1
|
Nagayama M, Gogokhia L, Longman RS. Precision microbiota therapy for IBD: premise and promise. Gut Microbes 2025; 17:2489067. [PMID: 40190259 PMCID: PMC11980506 DOI: 10.1080/19490976.2025.2489067] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/16/2024] [Revised: 12/19/2024] [Accepted: 03/28/2025] [Indexed: 04/11/2025] Open
Abstract
Inflammatory Bowel Disease (IBD) is a spectrum of chronic inflammatory diseases of the intestine that includes subtypes of ulcerative colitis (UC) and Crohn's Disease (CD) and currently has no cure. While IBD results from a complex interplay between genetic, environmental, and immunological factors, sequencing advances over the last 10-15 years revealed signature changes in gut microbiota that contribute to the pathogenesis of IBD. These findings highlight IBD as a disease target for microbiome-based therapies, with the potential to treat the underlying microbial pathogenesis and provide adjuvant therapy to the emerging spectrum of advanced therapies for IBD. Building on the success of fecal microbiota transplantation (FMT) for Clostridioides difficile infection, therapies targeting gut microbiota have emerged as promising approaches for treating IBD; however, unique aspects of IBD pathogenesis highlight the need for more precision in the approach to microbiome therapeutics that leverage aspects of recipient and donor selection, diet and xenobiotics, and strain-specific interactions to enhance the efficacy and safety of IBD therapy. This review focuses on both pre-clinical and clinical studies that support the premise for microbial therapeutics for IBD and aims to provide a framework for the development of precision microbiome therapeutics to optimize clinical outcomes for patients with IBD.
Collapse
Affiliation(s)
- Manabu Nagayama
- Jill Roberts Institute for Research in Inflammatory Bowel Disease, Weill Cornell Medicine, New York, NY, USA
- Jill Roberts Center for Inflammatory Bowel Disease, Division of Gastroenterology and Hepatology, Department of Medicine, Weill Cornell Medicine, New York, NY, USA
| | - Lasha Gogokhia
- Jill Roberts Center for Inflammatory Bowel Disease, Division of Gastroenterology and Hepatology, Department of Medicine, Weill Cornell Medicine, New York, NY, USA
| | - Randy S. Longman
- Jill Roberts Institute for Research in Inflammatory Bowel Disease, Weill Cornell Medicine, New York, NY, USA
- Jill Roberts Center for Inflammatory Bowel Disease, Division of Gastroenterology and Hepatology, Department of Medicine, Weill Cornell Medicine, New York, NY, USA
| |
Collapse
|
2
|
Marques de Souza PR, Keenan CM, Wallace LE, Habibyan YB, Davoli-Ferreira M, Ohland C, Vicentini FA, McCoy KD, Sharkey KA. T cells regulate intestinal motility and shape enteric neuronal responses to intestinal microbiota. Gut Microbes 2025; 17:2442528. [PMID: 39704079 DOI: 10.1080/19490976.2024.2442528] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/18/2024] [Revised: 11/18/2024] [Accepted: 12/09/2024] [Indexed: 12/21/2024] Open
Abstract
How the gut microbiota and immune system maintain intestinal homeostasis in concert with the enteric nervous system (ENS) remains incompletely understood. To address this gap, we assessed small intestinal transit, enteric neuronal density, enteric neurogenesis, intestinal microbiota, immune cell populations and cytokines in wildtype and T-cell deficient germ-free mice colonized with specific pathogen-free (SPF) microbiota, conventionally raised SPF and segmented filamentous bacteria (SFB)-monocolonized mice. SPF microbiota increased small intestinal transit in a T cell-dependent manner. SPF microbiota increased neuronal density in the myenteric and submucosal plexuses of the ileum and colon, similar to conventionally raised SPF mice, independently of T cells. SFB increased neuronal density in the ileum in a T cell-dependent manner, but independently of T cells in the colon. SPF microbiota stimulated enteric neurogenesis (Sox2 expression in enteric neurons) in the ileum in a T cell-dependent manner, but in the colon this effect was T cell-independent. T cells regulated nestin expression in the ENS. SPF colonization increased Th17 cells, RORγT+ Treg cells, and IL-1β and IL-17A levels in the ileum and colon. By neutralizing IL-1β and IL-17A, we observed that they control microbiota-mediated enteric neurogenesis but were not involved in the regulation of motility. Together, these findings provide new insights into the microbiota-neuroimmune dialog that regulates intestinal physiology.
Collapse
Affiliation(s)
- Patricia Rodrigues Marques de Souza
- Department of Health Education, Federal University of Sergipe, Aracaju, SE, Brazil
- Department of Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | - Catherine M Keenan
- Department of Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
- Hotchkiss Brain Institute, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
- Snyder Institute for Chronic Diseases, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | - Laurie E Wallace
- Department of Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
- Hotchkiss Brain Institute, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
- Snyder Institute for Chronic Diseases, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | - Yasaman Bahojb Habibyan
- Department of Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
- Hotchkiss Brain Institute, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
- Snyder Institute for Chronic Diseases, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | - Marcela Davoli-Ferreira
- Department of Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
- Snyder Institute for Chronic Diseases, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | - Christina Ohland
- Department of Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
- Snyder Institute for Chronic Diseases, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
- International Microbiome Centre, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | - Fernando A Vicentini
- Farncombe Family Digestive Health Research Institute, Department of Medicine, McMaster University, Hamilton, ON, Canada
| | - Kathy D McCoy
- Department of Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
- Snyder Institute for Chronic Diseases, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
- International Microbiome Centre, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | - Keith A Sharkey
- Department of Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
- Hotchkiss Brain Institute, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
- Snyder Institute for Chronic Diseases, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| |
Collapse
|
3
|
Okunlola FO, Okunlola AR, Adetuyi BO, Soliman MES, Alexiou A, Papadakis M, Fawzy MN, El-Saber Batiha G. Beyond the gut: Unraveling the multifaceted influence of microbiome on cardiovascular health. Clin Nutr ESPEN 2025; 67:71-89. [PMID: 40064239 DOI: 10.1016/j.clnesp.2025.03.002] [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: 10/09/2024] [Revised: 03/02/2025] [Accepted: 03/04/2025] [Indexed: 03/15/2025]
Abstract
Cardiovascular disease is one of the leading causes of death worldwide. Even while receiving adequate pharmacological treatment for their hypertension, people are nonetheless at greater risk for cardiovascular disease. There is growing evidence that the gut microbiota may have major positive and negative effects on blood pressure and illnesses related with it as more study into this topic is conducted. Trimethylamine n-oxide (TMAO) and short-chain fatty acids (SCFA) are two major by-products of the gut microbiota. TMAO is involved in the formation of other coronary artery diseases, including atherosclerosis and hypertension, while SCFAs play an important role in controlling blood pressure. Numerous investigations have confirmed the established link between dietary salt intake and hypertension. Reducing sodium in the diet is linked to lower rates of cardiovascular disease morbidity and mortality as well as lower rates of blood pressure and hypertension. In both human and animal research, high salt diets increase local and systemic tissue inflammation and compromise gut architecture. Given that the gut microbiota constantly interacts with the immune system and is required for the correct maturation of immune cells, it is scientifically conceivable that it mediates the inflammatory response. This review highlights the therapeutic possibilities for focusing on intestinal microbiomes as well as the potential functions of the gut microbiota and its metabolites in the development of hypertension.
Collapse
Affiliation(s)
- Felix Oladele Okunlola
- Department of Natural Sciences (Biochemistry Option), Faculty of Pure and Applied Sciences, Precious Cornerstone University, Ibadan, Nigeria.
| | - Abimbola Rafiat Okunlola
- Department of Natural Sciences (Biochemistry Option), Faculty of Pure and Applied Sciences, Precious Cornerstone University, Ibadan, Nigeria.
| | - Babatunde Oluwafemi Adetuyi
- Department of Natural Sciences (Biochemistry Option), Faculty of Pure and Applied Sciences, Precious Cornerstone University, Ibadan, Nigeria.
| | - Mahmoud E S Soliman
- Molecular Bio-computation and Drug Design Laboratory, School of Health Sciences, University of KwaZulu-Natal, Westville Campus, Durban 4001, South Africa.
| | - Athanasios Alexiou
- University Centre for Research & Development, Chandigarh University, Chandigarh-Ludhiana Highway, Mohali, Punjab, India; Department of Research & Development, Funogen, Athens, 11741, Greece.
| | - Marios Papadakis
- University Hospital Witten-Herdecke, University of Witten-Herdecke, Heusnerstrasse 40, 42283, Wuppertal, Germany.
| | - Mohamed N Fawzy
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Sinai University- Arish Branch, Arish, 45511, Egypt.
| | - Gaber El-Saber Batiha
- Department of Pharmacology and Therapeutics, Faculty of Veterinary Medicine, Damanhour University, Damanhour, AlBeheira, 22511, Egypt.
| |
Collapse
|
4
|
Cherrier M, Teo TH, Corrêa RO, Picard M, Couesnon A, Lebreton C, Carbone F, Masson C, Schnupf P, Cerf-Bensussan N, Gaboriau-Routhiau V. Hematopoietic MyD88 orchestrates the control of gut colonization by segmented filamentous bacteria. Mucosal Immunol 2025; 18:717-729. [PMID: 40090466 DOI: 10.1016/j.mucimm.2025.03.002] [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: 04/26/2024] [Revised: 02/22/2025] [Accepted: 03/10/2025] [Indexed: 03/18/2025]
Abstract
Host-microbiota cooperation is critical for successful intestinal homeostasis. The commensal segmented filamentous bacteria (SFB) are crucial for orchestrating the post-natal maturation of the host gut immune system and establishing a healthy state of physiological inflammation, which largely depends on their intimate attachment to the ileal mucosa. However, the signaling pathways used by SFB to induce gut immune responses and how such responses ultimately control SFB colonization remain controversial. Using gnotobiotic approaches, we showed that SFB load is controlled by complex interactions involving the gut microbiota and the host immune system. Therefore, to clearly determine the role of host immune responses induced by SFB in directly controlling their growth, immunodeficient mice monocolonized with SFB were used. Here, we show that in the absence of a complex microbiota, the humoral immune response is dispensable to control SFB growth in the jejunum and ileum, shortly and later after colonization. In contrast, MyD88 signaling in myeloid cells is critical for licensing interleukin (IL)-22 production by type 3 innate lymphoid cells (ILC3) and CD4+ T cells, which ultimately limits SFB expansion. Thus, by revisiting the hierarchy of immune mechanisms that directly control SFB growth, our results emphasize the necessary and sufficient role of a hematopoietic MyD88/IL-22 axis.
Collapse
Affiliation(s)
- Marie Cherrier
- Université Paris Cité, Imagine Institute, INSERM UMR1163, Laboratory of Intestinal Immunity, 75015 Paris, France
| | - Teck Hui Teo
- Université Paris Cité, INSERM UMR-S1151, CNRS UMR-S8253, Institut Necker Enfants Malades, F-75015 Paris, France; A*STAR Infectious Diseases Labs (A*STAR ID Labs), Agency for Science, Technology and Research, Singapore
| | - Renan Oliveira Corrêa
- Université Paris Cité, Imagine Institute, INSERM UMR1163, Laboratory of Intestinal Immunity, 75015 Paris, France
| | - Marion Picard
- Université Paris Cité, Imagine Institute, INSERM UMR1163, Laboratory of Intestinal Immunity, 75015 Paris, France
| | - Aurélie Couesnon
- Université Paris Cité, Imagine Institute, INSERM UMR1163, Laboratory of Intestinal Immunity, 75015 Paris, France
| | - Corinne Lebreton
- Université Paris Cité, Imagine Institute, INSERM UMR1163, Laboratory of Intestinal Immunity, 75015 Paris, France
| | - Francesco Carbone
- Université Paris Cité, Imagine Institute, INSERM UMR 1163, Labtech Single-Cell@Imagine, 75015 Paris, France
| | - Cécile Masson
- Université Paris Cité, Imagine Institute, Structure Fédérative de Recherche Necker, Bioinformatics Core Facility, 75015 Paris, France
| | - Pamela Schnupf
- Université Paris Cité, INSERM UMR-S1151, CNRS UMR-S8253, Institut Necker Enfants Malades, F-75015 Paris, France
| | - Nadine Cerf-Bensussan
- Université Paris Cité, Imagine Institute, INSERM UMR1163, Laboratory of Intestinal Immunity, 75015 Paris, France
| | - Valérie Gaboriau-Routhiau
- Université Paris Cité, Imagine Institute, INSERM UMR1163, Laboratory of Intestinal Immunity, 75015 Paris, France; Université Paris-Saclay, INRAe, AgroParisTech, Micalis Institute, 78350 Jouy-en-Josas, France.
| |
Collapse
|
5
|
Chen J, Yuan Q, Yu C, Fu J, Wang P, Tang S, Lin X, Shou Q, Fu H. Integrating 16srRNA sequencing, non-targeted metabolomics, and transcriptome sequencing to explore the mechanism of Total glucosides of paeony alleviating ulcerative colitis. J Chromatogr B Analyt Technol Biomed Life Sci 2025; 1259:124600. [PMID: 40286482 DOI: 10.1016/j.jchromb.2025.124600] [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] [Revised: 04/02/2025] [Accepted: 04/11/2025] [Indexed: 04/29/2025]
Abstract
Total glucosides of paeony (TGP), an active ingredient extracted from the dried root of Paeonia lactiflora Pall., has been approved in China for the treatment of various autoimmune diseases. However, the role and mechanism of TGP in UC have yet to be fully elucidated. This study aims to investigate the regulatory effects and underlying mechanisms of TGP on intestinal homeostasis disruption and immune imbalance in a mouse model of dextran sulfate sodium (DSS)-induced colitis. The results showed that TGP alleviated DSS induced body weight loss, colonic shortening and histopathological changes in mice, and also enhanced the integrity of the intestinal barrier by up-regulating the expression of ZO-1, Occludin and tight junction protein in the colon. The results of 16S and antibiotic cocktail (ABX) experiments showed that TGP alleviated colitis by inhibiting Th17 cell differentiation by correcting intestinal microbial imbalance in UC mice. Mechanism studies showed that TGP inhibited the activation of JAK2/STAT3 signaling pathway in UC mice, and decreased the levels of inflammatory factors in colon supernatant and serum. Importantly, TGP regulates JAK2/STAT3 to inhibit Th17 cell differentiation depending on gut flora. In addition, TGP can also improve the metabolic imbalance in UC mice, especially purine metabolism. In conclusion, TGP promotes the normalization of purine metabolism and relies on gut microbiota to regulate JAK2/STAT3 pathway, inhibit Th17 cell differentiation, and alleviate colitis. Our findings highlight TGP as a promising treatment candidate for ulcerative colitis.
Collapse
Affiliation(s)
- Jianglin Chen
- Second Clinical Medical School, Jinhua Academy, Zhejiang Chinese Medical University, Hangzhou, 310053, China; School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, 311402, China
| | - Qi Yuan
- Second Clinical Medical School, Jinhua Academy, Zhejiang Chinese Medical University, Hangzhou, 310053, China; School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, 311402, China
| | - Cui Yu
- Second Clinical Medical School, Jinhua Academy, Zhejiang Chinese Medical University, Hangzhou, 310053, China; School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, 311402, China
| | - Jie Fu
- Second Clinical Medical School, Jinhua Academy, Zhejiang Chinese Medical University, Hangzhou, 310053, China
| | - Penghao Wang
- Second Clinical Medical School, Jinhua Academy, Zhejiang Chinese Medical University, Hangzhou, 310053, China; School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, 311402, China
| | - Shuiyan Tang
- Second Clinical Medical School, Jinhua Academy, Zhejiang Chinese Medical University, Hangzhou, 310053, China; School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, 311402, China
| | - Xiaochen Lin
- Second Clinical Medical School, Jinhua Academy, Zhejiang Chinese Medical University, Hangzhou, 310053, China
| | - Qiyang Shou
- Second Clinical Medical School, Jinhua Academy, Zhejiang Chinese Medical University, Hangzhou, 310053, China; School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, 311402, China.
| | - Huiying Fu
- Second Clinical Medical School, Jinhua Academy, Zhejiang Chinese Medical University, Hangzhou, 310053, China; School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, 311402, China.
| |
Collapse
|
6
|
Klak K, Maciuszek M, Michalik A, Mazur M, Zawisza M, Pecio A, Nowak B, Chadzinska M. Fire in the belly: Stress and antibiotics induce dysbiosis and inflammation in the gut of common carp. FISH & SHELLFISH IMMUNOLOGY 2025; 161:110301. [PMID: 40157582 DOI: 10.1016/j.fsi.2025.110301] [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: 09/27/2024] [Revised: 03/12/2025] [Accepted: 03/26/2025] [Indexed: 04/01/2025]
Abstract
Fish are exposed to numerous stressors which negatively affect their immune response and increase infection susceptibility. The risk of bacterial infections results in the excessive and preventive use of antibiotics. Therefore, we aimed to study how antibiotic treatment and restraint stress will affect the stress response, microbiota composition, gut morphology, and inflammatory reaction in common carp. Both restraint stress and antibiotic treatment increased cortisol level. Moreover, antibiotics induced dysbiosis in fish gut, manifested by a decrease in the total abundance of bacteria, and a shift in bacteria diversity, including a reduced number of Aeromonas, Bacteroides, Barnesiellaceae, Cetobacterium and Shewanella and an increased abundance of Flavobacterium. To a lesser extent, stress modified gut microbiota, as it decreased bacteria number and slightly changed the microbiota composition by decreasing Cetobacterium abundance and increasing Vibrio abundance. Microbiota of the antibiotic-treated and stressed fish shifted from the beneficial bacterial genera - Cetobacterium and Bacteroides, to the increased presence of unfavorable bacteria such as Brevinema, Flavobacterium and Desulfovibrionaceae. Stress and antibiotic-induced changes in the gut microbiota were related to the changes in the gut morphology when the higher abundance of goblet and rodlet cells and increased secretion activity of goblet cells were observed. Moreover, up-regulation of the expression of genes encoding pro-inflammatory mediators and cytokines involved in the Th17 immune response was present in the gut of the antibiotic-treated and stressed fish. We conclude that in carp antibiotics and stress alter the abundance and composition of the microbiota and induce Th17-dependent inflammatory reaction in the gut. Moreover, our results strongly suggest the interplay of the stress axis and the brain-gut-microbiota axis.
Collapse
Affiliation(s)
- Katarzyna Klak
- Department of Evolutionary Immunology, Institute of Zoology and Biomedical Research, Faculty of Biology, Jagiellonian University, Krakow, Poland; Doctoral School of Exact and Natural Sciences, Jagiellonian University, Krakow, Poland.
| | - Magdalena Maciuszek
- Department of Evolutionary Immunology, Institute of Zoology and Biomedical Research, Faculty of Biology, Jagiellonian University, Krakow, Poland.
| | - Anna Michalik
- Department of Invertebrate Development and Morphology, Institute of Zoology and Biomedical Research, Faculty of Biology, Jagiellonian University, Krakow, Poland.
| | - Mikolaj Mazur
- Department of Evolutionary Immunology, Institute of Zoology and Biomedical Research, Faculty of Biology, Jagiellonian University, Krakow, Poland; Doctoral School of Exact and Natural Sciences, Jagiellonian University, Krakow, Poland.
| | - Maria Zawisza
- Department of Evolutionary Immunology, Institute of Zoology and Biomedical Research, Faculty of Biology, Jagiellonian University, Krakow, Poland; Doctoral School of Exact and Natural Sciences, Jagiellonian University, Krakow, Poland.
| | - Anna Pecio
- Department of Comparative Anatomy, Institute of Zoology and Biomedical Research, Faculty of Biology, Jagiellonian University, Krakow, Poland.
| | - Barbara Nowak
- Institute for Marine and Antarctic Studies - Launceston, University of Tasmania, Launceston, Tasmania, Australia.
| | - Magdalena Chadzinska
- Department of Evolutionary Immunology, Institute of Zoology and Biomedical Research, Faculty of Biology, Jagiellonian University, Krakow, Poland.
| |
Collapse
|
7
|
Park EJ, Lee YS, Jun EM, Lee BW, Park SM, Lee HJ. Immune-Enhancing Effects of Two Potential Probiotic Strains Latilactobacillus curvatus WiKim0169 and Pediococcus acidilactici WiKim0170 in a Cyclophosphamide-Induced Immunosuppression Rat Model. Probiotics Antimicrob Proteins 2025:10.1007/s12602-025-10585-4. [PMID: 40423876 DOI: 10.1007/s12602-025-10585-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/07/2025] [Indexed: 05/28/2025]
Abstract
Emerging evidence suggests that probiotics are beneficial for immunity and play a crucial role in regulating gut microbiota. However, the immune-enhancing effects of specific bacterial species remain uncertain. This study investigated the effects of two potential probiotic strains, Latilactobacillus curvatus WiKim0169 (Wilac L004, W4) and Pediococcus acidilactici WiKim0170 (Wilac L002, W2) isolated from fermented cabbage, on immune enhancement and gut microbiota changes in a cyclophosphamide-induced immunosuppression rat model. The results revealed that W4 and W2 improved natural killer cell activity, serum nitrite, and immunoglobulin levels. They also increased cytokine levels and activated the nuclear factor-κB pathway, substantiating the underlying mechanism of the immune-enhancing effects. Additionally, both strains altered gut microbiota composition by increasing bacteria that are being studied for their potential association with immune function. Taken together, both strains improved immune-related biomarkers and the gut microbiota. These findings suggest W4 and W2 as promising probiotics with immune-enhancing properties.
