young mice, a sham + old mice, an OVX + soybean oil, an OVX + soybean oil with rice bran, an OVX + tea seed oil (TO), and an OVX + TO with rice bran diet group. The OVX groups were subcutaneously injected with d-galactose (100 mg/kg/day) and received a 15% (v/v) fructose drink. The rice bran and tea seed oil supplementation formed 10% of the diet (w/w). The results showed that the rice bran with TO diet increased the number of short-chain fatty acid (SCFA)-producing Clostridia and reduced the number of endotoxin-producing Tannerellaceae, which mitigated imbalances in the gut–liver–brain axis. Rice bran supplementation reduced the relative weight of the liver, levels of hepatic triglycerides and total cholesterol; aspartate transaminase and alanine aminotransferase activity; brain levels of proinflammatory cytokines, including interleukin-1β and tumor necrosis factor-α; and plasma 8-hydroxy-2-deoxyguanosine. This study concludes that rice bran inhibits hepatic fat accumulation, which mitigates peripheral metaflammation and oxidative damage and reduces neuroinflammation in the brain. Full article
(This article belongs to the Special Issue Dietary Fiber, Gut Microbiota and Metabolic Disorder)
Show Figures

Figure 1

18 pages, 9761 KiB  
Article
Phenylethanol Glycoside from Cistanche tubulosa Attenuates BSA-Induced Liver Fibrosis in Rats by Modulating the Gut Microbiota–Liver Axis
by Xinxin Qi, Hongguang Sun, Jincun Liu, Meili Cong, Xinxuan Zhang, Yuxin Yan, Zhaolin Xia, Tao Liu and Jun Zhao
Pharmaceuticals 2024, 17(9), 1149; https://doi.org/10.3390/ph17091149 - 30 Aug 2024
Viewed by 653
Abstract
This study aimed to investigate the effect of phenylethanol glycoside from Cistanche tubulosa (CPhGs) on the prevention of bovine serum albumin (BSA)-induced hepatic fibrosis in rats. Investigation of the mechanisms of the anti-hepatic fibrosis effect was focused on CPhGs’ influence on the “gut–liver” [...] Read more.
This study aimed to investigate the effect of phenylethanol glycoside from Cistanche tubulosa (CPhGs) on the prevention of bovine serum albumin (BSA)-induced hepatic fibrosis in rats. Investigation of the mechanisms of the anti-hepatic fibrosis effect was focused on CPhGs’ influence on the “gut–liver” regulation, including the gut microbiota, intestinal barrier, systemic lipopolysaccharide (LPS) concentration, and LPS-related signaling pathway. The results show that CPhGs restored the diversity of gut microbiota, increased the relative abundance of Bacteroidetes, and decreased the relative abundance of Firmicutes and Proteobacteria in the fibrotic rats. In addition, CPhGs promoted the enrichment of probiotics such as Blautia, Oscillospira, Ruminococcus, Odoribacter, Bacteroides, and Parabacteroides in intestines of these rats. Furthermore, CPhGs reduced histopathological injury in the intestine and restored the tight junctions of the intestine by increasing the expression of ZO-1, occludin, and E-cadherin. CPhGs efficiently reduced serum LPS and liver lipopolysaccharide-binding protein (LBP) levels and inhibited the LPS-TLR4/MyD88/NF-κB pathway, which is related to protein expression in the liver. Correlation analysis confirmed that these beneficial bacteria were negatively associated with pathological damage, while LPS and harmful bacteria were positively associated with liver injury. Our fecal microbiota transplantation (FMT) experiment confirmed that gut microbiota is an important part of disease progression and that CPhGs is useful for the prevention and treatment of hepatic fibrosis. Our data demonstrate that the anti-hepatic fibrosis mechanism of CPhGs was mediated by regulation of the “gut–liver” axis. These results can stimulate consideration for its use in clinical practices. Full article
Show Figures

