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

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

Article Types

Countries / Regions

remove_circle_outline
remove_circle_outline
remove_circle_outline

Search Results (400)

Search Parameters:
Keywords = gut liver axis

Order results
Result details
Results per page
Select all
Export citation of selected articles as:
33 pages, 1872 KiB  
Review
Exploring the Epidemiologic Burden, Pathogenetic Features, and Clinical Outcomes of Primary Liver Cancer in Patients with Type 2 Diabetes Mellitus (T2DM) and Metabolic Dysfunction-Associated Steatotic Liver Disease (MASLD): A Scoping Review
by Mario Romeo, Fiammetta Di Nardo, Carmine Napolitano, Claudio Basile, Carlo Palma, Paolo Vaia, Marcello Dallio and Alessandro Federico
Diabetology 2025, 6(8), 79; https://doi.org/10.3390/diabetology6080079 - 4 Aug 2025
Viewed by 72
Abstract
Background/Objectives: Primary liver cancer (PLC), encompassing hepatocellular carcinoma (HCC) and cholangiocarcinoma (CCA), constitutes a growing global health concern. Metabolic dysfunction-associated Steatotic Liver Disease (MASLD) and Type 2 diabetes mellitus (T2DM) represent a recurrent epidemiological overlap. Individuals with MASLD and T2DM (MASLD-T2DM) are [...] Read more.
Background/Objectives: Primary liver cancer (PLC), encompassing hepatocellular carcinoma (HCC) and cholangiocarcinoma (CCA), constitutes a growing global health concern. Metabolic dysfunction-associated Steatotic Liver Disease (MASLD) and Type 2 diabetes mellitus (T2DM) represent a recurrent epidemiological overlap. Individuals with MASLD and T2DM (MASLD-T2DM) are at a higher risk of PLC. This scoping review highlights the epidemiological burden, the classic and novel pathogenetic frontiers, and the potential strategies optimizing the management of PLC in MASLD-T2DM. Methods: A systematic search of the PubMed, Medline, and SCOPUS electronic databases was conducted to identify evidence investigating the pathogenetic mechanisms linking MASLD and T2DM to hepatic carcinogenesis, highlighting the most relevant targets and the relatively emerging therapeutic strategies. The search algorithm included in sequence the filter words: “MASLD”, “liver steatosis”, “obesity”, “metabolic syndrome”, “body composition”, “insulin resistance”, “inflammation”, “oxidative stress”, “metabolic dysfunction”, “microbiota”, “glucose”, “immunometabolism”, “trained immunity”. Results: In the MASD-T2DM setting, insulin resistance (IR) and IR-induced mechanisms (including chronic inflammation, insulin/IGF-1 axis dysregulation, and autophagy), simultaneously with the alterations of gut microbiota composition and functioning, represent crucial pathogenetic factors in hepatocarcinogenesis. Besides, the glucose-related metabolic reprogramming emerged as a crucial pathogenetic moment contributing to cancer progression and immune evasion. In this scenario, lifestyle changes, simultaneously with antidiabetic drugs targeting IR-related effects and gut-liver axis, in parallel with novel approaches modulating immunometabolic pathways, represent promising strategies. Conclusions: Metabolic dysfunction, classically featuring MASLD-T2DM, constitutes a continuously expanding global issue, as well as a critical driver in PLC progression, demanding integrated and personalized interventions to reduce the future burden of disease. Full article
Show Figures