Collapse
Affiliation(s)
- Eun-Jung Park
- Department of Food and Nutrition, College of BioNano Technology, Gachon University, Seongnam, Gyeonggi-Do, 13120, Republic of Korea
- Institute for Aging and Clinical Nutrition Research, Gachon University, Seongnam, Gyeonggi-Do, 13120, Republic of Korea
| | - You-Suk Lee
- Department of Food and Nutrition, College of BioNano Technology, Gachon University, Seongnam, Gyeonggi-Do, 13120, Republic of Korea
- Institute for Aging and Clinical Nutrition Research, Gachon University, Seongnam, Gyeonggi-Do, 13120, Republic of Korea
| | - Eun-Min Jun
- Department of Food and Nutrition, College of BioNano Technology, Gachon University, Seongnam, Gyeonggi-Do, 13120, Republic of Korea
| | - Byung Wook Lee
- Microbiome R&D Center, Pharmsville Co., Ltd, Seoul, 07793, Republic of Korea
| | - Sang Min Park
- Microbiome R&D Center, Pharmsville Co., Ltd, Seoul, 07793, Republic of Korea
| | - Hae-Jeung Lee
- Department of Food and Nutrition, College of BioNano Technology, Gachon University, Seongnam, Gyeonggi-Do, 13120, Republic of Korea.
- Institute for Aging and Clinical Nutrition Research, Gachon University, Seongnam, Gyeonggi-Do, 13120, Republic of Korea.
- Department of Health Sciences and Technology, GAIHST, Gachon University, Incheon, 21999, Republic of Korea.
- Gachon Biomedical Convergence Institute, Gachon University Gil Medical Center, Incheon, 21565, Republic of Korea.
| |
Collapse
|
8
|
Peng Z, Hou T, Yang K, Zhang J, Mao YH, Hou X. Microecologics and Exercise: Targeting the Microbiota-Gut-Brain Axis for Central Nervous System Disease Intervention. Nutrients 2025; 17:1769. [PMID: 40507038 PMCID: PMC12157277 DOI: 10.3390/nu17111769] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2025] [Revised: 05/20/2025] [Accepted: 05/22/2025] [Indexed: 06/16/2025] Open
Abstract
The gut microbiota (GM) may play a crucial role in the development and progression of central nervous system (CNS) diseases. Microecologics and exercise can influence the composition and function of GM, thereby exerting positive effects on the CNS. Combined interventions of exercise and microecologics are expected to more comprehensively and effectively address CNS diseases through the microbiota-gut-brain axis (MGBA), potentially outperforming single interventions. However, there is currently a lack of relevant reviews on this topic. In this review, we examine the associations between changes in the microbiota and CNS diseases, including Alzheimer's disease (AD), Parkinson's disease (PD), multiple sclerosis (MS), and autism spectrum disorder (ASD). We also summarize studies on various types of microecologics (such as probiotics, prebiotics, synbiotics, and postbiotics) and exercise in improving CNS disease symptoms. Although current individual studies on microecologics and exercise have achieved certain results, the mechanisms underlying their synergistic effects remain unclear. This review aims to explore the theoretical basis, potential mechanisms, and clinical application prospects of combined interventions of microecologics and exercise in improving CNS diseases through the MGBA, providing a scientific basis for the development of more comprehensive and effective therapeutic interventions.
Collapse
Affiliation(s)
- Zhixing Peng
- School of Exercise and Health, Guangzhou Sport University, Guangzhou 510500, China; (Z.P.); (T.H.); (K.Y.); (J.Z.)
| | - Tingting Hou
- School of Exercise and Health, Guangzhou Sport University, Guangzhou 510500, China; (Z.P.); (T.H.); (K.Y.); (J.Z.)
| | - Keer Yang
- School of Exercise and Health, Guangzhou Sport University, Guangzhou 510500, China; (Z.P.); (T.H.); (K.Y.); (J.Z.)
| | - Jiangyu Zhang
- School of Exercise and Health, Guangzhou Sport University, Guangzhou 510500, China; (Z.P.); (T.H.); (K.Y.); (J.Z.)
| | - Yu-Heng Mao
- School of Exercise and Health, Guangzhou Sport University, Guangzhou 510500, China; (Z.P.); (T.H.); (K.Y.); (J.Z.)
- Guangdong Key Laboratory of Human Sports Performance Science, Guangzhou Sport University, Guangzhou 510500, China
| | - Xiaohui Hou
- School of Exercise and Health, Guangzhou Sport University, Guangzhou 510500, China; (Z.P.); (T.H.); (K.Y.); (J.Z.)
- Guangdong Key Laboratory of Human Sports Performance Science, Guangzhou Sport University, Guangzhou 510500, China
| |
Collapse
|
9
|
Zhou YD, Komnick MR, Sepulveda F, Liu G, Nieves-Ortiz E, Meador K, Ndatabaye O, Fatkhullina A, Bozicevich A, Juengel B, Wu-Woods NJ, Naydenkov PM, Kent J, Christiansen N, Madariaga ML, Witkowski P, Ismagilov RF, Esterházy D. Inducible, but not constitutive, pancreatic REG/Reg isoforms are regulated by intestinal microbiota and pancreatic diseases. Mucosal Immunol 2025:S1933-0219(25)00050-9. [PMID: 40398680 DOI: 10.1016/j.mucimm.2025.05.003] [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: 10/19/2024] [Revised: 04/21/2025] [Accepted: 05/09/2025] [Indexed: 05/23/2025]
Abstract
The REG/Reg gene locus encodes a conserved family of potent antimicrobial but also pancreatitis-associated proteins. Here we investigated whether REG/Reg family members differ in their baseline expression levels and abilities to be regulated in the pancreas and gut upon perturbations. We found, in humans and mice, the pancreas and gut differed in REG/Reg isoform levels and preferences, with the duodenum most resembling the pancreas. Pancreatic acinar cells and intestinal enterocytes were the dominant REG producers. Intestinal symbiotic microbes regulated the expression of the same, select Reg members in gut and pancreas. These Reg members had the most STAT3-binding sites close to the transcription start sites and were partially IL-22 dependent. We thus categorized them as "inducible" and others as "constitutive". Indeed, in pancreatic ductal adenocarcinoma and pancreatitis models, only inducible Reg members were upregulated in the pancreas. While intestinal Reg expression remained unchanged upon pancreatic perturbation, pancreatitis altered the microbial composition of the duodenum and feces shortly after disease onset. Our study reveals differential usage and regulation of REG/Reg isoforms as a mechanism for tissue-specific innate immunity, highlights the intimate connection of pancreas and duodenum, and implies a gut-to-pancreas communication axis resulting in a coordinated Reg response.
Collapse
Affiliation(s)
- Yixuan D Zhou
- Department of Pathology, University of Chicago, Chicago, IL, USA; Committee on Immunology, University of Chicago, Chicago, IL, USA
| | - Macy R Komnick
- Department of Pathology, University of Chicago, Chicago, IL, USA; Committee on Immunology, University of Chicago, Chicago, IL, USA
| | | | - Grace Liu
- The College, University of Chicago, Chicago, IL, USA
| | - Elida Nieves-Ortiz
- Department of Pathology, University of Chicago, Chicago, IL, USA; Committee on Immunology, University of Chicago, Chicago, IL, USA
| | - Kelsey Meador
- Department of Pathology, University of Chicago, Chicago, IL, USA; Committee on Immunology, University of Chicago, Chicago, IL, USA
| | | | - Aliia Fatkhullina
- Department of Pathology, University of Chicago, Chicago, IL, USA; Committee on Immunology, University of Chicago, Chicago, IL, USA
| | - Asha Bozicevich
- Department of Pathology, University of Chicago, Chicago, IL, USA; Committee on Immunology, University of Chicago, Chicago, IL, USA
| | - Braden Juengel
- The Transplantation Institute, University of Chicago, Chicago, IL, USA
| | - Natalie J Wu-Woods
- Biology and Bioengineering, California Institute of Technology, Pasadena, CA, USA
| | - Paulina M Naydenkov
- Chemistry and Chemical Engineering, California Institute of Technology, Pasadena, USA
| | - Johnathan Kent
- Department of Surgery, University of Chicago, Chicago, IL, USA
| | | | | | - Piotr Witkowski
- The Transplantation Institute, University of Chicago, Chicago, IL, USA
| | - Rustem F Ismagilov
- Biology and Bioengineering, California Institute of Technology, Pasadena, CA, USA; Chemistry and Chemical Engineering, California Institute of Technology, Pasadena, USA
| | - Daria Esterházy
- Department of Pathology, University of Chicago, Chicago, IL, USA; Committee on Immunology, University of Chicago, Chicago, IL, USA.
| |
Collapse
|
10
|
Pastras P, Aggeletopoulou I, Papantoniou K, Triantos C. Targeting the IL-23 Receptor Gene: A Promising Approach in Inflammatory Bowel Disease Treatment. Int J Mol Sci 2025; 26:4775. [PMID: 40429917 PMCID: PMC12112539 DOI: 10.3390/ijms26104775] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2025] [Revised: 05/09/2025] [Accepted: 05/14/2025] [Indexed: 05/29/2025] Open
Abstract
Inflammatory bowel disease (IBD), which includes Crohn's Disease (CD) and ulcerative colitis (UC), is characterized by chronic inflammation of the gastrointestinal tract. A key component of the inflammatory pathway in IBD is interleukin 23 (IL-23), which promotes the differentiation and maintenance of Th17 cells. These cells are major contributors to intestinal inflammation and the release of pro-inflammatory cytokines. A dysregulated IL-23/Th17 axis can lead to excessive gut inflammation. Notably, IL-23 affects Th17 cell responses differently in UC and CD, fostering IL-17 production in UC and interferon-gamma (IFN-γ) production in CD. Genetic studies have pinpointed specific variants of the IL-23 receptor (IL23R) gene that confer protection against IBD. The R381Q (rs11209026) variant has been linked to a reduced risk of developing both CD and UC. Additionally, other variants, such as G149R (rs76418789) and V362I (rs41313262), inhibit IL23R function by disrupting intracellular trafficking and protein stability. This disruption results in decreased phosphorylation of downstream signal transducers, such as STAT3 and STAT4, and reduced IL23R expression on the cell surface, ultimately dampening the activation of pro-inflammatory pathways. The protective effects of these genetic variants underscore the IL-23/IL23R pathway as a significant therapeutic target in IBD management. Therapies designed to modulate this pathway have the potential to reduce pro-inflammatory cytokine production and enhance anti-inflammatory mechanisms. Ongoing research into the IL23R gene and its variants continues to provide valuable insights, paving the way for more targeted and effective treatments for IBD patients.
Collapse
Affiliation(s)
| | - Ioanna Aggeletopoulou
- Division of Gastroenterology, Department of Internal Medicine, University of Patras, 26504 Patras, Greece; (P.P.); (K.P.); (C.T.)
| | | | | |
Collapse
|
11
|
Andreou E, Papaneophytou C. Boosting Immunity Through Nutrition and Gut Health: A Narrative Review on Managing Allergies and Multimorbidity. Nutrients 2025; 17:1685. [PMID: 40431425 PMCID: PMC12114198 DOI: 10.3390/nu17101685] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2025] [Revised: 05/12/2025] [Accepted: 05/13/2025] [Indexed: 05/29/2025] Open
Abstract
The increasing global burden of allergic diseases and multimorbidity underscores the urgent need for innovative strategies to strengthen immune health. This review explores the complex relationships among nutrition, gut microbiota, immune regulation, allergic diseases, and multimorbidity. It highlights how targeted nutritional and microbial interventions may influence disease outcomes. Dietary components and microbial metabolites dynamically modulated immune function, highlighting the critical role of the gut-immune-metabolism axis in disease pathogenesis and management. Personalized nutrition, guided by advances in diagnostics such as component-resolved diagnostics, basophil activation tests, and epigenetic biomarkers, allows for precise dietary interventions tailored to individual allergy phenotypes and multimorbidity profiles. The Mediterranean diet, breastfeeding, and microbiota-targeted therapies have emerged as effective strategies to enhance immune resilience, reduce inflammation, and manage allergic reactions. Technological advancements, including artificial intelligence-driven dietary assessments, wearable devices, and mobile applications, have further revolutionized personalized dietary management, enabling real-time, precise nutritional monitoring and intervention. Despite these advances, challenges in implementing personalized nutrition persist, including variability in dietary patterns, cultural and socioeconomic factors, and accessibility concerns. Future research should focus on long-term interventional and longitudinal studies to validate precision nutrition strategies and enhance clinical applicability. This integrative approach, combining nutrition, microbiome science, technology, and personalized healthcare, holds substantial promises for sustainable disease prevention and enhanced immune resilience across diverse populations.
Collapse
Affiliation(s)
| | - Christos Papaneophytou
- Department of Life Sciences, School of Life and Health Sciences, University of Nicosia, 2417 Nicosia, Cyprus;
| |
Collapse
|
12
|
Yang T, Hu X, Cao F, Yun F, Jia K, Zhang M, Kong G, Nie B, Liu Y, Zhang H, Li X, Gao H, Shi J, Liang G, Hu G, Kasper DL, Song X, Qian Y. Targeting symbionts by apolipoprotein L proteins modulates gut immunity. Nature 2025:10.1038/s41586-025-08990-4. [PMID: 40369072 DOI: 10.1038/s41586-025-08990-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2024] [Accepted: 04/07/2025] [Indexed: 05/16/2025]
Abstract
The mammalian gut harbours trillions of commensal bacteria that interact with their hosts through various bioactive molecules1,2. However, the mutualistic strategies that hosts evolve to benefit from these symbiotic relationships are largely unexplored. Here we report that mouse enterocytes secrete apolipoprotein L9a and b (APOL9a/b) in the presence of microbiota. By integrating flow cytometry sorting of APOL9-binding bacterial taxa with 16S ribosomal RNA gene sequencing (APOL9-seq), we identify that APOL9a/b, as well as their human equivalent APOL2, coat gut bacteria belonging to the order of Bacteroidales with a high degree of specificity through commensal ceramide-1-phosphate (Cer1P) lipids. Genetic abolition of ceramide-1-phosphate synthesis pathways in gut-dominant symbiote Bacteroides thetaiotaomicron significantly decreases the binding of APOL9a/b to the bacterium. Instead of lysing the bacterial cells, coating of APOL9a/b induces the production of outer membrane vesicles (OMVs) from the target bacteria. Subsequently, the Bacteroides-elicited outer membrane vesicles enhance the host's interferon-γ signalling to promote major histocompatibility complex class II expression in the intestinal epithelial cells. In mice, the loss of Apol9a/b compromises the gut major histocompatibility complex class II-instructed immune barrier function, leading to early mortality from infection by intestinal pathogens. Our data show how a host-elicited factor benefits gut immunological homeostasis by selectively targeting commensal ceramide molecules.
Collapse
Affiliation(s)
- Tao Yang
- Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Xiaohu Hu
- Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Fei Cao
- Key Laboratory of Multi-Cell Systems, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, China
| | - Fenglin Yun
- Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Kaiwen Jia
- Key Laboratory of RNA Science and Engineering, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, China
| | - Mingxiang Zhang
- School of Life Science and Technology, ShanghaiTech University, Shanghai, China
| | - Gaohui Kong
- Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Biyu Nie
- Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Yuexing Liu
- Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Haohao Zhang
- Key Laboratory of Multi-Cell Systems, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, China
| | - Xiaoyu Li
- Center for Inflammatory Bowel Disease Research and Department of Gastroenterology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China
| | - Hongyan Gao
- Center for Infectious Disease Research, School of Medicine, Tsinghua University, Beijing, China
| | - Jiantao Shi
- Key Laboratory of RNA Science and Engineering, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, China
| | - Guanxiang Liang
- Center for Infectious Disease Research, School of Medicine, Tsinghua University, Beijing, China
| | - Guohong Hu
- Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Dennis L Kasper
- Department of Immunology, Blavatnik Institute, Harvard Medical School, Boston, MA, USA
| | - Xinyang Song
- Key Laboratory of Multi-Cell Systems, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, China.
| | - Youcun Qian
- Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China.
- School of Life Science and Technology, ShanghaiTech University, Shanghai, China.
| |
Collapse
|
13
|
Goepp M, Milburn JV, Zhang B, Dong Y, Tyrrell V, Zheng X, Marshall JM, Bolsega S, Basic M, Glendinning L, Ho GT, Satsangi J, Breyer RM, Narumiya S, McSorley HJ, Schwarze JKJ, Anderson CJ, Dockrell DH, Rossi AG, Bleich A, Lucas CD, O'Donnell VB, Mole D, Arends MJ, Zhou Y, Yao C. Age-related impairment of intestinal inflammation resolution through an eicosanoid-immune-microbiota axis. Cell Host Microbe 2025; 33:671-687.e6. [PMID: 40373750 DOI: 10.1016/j.chom.2025.04.014] [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: 07/09/2024] [Revised: 02/27/2025] [Accepted: 04/15/2025] [Indexed: 05/17/2025]
Abstract
Aging manifests a decline of immune function, induces microbiome dysbiosis, drives organ inflammation, and impedes the resolution of inflammation. However, the mechanisms underlying age-related intestinal inflammation remain poorly described. Here, we find that the resolution of T cell-initiated intestinal inflammation is impaired with aging. This impairment is mediated by disrupting the immune-microbiota interplay, controlled by intestinal eicosanoid metabolism. Pharmacologically inhibiting eicosanoid biosynthesis, blocking the prostaglandin E receptor subtype 4 (EP4), or genetically ablating EP4 diminishes age-related impairment of intestinal inflammation resolution. Mechanistically, mononuclear phagocyte-intrinsic eicosanoid-EP4 signaling impedes the resolution of intestinal inflammation through fostering gut microbial dysbiosis and, more importantly, interrupting segmented filamentous bacterial adhesion to the intestinal epithelium. Colonization with EP4-ablated mouse microbiota or segmented filamentous bacteria improves the resolution of intestinal inflammation. These findings reveal that eicosanoid-dependent immune-microbiota interactions impair inflammation resolution in the aged intestine, highlighting potential intervention strategies for improving age-related gut health.