Figure 1

11 pages, 920 KiB  
Systematic Review
Effect of Saccharomyces boulardii on Liver Diseases: A Systematic Review
by Roman Maslennikov, Nona Benuni, Anna Levshina, Farida Adzhieva, Tatyana Demina, Alina Kucher, Ekaterina Pervushova, Evgeniya Yuryeva, Elena Poluektova, Oxana Zolnikova, Evgenii Kozlov, Alexey Sigidaev and Vladimir Ivashkin
Microorganisms 2024, 12(8), 1678; https://doi.org/10.3390/microorganisms12081678 - 15 Aug 2024
Viewed by 979
Abstract
We aimed to systematize the results of published studies on the use of Saccharomyces boulardii (SB) for the treatment of various liver disorders (CRD42022378050). Searches were conducted using PubMed and Scopus on 1 August 2022. The PubMed search was updated on 15 June [...] Read more.
We aimed to systematize the results of published studies on the use of Saccharomyces boulardii (SB) for the treatment of various liver disorders (CRD42022378050). Searches were conducted using PubMed and Scopus on 1 August 2022. The PubMed search was updated on 15 June 2024. The review included sixteen studies: ten experimental animal studies (EASs) and six randomized controlled trials (RCTs). The CNCM I-745 strain was used in 68.8% of the included studies. SB reduced the severity of many manifestations of cirrhosis, and lowered the Child–Pugh scores in RCT. SB reduced the serum concentrations of TNF-α, IL-1β, IL-6, and IL-4 in animals with metabolic dysfunction-associated steatotic liver disease (MASLD); lowered the serum TNF-α and IL-6 levels in experimental cirrhosis in rats; and reduced the CRP levels in decompensated cirrhosis. The EAS of MASLD revealed that SB reduced liver steatosis and inflammation and lowered the liver expression of genes of TNF-α, IL-1β, interferon-γ, and IL-10. In studies on experimental cirrhosis and MASLD, SB reduced the liver expression of genes of TGF-β, α-SMA, and collagen as well as liver fibrosis. SB reduced the abundance of Escherichia (Proteobacteria), increased the abundance of Bacteroidetes in the gut microbiota, prevented an increase in intestinal barrier permeability, and reduced bacterial translocation and endotoxemia. Full article
(This article belongs to the Special Issue Probiotics, Prebiotics, and Gut Microbes)
Show Figures

Figure 1

17 pages, 3701 KiB  
Review
Unearthing the Potential Therapeutic Effects of Oxyresveratrol Based on Intrinsic Links between Pharmacological Effects: Implications for the Gut–Liver–Brain Axis
by Lijuan Zhao, Yan Duan, Zhaoxing Li, Juan Li and Shunxiang Li
Pharmaceuticals 2024, 17(8), 1063; https://doi.org/10.3390/ph17081063 - 13 Aug 2024
Viewed by 733
Abstract
Oxyresveratrol is a stilbene compound with a simple chemical structure and various therapeutic potentials. This study summarized and analyzed the multiple pharmacological effects and mechanisms of oxyresveratrol, identifying its prominent performance in neuroprotection, hepatoprotection, and anti-inflammatory activities in the intestines. By integrating the [...] Read more.
Oxyresveratrol is a stilbene compound with a simple chemical structure and various therapeutic potentials. This study summarized and analyzed the multiple pharmacological effects and mechanisms of oxyresveratrol, identifying its prominent performance in neuroprotection, hepatoprotection, and anti-inflammatory activities in the intestines. By integrating the pharmacological effects of oxyresveratrol with insights from the network pharmacology and molecular docking of its interactions with targets linked to gut–liver–brain axis disorders, it has been shown that oxyresveratrol may hold promise for the treatment of gut–liver–brain axis-related disorders. The synergistic effect between various mechanisms has inspired further research and the development of oxyresveratrol’s application value. Full article
Show Figures

