1
|
Jiang J, Peng W, Sun N, Zhao D, Cui W, Lai Y, Zhang C, Duan C, Zeng W. Unraveling the anoikis-cancer nexus: a bibliometric analysis of research trends and mechanisms. Future Sci OA 2025; 11:2484159. [PMID: 40160087 PMCID: PMC11959893 DOI: 10.1080/20565623.2025.2484159] [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: 09/24/2024] [Accepted: 03/12/2025] [Indexed: 04/02/2025] Open
Abstract
BACKGROUND Cancer, influenced by genetics and the environment, involves anoikis, a cell death mechanism upon extracellular matrix detachment crucial for metastasis. Understanding this relationship is key for therapy. We analyze cancer and anoikis trends using bibliometrics. METHODS A search was conducted from Web of Science Core, PubMed, Scopus and non-English databases such as the CNKI (inception- 21 December 2024). Data analysis employed Microsoft Excel, VOSviewer, CiteSpace, R software, and the online platform (https://bibliometric.com/). RESULTS 2510 publications were retrieved, with a significant increase in the last decade. China led, the University of Texas system was productive, and the Oncogene Journal was popular. Breast, and colorectal cancers were frequently studied. Among them, representative tumor-related mechanisms were identified, commonalities such as (EMT, ECM, autophagy) and respective specific mechanisms were summarized. CONCLUSION This bibliometric analysis highlights rapid advances in anoikis research in cancer, emphasizing EMT and FAK pathways' translational potential, guiding targeted therapies, and improving cancer treatment outcomes.
Collapse
Affiliation(s)
- Junjie Jiang
- Department of Thoracic Surgery, Xiangya Hospital, Central South University, Changsha, Hunan, People’s Republic of China
- Hunan Engineering Research Center for Pulmonary Nodules Precise Diagnosis & Treatment, Changsha, Hunan, People’s Republic of China
| | - Wei Peng
- Department of Oncology, Hunan Provincial People’s Hospital, the First Affiliated Hospital of Hunan Normal University, Changsha, Hunan, People’s Republic of China
| | - Nianzhe Sun
- Department of Orthopedics, Xiangya Hospital, Central South University, Changsha, Hunan, People’s Republic of China
- National Clinical Research Center of Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, People’s Republic of China
| | - Deze Zhao
- Department of Thoracic Surgery, Xiangya Hospital, Central South University, Changsha, Hunan, People’s Republic of China
- Hunan Engineering Research Center for Pulmonary Nodules Precise Diagnosis & Treatment, Changsha, Hunan, People’s Republic of China
| | - Weifang Cui
- Department of Thoracic Surgery, Xiangya Hospital, Central South University, Changsha, Hunan, People’s Republic of China
- Hunan Engineering Research Center for Pulmonary Nodules Precise Diagnosis & Treatment, Changsha, Hunan, People’s Republic of China
| | - Yuwei Lai
- Department of Thoracic Surgery, Xiangya Hospital, Central South University, Changsha, Hunan, People’s Republic of China
- Hunan Engineering Research Center for Pulmonary Nodules Precise Diagnosis & Treatment, Changsha, Hunan, People’s Republic of China
| | - Chunfang Zhang
- Department of Thoracic Surgery, Xiangya Hospital, Central South University, Changsha, Hunan, People’s Republic of China
- Hunan Engineering Research Center for Pulmonary Nodules Precise Diagnosis & Treatment, Changsha, Hunan, People’s Republic of China
| | - Chaojun Duan
- Department of Thoracic Surgery, Xiangya Hospital, Central South University, Changsha, Hunan, People’s Republic of China
- Hunan Engineering Research Center for Pulmonary Nodules Precise Diagnosis & Treatment, Changsha, Hunan, People’s Republic of China
- Institute of Medical Sciences, Xiangya Lung Cancer Center, Xiangya Hospital, Central South University, Changsha, Hunan, People’s Republic of China
| | - Wei Zeng
- Department of Thoracic Surgery, Xiangya Hospital, Central South University, Changsha, Hunan, People’s Republic of China
- Hunan Engineering Research Center for Pulmonary Nodules Precise Diagnosis & Treatment, Changsha, Hunan, People’s Republic of China
- National Clinical Research Center of Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, People’s Republic of China
| |
Collapse
|
2
|
Liang H, Bai X, Zhu R, Song H, Miao Y, Wen Y, Niu J, Zhang F. Diminution of HSP75 disrupts intestinal epithelial barrier by inciting mPTP opening in ulcerative colitis. Cell Signal 2025; 132:111837. [PMID: 40294832 DOI: 10.1016/j.cellsig.2025.111837] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2025] [Revised: 04/21/2025] [Accepted: 04/25/2025] [Indexed: 04/30/2025]
Abstract
Ulcerative colitis is an idiopathic, chronic inflammatory disorder. The disruption of intestinal epithelial barrier caused by excessive apoptosis of intestinal epithelial cells is a pivotal factor in the etiology and pathology. The mitochondrial pathway is the most significant apoptosis mode of intestinal epithelial cells, which was regulated by the mitochondrial permeability transition pore(mPTP). However, the precise mechanism remains elusive. As a crucial molecule in combating stress and maintaining mitochondrial homeostasis, the heat shock protein 75(HSP75) may play a vital role in regulating the openness of the mPTP. In our research, we ascertained that HSP75 was significantly diminished in the intestinal mucosal of UC patients and experimental colitis mice, concomitantly with the disruption of intestinal epithelial barrier. Furthermore, a negative correlation between HSP75 and the openness of mPTP, mitochondrial-driven apoptosis, and disruption of intestinal epithelial barrier has been demonstrated in vivo and vitro. Secondly, HSP75 level is negatively correlated with the expression of ANT, VDAC, and PiC, which considered to be the components of mPTP. However, the CypD is unaffected by HSP75. Finally, HSP75 altered the synthesis of ANT, VDAC, PiC and the acetylation modification of ANT, but there is no direct interaction between HSP75 and mPTP component proteins. In conclusion, the present study demonstrated that HSP75 significantly decreased in the intestinal mucosa of UC, and preliminarily revealed a novel mechanism of HSP75 regulating the synthesis and openness of mPTP in the intestinal epithelial cells(IECs) of UC, suggesting that the targeted intestinal mucosa supplementation of HSP75 is anticipated to reverse the pathological process.
Collapse
Affiliation(s)
- Hao Liang
- Department of Gastroenterology, The First Affiliated Hospital of Kunming Medical University, Kunming 650032, Yunnan, China; Yunnan Province Clinical Research Center for Digestive Diseases, Kunming 650032, Yunnan, China
| | - Xinyu Bai
- Department of Gastroenterology, The First Affiliated Hospital of Kunming Medical University, Kunming 650032, Yunnan, China; Yunnan Province Clinical Research Center for Digestive Diseases, Kunming 650032, Yunnan, China
| | - Rui Zhu
- Department of Gastroenterology, The First Affiliated Hospital of Kunming Medical University, Kunming 650032, Yunnan, China; Yunnan Province Clinical Research Center for Digestive Diseases, Kunming 650032, Yunnan, China
| | - Huixian Song
- Department of Gastroenterology, The First Affiliated Hospital of Kunming Medical University, Kunming 650032, Yunnan, China; Yunnan Province Clinical Research Center for Digestive Diseases, Kunming 650032, Yunnan, China
| | - Yinglei Miao
- Department of Gastroenterology, The First Affiliated Hospital of Kunming Medical University, Kunming 650032, Yunnan, China; Yunnan Province Clinical Research Center for Digestive Diseases, Kunming 650032, Yunnan, China
| | - Yunling Wen
- Department of Gastroenterology, The First Affiliated Hospital of Kunming Medical University, Kunming 650032, Yunnan, China; Yunnan Province Clinical Research Center for Digestive Diseases, Kunming 650032, Yunnan, China
| | - Junkun Niu
- Department of Gastroenterology, The First Affiliated Hospital of Kunming Medical University, Kunming 650032, Yunnan, China; Yunnan Province Clinical Research Center for Digestive Diseases, Kunming 650032, Yunnan, China.
| | - Fengrui Zhang
- Department of Gastroenterology, The First Affiliated Hospital of Kunming Medical University, Kunming 650032, Yunnan, China; Yunnan Province Clinical Research Center for Digestive Diseases, Kunming 650032, Yunnan, China.
| |
Collapse
|
3
|
Obeng EM, Hodge C, You J. Microplastic pollution: a review of specific blood-tissue barrier breaches and health effects. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2025; 376:126416. [PMID: 40355068 DOI: 10.1016/j.envpol.2025.126416] [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: 02/23/2025] [Revised: 05/09/2025] [Accepted: 05/09/2025] [Indexed: 05/14/2025]
Abstract
Microplastic (1 μm - 5 mm) and nanoplastic (<1 μm) pollution is a heightening global challenge affecting the environment and the health of living creatures within. As primary precursors for plastic manufacturing, microplastics predominantly get into the environment through plastic product degradation and integrate into water, food chain and consumer products leading to potential health consequences. The mammalian system is equipped with several blood-tissue barriers with exclusive tight junctions that selectively regulate material transfer and protect vulnerable and functionally important organs. Nonetheless, emerging evidence indicates microplastics interact, traverse and compromise the integrity of these complex barriers. This review summarises the known and potential impact of microplastics on human health, focusing on specific organ barrier breaches. Evidence of microplastic traversal and deposition in distal mammalian organs are discussed. We further highlight current challenges facing both researchers and clinicians and provide an outlook for expanding our understanding of the impact of microplastic on health.
Collapse
Affiliation(s)
- Eugene M Obeng
- School of Medical Sciences, Faculty of Medicine and Health, University of Sydney, Camperdown, NSW, 2006, Australia; The University of Sydney Nano Institute (Sydney Nano), University of Sydney, Camperdown, NSW, 2006, Australia; Net Zero Institute, University of Sydney, Darlington, NSW, 2006, Australia.
| | - Christopher Hodge
- School of Medical Sciences, Faculty of Medicine and Health, University of Sydney, Camperdown, NSW, 2006, Australia; Net Zero Institute, University of Sydney, Darlington, NSW, 2006, Australia; Save Sight Institute, University of Sydney, Sydney, NSW, 2000, Australia; Vision Eye Institute, Chatswood, NSW, 2067, Australia
| | - Jingjing You
- School of Medical Sciences, Faculty of Medicine and Health, University of Sydney, Camperdown, NSW, 2006, Australia; Net Zero Institute, University of Sydney, Darlington, NSW, 2006, Australia; Vision Eye Institute, Chatswood, NSW, 2067, Australia; School of Optometry and Vision Science, University of New South Wales, Sydney, New South Wales, Australia.
| |
Collapse
|
4
|
So YJ, Park OJ, Kwon Y, Im J, Lee D, Yun SH, Cho K, Yun CH, Han SH. Bacillus subtilis Induces Human Beta Defensin-2 Through its Lipoproteins in Human Intestinal Epithelial Cells. Probiotics Antimicrob Proteins 2025; 17:1648-1662. [PMID: 38376819 PMCID: PMC12055916 DOI: 10.1007/s12602-024-10224-4] [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] [Accepted: 01/24/2024] [Indexed: 02/21/2024]
Abstract
Human intestinal epithelial cells (IECs) play an important role in maintaining gut homeostasis by producing antimicrobial peptides (AMPs). Bacillus subtilis, a commensal bacterium, is considered a probiotic. Although its protective effects on intestinal health are widely reported, the key component of B. subtilis responsible for its beneficial effects remains elusive. In this study, we tried to identify the key molecules responsible for B. subtilis-induced AMPs and their molecular mechanisms in a human IEC line, Caco-2. B. subtilis increased human beta defensin (HBD)-2 mRNA expression in a dose- and time-dependent manner. Among the B. subtilis microbe-associated molecular patterns, lipoprotein (LPP) substantially increased the mRNA expression and protein production of HBD-2, whereas lipoteichoic acid and peptidoglycan did not show such effects. Those results were confirmed in primary human IECs. In addition, both LPP recognition and HBD-2 secretion mainly took place on the apical side of fully differentiated and polarized Caco-2 cells through Toll-like receptor 2-mediated JNK/p38 MAP kinase/AP-1 and NF-κB pathways. HBD-2 efficiently inhibited the growth of the intestinal pathogens Staphylococcus aureus and Bacillus cereus. Furthermore, LPPs pre-incubated with lipase or proteinase K decreased LPP-induced HBD-2 expression, suggesting that the lipid and protein moieties of LPP are crucial for HBD-2 expression. Q Exactive Plus mass spectrometry identified 35 B. subtilis LPP candidates within the LPP preparation, and most of them were ABC transporters. Taken together, these results suggest that B. subtilis promotes HBD-2 secretion in human IECs mainly with its LPPs, which might enhance the protection from intestinal pathogens.