Collapse
Affiliation(s)
- Marie Goepp
- Centre for Inflammation Research, Institute for Regeneration and Repair, The University of Edinburgh, Edinburgh EH16 4UU, UK
| | - Jemma V Milburn
- Centre for Inflammation Research, Institute for Regeneration and Repair, The University of Edinburgh, Edinburgh EH16 4UU, UK
| | - Birong Zhang
- Systems Immunity University Research Institute and Division of Infection and Immunity, Cardiff University, Cardiff CF14 4XN, UK
| | - Yijia Dong
- Centre for Inflammation Research, Institute for Regeneration and Repair, The University of Edinburgh, Edinburgh EH16 4UU, UK
| | - Victoria Tyrrell
- Systems Immunity University Research Institute and Division of Infection and Immunity, Cardiff University, Cardiff CF14 4XN, UK
| | - Xiaozhong Zheng
- Centre for Inflammation Research, Institute for Regeneration and Repair, The University of Edinburgh, Edinburgh EH16 4UU, UK
| | - Jennifer M Marshall
- Centre for Inflammation Research, Institute for Regeneration and Repair, The University of Edinburgh, Edinburgh EH16 4UU, UK
| | - Silvia Bolsega
- Institute for Laboratory Animal Science, Hannover Medical School, Hannover 30625, Germany
| | - Marijana Basic
- Institute for Laboratory Animal Science, Hannover Medical School, Hannover 30625, Germany
| | - Laura Glendinning
- The Roslin Institute & Royal (Dick) School of Veterinary Studies, The University of Edinburgh, Edinburgh EH25 9RG, UK
| | - Gwo-Tzer Ho
- Centre for Inflammation Research, Institute for Regeneration and Repair, The University of Edinburgh, Edinburgh EH16 4UU, UK
| | - Jack Satsangi
- Translational Gastroenterology Unit, Nuffield Department of Medicine, The University of Oxford, Oxford OX3 9DU, UK
| | - Richard M Breyer
- Department of Veterans Affairs, Tennessee Valley Health Authority, and Department of Medicine, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Shuh Narumiya
- Alliance Laboratory for Advanced Medical Research and Department of Drug Discovery Medicine, Medical Innovation Center, Kyoto University Graduate School of Medicine, Kyoto 606-8507, Japan
| | - Henry J McSorley
- Division of Cell Signaling and Immunology, School of Life Sciences, Wellcome Trust Building, The University of Dundee, Dundee DD1 4HN, UK
| | - Jürgen K J Schwarze
- Centre for Inflammation Research, Institute for Regeneration and Repair, The University of Edinburgh, Edinburgh EH16 4UU, UK
| | - Christopher J Anderson
- Centre for Inflammation Research, Institute for Regeneration and Repair, The University of Edinburgh, Edinburgh EH16 4UU, UK
| | - David H Dockrell
- Centre for Inflammation Research, Institute for Regeneration and Repair, The University of Edinburgh, Edinburgh EH16 4UU, UK
| | - Adriano G Rossi
- Centre for Inflammation Research, Institute for Regeneration and Repair, The University of Edinburgh, Edinburgh EH16 4UU, UK
| | - André Bleich
- Institute for Laboratory Animal Science, Hannover Medical School, Hannover 30625, Germany
| | - Christopher D Lucas
- Centre for Inflammation Research, Institute for Regeneration and Repair, The University of Edinburgh, Edinburgh EH16 4UU, UK
| | - Valerie B O'Donnell
- Systems Immunity University Research Institute and Division of Infection and Immunity, Cardiff University, Cardiff CF14 4XN, UK
| | - Damian Mole
- Centre for Inflammation Research, Institute for Regeneration and Repair, The University of Edinburgh, Edinburgh EH16 4UU, UK
| | - Mark J Arends
- Edinburgh Pathology, Cancer Research UK Scotland Centre, Institute of Genetics & Cancer, The University of Edinburgh, Institute of Genetics & Cancer, Edinburgh EH4 2XR, UK
| | - You Zhou
- Systems Immunity University Research Institute and Division of Infection and Immunity, Cardiff University, Cardiff CF14 4XN, UK
| | - Chengcan Yao
- Centre for Inflammation Research, Institute for Regeneration and Repair, The University of Edinburgh, Edinburgh EH16 4UU, UK.
| |
Collapse
|
14
|
Yi J, Jung J, Horton D, Hsieh P, Peng Y, Wang SJ, Newberry R, Ericsson AC, Kim KS, Kau AL, Hsieh CS. A hierarchy of intestinal antigens instructs the CD4 + T cell receptor repertoire. Immunity 2025; 58:1217-1235.e4. [PMID: 40318631 DOI: 10.1016/j.immuni.2025.04.011] [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: 06/03/2024] [Revised: 11/12/2024] [Accepted: 04/10/2025] [Indexed: 05/07/2025]
Abstract
Intestinal CD4+ T cells that are specific for self-, diet-, or commensal-derived antigens are critical for host tolerance but must also be tightly regulated to prevent aberrant activation and conditions like inflammatory bowel disease (IBD). However, it is unclear how the antigen source and location dictate the intestinal TCR repertoire. Here, we hierarchically classified self-, diet-, or microbiota-dependent TCRs using TCliβ TCRβ transgenic mice. This demonstrated that microbiota had a greater influence than diet on CD4+ T cell responses throughout the intestine at homeostasis. Complex bi-directional interactions between microbes and diet were also observed. In the context of murine colitis as a model of IBD, we showed that antigen-free diet substantially altered the microbiota and associated T cell responses, which ameliorated intestinal inflammation. Collectively, these findings suggest how deconvoluting the gut immune interactome may facilitate identifying primary microbial and dietary drivers of T cell responses during health and disease.
Collapse
MESH Headings
- Animals
- Mice
- CD4-Positive T-Lymphocytes/immunology
- Mice, Transgenic
- Gastrointestinal Microbiome/immunology
- Mice, Inbred C57BL
- Inflammatory Bowel Diseases/immunology
- Inflammatory Bowel Diseases/microbiology
- Receptors, Antigen, T-Cell, alpha-beta/genetics
- Receptors, Antigen, T-Cell, alpha-beta/immunology
- Antigens/immunology
- Colitis/immunology
- Colitis/microbiology
- Receptors, Antigen, T-Cell/immunology
- Receptors, Antigen, T-Cell/metabolism
- Intestinal Mucosa/immunology
- Disease Models, Animal
- Intestines/immunology
- Intestines/microbiology
Collapse
Affiliation(s)
- Jaeu Yi
- Department of Internal Medicine, Division of Rheumatology, Washington University School of Medicine, St. Louis, MO 63110, USA; Department of Biological Science, Ajou University, Suwon 16499, Republic of Korea
| | - Jisun Jung
- Department of Internal Medicine, Division of Rheumatology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - David Horton
- Department of Internal Medicine, Division of Rheumatology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Patricia Hsieh
- Department of Internal Medicine, Division of Rheumatology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Yangqing Peng
- Department of Internal Medicine, Division of Rheumatology, Washington University School of Medicine, St. Louis, MO 63110, USA; Department of Molecular Microbiology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Sean J Wang
- Department of Internal Medicine, Division of Rheumatology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Rodney Newberry
- Department of Internal Medicine, Division of Gastroenterology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Aaron C Ericsson
- The Mutant Mouse Resource and Research Center, University of Missouri (MU-MMRRC), Columbia, MO 65201, USA
| | - Kwang Soon Kim
- Department of Life Sciences, Pohang University of Science and Technology, Pohang 37673, Republic of Korea
| | - Andrew L Kau
- Division of Allergy and Immunology, Department of Medicine and Center for Women's Infectious Disease Research, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Chyi-Song Hsieh
- Department of Internal Medicine, Division of Rheumatology, Washington University School of Medicine, St. Louis, MO 63110, USA.
| |
Collapse
|
15
|
Bian Q, Chen Y, Zhang J, Du X, Zhou Y, Zhang Q, Ding C, Wei H, Fu B. Maternal natural killer cells drive neuroimmune disorders in offspring through aberrant secretion of extracellular granzyme B. Immunity 2025:S1074-7613(25)00191-8. [PMID: 40381623 DOI: 10.1016/j.immuni.2025.04.028] [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: 09/28/2023] [Revised: 05/28/2024] [Accepted: 04/22/2025] [Indexed: 05/20/2025]
Abstract
Prenatal viral infections can lead to neurodevelopmental disorders in offspring. However, how viral-induced maternal perturbations impact fetal brain macrophages remains insufficiently clear. Here, we demonstrated that inflammatory decidual natural killer (NK) cells, triggered by viral infection-induced maternal immune activation, drove macrophage activation, neurodevelopmental disorders, and behavioral deficits in offspring. Extracellular granzyme B (GzmB), predominantly released by the maternal CD49a+ tissue-resident NK subset under type I interferon stimulation, crossed the maternal-fetal barrier and promoted interferon-stimulated gene (ISG)-expressing fetal macrophage accumulation and microglial activation. Targeting extracellular GzmB through the systemic administration of the serine protease inhibitor Serpina3n or genetic ablation of Gzmb in maternal NK cells mitigated neuroimmune disorders in the fetal brain. These findings suggest that exposure to a perturbed maternal milieu reprograms decidual NK cell immunity, disrupting fetal neuroimmune homeostasis and increasing offspring susceptibility to neurodevelopmental disorders later in life.
Collapse
Affiliation(s)
- Qiwu Bian
- National Key Laboratory of Immune Response and Immunotherapy, Department of Obstetrics and Gynecology, The First Affiliated Hospital of USTC, Center for Advanced Interdisciplinary Science & Biomedicine of IHM, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230001, Anhui, China
| | - Yihang Chen
- National Key Laboratory of Immune Response and Immunotherapy, Department of Obstetrics and Gynecology, The First Affiliated Hospital of USTC, Center for Advanced Interdisciplinary Science & Biomedicine of IHM, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230001, Anhui, China
| | - Jinghe Zhang
- National Key Laboratory of Immune Response and Immunotherapy, Department of Obstetrics and Gynecology, The First Affiliated Hospital of USTC, Center for Advanced Interdisciplinary Science & Biomedicine of IHM, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230001, Anhui, China
| | - Xianghui Du
- National Key Laboratory of Immune Response and Immunotherapy, Department of Obstetrics and Gynecology, The First Affiliated Hospital of USTC, Center for Advanced Interdisciplinary Science & Biomedicine of IHM, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230001, Anhui, China
| | - Yonggang Zhou
- National Key Laboratory of Immune Response and Immunotherapy, Department of Obstetrics and Gynecology, The First Affiliated Hospital of USTC, Center for Advanced Interdisciplinary Science & Biomedicine of IHM, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230001, Anhui, China
| | - Qiao Zhang
- State Key Laboratory of Genetic Engineering and Collaborative Innovation Center for Genetics and Development, School of Life Sciences, Institute of Biomedical Sciences, Human Phenome Institute, Department of Pathology, Zhongshan Hospital, Fudan University, Shanghai 200433, China
| | - Chen Ding
- State Key Laboratory of Genetic Engineering and Collaborative Innovation Center for Genetics and Development, School of Life Sciences, Institute of Biomedical Sciences, Human Phenome Institute, Department of Pathology, Zhongshan Hospital, Fudan University, Shanghai 200433, China
| | - Haiming Wei
- National Key Laboratory of Immune Response and Immunotherapy, Department of Obstetrics and Gynecology, The First Affiliated Hospital of USTC, Center for Advanced Interdisciplinary Science & Biomedicine of IHM, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230001, Anhui, China.
| | - Binqing Fu
- National Key Laboratory of Immune Response and Immunotherapy, Department of Obstetrics and Gynecology, The First Affiliated Hospital of USTC, Center for Advanced Interdisciplinary Science & Biomedicine of IHM, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230001, Anhui, China.
| |
Collapse
|
16
|
Kim SH, White Z, Gainullina A, Kang S, Kim J, Dominguez JR, Choi Y, Cabrera I, Plaster M, Takahama M, Czepielewski RS, Yeom J, Gunzer M, Hay N, David O, Chevrier N, Sano T, Kim KW. IL-10 sensing by lung interstitial macrophages prevents bacterial dysbiosis-driven pulmonary inflammation and maintains immune homeostasis. Immunity 2025; 58:1306-1326.e7. [PMID: 40306274 DOI: 10.1016/j.immuni.2025.04.004] [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: 02/02/2024] [Revised: 10/02/2024] [Accepted: 04/03/2025] [Indexed: 05/02/2025]
Abstract
Crosstalk between the immune system and the microbiome is critical for maintaining immune homeostasis. Here, we examined this communication and the impact of immune-suppressive IL-10 signaling on pulmonary homeostasis. We found that IL-10 sensing by interstitial macrophages (IMs) is required to prevent spontaneous lung inflammation. Loss of IL-10 signaling in IMs initiated an inflammatory cascade through the activation of classical monocytes and CD4+ T cell subsets, leading to chronic lung inflammation with age. Analyses of antibiotic-treated and germ-free mice established that lung inflammation in the animals lacking IL-10 signaling was triggered by commensal bacteria. 16S rRNA sequencing revealed Delftia acidovorans and Rhodococcus erythropolis as potential drivers of lung inflammation. Intranasal administration of these bacteria or transplantation of human fecal microbiota elicited lung inflammation in gnotobiotic Il10-deficient mice. These findings highlight that IL-10 sensing by IMs contributes to pulmonary homeostasis by preventing lung inflammation caused by commensal dysbiosis.
Collapse
Affiliation(s)
- Seung Hyeon Kim
- Department of Pharmacology and Regenerative Medicine, University of Illinois College of Medicine, Chicago, IL, USA
| | - Zachary White
- Department of Microbiology and Immunology, University of Illinois College of Medicine, Chicago, IL, USA
| | | | - Soeun Kang
- Department of Biochemistry and Genetics, University of Illinois College of Medicine, Chicago, IL, USA
| | - Jiseon Kim
- Department of Pharmacology and Regenerative Medicine, University of Illinois College of Medicine, Chicago, IL, USA
| | - Joseph R Dominguez
- Department of Pharmacology and Regenerative Medicine, University of Illinois College of Medicine, Chicago, IL, USA
| | - Yeonwoo Choi
- Department of Pharmacology and Regenerative Medicine, University of Illinois College of Medicine, Chicago, IL, USA
| | - Ivan Cabrera
- Department of Microbiology and Immunology, University of Illinois College of Medicine, Chicago, IL, USA
| | - Madison Plaster
- Pritzker School of Molecular Engineering, University of Chicago, Chicago, IL, USA
| | - Michihiro Takahama
- Pritzker School of Molecular Engineering, University of Chicago, Chicago, IL, USA
| | - Rafael S Czepielewski
- Immunology Center of Georgia, Department of Physiology, Medical College of Georgia, Augusta University, Augusta, GA, USA; Georgia Cancer Center, Augusta University, Augusta, GA, USA
| | - Jinki Yeom
- Department of Microbiology and Immunology, College of Medicine, Seoul National University, Seoul, Republic of Korea; Cancer Research Institute, Seoul National University, Seoul, Republic of Korea
| | - Matthias Gunzer
- Institute for Experimental Immunology and Imaging, University Hospital Essen, University of Duisburg-Essen, Essen, Germany; Leibniz-Institut für Analytische Wissenschaften - ISAS - e.V., Dortmund, Germany
| | - Nissim Hay
- Department of Biochemistry and Genetics, University of Illinois College of Medicine, Chicago, IL, USA; University of Illinois Cancer Center, University of Illinois at Chicago, Chicago, IL, USA
| | - Odile David
- University of Illinois Cancer Center, University of Illinois at Chicago, Chicago, IL, USA; Department of Pathology, University of Illinois College of Medicine, Chicago, IL, USA
| | - Nicolas Chevrier
- Pritzker School of Molecular Engineering, University of Chicago, Chicago, IL, USA
| | - Teruyuki Sano
- Department of Microbiology and Immunology, University of Illinois College of Medicine, Chicago, IL, USA; University of Illinois Cancer Center, University of Illinois at Chicago, Chicago, IL, USA.
| | - Ki-Wook Kim
- Department of Pharmacology and Regenerative Medicine, University of Illinois College of Medicine, Chicago, IL, USA; University of Illinois Cancer Center, University of Illinois at Chicago, Chicago, IL, USA.
| |
Collapse
|
17
|
Huang Y, Fang R, Xiong T, Li W, Yu N. The Relationship Between Gut Microbiota and Recurrent Spontaneous Abortion. Microorganisms 2025; 13:1073. [PMID: 40431246 PMCID: PMC12114443 DOI: 10.3390/microorganisms13051073] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2025] [Revised: 04/20/2025] [Accepted: 05/02/2025] [Indexed: 05/29/2025] Open
Abstract
Recently, the gut microbiota has been found to be associated with multiple organs and systems in the human body, playing a key role in the occurrence and development of various diseases, such as the gut-brain axis and the gut-liver axis. However, its interaction with miscarriages remains poorly understood. This article reviews the characteristics of gut microbiota and its metabolites in patients with recurrent spontaneous abortion (RSA), the mechanism of gut microbiota inducing RSA, and potential therapeutic strategies. Therefore, it provides a new perspective on the gut microbiota in the pathogenesis and treatment of recurrent abortion, and the prospect of the future research direction of gut microbiota and recurrent abortion is proposed based on existing studies.
Collapse
Affiliation(s)
- Yiyao Huang
- Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430000, China; (Y.H.); (R.F.)
| | - Ruijie Fang
- Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430000, China; (Y.H.); (R.F.)
| | - Ting Xiong
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430000, China; (T.X.); (W.L.)
| | - Wei Li
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430000, China; (T.X.); (W.L.)
| | - Nan Yu
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430000, China; (T.X.); (W.L.)
| |
Collapse
|
18
|
Wang J, Cheng W, Yang R. Nervous system-gut microbiota-immune system axis: future directions for preventing tumor. Front Immunol 2025; 16:1535955. [PMID: 40376000 PMCID: PMC12078214 DOI: 10.3389/fimmu.2025.1535955] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2024] [Accepted: 04/01/2025] [Indexed: 05/18/2025] Open
Abstract
Tumor is one of the leading causes of death worldwide. The occurrence and development of tumors are related to multiple systems and factors such as the immune system, gut microbiota, and nervous system. The immune system plays a critical role in tumor development. Studies have also found that the gut microbiota can directly or indirectly affect tumorigenesis and tumor development. With increasing attention on the tumor microenvironment in recent years, the nervous system has emerged as a novel regulator of tumor development. Some tumor therapies based on the nervous system have also been tested in clinical trials. However, the nervous system can not only directly interact with tumor cells but also indirectly affect tumor development through the gut microbiota. The nervous system-mediated gut microbiota can regulate tumorigenesis, growth, invasion, and metastasis through the immune system. Here, we mainly explore the potential effects of the nervous system-gut microbiota-immune system axis on tumorigenesis and tumor development. The effects of the nervous system-gut microbiota-immune system axis on tumors involve the nervous system regulating immune cells through the gut microbiota, which can prevent tumor development. Meanwhile, the direct effects of the gut microbiota on tumors and the regulation of the immune system by the nervous system, which can affect tumor development, are also reviewed.
Collapse
Affiliation(s)
- Juanjuan Wang
- Department of Immunology, Nankai University School of Medicine, Nankai University, Tianjin, China
| | - Wenyue Cheng
- Department of Immunology, Nankai University School of Medicine, Nankai University, Tianjin, China
| | - Rongcun Yang
- Department of Immunology, Nankai University School of Medicine, Nankai University, Tianjin, China
- State Key Laboratory of Medicinal Chemical Biology, Nankai University, Tianjin, China
- Translational Medicine Institute, Affiliated Tianjin Union Medical Center of Nankai University, Nankai University, Tianjin, China
| |
Collapse
|
19
|
Huang M, Zhang Y, Chen Z, Yu X, Luo S, Peng X, Li X. Gut microbiota reshapes the TNBC immune microenvironment: Emerging immunotherapeutic strategies. Pharmacol Res 2025; 215:107726. [PMID: 40184763 DOI: 10.1016/j.phrs.2025.107726] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/14/2025] [Revised: 03/27/2025] [Accepted: 04/01/2025] [Indexed: 04/07/2025]
Abstract
Triple-negative breast cancer (TNBC) is an aggressive subtype of breast cancer with limited treatment options and poor prognosis. The gut microbiota, a diverse community of microorganisms in the gastrointestinal tract, plays a crucial role in regulating immune responses through the gut-immune axis. Recent studies have highlighted its significant impact on TNBC progression and the efficacy of immunotherapies. This review examines the interactions between gut microbiota and the immune system in TNBC, focusing on key immune cells and pathways involved in tumor immunity. It also explores microbiota modulation strategies, including probiotics, prebiotics, dietary interventions, and fecal microbiota transplantation, as potential methods to enhance immunotherapeutic outcomes. Understanding these mechanisms offers promising avenues for improving treatment efficacy and patient prognosis in TNBC.
