1
|
Tüsüz Önata E, Özdemir Ö. Fecal microbiota transplantation in allergic diseases. World J Methodol 2025; 15:101430. [DOI: 10.5662/wjm.v15.i2.101430] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/13/2024] [Revised: 10/17/2024] [Accepted: 11/01/2024] [Indexed: 11/27/2024] Open
Abstract
Microorganisms such as bacteria, fungi, viruses, parasites living in the human intestine constitute the human intestinal microbiota. Dysbiosis refers to compositional and quantitative changes that negatively affect healthy gut microbiota. In recent years, with the demonstration that many diseases are associated with dysbiosis, treatment strategies targeting the correction of dysbiosis in the treatment of these diseases have begun to be investigated. Faecal microbiota transplantation (FMT) is the process of transferring faeces from a healthy donor to another recipient in order to restore the gut microbiota and provide a therapeutic benefit. FMT studies have gained popularity after probiotic, prebiotic, symbiotic studies in the treatment of dysbiosis and related diseases. FMT has emerged as a potential new therapy in the treatment of allergic diseases as it is associated with the maintenance of intestinal microbiota and immunological balance (T helper 1/T helper 2 cells) and thus suppression of allergic responses. In this article, the definition, application, safety and use of FMT in allergic diseases will be discussed with current data.
Collapse
Affiliation(s)
- Ece Tüsüz Önata
- Division of Pediatric Allergy and Immunology, Medical Faculty, Sakarya University, Adapazarı 54100, Sakarya, Türkiye
| | - Öner Özdemir
- Division of Pediatric Allergy and Immunology, Medical Faculty, Sakarya University, Adapazarı 54100, Sakarya, Türkiye
| |
Collapse
|
2
|
Das M, Kiruthiga C, Shafreen RB, Nachammai K, Selvaraj C, Langeswaran K. Harnessing the human microbiome and its impact on immuno-oncology and nanotechnology for next-generation cancer therapies. Eur J Pharmacol 2025; 996:177436. [PMID: 40023356 DOI: 10.1016/j.ejphar.2025.177436] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2024] [Revised: 02/14/2025] [Accepted: 02/26/2025] [Indexed: 03/04/2025]
Abstract
The integration of microbiome research and nanotechnology represents a significant advancement in immuno-oncology, potentially improving the effectiveness of cancer immunotherapies. Recent studies highlight the influential role of the human microbiome in modulating immune responses, presenting new opportunities to enhance immune checkpoint inhibitors (ICIs) and other cancer therapies. Nanotechnology offers precise drug delivery and immune modulation capabilities, minimizing off-target effects while maximizing therapeutic outcomes. This review consolidates current knowledge on the interactions between the microbiome and the immune system, emphasizing the microbiome's impact on ICIs, and explores the incorporation of nanotechnology in cancer treatment strategies. Additionally, it provides a forward-looking perspective on the synergistic potential of microbiome modulation and nanotechnology to overcome existing challenges in immuno-oncology. This integrated approach may enhance the personalization and effectiveness of next-generation cancer treatments, paving the way for transformative patient care.
Collapse
Affiliation(s)
- Mamali Das
- Department of Biomedical Science, Alagappa University, Karaikudi, 630003, India
| | | | - R Beema Shafreen
- Department of Biomedical Science, Alagappa University, Karaikudi, 630003, India
| | - Kathiresan Nachammai
- Department of Biotechnology, Alagappa University, Science Campus, Karaikudi, Tamil Nadu, India
| | - Chandrabose Selvaraj
- CsrDD Lab, Department of Microbiology, Dr. D. Y. Patil Medical College Hospital & Research Centre, Dr. D. Y. Patil Vidyapeeth (Deemed to Be University), Pimpri, Pune, 411018, India.
| | - K Langeswaran
- Department of Biomedical Science, Alagappa University, Karaikudi, 630003, India; Department of Biotechnology, Alagappa University, Science Campus, Karaikudi, Tamil Nadu, India.
| |
Collapse
|
3
|
Sadri M, Shafaghat Z, Roozbehani M, Hoseinzadeh A, Mohammadi F, Arab FL, Minaeian S, Fard SR, Faraji F. Effects of Probiotics on Liver Diseases: Current In Vitro and In Vivo Studies. Probiotics Antimicrob Proteins 2025; 17:1688-1710. [PMID: 39739162 DOI: 10.1007/s12602-024-10431-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/06/2024] [Indexed: 01/02/2025]
Abstract
Various types of liver or hepatic diseases cause the death of about 2 million people worldwide every year, of which 1 million die from the complications of cirrhosis and another million from hepatocellular carcinoma and viral hepatitis. Currently, the second most common solid organ transplant is the liver, and the current rate represents less than 10% of global transplant requests. Hence, finding new approaches to treat and prevent liver diseases is essential. In liver diseases, the interaction between the liver, gut, and immune system is crucial, and probiotics positively affect the human microbiota. Probiotics are a non-toxic and biosafe alternative to synthetic chemical compounds. Health promotion by lowering cholesterol levels, stimulating host immunity, the natural gut microbiota, and other functions are some of the activities of probiotics, and their metabolites, including bacteriocins, can exert antimicrobial effects against a broad range of pathogenic bacteria. The present review discusses the available data on the results of preclinical and clinical studies on the effects of probiotic administration on different types of liver diseases.
Collapse
Affiliation(s)
- Maryam Sadri
- Department of Immunology, Iran University of Medical Sciences, Tehran, Iran
| | - Zahra Shafaghat
- Department of Immunology, Iran University of Medical Sciences, Tehran, Iran
| | - Mona Roozbehani
- Vaccine Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Akram Hoseinzadeh
- Cancer Research Center, Faculty of Medicine, Semnan University of Medical Sciences, Semnan, Iran
| | - Fatemeh Mohammadi
- Department of Immunology, School of Medicine, Mashhad University of Medicine Sciences, Mashhad, Iran
| | - Fahimeh Lavi Arab
- Department of Immunology, School of Medicine, Mashhad University of Medicine Sciences, Mashhad, Iran
| | - Sara Minaeian
- Antimicrobial Resistance Research Center, Institute of Immunology and Infectious Diseases, Iran University of Medicine Sciences, Tehran, Iran
| | - Soheil Rahmani Fard
- Antimicrobial Resistance Research Center, Institute of Immunology and Infectious Diseases, Iran University of Medicine Sciences, Tehran, Iran
| | - Fatemeh Faraji
- Antimicrobial Resistance Research Center, Institute of Immunology and Infectious Diseases, Iran University of Medicine Sciences, Tehran, Iran.
| |
Collapse
|
4
|
Ebrahim YM, Sadek MA, Sabry MO, Lotfy RM, El-Dessouki AM, Abou-Hussein D, El-Shiekh RA, ElBishbishy RM. Integrative sleep management: from molecular pathways to conventional and herbal treatments. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2025:10.1007/s00210-025-04183-y. [PMID: 40338321 DOI: 10.1007/s00210-025-04183-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/08/2025] [Accepted: 04/13/2025] [Indexed: 05/09/2025]
Abstract
Sleep is regarded as one of the most crucial factors in keeping a healthy lifestyle. To function normally, a person needs at least 6-8 h of sleep per day. Sleep influences not only our mood but also the efficiency with which we complete tasks. Sleep disorders exhibit diverse etiologies across different conditions and populations, with genetic and environmental factors playing a significant role in their development. Many issues emerge as a result of inadequate sleep. Unhealthy food and lifestyle choices have increased our susceptibility to sleep disorders. A well-balanced diet rich in essential vitamins and minerals can have a profound impact on sleep patterns, enhancing both the duration and quality of rest. The primary categories of sleep disorders include insomnia, sleep apnea (SA), narcolepsy, parasomnias, circadian rhythm disorders, and restless legs syndrome (RLS). The drugs used to treat sleep disorders are primarily habit-forming and have a history of withdrawal effects. This insufficiency in medication has prompted the hunt for newer, better options. Nutraceuticals are well-suited to the treatment of such illnesses. Its non-toxic, non-habit-forming properties, and practical efficiency have made it an outstanding choice. This review provides nutraceuticals used in sleep disorders. A comprehensive literature search was conducted utilizing several databases, including Google Scholar, Elsevier, Springer Nature, Wiley, PubMed, and EKB. Nutraceuticals are products that employ food or dietary components to treat or prevent disease. In the therapy of sleep disorders, nutraceuticals such as Artemisia annua, valerian, rosemary, jujube, Passionflower, lemon balm, ashwagandha, kava-kava, lavender, and chamomile have been shown to have remarkable benefits. These remedies exert their effects through multiple mechanisms, both directly by modulating neurotransmitter and hormonal pathways within sleep circuits, and indirectly by enhancing sleep quality through the alleviation of stress, inflammation, and oxidative stress. Clinical studies were piloted to validate the efficacy of natural sleep aids. Future research should focus on elucidating the precise mechanisms through which natural products influence sleep.
Collapse
Affiliation(s)
- Yasmina M Ebrahim
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Cairo University, Cairo, 11562, Egypt
| | - Mohamed A Sadek
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Cairo University, Cairo, 11562, Egypt
- Department of Pharmacology and Toxicology, College of Medicine, University of Arkansas for Medical Sciences, Little Rock, AR, 72205, USA
| | - Miral O Sabry
- Faculty of Science, National University of Singapore, Singapore Institute of Manufacturing Technology (SIMTech), Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
| | - Rana M Lotfy
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Cairo University, Cairo, 11562, Egypt
| | - Ahmed M El-Dessouki
- Pharmacology and Toxicology Department, Faculty of Pharmacy, Ahram Canadian University, 6th of October City, 12566, Giza, Egypt
| | - Dina Abou-Hussein
- Department of Pharmacognosy, Faculty of Pharmacy, Cairo University, Cairo, 11562, Egypt
| | - Riham A El-Shiekh
- Department of Pharmacognosy, Faculty of Pharmacy, Cairo University, Cairo, 11562, Egypt.
| | - Rana M ElBishbishy
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Cairo University, Cairo, 11562, Egypt
| |
Collapse
|
5
|
Moustafa PE, Farouk H, Khattab MS, El-Marasy SA. Diacerein counteracts amiodarone‑induced hepatotoxicity in rats via targeting TLR4/NF-kB/NLRP3 pathways. Toxicol Mech Methods 2025:1-13. [PMID: 40331897 DOI: 10.1080/15376516.2025.2499024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2025] [Revised: 04/14/2025] [Accepted: 04/23/2025] [Indexed: 05/08/2025]
Abstract
This study investigates the protective effects of diacerein (DCN) against amiodarone (AMIO)-induced hepatotoxicity in a rat model. AMIO administration resulted in significant elevations of liver enzymes, ALT and AST, indicating hepatocellular membrane disruption and oxidative stress, as demonstrated by elevated levels of malondialdehyde (MDA) and decreased glutathione (GSH). Additionally, pro-inflammatory cytokines including TNF-α and IL-1β were expressed more when AMIO triggered the Toll-like receptor 4/nuclear factor kappa B/inflammasome 3 (TLR4/NF-κB/NLRP3) inflammatory pathway, along with elevated caspase-1 (CASP1) levels, which promoted apoptosis. In contrast, oral administration of DCN for two weeks effectively mitigated these effects by reducing liver enzyme levels and improving histopathological alterations. DCN also demonstrated anti-oxidant properties by decreasing MDA levels and increasing nuclear factor erythroid 2-related factor 2 (Nrf2) and GSH content. Furthermore, DCN downregulated the hepatic content of TLR4, NF-κB p65, NLRP3, CASP1, and pro-inflammatory cytokines, thereby inhibiting the activation of the inflammatory cascade. Moreover, DCN reduced protein expression of caspase 3. Those findings suggest that DCN exerts its hepatoprotective effects through its anti-oxidant activity, modulation of TLR4/NF-κB/NLRP3 inflammatory pathways, and reduction of apoptosis. These results provide new insights into potential therapeutic strategies for managing AMIO-induced hepatotoxicity, warranting further investigation into the underlying molecular mechanisms of DCN's protective effects.
Collapse
Affiliation(s)
- Passant E Moustafa
- Department of Pharmacology, Medical Research and Clinical Studies Institute, National Research Centre, Giza, Egypt
| | - Hadir Farouk
- Department of Pharmacology, Medical Research and Clinical Studies Institute, National Research Centre, Giza, Egypt
| | - Marwa S Khattab
- Department of Pathology, Faculty of Veterinary Medicine, Cairo University, Giza, Egypt
| | - Salma A El-Marasy
- Department of Pharmacology, Medical Research and Clinical Studies Institute, National Research Centre, Giza, Egypt
| |
Collapse
|
6
|
Yi Y, Qin S, Ding S, Fang J. Polysaccharides in the medicine and food homology to combat obesity via gut-liver axis: A review of possible mechanisms. Int J Biol Macromol 2025:144044. [PMID: 40345304 DOI: 10.1016/j.ijbiomac.2025.144044] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2025] [Revised: 04/13/2025] [Accepted: 05/06/2025] [Indexed: 05/11/2025]
Abstract
Polysaccharides, as macromolecular carbohydrates present in various medicine and food homology, have gained growing recognition for their potential in combating obesity through multiple mechanisms. Their natural origin and favorable safety profile have made polysaccharides from medicine and food homology (PMFH) an area of significant research interest, particularly in the context of developing effective, safe, and sustainable interventions for obesity management. This review summarized the classification and biological properties of PMFH and then elucidated the pathological characteristics of obesity. We primarily focused on the effects of PMFHs on obesity, with particular attention to the potential mechanisms mediated through the gut-liver axis. These mechanisms encompassed the improvement of fat metabolism imbalances, manager of appetite and energy balance, adjustment of intestinal microbial imbalances, and alleviation of oxidative stress and inflammation. The findings provided critical theoretical insights and data to support the development of anti-obesity dietary and pharmaceutical products. In brief, this review outlined future research directions regarding the potential mechanisms underlying the anti-obesity effects of PMFH, particularly those involving the gut-liver axis.
Collapse
Affiliation(s)
- Yuhang Yi
- College of Bioscience and Biotechnology, Hunan Agricultural University, Hunan Engineering Laboratory for Pollution Control and Waste Utilization in Swine Production, Changsha, Hunan 410128, China
| | - Si Qin
- Laboratory of Food Function and Nutrigenomics, College of Food Science and Technology, Hunan Agricultural University, Changsha 410128, China
| | - Sujuan Ding
- College of Bioscience and Biotechnology, Hunan Agricultural University, Hunan Engineering Laboratory for Pollution Control and Waste Utilization in Swine Production, Changsha, Hunan 410128, China.
| | - Jun Fang
- College of Bioscience and Biotechnology, Hunan Agricultural University, Hunan Engineering Laboratory for Pollution Control and Waste Utilization in Swine Production, Changsha, Hunan 410128, China.
| |
Collapse
|
7
|
Yue Y, Zhang Y, Cheng Y, Jiao H, Yan M. Echinococcus granulosus antigen B regulates T-cell function through inhibition of signal transducer and activator of transcription 3 in experimental immune thrombocytopenia. Br J Haematol 2025. [PMID: 40325612 DOI: 10.1111/bjh.20064] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2024] [Accepted: 03/21/2025] [Indexed: 05/07/2025]
Abstract
Dysregulated T-cell homeostasis is central to the development of immune thrombocytopenia (ITP), characterized by reduced platelet counts. Antigen B (AgB), a key protein in Echinococcus granulosus cyst fluid, modulates T-cell differentiation and reduces inflammation. Here, we explored the role of AgB in ITP and found that it enhances the generation and function of regulatory T cells (Tregs), boosting their immunosuppressive activity. In our passive ITP murine model, AgB treatment alleviated thrombocytopenia and restored the Treg-helper T-cell (Th) balance. However, the therapeutic effects of AgB on CD4+ T cells were abolished by Treg depletion, highlighting the essential role of Tregs in AgB's mechanism of action. Moreover, AgB reduced proinflammatory cytokine production and inhibited signal transducer and activator of transcription 3 (STAT3) activation in ITP mice, with STAT3 inhibition negating the effects of AgB in Tregs. AgB promoted STAT3 degradation via tumour necrosis factor receptor-associated factor 6 (TRAF6)-mediated ubiquitination. In conclusion, by facilitating TRAF6-mediated STAT3 ubiquitination, AgB restores T-cell homeostasis and strengthens Treg immunosuppression, affording a potential therapeutic strategy for ITP.
Collapse
Affiliation(s)
- Yingbin Yue
- The First Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang, China
- State Key Laboratory of Pathogenesis, Prevention and Treatment of High Incidence Diseases in Central Asia, Xinjiang Medical University, Urumqi, China
| | - Yunfei Zhang
- The First Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang, China
| | - Yongfeng Cheng
- The First Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang, China
| | - Hongjie Jiao
- The First Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang, China
| | - Mei Yan
- The First Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang, China
| |
Collapse
|
8
|
Jin J, Yue L, Du M, Geng F, Gao X, Zhou Y, Lu Q, Pan X. Molecular Hydrogen Therapy: Mechanisms, Delivery Methods, Preventive, and Therapeutic Application. MedComm (Beijing) 2025; 6:e70194. [PMID: 40297245 PMCID: PMC12035766 DOI: 10.1002/mco2.70194] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2024] [Revised: 03/22/2025] [Accepted: 03/26/2025] [Indexed: 04/30/2025] Open
Abstract
Molecular hydrogen (H2), recognized as the smallest gas molecule, is capable of permeating cellular membranes and diffusing throughout the body. Due to its high bioavailability, H2 is considered a therapeutic gas for the treatment of various diseases. The therapeutic efficacy of hydrogen is contingent upon factors such as the administration method, duration of contact with diseased tissue, and concentration at targeted sites. H2 can be administered exogenously and is also produced endogenously within the intestinal tract. A comprehensive understanding of its delivery mechanisms and modes of action is crucial for advancing hydrogen medicine. This review highlights H₂'s mechanisms of action, summarizes its administration methods, and explores advancements in treating intestinal diseases (e.g., inflammatory bowel disease, intestinal ischemia-reperfusion, colorectal cancer). Additionally, its applications in managing other diseases are discussed. Finally, the challenges associated with its clinical application and potential solutions are explored. We propose that current delivery challenges faced by H2 can be effectively addressed through the use of nanoplatforms; furthermore, interactions between hydrogen and gut microbiota may provide insights into its mechanisms for treating intestinal diseases. Future research should explore the synergistic effects of H2 in conjunction with conventional therapies and develop personalized treatment plans to achieve precision medicine.