Collapse
Affiliation(s)
- Yoon Ju So
- Department of Oral Microbiology and Immunology, and Dental Research Institute, School of Dentistry, Seoul National University, Seoul, 08826, Republic of Korea
| | - Ok-Jin Park
- Department of Oral Microbiology and Immunology, and Dental Research Institute, School of Dentistry, Seoul National University, Seoul, 08826, Republic of Korea
| | - Yeongkag Kwon
- Department of Oral Microbiology and Immunology, and Dental Research Institute, School of Dentistry, Seoul National University, Seoul, 08826, Republic of Korea
| | - Jintaek Im
- Department of Oral Microbiology and Immunology, and Dental Research Institute, School of Dentistry, Seoul National University, Seoul, 08826, Republic of Korea
| | - Dongwook Lee
- Department of Oral Microbiology and Immunology, and Dental Research Institute, School of Dentistry, Seoul National University, Seoul, 08826, Republic of Korea
| | - Sung-Ho Yun
- Center for Research Equipment, Korea Basic Science Institute, Ochang, 28119, Republic of Korea
| | - Kun Cho
- Center for Research Equipment, Korea Basic Science Institute, Ochang, 28119, Republic of Korea
| | - Cheol-Heui Yun
- Department of Agricultural Biotechnology, and Research Institute of Agriculture and Life Sciences, Seoul National University, Seoul, 08826, Republic of Korea
- Institutes of Green Bio Science & Technology, Seoul National University, Pyeongchang, 25354, Republic of Korea
| | - Seung Hyun Han
- Department of Oral Microbiology and Immunology, and Dental Research Institute, School of Dentistry, Seoul National University, Seoul, 08826, Republic of Korea.
| |
Collapse
|
5
|
Neurath MF, Artis D, Becker C. The intestinal barrier: a pivotal role in health, inflammation, and cancer. Lancet Gastroenterol Hepatol 2025; 10:573-592. [PMID: 40086468 DOI: 10.1016/s2468-1253(24)00390-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/26/2024] [Revised: 11/08/2024] [Accepted: 11/15/2024] [Indexed: 03/16/2025]
Abstract
The intestinal barrier serves as a boundary between the mucosal immune system in the lamina propria and the external environment of the intestinal lumen, which contains a diverse array of microorganisms and ingested environmental factors, including pathogens, food antigens, toxins, and other foreign substances. This barrier has a central role in regulating the controlled interaction between luminal factors and the intestinal immune system. Disruptions of intestinal epithelial cells, which serve as a physical barrier, or the antimicrobial peptides and mucins they produce, which act as a chemical barrier, can lead to a leaky gut. In this state, the intestinal wall is unable to efficiently separate the intestinal flora and luminal contents from the intestinal immune system. The subsequent activation of the immune system has an important role in the pathogenesis of inflammatory bowel disease, as well as in metabolic dysfunction-associated steatohepatitis, primary sclerosing cholangitis, and colorectal cancer. Dysregulated intestinal barrier integrity has also been described in patients with chronic inflammatory diseases outside the gastrointestinal tract, including rheumatoid arthritis and neurodegenerative disorders. Mechanistic studies of barrier dysfunction have revealed that the subsequent local activation and systemic circulation of activated immune cells and the cytokines they secrete, as well as extracellular vesicles, promote proinflammatory processes within and outside the gastrointestinal tract. In this Review, we summarise these findings and highlight several new therapeutic concepts currently being developed that attempt to control inflammatory processes via direct or indirect modulation of intestinal barrier function.
Collapse
Affiliation(s)
- Markus F Neurath
- Medical Clinic 1, Department of Gastroenterology, Ludwig Demling Endoscopy Center of Excellence, University Hospital Erlangen, Friedrich-Alexander University Erlangen-Nürnberg, Erlangen, Germany; Deutsches Zentrum Immuntherapie (DZI), Friedrich-Alexander University Erlangen-Nürnberg, Erlangen, Germany.
| | - David Artis
- Jill Roberts Institute for Research in Inflammatory Bowel Disease, Weill Cornell Medicine, Cornell University, New York, NY, USA; Friedman Center for Nutrition and Inflammation, Weill Cornell Medicine, Cornell University, New York, NY, USA; Joan and Sanford I Weill Department of Medicine, Weill Cornell Medicine, Cornell University, New York, NY, USA; Department of Microbiology and Immunology, Weill Cornell Medicine, Cornell University, New York, NY, USA; Allen Discovery Center for Neuroimmune Interactions, Weill Cornell Medicine, Cornell University, New York, NY, USA
| | - Christoph Becker
- Medical Clinic 1, Department of Gastroenterology, Ludwig Demling Endoscopy Center of Excellence, University Hospital Erlangen, Friedrich-Alexander University Erlangen-Nürnberg, Erlangen, Germany; Deutsches Zentrum Immuntherapie (DZI), Friedrich-Alexander University Erlangen-Nürnberg, Erlangen, Germany
| |
Collapse
|
6
|
Li Y, Li Q, Xu W, Liu R, Gong Y, Li M. Resveratrol Alleviated Intensive Exercise-Induced Fatigue Involving in Inhibiting Gut Inflammation and Regulating Gut Microbiota. Food Sci Nutr 2025; 13:e70304. [PMID: 40417740 PMCID: PMC12102494 DOI: 10.1002/fsn3.70304] [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: 02/12/2025] [Revised: 04/28/2025] [Accepted: 05/05/2025] [Indexed: 05/27/2025] Open
Abstract
Resveratrol (trans-3,4',5-trihydroxystilbene, RES) is a stilbenoid naturally present in a variety of plants. Although there are several reports about its anti-fatigue activity, its impact on intensive exercise-induced fatigue and the underlying mechanisms are yet not well understood. In the present study, we established a swimming exercise protocol in mice that is similar to the fatigue condition induced by a long period of intensive exercise and explored the effect of RES on fatigue and the mechanisms from the perspective of intestinal injury and gut microbiota. The results revealed that RES significantly prolonged exhaustive swimming time in fatigued mice and improved the serum indexes associated with fatigue, including serum glucose, lactic acid (LA), urea nitrogen (BUN), lactate dehydrogenase (LDH), creatine kinase (CK), catalase (CAT), glutathione peroxidase (GSH-Px), and glycogen storage in liver and muscle. Meanwhile, RES increased the expressions of ZO-1, Occludin, and Claudin-1, thereby enhancing intestinal barrier integrity and inhibiting mRNA expressions of tumor necrosis factor-α (TNF-α), interleukin-6 (IL-6), and interleukin-1β (IL-1β) in the colon, thereby improving the pathological injury in the colon. Importantly, RES modified gut microbiota dysbiosis by increasing the diversity of gut microbiota, regulating microbiota associated with inflammation and fatty acid metabolism at the phylum (Bacteroidetes and Firmicutes), family (Erysipelotrichaceae, Enterobacteriaceae, and Prevotellaceae), and genus (Brevundimonas diminuta, Coprobacillus, Megasphaera, and Lactobacillus) levels, respectively. The results supplemented the anti-fatigue mechanism for RES from the perspective of intestinal injury and gut microbiota. The detailed mechanisms and associated metabonomics analysis remain for further study.
Collapse
Affiliation(s)
- Yuening Li
- College of Competitive SportsWuhan Sports UniversityWuhanChina
| | - Qinsheng Li
- Physical Education InstituteTaishan UniversityTaianChina
| | - Wenxiu Xu
- College of Chemical EngineeringQingdao University of Science and TechnologyQingdaoChina
| | - Ruiqing Liu
- College of Physical Education and HealthLinyi UniversityLinyiChina
| | - Yanling Gong
- College of Chemical EngineeringQingdao University of Science and TechnologyQingdaoChina
| | - Ming Li
- College of Physical Education and HealthLinyi UniversityLinyiChina
| |
Collapse
|
7
|
Wang M, Li D, Ouyang S, Tong B, Chen Y, Ding B, Wang J, Jiang Z, Xu H, Hu S. Hydrogel derived from decellularized pig small intestine submucosa boosted the therapeutic effect of FGF-20 on TNBS-induced colitis in rats via restoring gut mucosal integrity. Mater Today Bio 2025; 32:101783. [PMID: 40321695 PMCID: PMC12049826 DOI: 10.1016/j.mtbio.2025.101783] [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: 03/01/2025] [Revised: 04/07/2025] [Accepted: 04/19/2025] [Indexed: 05/08/2025] Open
Abstract
Ulcerative colitis (UC) is a chronic inflammatory bowel disease characterized by impaired intestinal mucosal barrier function, leading to persistent inflammation and tissue damage. Current therapies often fail to address barrier dysfunction, highlighting the need for innovative treatments. This study developed a novel therapeutic strategy by combining decellularized porcine small intestinal submucosa (D-SIS) with fibroblast growth factor 20 (FGF-20) to promote mucosal repair and restore barrier integrity in a TNBS-induced colitis rat model. The D-SIS-based hydrogel, supplemented with hyaluronic acid (HA), was designed to enhance FGF-20 stability and enable sustained drug release. Results showed that the FGF-20-loaded hydrogel (MAF) exhibited excellent rheological properties, erosion resistance, and controlled drug release, making it suitable for rectal administration. In vitro cell experiments demonstrated that MAF enhanced Caco-2 cell proliferation, migration, and tight junction protein expression, restoring epithelial barrier integrity. In the colitis model, MAF significantly reduced disease activity index (DAI) scores, attenuated inflammation, and restored mucosal morphology. Additionally, MAF promoted goblet cell regeneration, enhanced mucus secretion, and upregulated intestinal stem cell markers, indicating its ability to repair both epithelial and mucus barriers. In conclusion, the MAF hydrogel represents a promising therapeutic approach for UC by combining the regenerative properties of FGF-20 with the bioactive support of D-SIS.