Figure 1

22 pages, 1013 KiB  
Review
Genomic Alterations and Microbiota Crosstalk in Hepatic Cancers: The Gut–Liver Axis in Tumorigenesis and Therapy
by Yuanji Fu, Jenny Bonifacio-Mundaca, Christophe Desterke, Íñigo Casafont and Jorge Mata-Garrido
Genes 2025, 16(8), 920; https://doi.org/10.3390/genes16080920 - 30 Jul 2025
Viewed by 221
Abstract
Background/Objectives: Hepatic cancers, including hepatocellular carcinoma (HCC) and cholangiocarcinoma (CCA), are major global health concerns due to rising incidence and limited therapeutic success. While traditional risk factors include chronic liver disease and environmental exposures, recent evidence underscores the significance of genetic alterations and [...] Read more.
Background/Objectives: Hepatic cancers, including hepatocellular carcinoma (HCC) and cholangiocarcinoma (CCA), are major global health concerns due to rising incidence and limited therapeutic success. While traditional risk factors include chronic liver disease and environmental exposures, recent evidence underscores the significance of genetic alterations and gut microbiota in liver cancer development and progression. This review aims to integrate emerging knowledge on the interplay between host genomic changes and gut microbial dynamics in the pathogenesis and treatment of hepatic cancers. Methods: We conducted a comprehensive review of current literature on genetic and epigenetic drivers of HCC and CCA, focusing on commonly mutated genes such as TP53, CTNNB1, TERT, IDH1/2, and FGFR2. In parallel, we evaluated studies addressing the gut–liver axis, including the roles of dysbiosis, microbial metabolites, and immune modulation. Key clinical and preclinical findings were synthesized to explore how host–microbe interactions influence tumorigenesis and therapeutic response. Results: HCC and CCA exhibit distinct but overlapping genomic landscapes marked by recurrent mutations and epigenetic reprogramming. Alterations in the gut microbiota contribute to hepatic inflammation, genomic instability, and immune evasion, potentially enhancing oncogenic signaling pathways. Furthermore, microbiota composition appears to affect responses to immune checkpoint inhibitors. Emerging therapeutic strategies such as probiotics, fecal microbiota transplantation, and precision oncology based on mutational profiling demonstrate potential for personalized interventions. Conclusions: The integration of host genomics with microbial ecology provides a promising paradigm for advancing diagnostics and therapies in liver cancer. Targeting the gut–liver axis may complement genome-informed strategies to improve outcomes for patients with HCC and CCA. Full article
(This article belongs to the Special Issue Feature Papers in Microbial Genetics and Genomics)
Show Figures

Figure 1

3 pages, 153 KiB  
Reply
Reply to Scarlata, G.G.M.; Abenavoli, L. Comment on “Rotaru et al. Lean MASLD and IBD: Exploring the Intersection of Metabolic Dysfunction and the Gut–Liver Axis. Life 2025, 15, 288”
by Adrian Rotaru, Remus Stafie, Ermina Stratina, Sebastian Zenovia, Robert Nastasa, Horia Minea, Laura Huiban, Tudor Cuciureanu, Cristina Muzica, Stefan Chiriac, Irina Girleanu, Ana-Maria Singeap, Catalin Sfarti, Carol Stanciu and Anca Trifan
Life 2025, 15(8), 1211; https://doi.org/10.3390/life15081211 - 30 Jul 2025
Viewed by 242
Abstract
We are grateful to Dr [...] Full article
(This article belongs to the Section Medical Research)
2 pages, 144 KiB  
Comment
Comment on Rotaru et al. Lean MASLD and IBD: Exploring the Intersection of Metabolic Dysfunction and the Gut–Liver Axis. Life 2025, 15, 288
by Giuseppe Guido Maria Scarlata and Ludovico Abenavoli
Life 2025, 15(8), 1210; https://doi.org/10.3390/life15081210 - 30 Jul 2025
Viewed by 319
Abstract
We read with great interest the recent article by Rotaru et al [...] Full article
18 pages, 2629 KiB  
Article
Dietary Interventions with Bletilla striata Polysaccharides and/or Composite Polysaccharides Remodel Liver Lipid Profiles and Ameliorate Gut Metabolic Disturbances in High-Fat Diet-Induced Obese Mice
by Peiting Zhang, Jinjin Dong, Jiamin Lu, Zijian Cai, Bingde Zhou, Qian Zhang, Chenglin Zhu and Luca Laghi
Foods 2025, 14(15), 2653; https://doi.org/10.3390/foods14152653 - 29 Jul 2025
Viewed by 186
Abstract
The global obesity epidemic and associated metabolic disorders present urgent public health challenges. This study employed a multi-omics approach (lipidomics, metabolomics, and gut microbiome analysis) to investigate how Bletilla striata polysaccharides (BSPs) and composite polysaccharides modulate liver lipid metabolism and gut microbiota in [...] Read more.
The global obesity epidemic and associated metabolic disorders present urgent public health challenges. This study employed a multi-omics approach (lipidomics, metabolomics, and gut microbiome analysis) to investigate how Bletilla striata polysaccharides (BSPs) and composite polysaccharides modulate liver lipid metabolism and gut microbiota in high-fat diet (HFD)-induced obese mice. HFD elevated hepatic phosphatidylcholines, cholesteryl esters (CEs), and acylcarnitines (CARs), alongside increased cecal choline and trimethylamine. BSP interventions reduced hepatic CEs, free fatty acids (FAs), CARs, and cecal sarcosine while restoring gut microbial diversity. Notably, BSP enriched beneficial genera, including Jeotgalicoccus and Atopostipes, and the network analysis revealed negative correlations between these genera and hepatic triglycerides (TGs), implicating the gut–liver axis in lipid metabolism regulation. These findings elucidate the anti-obesity mechanisms of polysaccharides through gut microbiota remodeling and cross-tissue metabolic interactions, providing a foundation for leveraging plant polysaccharides in developing safer, effective obesity therapies. Full article
(This article belongs to the Section Food Nutrition)
Show Figures