Collapse
Affiliation(s)
- Jiayi Jin
- School of PharmacyBinzhou Medical UniversityYantaiChina
| | - Lijun Yue
- School of PharmacyBinzhou Medical UniversityYantaiChina
| | - Maoru Du
- School of PharmacyBinzhou Medical UniversityYantaiChina
| | - Feng Geng
- School of PharmacyBinzhou Medical UniversityYantaiChina
| | - Xue Gao
- School of PharmacyBinzhou Medical UniversityYantaiChina
| | - Yuming Zhou
- Department of Laboratory MedicineYantai Affiliated Hospital of Binzhou Medical UniversityYantaiChina
| | - Qianqian Lu
- Department of OncologyYantai Affiliated Hospital of Binzhou Medical UniversityYantaiChina
| | - Xiaohong Pan
- School of PharmacyBinzhou Medical UniversityYantaiChina
| |
Collapse
|
9
|
Morais VND, Moreira LDPD, Gomes MJC, Grancieri M, Lucio HG, Toledo RCL, Mishima MDV, Costa NMB, da Silva BP, Stampini Duarte Martino H. Chia Oil ( Salvia hispanica L.) Improves the Intestinal Health of Wistar Rats Fed a Hypercaloric Diet. JOURNAL OF THE AMERICAN NUTRITION ASSOCIATION 2025; 44:322-331. [PMID: 39689242 DOI: 10.1080/27697061.2024.2431271] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/06/2024] [Revised: 10/04/2024] [Accepted: 11/13/2024] [Indexed: 12/19/2024]
Abstract
BACKGROUND A diet rich in fat and sugar is present in society everyday life, leading to the development of metabolic changes, especially in intestinal microbiota. Chia oil is a source of alpha-linolenic acid, which has antioxidant and anti-glycemic effects. Based on this, we hypothesized that chia oil may promote intestinal health. OBJECTIVE The study aims to investigate the effects of chia oil on gut microbiota and intestinal health in Wistar rats fed a high-fat and high-fructose diet (HFHF). METHODS The animals were separated into two groups and received the following diets: standard murine diet (AIN-93M) (n = 10) and HFHF (n = 20) to induce metabolic changes (phase I) during eight weeks. After that, the AIN-93M group remained unchanged, while the HFHF group was divided into two groups: HFHF (n = 10) and HFHF with chia oil (HFHF+CO) (n = 10) for ten weeks (phase II, chia oil treatment). We analyzed immunoglobulin A (IgA) levels, cecal pH, short-chain fatty acids (SCFAs), intestinal permeability, intestinal microbiome composition, histomorphometry, and murinometric parameters. RESULTS Chia oil consumption increased alpha-linolenic acid intake, IgA levels, propionic acid production, cecum weight, goblet cell number, thickness and depth of intestinal crypts, and the thickness of both circular and longitudinal muscle layers of the colon, and decreased cecal pH. No change was observed in the alpha and beta diversity between the HFHF and HFHF+CO groups. The HFHF+CO diet increased the relative abundance of genera Lactobacillus sp., Faecalibacterium sp., and Erysipelatoclostridium sp., compared to the AIN-93M group. No difference was observed in the intestinal permeability among the groups. CONCLUSION Chia oil consumption is an alternative for improving the intestinal health of rats fed a HFHF diet.
Collapse
Affiliation(s)
- Violeta Nunes de Morais
- Department of Nutrition and Health, Federal University of Viçosa, Viçosa, Minas Gerais, Brazil
| | | | | | - Mariana Grancieri
- Department of Pharmacy and Nutrition, Center for Exact, Natural and Health Sciences, Federal University of Espírito Santo, Alegre, Espírito Santo, Brazil
| | - Haira Guedes Lucio
- Department of Nutrition and Health, Federal University of Viçosa, Viçosa, Minas Gerais, Brazil
| | | | | | - Neuza Maria Brunoro Costa
- Department of Pharmacy and Nutrition, Center for Exact, Natural and Health Sciences, Federal University of Espírito Santo, Alegre, Espírito Santo, Brazil
| | | | | |
Collapse
|
10
|
Alenezi FO, Nader MA, El-Kashef DH, Abdelmageed ME. Dapansutrile mitigates concanavalin A- induced autoimmune hepatitis: Involvement of NLRP3/IL-1β and JNK/ p38 MAPK pathways. Biomed Pharmacother 2025; 186:118026. [PMID: 40164046 DOI: 10.1016/j.biopha.2025.118026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2025] [Revised: 03/25/2025] [Accepted: 03/27/2025] [Indexed: 04/02/2025] Open
Abstract
AIM Dapansutrile (Dapan) is a newly developed anti-inflammatory molecule that supresses the production of NLRP3 inflammasome-dependent IL-1β. Its hepatoprotective effects against autoimmune hepatitis (AIH) have not yet been explored. Hence, this study was conducted to examine the possible protective effects of Dapan against concanavalin A (Con A)-induced hepatitis in mice. MAIN METHODS Mice were randomly divided into five groups (n = 6): control, Con A (15 mg/kg), Dapan (60 mg/kg), Dapan (6 mg/kg) + Con A, and Dapan (60 mg/kg) + Con A. Mice were euthanised at the end of the study, and blood and hepatic tissues were collected. KEY FINDINGS Hepatic function testing using lactate dehydrogenase, alanine aminotransferase, and aspartate aminotransferase levels, in addition to hepatic tissue histological examination, revealed that intraperitoneal administration of Dapan noticeably ameliorated Con A-induced hepatic enzyme impairment and histopathological disruption. Moreover, Dapan-treated mice had significantly lower malondialdehyde hepatic content and elevated reduced glutathione, superoxide dismutase, and total antioxidant capacity levels than non-treated mice in a dose-dependent manner. The Dapan-treated groups showed significantly lower levels of the inflammatory mediators, NLRP3, TNF-α, IL-6, and IL-1β, in addition to the immunomodulators CD8, CD4, INF-γ, and NFκB and inhibition of JNK and p38 MAPK levels compared to the Con A-treated group. SIGNIFICANCE Our results showed that intraperitoneal administration of Dapan could be a therapeutic opportunity to inhibit the development of AIH via inhibition of inflammatory pathways.
Collapse
Affiliation(s)
- Fahad O Alenezi
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Mansoura University, Mansoura, Egypt; Forensic Toxicology Services Center, Ministry of health, Qassim, Saudi Arabia
| | - Manar A Nader
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Mansoura University, Mansoura, Egypt
| | - Dalia H El-Kashef
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Mansoura University, Mansoura, Egypt
| | - Marwa E Abdelmageed
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Mansoura University, Mansoura, Egypt.
| |
Collapse
|
11
|
Rust C, van den Heuvel LL, Bardien S, Carr J, Pretorius E, Seedat S, Hemmings SMJ. Association between the relative abundance of butyrate-producing and mucin-degrading taxa and Parkinson's disease. Neuroscience 2025; 576:149-154. [PMID: 40318838 DOI: 10.1016/j.neuroscience.2025.04.050] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2025] [Revised: 04/29/2025] [Accepted: 04/30/2025] [Indexed: 05/07/2025]
Abstract
Parkinson's disease (PD) is a neurodegenerative disorder characterised by motor and non-motor symptoms. Recent evidence suggests a role for gut microbiome composition and diversity in PD aetiology. This study aimed to explore the association between the gut microbiome and PD in a South African population. Gut microbial sequencing data (cases: n = 16; controls: n = 42) was generated using a 16S rRNA gene (V4) primer pair. Alpha- and beta-diversity were calculated using QIIME2, and differential abundance of taxa was evaluated using Analysis of Compositions of Microbiomes with Bias Correction (ANCOM-BC). Beta-diversity was found to differ significantly between cases and controls, with depletion in the relative abundance of Faecalibacterium, Roseburia, Dorea, and Veillonella, and enrichment of the relative abundance of Akkermansia and Victivallis. Our study found a reduction in butyrate-producing bacteria (e.g. Faecalibacterium and Roseburia) and an increase in mucin-degrading bacteria (Akkermansia) in PD cases compared to controls. These alterations might be associated with heightened gut permeability and inflammation. Longitudinal studies should address the question of whether these microbiome differences are a risk factor for, or are consequent to, the development of PD.
Collapse
Affiliation(s)
- C Rust
- Department of Psychiatry, Faculty of Medicine and Health Sciences, Stellenbosch University, Cape Town, South Africa; South African Medical Research Council / Stellenbosch University Genomics of Brain Disorders Research Unit, Stellenbosch University, Cape Town, South Africa.
| | - L L van den Heuvel
- Department of Psychiatry, Faculty of Medicine and Health Sciences, Stellenbosch University, Cape Town, South Africa; South African Medical Research Council / Stellenbosch University Genomics of Brain Disorders Research Unit, Stellenbosch University, Cape Town, South Africa
| | - S Bardien
- South African Medical Research Council / Stellenbosch University Genomics of Brain Disorders Research Unit, Stellenbosch University, Cape Town, South Africa; Division of Molecular Biology and Human Genetics, Faculty of Medicine and Health Sciences, Stellenbosch University, Cape Town, South Africa
| | - J Carr
- Department of Psychiatry, Faculty of Medicine and Health Sciences, Stellenbosch University, Cape Town, South Africa; South African Medical Research Council / Stellenbosch University Genomics of Brain Disorders Research Unit, Stellenbosch University, Cape Town, South Africa
| | - E Pretorius
- Department of Physiological Sciences, Faculty of Science, Stellenbosch University, Stellenbosch, South Africa; Department of Biochemistry and Systems Biology, Institute of Systems, Molecular and Integrative Biology Biosciences Building, University of Liverpool, Liverpool, United Kingdom
| | - S Seedat
- Department of Psychiatry, Faculty of Medicine and Health Sciences, Stellenbosch University, Cape Town, South Africa; South African Medical Research Council / Stellenbosch University Genomics of Brain Disorders Research Unit, Stellenbosch University, Cape Town, South Africa
| | - S M J Hemmings
- Department of Psychiatry, Faculty of Medicine and Health Sciences, Stellenbosch University, Cape Town, South Africa; South African Medical Research Council / Stellenbosch University Genomics of Brain Disorders Research Unit, Stellenbosch University, Cape Town, South Africa.
| |
Collapse
|
12
|
Alum EU, Uti DE, Ugwu OPC, Alum BN, Edeh FO, Ainebyoona C. Unveiling the microbial orchestra: exploring the role of microbiota in cancer development and treatment. Discov Oncol 2025; 16:646. [PMID: 40304829 PMCID: PMC12044139 DOI: 10.1007/s12672-025-02352-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/02/2025] [Accepted: 04/09/2025] [Indexed: 05/02/2025] Open
Abstract
The human microbiota comprises a diverse microbial ecosystem that significantly impacts health and disease. Among its components, the gut microbiota plays a crucial role in regulating metabolic, immunologic, and inflammatory responses. Dysbiosis, an imbalance in microbial composition, has been linked to carcinogenesis through mechanisms such as chronic inflammation, metabolic disturbances, epigenetic modifications, and immune system dysregulation. Additionally, dysbiosis influences the efficacy and toxicity of cancer therapies. Given these associations, there is growing interest in leveraging the microbiota as a biomarker for cancer detection and outcome prediction. Notably, distinct microbial signatures have been identified across various cancer types, suggesting their potential as diagnostic markers. Furthermore, modulation of the microbiota presents a promising avenue for improving cancer treatment outcomes through strategies such as antibiotics, prebiotics, probiotics, fecal microbiota transplantation, dietary interventions, small-molecule inhibitors, and phage therapy. To explore these relationships, we conducted a comprehensive literature review using Web of Science, Scopus, PubMed, MEDLINE, Embase, and Google Scholar as our primary online databases, focusing on indexed peer-reviewed articles up to the present year. This review aims to elucidate the role of dysbiosis in cancer development, examine the molecular mechanisms involved, and assess the impact of microbiota on cancer therapies. Additionally, we highlight microbiota-based therapeutic strategies and discuss their potential applications in cancer management. A deeper understanding of the intricate interplay between the microbiota and cancer may pave the way for novel approaches to cancer prevention, early detection, and treatment optimization.
Collapse
Affiliation(s)
- Esther Ugo Alum
- Department of Research and Publications, Kampala International University, Main Campus, P. O. Box 20000, Kampala, Uganda.
| | - Daniel Ejim Uti
- Department of Research and Publications, Kampala International University, Main Campus, P. O. Box 20000, Kampala, Uganda.
- Department of Biochemistry, Faculty of Basic Medical Sciences, College of Medicine, Federal University of Health Sciences, Otukpo, Benue State, Nigeria.
| | - Okechukwu Paul-Chima Ugwu
- Department of Research and Publications, Kampala International University, Main Campus, P. O. Box 20000, Kampala, Uganda
| | - Benedict Nnachi Alum
- Department of Research and Publications, Kampala International University, Main Campus, P. O. Box 20000, Kampala, Uganda
| | - Friday Ogbu Edeh
- College of Economics and Management, Kampala International University, Kampala, Uganda
| | - Christine Ainebyoona
- Faculty of Business and Management Sciences, Kampala International University, Kampala, Uganda
| |
Collapse
|
13
|
Wen L, Yang K, Wang J, Zhou H, Ding W. Gut microbiota-mitochondrial crosstalk in obesity: novel mechanistic insights and therapeutic strategies with traditional Chinese medicine. Front Pharmacol 2025; 16:1574887. [PMID: 40331200 PMCID: PMC12052897 DOI: 10.3389/fphar.2025.1574887] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2025] [Accepted: 03/24/2025] [Indexed: 05/08/2025] Open
Abstract
Obesity rates are rising globally and have become a major public health issue. Recent research emphasizes the bidirectional communication between gut microbiota and mitochondrial function in obesity development. Gut microbiota regulates energy metabolism through metabolites that impact mitochondrial processes, such as oxidative phosphorylation, biogenesis, and autophagy. In turn, alterations in mitochondrial function impact microbiota homeostasis. Traditional Chinese medicine (TCM), which encompasses TCM formulas and the metabolites of botanical drugs, employs a holistic and integrative approach that shows promise in regulating gut microbiota-mitochondrial crosstalk. This review systematically explores the intricate interactions between gut microbiota and mitochondrial function, underscoring their crosstalk as a critical mechanistic axis in obesity pathogenesis. Furthermore, it highlights the potential of TCM in developing innovative, targeted interventions, paving the way for personalized approaches in obesity treatment through the precise modulation of gut microbiota-mitochondrial interactions, offering more effective and individualized therapeutic options.
Collapse
Affiliation(s)
| | | | | | | | - Weijun Ding
- School of Basic Medical Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| |
Collapse
|
14
|
Fatima N, Fatima H, Ahmad S, Hashmi MATS, Sheikh N. Understanding the role of Hedgehog signaling pathway and gut dysbiosis in fueling liver cancer. Mol Biol Rep 2025; 52:411. [PMID: 40261446 DOI: 10.1007/s11033-025-10504-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2024] [Accepted: 04/09/2025] [Indexed: 04/24/2025]
Abstract
Liver cancer is one of the most prevalent types of cancer worldwide with less than 20% of patients surviving in the past half a decade. Several molecular pathways have been uncovered that may lead to the development of liver cancer but more recently the Hedgehog pathway (HH) and its interactions with the gut microbiota has emerged as an underlying cause of the development of liver cancer. Gut-liver axis is vital to maintaining homeostasis. The HH pathway controls cellular differentiation, proliferation, and apoptosis evasions, its abnormal activation can lead to uncontrolled proliferation of liver cancer stem cells. Additionally, the intricate interplay between HH signaling and the gut microbiota introduces a novel dimension. Recent investigations suggest that potential modulation of HH activity by gut microbiota influence HCC progression. This review explores a crosstalk between HH signaling and the gut microbiota, uncovering intricate mechanisms by which it fuels liver cancer development. This interplay provides insights into gut dysbiosis, HCC etiology and potential therapeutic avenues, highlighting the cooperative role of HH signaling and gut microbiota in shaping the overall HCC landscape.
Collapse
Affiliation(s)
- Naz Fatima
- Department of Zoology, Faculty of Science and Technology, University of Central Punjab, Lahore, Pakistan.
- Department of Internal Medicine & Gastroenterology, University of Michigan, Ann Arbor, 48109, USA.
| | - Hooriya Fatima
- Institute of Zoology, University of Punjab (Quaid-i-Azam Campus), Lahore, 54590, Pakistan
| | - Sadia Ahmad
- Institute of Zoology, University of Punjab (Quaid-i-Azam Campus), Lahore, 54590, Pakistan
| | | | - Nadeem Sheikh
- Institute of Zoology, University of Punjab (Quaid-i-Azam Campus), Lahore, 54590, Pakistan
| |
Collapse
|
15
|
Gao J, Zhang J, Tang L. The association of trimethylamine N-oxide with diabetic retinopathy Pathology: Insights from network toxicology and molecular docking analysis. Exp Eye Res 2025; 256:110399. [PMID: 40274183 DOI: 10.1016/j.exer.2025.110399] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2025] [Revised: 04/17/2025] [Accepted: 04/21/2025] [Indexed: 04/26/2025]
Abstract
Trimethylamine N-oxide (TMAO), a gut microbiota-derived metabolite, has emerged as a potential contributor to diabetic retinopathy (DR) progression. However, its molecular mechanisms in DR remain unclear. This study integrates network toxicology and multi-omics analyses to elucidate TMAO's role in DR pathogenesis. We identified TMAO-related targets through integration of CTD, SuperPred, and GeneCards databases. Differential expression analysis of DR-related genes was performed using GSE60436 and GSE102485 datasets. We intersected these with TMAO targets to identify key genes. Functional enrichment and pathway analyses were conducted, followed by immune cell infiltration assessment using ssGSEA. Machine learning algorithms (LASSO and RF) identified key marker genes, validated through GSE94019 dataset and in vitro experiments. Molecular docking explored interactions between TMAO and key proteins. We identified 45 TMAO-related targets implicated in DR. Functional analysis revealed enrichment in stress response and inflammatory pathways. Differential pathway analysis indicated significant upregulation of immune and apoptotic pathways in DR. Immune cell infiltration analysis showed increased levels of cytotoxic and inflammatory cells in DR. CASP3, CXCR4, and MAPK1 emerged as key marker genes, their expression significantly upregulated in PDR patients. Molecular docking highlighted stable interactions between TMAO and these proteins, suggesting potential modulation of their activity. TMAO-associated targets are enriched in inflammatory, oxidative, and apoptotic pathways in PDR tissues, suggesting a potential (but not causal) link to DR pathology. Our findings highlight the gut-retina axis in DR and provide a framework for targeting TMAO-mediated mechanisms in diabetic complications.