Collapse
Affiliation(s)
- Minmin Wang
- Department of Gastrointestinal Surgery Nursing Unit, Ward 442, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou City, Zhejiang Province, 325000, China
| | - Dingwei Li
- Department of Pharmaceutics, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou City, Zhejiang Province, 325035, China
| | - Shenyuan Ouyang
- Department of Pharmaceutics, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou City, Zhejiang Province, 325035, China
| | - Bingjie Tong
- Department of Pharmaceutics, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou City, Zhejiang Province, 325035, China
| | - Yumo Chen
- Department of Pharmaceutics, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou City, Zhejiang Province, 325035, China
| | - Bingyu Ding
- Department of Pharmaceutics, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou City, Zhejiang Province, 325035, China
| | - Jie Wang
- Department of Pharmaceutics, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou City, Zhejiang Province, 325035, China
| | - Zhijiang Jiang
- Department of Pharmaceutics, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou City, Zhejiang Province, 325035, China
| | - Helin Xu
- Department of Pharmaceutics, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou City, Zhejiang Province, 325035, China
| | - Sunkuan Hu
- Department of Gastroenterology, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou City, Zhejiang Province, 325000, China
| |
Collapse
|
8
|
Tang B, Liu Z, Xiong H, Zhang J, Dai J. IFN-λ: Unleashing Its Potential in Disease Therapies From Acute Inflammation Regulation to Cancer Immunotherapy. Immunology 2025. [PMID: 40421666 DOI: 10.1111/imm.13954] [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: 01/04/2025] [Revised: 04/21/2025] [Accepted: 05/14/2025] [Indexed: 05/28/2025] Open
Abstract
Type III interferons (IFN-λ), which include IFN-λ1 (or interleukin [IL]-29), IFN-λ2 (IL-28A), IFN-λ3 (IL-28B) and IFN-λ4, exert their effects through a unique receptor complex composed of interferon lambda receptor 1 (IFNLR1) and IL-10 receptor subunit beta (IL-10R2). Studies have highlighted their critical role in modulating immune response, particularly in the context of autoimmune diseases, viral infections and cancer. Unlike type I IFNs, which are broadly expressed, IFN-λ displays a more tissue-specific expression pattern, predominantly acting on epithelial cells and certain immune cell types, such as neutrophils and B cells. This specificity allows IFN-λ to play a pivotal role in mucosal immunity, particularly at barrier sites, such as the respiratory and gastrointestinal tracts. Emerging evidence suggests that IFN-λ has a dual role in both enhancing antiviral immunity and regulating inflammation, thus offering a promising therapeutic strategy for diseases like systemic lupus erythematosus, rheumatoid arthritis, asthma and various cancers. However, the precise mechanisms by which IFN-λ influence immune modulation and disease progression remain an area of active investigation. This review aims to provide an overview of the structure, function and signalling pathways of IFN-λ, exploring their role in immune-related diseases and discussing potential avenues for therapeutic intervention.
Collapse
Affiliation(s)
- Benfeng Tang
- Institute of Immunology and Molecular Medicine, Jining Medical University, Jining, China
- Jining Key Laboratory of Immunology, Jining Medical University, Jining, China
| | - Zhihong Liu
- Institute of Immunology and Molecular Medicine, Jining Medical University, Jining, China
- School of Basic Medicine, Shandong First Medical University, Jinan, China
| | - Huabao Xiong
- Institute of Immunology and Molecular Medicine, Jining Medical University, Jining, China
| | - Junfeng Zhang
- Institute of Immunology and Molecular Medicine, Jining Medical University, Jining, China
| | - Jun Dai
- Institute of Immunology and Molecular Medicine, Jining Medical University, Jining, China
| |
Collapse
|
9
|
Chen Y, Fu KX, Cotton R, Ou Z, Kwak JW, Chien JC, Kesler V, Nyein HYY, Eisenstein M, Tom Soh H. A biochemical sensor with continuous extended stability in vivo. Nat Biomed Eng 2025:10.1038/s41551-025-01389-6. [PMID: 40410556 DOI: 10.1038/s41551-025-01389-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2023] [Accepted: 04/03/2025] [Indexed: 05/25/2025]
Abstract
The development of biosensors that can detect specific analytes continuously, in vivo, in real time has proven difficult due to biofouling, probe degradation and signal drift that often occur in vivo. By drawing inspiration from intestinal mucosa that can protect host cell receptors in the presence of the gut microbiome, we develop a synthetic biosensor that can continuously detect specific target molecules in vivo. The biomimetic multicomponent sensor features the hierarchical nano-bio interface design with three-dimensional bicontinuous nanoporous structure, polymer coating and aptamer switches, balancing small-molecule sensing and surface protection in complex biological environments. Our system is stable for at least 1 month in undiluted serum in vitro or 1 week implanted within the blood vessels of free-moving rats, retaining over 50% baseline signal and reproducible calibration curves. We demonstrate that the implanted system can intravenously track pharmacokinetics in real time even after 4 days of continuous exposure to flowing blood within rat femoral vein. In this way, our work provides a generalizable design foundation for biosensors that can continuously operate in vivo for extended durations.
Collapse
Affiliation(s)
- Yihang Chen
- Department of Materials Science and Engineering, Stanford University, Stanford, CA, USA
- Department of Radiology, Stanford University, Stanford, CA, USA
| | - Kaiyu X Fu
- Department of Radiology, Stanford University, Stanford, CA, USA
- Department of Electrical Engineering, Stanford University, Stanford, CA, USA
- Department of Chemistry and Biochemistry, University of Notre Dame, Notre Dame, IN, USA
| | - Renee Cotton
- Department of Comparative Medicine, Stanford University, Stanford, CA, USA
| | - Zihao Ou
- Department of Materials Science and Engineering, Stanford University, Stanford, CA, USA
| | - Jean Won Kwak
- Department of Radiology, Stanford University, Stanford, CA, USA
| | - Jun-Chau Chien
- Department of Radiology, Stanford University, Stanford, CA, USA
- Department of Electrical Engineering, Stanford University, Stanford, CA, USA
| | - Vladimir Kesler
- Department of Radiology, Stanford University, Stanford, CA, USA
- Department of Electrical Engineering, Stanford University, Stanford, CA, USA
| | - Hnin Yin Yin Nyein
- Department of Radiology, Stanford University, Stanford, CA, USA
- Department of Electrical Engineering, Stanford University, Stanford, CA, USA
| | - Michael Eisenstein
- Department of Radiology, Stanford University, Stanford, CA, USA
- Department of Electrical Engineering, Stanford University, Stanford, CA, USA
| | - H Tom Soh
- Department of Radiology, Stanford University, Stanford, CA, USA.
- Department of Electrical Engineering, Stanford University, Stanford, CA, USA.
- Department of Bioengineering, Stanford University, Stanford, CA, USA.
- Chan Zuckerberg Biohub, San Francisco, CA, USA.
| |
Collapse
|
10
|
An C, Jiang C, Pei W, Li A, Wang M, Wang Y, Wang H, Zuo L. Intestinal epithelial cells in health and disease. Tissue Barriers 2025:2504744. [PMID: 40401816 DOI: 10.1080/21688370.2025.2504744] [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: 12/25/2024] [Revised: 04/23/2025] [Accepted: 05/06/2025] [Indexed: 05/23/2025] Open
Abstract
This comprehensive review delves into the pivotal role of intestinal epithelial cells in the context of various diseases. It provides an in-depth analysis of the diverse types and functions of these cells, explores the influence of multiple signaling pathways on their differentiation, and elucidates their critical roles in a spectrum of diseases. The significance of the gastrointestinal tract in maintaining overall health is extremely important and cannot be exaggerated. This complex and elongated organ acts as a crucial link between the internal and external environments, making it vulnerable to various harmful influences. Preserving the normal structure and function of the gut is essential for well-being. Intestinal epithelial cells serve as the primary defense mechanism within the gastrointestinal tract and play a crucial role in preventing harmful substances from infiltrating the body. As the main components of the digestive system, they not only participate in the absorption and secretion of nutrients and the maintenance of barrier function but also play a pivotal role in immune defense. Therefore, the health of intestinal epithelial cells is of vital importance for overall health.
Collapse
Affiliation(s)
- Chenchen An
- Laboratory of Molecular Biology, Department of Biochemistry, School of Basic Medical Science, Anhui Medical University, Hefei, China
- Innovation and Entrepreneurship Laboratory for college students, Anhui Medical University, Hefei, China
| | - Chonggui Jiang
- Laboratory of Molecular Biology, Department of Biochemistry, School of Basic Medical Science, Anhui Medical University, Hefei, China
- Innovation and Entrepreneurship Laboratory for college students, Anhui Medical University, Hefei, China
| | - Wangxiang Pei
- Laboratory of Molecular Biology, Department of Biochemistry, School of Basic Medical Science, Anhui Medical University, Hefei, China
- Innovation and Entrepreneurship Laboratory for college students, Anhui Medical University, Hefei, China
| | - Ao Li
- Innovation and Entrepreneurship Laboratory for college students, Anhui Medical University, Hefei, China
- The 904th Hospital of PLA, Medical School of Anhui Medical University, Wuxi, China
| | - Minghui Wang
- Innovation and Entrepreneurship Laboratory for college students, Anhui Medical University, Hefei, China
- The First College of Clinical Medicine, Anhui Medical University, Hefei, China
| | - Yufei Wang
- Innovation and Entrepreneurship Laboratory for college students, Anhui Medical University, Hefei, China
- The First College of Clinical Medicine, Anhui Medical University, Hefei, China
| | - Hua Wang
- Inflammation and Immune- Mediated Diseases Laboratory of Anhui Province, Anhui Medical University, Hefei, China
- Department of Oncology, the First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Li Zuo
- Laboratory of Molecular Biology, Department of Biochemistry, School of Basic Medical Science, Anhui Medical University, Hefei, China
- Innovation and Entrepreneurship Laboratory for college students, Anhui Medical University, Hefei, China
| |
Collapse
|
11
|
Hu K, Ren H, Yuan C, Liu Z, Zhang X, Zhang H, Zhou N, Chen L, Wang N, Zhang G. Comparison of Different Methods of Constructing a Celiac Disease Model in BALB/c Mice. Mol Nutr Food Res 2025:e70118. [PMID: 40395135 DOI: 10.1002/mnfr.70118] [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: 09/10/2024] [Revised: 04/24/2025] [Accepted: 05/02/2025] [Indexed: 05/22/2025]
Abstract
Efficient and stable animal models of celiac disease (CD) are crucial for CD research, dietary supplementation research, and new drug development. This study aimed to establish CD models by administering gliadin to parental and first-generation mice on a gluten-free diet using two intraperitoneal injection and a combination of sensitization by intraperitoneal injection and gavage. Various indicators, including clinical manifestations, characteristic indicators of CD, inflammatory factors, intestinal barriers, immune cells, and other related indicators, were used to compare different modeling methods. The results showed that all four methods induced varying degrees of CD symptoms in the mice. The analysis revealed that intraperitoneal injection in first-generation mice significantly increased specific IgG and total IgE antibody levels; markedly shortened intestinal villus and crypt hyperplasia in the jejunum; significantly increased pro-inflammatory cytokines IFN-γ, IL-17, IL-15, and TNF-α; and significantly decreased the anti-inflammatory cytokine IL-4. Additionally, immune cells in the spleen and intestinal lymph nodes were severely imbalanced. These results suggest that the intraperitoneal injection model in first-generation mice is more efficient, stable, and significant. This study provides a theoretical basis for the efficient construction of CD models.