Figure 1

15 pages, 1073 KiB  
Review
Revisiting the Immunometabolic Basis for the Metabolic Syndrome from an Immunonutritional View
by César Jeri Apaza, Juan Francisco Cerezo, Aurora García-Tejedor, Juan Antonio Giménez-Bastida and José Moisés Laparra-Llopis
Biomedicines 2024, 12(8), 1825; https://doi.org/10.3390/biomedicines12081825 - 12 Aug 2024
Viewed by 711
Abstract
Metabolic syndrome (MetS) implies different conditions where insulin resistance constitutes a major hallmark of the disease. The disease incurs a high risk for the development of cardiovascular complications, and takes its toll in regard to the gut–liver axis (pancreas, primary liver and colorectal)-associated [...] Read more.
Metabolic syndrome (MetS) implies different conditions where insulin resistance constitutes a major hallmark of the disease. The disease incurs a high risk for the development of cardiovascular complications, and takes its toll in regard to the gut–liver axis (pancreas, primary liver and colorectal)-associated immunity. The modulation of immunometabolic responses by immunonutritional factors (IFs) has emerged as a key determinant of the gut–liver axis’ metabolic and immune health. IFs from plant seeds have shown in vitro and pre-clinical effectiveness primarily in dealing with various immunometabolic and inflammatory diseases. Only recently have immunonutritional studies established the engagement of innate intestinal immunity to effectively control immune alterations in inflamed livers preceding the major features of the MetS. However, integrative analyses and the demonstration of causality between IFs and specific gut–liver axis-associated immunometabolic imbalances for the MetS remain ill-defined in the field. Herein, a better understanding of the IFs with a significant role in the MetS, as well as within the dynamic interplay in the functional differentiation of innate immune key effectors (i.e., monocytes/macrophages), worsening or improving the disease, could be of crucial relevance. The development of an adequate intermediary phenotype of these cells can significantly contribute to maintaining the function of Tregs and innate lymphoid cells for the prevention and treatment of MetS and associated comorbidities. Full article
(This article belongs to the Special Issue Immunometabolic Determinants of Gut–Liver Axis Health)
Show Figures

Figure 1

20 pages, 7499 KiB  
Article
Bacteroides uniformis Ameliorates Carbohydrate and Lipid Metabolism Disorders in Diabetic Mice by Regulating Bile Acid Metabolism via the Gut–Liver Axis
by Xue-Xue Zhu, Chen-Yang Zhao, Xin-Yu Meng, Xiao-Yi Yu, Lin-Chun Ma, Tian-Xiao Chen, Chang Chang, Xin-Yu Chen, Yuan Zhang, Bao Hou, Wei-Wei Cai, Bin Du, Zhi-Jun Han, Li-Ying Qiu and Hai-Jian Sun
Pharmaceuticals 2024, 17(8), 1015; https://doi.org/10.3390/ph17081015 - 1 Aug 2024
Viewed by 953
Abstract
Background: Type 2 diabetes mellitus (T2DM) is a metabolic syndrome characterized by chronic inflammation, insulin resistance, and islet cell damage. The prevention of T2DM and its associated complications is an urgent public health issue that affects hundreds of millions of people globally. Numerous [...] Read more.
Background: Type 2 diabetes mellitus (T2DM) is a metabolic syndrome characterized by chronic inflammation, insulin resistance, and islet cell damage. The prevention of T2DM and its associated complications is an urgent public health issue that affects hundreds of millions of people globally. Numerous studies suggest that disturbances in gut metabolites are important driving forces for the pathogenesis of diabetes. However, the functions and mechanisms of action of most commensal bacteria in T2DM remain largely unknown. Methods: The quantification of bile acids (BAs) in fecal samples was performed using ultra-performance liquid chromatography–tandem mass spectrometer (UPLC-MS/MS). The anti-diabetic effects of Bacteroides uniformis (B. uniformis) and its metabolites cholic acid (CA) and chenodeoxycholic acid (CDCA) were assessed in T2DM mice induced by streptozocin (STZ) plus high-fat diet (HFD). Results: We found that the abundance of B. uniformis in the feces and the contents of CA and CDCA were significantly downregulated in T2DM mice. B. uniformis was diminished in diabetic individuals and this bacterium was sufficient to promote the production of BAs. Colonization of B. uniformis and intragastric gavage of CA and CDCA effectively improved the disorder of glucose and lipid metabolism in T2DM mice by inhibiting gluconeogenesis and lipolysis in the liver. CA and CDCA improved hepatic glucose and lipid metabolism by acting on the Takeda G protein-coupled receptor 5 (TGR5)/adenosine monophosphate-activated protein kinase (AMPK) signaling pathway since knockdown of TGR5 minimized the benefit of CA and CDCA. Furthermore, we screened a natural product—vaccarin (VAC)—that exhibited anti-diabetic effects by promoting the growth of B. uniformis in vitro and in vivo. Gut microbiota pre-depletion abolished the favorable effects of VAC in diabetic mice. Conclusions: These data suggest that supplementation of B. uniformis may be a promising avenue to ameliorate T2DM by linking the gut and liver. Full article
(This article belongs to the Section Natural Products)
Show Figures