Collapse
Affiliation(s)
- Jianping Gao
- Department of Ophthalmology, The First People's Hospital of Changde City (Changde Hospital, Xiangya School of Medicine, Central South University), Changde, 415000, Hunan, China.
| | - Jian Zhang
- Department of Ophthalmology, The First People's Hospital of Changde City (Changde Hospital, Xiangya School of Medicine, Central South University), Changde, 415000, Hunan, China
| | - Lei Tang
- Department of Ophthalmology, The First People's Hospital of Changde City (Changde Hospital, Xiangya School of Medicine, Central South University), Changde, 415000, Hunan, China
| |
Collapse
|
16
|
Wang W, Zhao Y, Wang Z, Wang C, Bi L, Wang Y. Thlaspi arvense suppresses gut microbiota related TNF inflammatory pathway to alleviates ulcerative colitis. Front Immunol 2025; 16:1537325. [PMID: 40330488 PMCID: PMC12053237 DOI: 10.3389/fimmu.2025.1537325] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2024] [Accepted: 03/19/2025] [Indexed: 05/08/2025] Open
Abstract
Introduction Thlaspi arvense (TA), commonly known as "Ximi" or "Subaijiang," is a traditional Chinese medicinal herb used to prevent and treat ulcerative colitis (UC). However, the precise mechanisms underlying its therapeutic effects remain unclear, necessitating further investigation to identify potential pharmaceutical applications for UC management. This study aims to elucidate the efficacy and mechanisms of TA and its active constituents in UC treatment. Methods This study first evaluated the effects of varying TA doses on 3% dextran sulfate sodium (DSS)-induced UC. Gut microbiota alterations in UC mice were analyzed via 16S rRNA sequencing, with correlation analyses to reveal the relationship between gut microbiota and cytokines. Then, network pharmacology was utilized to identified potential TA targets for UC treatment. Protein-protein interaction (PPI) networks, Gene Ontology (GO), and Kyoto Encyclopedia of Genes and Genomes (KEGG) enrichment analyses were employed to explore TA's mechanisms. Molecular docking and dynamics simulations validated interactions between TA's active compounds and UC-related targets. Finally, TNF pathway modulation by TA and its active component, isovitexin, was verified in vitro and in vivo. Results TA alleviated DSS-induced weight loss in a dose-dependent manner, reduced disease activity indices, and preserved intestinal mucosal barrier integrity. Subsequently, fluorescence in situ hybridization (FISH) revealed TA suppressed microbial translocation in intestinal tissues. To further characterize inflammatory responses, ELISA demonstrated that TA modulated levels of key cytokines (TNF-α, IL-1β, IL-6, IL-10) and oxidative stress markers (SOD, MDA), indicating systemic anti-inflammatory effects. Building on these findings, 16S rRNA sequencing analyses showed that TA regulated gut microbiota alpha/beta diversity and inhibited infectious disease-related pathways. Notably, correlation heatmaps highlighted a strong association between TNF-α levels and Escherichia-Shigella abundance, with high-dose TA significantly reducing this pathogenic bacterial genus. To systematically explore molecular mechanisms, network pharmacology identified 220 potential TA targets for UC treatment. Consistent with experimental data, PPI and KEGG analyses implicated TNF-α, IL-6, and AKT as key targets, primarily through TNF signaling pathway modulation. To validate these predictions, molecular docking confirmed stable interactions between TA compounds and identified targets, while dynamics simulations specifically emphasized isovitexin's high affinity for TNF-α. Finally, experiments in vivo demonstrated TA's inhibition of TNF-α-mediated NF-κB pathway activation, and in vitro studies confirmed that isovitexin directly mitigated TNF-α-induced intestinal epithelial damage. Furthermore, TA demonstrated potent inhibition of TNF-α-mediated NF-κB inflammatory pathway activation in intestinal tissues, while its active constituent isovitexin effectively mitigated TNF-α-induced epithelial cell damage, collectively highlighting their complementary anti-inflammatory mechanisms. Discussion Collectively, Thlaspi arvense (TA) ameliorates ulcerative colitis through synergistic mechanisms involving gut microbiota modulation, inflammatory pathway suppression, and intestinal barrier preservation. By remodeling microbial communities to reduce Escherichia-Shigella colonization and microbial translocation. TA concurrently inhibits TNF-α/NF-κB-driven inflammation, and oxidative stress regulation. Furthermore, its active constituent isovitexin directly attenuates TNF-α-induced epithelial damage, demonstrating multi-scale therapeutic efficacy. These findings establish TA's multi-target pharmacology spanning host-microbe interactions and intracellular signaling, while providing a rationale for standardizing TA-based formulations and advancing isovitexin as a precision therapeutic agent for inflammatory bowel diseases.
Collapse
MESH Headings
- Animals
- Colitis, Ulcerative/drug therapy
- Colitis, Ulcerative/metabolism
- Colitis, Ulcerative/chemically induced
- Colitis, Ulcerative/microbiology
- Colitis, Ulcerative/immunology
- Gastrointestinal Microbiome/drug effects
- Mice
- Tumor Necrosis Factor-alpha/metabolism
- Drugs, Chinese Herbal/pharmacology
- Dextran Sulfate
- Signal Transduction/drug effects
- Disease Models, Animal
- Male
- Molecular Docking Simulation
- Mice, Inbred C57BL
- Anti-Inflammatory Agents/pharmacology
- Protein Interaction Maps
- RNA, Ribosomal, 16S/genetics
Collapse
Affiliation(s)
- Wenkai Wang
- Department of Oncology, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Yiyang Zhao
- Department of Oncology, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Ziwei Wang
- Department of Oncology, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Chaowei Wang
- Department of Oncology, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Ling Bi
- Department of Oncology, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Yan Wang
- Department of Oncology, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
- The Second Clinical Medical College of Guizhou University of Traditional Chinese Medicine, Guizhou, China
| |
Collapse
|
17
|
Feng J, Wang JP, Hu JR, Li P, Lv P, He HC, Cheng XW, Cao Z, Han JJ, Wang Q, Su Q, Liu LX. Multi-omics reveals the associations among the fecal metabolome, intestinal bacteria, and serum indicators in patients with hepatocellular carcinoma. World J Gastroenterol 2025; 31:104996. [PMID: 40309232 PMCID: PMC12038548 DOI: 10.3748/wjg.v31.i15.104996] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/09/2025] [Revised: 02/18/2025] [Accepted: 03/24/2025] [Indexed: 04/18/2025] Open
Abstract
BACKGROUND Hepatocellular carcinoma (HCC), the predominant form of primary liver cancer, is a key contributor to cancer-related deaths globally. However, HCC diagnosis solely based on blood biochemical markers lacks both sensitivity and specificity. AIM To investigate alterations of the fecal metabolome and intestinal bacteria and reveal the correlations among differential metabolites, distinct bacteria, and serum indicators. METHODS To uncover potentially effective therapeutic targets for HCC, we utilized non-targeted liquid chromatography-mass spectrometry and high-throughput DNA sequencing targeting the 16S rRNA gene. This comprehensive approach allowed us to investigate the metabolome and microbial community structure of feces samples obtained from patients with HCC. Furthermore, we conducted an analysis to assess the interplay between the fecal metabolome and intestinal bacterial population. RESULTS In comparison to healthy controls, a notable overlap of 161 differential metabolites and 3 enriched Kyoto Encyclopedia of Genes and Genomes pathways was observed in the HCC12 (comprising patients with stage I and II HCC) and HCC34 groups (comprising patients with stage III and IV HCC). Lachnospira, Streptococcus, and Veillonella had significant differences in abundance in patients with HCC. Notably, Streptococcus and Veillonella exhibited significant correlations with serum indicators such as alpha-fetoprotein (AFP). Meanwhile, several differential metabolites [e.g., 4-keto-2-undecylpyrroline, dihydrojasmonic acid, 1,8-heptadecadiene-4,6-diyne-3,10-diol, 9(S)-HOTrE] also exhibited significant correlations with serum indicators such as γ-glutamyl transferase, total bilirubin, AFP, aspartate aminotransferase, and albumin. Additionally, these two genera also had significant associations with differential metabolites such as 1,2-Dipentadecanoyl-rac-glycerol (15:0/20:0/0:0), arachidoyl ethanolamide, and 4-keto-2-undecylpyrroline. CONCLUSION Our results suggest that the metabolome of fecal samples and the composition of intestinal bacteria hold promise as potential biomarkers for HCC diagnosis.
Collapse
Affiliation(s)
- Jing Feng
- Department of Gastroenterology, The First Hospital of Shanxi Medical University, Taiyuan 030001, Shanxi Province, China
- Department of Infectious Diseases and Hepatology, Shanxi Provincial People’s Hospital, Affiliated to Shanxi Medical University, Taiyuan 030012, Shanxi Province, China
| | - Jun-Ping Wang
- Department of Gastroenterology, Shanxi Provincial People’s Hospital, Affiliated to Shanxi Medical University, Taiyuan 030012, Shanxi Province, China
| | - Jian-Ran Hu
- Department of Biological Science and Technology, Jinzhong University, Jinzhong 030619, Shanxi Province, China
| | - Ping Li
- Department of Biological Science and Technology, Jinzhong University, Jinzhong 030619, Shanxi Province, China
| | - Pin Lv
- Department of Gastroenterology, Shanxi Provincial People’s Hospital, Affiliated to Shanxi Medical University, Taiyuan 030012, Shanxi Province, China
| | - Hu-Cheng He
- Department of Gastroenterology, Shanxi Provincial People’s Hospital, Affiliated to Shanxi Medical University, Taiyuan 030012, Shanxi Province, China
| | - Xiao-Wei Cheng
- Department of Interventional Therapy, Shanxi Provincial People’s Hospital, Affiliated to Shanxi Medical University, Taiyuan 030012, Shanxi Province, China
| | - Zheng Cao
- Department of Gastroenterology, Shanxi Provincial People’s Hospital, Affiliated to Shanxi Medical University, Taiyuan 030012, Shanxi Province, China
| | - Jia-Jing Han
- Department of Gastroenterology, Shanxi Provincial People’s Hospital, Affiliated to Shanxi Medical University, Taiyuan 030012, Shanxi Province, China
| | - Qiang Wang
- Department of Infectious Diseases and Hepatology, Shanxi Provincial People’s Hospital, Affiliated to Shanxi Medical University, Taiyuan 030012, Shanxi Province, China
| | - Qian Su
- Department of Infectious Diseases and Hepatology, Shanxi Provincial People’s Hospital, Affiliated to Shanxi Medical University, Taiyuan 030012, Shanxi Province, China
| | - Li-Xin Liu
- Department of Gastroenterology, The First Hospital of Shanxi Medical University, Taiyuan 030001, Shanxi Province, China
| |
Collapse
|
18
|
Zaher A, Moura Nascimento Santos MJ, Elsaygh H, Peterson SJ, Colli Cruz C, Thomas AS, Wang Y. Management of refractory checkpoint inhibitor-induced colitis. Expert Opin Drug Saf 2025:1-10. [PMID: 40251944 DOI: 10.1080/14740338.2025.2496431] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2025] [Revised: 04/06/2025] [Accepted: 04/17/2025] [Indexed: 04/21/2025]
Abstract
INTRODUCTION This review discusses the epidemiology, pathophysiology, and factors associated with refractory immune-mediated diarrhea and colitis (r-IMDC), emphasizing tailored treatment strategies. AREAS COVERED The current literature on r-IMDC was reviewed using PubMed (2015-2025), focusing on clinical trials, meta-analyses, and case reports relevant to its management. EXPERT OPINION Effectively managing r-IMDC is crucial for balancing toxicities and antitumor response. Available second and third-line management options for r-IMDC cases must be carefully evaluated. Future perspectives include development of standardized protocols beyond second-line therapies and predictive biomarkers to enable personalized treatment.
Collapse
Affiliation(s)
- Anas Zaher
- Department of Internal Medicine, New York Presbyterian - Brooklyn Methodist/Weill Cornell Medicine, Brooklyn, NY, USA
| | | | - Hassan Elsaygh
- Department of Internal Medicine, University of Debrecen, Debrecen, Hungary
| | - Stephen J Peterson
- Department of Internal Medicine, New York Presbyterian - Brooklyn Methodist/Weill Cornell Medicine, Brooklyn, NY, USA
| | - Carolina Colli Cruz
- Department of Gastroenterology, Hepatology & Nutrition, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Anusha Shirwaikar Thomas
- Department of Gastroenterology, Hepatology & Nutrition, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Yinghong Wang
- Department of Gastroenterology, Hepatology & Nutrition, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| |
Collapse
|
19
|
Xu Y, Dong B, Tang Y, Jiang Y, Huang T, Jiang F, Xing W, Chen J, Zhu F. Association between thyroid stimulating hormone levels and nonproliferative diabetic retinopathy: a cross-sectional study. BMC Endocr Disord 2025; 25:106. [PMID: 40259281 PMCID: PMC12010677 DOI: 10.1186/s12902-025-01928-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/14/2024] [Accepted: 04/08/2025] [Indexed: 04/23/2025] Open
Abstract
BACKGROUND The association between thyroid-stimulating hormone (TSH) and type 2 diabetes mellitus (T2DM) is well known. However, whether TSH is related to nonproliferative diabetic retinopathy (NPDR) has not been studied. This study aimed to explore the relationship between TSH and NPDR in Chinese patients with T2DM. METHODS In this cross-sectional study, 427 patients with T2DM were enrolled. The individuals were classified into two groups according to the fundus oculi examination: the non-diabetic retinopathy (NDR) group (n = 224) and the non-proliferative diabetic retinopathy (NPDR) group (n = 203). The individuals' demographic and clinical data were collected by reviewing medical records and direct interviews. The demographic data and biochemical parameters were compared between groups using the Student's t - test or the Mann‒Whitney U test, anthropometric measurements, thyroid function, and NPDR were evaluated, and the associations between TSH and NPDR were assessed using logistic regression models. RESULTS No significant differences in age, sex, body mass index (BMI), incidence of alcohol consumption, and duration of diabetes were found between these two groups. The systolic blood pressure (SBP), incidence of smoking, TSH, blood urea nitrogen (BUN), and urinary micro-albumin (mALB) were significantly higher in the NPDR group than in the NDR group (P < 0.05). Individuals in the NDR group had higher levels of thyroxine (T4), glutamic pyruvic transaminase (ALT), fasting C-peptide (FCP), and 2-hour C-peptide (2hCP) than individuals in the NPDR group (P < 0.05). Spearman's correlation analysis revealed that the serum TSH levels were negatively associated with the HbA1c levels in all patients (r=-0.11, P < 0.05). Serum TSH levels were negatively correlated with HbA1c levels (r = -0.19, P < 0.01) and positively correlated with diabetes duration (r = 0.14, P < 0.05) in the NPDR group. Multivariate logistic regression analysis revealed that high TSH levels, sex, diabetes duration, high-density lipoprotein cholesterol (HDL-C), glycosylated hemoglobin (HbA1c), FCP, and SBP were associated with NPDR [odds ratio (OR) > 1, P < 0.05]. Receiver operating characteristic curve analysis revealed that the optimal cutoff point of TSH for predicting NPDR was 2.235 mIU/L. CONCLUSION The TSH level is independently associated with NPDR in the Chinese population with T2DM. A high serum TSH level may be a potential risk factor for NPDR and an indicator for screening for diabetic microangiopathy. TRIAL REGISTRATION This study is registered with the Chinese Clinical Trial Registry (02/21/2025 ChiCTR2500097614).
Collapse
Affiliation(s)
- Yiqi Xu
- Department of Endocrinology and Genetic Metabolism, The First Affiliated Hospital of Anhui Health College (the Second People's Hospital of Chizhou), Chizhou, Anhui, China
| | - Biwu Dong
- Department of Cardiology, The First Affiliated Hospital of Anhui Health College (the Second People's Hospital of Chizhou), Chizhou, Anhui, China
| | - Youyun Tang
- Department of Endocrinology and Genetic Metabolism, The First Affiliated Hospital of Anhui Health College (the Second People's Hospital of Chizhou), Chizhou, Anhui, China
| | - Yan Jiang
- Department of Endocrinology and Genetic Metabolism, The First Affiliated Hospital of Anhui Health College (the Second People's Hospital of Chizhou), Chizhou, Anhui, China
| | - Tingting Huang
- Department of Endocrinology and Genetic Metabolism, The First Affiliated Hospital of Anhui Health College (the Second People's Hospital of Chizhou), Chizhou, Anhui, China
| | - Feng Jiang
- Department of Endocrinology and Genetic Metabolism, The First Affiliated Hospital of Anhui Health College (the Second People's Hospital of Chizhou), Chizhou, Anhui, China
| | - Wei Xing
- Department of Endocrinology and Genetic Metabolism, The First Affiliated Hospital of Anhui Health College (the Second People's Hospital of Chizhou), Chizhou, Anhui, China
| | - Junsheng Chen
- Department of Endocrinology and Genetic Metabolism, The First Affiliated Hospital of Anhui Health College (the Second People's Hospital of Chizhou), Chizhou, Anhui, China
| | - Fengping Zhu
- Department of Endocrinology and Genetic Metabolism, The First Affiliated Hospital of Anhui Health College (the Second People's Hospital of Chizhou), Chizhou, Anhui, China.
| |
Collapse
|
20
|
Thonusin C, Suparan K, Kunasol C, Lungruammit N, Nawara W, Arunsak B, Kerdphoo S, Kongkaew A, Songtrai S, Pintana H, Maneechote C, Pratchayasakul W, Kaewsuwan S, Chattipakorn N, Chattipakorn SC. Interruptins Extracted from Cyclosorus terminans Protect Gut Pathologies Induced by High-Fat Diet in Rats. Nutrients 2025; 17:1387. [PMID: 40284250 PMCID: PMC12030309 DOI: 10.3390/nu17081387] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2025] [Revised: 04/16/2025] [Accepted: 04/19/2025] [Indexed: 04/29/2025] Open
Abstract
BACKGROUND/OBJECTIVES The fern "Cyclosorus terminans" (C. terminans) or "Maiden Fern" contains interruptin A and interruptin B. This plant could attenuate obesity, insulin resistance, and fatty liver in rats fed a high-fat/calorie diet. However, the benefits of C. terminans to the gut remain unknown. We investigated the protective effect of C. terminans extract against gut dysfunction in rats exposed to a high-fat/calorie diet. METHODS Male Wistar rats were assigned to receive either (1) a normal diet treated with vehicle, (2) a high-fat/calorie diet treated with vehicle, (3) a high-fat/calorie diet treated with 100 mg per kg per day (mg·kg-1·day-1) of C. terminans extract, or (4) a high-fat/calorie diet treated with 200 mg·kg-1·day-1 of C. terminans extract. The rats were euthanized after 12 weeks of treatment to enable feces and colon tissue collection. RESULTS Both 100 and 200 mg·kg-1·day-1 of C. terminans extract reduced body weight (-10.49%; p = 0.030 and -10.54%; p = 0.037, respectively) and ameliorated gut inflammation, gut barrier disruption, changes in short-chain fatty acid levels, and gut dysbiosis caused by high-fat/calorie diet. CONCLUSIONS C. terminans extract attenuated an increase in body weight and exerted prophylactic effects against gut pathologies induced by high-fat/calorie diet.