Collapse
Affiliation(s)
- Kexin Hu
- Zhengzhou Key Laboratory of Nutrition and Health Food, College of Food Science and Technology, Henan Agricultural University, Zhengzhou, China
- Longhu Laboratory, Zhengzhou, China
| | - Hongtao Ren
- Zhengzhou Key Laboratory of Nutrition and Health Food, College of Food Science and Technology, Henan Agricultural University, Zhengzhou, China
- Longhu Laboratory, Zhengzhou, China
| | - Chong Yuan
- Zhengzhou Key Laboratory of Nutrition and Health Food, College of Food Science and Technology, Henan Agricultural University, Zhengzhou, China
- Longhu Laboratory, Zhengzhou, China
| | - Zhigang Liu
- Longhu Laboratory, Zhengzhou, China
- College of Food Science and Engineering, Northwest A&F University, Shaanxi, China
| | - Xing Zhang
- Longhu Laboratory, Zhengzhou, China
- International Joint Research Center of National Animal Immunology, College of Veterinary Medicine, Henan Agricultural University, Zhengzhou, China
| | - Huaqiang Zhang
- Longhu Laboratory, Zhengzhou, China
- International Joint Research Center of National Animal Immunology, College of Veterinary Medicine, Henan Agricultural University, Zhengzhou, China
| | - Ning Zhou
- Zhengzhou Key Laboratory of Nutrition and Health Food, College of Food Science and Technology, Henan Agricultural University, Zhengzhou, China
- Longhu Laboratory, Zhengzhou, China
| | - Linlin Chen
- Zhengzhou Key Laboratory of Nutrition and Health Food, College of Food Science and Technology, Henan Agricultural University, Zhengzhou, China
- Longhu Laboratory, Zhengzhou, China
| | - Na Wang
- Zhengzhou Key Laboratory of Nutrition and Health Food, College of Food Science and Technology, Henan Agricultural University, Zhengzhou, China
- Longhu Laboratory, Zhengzhou, China
- International Joint Research Center of National Animal Immunology, College of Veterinary Medicine, Henan Agricultural University, Zhengzhou, China
| | - Gaiping Zhang
- Longhu Laboratory, Zhengzhou, China
- International Joint Research Center of National Animal Immunology, College of Veterinary Medicine, Henan Agricultural University, Zhengzhou, China
- School of Advanced Agricultural Sciences, Peking University, Beijing, China
| |
Collapse
|
12
|
Gao X, Yang C, Feng Z, Liu P, Liu Z. The signature of the small intestinal epithelial and immune cells in health and diseases. Chin Med J (Engl) 2025:00029330-990000000-01558. [PMID: 40394804 DOI: 10.1097/cm9.0000000000003615] [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: 12/05/2024] [Indexed: 05/22/2025] Open
Abstract
ABSTRACT The small intestine is essential for digestion, nutrient absorption, immune regulation, and microbial balance. Its epithelial lining, containing specialized cells like Paneth and tuft cells, is crucial for maintaining intestinal homeostasis. Paneth cells produce antimicrobial peptides and growth factors that support microbial regulation and intestinal stem cells, while tuft cells act as chemosensors, detecting environmental changes and modulating immune responses. Along with immune cells such as intraepithelial lymphocytes, innate lymphoid cells, T cells, and macrophages, they form a strong defense system that protects the epithelial barrier. Disruptions in this balance contribute to chronic inflammation, microbial dysbiosis, and compromised barrier function-key features of inflammatory bowel disease, celiac disease, and metabolic syndromes. Furthermore, dysfunctions in the small intestine and immune cells are linked to systemic diseases like obesity, diabetes, and autoimmune disorders. Recent research highlights promising therapeutic strategies, including modulation of epithelial and immune cell functions, probiotics, and gene editing to restore gut health and address systemic effects. This review emphasizes the pivotal roles of small intestinal epithelia and immune cells in maintaining intestinal homeostasis, their involvement in disease development, and emerging treatments for intestinal and systemic disorders.
Collapse
Affiliation(s)
- Xiang Gao
- Center for Inflammatory Bowel Disease Research, Department of Gastroenterology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai 200072, China
| | - Cuiping Yang
- Department of Gastroenterology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 201801, China
| | - Zhongsheng Feng
- Center for Inflammatory Bowel Disease Research, Department of Gastroenterology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai 200072, China
| | - Ping Liu
- Department of Gastroenterology, Wuhu First People's Hospital, Wuhu, Anhui 241000, China
| | - Zhanju Liu
- Center for Inflammatory Bowel Disease Research, Department of Gastroenterology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai 200072, China
| |
Collapse
|
13
|
Lin H, Lv Y, Liu H, Lin Z, Zhu N, Huang B. Baicalein alleviates chronic acute stress-induced irritable bowel syndrome-like symptoms in rats via modulating the ODC1/NF-κB pathway and oxidative stress. Biochem Biophys Res Commun 2025; 771:152058. [PMID: 40409117 DOI: 10.1016/j.bbrc.2025.152058] [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: 02/13/2025] [Revised: 04/14/2025] [Accepted: 05/19/2025] [Indexed: 05/25/2025]
Abstract
BACKGROUND As a persistent gastrointestinal disorder, irritable bowel syndrome (IBS) influences the gut-brain connection, leading to abdominal pain and altered bowel irregularities. Baicalein, a flavonoid extracted from Scutellaria baicalensis, is frequently utilized in anti-inflammatory treatments. This study aimed to explore baicalein's effectiveness in mitigating IBS symptoms triggered by both chronic and acute stress (CAS) and to uncover its fundamental mechanisms. METHODS Sprague-Dawley rats were employed to develop an IBS rat model by inducing CAS for five weeks. Each rat was randomly allocated to one of four experimental groups: model (M), low-dose baicalein (L), high-dose baicalein (H), and control (C). Baicalein was orally administered throughout the experiment. Behavioral assessments were conducted, including forced swimming, marble-burying, intestinal motility, and visceral sensitivity tests. Colonic tissues were collected for histopathological examination, evaluation of oxidative stress (MDA and SOD levels), and analysis of inflammatory cytokines, and ODC1/NF-κB pathway activation using Western blot assay, ELISA, and immunofluorescence. RESULTS Baicalein treatment notably ameliorated IBS-like symptoms, such as fecal pellet output and AWR scores, by alleviating stress-induced behavioral changes. Baicalein bolstered antioxidant defenses through boosting SOD activity and lowering MDA levels. Moreover, baicalein inhibited inflammatory responses, targeting IL-6, IL-1β, and TNF-α, while suppressing the expression of ODC1 and restraining NF-κB p65 phosphorylation within the colon. These results indicate that baicalein modulates oxidative stress and inflammation in CAS-associated IBS. CONCLUSION Baicalein demonstrates protection against CAS-induced IBS by diminishing intestinal inflammation, oxidative damage, and visceral hypersensitivity via suppression of the ODC1/NF-κB pathway. These findings underscore baicalein's potential as a therapeutic approach for IBS.
Collapse
Affiliation(s)
- Han Lin
- Department of Anesthesiology and Perioperative Medicine, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Key Laboratory of Pediatric Anesthesiology, Ministry of Education, Key Laboratory of Anesthesiology of Zhejiang Province, Wenzhou Medical University, People's Republic of China
| | - Yunlong Lv
- Department of Anesthesiology and Perioperative Medicine, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Key Laboratory of Pediatric Anesthesiology, Ministry of Education, Key Laboratory of Anesthesiology of Zhejiang Province, Wenzhou Medical University, People's Republic of China
| | - Huacheng Liu
- Department of Anesthesiology and Perioperative Medicine, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Key Laboratory of Pediatric Anesthesiology, Ministry of Education, Key Laboratory of Anesthesiology of Zhejiang Province, Wenzhou Medical University, People's Republic of China
| | - Zheng Lin
- Department of Anesthesiology and Perioperative Medicine, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Key Laboratory of Pediatric Anesthesiology, Ministry of Education, Key Laboratory of Anesthesiology of Zhejiang Province, Wenzhou Medical University, People's Republic of China
| | - Ning Zhu
- Department of Cardiology, The Third Affiliated Hospital of Shanghai University (Wenzhou People's Hospital), The Wenzhou Third Clinical Institute Affiliated to Wenzhou Medical University, No. 299 Guan Road, Wenzhou, 325000, Zhejiang Province, People's Republic of China.
| | - Bingwu Huang
- Department of Anesthesiology and Perioperative Medicine, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Key Laboratory of Pediatric Anesthesiology, Ministry of Education, Key Laboratory of Anesthesiology of Zhejiang Province, Wenzhou Medical University, People's Republic of China.
| |
Collapse
|
14
|
Prisco SZ, Blake M, Kazmirczak F, Moon R, Kremer BP, Hartweck LM, Kim M, Vogel N, Mendelson JB, Moutsoglou D, Thenappan T, Prins KW. Lactobacillus Restructures the Micro/Mycobiome to Combat Inflammation-Mediated Right Ventricular Dysfunction in Pulmonary Arterial Hypertension. Circ Heart Fail 2025:e012524. [PMID: 40376801 DOI: 10.1161/circheartfailure.124.012524] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/07/2024] [Accepted: 05/01/2025] [Indexed: 05/18/2025]
Abstract
BACKGROUND Inflammation suppresses right ventricular (RV) function in pulmonary arterial hypertension (PAH). In particular, we showed GP130 (glycoprotein-130) signaling promotes pathological microtubule remodeling and RV dysfunction in rodent PAH. Emerging data demonstrate the intestinal microbiome regulates systemic inflammation, but the impact of modulating the gut microbiome on the GP130-microtubule axis in RV failure is unknown. METHODS Two weeks following monocrotaline injection, rats were administered daily Lactobacillus rhamnosus (4×107 colony-forming units) via oral gavage for 10 days. Next-generation metagenomics and internal transcribed spacer 2 sequencing delineated fecal bacterial and fungal compositions. SomaScan proteomics measured levels of 7596 serum proteins. RV immunoblots quantified protein abundances. Light or super resolution confocal microscopy assessed RV, lung, and jejunal morphology. Echocardiography and invasive closed-chest pressure-volume loops evaluated PAH severity and RV function. The relationship between Lactobacillus abundance and RV function was assessed in 65 patients with PAH. RESULTS Lactobacillus administration restructured both the intestinal micro- and mycobiome. The alteration in the gut ecosystem improved intestinal health as demonstrated by increased jejunal villus length and glycocalyx thickness and diminished intestinal permeability biomarkers. Serum proteomics revealed Lactobacillus modulated systemic inflammation and decreased circulating GP130 ligands. Lactobacillus-mediated suppression of GP130 signaling blunted pathological microtubule remodeling in RV cardiomyocytes. Microtubule-associated phenotypes, including RV cardiomyocyte and nuclear hypertrophy, transverse tubule integrity, and connexin-43 localization, were all corrected with Lactobacillus. These cellular changes manifested as improved RV function despite no significant alteration in PAH severity. Finally, patients with PAH and detectable fecal Lactobacillus had superior RV function despite similar mean pulmonary arterial pressure and pulmonary vascular resistance as compared with those without detectable Lactobacillus. CONCLUSIONS Lactobacillus supplementation restructures the gut micro/mycobiome, restores intestinal health, dampens systemic inflammation, and reduces GP130 ligands and associated RV cardiomyocyte microtubule remodeling. These data identify a novel microbiome-inflammation-microtubule axis that has therapeutic relevance for RV dysfunction.