Graphical abstract

28 pages, 1362 KiB  
Review
Multifaceted Interactions Between Bile Acids, Their Receptors, and MASH: From Molecular Mechanisms to Clinical Therapeutics
by Xuan Tang, Yuanjiao Zhou, Li Xia, Xiulian Lin, Yao Zhu, Menghan Chen, Jiayao Wang and Yamei Li
Molecules 2025, 30(15), 3066; https://doi.org/10.3390/molecules30153066 - 22 Jul 2025
Viewed by 387
Abstract
Metabolic dysfunction-associated steatohepatitis (MASH) represents a critical hepatic manifestation within the broader spectrum of metabolic syndrome. The pathogenesis of MASH is characterized by disruptions in lipid metabolism, inflammation, and fibrosis. Bile acids and their receptors are integral to the progression of MASH, primarily [...] Read more.
Metabolic dysfunction-associated steatohepatitis (MASH) represents a critical hepatic manifestation within the broader spectrum of metabolic syndrome. The pathogenesis of MASH is characterized by disruptions in lipid metabolism, inflammation, and fibrosis. Bile acids and their receptors are integral to the progression of MASH, primarily through their regulatory influence on the metabolic networks of the gut–liver axis. This review offers a comprehensive and systematic examination of the molecular mechanisms underlying bile acid biosynthesis, metabolic dysregulation, and receptor signaling anomalies in MASH. Furthermore, it explores the translational potential of these insights into clinical therapies. Bile acids and their receptors emerge as pivotal therapeutic targets for MASH. Future research should focus on an in-depth analysis of dynamic regulatory mechanisms and the optimization of multi-target combination therapies, thereby paving the way for significant clinical advancements. Full article
(This article belongs to the Special Issue Chemical Biology in Asia—Second Edition)
Show Figures

Figure 1

22 pages, 2637 KiB  
Article
Anti-Obesity and Hepatoprotective Effects of Probiotic Goat Milk in Mice: Insights from Hepatic Proteomics
by Antonela Marquez, Estefanía Andrada, Matias Russo, Jaime Daniel Babot, Roxana Medina and Paola Gauffin-Cano
Fermentation 2025, 11(7), 419; https://doi.org/10.3390/fermentation11070419 - 20 Jul 2025
Viewed by 543
Abstract
Dietary administration of fermented goat milk (FGM) with the starter strain Lactobacillus delbrueckii subsp. indicus CRL1447 and supplemented with different functional cultures (FCs) of lactobacilli strains (FC1: Limosilactobacillus fermentum CRL1446 + Lactiplantibacillus paraplantarum CRL1449 + Lactiplantibacillus paraplantarum CRL1472; FC2: CRL1446 + CRL1449; FC3: [...] Read more.
Dietary administration of fermented goat milk (FGM) with the starter strain Lactobacillus delbrueckii subsp. indicus CRL1447 and supplemented with different functional cultures (FCs) of lactobacilli strains (FC1: Limosilactobacillus fermentum CRL1446 + Lactiplantibacillus paraplantarum CRL1449 + Lactiplantibacillus paraplantarum CRL1472; FC2: CRL1446 + CRL1449; FC3: CRL1446 + CRL1472; and FC4: CRL1449 + CRL1472) was investigated in mice fed a high-fat diet (HFD). FGM supplemented with different FCs, referred to as Probiotic Goat Milk (PGM), demonstrated significant anti-obesity activity by reducing body weight and improving blood lipid profiles in obese mice. The animals that received the PGM showed less fat infiltration in the hepatocytes compared to the obese mice fed FGM. Hepatic proteomics data show that HFD generally upregulates proteins involved in fatty acid oxidation and downregulates proteins implicated in lipid synthesis, whereas the administration of FGM supplemented with FC3 (PGM3) improves the proteomic profile. These results suggest that PGM exerts systemic metabolic effects through modulation of the gut–liver axis, highlighting its potential as a dietary strategy against obesity-related disorders. Full article
Show Figures