Collapse
Affiliation(s)
- Mingyao Huang
- School of Basic Medicine, Putian University, Putian, Fujian 351100, China
| | - Yikai Zhang
- School of Basic Medicine, Putian University, Putian, Fujian 351100, China
| | - Zhaoji Chen
- School of Basic Medicine, Putian University, Putian, Fujian 351100, China
| | - Xin Yu
- Department of Breast Surgery, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital (Fujian Branch of Fudan University Shanghai Cancer Center), Fuzhou, Fujian 350011, China
| | - Shiping Luo
- Department of Breast Surgery, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital (Fujian Branch of Fudan University Shanghai Cancer Center), Fuzhou, Fujian 350011, China.
| | - Xueqiang Peng
- Department of General Surgery, The Fourth Affiliated Hospital, China Medical University, Shenyang 110032, China; Shenyang Clinical Medical Research Center for Diagnosis, Treatment and Health Management, China.
| | - Xuexin Li
- Department of General Surgery, The Fourth Affiliated Hospital, China Medical University, Shenyang 110032, China; Key Laboratory of Precision Diagnosis and Treatment of Gastrointestinal Tumors, Ministry of Education, China Medical University, Shenyang, Liaoning 110122, China; Institute of Health Sciences, China Medical University, Shenyang, Liaoning 110122, China; Department of Physiologyand Pharmacology, Karolinska Institutet, Solna 171 65, Sweden.
| |
Collapse
|
20
|
Zhai J, Li Y, Liu J, Dai C. Neuroimmune interactions: The bridge between inflammatory bowel disease and the gut microbiota. Clin Transl Med 2025; 15:e70329. [PMID: 40400119 PMCID: PMC12095209 DOI: 10.1002/ctm2.70329] [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: 11/08/2024] [Revised: 04/16/2025] [Accepted: 04/21/2025] [Indexed: 05/23/2025] Open
Abstract
BACKGROUND The multidimensional regulatory mechanism of the gut-brain-immune axis in the context of inflammatory bowel disease (IBD) has garnered significant attention, particularly regarding how intestinal microbiota finely regulates immune responses through immune cells and sensory neurons. MAIN BODY Metabolites produced by intestinal microbiota influence the phenotype switching of immune cells via complex signalling pathways, thereby modulating their anti-inflammatory and pro-inflammatory functions during intestinal inflammation. Furthermore, sensory neurons exhibit heightened sensitivity to microbial-derived signals, which is essential for preserving intestinal balance and controlling pathological inflammation by integrating peripheral environmental signals with local immune responses. The dynamic equilibrium between immune cells and the neuroimmunoregulation mediated by sensory neurons collectively sustains immune homeostasis within the intestine. However, this coordination mechanism is markedly disrupted under the pathological conditions associated with IBD. CONCLUSION An in-depth exploration of the interactions among immune cells, gut microbiota and sensory neurons may yield significant insights into the pathological mechanisms underlying IBD and guide the creation of new treatment approaches. KEY POINTS The gut microbiota regulates the gut-brain-immune axis, modulating neuroimmune interactions in IBD. Microbiota-derived metabolites influence immune cells, thereby affecting neurons. Neurons secrete mediators, enabling bidirectional neuroimmune communication essential for intestinal homeostasis. Disruptions contribute to IBD, offering therapeutic targets.
Collapse
Affiliation(s)
- Jinxia Zhai
- Department of GastroenterologyFirst Affiliated Hospital, China Medical UniversityShenyang CityLiaoning ProvinceChina
| | - Yingjie Li
- Department of GastroenterologyFirst Affiliated Hospital, Jinzhou Medical UniversityJinzhou CityLiaoning ProvinceChina
| | - Jiameng Liu
- Department of GastroenterologyFirst Affiliated Hospital, China Medical UniversityShenyang CityLiaoning ProvinceChina
| | - Cong Dai
- Department of GastroenterologyFirst Affiliated Hospital, China Medical UniversityShenyang CityLiaoning ProvinceChina
| |
Collapse
|
21
|
Beaufrère M, Jacoutot M, Nahal RS, Cosentino G, Hutteau-Hamel T, Clavel G, Malfait AJ, Araujo LM, Breban M, Glatigny S. Interleukin 17-producing C-C motif chemokine receptor 6 + conventional CD4 + T cells are arthritogenic in an animal model of spondyloarthritis. J Autoimmun 2025; 153:103413. [PMID: 40163937 DOI: 10.1016/j.jaut.2025.103413] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2024] [Revised: 03/03/2025] [Accepted: 03/24/2025] [Indexed: 04/02/2025]
Abstract
OBJECTIVE Spondyloarthritis (SpA) is a group of chronic inflammatory disorders associated with the human leukocyte antigen (HLA) class I allele HLA-B27. Transgenic rats expressing HLA-B27 and human β2-microglobulin (B27 rats) develop clinical manifestations resembling SpA called rat SpA. IL-17 and TNF are key proinflammatory cytokines implicated in both human and rat SpA. We aimed to determine which T cell subset(s) produce IL-17 and TNF during rat SpA, characterize their tissue distribution and tested their pathogenicity in vivo. METHODS Cytokine production by T cell subsets was evaluated in target tissues and lymphoid organs during rat SpA. Pathogenicity of purified IL-17+ cells was assessed in vivo by cell transfer. Blood samples were used to translate B27 rats findings to SpA patients. RESULTS Conventional CD4+ T cells (Foxp3-; Tconv) and γδ T cells were the main producers of both IL-17 and TNF in B27 rats. IL-17-producing Tconv and γδ T cells were expanded in the colon of premorbid 3-weeks-old B27 rats. C-C motif chemokine receptor 6 (CCR6) allowed the isolation of IL-17+ Tconv (Th17) in rat. Transfer of B27 rat IL-17-producing CCR6+ Tconv but not of γδ T cells into disease-free nude B27 rats induced arthritis, directly demonstrating for the first time the arthritogenic potential of Th17 cells in SpA. Finally, a CCR6+ IL-17+ Tconv expansion enriched for IL-17F production was evidenced in SpA patients. CONCLUSION Our study demonstrates that IL-17+TNF+CCR6+ Th17 cells and IL-17+ γδ T cells are expanded preceding SpA onset in B27 rats and that only IL-17+TNF+CCR6+ Th17 cells can trigger arthritis.
Collapse
Affiliation(s)
- Marie Beaufrère
- UMR1173, Université Paris Saclay, Université de Versailles St Quentin en Yvelines, Inserm, Infection et Inflammation, Montigny le Btx, France; INFLAMEX, Laboratoire d'Excellence, Université Paris Cité, France; Rheumatology Division, Ambroise Paré Hospital (AP-HP), Boulogne-Billancourt, France
| | - Manon Jacoutot
- UMR1173, Université Paris Saclay, Université de Versailles St Quentin en Yvelines, Inserm, Infection et Inflammation, Montigny le Btx, France; INFLAMEX, Laboratoire d'Excellence, Université Paris Cité, France
| | - Roula Said Nahal
- UMR1173, Université Paris Saclay, Université de Versailles St Quentin en Yvelines, Inserm, Infection et Inflammation, Montigny le Btx, France; Rheumatology Division, Ambroise Paré Hospital (AP-HP), Boulogne-Billancourt, France
| | - Gina Cosentino
- UMR1173, Université Paris Saclay, Université de Versailles St Quentin en Yvelines, Inserm, Infection et Inflammation, Montigny le Btx, France
| | - Tom Hutteau-Hamel
- UMR1173, Université Paris Saclay, Université de Versailles St Quentin en Yvelines, Inserm, Infection et Inflammation, Montigny le Btx, France
| | - Gaelle Clavel
- Institut National de la Santé et de la Recherche Médicale, UMR 1125, Université Sorbonne Paris Cité, Paris, France
| | - Aude Jobart Malfait
- UMR1173, Université Paris Saclay, Université de Versailles St Quentin en Yvelines, Inserm, Infection et Inflammation, Montigny le Btx, France; INFLAMEX, Laboratoire d'Excellence, Université Paris Cité, France
| | - Luiza M Araujo
- UMR1173, Université Paris Saclay, Université de Versailles St Quentin en Yvelines, Inserm, Infection et Inflammation, Montigny le Btx, France; INFLAMEX, Laboratoire d'Excellence, Université Paris Cité, France
| | - Maxime Breban
- UMR1173, Université Paris Saclay, Université de Versailles St Quentin en Yvelines, Inserm, Infection et Inflammation, Montigny le Btx, France; INFLAMEX, Laboratoire d'Excellence, Université Paris Cité, France; Rheumatology Division, Ambroise Paré Hospital (AP-HP), Boulogne-Billancourt, France.
| | - Simon Glatigny
- UMR1173, Université Paris Saclay, Université de Versailles St Quentin en Yvelines, Inserm, Infection et Inflammation, Montigny le Btx, France; INFLAMEX, Laboratoire d'Excellence, Université Paris Cité, France.
| |
Collapse
|
22
|
Yang H, Zhao Y, Zhang R, Zhao L, Yang H, Liao X. CiLi (Rosa roxburghii Tratt.) polyphenols improve colitis via gut microbiota-lipid mediator-immunity axis. Food Res Int 2025; 209:116257. [PMID: 40253185 DOI: 10.1016/j.foodres.2025.116257] [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: 12/28/2024] [Revised: 02/12/2025] [Accepted: 03/11/2025] [Indexed: 04/21/2025]
Abstract
Dysbiosis of gut microbiome is one of the most important factors leading to inflammatory bowel disease (IBD). Intake of phytochemicals from fruits and vegetables is an effective way to improve IBD, but how these bioactivators regulate gut microbiota to exert healthy effects remains unclear. Here, we found that pretreatment with CiLi juice, particularly its polyphenol component, alleviated dextran sulfate sodium (DSS)-induced colitis while preserving intestinal barrier integrity. CiLi polyphenols (CL_PP) reduced inflammation and oxidative stress in colon tissue and enriched fecal short-chain fatty acids. Importantly, CL_PP significantly regulated the gut microbiome diversity, increasing beneficial bacteria (e.g., Clostridia_UCG-014, f_Muribaculaceae and Ileibacterium_valens) while decreasing harmful bacteria (Escherichia-Shigella and Romboutsia). Multiomics analysis revealed that CL_PP upregulated bioactive lipid metabolites, particularly those derived from polyunsaturated fatty acids (e.g., resolvin D2, prostaglandin A1, and glycerophosphocholine) related gene expressions (Pltp, Alox15 and Pld4). Additionally, CL-PP downregulated the oxidative stress markers (oxidized glutathione and glutathione peroxidase 3), and immune cell markers (CD8 and CD68). Fecal microbiota transplantation confirmed that the fecal microbiota from CL_PP-treated mice exhibited anti-colitis effects. These effects were diminished in antibiotic-treated mice, underscoring the importance of the gut microbiota in mediating the CL_PP's anti-inflammatory benefits. This study suggests that CL_PP is a potential modulator of gut microbiome dysbiosis for the prevention and treatment of colitis.
Collapse
Affiliation(s)
- Huanzhi Yang
- College of Food Science and Nutritional Engineering, China Agricultural University, Beijing 100083, China
| | - Yang Zhao
- College of Food Science and Nutritional Engineering, China Agricultural University, Beijing 100083, China
| | - Ruiqi Zhang
- College of Food Science and Nutritional Engineering, China Agricultural University, Beijing 100083, China
| | - Liang Zhao
- College of Food Science and Nutritional Engineering, China Agricultural University, Beijing 100083, China
| | - Haixia Yang
- College of Food Science and Nutritional Engineering, China Agricultural University, Beijing 100083, China.
| | - Xiaojun Liao
- College of Food Science and Nutritional Engineering, China Agricultural University, Beijing 100083, China.
| |
Collapse
|
23
|
Fan T, Tai C, Sleiman KC, Cutcliffe MP, Kim H, Liu Y, Li J, Xin G, Grashel M, Baert L, Ekeocha C, Vergenes P, Lima S, Lo WL, Lin J, Hanaoka B, Tankersley TN, Wang M, Zhang X, Tsokos GC, Jarjour W, Longman R, Wu HJJ. Aberrant T follicular helper cells generated by T H17 cell plasticity in the gut promote extraintestinal autoimmunity. Nat Immunol 2025; 26:790-804. [PMID: 40307450 DOI: 10.1038/s41590-025-02125-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2024] [Accepted: 03/12/2025] [Indexed: 05/02/2025]
Abstract
Much remains unknown regarding T follicular helper 17 (TFH17) cells commonly found in autoimmune patients. We previously showed that (and here ask why) egress of gut segmented filamentous bacteria (SFB)-induced TFH cells from Peyer's patches (PP) to systemic sites promotes arthritis. We found splenic TFH17 cells are gut derived. Functional analyses using fate-mapping mice revealed a c-Maf-dependent and SFB-induced TH17-to-TFH cell reprogramming that dominantly occurs in PPs. Unlike conventional TFH cells, TH17-derived TFH cells are highly migratory and atypically concentrated in the dark zone of germinal centers (GCs). Compared to conventional TFH cells, TH17-derived TFH cells express higher levels of TFH-associated functional molecules and more robustly conjugate with B cells. Gain- and loss-of-function studies demonstrated their dominance in promoting GC B cells and arthritis. Notably, murine gut TH17-derived TFH signatures exist in rheumatoid arthritis patients. Thus, gut T cell plasticity generates atypical, potent TFH cells promoting systemic autoimmunity.
Collapse
Affiliation(s)
- Tingting Fan
- Division of Rheumatology and Immunology, Department of Internal Medicine, The Ohio State University Wexner Medical Center, Columbus, OH, USA
- Department of Immunobiology, College of Medicine, University of Arizona, Tucson, AZ, USA
| | - Chi Tai
- Division of Rheumatology and Immunology, Department of Internal Medicine, The Ohio State University Wexner Medical Center, Columbus, OH, USA
| | - Kiah C Sleiman
- Department of Immunobiology, College of Medicine, University of Arizona, Tucson, AZ, USA
| | - Madeline P Cutcliffe
- Division of Rheumatology and Immunology, Department of Internal Medicine, The Ohio State University Wexner Medical Center, Columbus, OH, USA
- Department of Immunobiology, College of Medicine, University of Arizona, Tucson, AZ, USA
| | - Haram Kim
- Division of Rheumatology and Immunology, Department of Internal Medicine, The Ohio State University Wexner Medical Center, Columbus, OH, USA
| | - Ye Liu
- Division of Rheumatology and Immunology, Department of Internal Medicine, The Ohio State University Wexner Medical Center, Columbus, OH, USA
| | - Jianying Li
- Department of Microbial Infection and Immunity, The Ohio State University, Columbus, OH, USA
| | - Gang Xin
- Department of Microbial Infection and Immunity, The Ohio State University, Columbus, OH, USA
- Pelotonia Institute for Immuno-Oncology; The Ohio State Comprehensive Cancer Center, Columbus, Ohio, USA
| | - Mollyanna Grashel
- Division of Rheumatology and Immunology, Department of Internal Medicine, The Ohio State University Wexner Medical Center, Columbus, OH, USA
| | - Laurie Baert
- Division of Rheumatology and Immunology, Department of Internal Medicine, The Ohio State University Wexner Medical Center, Columbus, OH, USA
- Department of Immunobiology, College of Medicine, University of Arizona, Tucson, AZ, USA
| | - Chinwe Ekeocha
- Division of Rheumatology and Immunology, Department of Internal Medicine, The Ohio State University Wexner Medical Center, Columbus, OH, USA
| | - Paige Vergenes
- Division of Rheumatology and Immunology, Department of Internal Medicine, The Ohio State University Wexner Medical Center, Columbus, OH, USA
| | - Svetlana Lima
- Jill Roberts Center and Institute for Research in Inflammatory Bowel Disease, Division of Gastroenterology and Hepatology, Weill Cornell Medicine, New York, NY, USA
| | - Wan-Lin Lo
- Division of Microbiology and Immunology, Department of Pathology, University of Utah School of Medicine, Salt Lake City, UT, USA
| | - Judith Lin
- Division of Rheumatology and Immunology, Department of Internal Medicine, The Ohio State University Wexner Medical Center, Columbus, OH, USA
| | - Beatriz Hanaoka
- Division of Rheumatology and Immunology, Department of Internal Medicine, The Ohio State University Wexner Medical Center, Columbus, OH, USA
| | - Trevor N Tankersley
- Department of Immunobiology, College of Medicine, University of Arizona, Tucson, AZ, USA
| | - Min Wang
- Department of Rheumatology, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Beijing, China
| | - Xuan Zhang
- Department of Rheumatology, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Beijing, China
| | - George C Tsokos
- Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Wael Jarjour
- Division of Rheumatology and Immunology, Department of Internal Medicine, The Ohio State University Wexner Medical Center, Columbus, OH, USA
| | - Randy Longman
- Jill Roberts Center and Institute for Research in Inflammatory Bowel Disease, Division of Gastroenterology and Hepatology, Weill Cornell Medicine, New York, NY, USA
| | - Hsin-Jung Joyce Wu
- Division of Rheumatology and Immunology, Department of Internal Medicine, The Ohio State University Wexner Medical Center, Columbus, OH, USA.
- Department of Immunobiology, College of Medicine, University of Arizona, Tucson, AZ, USA.
- Department of Microbial Infection and Immunity, The Ohio State University, Columbus, OH, USA.
- Pelotonia Institute for Immuno-Oncology; The Ohio State Comprehensive Cancer Center, Columbus, Ohio, USA.
- Arizona Arthritis Center, College of Medicine, University of Arizona, Tucson, AZ, USA.
| |
Collapse
|
24
|
Xie X, Chen X, Wang X, Wang S, Qi P. Dual regulatory effects of gut microbiota and their metabolites in rheumatoid arthritis: balancing pathogenic and protective mechanisms. Front Immunol 2025; 16:1584023. [PMID: 40370449 PMCID: PMC12075411 DOI: 10.3389/fimmu.2025.1584023] [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: 02/26/2025] [Accepted: 04/07/2025] [Indexed: 05/16/2025] Open
Abstract
Rheumatoid arthritis is a chronic autoimmune disorder characterized by destructive, symmetric joint inflammation and synovitis, resulting in substantial disability that profoundly compromises patients' quality of life. Its pathogenesis encompasses complex interactions between genetic and environmental factors. Recent advances in bacterial DNA sequencing technologies have uncovered a significant correlation between the human gut microbiota composition and rheumatoid arthritis progression. Growing clinical and experimental evidence establishes the gut-joint axis as a crucial mediator in rheumatoid arthritis pathogenesis. Comprehensive investigation of gut microbial communities and their metabolites' influence on rheumatoid arthritis mechanisms, coupled with the elucidation of microbiome's bidirectional regulatory effects in disease development, not only deepens our understanding of pathological processes but also establishes a theoretical framework for developing novel diagnostic biomarkers and personalized therapeutic interventions to enhance patient outcomes.