Collapse
Affiliation(s)
- Chanisa Thonusin
- Cardiac Electrophysiology Unit, Department of Physiology, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand; (C.T.); (W.P.); (N.C.)
- Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand; (K.S.); (C.K.); (W.N.); (B.A.); (S.K.); (H.P.); (C.M.)
- Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, Chiang Mai 50200, Thailand
| | - Kanokphong Suparan
- Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand; (K.S.); (C.K.); (W.N.); (B.A.); (S.K.); (H.P.); (C.M.)
- Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, Chiang Mai 50200, Thailand
| | - Chanon Kunasol
- Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand; (K.S.); (C.K.); (W.N.); (B.A.); (S.K.); (H.P.); (C.M.)
- Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, Chiang Mai 50200, Thailand
| | | | - Wichwara Nawara
- Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand; (K.S.); (C.K.); (W.N.); (B.A.); (S.K.); (H.P.); (C.M.)
- Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, Chiang Mai 50200, Thailand
| | - Busarin Arunsak
- Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand; (K.S.); (C.K.); (W.N.); (B.A.); (S.K.); (H.P.); (C.M.)
- Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, Chiang Mai 50200, Thailand
| | - Sasiwan Kerdphoo
- Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand; (K.S.); (C.K.); (W.N.); (B.A.); (S.K.); (H.P.); (C.M.)
- Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, Chiang Mai 50200, Thailand
| | - Aphisek Kongkaew
- Research Administration Section, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand;
| | - Sujinda Songtrai
- Faculty of Medical Technology, Rangsit University, Pathumthani 12000, Thailand;
| | - Hiranya Pintana
- Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand; (K.S.); (C.K.); (W.N.); (B.A.); (S.K.); (H.P.); (C.M.)
- Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, Chiang Mai 50200, Thailand
| | - Chayodom Maneechote
- Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand; (K.S.); (C.K.); (W.N.); (B.A.); (S.K.); (H.P.); (C.M.)
- Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, Chiang Mai 50200, Thailand
| | - Wasana Pratchayasakul
- Cardiac Electrophysiology Unit, Department of Physiology, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand; (C.T.); (W.P.); (N.C.)
- Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand; (K.S.); (C.K.); (W.N.); (B.A.); (S.K.); (H.P.); (C.M.)
- Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, Chiang Mai 50200, Thailand
| | - Sireewan Kaewsuwan
- Department of Pharmacognosy and Pharmaceutical Botany, Faculty of Pharmaceutical Sciences, Prince of Songkhla University, Songkhla 90110, Thailand;
- Phytomedicine and Pharmaceutical Biotechnology Excellence Center, Faculty of Pharmaceutical Sciences, Prince of Songkhla University, Songkhla 90110, Thailand
| | - Nipon Chattipakorn
- Cardiac Electrophysiology Unit, Department of Physiology, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand; (C.T.); (W.P.); (N.C.)
- Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand; (K.S.); (C.K.); (W.N.); (B.A.); (S.K.); (H.P.); (C.M.)
- Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, Chiang Mai 50200, Thailand
| | - Siriporn C. Chattipakorn
- Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand; (K.S.); (C.K.); (W.N.); (B.A.); (S.K.); (H.P.); (C.M.)
- Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, Chiang Mai 50200, Thailand
- Department of Oral Biology and Diagnostic Sciences, Faculty of Dentistry, Chiang Mai University, Chiang Mai 50200, Thailand
| |
Collapse
|
21
|
Sun X, Li W, Chen G, Hu G, Jia J. Faecalibacterium duncaniae Mitigates Intestinal Barrier Damage in Mice Induced by High-Altitude Exposure by Increasing Levels of 2-Ketoglutaric Acid. Nutrients 2025; 17:1380. [PMID: 40284246 PMCID: PMC12030221 DOI: 10.3390/nu17081380] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2025] [Revised: 04/06/2025] [Accepted: 04/17/2025] [Indexed: 04/29/2025] Open
Abstract
Background/Objectives: Exposure to high altitudes often results in gastrointestinal disorders. This study aimed to identify probiotic strains that can alleviate such disorders. Methods: We conducted a microbiome analysis to investigate the differences in gut microbiota among volunteers during the acute response and acclimatization phases at high altitudes. Subsequently, we established a mouse model of intestinal barrier damage induced by high-altitude exposure to further investigate the roles of probiotic strains and 2-ketoglutaric acid. Additionally, we performed untargeted metabolomics and transcriptomic analyses to elucidate the underlying mechanisms. Results: The microbiome analysis revealed a significant increase in the abundance of Faecalibacterium prausnitzii during the acclimatization phase. Faecalibacterium duncaniae (F. duncaniae) significantly mitigated damage to the intestinal barrier and the reduction of 2-ketoglutaric acid levels in the cecal contents induced by high-altitude exposure in mice. Immunohistochemistry and TUNEL staining demonstrated that high-altitude exposure significantly decreased the expression of ZO-1 and occludin while increasing apoptosis in ileal tissues. In contrast, treatment with F. duncaniae alleviated the loss of ZO-1 and occludin, as well as the apoptosis induced by high-altitude exposure. Furthermore, 2-ketoglutaric acid also mitigated this damage, reducing the loss of occludin and apoptosis in mice. Transcriptomic analysis indicated that high-altitude exposure significantly affects the calcium signaling pathway; conversely, the administration of F. duncaniae significantly influenced the PPAR signaling pathway, mineral absorption, and the regulation of lipolysis in adipocytes. Additionally, the expression of the FBJ osteosarcoma oncogene (Fos) was markedly reduced following the administration of F. duncaniae. Conclusions:F. duncaniae mitigates hypoxia-induced intestinal barrier damage by increasing levels of 2-ketoglutaric acid and shows promise as a probiotic, ultimately aiding travelers in adapting to high-altitude environments.
Collapse
Affiliation(s)
| | | | | | - Gaosheng Hu
- School of Traditional Chinese Materia Medica, Shenyang Pharmaceutical University, Shenyang 110016, China; (X.S.); (W.L.); (G.C.)
| | - Jingming Jia
- School of Traditional Chinese Materia Medica, Shenyang Pharmaceutical University, Shenyang 110016, China; (X.S.); (W.L.); (G.C.)
| |
Collapse
|
22
|
Gong Q, Zhu T, Zhang L, Wu H, Miao Y, Hu Y. Synthesis of AgInS 2 quantum dots loaded with celastrol for induction of apoptosis and autophagy in hepatocellular carcinoma cells. Discov Oncol 2025; 16:538. [PMID: 40238029 PMCID: PMC12003245 DOI: 10.1007/s12672-025-02332-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/26/2025] [Accepted: 04/08/2025] [Indexed: 04/18/2025] Open
Abstract
Hepatocellular carcinoma (HCC) is a predominant form of liver cancer and one of the leading causes of cancer-related death globally. Therefore, there is an urgent need for innovative therapeutic strategies that target the molecular mechanisms underlying HCC progression and metastasis, aiming to improve treatment efficacy and patient survival. The natural product celastrol (Cel) has demonstrated inhibitory effects in various cancer cell lines. However, its clinical application has been hindered by high toxicity and a low safety threshold. Metal-free quantum dots (QDs), AgInS2 (AIS QDs) not only eliminate toxic risks associated with heavy metals but also exhibit high biocompatibility in the biomedical field. By developing AIS QD@Cel, an AIS QDs nano-delivery system for Cel, the cell selectivity and inhibitory effects of Cel on HCC were enhanced. Fourier-transform infrared spectroscopy (FTIR) analysis revealed that AIS QDs can interact with Cel via amide bonds. The encapsulation rate of AIS QDs to Cel reached 27.5%. AIS QD@Cel eliminated toxicity on 293T and enhanced inhibition on HCC cells by over 10 times. Furthermore, the western blotting and flow cytometry experiments showed that AIS QD@Cel promoted apoptosis and autophagy signal pathway. Finally, transcriptome sequencing revealed that AIS QD@Cel effect on HCC by regulating gene expression involved in critical signaling pathways that are implicated in the progression of cancer. This strategy holds the potential to increase safety threshold and clinical applicability of Cel, offering significant clinical value for the treatment of HCC patients.
Collapse
Affiliation(s)
- Qineng Gong
- Medical Research Center, Affiliated Hospital 2, Nantong University, Nantong, 226001, People's Republic of China
- Jiangsu Provincial Medical Key Discipline (Laboratory) Cultivation Unit, Medical Research Center, Nantong First People's Hospital, Nantong, 226001, People's Republic of China
| | - Tianyu Zhu
- Medical Research Center, Affiliated Hospital 2, Nantong University, Nantong, 226001, People's Republic of China
- Jiangsu Provincial Medical Key Discipline (Laboratory) Cultivation Unit, Medical Research Center, Nantong First People's Hospital, Nantong, 226001, People's Republic of China
| | - Linlin Zhang
- Department of Pharmacology, The First People's Hospital of Yancheng, Yancheng, 224000, People's Republic of China
- Department of Pharmacology, The Fourth Affiliated Hospital of Nantong University, Yancheng, 224000, People's Republic of China
| | - Hui Wu
- Department of Pharmacology, The First People's Hospital of Yancheng, Yancheng, 224000, People's Republic of China
- Department of Pharmacology, The Fourth Affiliated Hospital of Nantong University, Yancheng, 224000, People's Republic of China
| | - Yang Miao
- Department of Pharmacology, The First People's Hospital of Yancheng, Yancheng, 224000, People's Republic of China.
- Department of Pharmacology, The Fourth Affiliated Hospital of Nantong University, Yancheng, 224000, People's Republic of China.
| | - Ye Hu
- Department of Pharmacology, The First People's Hospital of Yancheng, Yancheng, 224000, People's Republic of China.
- Department of Pharmacology, The Fourth Affiliated Hospital of Nantong University, Yancheng, 224000, People's Republic of China.
| |
Collapse
|
23
|
Qu Q, Liu M, Hu Y, Huang G, Xuan Z, Lun J, Chen X, Lv W, Guo S. Modulatory effects of polyherbal mixture on the immuno-antioxidant capacity and intestinal health of chicks infected with Escherichia coli O78. Poult Sci 2025; 104:105156. [PMID: 40239311 PMCID: PMC12032338 DOI: 10.1016/j.psj.2025.105156] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2025] [Revised: 04/02/2025] [Accepted: 04/11/2025] [Indexed: 04/18/2025] Open
Abstract
A total of 180 one-day-old white-feathered broiler chicks were selected and randomly divided into 4 treatments, namely the control group (CON), Escherichia coli groups (E. coli), 2 g/kg polyherbal mixture group (PHM2), and the 4 g/kg polyherbal mixture group (PHM4). The CON and E. coli groups were fed a basal diet, while the PHM2 and PHM4 groups were fed the basal diet supplemented with 2 g/kg and 4 g/kg PHM, respectively. Each group had 3 replicates, with 15 broilers per replicate. On day 17 of the experiment, broilers in the E. coli, PHM2, and PHM4 groups were intraperitoneally injected with 0.8 mL of 1 × 108 CFU/mL of E. coli O78. Broilers in the control group received an equivalent volume of saline. Chicks were euthanized 48 h postinjection for collecting serum, liver, spleen, jejunum, ileum, ileal mucosa, and cecal contents. Our results showed that PHM significantly reversed the weight loss and decreased the diarrhea rate and the mortality of chicks caused by E. coli infection (P < 0.05). In the serum of chicks infected with E. coli, PHM significantly enhanced the antioxidant capacity (P < 0.05), increased the levels of immunoglobulins and anti-inflammatory cytokines (P < 0.05), and decreased the concentrations of proinflammatory cytokines (P < 0.05). Meanwhile, PHM also promoted the mRNA expression of antioxidant-related genes and decreased the expression of proinflammatory cytokines and apoptosis-related genes in the liver, spleen, jejunum, and ileum (P < 0.05). In addition, PHM repaired the intestinal barrier and injury to further reduce the serum concentrations of d-lactate (DAO) and lipopolysaccharide (LPS) (P < 0.05). More importantly, PHM significantly regulated the composition of cecal microbiota, especially by up-regulating the relative abundance of beneficial bacteria, including Faecalibacterium, Bacteroides, Butyricicoccus, and Lactobacillus, and down-regulating the relative abundance of pathogenic bacteria, including Enterococcus, Escherichia, and Shigella (P < 0.05). These beneficial bacteria were significantly positively correlated with antioxidant capacity and intestinal barrier function, while pathogenic bacteria were significantly positively correlated with proinflammatory cytokines (P < 0.05). In conclusion, PHM may be a potential preventive strategy for E. coli-infected poultry, which is closely related to its modulation of gut microbiota.
Collapse
Affiliation(s)
- Qian Qu
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
| | - Mengjie Liu
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, China; School of Animal Science and Technology, Foshan University, Foshan, China
| | - Yifan Hu
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
| | - Gengxiong Huang
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
| | - Zhaoying Xuan
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
| | - Jianchi Lun
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
| | - Xiaoli Chen
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
| | - Weijie Lv
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, China; Guangdong Technology Research Center for Traditional Chinese Veterinary Medicine and Natural Medicine, Guangzhou, China
| | - Shining Guo
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, China; Guangdong Technology Research Center for Traditional Chinese Veterinary Medicine and Natural Medicine, Guangzhou, China.
| |
Collapse
|
24
|
Shimokawa C, Mizutani W, Motegi H, Gokan N, Tomita J, Hisaeda H. Prebiotic Effects of Insoluble Konjac Glucomannan Derived from Edible "Konnyaku" on Weight Control. Microorganisms 2025; 13:877. [PMID: 40284712 PMCID: PMC12029870 DOI: 10.3390/microorganisms13040877] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2025] [Revised: 04/04/2025] [Accepted: 04/04/2025] [Indexed: 04/29/2025] Open
Abstract
Obesity is a major global health issue, and novel dietary approaches are needed for prevention and management. This study investigates the effect of insoluble konjac glucomannan (iKGM) derived from edible konnyaku, a traditional Japanese food, on weight gain suppression in mice. Mice treated with iKGM showed increased fecal volume, reduced food intake, and suppressed weight gain (Day 21; p < 0.01). This weight-suppression effect was prebiotic rather than physical properties of iKGM, as antibiotic treatment abolished the weight-suppressing effect despite increased fecal volume. iKGM treatment altered the gut microbiota, notably increasing Akkermansia muciniphila (Day 21; p < 0.01), a bacterium associated with weight loss, along with elevated levels of short-chain fatty acids (SCFAs) such as butyrate and propionate (Day 21; p < 0.01). Furthermore, iKGM-induced weight suppression was linked to elevated leptin levels (Day 21; p < 0.01), an appetite suppressant induced by SCFAs. These results suggest that iKGM modulates gut microbiota, increases A. muciniphila, induces leptin production, and reduces food intake, inhibiting weight gain. This study indicates that iKGM may represent a promising approach for obesity prevention through gut microbiota modulation. Future research should investigate the mechanisms of iKGM's effects on microbiota and explore its long-term safety and efficacy in clinical trials.
Collapse
Affiliation(s)
- Chikako Shimokawa
- Department of Parasitology, National Institute of Infectious Diseases, 1-23-1 Toyama, Shinjuku 162-8640, Tokyo, Japan;
| | - Wakana Mizutani
- Department of Parasitology, Graduate School of Medical Science, Gunma University, 3-39-15 Showa, Maebashi 371-8511, Gunma, Japan;
| | - Haruhisa Motegi
- ORIHIRO Plantdew Co., Ltd., 613 Shimooshima-machi, Takasaki 370-0886, Gunma, Japan; (H.M.); (N.G.); (J.T.)
| | - Naomi Gokan
- ORIHIRO Plantdew Co., Ltd., 613 Shimooshima-machi, Takasaki 370-0886, Gunma, Japan; (H.M.); (N.G.); (J.T.)
| | - Junichi Tomita
- ORIHIRO Plantdew Co., Ltd., 613 Shimooshima-machi, Takasaki 370-0886, Gunma, Japan; (H.M.); (N.G.); (J.T.)
| | - Hajime Hisaeda
- Department of Parasitology, National Institute of Infectious Diseases, 1-23-1 Toyama, Shinjuku 162-8640, Tokyo, Japan;
| |
Collapse
|
25
|
Wu Y, Mo J, Wang Q, Li J, Wei J, Zhang N, Dong Y, Zhu X, Lu T, Huang S. Microbiome and metabolome explain the high-fat diet-induced diabetes development and diabetes resistance in Guizhou mini-pigs. Front Microbiol 2025; 16:1555069. [PMID: 40291804 PMCID: PMC12023756 DOI: 10.3389/fmicb.2025.1555069] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2025] [Accepted: 03/18/2025] [Indexed: 04/30/2025] Open
Abstract
Type 2 diabetes mellitus (T2DM) is an obesity-related disease claiming substantial global mortality annually. Current animal models of T2DM remain limited, with low success rates in establishing porcine models of high-fat diet (HFD)-induced T2DM. Our experimental design employed 35 Guizhou mini-pigs to develop a T2DM model via HFD induction, aiming to identify microbial and metabolic signatures associated with disease pathogenesis and resistance. At month 10, five individuals from the control (CTR), T2DM (DM), and T2DM resistant (anti-DM) groups were slaughtered, samples were collected, and relevant indices were measured. Metagenomics, metabolomics, and 16S rRNA sequencing were performed to identify microbes and metabolites linked to T2DM progression and resistance. Key findings demonstrated anti-DM group parameters-including metabolic indices (fasting blood glucose, insulin levels, HbA1c, IVGTT), histopathology (HE-stained pancreatic/hepatic tissues), microbial profiles (structural, compositional, functional), and metabolomic signatures-occupied intermediate positions between CTR and DM groups. Network analyses revealed: (1) Lactobacillus, L. amylovorus, fingolimod, polyoxyethylene sorbitan monooleate, thiamine, and atrazine in HFD-associated networks; (2) Limosilactobacillus reuteri, N-oleoyl-L-serine, tolbutamide, tetradecanoyl carnitine, 3'-sulfogalactosylceramide, and guggulsterone in T2DM resistance networks; (3) Ruminococcaceae NK4A214 group, diethyl phthalate, zingerone, enalapril, 5-hydroxytryptophol, 2'-deoxyinosine, icariin, and emetine in T2DM progression networks. These results further clarify the role of the gut microbiota and serum metabolites in the development of T2DM in the Guizhou mini-pig model.