Collapse
Affiliation(s)
- Sasha Z Prisco
- Lillehei Heart Institute, Cardiovascular Division, Department of Medicine, University of Minnesota, Minneapolis (S.Z.P., M.B., F.K., R.M., B.P.K., L.M.H., M.K., N.V., J.B.M., T.T., K.W.P.)
| | - Madelyn Blake
- Lillehei Heart Institute, Cardiovascular Division, Department of Medicine, University of Minnesota, Minneapolis (S.Z.P., M.B., F.K., R.M., B.P.K., L.M.H., M.K., N.V., J.B.M., T.T., K.W.P.)
| | - Felipe Kazmirczak
- Lillehei Heart Institute, Cardiovascular Division, Department of Medicine, University of Minnesota, Minneapolis (S.Z.P., M.B., F.K., R.M., B.P.K., L.M.H., M.K., N.V., J.B.M., T.T., K.W.P.)
| | - Ryan Moon
- Lillehei Heart Institute, Cardiovascular Division, Department of Medicine, University of Minnesota, Minneapolis (S.Z.P., M.B., F.K., R.M., B.P.K., L.M.H., M.K., N.V., J.B.M., T.T., K.W.P.)
| | - Benjamin P Kremer
- Lillehei Heart Institute, Cardiovascular Division, Department of Medicine, University of Minnesota, Minneapolis (S.Z.P., M.B., F.K., R.M., B.P.K., L.M.H., M.K., N.V., J.B.M., T.T., K.W.P.)
| | - Lynn M Hartweck
- Lillehei Heart Institute, Cardiovascular Division, Department of Medicine, University of Minnesota, Minneapolis (S.Z.P., M.B., F.K., R.M., B.P.K., L.M.H., M.K., N.V., J.B.M., T.T., K.W.P.)
| | - Minwoo Kim
- Lillehei Heart Institute, Cardiovascular Division, Department of Medicine, University of Minnesota, Minneapolis (S.Z.P., M.B., F.K., R.M., B.P.K., L.M.H., M.K., N.V., J.B.M., T.T., K.W.P.)
| | - Neal Vogel
- Lillehei Heart Institute, Cardiovascular Division, Department of Medicine, University of Minnesota, Minneapolis (S.Z.P., M.B., F.K., R.M., B.P.K., L.M.H., M.K., N.V., J.B.M., T.T., K.W.P.)
| | - Jenna B Mendelson
- Lillehei Heart Institute, Cardiovascular Division, Department of Medicine, University of Minnesota, Minneapolis (S.Z.P., M.B., F.K., R.M., B.P.K., L.M.H., M.K., N.V., J.B.M., T.T., K.W.P.)
| | - Daphne Moutsoglou
- Gastroenterology Section, Minneapolis VA Health Care System, MN (D.M.)
- Department of Medicine, University of Minnesota, Minneapolis (D.M.)
| | - Thenappan Thenappan
- Lillehei Heart Institute, Cardiovascular Division, Department of Medicine, University of Minnesota, Minneapolis (S.Z.P., M.B., F.K., R.M., B.P.K., L.M.H., M.K., N.V., J.B.M., T.T., K.W.P.)
| | - Kurt W Prins
- Lillehei Heart Institute, Cardiovascular Division, Department of Medicine, University of Minnesota, Minneapolis (S.Z.P., M.B., F.K., R.M., B.P.K., L.M.H., M.K., N.V., J.B.M., T.T., K.W.P.)
| |
Collapse
|
15
|
Shi JY, Wang YJ, Bao QW, Qin YM, Li PP, Wu QQ, Xia CK, Wu DL, Xie SZ. Polygonatum cyrtonema Hua polysaccharide alleviates ulcerative colitis via gut microbiota-independent modulation of inflammatory immune response. Carbohydr Polym 2025; 356:123387. [PMID: 40049966 DOI: 10.1016/j.carbpol.2025.123387] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2024] [Revised: 01/30/2025] [Accepted: 02/13/2025] [Indexed: 05/13/2025]
Abstract
Polygonatum cyrtonema polysaccharides (PCP) exhibit ameliorative effects on colitis. However, whether the protective effects of PCP depend on the gut microbiota and how PCP regulates intestinal immune responses to alleviate colitis remain unclear. Therefore, this study investigated the effect of PCP against colitis focusing on the regulation of intestinal immune response. The PCP structure was reclassified as fructan. PCP treatment significantly reduced the symptoms of colitis. PCP restored IgA, ZO-1, Occludin, and MUC2 expression to enhance intestinal barrier function. Oral PCP administration markedly inhibited excessive inflammation-mediated immune response by modulating inflammatory cytokines secretion and Th17/Tregs cell balance and restored gut microbial composition. Interestingly, PCP still had a significant ameliorating effect on intestinal inflammation in colitis mice with gut microbial depletion by antibiotics. In the Caco-2/RAW264.7 co-culture inflammation model, PCP treatment improved the intestinal epithelial barrier function by regulating the inflammatory immune response through signal transduction pathways. Overall, these findings suggested that the alleviating effects of PCP on colitis are independent of gut microbiota, and that PCP can directly modulate the inflammatory immune response and intestinal barrier function, which in turn regulates gut microbiota. These findings will provide new insights into the action mechanism of natural polysaccharides in relieving colitis.
Collapse
Affiliation(s)
- Jin-Yang Shi
- School of Pharmacy, Anhui Province Key Laboratory of Bioactive Natural Products, Anhui Province Key Laboratory of Traditional Chinese Medicine Decoction Pieces of New Manufacturing Technology, Anhui University of Chinese Medicine, Hefei, Anhui 230012, China
| | - Yong-Jian Wang
- School of Pharmacy, Anhui Province Key Laboratory of Bioactive Natural Products, Anhui Province Key Laboratory of Traditional Chinese Medicine Decoction Pieces of New Manufacturing Technology, Anhui University of Chinese Medicine, Hefei, Anhui 230012, China
| | - Qian-Wen Bao
- School of Pharmacy, Anhui Province Key Laboratory of Bioactive Natural Products, Anhui Province Key Laboratory of Traditional Chinese Medicine Decoction Pieces of New Manufacturing Technology, Anhui University of Chinese Medicine, Hefei, Anhui 230012, China
| | - Ya-Min Qin
- School of Pharmacy, Anhui Province Key Laboratory of Bioactive Natural Products, Anhui Province Key Laboratory of Traditional Chinese Medicine Decoction Pieces of New Manufacturing Technology, Anhui University of Chinese Medicine, Hefei, Anhui 230012, China
| | - Pei-Pei Li
- School of Pharmacy, Anhui Province Key Laboratory of Bioactive Natural Products, Anhui Province Key Laboratory of Traditional Chinese Medicine Decoction Pieces of New Manufacturing Technology, Anhui University of Chinese Medicine, Hefei, Anhui 230012, China
| | - Qiao-Qiao Wu
- School of Pharmacy, Anhui Province Key Laboratory of Bioactive Natural Products, Anhui Province Key Laboratory of Traditional Chinese Medicine Decoction Pieces of New Manufacturing Technology, Anhui University of Chinese Medicine, Hefei, Anhui 230012, China
| | - Cheng-Kai Xia
- School of Pharmacy, Anhui Province Key Laboratory of Bioactive Natural Products, Anhui Province Key Laboratory of Traditional Chinese Medicine Decoction Pieces of New Manufacturing Technology, Anhui University of Chinese Medicine, Hefei, Anhui 230012, China
| | - De-Ling Wu
- School of Pharmacy, Anhui Province Key Laboratory of Bioactive Natural Products, Anhui Province Key Laboratory of Traditional Chinese Medicine Decoction Pieces of New Manufacturing Technology, Anhui University of Chinese Medicine, Hefei, Anhui 230012, China; Bozhou University, Bozhou, Anhui 236800, China.
| | - Song-Zi Xie
- School of Pharmacy, Anhui Province Key Laboratory of Bioactive Natural Products, Anhui Province Key Laboratory of Traditional Chinese Medicine Decoction Pieces of New Manufacturing Technology, Anhui University of Chinese Medicine, Hefei, Anhui 230012, China.
| |
Collapse
|
16
|
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
|
17
|
Adelfio M, Callen GE, He X, Paster BJ, Hasturk H, Ghezzi CE. Engineered Tissue Models to Decode Host-Microbiota Interactions. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025:e2417687. [PMID: 40364768 DOI: 10.1002/advs.202417687] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/28/2024] [Revised: 04/13/2025] [Indexed: 05/15/2025]
Abstract
A mutualistic co-evolution exists between the host and its associated microbiota in the human body. Bacteria establish ecological niches in various tissues of the body, locally influencing their physiology and functions, but also contributing to the well-being of the whole organism through systemic communication with other distant niches (axis). Emerging evidence indicates that when the composition of the microbiota inhabiting the niche changes toward a pathogenic state (dysbiosis) and interactions with the host become unbalanced, diseases may present. In addition, imbalances within a single niche can cause dysbiosis in distant organs. Current research efforts are focused on elucidating the mechanisms leading to dysbiosis, with the goal of restoring tissue homeostasis. In vitro models can provide critical experimental platforms to address this need, by reproducing the niche cyto-architecture and physiology with high fidelity. This review surveys current in in vitro host-microbiota research strategies and provides a roadmap that can guide the field in further developing physiologically relevant in vitro models of ecological niches, thus enabling investigation of the role of the microbiota in human health and diseases. Lastly, given the Food and Drug Administration Modernization Act 2.0, this review highlights emerging in vitro strategies to support the development and validation of new therapies on the market.