Figure 1

8 pages, 493 KiB  
Article
Genomic Characterization of Selected Escherichia coli Strains from Catfish (Clarias gariepinus) in Nigeria
by Chibuzo Linda Ekwuazi, Frank C. Ogbo, Anna Stöger, Werner Ruppitsch and Adriana Cabal Rosel
Appl. Microbiol. 2024, 4(3), 1142-1149; https://doi.org/10.3390/applmicrobiol4030077 - 24 Jul 2024
Viewed by 539
Abstract
According to a report by the World Health Organization (WHO), each year, over 550 million individuals worldwide suffer from and 230,000 die from diarrheal illnesses, which accounts for more than half of the global foodborne disease burden. Among them, children face a heightened [...] Read more.
According to a report by the World Health Organization (WHO), each year, over 550 million individuals worldwide suffer from and 230,000 die from diarrheal illnesses, which accounts for more than half of the global foodborne disease burden. Among them, children face a heightened vulnerability, with approximately 220 million falling ill and 96,000 succumbing to these diseases annually. This work aimed to study the genomic characterization of selected E. coli strains from catfish (Clarias (C.) gariepinus) caught from the Onitsha North axis of the River Niger in Anambra state, Nigeria. A total of 50 fish were randomly purchased from different fishermen over a period of four months. Samples that comprised six different organs (skin, flesh, gills, gonads, guts, and liver) were screened for E. coli strains using cultural and biochemical methods. Multilocus sequence typing (MLST) and core genome (cg)MLST were performed using Ridom SeqSphere+ software. The aerobic plate count (APC) and coliform count ranged from 0.5 × 104 to 3.7 × 104 cfu/g and 0 to 3.0 × 104 cfu/g, respectively. Whole-genome sequencing (WGS) confirmed the presence of E. coli and Klebsiella quasipneumoniae isolates in our samples. We could identify only two serotypes (O102:H7 and O40:H4) of E. coli. Antimicrobial resistance genes (ARGs) and point mutations that conferred antibiotic resistance were extracted from the genome assemblies. Good hygiene is recommended to avoid the cross-contamination of raw C. gariepinus with ready-to-eat food. Full article
Show Figures

Figure 1

24 pages, 1189 KiB  
Review
Role of Gut Microbial Metabolites in the Pathogenesis of Primary Liver Cancers
by Maria Pallozzi, Valeria De Gaetano, Natalia Di Tommaso, Lucia Cerrito, Francesco Santopaolo, Leonardo Stella, Antonio Gasbarrini and Francesca Romana Ponziani
Nutrients 2024, 16(14), 2372; https://doi.org/10.3390/nu16142372 - 22 Jul 2024
Cited by 2 | Viewed by 1712
Abstract
Hepatobiliary malignancies, which include hepatocellular carcinoma (HCC) and cholangiocarcinoma (CCA), are the sixth most common cancers and the third leading cause of cancer-related death worldwide. Hepatic carcinogenesis is highly stimulated by chronic inflammation, defined as fibrosis deposition, and an aberrant imbalance between liver [...] Read more.
Hepatobiliary malignancies, which include hepatocellular carcinoma (HCC) and cholangiocarcinoma (CCA), are the sixth most common cancers and the third leading cause of cancer-related death worldwide. Hepatic carcinogenesis is highly stimulated by chronic inflammation, defined as fibrosis deposition, and an aberrant imbalance between liver necrosis and nodular regeneration. In this context, the gut–liver axis and gut microbiota have demonstrated a critical role in the pathogenesis of HCC, as dysbiosis and altered intestinal permeability promote bacterial translocation, leading to chronic liver inflammation and tumorigenesis through several pathways. A few data exist on the role of the gut microbiota or bacteria resident in the biliary tract in the pathogenesis of CCA, and some microbial metabolites, such as choline and bile acids, seem to show an association. In this review, we analyze the impact of the gut microbiota and its metabolites on HCC and CCA development and the role of gut dysbiosis as a biomarker of hepatobiliary cancer risk and of response during anti-tumor therapy. We also discuss the future application of gut microbiota in hepatobiliary cancer management. Full article
(This article belongs to the Special Issue Diet, Gut Microbiota, and Gastrointestinal Disease)
Show Figures