Collapse
Affiliation(s)
- Xingwen Xie
- Affiliated Hospital of Gansu University of Traditional Chinese Medicine, Lanzhou, China
- Gansu University of Traditional Chinese Medicine, Lanzhou, China
| | - Xin Chen
- Affiliated Hospital of Gansu University of Traditional Chinese Medicine, Lanzhou, China
| | - Xuetao Wang
- Gansu University of Traditional Chinese Medicine, Lanzhou, China
| | - Sunli Wang
- Gansu University of Traditional Chinese Medicine, Lanzhou, China
| | - Peng Qi
- Gansu University of Traditional Chinese Medicine, Lanzhou, China
| |
Collapse
|
25
|
Yu Z, Wang Y, Guo Y, Zhu R, Fang Y, Yao Q, Fu H, Zhou A, Ma L, Shou Q. Exploring the Therapeutic and Gut Microbiota-Modulating Effects of Qingreliangxuefang on IMQ-Induced Psoriasis. Drug Des Devel Ther 2025; 19:3269-3291. [PMID: 40322026 PMCID: PMC12048299 DOI: 10.2147/dddt.s492044] [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: 09/08/2024] [Accepted: 03/24/2025] [Indexed: 05/08/2025] Open
Abstract
Purpose To investigate the therapeutic and gut microbiota-modulating effects of Qingreliangxuefang (QRLXF) on psoriasis. Materials and Methods We used network pharmacology, a computational approach, to identify key bioactive compounds and biological targets, and explored the molecular mechanisms of QRLXF. The effects of QRLXF on keratinocyte proliferation and inflammation were evaluated using a mouse model of psoriasis. Changes in the gut microbiota were analyzed via 16SrDNA sequencing, and T cell subsets were assessed using flow cytometry. Results Network pharmacology analysis suggested that QRLXF ameliorated psoriasis by modulating Th17 cell differentiation. Further experiments confirmed the anti-inflammatory effects and relief of psoriatic lesions in IMQ-induced mice. 16SrDNA sequencing revealed significant shifts in the gut microbiota, notably increases in Ligilactobacillus and Lactobacillus genera and decreases in Anaerotruncus, Negativibacillus, Bilophila, and Mucispirillum, suggesting a potential relationship between specific microbiota changes and Th17 cell differentiation. Conclusion QRLXF alleviated psoriatic dermatitis by regulating Th17 cell responses and modifying gut microbiota profiles, highlighting its therapeutic potential for psoriasis treatment.
Collapse
Affiliation(s)
- Zhengyao Yu
- The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Traditional Chinese Medicine), Hangzhou, Zhejiang, 310006, People’s Republic of China
- The Second School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, 310053, People’s Republic of China
- Jinhua Academy, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, 310053, People’s Republic of China
- The First School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, 310053, People’s Republic of China
- Yongkang Hospital of Traditional Chinese Medicine, Jinhua, Zhejiang, 321300, People’s Republic of China
| | - Yingying Wang
- The Second School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, 310053, People’s Republic of China
- Jinhua Academy, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, 310053, People’s Republic of China
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, 310053, People’s Republic of China
| | - Yingxue Guo
- The Second School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, 310053, People’s Republic of China
- Jinhua Academy, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, 310053, People’s Republic of China
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, 310053, People’s Republic of China
| | - Ruotong Zhu
- The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Traditional Chinese Medicine), Hangzhou, Zhejiang, 310006, People’s Republic of China
- The First School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, 310053, People’s Republic of China
| | - Yimiao Fang
- The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Traditional Chinese Medicine), Hangzhou, Zhejiang, 310006, People’s Republic of China
| | - Qinghua Yao
- The Second Affiliated Hospital of Zhejiang Chinese Medical University, Xinhua Hospital of Zhejiang Province, Hangzhou, Zhejiang, 310005, People’s Republic of China
| | - Huiying Fu
- The Second School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, 310053, People’s Republic of China
- Jinhua Academy, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, 310053, People’s Republic of China
- Yongkang Hospital of Traditional Chinese Medicine, Jinhua, Zhejiang, 321300, People’s Republic of China
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, 310053, People’s Republic of China
| | - Ang Zhou
- Department of Dermatology, Yiwu Central Hospital Medical Community Choujiang Hospital District, Yiwu, Zhejiang, 322000, People’s Republic of China
| | - Lili Ma
- The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Traditional Chinese Medicine), Hangzhou, Zhejiang, 310006, People’s Republic of China
| | - Qiyang Shou
- The Second School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, 310053, People’s Republic of China
- Jinhua Academy, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, 310053, People’s Republic of China
- Yongkang Hospital of Traditional Chinese Medicine, Jinhua, Zhejiang, 321300, People’s Republic of China
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, 310053, People’s Republic of China
| |
Collapse
|
26
|
Kolland D, Kuhlmann M, de Almeida GP, Köhler A, Arifovic A, von Strempel A, Pourjam M, Bolsega S, Wurmser C, Steiger K, Basic M, Neuhaus K, Schmidt-Weber CB, Stecher B, Zehn D, Ohnmacht C. A specific microbial consortium enhances Th1 immunity, improves LCMV viral clearance but aggravates LCMV disease pathology in mice. Nat Commun 2025; 16:3902. [PMID: 40274773 PMCID: PMC12022176 DOI: 10.1038/s41467-025-59073-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] [Subscribe] [Scholar Register] [Received: 02/07/2024] [Accepted: 04/10/2025] [Indexed: 04/26/2025] Open
Abstract
Anti-viral immunity can vary tremendously from individual to individual but mechanistic understanding is still scarce. Here, we show that a defined, low complex bacterial community (OMM12) but not the general absence of microbes in germ-free mice leads to a more potent immune response compared to the microbiome of specific-pathogen-free (SPF) mice after a systemic viral infection with LCMV Clone-13. Consequently, gnotobiotic mice colonized with OMM12 have more severe LCMV-induced disease pathology but also enhance viral clearance in the intestinal tract. Mechanistically, single-cell RNA sequencing analysis of adoptively transferred virus-specific T helper cells and endogenous T helper cells in the intestinal tract reveal a stronger pro-inflammatory Th1 profile and a more vigorous expansion in OMM12 than SPF mice. Altogether, our work highlights the causative function of the intestinal microbiome for shaping adaptive anti-viral immunity with implications for vaccination strategies and anti-cancer treatment regimens.
Collapse
Affiliation(s)
- Daphne Kolland
- Center of Allergy and Environment (ZAUM), Technical University and Helmholtz Center, Munich, Germany
| | - Miriam Kuhlmann
- Division of Animal Physiology and Immunology, School of Life Sciences Weihenstephan, Technical University of Munich, Freising, Germany
| | - Gustavo P de Almeida
- Division of Animal Physiology and Immunology, School of Life Sciences Weihenstephan, Technical University of Munich, Freising, Germany
- Center for Infection Prevention (ZIP), School of Life Sciences Weihenstephan, Technical University of Munich, Freising, Germany
| | - Amelie Köhler
- Center of Allergy and Environment (ZAUM), Technical University and Helmholtz Center, Munich, Germany
| | - Anela Arifovic
- Center of Allergy and Environment (ZAUM), Technical University and Helmholtz Center, Munich, Germany
| | - Alexandra von Strempel
- Max von Pettenkofer Institute of Hygiene and Medical Microbiology, Faculty of Medicine, LMU, Munich, Germany
| | - Mohsen Pourjam
- Core Facility Microbiome ZIEL - Institute for Food & Health, Technical University of Munich, Freising, Germany
| | - Silvia Bolsega
- Institute for Laboratory Animal Science and Central Animal Facility, Hannover Medical School, Hannover, Germany
| | - Christine Wurmser
- Division of Animal Physiology and Immunology, School of Life Sciences Weihenstephan, Technical University of Munich, Freising, Germany
- Center for Infection Prevention (ZIP), School of Life Sciences Weihenstephan, Technical University of Munich, Freising, Germany
| | - Katja Steiger
- Institute of Pathology, School of Medicine and Health, Technical University Munich, Munich, Germany
| | - Marijana Basic
- Institute for Laboratory Animal Science and Central Animal Facility, Hannover Medical School, Hannover, Germany
| | - Klaus Neuhaus
- Core Facility Microbiome ZIEL - Institute for Food & Health, Technical University of Munich, Freising, Germany
| | - Carsten B Schmidt-Weber
- Center of Allergy and Environment (ZAUM), Technical University and Helmholtz Center, Munich, Germany
- Member of the German Center of Lung Research (DZL), Partner Site Munich, Munich, Germany
| | - Bärbel Stecher
- Max von Pettenkofer Institute of Hygiene and Medical Microbiology, Faculty of Medicine, LMU, Munich, Germany
- German Center for Infection Research (DZIF), partner site LMU, Munich, Germany
| | - Dietmar Zehn
- Division of Animal Physiology and Immunology, School of Life Sciences Weihenstephan, Technical University of Munich, Freising, Germany.
- Center for Infection Prevention (ZIP), School of Life Sciences Weihenstephan, Technical University of Munich, Freising, Germany.
| | - Caspar Ohnmacht
- Center of Allergy and Environment (ZAUM), Technical University and Helmholtz Center, Munich, Germany.
| |
Collapse
|
27
|
Mears KS, Denny JE, Maslanka JR, Mdluli NV, Hulit EN, Matsuda R, Furth EE, Buffie CG, Abt MC. Therapeutic activation of IL-22-producing innate lymphoid cells enhances host defenses to Clostridioides difficile infection. Cell Rep 2025; 44:115438. [PMID: 40138315 PMCID: PMC12115236 DOI: 10.1016/j.celrep.2025.115438] [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: 06/21/2024] [Revised: 01/02/2025] [Accepted: 02/26/2025] [Indexed: 03/29/2025] Open
Abstract
Clostridioides difficile causes debilitating colitis via secreted toxins that disrupt the intestinal barrier, and toxemia is associated with severe disease. Thus, therapies that fortify the intestinal barrier will reduce the severity of infection. Innate lymphoid cells (ILCs) are critical in the defense against acute C. difficile infection and represent a promising therapeutic target to limit disease. Here, we report that oral administration of the Toll-like receptor (TLR) 7 agonist R848 limits intestinal damage and protects mice from lethal C. difficile infection without impacting pathogen burden or altering the intestinal microbiome. R848 induced interleukin (IL)-22 secretion by ILCs, leading to STAT3 phosphorylation in the intestinal epithelium and increased stem cell proliferation. Genetic ablation of ILCs, IL-22, or epithelial-specific STAT3 abrogated R848-mediated protection. R848 reduced intestinal permeability following infection and limited systemic toxin dissemination. Combined, these data identify an immunostimulatory molecule that activates IL-22 production in ILCs to enhance host tissue defenses following C. difficile infection.
Collapse
Affiliation(s)
- Kevin S Mears
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Joshua E Denny
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Jeffrey R Maslanka
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Nontokozo V Mdluli
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Ellie N Hulit
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Rina Matsuda
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA; Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX, USA
| | - Emma E Furth
- Department of Pathology, University of Pennsylvania Medical Center, Philadelphia, PA, USA
| | - Charlie G Buffie
- Laboratory of Genetically Encoded Small Molecules, The Rockefeller University, New York, NY, USA
| | - Michael C Abt
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA; Institute for Immunology and Immune Health, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
| |
Collapse
|
28
|
Nemati MH, Yazdanpanah E, Kazemi R, Orooji N, Dadfar S, Oksenych V, Haghmorad D. Microbiota-Driven Mechanisms in Multiple Sclerosis: Pathogenesis, Therapeutic Strategies, and Biomarker Potential. BIOLOGY 2025; 14:435. [PMID: 40282300 PMCID: PMC12025160 DOI: 10.3390/biology14040435] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/25/2025] [Revised: 04/11/2025] [Accepted: 04/16/2025] [Indexed: 04/29/2025]
Abstract
Multiple sclerosis (MS) is a well-known, chronic autoimmune disorder of the central nervous system (CNS) involving demyelination and neurodegeneration. Research previously conducted in the area of the gut microbiome has highlighted it as a critical contributor to MS pathogenesis. Changes in the commensal microbiota, or dysbiosis, have been shown to affect immune homeostasis, leading to elevated levels of pro-inflammatory cytokines and disruption of the gut-brain axis. In this review, we provide a comprehensive overview of interactions between the gut microbiota and MS, especially focusing on the immunomodulatory actions of microbiota, such as influencing T-cell balance and control of metabolites, e.g., short-chain fatty acids. Various microbial taxa (e.g., Prevotella and Faecalibacterium) were suggested to lay protective roles, whereas Akkermansia muciniphila was associated with disease aggravation. Interventions focusing on microbiota, including probiotics, prebiotics, fecal microbiota transplantation (FMT), and dietary therapies to normalize gut microbial homeostasis, suppress inflammation and are proven to improve clinical benefits in MS patients. Alterations in gut microbiota represent opportunities for identifying biomarkers for early diagnosis, disease progression and treatment response monitoring. Further studies need to be conducted to potentially address the interplay between genetic predispositions, environmental cues, and microbiota composition to get the precise mechanisms of the gut-brain axis in MS. In conclusion, the gut microbiota plays a central role in MS pathogenesis and offers potential for novel therapeutic approaches, providing a promising avenue for improving clinical outcomes in MS management.
Collapse
Affiliation(s)
- Mohammad Hosein Nemati
- Student Research Committee, Semnan University of Medical Sciences, Semnan 3514799442, Iran
- Department of Immunology, School of Medicine, Semnan University of Medical Sciences, Semnan 3514799442, Iran
| | - Esmaeil Yazdanpanah
- Immunology Research Center, Mashhad University of Medical Sciences, Mashhad 9177948564, Iran
| | - Roya Kazemi
- Student Research Committee, Semnan University of Medical Sciences, Semnan 3514799442, Iran
- Department of Immunology, School of Medicine, Semnan University of Medical Sciences, Semnan 3514799442, Iran
| | - Niloufar Orooji
- Student Research Committee, Semnan University of Medical Sciences, Semnan 3514799442, Iran
- Department of Immunology, School of Medicine, Semnan University of Medical Sciences, Semnan 3514799442, Iran
| | - Sepehr Dadfar
- Student Research Committee, Semnan University of Medical Sciences, Semnan 3514799442, Iran
- Department of Immunology, School of Medicine, Semnan University of Medical Sciences, Semnan 3514799442, Iran
| | - Valentyn Oksenych
- Broegelmann Research Laboratory, Department of Clinical Science, University of Bergen, 5020 Bergen, Norway
| | - Dariush Haghmorad
- Department of Immunology, School of Medicine, Semnan University of Medical Sciences, Semnan 3514799442, Iran
| |
Collapse
|
29
|
Lejeune A, Zhou C, Ercelen D, Putzel G, Yao X, Guy AR, Pawline M, Podkowik M, Pironti A, Torres VJ, Shopsin B, Cadwell K. Sex-dependent gastrointestinal colonization resistance to MRSA is microbiota and Th17 dependent. eLife 2025; 13:RP101606. [PMID: 40197396 PMCID: PMC11978300 DOI: 10.7554/elife.101606] [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] [Indexed: 04/10/2025] Open
Abstract
Gastrointestinal (GI) colonization by methicillin-resistant Staphylococcus aureus (MRSA) is associated with a high risk of transmission and invasive disease in vulnerable populations. The immune and microbial factors that permit GI colonization remain unknown. Male sex is correlated with enhanced Staphylococcus aureus nasal carriage, skin and soft tissue infections, and bacterial sepsis. Here, we established a mouse model of sexual dimorphism during GI colonization by MRSA. Our results show that in contrast to male mice that were susceptible to persistent colonization, female mice rapidly cleared MRSA from the GI tract following oral inoculation in a manner dependent on the gut microbiota. This colonization resistance displayed by female mice was mediated by an increase in IL-17A+ CD4+ T cells (Th17) and dependent on neutrophils. Ovariectomy of female mice increased MRSA burden, but gonadal female mice that have the Y chromosome retained enhanced Th17 responses and colonization resistance. Our study reveals a novel intersection between sex and gut microbiota underlying colonization resistance against a major widespread pathogen.
Collapse
Affiliation(s)
- Alannah Lejeune
- Department of Microbiology, New York University School of MedicineNew YorkUnited States
- Department of Medicine, Division of Infectious Diseases, New York University School of MedicineNew YorkUnited States
| | - Chunyi Zhou
- Department of Microbiology, New York University School of MedicineNew YorkUnited States
- Department of Medicine, Division of Infectious Diseases, New York University School of MedicineNew YorkUnited States
| | - Defne Ercelen
- Department of Medicine, Division of Gastroenterology and Hepatology, New York University Langone HealthNew YorkUnited States
| | - Gregory Putzel
- Department of Microbiology, New York University School of MedicineNew YorkUnited States
- Antimicrobial-Resistant Pathogens Program, New York University School of MedicineNew YorkUnited States
| | - Xiaomin Yao
- Department of Medicine, Division of Infectious Diseases, New York University School of MedicineNew YorkUnited States
| | - Alyson R Guy
- NYU-Regeneron Veterinary Postdoctoral Training Program in Laboratory Animal Medicine, Division of Comparative Medicine, New York University School of MedicineNew YorkUnited States
| | - Miranda Pawline
- Department of Medicine, Division of Gastroenterology and Hepatology, New York University Langone HealthNew YorkUnited States
| | - Magdalena Podkowik
- Department of Medicine, Division of Infectious Diseases, New York University School of MedicineNew YorkUnited States
- Antimicrobial-Resistant Pathogens Program, New York University School of MedicineNew YorkUnited States
| | - Alejandro Pironti
- Department of Microbiology, New York University School of MedicineNew YorkUnited States
- Antimicrobial-Resistant Pathogens Program, New York University School of MedicineNew YorkUnited States
| | - Victor J Torres
- Department of Microbiology, New York University School of MedicineNew YorkUnited States
- Department of Host-Microbe Interactions, St. Jude Children’s Research HospitalMemphisUnited States
| | - Bo Shopsin
- Department of Microbiology, New York University School of MedicineNew YorkUnited States
- Department of Medicine, Division of Infectious Diseases, New York University School of MedicineNew YorkUnited States
- Antimicrobial-Resistant Pathogens Program, New York University School of MedicineNew YorkUnited States
| | - Ken Cadwell
- Department of Medicine, Division of Gastroenterology and Hepatology, University of Pennsylvania Perelman School of MedicinePhiladelphiaUnited States
- Department of Pathobiology, University of Pennsylvania Perelman School of Veterinary MedicinePhiladelphiaUnited States
| |
Collapse
|
30
|
Zhou Y, Zhang L, Lin L, Liu Y, Li Q, Zhao Y, Zhang Y. Associations of prenatal organophosphate esters exposure with risk of eczema in early childhood, mediating role of gut microbiota. JOURNAL OF HAZARDOUS MATERIALS 2025; 487:137250. [PMID: 39827805 DOI: 10.1016/j.jhazmat.2025.137250] [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: 11/16/2024] [Revised: 01/01/2025] [Accepted: 01/15/2025] [Indexed: 01/22/2025]
Abstract
Few epidemiological evidence has focused on the impact of organophosphate esters (OPEs) and the risk of eczema, and underlying role of gut microbiota. Based on the Shanghai Maternal-Child Pairs Cohort, a nested case-control study including 332 eczema cases and 332 controls was conducted. Umbilical cord blood and stools were collected for OPEs detection and gut microbiota sequencing, separately. Eczema cases were identified using the International Study of Asthma and Allergies in Childhood core questionnaire and clinical diagnosis. The environmental risk score (ERS) for OPEs was developed to quantify OPEs burden. Conditional logistic regression models, multivariate analysis by linear models, negative-binomial hurdle regression, and mediation analysis were employed. Tris(2-butoxyethyl) phosphate (TBP), tris (2-butoxy ethyl) phosphate (TBEP), 2-ethylhexyl diphenyl phosphate (EHDPP), and tris(1,3-dichloro-2-propyl) phosphate (TDCPP) had detection rates > 50 %, with median concentrations ranged from 0.11 to 2.71 μg/L. TBP (OR = 1.12, 95 % CI: 1.01, 1.25), TDCPP (OR = 1.32, 95 % CI: 1.09, 1.59), and ERS (OR = 6.44, 95 % CI: 3.47, 11.94) were associated with elevated risk of eczema. OPEs exposure was correlated with increased alpha diversity and the abundance of several pathogenic bacteria, such as Klebsiella. Negative associations were observed between OPEs exposure and the abundances of Lachnospiraceae genera. Additionally, a positive correlation was identified between alpha diversity and the risk of eczema during childhood. Alpha diversity indices and Lachnospiraceae serve as significant mediators in this relationship. Results of this study indicate that prenatal exposure to OPEs is linked to an elevated risk of eczema and gut microbiota dysbiosis, potentially contributing to immunotoxicity of OPEs during early life.