Figure 1

15 pages, 4789 KiB  
Article
Lacticaseibacillus paracasei 36 Mitigates Alcoholic-Associated Liver Disease Through Modulation of Microbiota and AMPK Signaling
by Chongyu Wang, Xi Chen, Fei Wang, Tianyu Chen, Mengqiu Yin, Ziyu Liu, Weifen Li and Jinhui Zhu
Nutrients 2025, 17(14), 2340; https://doi.org/10.3390/nu17142340 - 17 Jul 2025
Viewed by 365
Abstract
Background: Alcohol-associated liver disease (ALD) is characterized by gut–liver axis dysfunction and metabolic dysregulation, yet the therapeutic potential of probiotics remains underexplored. This study aimed to investigate the protective effects and mechanisms of Lacticaseibacillus paracasei 36 (LP36) against ethanol-induced ALD in mice. Methods: [...] Read more.
Background: Alcohol-associated liver disease (ALD) is characterized by gut–liver axis dysfunction and metabolic dysregulation, yet the therapeutic potential of probiotics remains underexplored. This study aimed to investigate the protective effects and mechanisms of Lacticaseibacillus paracasei 36 (LP36) against ethanol-induced ALD in mice. Methods: Mice were pretreated with LP36 prior to ethanol exposure. Liver injury was assessed through serum ALT/AST levels, hepatic steatosis (TC/TG content), and ethanol detoxification capacity (ADH/ALDH activity). Intestinal barrier integrity was evaluated via Mucin2 and ZO-1 expression, and gut microbiota alterations were analyzed by 16S rRNA sequencing. Hepatic transcriptomics (RNA-seq) was performed to identify key regulatory pathways. Results: LP36 significantly attenuated ethanol-induced liver injury, evidenced by reduced ALT/AST, improved hepatic steatosis (lower TC/TG), and enhanced ADH/ALDH activity. Mechanistically, LP36 restored intestinal barrier function (upregulated Mucin2 and ZO-1), modulated gut microbiota (suppressed Parasutterella, Romboutsia, and Christensenellaceae_R-7_group; enriched Faecalibaculum and Tuzzerella), and reduced systemic inflammation. Transcriptomics revealed LP36-mediated rescue of AMPK signaling, involving regulation of Stk11, Prkag3, lipid synthesis genes (Fasn, Acaca), and metabolic modulators (Creb3l3, G6pc3, mTOR, Rps6kb2).Conclusions: LP36 ameliorates ethanol-induced ALD by enhancing intestinal barrier integrity, reshaping gut microbiota, and restoring AMPK-dependent metabolic homeostasis. These findings highlight LP36 as a promising probiotic candidate for ALD prevention. Full article
(This article belongs to the Section Prebiotics and Probiotics)
Show Figures