Figure 1

30 pages, 3778 KiB  
Review
Metabolic Crosstalk between Liver and Brain: From Diseases to Mechanisms
by Xiaoyue Yang, Kangli Qiu, Yaoyao Jiang, Yumei Huang, Yajuan Zhang and Yunfei Liao
Int. J. Mol. Sci. 2024, 25(14), 7621; https://doi.org/10.3390/ijms25147621 - 11 Jul 2024
Viewed by 1381
Abstract
Multiple organs and tissues coordinate to respond to dietary and environmental challenges. It is interorgan crosstalk that contributes to systemic metabolic homeostasis. The liver and brain, as key metabolic organs, have their unique dialogue to transmit metabolic messages. The interconnected pathogenesis of liver [...] Read more.
Multiple organs and tissues coordinate to respond to dietary and environmental challenges. It is interorgan crosstalk that contributes to systemic metabolic homeostasis. The liver and brain, as key metabolic organs, have their unique dialogue to transmit metabolic messages. The interconnected pathogenesis of liver and brain is implicated in numerous metabolic and neurodegenerative disorders. Recent insights have positioned the liver not only as a central metabolic hub but also as an endocrine organ, capable of secreting hepatokines that transmit metabolic signals throughout the body via the bloodstream. Metabolites from the liver or gut microbiota also facilitate a complex dialogue between liver and brain. In parallel to humoral factors, the neural pathways, particularly the hypothalamic nuclei and autonomic nervous system, are pivotal in modulating the bilateral metabolic interplay between the cerebral and hepatic compartments. The term “liver–brain axis” vividly portrays this interaction. At the end of this review, we summarize cutting-edge technical advancements that have enabled the observation and manipulation of these signals, including genetic engineering, molecular tracing, and delivery technologies. These innovations are paving the way for a deeper understanding of the liver–brain axis and its role in metabolic homeostasis. Full article
(This article belongs to the Special Issue Crosstalk between Metabolism and Neuroprotection)
Show Figures

Graphical abstract

14 pages, 4016 KiB  
Article
Mitigative Effect of Graphene Oxide Nanoparticles in Maintaining Gut–Liver Homeostasis against Alcohol Injury
by Hiral Aghara, Prashsti Chadha and Palash Mandal
Gastroenterol. Insights 2024, 15(3), 574-587; https://doi.org/10.3390/gastroent15030042 - 2 Jul 2024
Viewed by 754
Abstract
Alcoholic liver disease (ALD) develops when the immunotolerant environment of the liver is compromised due to excessive alcohol consumption. ALD progression involves variations in the expressions of multiple genes, resulting in liver inflammation and the development of a leaky gut. It is still [...] Read more.
Alcoholic liver disease (ALD) develops when the immunotolerant environment of the liver is compromised due to excessive alcohol consumption. ALD progression involves variations in the expressions of multiple genes, resulting in liver inflammation and the development of a leaky gut. It is still unclear which molecular mechanism is involved in ALD progression, and due to that, there are currently no FDA-approved drugs available for its treatment. In this study, the protective effects of graphene oxide (GO) nanoparticles were investigated against ethanol-induced damage in the gut–liver axis in in vitro. GO was synthesized using a modified Hummer’s method, and characterization was performed. Given the general concerns regarding nanoparticle toxicity, assessments of cell viability, lipid accumulation, DNA damage, cell death, and the generation of reactive oxygen species (ROS) were conducted using various techniques. Furthermore, the gene expressions of pro- and anti-inflammatory cytokines were determined using RT-qPCR. The findings reveal that GO promoted cell viability even against ethanol treatment. Additionally, lipid accumulation significantly decreased when cells were treated with GO alongside ethanol compared to ethanol treatment alone, with similar trends observed for other assays. A gene expression analysis indicated that GO treatment reduced the expression of proinflammatory cytokines while enhancing the expression of antioxidant genes. Moreover, GO treatment led to improvements in gut integrity and a reduction in proinflammatory cytokines in colon cells damaged by ethanol. These findings suggest that GO holds promise as a drug carrier, exhibiting no observed toxic effects. By shedding light on the protective effects of GO against ethanol-induced damage, this study contributes to the burgeoning field of nanoparticle-mediated therapy for ALD. Full article
(This article belongs to the Section Liver)
Show Figures

Figure 1

Back to TopTop