Collapse
Affiliation(s)
- Yuhan Zhou
- School of Exercise and Health, Shanghai University of Sport, Shanghai, China; Key Lab of Health Technology Assessment, National Health Commission of the People's Republic of China, Fudan University, Shanghai 200032, China
| | - Liyi Zhang
- Key Lab of Health Technology Assessment, National Health Commission of the People's Republic of China, Fudan University, Shanghai 200032, China; Key Laboratory of Public Health Safety, Ministry of Education, School of Public Health, Fudan University, Shanghai 200032, China
| | - Ling Lin
- Nantong Center for Disease Control & Prevention, Jiangsu 226007, China
| | - Yang Liu
- Key Lab of Health Technology Assessment, National Health Commission of the People's Republic of China, Fudan University, Shanghai 200032, China; Key Laboratory of Public Health Safety, Ministry of Education, School of Public Health, Fudan University, Shanghai 200032, China
| | - Qiang Li
- Putuo District Center for Disease Control & Prevention, Shanghai 200333, China
| | - Yingya Zhao
- Key Lab of Health Technology Assessment, National Health Commission of the People's Republic of China, Fudan University, Shanghai 200032, China; Key Laboratory of Public Health Safety, Ministry of Education, School of Public Health, Fudan University, Shanghai 200032, China
| | - Yunhui Zhang
- Key Lab of Health Technology Assessment, National Health Commission of the People's Republic of China, Fudan University, Shanghai 200032, China; Key Laboratory of Public Health Safety, Ministry of Education, School of Public Health, Fudan University, Shanghai 200032, China.
| |
Collapse
|
31
|
Saadawi A, Mair F, Rosenwald E, Hoces D, Slack E, Kopf M. Investigating Polyreactivity of CD4 + T Cells to the Intestinal Microbiota. Eur J Immunol 2025; 55:e202451484. [PMID: 40223653 DOI: 10.1002/eji.202451484] [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: 08/28/2024] [Revised: 03/12/2025] [Accepted: 03/13/2025] [Indexed: 04/15/2025]
Abstract
Antigen-specific recognition of microbiota by T cells enforces tolerance at homeostasis. Conversely, dysbiosis leads to imbalanced T-cell responses, triggering inflammatory and autoimmune diseases. Despite their significance, the identities of immunogenic microbial antigens remain largely enigmatic. Here, we leveraged a sensitive, unbiased, genome-wide screening platform to identify peptides from Akkermansia muciniphila (AKK) and Bacteroides thetaiotaomicron (BT) recognized by CD4+ T cells. The platform is based on screening peptide libraries using an NFAT-fluorescence reporter cell line transduced with a retrovirus encoding an MHC-TCR (MCR) hybrid molecule. We discovered several novel epitopes from AKK and BT. T-cell hybridomas reactive to AKK and BT bacteria demonstrated polyreactivity to microbiota-derived peptides in co-cultures with MCR reporter cells. Steady-state T cells recognized these epitopes in an MHC-restricted fashion. Intriguingly, most of the identified epitopes are broadly conserved within the given phylum and originate from membrane and intracellular proteins. Ex vivo stimulation of CD4+ T cells from mice vaccinated with the identified peptides revealed mono-specific IFN-γ and IL-17 responses. Our work showcases the potential of the MCR system for identifying immunogenic microbial epitopes, providing a valuable resource. Our study facilitates decoding antigen specificity in immune system-bacterial interactions, with applications in understanding microbiome and pathogenic bacterial immunity.
Collapse
Affiliation(s)
- Ahmed Saadawi
- Department of Biology, Institute of Molecular Health Sciences, ETH Zürich, Zürich, Switzerland
| | - Florian Mair
- Department of Biology, Institute of Molecular Health Sciences, ETH Zürich, Zürich, Switzerland
| | - Esther Rosenwald
- Department of Biology, Institute of Molecular Health Sciences, ETH Zürich, Zürich, Switzerland
| | - Daniel Hoces
- Department of Health Sciences and Technology, ETH Zürich, Zürich, Switzerland
| | - Emma Slack
- Department of Health Sciences and Technology, ETH Zürich, Zürich, Switzerland
| | - Manfred Kopf
- Department of Biology, Institute of Molecular Health Sciences, ETH Zürich, Zürich, Switzerland
| |
Collapse
|
32
|
Schütz B, Krause FF, Taudte RV, Zaiss MM, Luu M, Visekruna A. Modulation of Host Immunity by Microbiome-Derived Indole-3-Propionic Acid and Other Bacterial Metabolites. Eur J Immunol 2025; 55:e202451594. [PMID: 40170399 PMCID: PMC11962249 DOI: 10.1002/eji.202451594] [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: 11/07/2024] [Revised: 03/05/2025] [Accepted: 03/06/2025] [Indexed: 04/03/2025]
Abstract
In recent years, we have witnessed a rapidly growing interest in the intricate communications between intestinal microorganisms and the host immune system. Research on the human microbiome is evolving from merely descriptive and correlative studies to a deeper mechanistic understanding of the bidirectional interactions between gut microbiota and the mucosal immune system. Despite numerous challenges, it has become increasingly evident that an imbalance in gut microbiota composition, known as dysbiosis, is associated with the development and progression of various metabolic, immune, cancer, and neurodegenerative disorders. A growing body of evidence highlights the importance of small molecules produced by intestinal commensal bacteria, collectively referred to as gut microbial metabolites. These metabolites serve as crucial diffusible messengers, translating the microbial language to host cells. This review aims to explore the complex and not yet fully understood molecular mechanisms through which microbiota-derived metabolites influence the activity of the immune cells and shape immune reactions in the gut and other organs. Specifically, we will discuss recent research that reveals the close relationship between microbial indole-3-propionic acid (IPA) and mucosal immunity. Furthermore, we will emphasize the beneficial effects of IPA on intestinal inflammation and discuss its potential clinical implications.
Collapse
Affiliation(s)
- Burkhard Schütz
- Institute of Anatomy and Cell BiologyPhilipps‐University MarburgMarburgGermany
| | - Felix F. Krause
- Institute for Medical Microbiology and HygienePhilipps‐University MarburgMarburgGermany
| | - R. Verena Taudte
- Core Facility for MetabolomicsDepartment of MedicinePhilipps‐University MarburgMarburgGermany
| | - Mario M. Zaiss
- Department of Internal Medicine 3Rheumatology and ImmunologyFriedrich‐Alexander‐Universität Erlangen‐Nürnberg (FAU) and Universitätsklinikum ErlangenErlangenGermany
- Deutsches Zentrum Immuntherapie (DZI)Friedrich‐Alexander‐Universität Erlangen‐Nürnberg (FAU) and Universitätsklinikum ErlangenErlangenGermany
| | - Maik Luu
- Lehrstuhl für Zelluläre Immuntherapie, Medizinische Klinik und Poliklinik IIUniversitätsklinikum WürzburgWürzburgGermany
| | - Alexander Visekruna
- Institute for Medical Microbiology and HygienePhilipps‐University MarburgMarburgGermany
| |
Collapse
|
33
|
Yoo E, Jo Y, Park J, Hong SW. Immune tolerance to foreign antigens in the intestine: mechanisms mediated by CD4+ T cells. BMB Rep 2025; 58:158-168. [PMID: 40176601 PMCID: PMC12041928] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2025] [Revised: 02/24/2025] [Accepted: 03/17/2025] [Indexed: 04/04/2025] Open
Abstract
The immune system encounters a diverse array of antigens, both self and foreign, necessitating mechanisms to maintain tolerance and prevent harmful inflammatory responses. CD4+ T cells, crucial in orchestrating immune responses, play a critical role in mediating tolerance to both self and foreign antigens. While the mechanisms of CD4+ T cell-mediated tolerance to self-antigens are well-documented, the understanding of tolerance to foreign antigens, including those from commensal microbes and food, remains incomplete. This review discusses recent progress in the mechanisms underlying immune tolerance to foreign antigens, with a focus on the role of CD4+ T cells. We explore how inflammatory and tolerogenic CD4+ T cell subsets are developed and maintained. Moreover, we delve into the complexities of immune responses to commensal microbes and food antigens by reviewing recent findings, highlighting the immunological contexts that shape immune tolerance. Understanding these mechanisms enhances our comprehension of how immune tolerance is established and sustained, providing insights into potential therapeutic approaches for managing chronic inflammatory diseases resulting from a loss of immune tolerance to foreign antigens. [BMB Reports 2025; 58(4): 158-168].
Collapse
Affiliation(s)
- Eunbi Yoo
- Department of Biotechnology, College of Life Science and Biotechnology, Yonsei University, Seoul 03722, Korea
| | - Yeleen Jo
- Department of Biotechnology, College of Life Science and Biotechnology, Yonsei University, Seoul 03722, Korea
| | - Jooyoun Park
- Department of Biotechnology, College of Life Science and Biotechnology, Yonsei University, Seoul 03722, Korea
| | - Sung-Wook Hong
- Department of Biotechnology, College of Life Science and Biotechnology, Yonsei University, Seoul 03722, Korea
| |
Collapse
|
34
|
Mikhail SG, O'Dwyer DN. The lung microbiome in interstitial lung disease. Breathe (Sheff) 2025; 21:240167. [PMID: 40255291 PMCID: PMC12004254 DOI: 10.1183/20734735.0167-2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2024] [Accepted: 01/17/2025] [Indexed: 04/22/2025] Open
Abstract
Interstitial lung disease (ILD) is a heterogeneous chronic form of lung disease. The pathogenesis of ILD is poorly understood and a common form of ILD, idiopathic pulmonary fibrosis (IPF) is associated with poor prognosis. There is evidence for substantial dysregulated immune responses in ILD. The microbiome is a key regulator of the immune response, and the lung microbiome correlates with alveolar immunity and clinical outcomes in ILD. Most observational lung microbiome studies have been conducted in patients with IPF. A consistent observation in these studies is that the bacterial burden of the lung is elevated in patients with IPF and predicts mortality. However, our understanding of the mechanism is incomplete and our understanding of the role of the lung microbiome in other forms of ILD is limited. The microbiomes of the oropharynx and gut may have implications for the lung microbiome and pulmonary immunity in ILD but require substantial further research. Here, we discuss the studies supporting a role for the lung microbiome in the pathogenesis of IPF, and briefly describe the putative role of the oral-lung axis and the gut-lung axis in ILD.
Collapse
Affiliation(s)
- Sheridan G. Mikhail
- Division of Pulmonary and Critical Care Medicine, Dept. of Internal Medicine, University of Michigan Medical School, Ann Arbor, MI, USA
| | - David N. O'Dwyer
- Division of Pulmonary and Critical Care Medicine, Dept. of Internal Medicine, University of Michigan Medical School, Ann Arbor, MI, USA
- Dept. of Microbiology and Immunology, University of Michigan Medical School, Ann Arbor, MI, USA
| |
Collapse
|
35
|
Edwards M, Brockmann L. Microbiota-dependent modulation of intestinal anti-inflammatory CD4 + T cell responses. Semin Immunopathol 2025; 47:23. [PMID: 40167791 DOI: 10.1007/s00281-025-01049-6] [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: 10/10/2024] [Accepted: 03/08/2025] [Indexed: 04/02/2025]
Abstract
Barrier organs such as the gastrointestinal tract, lungs, and skin are colonized by diverse microbial strains, including bacteria, viruses, and fungi. These microorganisms, collectively known as the commensal microbiota, play critical roles in maintaining health by defending against pathogens, metabolizing nutrients, and providing essential metabolites. In the gut, commensal-derived antigens are frequently sensed by the intestinal immune system. Maintaining tolerance toward these beneficial microbial species is crucial, as failure to do so can lead to chronic inflammatory conditions like inflammatory bowel disease (IBD) and can even affect systemic immune or metabolic health. The immune system carefully regulates responses to commensals through various mechanisms, including the induction of anti-inflammatory CD4⁺ T cell responses. Foxp3⁺ regulatory T cells (Foxp3+ Tregs) and Type 1 regulatory T cells (Tr1) play a major role in promoting tolerance, as both cell types can produce the anti-inflammatory cytokine IL-10. In addition to these regulatory T cells, effector T cell subsets, such as Th17 cells, also adopt anti-inflammatory functions within the intestine in response to the microbiota. This process of anti-inflammatory CD4+ T cell induction is heavily influenced by the microbiota and their metabolites. Microbial metabolites affect intestinal epithelial cells, promoting the secretion of anti-inflammatory mediators that create a tolerogenic environment. They also modulate intestinal dendritic cells (DCs) and macrophages, inducing a tolerogenic state, and can interact directly with T cells to drive anti-inflammatory CD4⁺ T cell functionality. The disrupted balance of these signals may result in chronic inflammation, with broader implications for systemic health. In this review, we highlight the intricate interplays between commensal microorganisms and the immune system in the gut. We discuss how the microbiota influences the differentiation of commensal-specific anti-inflammatory CD4⁺ T cells, such as Foxp3⁺ Tregs, Tr1 cells, and Th17 cells, and explore the mechanisms through which microbial metabolites modulate these processes. We further discuss the innate signals that prime and commit these cells to an anti-inflammatory fate.
Collapse
Affiliation(s)
- Madeline Edwards
- Department of Microbiology and Immunology, Vagelos College of Physicians and Surgeons, Columbia University, New York, NY, 10032, USA
| | - Leonie Brockmann
- Department of Systems Biology, Vagelos College of Physicians and Surgeons, Columbia University, New York, NY, 10032, USA.
- Human Biology-Microbiome-Quantum Research Center (WPI-Bio2Q), Keio University, Tokyo, 108-8345, Japan.
| |
Collapse
|
36
|
Molina PA, Edell CJ, Dunaway LS, Kellum CE, Muir RQ, Jennings MS, Colson JC, De Miguel C, Rhoads MK, Buzzelli AA, Harrington LE, Meza-Perez S, Randall TD, Botta D, Müller DN, Pollock DM, Maynard CL, Pollock JS. Aryl Hydrocarbon Receptor Activation Promotes Effector CD4+ T Cell Homeostasis and Restrains Salt-Sensitive Hypertension. FUNCTION 2025; 6:zqaf001. [PMID: 39779302 PMCID: PMC11931625 DOI: 10.1093/function/zqaf001] [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: 06/25/2024] [Revised: 12/18/2024] [Accepted: 01/06/2025] [Indexed: 01/11/2025] Open
Abstract
Excess dietary salt and salt-sensitivity contribute to cardiovascular disease. Distinct T cell phenotypic responses to high salt and hypertension, as well as influences from environmental cues, are not well understood. The aryl hydrocarbon receptor (AhR) is activated by dietary ligands, promoting T cell and systemic homeostasis. We hypothesized that activating AhR supports CD4+ homeostatic functions, such as cytokine production and mobilization, in response to high salt intake while mitigating salt-sensitive hypertension. In the intestinal mucosa, we demonstrate that a high-salt diet (HSD) is a key driving factor, independent of hypertension, in diminishing interleukin 17A (IL-17A) production by CD4+ T (Th17) cells without disrupting circulating cytokines associated with Th17 function. Previous studies suggest that hypertensive patients and individuals on a HSD are deficient in AhR ligands or agonistic metabolites. We found that activating AhR augments Th17 cells during experimental salt-sensitive hypertension. Further, we demonstrate that activating AhR in vitro contributes to sustaining Th17 cells in the setting of excess salt. Using photoconvertible Kikume Green-Red mice, we also revealed that HSD drives CD4+ T cell mobilization. Next, we found that excess salt augments T cell mobilization markers, validating HSD-driven T cell migration. Also, we found that activating AhR mitigates HSD-induced T cell migration markers. Using telemetry in a model of experimental salt-sensitivity, we found that activating AhR prevents the development of salt-sensitive hypertension. Collectively, stimulating AhR through dietary ligands facilitates immunologic and systemic functions amid excess salt intake and restrains the development of salt-sensitive hypertension.
Collapse
Affiliation(s)
- Patrick A Molina
- Cardio-Renal Physiology and Medicine Section, Division of Nephrology,
Department of Medicine, University of Alabama at Birmingham, Birmingham, AL 35223, USA
| | - Claudia J Edell
- Cardio-Renal Physiology and Medicine Section, Division of Nephrology,
Department of Medicine, University of Alabama at Birmingham, Birmingham, AL 35223, USA
| | - Luke S Dunaway
- Cardio-Renal Physiology and Medicine Section, Division of Nephrology,
Department of Medicine, University of Alabama at Birmingham, Birmingham, AL 35223, USA
| | - Cailin E Kellum
- Cardio-Renal Physiology and Medicine Section, Division of Nephrology,
Department of Medicine, University of Alabama at Birmingham, Birmingham, AL 35223, USA
| | - Rachel Q Muir
- Department of Pathology, University of Alabama at Birmingham, Birmingham, AL 35223, USA
| | - Melissa S Jennings
- Department of Pathology, University of Alabama at Birmingham, Birmingham, AL 35223, USA
| | - Jackson C Colson
- Cardio-Renal Physiology and Medicine Section, Division of Nephrology,
Department of Medicine, University of Alabama at Birmingham, Birmingham, AL 35223, USA
| | - Carmen De Miguel
- Cardio-Renal Physiology and Medicine Section, Division of Nephrology,
Department of Medicine, University of Alabama at Birmingham, Birmingham, AL 35223, USA
| | - Megan K Rhoads
- Cardio-Renal Physiology and Medicine Section, Division of Nephrology,
Department of Medicine, University of Alabama at Birmingham, Birmingham, AL 35223, USA
| | - Ashlyn A Buzzelli
- Department of Cell, Developmental and Integrative Biology, University of Alabama at Birmingham, Birmingham, AL 35223, USA
| | - Laurie E Harrington
- Department of Cell, Developmental and Integrative Biology, University of Alabama at Birmingham, Birmingham, AL 35223, USA
| | - Selene Meza-Perez
- Division of Clinical Immunology and Rheumatology, Department of Medicine, University of Alabama at Birmingham, Birmingham, AL 35223, USA
| | - Troy D Randall
- Division of Clinical Immunology and Rheumatology, Department of Medicine, University of Alabama at Birmingham, Birmingham, AL 35223, USA
| | - Davide Botta
- Department of Microbiology, University of Alabama at Birmingham, Birmingham, AL 35223, USA
- Immunology Institute, Heersink School of Medicine, University of Alabama at Birmingham, Birmingham, AL 35223, USA
| | - Dominik N Müller
- Experimental and Clinical Research Center, Max Delbrück Center for Molecular Medicine and Charité-Universitätsmedizin Berlin
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Robert-Rössle-Str. 10
- Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt Universität Zu Berlin, Lindenberger Weg 80, Berlin 13092, Germany
| | - David M Pollock
- Cardio-Renal Physiology and Medicine Section, Division of Nephrology,
Department of Medicine, University of Alabama at Birmingham, Birmingham, AL 35223, USA
| | - Craig L Maynard
- Department of Pathology, University of Alabama at Birmingham, Birmingham, AL 35223, USA
| | - Jennifer S Pollock
- Cardio-Renal Physiology and Medicine Section, Division of Nephrology,
Department of Medicine, University of Alabama at Birmingham, Birmingham, AL 35223, USA
| |
Collapse
|
37
|
Wilson RP, Rink L, Tükel Ç. Microbiota and cancer: unraveling the significant influence of microbial communities on cancer treatment. Cancer Metastasis Rev 2025; 44:42. [PMID: 40120010 DOI: 10.1007/s10555-025-10256-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 03/25/2025]
Affiliation(s)
- R Paul Wilson
- Center for Microbiology and Immunology, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, USA
| | - Lori Rink
- Fox Chase Cancer Center, Philadelphia, PA, USA.
| | - Çagla Tükel
- Center for Microbiology and Immunology, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, USA.