Collapse
Affiliation(s)
- Yanjun Wu
- The Provincial Key Miao Medicine Laboratory of Guizhou, Guizhou University of Traditional Chinese Medicine, Guiyang, China
| | - Jiayuan Mo
- College of Animal Science, Anhui Science and Technology University, Chuzhou, China
- Anhui Engineering Technology Research Center of Pork Quality Control and Enhance, Anhui Science and Technology University, Chuzhou, China
| | - Qianguang Wang
- The Provincial Key Miao Medicine Laboratory of Guizhou, Guizhou University of Traditional Chinese Medicine, Guiyang, China
| | - Jialong Li
- The Provincial Key Miao Medicine Laboratory of Guizhou, Guizhou University of Traditional Chinese Medicine, Guiyang, China
| | - Jia Wei
- The Provincial Key Miao Medicine Laboratory of Guizhou, Guizhou University of Traditional Chinese Medicine, Guiyang, China
| | - Nuo Zhang
- The Provincial Key Miao Medicine Laboratory of Guizhou, Guizhou University of Traditional Chinese Medicine, Guiyang, China
| | - Yuanqiu Dong
- The Provincial Key Miao Medicine Laboratory of Guizhou, Guizhou University of Traditional Chinese Medicine, Guiyang, China
| | - Xiang Zhu
- The Provincial Key Miao Medicine Laboratory of Guizhou, Guizhou University of Traditional Chinese Medicine, Guiyang, China
| | - Taofeng Lu
- The Provincial Key Miao Medicine Laboratory of Guizhou, Guizhou University of Traditional Chinese Medicine, Guiyang, China
| | - Sicheng Huang
- Department of Oncology, The Affiliated Hospital of Guizhou Medical University/ Guizhou Hospital of the First Affiliated Hospital, Sun Yat-sen University, Guiyang, China
- Department of Abdominal Oncology, The Affiliated Cancer Hospital of Guizhou Medical University, Guiyang, China
| |
Collapse
|
26
|
Zhu T, Liu S, Gao Y, Sun X, Hu H, Zhang Z, Zhang W, Xiong L, Liu H. Functional Characterization of a Highly Efficient Endoglucanase from Bacillus licheniformis BJ2022 and Its Application in the Preparation of Low-Molecular-Weight Konjac Glucomannan. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2025; 73:8367-8380. [PMID: 40139964 DOI: 10.1021/acs.jafc.4c10714] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/29/2025]
Abstract
Endoglucanases may not only act on β-1,4-linkages in β-glucan but also target d-glucose in glucomannan, which can expand their utility in the preparation of prebiotics. In this study, a highly efficient endoglucanase (BlGH5) from Bacillus licheniformis BJ2022 was expressed and characterized. BlGH5 exhibited the highest activity at pH 7.0 and 60 °C. It maintained over 80% activity at pH 4.0-7.0 and 30-60 °C. BlGH5 specifically cleaved β-1,4-glycosidic bonds linked to d-glucose. Site-directed mutagenesis results suggested that Arg91, Asn167, Glu168, Trp206, His228, Tyr230, Ser263, and Trp290 are key residues for its binding and catalytic activity. Moreover, BlGH5 displayed high activity against konjac glucomannan (KGM), indicating that BlGH5 could be used to prepare low-molecular-weight konjac glucomannan (KGM-L). Based on the physicochemical properties of KGM and KGM-L, KGM-L was characterized by reduced molecular weight and viscosity. Fecal fermentation experiments demonstrated that KGM and KGM-L could promote the production of short-chain fatty acids (SCFAs). Still, KGM-L was more conducive to the growth of the gut probiotics. In conclusion, we identified an endoglucanase for the degradation of KGM. Results indicated that KGM-L would have superior prebiotic effects. Thus, our study provides a basis for the future development and application of KGM-L as a prebiotic.
Collapse
Affiliation(s)
- Tianxiang Zhu
- College of Basic Medicine, Hubei University of Chinese Medicine, Wuhan 430065, China
- Hubei Shizhen Laboratory, Wuhan 430061, China
- Key Laboratory of Chinese Medicine Resource and Compound Prescription, Ministry of Education, Wuhan, Hubei 430065, China
| | - Songlin Liu
- Hubei Shizhen Laboratory, Wuhan 430061, China
- Key Laboratory of Chinese Medicine Resource and Compound Prescription, Ministry of Education, Wuhan, Hubei 430065, China
| | - Yu Gao
- College of Basic Medicine, Hubei University of Chinese Medicine, Wuhan 430065, China
- Hubei Shizhen Laboratory, Wuhan 430061, China
- Key Laboratory of Chinese Medicine Resource and Compound Prescription, Ministry of Education, Wuhan, Hubei 430065, China
| | - Xiongjie Sun
- College of Basic Medicine, Hubei University of Chinese Medicine, Wuhan 430065, China
- Hubei Shizhen Laboratory, Wuhan 430061, China
- Key Laboratory of Chinese Medicine Resource and Compound Prescription, Ministry of Education, Wuhan, Hubei 430065, China
| | - Haiming Hu
- Hubei Shizhen Laboratory, Wuhan 430061, China
- Key Laboratory of Chinese Medicine Resource and Compound Prescription, Ministry of Education, Wuhan, Hubei 430065, China
- School of Laboratory Medicine, Hubei University of Chinese Medicine, Wuhan, Hubei 430065, China
| | - Zhigang Zhang
- College of Basic Medicine, Hubei University of Chinese Medicine, Wuhan 430065, China
- Hubei Shizhen Laboratory, Wuhan 430061, China
- Key Laboratory of Chinese Medicine Resource and Compound Prescription, Ministry of Education, Wuhan, Hubei 430065, China
| | - Wenwen Zhang
- College of Basic Medicine, Hubei University of Chinese Medicine, Wuhan 430065, China
- Hubei Shizhen Laboratory, Wuhan 430061, China
- Key Laboratory of Chinese Medicine Resource and Compound Prescription, Ministry of Education, Wuhan, Hubei 430065, China
| | - Lei Xiong
- College of Basic Medicine, Hubei University of Chinese Medicine, Wuhan 430065, China
- Hubei Shizhen Laboratory, Wuhan 430061, China
- Key Laboratory of Chinese Medicine Resource and Compound Prescription, Ministry of Education, Wuhan, Hubei 430065, China
| | - Hongtao Liu
- College of Basic Medicine, Hubei University of Chinese Medicine, Wuhan 430065, China
- Hubei Shizhen Laboratory, Wuhan 430061, China
- Key Laboratory of Chinese Medicine Resource and Compound Prescription, Ministry of Education, Wuhan, Hubei 430065, China
| |
Collapse
|
27
|
Cheng X, He J, Yang Y, He Y, Chen G, Ling B, Wang A. Targeted metabolomics unravels the mechanism by phenylpropanoid-rich of the peel of Zea mays L. ameliorates metabolic disorders in diabetic mice through gut microbiota modulation. Front Pharmacol 2025; 16:1551713. [PMID: 40271058 PMCID: PMC12014729 DOI: 10.3389/fphar.2025.1551713] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2024] [Accepted: 03/18/2025] [Indexed: 04/25/2025] Open
Abstract
Background Diabetes is one common clinical symptoms of metabolic disorders. The peel of Zea mays L. is a folk remedy for diabetes that has not been thoroughly studied. The effects and mechanisms on diabetes complicated glucose and lipid metabolism disorders are still unknown now. Purpose The research is intended to elucidate the constituent of phenylpropanoid enriched of Zea mays L. (YMP), and investigate the treatment and mechanism on amending glucose and lipid metabolism disorders. Methods The constituents of YMP were systematacially identified by HPLC-Q-TOF-MS/MS and NMR. To assess the effects of varying YMP doses, diabetic mice induced by streptozotocin and a high-fat diet were divided into groups. Targeted serum metabolomics investigations were conducted using UHPLC-LTQ-Orbitrap MS. Moreover, 16S rRNA analysis was employed to elucidate the intricate mechanisms through the gut microbiota modulates lipid and glucose metabolism. Results It demonstrated that the primary component of YMP was luteolin. At a high dosage of 160 mg/kg/day, YMP considerably reduced the values of the oral glucose tolerance test, insulin, and blood glucose (p < 0.001). After administration, insulin resistance indexes decreased. YMP reversed the accumulation of glycogen in the liver and reduced hepatic lipid deposition. Compared to MOD group, the concentration of luteolin is higher and its metabolite, indicating that luteolin may be adequately absorbed and have an influence on the circulatory system. The results of 16S rRNA sequencing demonstrated that YMP and gut microbiota interacted to positively regulate beneficial bacteria such as Bifidobacterium, Ligilactobacillus, and Lactobacillus. Conclusion This work investigated the regulating effect of YMP on the liver glycolipid metabolism for the first time, and it also showed the underlying mechanism through gut microbiota. According to these studies, YMP has a lot of potential to be used as a supplemental treatment for complex metabolic illnesses like diabetes. It offered empirical support for the use of alternative medicine in the area to treat complex problems of glucose and lipid metabolism in diabetes.
Collapse
Affiliation(s)
- Xiaotian Cheng
- School of Pharmacy, Nantong University, Nantong, Jiangsu, China
- Department of Pharmacy, The Yancheng Clinical College of Xuzhou Medical University & The First people’s Hospital of Yancheng, Yancheng, Jiangsu, China
| | - Jinyan He
- School of Pharmacy, Nantong University, Nantong, Jiangsu, China
| | - Yuru Yang
- School of Pharmacy, Nantong University, Nantong, Jiangsu, China
| | - Yaonan He
- School of Pharmacy, Nantong University, Nantong, Jiangsu, China
| | - Guangtong Chen
- School of Pharmacy, Nantong University, Nantong, Jiangsu, China
| | - Bai Ling
- School of Pharmacy, Nantong University, Nantong, Jiangsu, China
- Department of Pharmacy, The Yancheng Clinical College of Xuzhou Medical University & The First people’s Hospital of Yancheng, Yancheng, Jiangsu, China
| | - Andong Wang
- School of Pharmacy, Nantong University, Nantong, Jiangsu, China
| |
Collapse
|
28
|
Zhang W, Wu Y, Cheng M, Wei H, Sun R, Peng H, Tian Z, Chen Y. Chronic hepatitis B virus infection imbalances short-chain fatty acids and amino acids in the liver and gut via microbiota modulation. Gut Pathog 2025; 17:18. [PMID: 40188120 PMCID: PMC11971750 DOI: 10.1186/s13099-025-00695-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/15/2024] [Accepted: 03/20/2025] [Indexed: 04/07/2025] Open
Abstract
The commensal microbiota is closely related to HBV infection and HBV-related liver diseases; however, how HBV and viral components dynamically affect the targeted organ liver microbiota is not well-known. In this study, an HBV-carrier mouse model established by HBsAg+ hepatocyte replacement in Fah-/- recipient mice, named HBs-HepR mice, was used to analyze the microbiota and metabolomics at the time of triggering the specific anti-HBV CD8+ T cell response in the liver. The composition and relative abundance of microbiota were both altered in the gut and liver of HBs-HepR mice. Particularly, increased Muribaculaceae and Alloprevotella, and decreased Lachnospiraceae-NK4A136 and Rikenella were observed in the gut; while increased Ralstonia and Geobacillus were observed in the liver of HBs-HepR mice. Furthermore, changes in microbial functions were revealed. There were no significant differences in the levels of SCFAs in fecal and serum; however, decreased propionic acid and acetic acid were detected in the livers of HBs-HepR mice, which was negatively related to the abundance of Geobacillus in the liver. Significantly decreased levels of 9 kinds of amino acids were detected in the feces of HBs-HepR mice, which was positively related to decreased Rikenella in the gut. A significant increase in L-glycine was observed in the liver and serum, positively related to the abundance of Geobaillus in the livers of HBs-HepR mice. In conclusion, chronic HBV infection imbalanced SCFA and amino acid metabolism by modulating microbiota in the liver, unlike in the gut, which was involved in the immune activation phase.
Collapse
Affiliation(s)
- Wendi Zhang
- Key Laboratory of Immune Response and Immunotherapy, the Institute of Immunology, Biomedical Sciences and Health Laboratory of Anhui Province, Center for Advanced Interdisciplinary Science and Biomedicine of IHM, School of Basic Medical Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, #443 Huangshan Road, Hefei, 230027, Anhui, China
| | - Yuwei Wu
- Key Laboratory of Immune Response and Immunotherapy, the Institute of Immunology, Biomedical Sciences and Health Laboratory of Anhui Province, Center for Advanced Interdisciplinary Science and Biomedicine of IHM, School of Basic Medical Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, #443 Huangshan Road, Hefei, 230027, Anhui, China
| | - Min Cheng
- Key Laboratory of Immune Response and Immunotherapy, the Institute of Immunology, Biomedical Sciences and Health Laboratory of Anhui Province, Center for Advanced Interdisciplinary Science and Biomedicine of IHM, School of Basic Medical Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, #443 Huangshan Road, Hefei, 230027, Anhui, China
- Department of Geriatrics, Gerontology Institute of Anhui Province, the First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, China
| | - Haiming Wei
- Key Laboratory of Immune Response and Immunotherapy, the Institute of Immunology, Biomedical Sciences and Health Laboratory of Anhui Province, Center for Advanced Interdisciplinary Science and Biomedicine of IHM, School of Basic Medical Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, #443 Huangshan Road, Hefei, 230027, Anhui, China
| | - Rui Sun
- Key Laboratory of Immune Response and Immunotherapy, the Institute of Immunology, Biomedical Sciences and Health Laboratory of Anhui Province, Center for Advanced Interdisciplinary Science and Biomedicine of IHM, School of Basic Medical Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, #443 Huangshan Road, Hefei, 230027, Anhui, China
| | - Hui Peng
- Key Laboratory of Immune Response and Immunotherapy, the Institute of Immunology, Biomedical Sciences and Health Laboratory of Anhui Province, Center for Advanced Interdisciplinary Science and Biomedicine of IHM, School of Basic Medical Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, #443 Huangshan Road, Hefei, 230027, Anhui, China
| | - Zhigang Tian
- Key Laboratory of Immune Response and Immunotherapy, the Institute of Immunology, Biomedical Sciences and Health Laboratory of Anhui Province, Center for Advanced Interdisciplinary Science and Biomedicine of IHM, School of Basic Medical Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, #443 Huangshan Road, Hefei, 230027, Anhui, China
| | - Yongyan Chen
- Key Laboratory of Immune Response and Immunotherapy, the Institute of Immunology, Biomedical Sciences and Health Laboratory of Anhui Province, Center for Advanced Interdisciplinary Science and Biomedicine of IHM, School of Basic Medical Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, #443 Huangshan Road, Hefei, 230027, Anhui, China.
| |
Collapse
|
29
|
Wang Y, Li Y, Lin Y, Cao C, Chen D, Huang X, Li C, Xu H, Lai H, Chen H, Zhou Y. Roles of the gut microbiota in hepatocellular carcinoma: from the gut dysbiosis to the intratumoral microbiota. Cell Death Discov 2025; 11:140. [PMID: 40185720 PMCID: PMC11971373 DOI: 10.1038/s41420-025-02413-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2024] [Revised: 02/23/2025] [Accepted: 03/18/2025] [Indexed: 04/07/2025] Open
Abstract
Hepatocellular carcinoma (HCC) is closely linked to alterations in the gut microbiota. This dysbiosis is characterized by significant changes in the microbial population, which correlate with the progression of HCC. Gut dysbiosis ultimately promotes HCC development in several ways: it damages the integrity of the gut-vascular barrier (GVB), alters the tumor microenvironment (TME), and even affects the intratumoral microbiota. Subsequently, intratumoral microbiota present a characteristic profile and play an essential role in HCC progression mainly by causing DNA damage, mediating tumor-related signaling pathways, altering the TME, promoting HCC metastasis, or through other mechanisms. Both gut microbiota and intratumoral microbiota have dual effects on HCC progression; a comprehensive understanding of their complex biological roles will provide a theoretical foundation for potential clinical applications in HCC treatment.
Collapse
Affiliation(s)
- Yiqin Wang
- Department of Gastroenterology and Hepatology, The Second Affiliated Hospital, School of Medicine, South China University of Technology, Guangzhou, China
- Department of Gastroenterology and Hepatology, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou, China
| | - Yongqiang Li
- Department of Gastroenterology and Hepatology, The Second Affiliated Hospital, School of Medicine, South China University of Technology, Guangzhou, China
- Department of Gastroenterology and Hepatology, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou, China
| | - Yong Lin
- Department of Gastroenterology and Hepatology, The Second Affiliated Hospital, School of Medicine, South China University of Technology, Guangzhou, China
- Department of Gastroenterology and Hepatology, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou, China
| | - Chuangyu Cao
- Department of Gastroenterology and Hepatology, The Second Affiliated Hospital, School of Medicine, South China University of Technology, Guangzhou, China
- Department of Gastroenterology and Hepatology, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou, China
| | - Dongcheng Chen
- Department of Gastroenterology and Hepatology, Baiyun Hospital of Guangzhou First People's Hospital (The Second People's Hospital of Baiyun District), Guangzhou, China
| | - Xianguang Huang
- Department of Gastroenterology and Hepatology, Baiyun Hospital of Guangzhou First People's Hospital (The Second People's Hospital of Baiyun District), Guangzhou, China
| | - Canhua Li
- Department of Gastroenterology and Hepatology, Baiyun Hospital of Guangzhou First People's Hospital (The Second People's Hospital of Baiyun District), Guangzhou, China
| | - Haoming Xu
- Department of Gastroenterology and Hepatology, The Second Affiliated Hospital, School of Medicine, South China University of Technology, Guangzhou, China
- Department of Gastroenterology and Hepatology, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou, China
| | - Huasheng Lai
- Department of Gastroenterology and Hepatology, The Second Affiliated Hospital, School of Medicine, South China University of Technology, Guangzhou, China
- Department of Gastroenterology and Hepatology, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou, China
| | - Huiting Chen
- Department of Gastroenterology and Hepatology, The Second Affiliated Hospital, School of Medicine, South China University of Technology, Guangzhou, China.
- Department of Gastroenterology and Hepatology, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou, China.
| | - Yongjian Zhou
- Department of Gastroenterology and Hepatology, The Second Affiliated Hospital, School of Medicine, South China University of Technology, Guangzhou, China.