Figure 1

27 pages, 5846 KiB  
Article
Agrocybe cylindracea Polysaccharides Ameliorate DSS-Induced Colitis by Restoring Intestinal Barrier Function and Reprogramming Immune Homeostasis via the Gut–Liver Axis
by Aamna Atta, Muhammad Naveed, Mujeeb Ur Rahman, Yamina Alioui, Immad Ansari, Sharafat Ali, Eslam Ghaleb, Nabeel Ahmed Farooqui, Mohammad Abusidu, Yi Xin and Bin Feng
Int. J. Mol. Sci. 2025, 26(14), 6805; https://doi.org/10.3390/ijms26146805 - 16 Jul 2025
Viewed by 434
Abstract
Ulcerative colitis (UC) is a chronic inflammatory bowel disease driven by immune dysregulation, microbiota imbalance, and intestinal barrier dysfunction. Despite its global burden, effective therapies remain limited. This study explores the therapeutic potential of Agrocybe cylindracea polysaccharides (ACP) in a dextran sulfate sodium [...] Read more.
Ulcerative colitis (UC) is a chronic inflammatory bowel disease driven by immune dysregulation, microbiota imbalance, and intestinal barrier dysfunction. Despite its global burden, effective therapies remain limited. This study explores the therapeutic potential of Agrocybe cylindracea polysaccharides (ACP) in a dextran sulfate sodium (DSS)-induced murine colitis model. High-performance liquid chromatography (HPLC)-characterized ACP was administered orally to BALB/c mice following colitis induction. ACP treatment significantly reduced Disease Activity Index (DAI) scores, preserved colon length, and restored intestinal barrier integrity by upregulating tight junction proteins. Mechanistically, ACP modulated immune homeostasis, suppressing pro-inflammatory cytokines (IL-17, IL-23, CRP) while enhancing anti-inflammatory mediators (IL-4, TGF-β). Furthermore, ACP inhibited hepatic TLR4/MyD88/NF-κB signaling, attenuated systemic inflammation, and reshaped gut microbiota composition by enriching beneficial taxa and reducing pathogenic Bacteroides. These findings demonstrate ACP multi-target efficacy in colitis, positioning it as a promising natural therapeutic for UC. Full article
Show Figures

Figure 1

16 pages, 3260 KiB  
Article
Rifaximin Attenuates Liver Fibrosis and Hepatocarcinogenesis in a Rat MASH Model by Suppressing the Gut–Liver Axis and Epiregulin–IL-8-Associated Angiogenesis
by Naoki Nishimura, Kosuke Kaji, Norihisa Nishimura, Junichi Hanatani, Tatsuya Nakatani, Masafumi Oyama, Akihiko Shibamoto, Yuki Tsuji, Koh Kitagawa, Shinya Sato, Tadashi Namisaki, Satoru Tamaoki and Hitoshi Yoshiji
Int. J. Mol. Sci. 2025, 26(14), 6710; https://doi.org/10.3390/ijms26146710 - 12 Jul 2025
Viewed by 400
Abstract
Metabolic dysfunction-associated steatohepatitis (MASH) is a progressive liver disease linked to fibrosis and hepatocellular carcinoma (HCC). Gut-derived lipopolysaccharide (LPS) promotes hepatic inflammation, fibrosis, and angiogenesis through toll-like receptor 4 (TLR4) signaling. This study examined the effects of rifaximin, a non-absorbable, gut-targeted antibiotic, on [...] Read more.
Metabolic dysfunction-associated steatohepatitis (MASH) is a progressive liver disease linked to fibrosis and hepatocellular carcinoma (HCC). Gut-derived lipopolysaccharide (LPS) promotes hepatic inflammation, fibrosis, and angiogenesis through toll-like receptor 4 (TLR4) signaling. This study examined the effects of rifaximin, a non-absorbable, gut-targeted antibiotic, on MASH-related liver fibrosis and early hepatocarcinogenesis, with a focus on the LPS–epiregulin–IL-8–angiogenesis axis.MASH was induced in Fischer 344 rats using a choline-deficient, L-amino acid-defined high-fat diet (CDAHFD). Rifaximin (30 mg/kg/day) was orally administered for 12 weeks. Liver histology, gene expression, intestinal permeability, LPS levels, and angiogenic markers were evaluated. Rifaximin reduced hepatic inflammation, fibrosis, hydroxyproline content, and fibrogenic gene expression. The number and size of GST-P-positive preneoplastic lesions and proliferation-related genes were decreased. Portal LPS levels and Kupffer cell activation declined, with downregulation of Lbp, Cd14, Tlr4, and inflammatory cytokines. Rifaximin decreased hepatic epiregulin and IL-8 expression, attenuated CD34-positive neovascularization, and suppressed proangiogenic gene expression, accompanied by improved intestinal barrier function and reduced gut permeability. Rifaximin mitigates MASH progression by restoring gut barrier integrity, limiting LPS translocation, and inhibiting fibrogenic and angiogenic pathways. These results suggest its potential as a chemopreventive agent in MASH-related hepatocarcinogenesis. Full article
(This article belongs to the Special Issue Liver Diseases: From Molecular Basis to Potential Therapy)
Show Figures