- Fox Chase Cancer Center, Philadelphia, PA, USA.
| |
Collapse
|
38
|
Peng C, Lei P, Qi H, Zhu Q, Huang C, Fu J, Zhao C. Effect of fecal microbiota transplantation on diabetic wound healing through the IL-17A-mTOR-HIF1α signaling axis. Appl Environ Microbiol 2025; 91:e0201924. [PMID: 40019272 PMCID: PMC11921319 DOI: 10.1128/aem.02019-24] [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: 10/12/2024] [Accepted: 01/24/2025] [Indexed: 03/01/2025] Open
Abstract
Diabetes is the third most common chronic disorder worldwide. Diabetic wounds are a severe complication that is costly and often results in non-traumatic lower limb amputation. Recent investigations have demonstrated that the gut microbiota as a "virtual organ" can regulate metabolic diseases like diabetes. Fecal microbiota transplantation (FMT) is an innovative therapeutic approach for promoting wound healing, but its function remains incompletely defined. A diabetes model was established by supplying mice with a high-fat diet and performing an intraperitoneal injection of streptozotocin. Diabetic wounds were then created, followed by bacterial transplantation. The relevant indexes of wound healing were evaluated to verify the promoting effect of FMT on the diabetic wounds. Human skin keratinocytes were also cultured, and cell scratch experiments were conducted to further investigate the underlying mechanism. The FMT regulated the levels of specific bacteria in the diabetic mice and helped restore the balance of intestinal microbes. This transplantation also enhanced wound healing in the diabetic mice by augmenting the closure rate, accelerating re-epithelialization, and boosting collagen deposition in skin wounds. Furthermore, FMT promoted the production of IL-17A, which significantly enhanced the growth and movement of human keratinocytes. Inhibiting molecules related to the IL-17A-mTOR-HIF1α signaling axis were shown to hinder wound re-epithelialization.This study clarifies the function of the IL-17A-mTOR-HIF1α signaling axis in the utilization of FMT in diabetic wound healing, providing a new therapeutic method and target for promoting the healing of diabetic wounds. IMPORTANCE The Intestinal microbiota, as the organ with the largest number of microorganisms in the body, plays a crucial role in the physiological functions of the human body. Normal microbiota can be involved in various functions such as energy absorption, metabolism, and immunity of the body, and microbiota imbalance is related to many diseases such as obesity and diabetes. Diabetes, as one of the world's three major chronic diseases, is a significant health issue that troubles more than a billion people globally. Diabetic wounds are a problem that all diabetic patients must confront when undergoing surgery, and it is an important cause of non-traumatic amputations. Exploring the role of intestinal microorganisms in the wound-healing process of diabetic mice can offer the possibility of using microorganisms as a therapeutic means to intervene in clinically related diseases.
Collapse
Affiliation(s)
- Chenmei Peng
- Qinghai University Affiliated Hospital, Qinghai University, Xining, China
| | - Pan Lei
- Department of General Practice Medicine, Qinghai University Affiliated Hospital, Xining, China
| | - Hongying Qi
- Department of Endocrinology, Qinghai University Affiliated Hospital, Xining, China
| | - Qianjun Zhu
- Department of Endocrinology, Qinghai Province People’s Hospital, Xining, China
| | - Chushun Huang
- Qinghai University Affiliated Hospital, Qinghai University, Xining, China
| | - Ju Fu
- Qinghai University Affiliated Hospital, Qinghai University, Xining, China
| | - Chengyu Zhao
- Department of Geriatrics, Qinghai University Affiliated Hospital, Xining, China
| |
Collapse
|
39
|
Oami T, Shimazui T, Yumoto T, Otani S, Hayashi Y, Coopersmith CM. Gut integrity in intensive care: alterations in host permeability and the microbiome as potential therapeutic targets. J Intensive Care 2025; 13:16. [PMID: 40098052 PMCID: PMC11916345 DOI: 10.1186/s40560-025-00786-y] [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: 01/31/2025] [Accepted: 02/21/2025] [Indexed: 03/19/2025] Open
Abstract
BACKGROUND The gut has long been hypothesized to be the "motor" of critical illness, propagating inflammation and playing a key role in multiple organ dysfunction. However, the exact mechanisms through which impaired gut integrity potentially contribute to worsened clinical outcome remain to be elucidated. Critical elements of gut dysregulation including intestinal hyperpermeability and a perturbed microbiome are now recognized as potential therapeutic targets in critical care. MAIN BODY The gut is a finely tuned ecosystem comprising ~ 40 trillion microorganisms, a single cell layer intestinal epithelia that separates the host from the microbiome and its products, and the mucosal immune system that actively communicates in a bidirectional manner. Under basal conditions, these elements cooperate to maintain a finely balanced homeostasis benefitting both the host and its internal microbial community. Tight junctions between adjacent epithelial cells selectively transport essential molecules while preventing translocation of pathogens. However, critical illness disrupts gut barrier function leading to increased gut permeability, epithelial apoptosis, and immune activation. This disruption is further exacerbated by a shift in the microbiome toward a "pathobiome" dominated by pathogenic microbes with increased expression of virulence factors, which intensifies systemic inflammation and accelerates organ dysfunction. Research has highlighted several potential therapeutic targets to restore gut integrity in the host, including the regulation of epithelial cell function, modulation of tight junction proteins, and inhibition of epithelial apoptosis. Additionally, microbiome-targeted therapies, such as prebiotics, probiotics, fecal microbiota transplantation, and selective decontamination of the digestive tract have also been extensively investigated to promote restoration of gut homeostasis in critically ill patients. Future research is needed to validate the potential efficacy of these interventions in clinical settings and to determine if the gut can be targeted in an individualized fashion. CONCLUSION Increased gut permeability and a disrupted microbiome are common in critical illness, potentially driving dysregulated systemic inflammation and organ dysfunction. Therapeutic strategies to modulate gut permeability and restore the composition of microbiome hold promise as novel treatments for critically ill patients.
Collapse
Affiliation(s)
- Takehiko Oami
- Department of Surgery and Emory Critical Care Center, Emory University School of Medicine, 101 Woodruff Circle, Suite WMB 5105, Atlanta, GA, 30322, USA
- Department of Emergency and Critical Care Medicine, Chiba University Graduate School of Medicine, Chiba, Japan
| | - Takashi Shimazui
- Department of Surgery and Emory Critical Care Center, Emory University School of Medicine, 101 Woodruff Circle, Suite WMB 5105, Atlanta, GA, 30322, USA
- Department of Emergency and Critical Care Medicine, Chiba University Graduate School of Medicine, Chiba, Japan
| | - Tetsuya Yumoto
- Department of Surgery and Emory Critical Care Center, Emory University School of Medicine, 101 Woodruff Circle, Suite WMB 5105, Atlanta, GA, 30322, USA
- Department of Emergency, Critical Care and Disaster Medicine, Faculty of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama, Japan
| | - Shunsuke Otani
- Department of Surgery and Emory Critical Care Center, Emory University School of Medicine, 101 Woodruff Circle, Suite WMB 5105, Atlanta, GA, 30322, USA
- Department of Emergency and Critical Care Medicine, Chiba University Graduate School of Medicine, Chiba, Japan
| | - Yosuke Hayashi
- Department of Surgery and Emory Critical Care Center, Emory University School of Medicine, 101 Woodruff Circle, Suite WMB 5105, Atlanta, GA, 30322, USA
- Department of Emergency and Critical Care Medicine, Chiba University Graduate School of Medicine, Chiba, Japan
| | - Craig M Coopersmith
- Department of Surgery and Emory Critical Care Center, Emory University School of Medicine, 101 Woodruff Circle, Suite WMB 5105, Atlanta, GA, 30322, USA.
| |
Collapse
|
40
|
Tang MH, Ligthart I, Varga S, Lebeer S, van Overveld FJ, Rijkers GT. Mutual Interactions Between Microbiota and the Human Immune System During the First 1000 Days of Life. BIOLOGY 2025; 14:299. [PMID: 40136555 PMCID: PMC11940030 DOI: 10.3390/biology14030299] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/03/2025] [Revised: 02/25/2025] [Accepted: 03/14/2025] [Indexed: 03/27/2025]
Abstract
The development of the human immune system starts during the fetal period in a largely, but probably not completely, sterile environment. During and after birth, the immune system is exposed to an increasingly complex microbiota. The first microbiota encountered during passage through the birth canal colonize the infant gut and induce the tolerance of the immune system. Transplacentally derived maternal IgG as well as IgA from breast milk protect the infant from infections during the first 100 days, during which the immune system further develops and immunological memory is formed. The Weaning and introduction of solid food expose the immune system to novel (food) antigens and allow for other microbiota to colonize. The cells and molecules involved in the mutual and intricate interactions between microbiota and the developing immune system are now beginning to be recognized. These include bacterial components such as polysaccharide A from Bacteroides fragilis, as well as bacterial metabolites such as the short-chain fatty acid butyrate, indole-3-aldehyde, and indole-3-propionic acid. All these, and probably more, bacterial metabolites have specific immunoregulatory functions which shape the development of the human immune system during the first 1000 days of life.
Collapse
Affiliation(s)
- Muy Heang Tang
- Department of Science and Engineering, University College Roosevelt, 4331 CB Middelburg, The Netherlands; (M.H.T.); (I.L.); (S.V.); (F.J.v.O.)
| | - Ishbel Ligthart
- Department of Science and Engineering, University College Roosevelt, 4331 CB Middelburg, The Netherlands; (M.H.T.); (I.L.); (S.V.); (F.J.v.O.)
| | - Samuel Varga
- Department of Science and Engineering, University College Roosevelt, 4331 CB Middelburg, The Netherlands; (M.H.T.); (I.L.); (S.V.); (F.J.v.O.)
| | - Sarah Lebeer
- Lab of Applied Microbiology and Biotechnology, Department of Bioscience Engineering, University of Antwerp, 2020 Antwerpen, Belgium;
| | - Frans J. van Overveld
- Department of Science and Engineering, University College Roosevelt, 4331 CB Middelburg, The Netherlands; (M.H.T.); (I.L.); (S.V.); (F.J.v.O.)
| | - Ger T. Rijkers
- Department of Science and Engineering, University College Roosevelt, 4331 CB Middelburg, The Netherlands; (M.H.T.); (I.L.); (S.V.); (F.J.v.O.)
| |
Collapse
|
41
|
Yan R, Jia D, Qi Y, Wang Q, Chen S. Intestinal tissue-resident memory T cells: Characteristics, functions under physiological and pathological conditions and spatial specificity. J Adv Res 2025:S2090-1232(25)00181-X. [PMID: 40096943 DOI: 10.1016/j.jare.2025.03.021] [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: 01/04/2025] [Revised: 03/08/2025] [Accepted: 03/14/2025] [Indexed: 03/19/2025] Open
Abstract
BACKGROUND Tissue-resident memory T (TRM) cells are a distinct subset of memory T cells that persist in non-lymphoid tissues, providing localized and rapid immune responses to infection and malignancy. Unlike circulating memory T cells, TRM cells have unique homing and functional characteristics that are shaped by the tissue microenvironment. In the gut, TRM cells play a pivotal role in maintaining mucosal immunity, exhibiting phenotypic and functional heterogeneity in different intestinal compartments and in response to aging and pathological conditions. AIM OF REVIEW This review aims to systematically examine the definition, spatial heterogeneity and functional roles of intestinal TRM (iTRM) cells. It highlights their contributions to physiological immunity, their involvement in pathological processes such as inflammatory bowel disease (IBD) and colorectal cancer (CRC), and their age-related dynamics. The review also explores emerging therapeutic implications of modulating iTRM cells for intestinal health and disease management. KEY SCIENTIFIC CONCEPTS OF REVIEW: iTRM cells are defined by surface markers like CD69 and CD103, transcriptional regulators such as Hobit, Runx3, Blimp-1, as well as cytokine signals including TGF-β, IFN-β, IL-12. They exhibit spatial and functional heterogeneity across intestinal layers (epithelium versus lamina propria) and regions (small intestine versus colon). In IBD, iTRM cells play a dual role, contributing to both inflammation and tissue repair, whereas in CRC, specific subsets of iTRM cells (e.g., CD8+ CD103+ CD39+) are associated with enhanced antitumor immunity. Aging impacts iTRM functionality, with shifts in the CD4+/CD8+ ratio and reduced cytokine production in elderly individuals. Insights into the metabolic, transcriptional, and environmental regulation of iTRM cells provide avenues for targeted therapies in intestinal diseases, cancer immunotherapy, and interventions to delay intestinal aging.
Collapse
Affiliation(s)
- Ruochen Yan
- Department of Gastroenterology, Sir Run Run Shaw Hospital, Zhejiang University, Hangzhou, Zhejiang Province 310058, China; Institution of Gastroenterology, Zhejiang University, Hangzhou, Zhejiang Province 310058, China
| | - Dingjiacheng Jia
- Department of Gastroenterology, Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, Zhejiang Province 310058, China; Institution of Gastroenterology, Zhejiang University, Hangzhou, Zhejiang Province 310058, China
| | - Yadong Qi
- Department of Gastroenterology, Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, Zhejiang Province 310058, China; Institution of Gastroenterology, Zhejiang University, Hangzhou, Zhejiang Province 310058, China
| | - Qiwen Wang
- Department of Gastroenterology, Sir Run Run Shaw Hospital, Zhejiang University, Hangzhou, Zhejiang Province 310058, China; Institution of Gastroenterology, Zhejiang University, Hangzhou, Zhejiang Province 310058, China
| | - Shujie Chen
- Department of Gastroenterology, Sir Run Run Shaw Hospital, Zhejiang University, Hangzhou, Zhejiang Province 310058, China; Institution of Gastroenterology, Zhejiang University, Hangzhou, Zhejiang Province 310058, China; Cancer Center, Zhejiang University, Hangzhou, Zhejiang Province 310001, China.
| |
Collapse
|
42
|
Xie Z, Chen Z, Chai Y, Yao W, Ma G. Unveiling the placental bacterial microbiota: implications for maternal and infant health. Front Physiol 2025; 16:1544216. [PMID: 40161970 PMCID: PMC11949977 DOI: 10.3389/fphys.2025.1544216] [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: 12/12/2024] [Accepted: 02/24/2025] [Indexed: 04/02/2025] Open
Abstract
The human placenta is a unique organ that forms under specific physiological conditions and plays a crucial role in nutrient and metabolite exchange between the mother and fetus. Research on the placenta is important for understanding maternal-fetal diseases. Traditionally, the placenta was considered "sterile," but advancements in detection techniques have revealed the presence of a low level of microorganisms. This discovery challenges the traditional notion that the uterine placenta is sterile. The revelation of this truth marks a significant breakthrough in medical research, prompting more researchers to focus on this vital organ, the placenta. Placental microbial communities may originate from the oral, vaginal, and intestinal microbiota of expectant mothers. These microorganisms may reach the maternal-fetal interface, collectively shaping the placental microbiota and contributing to the composition of normal placental microbial communities. Abnormal placental microbial communities may be associated with some pregnancy complications and fetal developmental issues such as preterm birth, gestational hypertension, fetal growth restriction, and gestational diabetes mellitus. Intervention strategies targeting microbial communities, which include modulation of placental microbiota composition or function, such as probiotics, may help prevent or treat complications related to abnormal placental microbiota during pregnancy.
Collapse
Affiliation(s)
- Zhuojun Xie
- General Medicine Department, Clinical Medical College & Affiliated Hospital of Chengdu University, Chengdu University, Chengdu, China
| | - Zhongsheng Chen
- Department of Colorectal Cancer Surgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Yang Chai
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Jinan University, Guangzhou, China
| | - Wang Yao
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Jinan University, Guangzhou, China
| | - Guangyu Ma
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Jinan University, Guangzhou, China
| |
Collapse
|
43
|
Strutzenberg TS, Mann MD, Li X, Shin H, Kelsey J, Aiyer S, Yu J, Gray G, Zhang Z, Shan Z, Zhou B, Zheng Y, Griffin PR, Lyumkis D. Nucleosome Engagement Regulates RORγt Structure and Dynamics. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.03.10.642251. [PMID: 40161694 PMCID: PMC11952427 DOI: 10.1101/2025.03.10.642251] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 04/02/2025]
Abstract
The retinoic acid-related orphan receptor gamma (RORγt) acts as the major transcriptional activator in Th17 cell development and function to mediate adaptive immune defenses against pathogenic infection. RORγt engages accessible DNA response elements in the genome and interplays with coactivator proteins and accessory transcription factors to drive gene expression. However, how the chromatin environment mediates RORγt structure, dynamics, and function remains unclear. Here, we profile how the nucleosome promotes or restricts access to the main RORγt DNA response elements found in native enhancers and promoters, revealing preferential binding in regions of free DNA and nucleosomal entry/exit sites, with single base-pair resolution. Solution phase measurements using hydrogen deuterium exchange coupled to mass spectrometry identify novel allosteric effects that influence RORγt binding and mediate chromatin dynamics. A high-resolution structure of RORγt bound to the nucleosome reveals how structured elements assemble to confer binding specificity and avidity to chromatin substrates. The observations suggest an activation model where RORγt binding to chromatinized DNA promotes coregulator recruitment and chromatin decompaction.
Collapse
Affiliation(s)
| | - Matthew D. Mann
- The Herbert Wertheim UF Scripps Institute for Biomedical Innovation & Technology, Jupiter, FL, USA
| | - Xiandu Li
- The Salk Institute for Biological Studies, La Jolla, CA, USA
| | - Hyejeong Shin
- The Salk Institute for Biological Studies, La Jolla, CA, USA
| | - Jordan Kelsey
- The Salk Institute for Biological Studies, La Jolla, CA, USA
| | - Sriram Aiyer
- The Salk Institute for Biological Studies, La Jolla, CA, USA
| | - Jingting Yu
- The Salk Institute for Biological Studies, La Jolla, CA, USA
| | - Gennavieve Gray
- The Salk Institute for Biological Studies, La Jolla, CA, USA
| | - Zeyuan Zhang
- The Salk Institute for Biological Studies, La Jolla, CA, USA
| | - Zelin Shan
- The Salk Institute for Biological Studies, La Jolla, CA, USA
| | - Bo Zhou
- The Salk Institute for Biological Studies, La Jolla, CA, USA
| | - Ye Zheng
- The Salk Institute for Biological Studies, La Jolla, CA, USA
| | - Patrick R. Griffin
- The Herbert Wertheim UF Scripps Institute for Biomedical Innovation & Technology, Jupiter, FL, USA
| | - Dmitry Lyumkis
- The Salk Institute for Biological Studies, La Jolla, CA, USA
- Graduate School for Biological Sciences, Section of Molecular Biology, University of California San Diego, La Jolla, California, USA
| |
Collapse
|
44
|
Dehghanizai AB, Stewart CJ, Thomas RH. The microbiome: what a neurologist needs to know. Pract Neurol 2025:pn-2024-004400. [PMID: 40081897 DOI: 10.1136/pn-2024-004400] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/27/2025] [Indexed: 03/16/2025]
Abstract
The gastrointestinal tract is inhabited by trillions of micro-organisms that form the gut microbiome, which serves various functions that can influence neurological pathways. It can release metabolites that could affect the nervous system. The bidirectional communication between the intestine and the central nervous system is known as the gut-brain axis. This communication can be impacted by the microbiota in various direct and indirect ways. There has been a suggested connection between the microbiome and many neurological disorders, including epilepsy, Alzheimer's disease, Parkinson's disease and multiple sclerosis. This has been explored in human and animal studies. While no microbial biomarkers have been identified yet, alterations in several taxa have been suggested to be associated with disease states. The potential of the microbiome to modulate neurological function has sparked multiple clinical trials using gut-altering treatments, some with positive preliminary results.