- Department of Gastroenterology and Hepatology, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou, China.
| |
Collapse
|
30
|
Sanhueza-Carrera EA, Fernández-Lainez C, Castro-De la Mora C, Ortega-Álvarez D, Mendoza-Camacho C, Cortéz-Sánchez JM, Pérez-Guillé B, de Vos P, López-Velázquez G. Swine Gut Lactic Acid Bacteria and Their Exopolysaccharides Differentially Modulate Toll-like Receptor Signaling Depending on the Agave Fructans Used as a Carbon Source. Animals (Basel) 2025; 15:1047. [PMID: 40218440 PMCID: PMC11988020 DOI: 10.3390/ani15071047] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2025] [Revised: 03/27/2025] [Accepted: 04/02/2025] [Indexed: 04/14/2025] Open
Abstract
Exopolysaccharides (EPSs) produced by probiotic bacteria have garnered attention due to their effects on the gut health of humans and animals. The nutrients that probiotics access during their growth are essential for producing beneficial effects on host health. Direct immunomodulatory effects of graminan-type fructans (GTFs) from Agave tequilana through toll-like receptors (TLRs) have been demonstrated. However, the immunomodulatory effects of these fructans, mediated through the EPSs produced by the probiotics cultivated with them, remain unexplored. We explored the immunomodulatory effects of lactic acid bacteria (LAB) strains isolated from swine and their EPSs, based on the GTFs used as carbon sources during their growth. While the LAB strains activated the NF-κB pathway independently of the GTF source, their EPSs activated it in a GTF source-dependent manner. LAB activation through TLR2 showed a GTF source dependency, whereas their EPSs activated TLR2 independently of the GTF source. The LAB and their EPSs activated TLR4 in a GTF source-dependent manner. Both the LAB and their EPSs inhibited the activation of TLR2 and TLR4 agonists, which exhibited a strong dependence on the GTF source. The strength of GTF C's immunomodulatory effects on LAB illustrates its specificity, its impact on the EPS structure, and its biological effects. Our results support the promising health benefits of this synbiotic model for swine health and lowering inflammation.
Collapse
Affiliation(s)
- Enrique A. Sanhueza-Carrera
- Laboratorio de Biomoléculas y Salud Infantil, Instituto Nacional de Pediatría, Ciudad de Mexico 04530, Mexico;
- Posgrado en Ciencias Biológicas, Universidad Nacional Autónoma de México, Cuidad de Mexico 04510, Mexico
| | - Cynthia Fernández-Lainez
- Laboratorio de Errores Innatos del Metabolismo y Tamiz, Instituto Nacional de Pediatría, Ciudad de Mexico 04530, Mexico;
| | | | - Daniel Ortega-Álvarez
- Layan Biotic Solutions, Guadalajara 44670, Mexico; (C.C.-D.l.M.); (D.O.-Á.); (C.M.-C.)
| | | | | | - Beatriz Pérez-Guillé
- Translational Research Center, Instituto Nacional de Pediatría, Ciudad de Mexico 04530, Mexico;
| | - Paul de Vos
- Immunoendocrinology, Division of Medical Biology, Department of Pathology and Medical Biology, University and Medical Center of Groningen, 9700 Groningen, The Netherlands;
| | - Gabriel López-Velázquez
- Laboratorio de Biomoléculas y Salud Infantil, Instituto Nacional de Pediatría, Ciudad de Mexico 04530, Mexico;
| |
Collapse
|
31
|
Liu T, Zhang M, Xie Q, Gu J, Zeng S, Huang D. Unveiling the Antiobesity Mechanism of Sweet Potato Extract by Microbiome, Transcriptome, and Metabolome Analyses in Mice. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2025; 73:7807-7821. [PMID: 39989409 DOI: 10.1021/acs.jafc.4c13173] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/25/2025]
Abstract
This study aimed to elucidate the antiobesity mechanisms of sweet potato extract (SPE) through biochemical, gut microbiome, liver transcriptome, and metabolome analyses. Administration of SPE to high-fat-diet-fed mice significantly reduced body weight gain, serum low-density lipoprotein cholesterol, hepatic lipid accumulation, and adipocyte hypertrophy, which were closely linked to gut microbiome composition. SPE notably increased the abundance of Eubacterium_coprostanoligenes_group_unclassified and decreased that of Kineothrix, both of which were strongly associated with short-chain fatty acid (SCFA) production. LC-QTOF-MS analysis identified resin glycoside compounds from SPE with reduced levels in mouse feces, suggesting their utilization in vivo. SPE also promoted dietary fat excretion. Liver transcriptomic and metabolomic profiling revealed that SPE may exert antiobesity effects by modulating the bile-sphingolipid metabolism, which was closely correlated with the reshaped gut microbiomes and SCFAs. These findings provide new insights into the antiobesity effects and mechanisms of SPE.
Collapse
Affiliation(s)
- Tiange Liu
- National University of Singapore (Suzhou) Research Institute, 377 Linquan Street, Suzhou 215123, Jiangsu, China
| | - Min Zhang
- National University of Singapore (Suzhou) Research Institute, 377 Linquan Street, Suzhou 215123, Jiangsu, China
| | - Qingtong Xie
- Department of Food Science and Technology, National University of Singapore, 2 Science Drive 2, Singapore 117542, Singapore
| | - Jia Gu
- National University of Singapore (Suzhou) Research Institute, 377 Linquan Street, Suzhou 215123, Jiangsu, China
| | - Shunjiang Zeng
- National University of Singapore (Suzhou) Research Institute, 377 Linquan Street, Suzhou 215123, Jiangsu, China
| | - Dejian Huang
- National University of Singapore (Suzhou) Research Institute, 377 Linquan Street, Suzhou 215123, Jiangsu, China
- Department of Food Science and Technology, National University of Singapore, 2 Science Drive 2, Singapore 117542, Singapore
| |
Collapse
|
32
|
He Y, Jia D, Chen W, Liu J, Liu C, Shi X. Discussion on the treatment of diabetic kidney disease based on the "gut-fat-kidney" axis. Int Urol Nephrol 2025; 57:1233-1243. [PMID: 39549180 DOI: 10.1007/s11255-024-04283-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2024] [Accepted: 11/05/2024] [Indexed: 11/18/2024]
Abstract
Diabetic kidney disease is the main cause of end-stage renal disease, and its prevention and treatment are still a major clinical problem. The human intestine has a complex flora of hundreds of millions of microorganisms, and intestinal microorganisms, and their derivatives are closely related to renal inflammatory response, immune response, and material metabolism. Brown adipose tissue is the main part of adaptive thermogenesis. Recent studies have shown that activating brown fat by regulating intestinal flora has good curative effects in diabetic kidney disease-related diseases. As an emerging medical concept, the "gut-fat-kidney" axis has received increasing attention in diabetic kidney disease and related diseases. However, the specific mechanism involved needs further study. A new theoretical basis for the prevention and treatment of diabetic kidney disease is presented in this article, based on the "gut-fat-kidney" axis.
Collapse
Affiliation(s)
- Yaping He
- Gansu University of Chinese Medicine, Lanzhou, 730000, China
| | - Dengke Jia
- Lanzhou University Second Clinical Medical College, Lanzhou University, Lanzhou, 730000, China
| | - Wenying Chen
- Gansu University of Chinese Medicine, Lanzhou, 730000, China
| | - Juan Liu
- Gansu University of Chinese Medicine, Lanzhou, 730000, China
| | - Congrong Liu
- Gansu University of Chinese Medicine, Lanzhou, 730000, China
| | - Xiaowei Shi
- Department of Endocrinology, Gansu Provincial Hospital of Traditional Chinese Medicine, Lanzhou, 730000, China.
| |
Collapse
|
33
|
Yakut A. Gut microbiota in the development and progression of chronic liver diseases: Gut microbiota-liver axis. World J Hepatol 2025; 17:104167. [PMID: 40177197 PMCID: PMC11959663 DOI: 10.4254/wjh.v17.i3.104167] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/12/2024] [Revised: 01/28/2025] [Accepted: 02/25/2025] [Indexed: 03/26/2025] Open
Abstract
The gut microbiota (GM) is a highly dynamic ecology whose density and composition can be influenced by a wide range of internal and external factors. Thus, "How do GM, which can have commensal, pathological, and mutualistic relationships with us, affect human health?" has become the most popular research issue in recent years. Numerous studies have demonstrated that the trillions of microorganisms that inhabit the human body can alter host physiology in a variety of systems, such as metabolism, immunology, cardiovascular health, and neurons. The GM may have a role in the development of a number of clinical disorders by producing bioactive peptides, including neurotransmitters, short-chain fatty acids, branched-chain amino acids, intestinal hormones, and secondary bile acid conversion. These bioactive peptides enter the portal circulatory system through the gut-liver axis and play a role in the development of chronic liver diseases, cirrhosis, and hepatic encephalopathy. This procedure is still unclear and quite complex. In this study, we aim to discuss the contribution of GM to the development of liver diseases, its effects on the progression of existing chronic liver disease, and to address the basic mechanisms of the intestinal microbiota-liver axis in the light of recent publications that may inspire the future.
Collapse
Affiliation(s)
- Aysun Yakut
- Department of Gastroenterology, İstanbul Medipol University Sefakoy Health Practice Research Center, İstanbul 38000, Türkiye.
| |
Collapse
|
34
|
Huang A, Ma J, Zhu H, Qi Y, Jin Y, Zhang M, Yin L, Luo M, Chen S, Xie C, Huang H. Blood metabolites mediate causal inference studies on the effect of gut microbiota on the risk of vascular calcification. J Adv Res 2025:S2090-1232(25)00198-5. [PMID: 40139524 DOI: 10.1016/j.jare.2025.03.038] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2025] [Revised: 03/18/2025] [Accepted: 03/20/2025] [Indexed: 03/29/2025] Open
Abstract
BACKGROUND Emerging evidence indicates a notable connection between gut microbiota and Vascular Calcification (VC). Gut microbiota influences various disease processes through host metabolic pathways; however, the causative link between gut microbiota and VC, along with the potential mediating role of metabolites, is still not well understood. METHODS We leveraged data from the largest Genome-Wide Association Studies (GWAS) concerning gut microbiota, blood metabolites, and VC. To explore the causal relationships among these variables, we conducted two-sample bidirectional Mendelian Randomization (MR) analyses. Furthermore, mediation analyses were conducted to determine if metabolites act as an intermediary in the impact of gut microbiota on VC. In addition, we recruited CKD patients for mass spectrometry and CT examination, and performed a correlation analysis between the expression of blood metabolites and VC score. Finally, we experimentally validated the effects of intermediate metabolites on VC. RESULTS We identified 19 positive gut microbiota species and 52 positive blood metabolites with causal effects on VC. Additionally, the onset of VC was found to induce changes in the abundance of 24 gut microbiota species and 56 metabolites. Further analyses revealed that up to 13 positive gut microbiota species regulate the expression of 20 positive metabolites. Mediation analysis suggests that the gut microbiota g_KLE1615 promotes VC by downregulating the methionine-to-phosphate ratio. Mass spectrometry results indicate that over half of the metabolites identified through MR analysis show altered expression during CKD progression. Among them, 7 metabolites were significantly associated with the progression of VC. Further in vitro experiments confirmed the inhibitory effect of the intermediate metabolite methionine on VC. CONCLUSION Gut microbiota and blood metabolites are causally linked to VC. These findings provide a theoretical basis for microbiome- and metabolome-based therapeutic strategies for targeting VC and enhances our comprehension of the gut-vascular axis.
Collapse
Affiliation(s)
- Aoran Huang
- Department of Cardiology, Joint Laboratory of Guangdong-Hong Kong-Macao Universities for Nutritional Metabolism and Precise Prevention and Control of Major Chronic Diseases, the Eighth Affiliated Hospital of Sun Yat-sen University, Shenzhen 518033, China
| | - Jianshuai Ma
- Department of Cardiology, Joint Laboratory of Guangdong-Hong Kong-Macao Universities for Nutritional Metabolism and Precise Prevention and Control of Major Chronic Diseases, the Eighth Affiliated Hospital of Sun Yat-sen University, Shenzhen 518033, China
| | - Huijin Zhu
- Department of Cardiology, Joint Laboratory of Guangdong-Hong Kong-Macao Universities for Nutritional Metabolism and Precise Prevention and Control of Major Chronic Diseases, the Eighth Affiliated Hospital of Sun Yat-sen University, Shenzhen 518033, China
| | - Yanli Qi
- Department of Cardiology, Joint Laboratory of Guangdong-Hong Kong-Macao Universities for Nutritional Metabolism and Precise Prevention and Control of Major Chronic Diseases, the Eighth Affiliated Hospital of Sun Yat-sen University, Shenzhen 518033, China
| | - Yang Jin
- Department of Cardiology, Joint Laboratory of Guangdong-Hong Kong-Macao Universities for Nutritional Metabolism and Precise Prevention and Control of Major Chronic Diseases, the Eighth Affiliated Hospital of Sun Yat-sen University, Shenzhen 518033, China
| | - Mingxuan Zhang
- Department of Cardiology, Joint Laboratory of Guangdong-Hong Kong-Macao Universities for Nutritional Metabolism and Precise Prevention and Control of Major Chronic Diseases, the Eighth Affiliated Hospital of Sun Yat-sen University, Shenzhen 518033, China
| | - Li Yin
- Department of Cardiology, Joint Laboratory of Guangdong-Hong Kong-Macao Universities for Nutritional Metabolism and Precise Prevention and Control of Major Chronic Diseases, the Eighth Affiliated Hospital of Sun Yat-sen University, Shenzhen 518033, China
| | - Minhong Luo
- Department of Nephrology, the Eighth Affiliated Hospital of Sun Yat-sen University, Shenzhen 518033, China
| | - Sifan Chen
- Medical Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510000, China
| | - Chen Xie
- Department of Cardiology, Joint Laboratory of Guangdong-Hong Kong-Macao Universities for Nutritional Metabolism and Precise Prevention and Control of Major Chronic Diseases, the Eighth Affiliated Hospital of Sun Yat-sen University, Shenzhen 518033, China.
| | - Hui Huang
- Department of Cardiology, Joint Laboratory of Guangdong-Hong Kong-Macao Universities for Nutritional Metabolism and Precise Prevention and Control of Major Chronic Diseases, the Eighth Affiliated Hospital of Sun Yat-sen University, Shenzhen 518033, China.
| |
Collapse
|
35
|
Xu H, Ma Y, Shao L, Fu Y, Luo B, Xia C, Min X. Pan-cancer analysis of DLAT reveals it as a prognostic Biomarker involved in immune infiltration of liver hepatocellular carcinoma. J Cancer 2025; 16:2167-2180. [PMID: 40302799 PMCID: PMC12036108 DOI: 10.7150/jca.102256] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2024] [Accepted: 03/06/2025] [Indexed: 05/02/2025] Open
Abstract
Background: Dihydrolipoamide S-acetyltransferase (DLAT) is one of the cuproptosis-related genes (CRGs). Increasing evidence suggests that DLAT plays a critical role in various cancers. However, information about its functions and potential mechanisms in liver hepatocellular carcinoma (HCC) is still limited. Methods: Bioinformatics analysis were used to evaluate the potential association of DLAT expression with N6-methyladenosine (m6A) modification, clinical features, survival prognosis, biological functions, immune infiltration, and immune checkpoint molecules (ICM) in tumor patients. In addition, the expression of DLAT in HCC tissues was verified using immunohistochemistry (IHC). Results: The aberrant expression of DLAT had a significant impact on the prognosis of patients with various tumors. Importantly, DLAT expression was strongly associated with immune cell infiltration and immune checkpoint molecules (ICM) in tumors. For the first time, we found a significant positive correlation between DLAT expression and m6A regulatory factors in liver cancer, and that DLAT is associated with the PLK1 pathway, PI3K-AKT signaling pathway, Notch signaling pathway, WNT signaling pathway, and Aurora B pathway. Conclusions: Our results showed a significant increase in DLAT expression in HCC. Furthermore, the prognosis of patients with high DLAT expression was poor. Importantly, DLAT was correlated with immune cell infiltration and immune checkpoint molecules in HCC patients. Together, our results indicate that DLAT represents a promising therapeutic target for HCC patients.
Collapse
Affiliation(s)
- Haitao Xu
- Department of Neurosurgery, The Second Affiliated Hospital of Kunming Medical University, Kunming, Yunnan 650101, P.R. China
| | - Yiqun Ma
- Department of Radiology, 3201 Hospital, Hanzhong, Shanxi 723000, P.R. China
| | - Lishi Shao
- Department of Radiology, Yan'an Hospital Affiliated to Kunming Medical University, Kunming, Yunnan 650051, P.R. China
| | - Yao Fu
- Department of Hepatobiliary Surgery, The Second Affiliated Hospital of Kunming Medical University, Kunming, Yunnan 650101, P.R. China
| | - Bosheng Luo
- Department of Radiology, The Second Affiliated Hospital of Kunming Medical University, Kunming, Yunnan 650101, P.R. China
| | - Chunjuan Xia
- Department of Ultrasonography, The Second Affiliated Hospital of Kunming Medical University, Kunming, Yunnan 650101, P.R. China
| | - Xiaoli Min
- Department of Cerebrovascular Diseases, The Second Affiliated Hospital of Kunming Medical University, Kunming, Yunnan 650101, P.R. China
| |
Collapse
|
36
|
Wu Y, Huang X, Li Q, Yang C, Huang X, Du H, Situ B, Zheng L, Ou Z. Reducing severity of inflammatory bowel disease through colonization of Lactiplantibacillus plantarum and its extracellular vesicles release. J Nanobiotechnology 2025; 23:227. [PMID: 40114208 PMCID: PMC11924789 DOI: 10.1186/s12951-025-03280-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2025] [Accepted: 02/24/2025] [Indexed: 03/22/2025] Open
Abstract
Inflammatory bowel disease (IBD) is characterized by compromised intestinal barrier function and a lack of effective treatments. Probiotics have shown promise in managing IBD due to their ability to modulate the gut microbiota, enhance intestinal barrier function, and exert anti-inflammatory effects. However, the specific mechanisms through which probiotics exert these therapeutic effects in IBD treatment remain poorly understood. Our research revealed a significant reduction of Lactiplantibacillus plantarum (L. plantarum) in the gut microbiota of IBD patients. L. plantarum is a well-known probiotic strain in the list of edible probiotics, recognized for its beneficial effects on gut health, including its ability to strengthen the intestinal barrier and reduce inflammation. We demonstrated that supplementation with L. plantarum could alleviate IBD symptoms in mice, primarily by inhibiting apoptosis in intestinal epithelial cells through L. plantarum's bacterial extracellular vesicles (L. plant-EVs). This protective effect is dependent on the efficient uptake of L. plant-EVs by intestinal cells. Intriguingly, watermelon enhances L. plantarum colonization and L. plant-EVs release, further promoting intestinal barrier repair. Our findings contribute to the understanding of L. plant-EVs in the probiotic-based therapeutic approach for IBD, as they are promising candidates for nanoparticle-based therapeutic methods that are enhanced by natural diets such as watermelon. This study thereby offers a potential breakthrough in the management and treatment of IBD.