Collapse
Affiliation(s)
- Miryam Adelfio
- Department of Biomedical Engineering, University of Massachusetts-Lowell, Lowell, MA, 01854, USA
| | - Grace E Callen
- Department of Biomedical Engineering, University of Massachusetts-Lowell, Lowell, MA, 01854, USA
| | - Xuesong He
- ADA Forsyth Institute, 245 First St, Cambridge, MA, 02142, USA
| | - Bruce J Paster
- ADA Forsyth Institute, 245 First St, Cambridge, MA, 02142, USA
| | - Hatice Hasturk
- ADA Forsyth Institute, 245 First St, Cambridge, MA, 02142, USA
| | - Chiara E Ghezzi
- Department of Biomedical Engineering, University of Massachusetts-Lowell, Lowell, MA, 01854, USA
| |
Collapse
|
18
|
You M, Li J, Wang X, Liu Y, Chen S, Wang P. Targeting SLC7 A11 Ameliorates Ulcerative Colitis by Promoting Efferocytosis Through the ERK1/2 Pathway. Inflammation 2025:10.1007/s10753-025-02312-6. [PMID: 40360947 DOI: 10.1007/s10753-025-02312-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2025] [Revised: 03/28/2025] [Accepted: 05/02/2025] [Indexed: 05/15/2025]
Abstract
OBJECTIVE AND DESIGN This study investigates the effect and underlying mechanism of targeting SLC7A11 in mitigating dextran sulfate sodium (DSS)-induced intestinal inflammation and injury in colitis. METHODS We utilized wild-type and SLC7A11-/+ mice to assess the inflammatory damage in DSS-induced colitis in vivo. In vitro, colon tissues from patients with ulcerative colitis were analyzed to compare SLC7A11 expression between inflamed and non-inflamed regions. Further mechanistic insights were obtained using Caco-2 cells and bone marrow-derived dendritic cells (BMDCs). RESULTS In human colon tissues, SLC7A11 expression was significantly elevated in inflamed regions compared to non-inflamed areas, particularly in dendritic cells. In vivo inhibition of SLC7A11 markedly alleviated DSS-induced colitis symptoms. In vitro, suppressing SLC7A11 restored the integrity of the Caco-2 monolayer intestinal epithelial model. Both knockout and inhibition of SLC7A11 enhanced ERK1/2 phosphorylation and increased efferocytosis in BMDCs. CONCLUSIONS Targeting SLC7A11 augments dendritic cell efferocytosis and preserves intestinal epithelial barrier function, potentially offering a therapeutic avenue for alleviating ulcerative colitis.
Collapse
Affiliation(s)
- Meiyi You
- Department of Gastrointestinal Surgery, Peking University First Hospital, Beijing, 100034, People's Republic of China
| | - Jichang Li
- Department of Gastrointestinal Surgery, Peking University First Hospital, Beijing, 100034, People's Republic of China
| | - Xin Wang
- Department of Gastrointestinal Surgery, Peking University First Hospital, Beijing, 100034, People's Republic of China
| | - Yucun Liu
- Department of Gastrointestinal Surgery, Peking University First Hospital, Beijing, 100034, People's Republic of China
| | - Shanwen Chen
- Department of Gastrointestinal Surgery, Peking University First Hospital, Beijing, 100034, People's Republic of China.
| | - Pengyuan Wang
- Department of Gastrointestinal Surgery, Peking University First Hospital, Beijing, 100034, People's Republic of China.
| |
Collapse
|
19
|
Chen C, Qi M, Xu Z, Wen J, Tang W, Diao H, Li Z, Chu Y, Feng F, Tang Z. Sesamin improved growth and overall health in young animals by enhancing gut-liver axis function. Food Funct 2025. [PMID: 40351157 DOI: 10.1039/d4fo05933f] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/14/2025]
Abstract
The immature gastrointestinal development of infants and young animals made them more vulnerable to stress-related damage, which affected the gut-liver axis and consequently impaired their health and growth. This study used weaned piglets as a model to investigate how dietary sesamin regulated the gut-liver axis and impacted young animal health. We assessed gut-liver tissue morphology, measured key indicators of intestinal barrier damage, mucosal repair, antioxidant and immune pathways in the gut-liver system and serum, and analyzed microbial composition. We further explored the interactions between sesamin and the gut-liver axis through PLS-PM and molecular docking analysis. Results showed that sesamin enhanced intestinal barrier function, reduced liver damage, decreased oxidative stress, promoted anti-inflammatory immune responses, and enriched beneficial microbes, thereby promoting overall growth. Sesamin can enhance the health of young animals by regulating the function of the gut-liver axis.
Collapse
Affiliation(s)
- Chen Chen
- Laboratory for Bio-feed and Molecular Nutrition, College of Animal Science and Technology, Southwest University, Chongqing 400715, China.
| | - Min Qi
- Yunnan Animal Husbandry Station, Kunming 650225, China
| | - Zhiran Xu
- Laboratory for Bio-feed and Molecular Nutrition, College of Animal Science and Technology, Southwest University, Chongqing 400715, China.
| | - Jincheng Wen
- Laboratory for Bio-feed and Molecular Nutrition, College of Animal Science and Technology, Southwest University, Chongqing 400715, China.
| | - Wenjie Tang
- Animal Breeding and Genetics Key Laboratory of Sichuan Province, Sichuan Animal Science Academy, Chengdu 610066, China
- Livestock and Poultry Biological Products Key Laboratory of Sichuan Province, Sichuan Animtche Group Co., Ltd., Chengdu 610066, China
| | - Hui Diao
- Animal Breeding and Genetics Key Laboratory of Sichuan Province, Sichuan Animal Science Academy, Chengdu 610066, China
- Livestock and Poultry Biological Products Key Laboratory of Sichuan Province, Sichuan Animtche Group Co., Ltd., Chengdu 610066, China
| | - Zhangcheng Li
- Laboratory for Bio-feed and Molecular Nutrition, College of Animal Science and Technology, Southwest University, Chongqing 400715, China.
| | - Yunyun Chu
- Laboratory for Bio-feed and Molecular Nutrition, College of Animal Science and Technology, Southwest University, Chongqing 400715, China.
| | - Fu Feng
- Laboratory for Bio-feed and Molecular Nutrition, College of Animal Science and Technology, Southwest University, Chongqing 400715, China.
| | - Zhiru Tang
- Laboratory for Bio-feed and Molecular Nutrition, College of Animal Science and Technology, Southwest University, Chongqing 400715, China.
| |
Collapse
|
20
|
Jiang Y, Chen J, Du Y, Fan M, Shen L. Immune modulation for the patterns of epithelial cell death in inflammatory bowel disease. Int Immunopharmacol 2025; 154:114462. [PMID: 40186907 DOI: 10.1016/j.intimp.2025.114462] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2025] [Revised: 02/23/2025] [Accepted: 03/08/2025] [Indexed: 04/07/2025]
Abstract
Inflammatory bowel disease (IBD) is an inflammatory disease of the intestine whose primary pathological presentation is the destruction of the intestinal epithelium. The intestinal epithelium, located between the lumen and lamina propria, transmits luminal microbial signals to the immune cells in the lamina propria, which also modulate the intestinal epithelium. In IBD patients, intestinal epithelial cells (IECs) die dysfunction and the mucosal barrier is disrupted, leading to the recruitment of immune cells and the release of cytokines. In this review, we describe the structure and functions of the intestinal epithelium and mucosal barrier in the physiological state and under IBD conditions, as well as the patterns of epithelial cell death and how immune cells modulate the intestinal epithelium providing a reference for clinical research and drug development of IBD. In addition, according to the targeting of epithelial apoptosis and necroptotic pathways and the regulation of immune cells, we summarized some new methods for the treatment of IBD, such as necroptosis inhibitors, microbiome regulation, which provide potential ideas for the treatment of IBD. This review also describes the potential for integrating AI-driven approaches into innovation in IBD treatments.
Collapse
Affiliation(s)
- Yuting Jiang
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China; Center for Pharmaceutics Research, Shanghai Institute of Materia Medica Chinese Academy of Sciences, Shanghai 201203, China
| | - Jie Chen
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China; Center for Pharmaceutics Research, Shanghai Institute of Materia Medica Chinese Academy of Sciences, Shanghai 201203, China
| | - Yaoyao Du
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China; Center for Pharmaceutics Research, Shanghai Institute of Materia Medica Chinese Academy of Sciences, Shanghai 201203, China
| | - Minwei Fan
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China.
| | - Lan Shen
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China.
| |
Collapse
|
21
|
Wang HT, Weng JY, Amadou I, Song J, Jiang MQ, Ci WJ, Zhu JJ. Ligninoformic acid improved DSS-induced chronic colitis in mice by regulating intestinal flora and intestinal barrier. Microb Pathog 2025; 205:107670. [PMID: 40339622 DOI: 10.1016/j.micpath.2025.107670] [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: 12/07/2024] [Revised: 02/23/2025] [Accepted: 05/04/2025] [Indexed: 05/10/2025]
Abstract
Inflammatory Bowel Disease (IBD) is a complex intestinal disorder that typically triggers inflammatory responses, immune dysregulation, and gut microbiota imbalance. Lignoformic acid (LFA) is a lignin-derived compound containing benzene rings and hydroxyl functional groups. It has antioxidant properties and can regulate intestinal pH. This study aimed to investigate the improve effects of LFA on dextran sulfate sodium (DSS)-induced chronic colitis in mice. The results showed that LFA treatment significantly improved body weight and Disease Activity Index (DAI) in mice and alleviated colon damage. In terms of oxidative stress and anti-inflammatory effects, the expression of antioxidant enzymes such as Glutathione Peroxidase (GSH-PX) and Superoxide Dismutase (SOD) was dose-dependently enhanced in DSS-induced mice. LFA reduced the expression of Tumor Necrosis Factor-alpha (TNF-α) by modulating the TLR4/MyD88/NF-κB signaling pathway. Furthermore, LFA dose-dependently increased the abundance of beneficial bacteria, including Akkermansia and Lachnospiraceae, and promoted the production of short-chain fatty acids (SCFAs). These findings suggest that LFA could serve as a therapeutic agent for colitis by enhancing intestinal barrier integrity, regulating inflammation, and restoring gut microbiota balance.
Collapse
Affiliation(s)
- Hong-Tao Wang
- School of Food Science and Technology, Jiangnan University, Wuxi, 214122, Jiangsu, China; State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi, 214122, Jiangsu, China
| | - Jia-Yi Weng
- School of Food Science and Technology, Jiangnan University, Wuxi, 214122, Jiangsu, China; State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi, 214122, Jiangsu, China
| | - Issoufou Amadou
- Laboratory of Food Science and Technology, Faculty of Agriculture and Environment Sciences, Dan Dicko Dankoulodo University of Maradi, Niger
| | - Jie Song
- School of Food Science and Technology, Jiangnan University, Wuxi, 214122, Jiangsu, China; State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi, 214122, Jiangsu, China
| | - Meng-Qi Jiang
- School of Food Science and Technology, Jiangnan University, Wuxi, 214122, Jiangsu, China; State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi, 214122, Jiangsu, China
| | - Wen-Jia Ci
- School of Food Science and Technology, Jiangnan University, Wuxi, 214122, Jiangsu, China; State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi, 214122, Jiangsu, China
| | - Jian-Jin Zhu
- School of Food Science and Technology, Jiangnan University, Wuxi, 214122, Jiangsu, China; State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi, 214122, Jiangsu, China.
| |
Collapse
|
22
|
Agulló V, García-Viguera C, Medina S, Domínguez-Perles R. Bioaccessible (Poly)phenols of Winery Byproducts Modulate Pathogenic Mediators of Intestinal Bowel Disease: In Vitro Evidence. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2025; 73:11007-11018. [PMID: 40267141 PMCID: PMC12063181 DOI: 10.1021/acs.jafc.5c00916] [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] [Subscribe] [Scholar Register] [Received: 01/20/2025] [Revised: 04/11/2025] [Accepted: 04/16/2025] [Indexed: 04/25/2025]
Abstract
Intestinal inflammation entails a multifactorial pathophysiology, frequently treated by using anti-inflammatory drugs with severe side effects. At the same time, bioactive compounds present in plant materials and derived residues could contribute to reducing the use of such medications in terms of dosage and treatment length. Thus, the phytochemicals of winery byproducts, mainly represented by (poly)phenols, display significant anti-inflammatory and antioxidant potential. However, the functionality of bioaccessible fractions remains underexplored. This study uncovers the capacity of bioaccessible (poly)phenols of winery byproducts to modulate inflammatory mediators and secondary oxidative stress (OS). After in vitro simulated digestion, bioaccessible (poly)phenols exhibited significant inhibitory capacity of nitric oxide, interleukin (IL)-6, IL-8, and TNF-α production and prevented OS, lowering reactive oxygen species (ROS) resulting from disturbed cell metabolism while preserving the molecular machinery of cells, involving glutathione, catalase, superoxide dismutase, and glutathione peroxidase. The results retrieved suggested the relevance of specific profiles for efficiently preventing inflammation.