Figure 1

18 pages, 5392 KiB  
Article
Kaempferol Alleviates Carbon Tetrachloride-Induced Liver Fibrosis in Mice by Regulating Intestinal Short-Chain Fatty Acids
by Siqi Zhang, Fei Tang, Zhe Zhou, Linhui Li, Yang Tang, Kaiwen Fu, Yang Tan and Ling Li
Int. J. Mol. Sci. 2025, 26(14), 6666; https://doi.org/10.3390/ijms26146666 - 11 Jul 2025
Viewed by 348
Abstract
Liver fibrosis remains a critical health concern with limited therapeutic options. Kaempferol (Kae) is a natural flavonoid widely present in natural plants, yet its role in modulating gut–liver axis interactions during fibrosis is unexplored. This study investigates the hepatoprotective effects of Kae on [...] Read more.
Liver fibrosis remains a critical health concern with limited therapeutic options. Kaempferol (Kae) is a natural flavonoid widely present in natural plants, yet its role in modulating gut–liver axis interactions during fibrosis is unexplored. This study investigates the hepatoprotective effects of Kae on alleviating carbon tetrachloride (CCl4)-induced liver fibrosis, and its underlying mechanisms, focusing on oxidative stress, gut microbiota, and short-chain fatty acids (SCFAs), are revealed. A mouse model of hepatic fibrosis was built by the subcutaneous injection of CCl4. Meanwhile, Kae was administered by gavage at doses of 25, 50, and 100 mg/kg body weight. Serum biomarkers, liver histopathology, oxidative damage markers, and nuclear factor erythroid 2-related factor 2 (Nrf2)/kelch-like ECH-associated protein 1 (Keap1)/heme oxygenase 1 (HO-1) signaling were analyzed. AML12 hepatocytes were pretreated with Kae or SCFAs (acetate, propionate, butyrate) before H2O2-induced oxidative injury. The changes in gut microbiota and the levels of SCFAs were assessed via 16S rRNA sequencing and GC-MS, respectively. Kae effectively alleviated the destruction of the liver morphology and tissue structure, reduced the infiltration of inflammatory cells, collagen deposition in the liver, and the expression of fibrotic factors, and downregulated the oxidative stress level in the liver of mice with liver fibrosis by activating the Nrf2/Keap1/HO-1 pathway (p < 0.05 or 0.01). In vitro, Kae significantly mitigated H2O2-induced cytotoxicity and oxidative damage (p < 0.05 or 0.01). Furthermore, Kae restored gut microbiota diversity, increased beneficial genera (e.g., Lactobacillus), and elevated both intestinal and hepatic SCFA levels (p < 0.01). The discrepant SCFA pretreatment similarly protected AML12 cells by activating Nrf2 signaling (p < 0.05 or 0.01). Our research suggests that Kae could inhibit CCl4-induced liver fibrosis by restoring the levels of intestinal metabolite SCFAs to reduce oxidative damage. Full article
(This article belongs to the Section Molecular Pharmacology)
Show Figures