Collapse
Affiliation(s)
- Anna B Dehghanizai
- Translational and Clinical Research Institute, Newcastle University, Newcastle upon Tyne, UK
| | | | - Rhys H Thomas
- Neurology, Royal Victoria Infirmary, Newcastle upon Tyne, UK
- Institute of Neuroscience, Newcastle University, Newcastle upon Tyne, UK
| |
Collapse
|
45
|
Band VI, Gribonika I, Stacy A, Bouladoux N, Mistry S, Burns A, Perez-Chaparro PJ, Chau J, Enamorado M, Nagai M, Bhushan V, Golec DP, Schwartzberg PL, Hourigan SK, Nita-Lazar A, Belkaid Y. Sulfide is a keystone metabolite for gut homeostasis and immunity. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.03.06.641928. [PMID: 40161817 PMCID: PMC11952330 DOI: 10.1101/2025.03.06.641928] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 04/02/2025]
Abstract
Hydrogen sulfide is a gaseous, reactive molecule specifically enriched in the gastrointestinal tract. Here, we uncover a non-redundant role for sulfide in the control of both microbial and immune homeostasis of the gut. Notably, depletion of sulfide via both pharmaceutical and dietary interventions led to a profound collapse of CD4 T cells in the ileum of the small intestine lamina propria and significant impact on microbial ecology. As a result, mice with reduced sulfide within the gut were deficient in their ability to mount T cell dependent antibody responses to oral vaccine. Mechanistically, our results support the idea that sulfide could act directly on CD4 T cells via enhanced AP-1 activation, leading to heightened proliferation and cytokine production. This study uncovers sulfides as keystone components in gut ecology and provides mechanistic insight between diet, gut sulfide production and mucosal immunity.
Collapse
Affiliation(s)
- Victor I. Band
- Metaorganism Immunity Section, Laboratory of Host Immunity and Microbiome, National Institute of Allergy and Infectious Diseases, National Institutes of Health; Bethesda, MD 20892, USA
| | - Inta Gribonika
- Metaorganism Immunity Section, Laboratory of Host Immunity and Microbiome, National Institute of Allergy and Infectious Diseases, National Institutes of Health; Bethesda, MD 20892, USA
| | - Apollo Stacy
- Metaorganism Immunity Section, Laboratory of Host Immunity and Microbiome, National Institute of Allergy and Infectious Diseases, National Institutes of Health; Bethesda, MD 20892, USA
| | - Nicolas Bouladoux
- Metaorganism Immunity Section, Laboratory of Host Immunity and Microbiome, National Institute of Allergy and Infectious Diseases, National Institutes of Health; Bethesda, MD 20892, USA
| | - Shreni Mistry
- NIAID Microbiome Program, National Institute of Allergy and Infectious Diseases, National Institutes of Health; Bethesda, MD 20892, USA
| | - Andrew Burns
- NIAID Microbiome Program, National Institute of Allergy and Infectious Diseases, National Institutes of Health; Bethesda, MD 20892, USA
| | - P. Juliana Perez-Chaparro
- NIAID Microbiome Program, National Institute of Allergy and Infectious Diseases, National Institutes of Health; Bethesda, MD 20892, USA
| | - Joanna Chau
- Metaorganism Immunity Section, Laboratory of Host Immunity and Microbiome, National Institute of Allergy and Infectious Diseases, National Institutes of Health; Bethesda, MD 20892, USA
| | - Michel Enamorado
- Metaorganism Immunity Section, Laboratory of Host Immunity and Microbiome, National Institute of Allergy and Infectious Diseases, National Institutes of Health; Bethesda, MD 20892, USA
| | - Motoyoshi Nagai
- Metaorganism Immunity Section, Laboratory of Host Immunity and Microbiome, National Institute of Allergy and Infectious Diseases, National Institutes of Health; Bethesda, MD 20892, USA
| | - Vanya Bhushan
- Functional Cellular Networks Section, Laboratory of Immune System Biology, National Institute of Allergy and Infectious Diseases, National Institutes of Health; Bethesda, MD 20892, USA
| | - Dominic P. Golec
- Cell Signaling and Immunity Section, Laboratory of Immune System Biology, National Institute of Allergy and Infectious Diseases, National Institutes of Health; Bethesda, MD 20892, USA
| | - Pamela L. Schwartzberg
- Cell Signaling and Immunity Section, Laboratory of Immune System Biology, National Institute of Allergy and Infectious Diseases, National Institutes of Health; Bethesda, MD 20892, USA
| | - Suchitra K. Hourigan
- Clinical Microbiome Unit, Laboratory of Host Immunity and Microbiome, National Institute of Allergy and Infectious Diseases, National Institutes of Health; Bethesda, MD 20892, USA
| | - Aleksandra Nita-Lazar
- Functional Cellular Networks Section, Laboratory of Immune System Biology, National Institute of Allergy and Infectious Diseases, National Institutes of Health; Bethesda, MD 20892, USA
| | - Yasmine Belkaid
- Metaorganism Immunity Section, Laboratory of Host Immunity and Microbiome, National Institute of Allergy and Infectious Diseases, National Institutes of Health; Bethesda, MD 20892, USA
| |
Collapse
|
46
|
Zhang M, Qin Z, Huang C, Liang B, Zhang X, Sun W. The gut microbiota modulates airway inflammation in allergic asthma through the gut-lung axis related immune modulation: A review. BIOMOLECULES & BIOMEDICINE 2025; 25:727-738. [PMID: 39465678 PMCID: PMC11959394 DOI: 10.17305/bb.2024.11280] [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: 09/09/2024] [Revised: 10/20/2024] [Accepted: 10/20/2024] [Indexed: 10/29/2024]
Abstract
The human gut microbiota is a vast and complex microbial community. According to statistics, the number of bacteria residing in the human intestinal tract is approximately ten times that of total human cells, with over 1000 different species. The interaction between the gut microbiota and various organ tissues plays a crucial role in the pathogenesis of local and systemic diseases, exerting a significant influence on disease progression. The relationship between the gut microbiota and intestinal diseases, along with its connection to the pulmonary immune environment and the development of lung diseases, is commonly referred to as the "gut-lung axis." The incidence of bronchial asthma is rising globally. With ongoing research on gut microbiota, it is widely believed that intestinal microorganisms and their metabolic products directly or indirectly participate in the occurrence and development of asthma. Based on the gut-lung axis, this review examines recent research suggesting that the intestinal microbiota can influence the occurrence and progression of allergic asthma through the modulation of cytokine immune balance and mucosal integrity. Though the precise immune pathways or microbial species influencing asthma through the gut-lung axis are still under exploration, summarizing the immune modulation through the gut-lung axis in allergic asthma may provide insights for the clinical management of the condition.
Collapse
Affiliation(s)
- Meng Zhang
- Department of Gastroenterology, People’s Hospital Affiliated to Shandong First Medical University, Jinan, Shandong Province, China
| | - Ziwen Qin
- The First Clinical Medical College, Shandong University of Traditional Chinese Medicine, Jinan, Shandong Province, China
| | - Chuanjun Huang
- Department of Respiratory and Critical Care Medicine, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong Province, China
| | - Bin Liang
- Department of Respiratory and Critical Care Medicine, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong Province, China
| | - Xiuqing Zhang
- Department of Radiology, Dongying City Dongying District People’s Hospital, Dongying, Shandong Province, China
| | - Weitao Sun
- Department of Respiratory Medicine, Dongying City Dongying District People’s Hospital, Dongying, Shandong Province, China
| |
Collapse
|
47
|
Stephens M, Keane K, Roizes S, Defaye M, Altier C, von der Weid PY. Uncovering the therapeutic potential of anti-tuberculoid agent Isoniazid in a model of microbial-driven Crohn's disease. J Crohns Colitis 2025; 19:jjaf032. [PMID: 39987456 PMCID: PMC11920797 DOI: 10.1093/ecco-jcc/jjaf032] [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: 07/02/2024] [Indexed: 02/24/2025]
Abstract
AIMS TNFα has long stood as a hallmark feature of both inflammatory bowel disease and arthritis with its therapeutic potential demonstrated in neutralizing monoclonal antibody treatments such as Infliximab. Due to the high global burden of latent Mycobacterium tuberculosis (TB) infections, prior to receiving anti-TNF therapy, patients testing positive for latent TB are given prophylactic treatment with anti-tuberculoid medications including the first described TB-selective antibiotic, Isoniazid. While this is common clinical practice to prevent the emergence of TB, little is known about whether Isoniazid modifies intestinal inflammation alone. The aim of this study, therefore, was to determine whether Isoniazid presents a novel TB-independent therapeutic option for the treatment of Crohn's disease (CD)-like ileitis and uncover new mechanisms predisposing the host to intestinal inflammation. METHODS The transgenic TNFΔARE mouse model of Crohn's-like terminal ileitis was used. The impact of Isoniazid administration (10 mg/kg/day dose in drinking water) on disease development was monitored between 8 and 12 weeks of age using a variety of behavioral and serological assays. Behavioral and motor functions were assessed using the LABORAS automated monitoring system while systemic and local tissue inflammation were determined at experimental termination using multiplex cytokine analysis. Whole-mount tissue immunofluorescence and fluorescent in situ hybridization were used to qualify changes within the host as well as the microbial compartment of the ileum and associated mesentery. Proposed cellular mechanisms of altered cytokine decay were performed on isolated primary splenocytes in vitro using selective pharmacological agents. RESULTS Compared to age-matched wild-type littermates, TNFΔARE mice display prominent progressive sickness behaviors from 8 through 12 weeks of age indicated by reduced movement, climbing, and rearing. Prophylactic administration of Isoniazid (10 mg/kg/day) is effectively able to protect TNFΔARE mice from this loss of function during the same period. Analysis revealed that Isoniazid was able to significantly reduce both systemic and intestinal inflammation compared to untreated vehicle controls impacting the epithelial colonization of known pathobiont segmented filamentous bacteria (SFB). Reduction in terminal ileal inflammation was also associated to the diminished formation of precursor-tertiary lymphoid organs within the associated ileal mesentery which were found to be associated with endospores derived SFB itself. Finally, we reveal that due to their genetic manipulation, TNFΔARE mice display accelerated posttranscriptional decay of IL-22 mRNA resulting in diminished IL-22 protein production and associated downstream antimicrobial peptide production. CONCLUSIONS Isoniazid protects against the development of intestinal and systemic inflammation in the TNFΔARE model of terminal ileitis by limiting the expansion of mucosal SFB and progression of the associated microbial-driven inflammation. This work highlights a possible mycobacterial-independent function of Isoniazid in limiting CD pathophysiology through limiting the mucosal establishment of pathobionts such as SFB and the association of such microbe-derived endospores linked to the formation of ectopic tertiary lymphoid organs seen commonly in patients.
Collapse
Affiliation(s)
- Matthew Stephens
- Department of Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, Alberta T2N4N1, Canada
- Inflammation Research Network Snyder Institute for Chronic Diseases, Cumming School of Medicine, University of Calgary, Calgary, Alberta T2N4N1, Canada
- Snyder Institute for Chronic Diseases, University of Calgary, HS 1665, 3330 Hospital Drive NW, Calgary, Alberta T2N4N1, Canada
| | - Keith Keane
- Department of Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, Alberta T2N4N1, Canada
- Inflammation Research Network Snyder Institute for Chronic Diseases, Cumming School of Medicine, University of Calgary, Calgary, Alberta T2N4N1, Canada
- Snyder Institute for Chronic Diseases, University of Calgary, HS 1665, 3330 Hospital Drive NW, Calgary, Alberta T2N4N1, Canada
| | - Simon Roizes
- Department of Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, Alberta T2N4N1, Canada
- Inflammation Research Network Snyder Institute for Chronic Diseases, Cumming School of Medicine, University of Calgary, Calgary, Alberta T2N4N1, Canada
- Snyder Institute for Chronic Diseases, University of Calgary, HS 1665, 3330 Hospital Drive NW, Calgary, Alberta T2N4N1, Canada
| | - Manon Defaye
- Department of Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, Alberta T2N4N1, Canada
- Inflammation Research Network Snyder Institute for Chronic Diseases, Cumming School of Medicine, University of Calgary, Calgary, Alberta T2N4N1, Canada
- Snyder Institute for Chronic Diseases, University of Calgary, HS 1665, 3330 Hospital Drive NW, Calgary, Alberta T2N4N1, Canada
- Hotchkiss Brain Institute, Cumming School of Medicine, University of Calgary, Calgary, Alberta T2N4N1, Canada
| | - Christophe Altier
- Department of Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, Alberta T2N4N1, Canada
- Inflammation Research Network Snyder Institute for Chronic Diseases, Cumming School of Medicine, University of Calgary, Calgary, Alberta T2N4N1, Canada
- Snyder Institute for Chronic Diseases, University of Calgary, HS 1665, 3330 Hospital Drive NW, Calgary, Alberta T2N4N1, Canada
- Hotchkiss Brain Institute, Cumming School of Medicine, University of Calgary, Calgary, Alberta T2N4N1, Canada
| | - Pierre-Yves von der Weid
- Department of Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, Alberta T2N4N1, Canada
- Inflammation Research Network Snyder Institute for Chronic Diseases, Cumming School of Medicine, University of Calgary, Calgary, Alberta T2N4N1, Canada
- Snyder Institute for Chronic Diseases, University of Calgary, HS 1665, 3330 Hospital Drive NW, Calgary, Alberta T2N4N1, Canada
| |
Collapse
|
48
|
Yang S, Liu H, Liu Y. Advances in intestinal epithelium and gut microbiota interaction. Front Microbiol 2025; 16:1499202. [PMID: 40104591 PMCID: PMC11914147 DOI: 10.3389/fmicb.2025.1499202] [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: 09/20/2024] [Accepted: 02/17/2025] [Indexed: 03/20/2025] Open
Abstract
The intestinal epithelium represents a critical interface between the host and external environment, serving as the second largest surface area in the human body after the lungs. This dynamic barrier is sustained by specialized epithelial cell types and their complex interactions with the gut microbiota. This review comprehensively examines the recent advances in understanding the bidirectional communication between intestinal epithelial cells and the microbiome. We briefly highlight the role of various intestinal epithelial cell types, such as Paneth cells, goblet cells, and enteroendocrine cells, in maintaining intestinal homeostasis and barrier function. Gut microbiota-derived metabolites, particularly short-chain fatty acids and bile acids, influence epithelial cell function and intestinal barrier integrity. Additionally, we highlight emerging evidence of the sophisticated cooperation between different epithelial cell types, with special emphasis on the interaction between tuft cells and Paneth cells in maintaining microbial balance. Understanding these complex interactions has important implications for developing targeted therapeutic strategies for various gastrointestinal disorders, including inflammatory bowel disease, metabolic disorders, and colorectal cancer.
Collapse
Affiliation(s)
- Sen Yang
- Department of Pediatric Pulmonology and Immunology, West China Second University Hospital, Sichuan University, Chengdu, China
- Department of Pediatrics, The Fifth Peoples Hospital of Chengdu, Chengdu, China
| | - Hanmin Liu
- Department of Pediatric Pulmonology and Immunology, West China Second University Hospital, Sichuan University, Chengdu, China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, Chengdu, China
- NHC Key Laboratory of Chronobiology (Sichuan University), Chengdu, China
- The Joint Laboratory for Lung Development and Related Diseases of West China Second University Hospital, Sichuan University and School of Life Sciences of Fudan University, West China Institute of Women and Children's Health, West China Second University Hospital, Sichuan University, Chengdu, China
| | - Yang Liu
- Department of Pediatric Pulmonology and Immunology, West China Second University Hospital, Sichuan University, Chengdu, China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, Chengdu, China
- NHC Key Laboratory of Chronobiology (Sichuan University), Chengdu, China
- The Joint Laboratory for Lung Development and Related Diseases of West China Second University Hospital, Sichuan University and School of Life Sciences of Fudan University, West China Institute of Women and Children's Health, West China Second University Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
49
|
Jiang J, Xie H, Cao S, Xu X, Zhou J, Liu Q, Ding C, Liu M. Post-stroke depression: exploring gut microbiota-mediated barrier dysfunction through immune regulation. Front Immunol 2025; 16:1547365. [PMID: 40098959 PMCID: PMC11911333 DOI: 10.3389/fimmu.2025.1547365] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2024] [Accepted: 02/17/2025] [Indexed: 03/19/2025] Open
Abstract
Post-stroke depression (PSD) is one of the most common and devastating neuropsychiatric complications in stroke patients, affecting more than one-third of survivors of ischemic stroke (IS). Despite its high incidence, PSD is often overlooked or undertreated in clinical practice, and effective preventive measures and therapeutic interventions remain limited. Although the exact mechanisms of PSD are not fully understood, emerging evidence suggests that the gut microbiota plays a key role in regulating gut-brain communication. This has sparked great interest in the relationship between the microbiota-gut-brain axis (MGBA) and PSD, especially in the context of cerebral ischemia. In addition to the gut microbiota, another important factor is the gut barrier, which acts as a frontline sensor distinguishing between beneficial and harmful microbes, regulating inflammatory responses and immunomodulation. Based on this, this paper proposes a new approach, the microbiota-immune-barrier axis, which is not only closely related to the pathophysiology of IS but may also play a critical role in the occurrence and progression of PSD. This review aims to systematically analyze how the gut microbiota affects the integrity and function of the barrier after IS through inflammatory responses and immunomodulation, leading to the production or exacerbation of depressive symptoms in the context of cerebral ischemia. In addition, we will explore existing technologies that can assess the MGBA and potential therapeutic strategies for PSD, with the hope of providing new insights for future research and clinical interventions.
Collapse
Affiliation(s)
- Jia Jiang
- The Second Affiliated Hospital, Hunan University of Chinese Medicine, Changsha, China
| | - Haihua Xie
- School of Acupuncture & Tuina and Rehabilitation, Hunan University of Chinese Medicine, Changsha, China
| | - Sihui Cao
- School of Acupuncture & Tuina and Rehabilitation, Hunan University of Chinese Medicine, Changsha, China
| | - Xuan Xu
- School of Acupuncture & Tuina and Rehabilitation, Hunan University of Chinese Medicine, Changsha, China
| | - Jingying Zhou
- School of Acupuncture & Tuina and Rehabilitation, Hunan University of Chinese Medicine, Changsha, China
| | - Qianyan Liu
- School of Acupuncture & Tuina and Rehabilitation, Hunan University of Chinese Medicine, Changsha, China
| | - Changsong Ding
- School of Information Science and Engineering, Hunan University of Chinese Medicine, Changsha, China
| | - Mi Liu
- School of Acupuncture & Tuina and Rehabilitation, Hunan University of Chinese Medicine, Changsha, China
| |
Collapse
|
50
|
Berzack S, Galor A. Microbiome-based therapeutics for ocular diseases. Clin Exp Optom 2025; 108:115-122. [PMID: 39617011 PMCID: PMC11875938 DOI: 10.1080/08164622.2024.2422479] [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: 09/16/2024] [Revised: 10/22/2024] [Accepted: 10/23/2024] [Indexed: 12/08/2024] Open
Abstract
The relationship between the gut microbiome and ocular health has garnered increasing attention within the scientific community. Recent research has focused on the gut-eye axis, examining whether imbalances within the gut microbiome can influence the development, progression and severity of ocular diseases, including dry eye disease, uveitis, and glaucoma. Dysbiosis within the gut microbiome is linked to immune dysregulation, chronic inflammation, and epithelial barrier dysfunction, all of which contribute to ocular pathology. This review synthesises current evidence on these associations, exploring how gut microbiome alterations drive disease mechanisms. Furthermore, it examines the therapeutic potential of microbiome-targeted interventions, including antibiotics, prebiotics, probiotics, and faecal microbiota transplantation, all of which aim to restore microbial balance and modulate immune responses. As the prevalence of these conditions continues to rise, a deeper understanding of the gut-eye axis may facilitate the development of novel, targeted therapies to address unmet needs in the management of ocular diseases.
Collapse
Affiliation(s)
- Shannan Berzack
- Herbert Wertheim College of Medicine, Florida International University, Miami, Florida, USA
| | - Anat Galor
- Bascom Palmer Eye Institute, University of Miami Miller School of Medicine, Miami, Florida, USA
| |
Collapse
|