Collapse
Affiliation(s)
- Vicente Agulló
- Departamento
de Tecnología Agroalimentaria, EPSO,
Universidad Miguel Hernández, Carretera Beniel km 3.2, 03312 Orihuela, Alicante, Spain
| | - Cristina García-Viguera
- Laboratorio
de Fitoquímica y Alimentos Saludables (LabFAS), CEBAS, CSIC, Campus Universitario de Espinardo, Edificio 25, 30100 Murcia, Spain
| | - Sonia Medina
- Laboratorio
de Fitoquímica y Alimentos Saludables (LabFAS), CEBAS, CSIC, Campus Universitario de Espinardo, Edificio 25, 30100 Murcia, Spain
| | - Raúl Domínguez-Perles
- Laboratorio
de Fitoquímica y Alimentos Saludables (LabFAS), CEBAS, CSIC, Campus Universitario de Espinardo, Edificio 25, 30100 Murcia, Spain
| |
Collapse
|
23
|
Valderas-García E, Zafra R, Rufino-Moya PJ, Martínez-Moreno FJ, Ruiz-Campillo MT, Molina-Hernández V, González-Miguel J, Siles-Lucas M, Pérez J, Martínez-Moreno Á, Buffoni L. Evaluation of a novel vaccine candidate derived from newly excysted juveniles of Fasciola hepatica in sheep. Sci Rep 2025; 15:15512. [PMID: 40319070 PMCID: PMC12049522 DOI: 10.1038/s41598-025-00109-z] [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: 12/21/2024] [Accepted: 04/24/2025] [Indexed: 05/07/2025] Open
Abstract
Vaccine approaches for controlling Fasciola hepatica present a promising avenue, particularly considering increasing resistance to anthelmintic treatments and concerns over chemical residues. Targeting vaccine candidates that are expressed and secreted during the early infective stage of F. hepatica could offer an effective alternative. This approach aims to inhibit the invasion and migration of juvenile parasites, which have not yet fully developed their immune evasion mechanisms, thereby preventing parasite establishment and development in the host. In this study, we evaluated the host immune response and the protective efficacy of a vaccine cocktail comprising four antigens -KTSPIDP, VGHC1, CRTA, and CAL- in sheep infected with F. hepatica. These parasitic antigens were selected based on a proteomic analysis coupled with an "in vitro" interaction model between newly excysted juvenile worms and mouse intestinal epithelial cell cultures. Despite inducing a strong IgG1 response, vaccination did not reduce liver fluke burden nor faecal egg counts. However, it reduced liver pathology caused by the parasite. Our findings highlight the need for further research into early-stage interactions between F. hepatica and the host. Understanding these interactions could facilitate the progress of vaccines capable of disrupting parasite development and transmission in livestock, potentially reducing the economic and health impacts associated with fasciolosis.
Collapse
Affiliation(s)
- Elora Valderas-García
- Departamento de Sanidad Animal, Área de Parasitología, Facultad de Veterinaria, Universidad de Córdoba, Ctra. Madrid-Cádiz, km 396, 14071, Córdoba, Spain
- Departamento de Sanidad Animal, Facultad de Veterinaria, Universidad de León, Campus de Vegazana s/n, 24007, León, Spain
| | - Rafael Zafra
- Departamento de Sanidad Animal, Área de Parasitología, Facultad de Veterinaria, UIC Zoonosis y Enfermedades Emergentes ENZOEM, Universidad de Córdoba, Ctra. Madrid-Cádiz, km 396, 14071, Córdoba, Spain
| | - Pablo J Rufino-Moya
- Departamento de Sanidad Animal, Área de Parasitología, Facultad de Veterinaria, UIC Zoonosis y Enfermedades Emergentes ENZOEM, Universidad de Córdoba, Ctra. Madrid-Cádiz, km 396, 14071, Córdoba, Spain
| | - F Javier Martínez-Moreno
- Departamento de Sanidad Animal, Área de Parasitología, Facultad de Veterinaria, UIC Zoonosis y Enfermedades Emergentes ENZOEM, Universidad de Córdoba, Ctra. Madrid-Cádiz, km 396, 14071, Córdoba, Spain
| | - María T Ruiz-Campillo
- Departamento de Anatomía y Anatomía Patológica Comparadas y Toxicología, Facultad de Veterinaria, UIC Zoonosis y Enfermedades Emergentes ENZOEM, Universidad de Córdoba, Ctra. Madrid-Cádiz, km 396, 14014, Córdoba, Spain
| | - Verónica Molina-Hernández
- Departamento de Anatomía y Anatomía Patológica Comparadas y Toxicología, Facultad de Veterinaria, UIC Zoonosis y Enfermedades Emergentes ENZOEM, Universidad de Córdoba, Ctra. Madrid-Cádiz, km 396, 14014, Córdoba, Spain
| | - Javier González-Miguel
- Laboratory of Helminth Parasites of Zoonotic Importance (ATENEA), Institute of Natural Resources and Agrobiology of Salamanca (IRNASA-CSIC), C/Cordel de Merinas 40-52, 37008, Salamanca, Spain
| | - Mar Siles-Lucas
- Laboratory of Helminth Parasites of Zoonotic Importance (ATENEA), Institute of Natural Resources and Agrobiology of Salamanca (IRNASA-CSIC), C/Cordel de Merinas 40-52, 37008, Salamanca, Spain
| | - José Pérez
- Departamento de Anatomía y Anatomía Patológica Comparadas y Toxicología, Facultad de Veterinaria, UIC Zoonosis y Enfermedades Emergentes ENZOEM, Universidad de Córdoba, Ctra. Madrid-Cádiz, km 396, 14014, Córdoba, Spain
| | - Álvaro Martínez-Moreno
- Departamento de Sanidad Animal, Área de Parasitología, Facultad de Veterinaria, UIC Zoonosis y Enfermedades Emergentes ENZOEM, Universidad de Córdoba, Ctra. Madrid-Cádiz, km 396, 14071, Córdoba, Spain
| | - Leandro Buffoni
- Departamento de Sanidad Animal, Área de Parasitología, Facultad de Veterinaria, UIC Zoonosis y Enfermedades Emergentes ENZOEM, Universidad de Córdoba, Ctra. Madrid-Cádiz, km 396, 14071, Córdoba, Spain.
| |
Collapse
|
24
|
Yang JZ, Li JH, Liu JL, Zhou AD, Wang H, Xie XL, Zhang KK, Wang Q. Multiomics analysis revealed the effects of polystyrene nanoplastics at different environmentally relevant concentrations on intestinal homeostasis. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2025; 372:126050. [PMID: 40086783 DOI: 10.1016/j.envpol.2025.126050] [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: 11/25/2024] [Revised: 02/16/2025] [Accepted: 03/11/2025] [Indexed: 03/16/2025]
Abstract
Nanoplastics pollution is a global issue, with the digestive tract being one of the first affected organs, requiring further research on its impact on intestinal health. This study involved orally exposing mice to polystyrene nanoplastics (PS-NPs) at doses of 0.1, 0.5, or 2.5 mg/d for 42 days. The effects on intestinal health were thoroughly assessed via microbiomics, metabolomics, transcriptomics, and molecular biology. Our study demonstrated that the administration of all three doses of PS-NPs resulted in increased colonic permeability, heightened colonic and peripheral inflammation, reduced levels of antimicrobial peptides, and shortened colonic length. These effects may be attributed to a reduction in the abundance of probiotic bacteria, such as Clostridia_UCG-014, Roseburia, and Akkermansia, alongside an increase in the abundance of the pathogenic bacterium Desulfovibrionaceae induced by PS-NPs. Furthermore, we underscored the crucial role of histidine metabolism in PS-NPs-induced colonic injury, characterized by a significant reduction of L-histidine, which is closely related to microbial ecological dysregulation. Corresponding to microbiota deterioration and metabolic dysregulation, transcriptome analysis revealed that PS-NPs may disrupt colonic immune homeostasis by activating the TLR4/MyD88/NF-κB/NLRP3 signaling pathway. In conclusion, this study provided novel insights into the mechanisms by which PS-NPs disrupt intestinal homeostasis through integrated multiomics analysis, revealing critical molecular pathway and providing a scientific basis for future risk assessment of nanoplastics exposure.
Collapse
Affiliation(s)
- Jian-Zheng Yang
- Guangzhou Key Laboratory of Forensic Multi-Omics for Precision Identification, School of Forensic Medicine, Southern Medical University, Guangzhou, Guangdong, 510515, China
| | - Ji-Hui Li
- Guangzhou Key Laboratory of Forensic Multi-Omics for Precision Identification, School of Forensic Medicine, Southern Medical University, Guangzhou, Guangdong, 510515, China
| | - Jia-Li Liu
- Guangzhou Key Laboratory of Forensic Multi-Omics for Precision Identification, School of Forensic Medicine, Southern Medical University, Guangzhou, Guangdong, 510515, China
| | - An-Ding Zhou
- Guangzhou Key Laboratory of Forensic Multi-Omics for Precision Identification, School of Forensic Medicine, Southern Medical University, Guangzhou, Guangdong, 510515, China
| | - Hui Wang
- Department of Pediatric Surgery, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, National Children's Medical Center for South Central Region, Guangzhou, 510623, China
| | - Xiao-Li Xie
- Department of Toxicology, School of Public Health, Southern Medical University (Guangdong Provincial Key Laboratory of Tropical Disease Research), Guangzhou, Guangdong, 510515, China
| | - Kai-Kai Zhang
- Guangzhou Key Laboratory of Forensic Multi-Omics for Precision Identification, School of Forensic Medicine, Southern Medical University, Guangzhou, Guangdong, 510515, China.
| | - Qi Wang
- Guangzhou Key Laboratory of Forensic Multi-Omics for Precision Identification, School of Forensic Medicine, Southern Medical University, Guangzhou, Guangdong, 510515, China.