Figure 1

22 pages, 4797 KiB  
Article
Silver Nanoparticles Synthesized from Enicostemma littorale Exhibit Gut Tight Junction Restoration and Hepatoprotective Activity via Regulation of the Inflammatory Pathway
by Hiral Aghara, Simran Samanta, Manali Patel, Prashsti Chadha, Divyesh Patel, Anamika Jha and Palash Mandal
Pharmaceutics 2025, 17(7), 895; https://doi.org/10.3390/pharmaceutics17070895 - 9 Jul 2025
Viewed by 504
Abstract
Background: Alcohol-associated liver disease (ALD) is a primary global health concern, exacerbated by oxidative stress, inflammation, and gut barrier dysfunction. Conventional phytocompounds exhibit hepatoprotective potential but are hindered by low bioavailability. This study aimed to evaluate the hepatoprotective and gut-barrier-restorative effects of green-synthesized [...] Read more.
Background: Alcohol-associated liver disease (ALD) is a primary global health concern, exacerbated by oxidative stress, inflammation, and gut barrier dysfunction. Conventional phytocompounds exhibit hepatoprotective potential but are hindered by low bioavailability. This study aimed to evaluate the hepatoprotective and gut-barrier-restorative effects of green-synthesized silver nanoparticles (AgNPs) derived from Enicostemma littorale, a medicinal plant known for its antioxidant and anti-inflammatory properties. Methods: AgNPs were synthesized using aqueous leaf extract of E. littorale and characterized using UV-Vis, XRD, FTIR, DLS, and SEM. HepG2 (liver) and Caco-2 (colon) cells were exposed to 0.2 M ethanol, AgNPs (1–100 µg/mL), or both, to simulate ethanol-induced toxicity. A range of in vitro assays was performed to assess cell viability, oxidative stress (H2DCFDA), nuclear and morphological integrity (DAPI and AO/EtBr staining), lipid accumulation (Oil Red O), and gene expression of pro- and anti-inflammatory, antioxidant, and tight-junction markers using RT-qPCR. Results: Ethanol exposure significantly increased ROS, lipid accumulation, and the expression of inflammatory genes, while decreasing antioxidant enzymes and tight-junction proteins. Green AgNPs at lower concentrations (1 and 10 µg/mL) restored cell viability, reduced ROS levels, preserved nuclear morphology, and downregulated CYP2E1 and SREBP expression. Notably, AgNPs improved the expression of Nrf2, HO-1, ZO-1, and IL-10, and reduced TNF-α and IL-6 expression in both cell lines, indicating protective effects on both liver and intestinal cells. Conclusions: Green-synthesized AgNPs from E. littorale exhibit potent hepatoprotective and gut-barrier-restoring effects through antioxidant, anti-inflammatory, and antilipidemic mechanisms. These findings support the therapeutic potential of plant-based nanoparticles in mitigating ethanol-induced gut–liver axis dysfunction. Full article
(This article belongs to the Special Issue Nanoparticles for Liver Diseases Therapy)
Show Figures

Figure 1

16 pages, 488 KiB  
Review
Exploring the Role of Probiotics, Prebiotics, and Synbiotics in the Treatment of Metabolic Dysfunction-Associated Steatotic Liver Disease—A Scoping Review
by Anastasia Ntikoudi, Anastasia Papachristou, Alketa Spirou, Eleni Evangelou, Athanasios Tsartsalis, Eugenia Vlachou and George Mastorakos
Livers 2025, 5(3), 31; https://doi.org/10.3390/livers5030031 - 7 Jul 2025
Viewed by 385
Abstract
Background: Metabolic dysfunction-associated steatotic liver disease (MASLD) has emerged as the most prevalent chronic liver condition. Its prevalence is estimated to further increase. The gut–liver axis, which represents both anatomical and functional connections, contributes significantly to the development of MASLD. Dysbiosis, characterized by [...] Read more.
Background: Metabolic dysfunction-associated steatotic liver disease (MASLD) has emerged as the most prevalent chronic liver condition. Its prevalence is estimated to further increase. The gut–liver axis, which represents both anatomical and functional connections, contributes significantly to the development of MASLD. Dysbiosis, characterized by an imbalance in gut microbiota, can exacerbate the disease by increasing intestinal permeability, which permits harmful bacteria and their components to enter the bloodstream. This review sought to explore the impact of probiotics, prebiotics, and synbiotics on the treatment of MASLD. Method: The methodology for scoping reviews in accordance with Prisma-ScR guidelines was followed. A comprehensive search was conducted in databases such as PubMed, Scopus, and Medline. Out of 1390 studies screened, 25 were selected for the final analysis. Results: The findings of this scoping review highlight the therapeutic potential of probiotics, prebiotics, and synbiotics in the management and treatment of MASLD, as showcased by the existing literature. Conclusions: This scoping review offers important insights into the advantages of probiotics, prebiotics, and synbiotics in the treatment of MASLD. The limitations identified in this study emphasize the necessity for larger, long-term, and geographically diverse studies in order to obtain more solid scientific results. Full article
Show Figures