Collapse
Affiliation(s)
- Yuanyuan Wu
- Department of Laboratory Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Xinyue Huang
- Department of Laboratory Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Qianbei Li
- Department of Laboratory Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Chaoqun Yang
- Department of Laboratory Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
- Ganzhou Hospital-Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Xixin Huang
- Department of Laboratory Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Hualongyue Du
- Department of Laboratory Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Bo Situ
- Department of Laboratory Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China.
| | - Lei Zheng
- Department of Laboratory Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China.
| | - Zihao Ou
- Department of Laboratory Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China.
| |
Collapse
|
37
|
Kim MJ, Lee YJ, Hussain Z, Park H. Effect of Probiotics on Improving Intestinal Mucosal Permeability and Inflammation after Surgery. Gut Liver 2025; 19:207-218. [PMID: 39327843 PMCID: PMC11907258 DOI: 10.5009/gnl240170] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/18/2024] [Revised: 06/10/2024] [Accepted: 06/11/2024] [Indexed: 09/28/2024] Open
Abstract
Background/Aims We explored the mechanisms underlying the improvement of postoperative ileus (POI) following probiotic pretreatment. We assessed intestinal permeability, inflammation, tight junction (TJ) protein expression in the gut epithelium, and plasma interleukin (IL)-17 levels in a guinea pig model of POI. Methods Guinea pigs were divided into control, POI, and probiotic groups. The POI and probiotic groups underwent surgery, but the probiotic group received probiotics before the procedure. The ileum and proximal colon were harvested. Intestinal permeability was measured via horseradish peroxidase permeability. Inflammation was evaluated via leukocyte count in the intestinal wall muscle layer, and calprotectin expression in each intestinal wall layer was analyzed immunohistochemically. TJ proteins were analyzed using immunohistochemical staining, and plasma IL-17 levels were measured using an enzyme-linked immunosorbent assay. Results The POI group exhibited increased intestinal permeability and inflammation, whereas probiotic pretreatment reduced the extent of these POI-induced changes. Probiotics restored the expression of TJ proteins occludin and zonula occludens-1 in the proximal colon, which were increased in the POI group. Calprotectin expression significantly increased in the muscle layer of the POI group and was downregulated in the probiotic group; however, no distinct differences were observed between the mucosal and submucosal layers. Plasma IL-17 levels did not significantly differ among the groups. Conclusions Probiotic pretreatment may relieve POI by reducing intestinal permeability and inflammation and TJ protein expression in the gut epithelium. These findings suggest a potential therapeutic approach for POI management.
Collapse
Affiliation(s)
- Min-Jae Kim
- Division of Gastroenterology, Department of Internal Medicine, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul, Korea
| | - Young Ju Lee
- Division of Gastroenterology, Department of Internal Medicine, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul, Korea
| | - Zahid Hussain
- Division of Gastroenterology, Department of Internal Medicine, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul, Korea
| | - Hyojin Park
- Division of Gastroenterology, Department of Internal Medicine, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul, Korea
| |
Collapse
|
38
|
Hyder N, Raza ML. Stress and the gut microbiota-brain axis. PROGRESS IN BRAIN RESEARCH 2025; 291:175-203. [PMID: 40222779 DOI: 10.1016/bs.pbr.2025.01.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/15/2025]
Abstract
The gut microbiota-brain axis is a complex system that links the bacteria in our gut with our brain, it plays a part in what way we respond to stress. This chapter explores how stress affects the types of bacteria in the gut and shows the two-way connection between them. Stress can change the bacteria in our gut, which can cause various problems related to stress, like depression, anxiety, and irritable bowel syndrome (IBS). Figuring out how these interactions may help us develop new treatments that focus on the connection between gut bacteria and the brain. This chapter looks at how gut bacteria could help identify stress-related problems. It also discusses the difficulties and possibilities of using this research in medical practice. In the end, the chapter talks about what comes next in this quickly changing area. It highlights how important it is to include research about the gut-brain connection in overall public health plans.
Collapse
Affiliation(s)
- Noorulain Hyder
- Department of Pharmacology, Faculty of Pharmacy, Hamdard University, Karachi, Pakistan; HEJ Research Institute of Chemistry, International Center for Chemical and Biological Sciences, University of Karachi, Karachi, Pakistan.
| | - Muhammad Liaquat Raza
- Department of Infection Prevention & Control, Ministry of National Guard Health Affairs, Riyadh, Saudi Arabia; King Abdullah International Medical Research Center, Riyadh, Saudi Arabia; King Saud bin Abdulaziz University for Health Sciences, Riyadh, Saudi Arabia.
| |
Collapse
|
39
|
Chen Y, Yue S, Yu L, Cao J, Liu Y, Deng A, Lu Y, Yang J, Li H, Du J, Xia J, Li Y, Xia Y. Regulation and Function of the cGAS-STING Pathway: Mechanisms, Post-Translational Modifications, and Therapeutic Potential in Immunotherapy. Drug Des Devel Ther 2025; 19:1721-1739. [PMID: 40098909 PMCID: PMC11911240 DOI: 10.2147/dddt.s501773] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2024] [Accepted: 02/24/2025] [Indexed: 03/19/2025] Open
Abstract
Autoimmune diseases arise when the immune system attacks healthy tissues, losing tolerance for self-tissues. Normally, the immune system recognizes and defends against pathogens like bacteria and viruses. The cGAS-STING pathway, activated by pattern-recognition receptors (PRRs), plays a key role in autoimmune responses. The cGAS protein senses pathogenic DNA and synthesizes cGAMP, which induces conformational changes in STING, activating kinases IKK and TBK1 and leading to the expression of interferon genes or inflammatory mediators. This pathway is crucial in immunotherapy, activating innate immunity, enhancing antigen presentation, modulating the tumor microenvironment, and integrating into therapeutic strategies. Modulation strategies include small molecule inhibitors, oligonucleotide therapies, protein and antibody therapies, genetic and epigenetic regulation, cytokine and metabolite modulation, and nanoscale delivery systems. Post-translational modifications (PTMs) of the cGAS-STING pathway, such as phosphorylation, acetylation, ubiquitination, methylation, palmitoylation, and glycosylation, fine-tune immune responses by regulating protein activity, stability, localization, and interactions. These modifications are interconnected and collectively influence pathway functionality. We summarize the functions of cGAS-STING and its PTMs in immune and non-immune cells across various diseases, and explore potential clinical applications.
Collapse
Affiliation(s)
- Yuhan Chen
- Laboratory Medicine Center, Department of Clinical Laboratory, Zhejiang Provincial People’s Hospital (Affiliated People’s Hospital), Hangzhou Medical College, Hangzhou, Zhejiang, People’s Republic of China
| | - Si Yue
- Laboratory Medicine Center, Department of Clinical Laboratory, Zhejiang Provincial People’s Hospital (Affiliated People’s Hospital), Hangzhou Medical College, Hangzhou, Zhejiang, People’s Republic of China
| | - Lingyan Yu
- Laboratory Medicine Center, Department of Clinical Laboratory, Zhejiang Provincial People’s Hospital (Affiliated People’s Hospital), Hangzhou Medical College, Hangzhou, Zhejiang, People’s Republic of China
| | - Jinghao Cao
- Laboratory Medicine Center, Department of Clinical Laboratory, Zhejiang Provincial People’s Hospital (Affiliated People’s Hospital), Hangzhou Medical College, Hangzhou, Zhejiang, People’s Republic of China
| | - Yingchao Liu
- Laboratory Medicine Center, Department of Clinical Laboratory, Zhejiang Provincial People’s Hospital (Affiliated People’s Hospital), Hangzhou Medical College, Hangzhou, Zhejiang, People’s Republic of China
| | - Aoli Deng
- Laboratory Medicine Center, Department of Clinical Laboratory, Zhejiang Provincial People’s Hospital (Affiliated People’s Hospital), Hangzhou Medical College, Hangzhou, Zhejiang, People’s Republic of China
| | - Yajuan Lu
- Laboratory Medicine Center, Department of Clinical Laboratory, Zhejiang Provincial People’s Hospital (Affiliated People’s Hospital), Hangzhou Medical College, Hangzhou, Zhejiang, People’s Republic of China
| | - Jing Yang
- Laboratory Medicine Center, Department of Clinical Laboratory, Zhejiang Provincial People’s Hospital (Affiliated People’s Hospital), Hangzhou Medical College, Hangzhou, Zhejiang, People’s Republic of China
| | - Huanjuan Li
- Laboratory Medicine Center, Department of Clinical Laboratory, Zhejiang Provincial People’s Hospital (Affiliated People’s Hospital), Hangzhou Medical College, Hangzhou, Zhejiang, People’s Republic of China
| | - Jing Du
- Laboratory Medicine Center, Department of Clinical Laboratory, Zhejiang Provincial People’s Hospital (Affiliated People’s Hospital), Hangzhou Medical College, Hangzhou, Zhejiang, People’s Republic of China
| | - Jun Xia
- Laboratory Medicine Center, Department of Clinical Laboratory, Zhejiang Provincial People’s Hospital (Affiliated People’s Hospital), Hangzhou Medical College, Hangzhou, Zhejiang, People’s Republic of China
| | - Yanchun Li
- Department of Clinical Laboratory, Affiliated Hangzhou First People’s Hospital, School of Medicine, Westlake University, Hangzhou, Zhejiang, People’s Republic of China
| | - Yongming Xia
- Department of Hematology, Yuyao People’s Hospital, Yuyao, Zhejiang, People’s Republic of China
| |
Collapse
|
40
|
Zhan J, Cheng J, Chang W, Su Y, Yue X, Wu C. Absolute Quantitative Metagenomic Analysis Provides More Accurate Insights for the Anti-Colitis Effect of Berberine via Modulation of Gut Microbiota. Biomolecules 2025; 15:400. [PMID: 40149936 PMCID: PMC11940175 DOI: 10.3390/biom15030400] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2025] [Revised: 03/01/2025] [Accepted: 03/07/2025] [Indexed: 03/29/2025] Open
Abstract
Current gut microbiota studies often rely on relative quantitative sequencing. However, under certain circumstances, while the relative quantitative abundance of these bacteria may remain stable, the absolute quantities of specific bacteria can vary considerably. Since the function of bacteria is directly linked to their total numbers, absolute quantification is crucial. This study aims to identify the optimal method for microbiome analysis by comparing relative and absolute quantitative sequencing. Using ulcerative colitis, which is closely associated with gut microbiota, as a disease model and berberine (which affects microbiota) versus sodium butyrate (which does not) as drugs, relative and absolute quantitative methods were used to evaluate the varying effects of the different drugs on the regulation of gut microbiota in UC-affected animals. The regulatory effects of BBR on gut microbiota were further synthesized as identified in earlier studies using an individual-based meta-analysis, and we compared these findings with our absolute sequencing results. The results from absolute sequencing were more consistent with the actual microbial community, suggesting that relative abundance measurements might not accurately reflect the true abundance of microbial species. Moreover, meta-analysis results were only partially consistent with absolute quantitative sequencing and sometimes directly opposed, suggesting that relative quantitative sequencing analyses are prone to misinterpretation and incorrect correlation of results. This study underscores the importance of absolute quantitative analysis in accurately representing the true microbial counts in a sample and evaluating the modulatory effects of drugs on the microbiome, which plays a vital role in the study of the microbiome.
Collapse
Affiliation(s)
- Jiaguo Zhan
- School of Chinese Materia Medica, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China; (J.Z.); (J.C.); (W.C.); (Y.S.); (X.Y.)
| | - Jiale Cheng
- School of Chinese Materia Medica, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China; (J.Z.); (J.C.); (W.C.); (Y.S.); (X.Y.)
| | - Wenhui Chang
- School of Chinese Materia Medica, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China; (J.Z.); (J.C.); (W.C.); (Y.S.); (X.Y.)
| | - Yuying Su
- School of Chinese Materia Medica, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China; (J.Z.); (J.C.); (W.C.); (Y.S.); (X.Y.)
| | - Xixin Yue
- School of Chinese Materia Medica, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China; (J.Z.); (J.C.); (W.C.); (Y.S.); (X.Y.)
| | - Chongming Wu
- School of Chinese Materia Medica, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China; (J.Z.); (J.C.); (W.C.); (Y.S.); (X.Y.)
- State Key Laboratory of Chinese Medicine Modernization, Tianjin 301617, China
- Tianjin Key Laboratory of Therapeutic Substance of Traditional Chinese Medicine, Tianjin 301617, China
| |
Collapse
|
41
|
Zhang X, Chen J, Zhang S, Wei B, Han Y, Zhao Z. Insight into the Potential of Somatostatin Vaccination with Goats as a Model: From a Perspective of the Gastrointestinal Microbiota. Animals (Basel) 2025; 15:728. [PMID: 40076011 PMCID: PMC11899232 DOI: 10.3390/ani15050728] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2024] [Revised: 02/01/2025] [Accepted: 02/07/2025] [Indexed: 03/14/2025] Open
Abstract
Deciphering the gastrointestinal microbial response to oral SS DNA vaccines with different doses is helpful for identifying the mechanism for effective utilization of the vaccine for improving animal production. Here, we conduct a comparative study with different doses of vaccine (control: empty plasmid; low dose: 1 × 107 CFU vaccine; high dose: 1 × 1012 CFU vaccine) using goat as a case to investigate the potential of somatostatin vaccination from the entire gastrointestinal microbiota perspective. Our results show that body weight gain and slaughter rate are greater in the L_SS group than in the C_SS group. Compared with the C_SS group, the GH concentration is reduced, while the SS concentration is elevated in the cecum of L_SS goats. Moreover, the SCFAs concentration is elevated in the L_SS goats, the acetate molar proportion is lower in the rumen, the proportion of the acetate is decreased, and propionate is increased in the cecum of L_SS goats. Our data indicate that the low-dose somatostatin vaccine possesses a more efficient improvement in the productivity of goats, emphasizing that the dosage should be considered to reach its optimal effect on the host. Moreover, we find that different doses of the SS vaccination select distinct microbial communities in the gastrointestinal tract. Beta diversity analysis shows a significant interaction. Microorganisms capable of converting nutrients, including Ruminococcacease, Butyrivibrio, Akkermansia, and Lachnospiraceae are enriched, altering the gastrointestinal fermentation response to SS DNA vaccination of ruminants. Moreover, the correlation analysis results revealing these biomarkers have a close association with the phenotypes of productivity. These results imply that somatostatin immunoneutralization might directly alter the gastrointestinal tract commensal bacterial structure, improving gastrointestinal homeostasis, and, thus, modifying the fermentability and effected hormone level to improve the productivity of goats. Our study extends the understanding of the somatostatin vaccine regulation of ruminants' growth through the entire gastrointestinal microbial perspective.
Collapse
Affiliation(s)
| | | | | | | | | | - Zhongquan Zhao
- Chongqing Key Laboratory of Herbivore Science, College of Animal Science and Technology, Southwest University, Chongqing 400715, China; (X.Z.)
| |
Collapse
|
42
|
Lim SYM, Chong EJ, Mah WY, Pan Y, Fang CM, Murugaiah C, Alshagga M. Exploring the anti-obesity effects of Lactobacillus in C57BL/6 mice: mechanisms, interventions, and future directions. Lett Appl Microbiol 2025; 78:ovaf024. [PMID: 39965784 DOI: 10.1093/lambio/ovaf024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2024] [Revised: 01/25/2025] [Accepted: 02/17/2025] [Indexed: 02/20/2025]
Abstract
Lactobacillus species show strong potential in fighting obesity-related inflammation and metabolic issues. Obesity causes inflammation in adipose tissue, which harms insulin sensitivity and leads to fat buildup. Lactobacillus strains like Lactobacillus gasseri, Lactobacillus reuteri, and Lactobacillus plantarum help regulate lipid metabolism by boosting key genes, preventing fat cell formation, and encouraging fat breakdown. They also produce short-chain fatty acids (SCFAs) that improve gut health, activate metabolic pathways, and reduce inflammation. Studies in animals have shown that Lactobacillus can reduce body weight, fat, and inflammation, with Lactobacillus plantarum being especially effective in improving gut microbiota and liver function. When combined with other probiotics or prebiotics, these strains work even better, enhancing lipid metabolism and reducing inflammation. These results suggest that Lactobacillus could be an effective way to manage obesity and related health problems by influencing metabolism, gut health, and inflammation. However, more research, particularly human clinical trials, is needed to confirm its potential as a dietary treatment for obesity.
Collapse
Affiliation(s)
- Sharoen Yu Ming Lim
- Division of Biomedical Sciences, School of Pharmacy, University of Nottingham Malaysia, 43500 Semenyih, Selangor, Malaysia
- Faculty of Business, Design and Arts, Swinburne University of Technology, 93350 Kuching, Sarawak, Malaysia
| | - E-Jayn Chong
- Division of Biomedical Sciences, School of Pharmacy, University of Nottingham Malaysia, 43500 Semenyih, Selangor, Malaysia
| | - Weng Yan Mah
- Division of Biomedical Sciences, School of Pharmacy, University of Nottingham Malaysia, 43500 Semenyih, Selangor, Malaysia
| | - Yan Pan
- Division of Biomedical Sciences, School of Pharmacy, University of Nottingham Malaysia, 43500 Semenyih, Selangor, Malaysia
| | - Chee Mun Fang
- Division of Biomedical Sciences, School of Pharmacy, University of Nottingham Malaysia, 43500 Semenyih, Selangor, Malaysia
| | - Chandrika Murugaiah
- Faculty of Medicine, Manipal University College Malaysia, Bukit Baru 75150 Melaka, Malaysia
| | - Mustafa Alshagga
- Division of Biomedical Sciences, School of Pharmacy, University of Nottingham Malaysia, 43500 Semenyih, Selangor, Malaysia
| |
Collapse
|
43
|
Li Z, Xu R, Li C, Chi C, Yang X, Yang S, Liu R. The protective effect and potential mechanism of Zanthoxylum bungeanum Maxim. on atherosclerosis. Fitoterapia 2025; 181:106394. [PMID: 39826616 DOI: 10.1016/j.fitote.2025.106394] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2024] [Revised: 01/06/2025] [Accepted: 01/12/2025] [Indexed: 01/22/2025]
Abstract
The pericarp of Zanthoxylum bungeanum Maxim. (ZBM) is an edible spice with medicinal value, and it has anti-obesity, anti-inflammatory, and cardiovascular protective effects. This study investigated the therapeutic effect of ZBM on atherosclerosis (AS) and its potential mechanisms. An in vivo experimental model of AS was established on apolipoprotein-E deficient (ApoE-/-) mice to evaluate the efficacy of ZBM. Serum metabolomics combined with the vascular smooth muscle cell (VSMC) proliferation model were subsequently conducted to analyze the mechanisms. Results showed that ZBM effectively alleviated blood lipid disorders, aortic lipid accumulation, and intimal thickness in mice. Metabolomics indicated that ZBM mainly regulated 5 major metabolic pathways, including TCA cycle, steroid hormone biosynthesis, sphingolipid metabolism, glyoxylate and dicarboxylate metabolism, glycerophospholipid metabolism, which affected lipid metabolism and cell proliferation. Further experiments showed that ZBM inhibited VSMC proliferation likely because it blocked the signal transducer activator of transcription 3 (STAT3) phosphorylation and activated nuclear factor E2-related factor-2 (Nrf2)/heme oxygenase-1 (HO-1) pathway, resulting in G0/G1 phase arrest and reactive oxygen species (ROS) clearance.