Figure 1

16 pages, 3254 KiB  
Article
Integrated Microbiome–Metabolome Analysis Reveals Intestine–Liver Metabolic Associations in the Moustache Toad
by Shui-Sheng Yu, Jing-Wen Xiang, Lin Zhang, Xiao-Hua Guo, Yu Wang, Guo-Hua Ding and Hua-Li Hu
Animals 2025, 15(13), 1973; https://doi.org/10.3390/ani15131973 - 4 Jul 2025
Viewed by 383
Abstract
The intestinal microbiota regulates host metabolic homeostasis through production of bioactive microbial metabolites. These microorganisms facilitate digestion, enhance immune function, maintain osmoregulation, and support physiological balance via these bioactive compounds, thereby enhancing environmental adaptation. Our study investigated intestinal microbiota–liver metabolic interactions in Leptobrachium [...] Read more.
The intestinal microbiota regulates host metabolic homeostasis through production of bioactive microbial metabolites. These microorganisms facilitate digestion, enhance immune function, maintain osmoregulation, and support physiological balance via these bioactive compounds, thereby enhancing environmental adaptation. Our study investigated intestinal microbiota–liver metabolic interactions in Leptobrachium liui using 16S rRNA gene sequencing and non-targeted liquid chromatography–tandem mass spectrometry metabolomics. Key findings include (1) comparable alpha diversity but distinct microbial community structures between the small intestine (SI) and large intestine (LI), with the SI dominated by Enterobacteriaceae (72.14%) and the LI by Chitinophagaceae (55.16%); (2) segment-specific microbe–metabolite correlations, with predominantly positive correlations in the SI and complex patterns in the LI involving fatty acids, amino acids, and energy metabolites; and (3) significant correlations between specific bacterial families (Aeromonadaceae, Enterobacteriaceae, Chitinophagaceae) and hepatic metabolites related to fatty acid metabolism, amino acid synthesis, and energy pathways, indicating potential gut–liver axis associations. These findings provide insights into amphibian intestinal microbiota–hepatic metabolite associations and may inform future studies of host–microbe interactions. Full article
Show Figures

Figure 1

26 pages, 1605 KiB  
Review
Thiamine Deficiency and Neuroinflammation Are Important Contributors to Alcohol Use Disorder
by Nikhila Kalapatapu, Samantha G. Skinner, Emma G. D’Addezio, Srija Ponna, Enrique Cadenas and Daryl L. Davies
Pathophysiology 2025, 32(3), 34; https://doi.org/10.3390/pathophysiology32030034 - 4 Jul 2025
Viewed by 964
Abstract
Despite the growing morbidity associated with alcohol use disorder (AUD), current FDA-approved therapeutics fail to adequately address the condition. This is in part due to the complex systemic effects of ethanol (EtOH), which have particularly negative consequences on the gut–liver–brain axis. Importantly, two [...] Read more.
Despite the growing morbidity associated with alcohol use disorder (AUD), current FDA-approved therapeutics fail to adequately address the condition. This is in part due to the complex systemic effects of ethanol (EtOH), which have particularly negative consequences on the gut–liver–brain axis. Importantly, two systemic mechanisms underlying the progression of AUD remain underemphasized in therapeutic development: thiamine deficiency and neuroinflammation. Alcohol-induced thiamine deficiency leads to reduced activity of key metabolic enzymes, thereby resulting in energy deficits, oxidative stress, and severe clinical implications. EtOH also activates TLR4 and NLRP3, both of which play critical roles in the regulation of neuroimmune responses. While research directly investigating the relationship between thiamine deficiency and neuroinflammation is still in its early stages, our review highlights the emerging connections between these two seemingly distinct pathomechanisms. Additionally, potential therapeutic approaches and targets for addressing AUD at a systemic level are discussed. Full article
(This article belongs to the Section Systemic Pathophysiology)
Show Figures

Graphical abstract

Back to TopTop