Collapse
Affiliation(s)
- Zongchao Li
- School of Pharmacy, Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), Ministry of Education, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Yantai University, Yantai, China
| | - Ruoxuan Xu
- School of Pharmacy, Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), Ministry of Education, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Yantai University, Yantai, China
| | - Can Li
- School of Pharmacy, Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), Ministry of Education, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Yantai University, Yantai, China
| | - Chenglin Chi
- School of Pharmacy, Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), Ministry of Education, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Yantai University, Yantai, China
| | - Xiaoli Yang
- School of Pharmacy, Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), Ministry of Education, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Yantai University, Yantai, China
| | - Shufang Yang
- School of Pharmacy, Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), Ministry of Education, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Yantai University, Yantai, China
| | - Rongxia Liu
- School of Pharmacy, Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), Ministry of Education, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Yantai University, Yantai, China.
| |
Collapse
|
44
|
Tao W, Zhang Y, Wang B, Nie S, Fang L, Xiao J, Wu Y. Advances in molecular mechanisms and therapeutic strategies for central nervous system diseases based on gut microbiota imbalance. J Adv Res 2025; 69:261-278. [PMID: 38579985 PMCID: PMC11954836 DOI: 10.1016/j.jare.2024.03.023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2024] [Revised: 03/12/2024] [Accepted: 03/29/2024] [Indexed: 04/07/2024] Open
Abstract
BACKGROUD Central nervous system (CNS) diseases pose a serious threat to human health, but the regulatory mechanisms and therapeutic strategies of CNS diseases need to be further explored. It has been demonstrated that the gut microbiota (GM) is closely related to CNS disease. GM structure disorders, abnormal microbial metabolites, intestinal barrier destruction and elevated inflammation exist in patients with CNS diseases and promote the development of CNS diseases. More importantly, GM remodeling alleviates CNS pathology to some extent. AIM OF REVIEW Here, we have summarized the regulatory mechanism of the GM in CNS diseases and the potential treatment strategies for CNS repair based on GM regulation, aiming to provide safer and more effective strategies for CNS repair from the perspective of GM regulation. KEY SCIENTIFIC CONCEPTS OF REVIEW The abundance and composition of GM is closely associated with the CNS diseases. On the basis of in-depth analysis of GM changes in mice with CNS disease, as well as the changes in its metabolites, therapeutic strategies, such as probiotics, prebiotics, and FMT, may be used to regulate GM balance and affect its microbial metabolites, thereby promoting the recovery of CNS diseases.
Collapse
Affiliation(s)
- Wei Tao
- The Institute of Life Sciences, Wenzhou University, Wenzhou 325035, China
| | - Yanren Zhang
- The Institute of Life Sciences, Wenzhou University, Wenzhou 325035, China
| | - Bingbin Wang
- The Institute of Life Sciences, Wenzhou University, Wenzhou 325035, China
| | - Saiqun Nie
- The Institute of Life Sciences, Wenzhou University, Wenzhou 325035, China
| | - Li Fang
- The Institute of Life Sciences, Wenzhou University, Wenzhou 325035, China
| | - Jian Xiao
- School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China.
| | - Yanqing Wu
- The Institute of Life Sciences, Wenzhou University, Wenzhou 325035, China.
| |
Collapse
|
45
|
Zheng Y, Gao D, Xie H, Geng H. Epicatechin protects mice against OVA-induced asthma through inhibiting airway inflammation and modulating gut microbiota. Cell Signal 2025; 127:111609. [PMID: 39826677 DOI: 10.1016/j.cellsig.2025.111609] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2024] [Revised: 12/21/2024] [Accepted: 01/13/2025] [Indexed: 01/22/2025]
Abstract
Allergic asthma is a chronic airway inflammatory reaction that seriously affects people's quality of life and even endangers their lives. The aim of this study was to explore the role of epicatechin (EC) on asthma and its potential mechanism. A mice model of allergic asthma was established by intraperitoneal injection of ovalbumin (OVA) with aluminum hydrogen solution, and nebulized inhalation of OVA to stimulate. EC (10, 20, 40 mg/kg) was administered 30 min before nebulization for three consecutive days. The results showed that EC attenuated OVA-induced lung injury, inflammatory cell infiltration, IgE, and inflammatory cytokine production. EC also inhibited OVA-induced NF-κB activation and increased Nrf2 and HO-1 expression. 16S rRNA sequencing analysis demonstrated that at genus level, EC significantly increased the abundance of Lachnospiraceae_NK4A136_group, Ligilactobacillus, Alloprevotella. Meanwhile, EC inhibited the abundance of Clostridia UCG-014, Helicobacter, Paramuribaculum, and Escherichia-Shigella. In conclusion, EC can effectively alleviate the symptoms of asthma in mice, which may through regulating the composition of gut microbiota and inhibiting inflammatory response.
Collapse
Affiliation(s)
- Yang Zheng
- Department of Gynecology, China-Japan Union Hospital of Jilin University, 126 Sendai Street, Changchun, Jilin 130033, China
| | - Dengyu Gao
- Department of Anesthesiology, China-Japan Union Hospital of Jilin University, 126 Sendai Street, Changchun, Jilin 130033, China
| | - Hongyang Xie
- Department of Gynecology, China-Japan Union Hospital of Jilin University, 126 Sendai Street, Changchun, Jilin 130033, China.
| | - Huafeng Geng
- Department of Gynecology, China-Japan Union Hospital of Jilin University, 126 Sendai Street, Changchun, Jilin 130033, China.
| |
Collapse
|
46
|
Jin L, Hu R, Qing Y, Rang Z, Cui F. Exploring the Role of Gut Vascular Barrier Proteins in HIV-Induced Mucosal Damage: A Comparative Study. AIDS Res Hum Retroviruses 2025; 41:159-166. [PMID: 39501662 DOI: 10.1089/aid.2024.0077] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/16/2025] Open
Abstract
This study aims to compare intestinal mucosal damage and the expression levels of occludin, zonula occludens-1 (ZO-1), vascular endothelial (VE)-cadherin, β-catenin, and plasmalemma vesicle-associated protein (PLVAP) in the gut vascular barrier (GVB) among people living with HIV (PLWH), asymptomatic PLWH, and healthy volunteers (non-PLWH). Three groups were selected for the study: PLWH, asymptomatic PLWH, and healthy volunteers. Colonic mucosal tissue samples were collected via colonoscopy from all participants. Histological examination of the colonic mucosa was conducted using hematoxylin and eosin staining. The expression levels of occludin, ZO-1, VE-cadherin, β-catenin, and PLVAP were assessed using RT-qPCR, immunohistochemistry, and western blot analyses. Pathological scores of colonic mucosa in PLWH and asymptomatic PLWH were significantly higher than those in non-PLWH (p < .001 and p = .0056, respectively). CD4+ T cell counts in asymptomatic PLWH and non-PLWH were significantly higher than in PLWH (p < 0.05). The CD4+/CD8+ T cell ratio in non-PLWH significantly exceeded those in PLWH and asymptomatic PLWH (p < .05). Analysis of protein and mRNA expression revealed: (1) no statistically significant differences in PLVAP-mRNA expression across all groups (p > .05); (2) higher PLVAP protein levels in PLWH compared with asymptomatic PLWH and non-PLWH (p < .05), with no significant differences between asymptomatic PLWH and non-PLWH (p = .632); (3) significantly higher PLVAP expression in the colonic mucosa of PLWH and asymptomatic PLWH compared with non-PLWH (p = .034 and p = .011, respectively), with no significant differences between PLWH and asymptomatic PLWH (p > .999). ZO-1 expression was significantly lower in PLWH than in non-PLWH (p = .012), with no notable differences between asymptomatic PLWH and other groups. PLWH, compared with healthy controls, exhibit significant inflammatory changes in the intestinal mucosa. PLVAP expression serves as a potential indicator to assess the extent of GVB damage and disease progression in PLWH.
Collapse
Affiliation(s)
- Li Jin
- Department of Dermatology, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Rong Hu
- Department of Dermatology, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Yong Qing
- Department of Proctology and Dermatology, Chengdu Anorectal Hospital, Chengdu, China
| | - Zhen Rang
- Department of Dermatology, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Fan Cui
- Department of Dermatology, The Affiliated Hospital of Southwest Medical University, Luzhou, China
- Department of Dermatology, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| |
Collapse
|
47
|
Sun Y, Sun Z, Fang B, Wang R, Liu Y, Li J, Lan H, Zhao W, Hung WL, Zhang M. Exploring the anti-inflammatory potential of Lacticaseibacillus paracasei postbiotics: Mechanistic insights and functional components. FOOD BIOSCI 2025; 65:106105. [DOI: 10.1016/j.fbio.2025.106105] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/04/2025]
|
48
|
Mahmod AI, Govindaraju K, Lokanathan Y, Said NABM, Ibrahim B. Exploring the Potential of Stem Cells in Modulating Gut Microbiota and Managing Hypertension. Stem Cells Dev 2025; 34:99-116. [PMID: 39836384 DOI: 10.1089/scd.2024.0195] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2025] Open
Abstract
Hypertension, commonly known as high blood pressure, is a significant health issue that increases the risk of cardiovascular diseases, stroke, and renal failure. This condition broadly encompasses both primary and secondary forms. Despite extensive research, the underlying mechanisms of systemic arterial hypertension-particularly primary hypertension, which has no identifiable cause and is affected by genetic and lifestyle agents-remain complex and not fully understood. Recent studies indicate that an imbalance in gut microbiota, referred to as dysbiosis, may promote hypertension, affecting blood pressure regulation through metabolites such as short-chain fatty acids and trimethylamine N-oxide. Current antihypertensive medications face limitations, including resistance and adherence issues, highlighting the need for novel therapeutic approaches. Stem cell therapy, an emerging field in regenerative medicine, shows promise in addressing these challenges. Stem cells, with mesenchymal stem cells being a prime example, have regenerative, anti-inflammatory, and immunomodulatory properties. Emerging research indicates that stem cells can modulate gut microbiota, reduce inflammation, and improve vascular health, potentially aiding in blood pressure management. Research has shown the positive impact of stem cells on gut microbiota in various disorders, suggesting their potential therapeutic role in treating hypertension. This review synthesizes the recent studies on the complex interactions between gut microbiota, stem cells, and systemic arterial hypertension. By offering a thorough analysis of the current literature, it highlights key insights, uncovers critical gaps, and identifies emerging trends that will inform and guide future investigations in this rapidly advancing field.
Collapse
Affiliation(s)
- Asma Ismail Mahmod
- Department of Pharmaceutical Life Sciences, Faculty of Pharmacy, University of Malaya, Kuala Lumpur, Malaysia
| | - Kayatri Govindaraju
- Department of Pharmaceutical Life Sciences, Faculty of Pharmacy, University of Malaya, Kuala Lumpur, Malaysia
| | - Yogeswaran Lokanathan
- Department of Tissue Engineering and Regenerative Medicine, Faculty of Medicine, Universiti Kebangsaan Malaysia, Kuala Lumpur, Malaysia
- Advance Bioactive Materials-Cells UKM Research Group, Universiti Kebangsaan Malaysia, Bangi, Malaysia
| | - Nur Akmarina B M Said
- Department of Pharmaceutical Life Sciences, Faculty of Pharmacy, University of Malaya, Kuala Lumpur, Malaysia
| | - Baharudin Ibrahim
- Department of Clinical Pharmacy and Pharmacy Practices, Faculty of Pharmacy, University Malaya, Kuala Lumpur, Malaysia
| |
Collapse
|
49
|
Jans M, Vereecke L. A guide to germ-free and gnotobiotic mouse technology to study health and disease. FEBS J 2025; 292:1228-1251. [PMID: 38523409 DOI: 10.1111/febs.17124] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2023] [Revised: 01/17/2024] [Accepted: 03/11/2024] [Indexed: 03/26/2024]
Abstract
The intestinal microbiota has major influence on human physiology and modulates health and disease. Complex host-microbe interactions regulate various homeostatic processes, including metabolism and immune function, while disturbances in microbiota composition (dysbiosis) are associated with a plethora of human diseases and are believed to modulate disease initiation, progression and therapy response. The vast complexity of the human microbiota and its metabolic output represents a great challenge in unraveling the molecular basis of host-microbe interactions in specific physiological contexts. To increase our understanding of these interactions, functional microbiota research using animal models in a reductionistic setting are essential. In the dynamic landscape of gut microbiota research, the use of germ-free and gnotobiotic mouse technology, in which causal disease-driving mechanisms can be dissected, represents a pivotal investigative tool for functional microbiota research in health and disease, in which causal disease-driving mechanisms can be dissected. A better understanding of the health-modulating functions of the microbiota opens perspectives for improved therapies in many diseases. In this review, we discuss practical considerations for the design and execution of germ-free and gnotobiotic experiments, including considerations around germ-free rederivation and housing conditions, route and timing of microbial administration, and dosing protocols. This comprehensive overview aims to provide researchers with valuable insights for improved experimental design in the field of functional microbiota research.
Collapse
Affiliation(s)
- Maude Jans
- VIB Center for Inflammation Research, Ghent, Belgium
- Department of Biomedical Molecular Biology, Ghent University, Belgium
| | - Lars Vereecke
- VIB Center for Inflammation Research, Ghent, Belgium
- Department of Internal Medicine and Pediatrics, Ghent University, Belgium
| |
Collapse
|
50
|
Wang J, Jian A, Sun D, Cui M, Piao C, Wang J, Mu B, Li T, Li G, Li H. Acer tegmeutosum Maxim extract alleviates acute alcohol-induced liver disease and regulates gut microbiota dysbiosis in mice. Arch Biochem Biophys 2025; 765:110314. [PMID: 39832610 DOI: 10.1016/j.abb.2025.110314] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2024] [Revised: 12/29/2024] [Accepted: 01/15/2025] [Indexed: 01/22/2025]
Abstract
Acer tegmentosum Maxim (AT) has a variety of pharmacological activities, however, the effects of AT on liver injury and gut microbiota in alcoholic liver disease (ALD) mice is still unclear. This study aimed to evaluate the preventive effect of AT extract on acute alcoholic liver disease. Six-week-old male C57BL/6J mice were randomly divided into 6 groups. Each group was intragastrically treated saline or different concentration of AT extract solution for 5 weeks continuously. After the last gavage, except for the NC group, all the other groups were gavaged twice with 56 % alcohol to establish the acute ALD model and biochemical indexes, histopathological, and gut microbiota were analyzed. Established an acute ALD mouse model and detected serum, liver oxidation levels, and alcohol metabolism-related gene expressions. Through 16S rRNA sequencing, analyzed gut microbiota, explored the relationship between gut microbiota and liver indicators. AT extract significantly decreased lipid levels, promoted ADH, ALDH, and increased the antioxidant activities. Meanwhile, AT extract significantly downregulated the expression of lipid oxidation and inflammatory factors, upregulated alcohol metabolism genes. In addition, 16S rRNA sequencing and analysis showed that AT extract effectively regulated the gut microbiota diversity of ALD mice, significantly improved the structural disturbance of intestinal microflora. AT extract regulated gut microbiota and had a strong correlation with serum, liver-related indexes, and gene expression levels. All these results showed that AT can alleviate alcohol induced liver injury by regulating oxidative stress, inflammatory response, alcohol metabolism, and gut microbiota disorder.
Collapse
Affiliation(s)
- Jianan Wang
- College of Agricultural, Yanbian University, Yanji, Jilin, 133002, China; Food Research Center, Yanbian University, Yanji, Jilin, 133002, China
| | - Aqing Jian
- College of Agricultural, Yanbian University, Yanji, Jilin, 133002, China; Food Research Center, Yanbian University, Yanji, Jilin, 133002, China
| | - Depeng Sun
- College of Agricultural, Yanbian University, Yanji, Jilin, 133002, China; Food Research Center, Yanbian University, Yanji, Jilin, 133002, China
| | - Mingxun Cui
- College of Agricultural, Yanbian University, Yanji, Jilin, 133002, China; Food Research Center, Yanbian University, Yanji, Jilin, 133002, China
| | - Chunxiang Piao
- College of Agricultural, Yanbian University, Yanji, Jilin, 133002, China; Food Research Center, Yanbian University, Yanji, Jilin, 133002, China
| | - Juan Wang
- College of Agricultural, Yanbian University, Yanji, Jilin, 133002, China; Food Research Center, Yanbian University, Yanji, Jilin, 133002, China
| | - Baide Mu
- College of Agricultural, Yanbian University, Yanji, Jilin, 133002, China; Food Research Center, Yanbian University, Yanji, Jilin, 133002, China
| | - Tingyu Li
- College of Agricultural, Yanbian University, Yanji, Jilin, 133002, China; Food Research Center, Yanbian University, Yanji, Jilin, 133002, China
| | - Guanhao Li
- College of Agricultural, Yanbian University, Yanji, Jilin, 133002, China; Food Research Center, Yanbian University, Yanji, Jilin, 133002, China.
| | - Hongmei Li
- College of Agricultural, Yanbian University, Yanji, Jilin, 133002, China; Food Research Center, Yanbian University, Yanji, Jilin, 133002, China.
| |
Collapse
|