| |
Collapse
|
25
|
Scalavino V, Piccinno E, Giannelli G, Serino G. miR-369-3p Ameliorates Inflammation and Apoptosis in Intestinal Epithelial Cells via the MEK/ERK Signaling Pathway. Int J Mol Sci 2025; 26:4288. [PMID: 40362525 PMCID: PMC12072081 DOI: 10.3390/ijms26094288] [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: 03/18/2025] [Revised: 04/28/2025] [Accepted: 04/29/2025] [Indexed: 05/15/2025] Open
Abstract
Inflammatory Bowel Disease (IBD) is a group of chronic and recurrent inflammatory diseases characterized by prolonged inflammation of the intestinal tract. Although it has been proven that the immune system plays a crucial role in the pathogenesis of IBD, a defective intestinal epithelium is also responsible for chronic inflammation, hence causing an over-activation of the immune response. For this reason, a therapeutic approach that acts by improving impaired intestinal homeostasis could ensure a greater therapeutic efficacy in IBD. Mitogen-activated protein kinases (MAPKs) signaling pathways may be involved in the pathogenesis of IBD. It has been demonstrated that the inhibition of mitogen-activated protein kinase kinase 1 (MEK1) may be a potential treatment against IBD since it may restore the normal epithelial function and reduce apoptosis of intestinal epithelial cells (IECs). New therapeutic strategies are emerging including small molecules such as microRNAs (miRNAs). In this study, we aimed to demonstrate that miR-369-3p was able to modulate the MEK/ERK signaling pathway. As reported by in silico analysis, miR-369-3p was capable of pairing the 3'UTR of the MAP2K1 gene. In vitro analysis demonstrated that mimic transfection with miR-369-3p in epithelial cells downregulated the expression of MEK1, reduced the activation of ERK signaling, and modulated apoptosis of epithelial cells in response to TNF-α. Moreover, miR-369-3p significantly decreased the release of pro-inflammatory cytokine IL-8. These results support the potential of miR-369-3p to prevent apoptosis of IECs, responsible for a persistent inflammatory condition in IBD, highlighting its application value in the treatment of inflammatory disorders.
Collapse
Affiliation(s)
| | | | | | - Grazia Serino
- National Institute of Gastroenterology S. De Bellis, IRCCS Research Hospital, Via Turi 27, Castellana Grotte, 70013 Bari, Italy; (V.S.); (E.P.); (G.G.)
| |
Collapse
|
26
|
Wang Y, Hong Y. Genetic insights into the immunological basis of male infertility: a translational perspective. F&S SCIENCE 2025; 6:130-140. [PMID: 39988235 DOI: 10.1016/j.xfss.2025.02.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/10/2025] [Revised: 02/14/2025] [Accepted: 02/14/2025] [Indexed: 02/25/2025]
Abstract
OBJECTIVE To elaborate the causal relationships between specific immunocyte phenotypes and male infertility. DESIGN Mendelian randomization using genome-wide association study data. SUBJECTS Large cohorts of European ancestry. EXPOSURE 731 immunocyte phenotypes or male infertility. MAIN OUTCOMES MEASURES Genetic variants were used as instrumental variables to infer causality, minimizing confounding and bias. The causal associations were assessed using the inverse variance-weighted (IVW) method for primary analysis, and the findings were validated using MR-Egger, Weighted Median, Simple Mode, and Weighted Mode approaches. Additional sensitivity analyses were performed to validate the robustness of the findings. RESULTS Our analysis identified significant causal associations between specific immunocyte phenotypes and male infertility. Phenotypes such as naive-mature B cell %lymphocyte (odds ratio [OR] = 1.257) and IgD- CD38dim %B cell (OR = 1.100) were positively associated with increased infertility risk, whereas phenotypes like CD39+ CD8br %T cell (OR = 0.856) and B cells activator of the TNF-α family receptor (BAFF-R) on transitional (OR = 0.833) were negatively associated, suggesting a protective effect. Additionally, reverse MR analysis revealed that male infertility might causally affect certain immunocyte phenotypes, including CD14- CD16+ monocyte %monocyte (OR = 1.049). CONCLUSION This study provides robust evidence for the causal role of specific immunocyte phenotypes in male infertility and highlights the bidirectional relationship between immune function and reproductive health. These findings provide new insights into the immunological factors contributing to male infertility and suggest potential biomarkers and therapeutic targets for future research and clinical interventions.
Collapse
Affiliation(s)
- Yi Wang
- The First School of Medicine, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Yanggang Hong
- The Second School of Medicine, Wenzhou Medical University, Wenzhou, Zhejiang, China.
| |
Collapse
|
27
|
Hao Y, Wang C, Wang L, Hu L, Duan T, Zhang R, Yang X, Li T. Nondigestible stachyose alleviates cyclophosphamide-induced small intestinal mucosal injury in mice by regulating intestinal exosomal miRNAs, independently of the gut microbiota. Food Res Int 2025; 209:116258. [PMID: 40253186 DOI: 10.1016/j.foodres.2025.116258] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2025] [Revised: 02/16/2025] [Accepted: 03/11/2025] [Indexed: 04/21/2025]
Abstract
Stachyose has traditionally been considered to exert prebiotic effects primarily through its interaction with gut microbiota. However, this study reveals a novel mechanism by which stachyose alleviates cyclophosphamide (CY)-induced small intestinal mucosa disruption by regulating the intestinal exosomal miRNAs, without relying on the gut microbiota. Specifically, stachyose significantly mitigates CY-caused damage to the intestinal permeability, oxidative stress, and the structure of intestinal villi and crypts in pseudo-germ-free (PGF) mice. The immunofluorescence staining and qPCR analyses show that stachyose treatment restores CY-caused abnormal changes on the levels of tight junction proteins including MUC2, Occludin, Claudin-1, and ZO-1, and pro-inflammatory cytokines including TNF-α, IL-1β, and IL-2. Furthermore, by conducting fecal miRNA transplantation experiment, we further demonstrated that, similar to stachyose, stachyose-shaped intestinal miRNAs protect against CY-induced intestinal mucosal damage in PGF mice. In summary, this study provides new scientific evidence for the direct interaction between nondigestible stachyose and the proximal small intestine. It also opens new avenues for further investigation into the systemic nutritional functions of stachyose, particularly the health benefits of stachyose in the upper gastrointestinal tract.
Collapse
Affiliation(s)
- Yuhang Hao
- Shaanxi Engineering Laboratory for Food Green Processing and Safety Control, and Shaanxi Key Laboratory for Hazard Factors Assessment in Processing and Storage of Agricultural Products, College of Food Engineering and Nutritional Science, Shaanxi Normal University, Xi'an 710119, China
| | - Chennan Wang
- Shaanxi Engineering Laboratory for Food Green Processing and Safety Control, and Shaanxi Key Laboratory for Hazard Factors Assessment in Processing and Storage of Agricultural Products, College of Food Engineering and Nutritional Science, Shaanxi Normal University, Xi'an 710119, China
| | - Lu Wang
- Shaanxi Engineering Laboratory for Food Green Processing and Safety Control, and Shaanxi Key Laboratory for Hazard Factors Assessment in Processing and Storage of Agricultural Products, College of Food Engineering and Nutritional Science, Shaanxi Normal University, Xi'an 710119, China
| | - Lili Hu
- Shaanxi Engineering Laboratory for Food Green Processing and Safety Control, and Shaanxi Key Laboratory for Hazard Factors Assessment in Processing and Storage of Agricultural Products, College of Food Engineering and Nutritional Science, Shaanxi Normal University, Xi'an 710119, China
| | - Tianchi Duan
- Shaanxi Engineering Laboratory for Food Green Processing and Safety Control, and Shaanxi Key Laboratory for Hazard Factors Assessment in Processing and Storage of Agricultural Products, College of Food Engineering and Nutritional Science, Shaanxi Normal University, Xi'an 710119, China
| | - Runguang Zhang
- Shaanxi Engineering Laboratory for Food Green Processing and Safety Control, and Shaanxi Key Laboratory for Hazard Factors Assessment in Processing and Storage of Agricultural Products, College of Food Engineering and Nutritional Science, Shaanxi Normal University, Xi'an 710119, China
| | - Xingbin Yang
- Shaanxi Engineering Laboratory for Food Green Processing and Safety Control, and Shaanxi Key Laboratory for Hazard Factors Assessment in Processing and Storage of Agricultural Products, College of Food Engineering and Nutritional Science, Shaanxi Normal University, Xi'an 710119, China
| | - Ting Li
- Shaanxi Engineering Laboratory for Food Green Processing and Safety Control, and Shaanxi Key Laboratory for Hazard Factors Assessment in Processing and Storage of Agricultural Products, College of Food Engineering and Nutritional Science, Shaanxi Normal University, Xi'an 710119, China..
| |
Collapse
|
28
|
Sun Y, Liang J, Ma J, Su W, Cheng S. Curcumin-loaded Pickering emulsions stabilized by Spanish mackerel protein-pectin for ameliorating ulcerative colitis through barrier repair and anti-inflammatory effects. Int J Biol Macromol 2025; 307:141934. [PMID: 40068742 DOI: 10.1016/j.ijbiomac.2025.141934] [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: 12/18/2024] [Revised: 02/18/2025] [Accepted: 03/08/2025] [Indexed: 03/14/2025]
Abstract
Curcumin (Cur) has gained considerable recognition because of its anti-inflammatory and antioxidant effects as a bioactive compound, but its water insolubility and low bioaccessibility limit its application in food industry. In this study, Pickering emulsion stabilized by Spanish mackerel protein and pectin complex (SMP/PEC) was prepared to deliver curcumin, and its alleviating effects on DSS-induced ulcerative colitis (UC) were investigated. The emulsions stabilized by SMP/PEC 1:1 inhibited phase separation, had good rheological properties and the emulsions were stable at high temperatures, centrifugation, salt ions, and pH conditions. Meanwhile, the bioaccessibility of Cur loaded in the emulsion was 2.6 times higher than that of free Cur in corn oil. Furthermore, in vivo experiments have demonstrated that Cur loaded Pickering emulsion stabilized by SMP/PEC could enhance the proliferation of goblet cells and the expression of tight junction proteins, and restore the structural integrity of colonic tissues. Additionally, it has been shown to downregulate pro-inflammatory cytokines, such as serum IL-6 and TNF-α, and reduce the levels of MPO, NO, and other biomarkers in colonic tissues. Concurrently, gut microbiota demonstrated that emulsion stabilized by SMP/PEC could regulate the relative abundance of intestinal microorganisms, facilitating the increase of beneficial bacteria and reducing the level of harmful bacteria in the gut. In conclusion, the findings of this study indicated that SPECPE might be a potentially beneficial dietary supplement for the prevention of ulcerative colitis.
Collapse
Affiliation(s)
- Yu Sun
- State Key Laboratory of Marine Food Processing and Safety Control, Dalian Polytechnic University, Dalian 116034, Liaoning, China; Academy of Food Interdisciplinary Science, School of Food Science and Technology, Dalian Polytechnic University, Dalian 116034, Liaoning, China; National Engineering Research Center of Seafood, Dalian Polytechnic University, Dalian 116034, Liaoning, China; Collaborative Innovation Center of Seafood Deep Processing, Dalian Polytechnic University, Dalian 116034, Liaoning, China; Dalian Key Laboratory for Precision Nutrition, Dalian Polytechnic University, Dalian 116034, Liaoning, China
| | - Jiayue Liang
- State Key Laboratory of Marine Food Processing and Safety Control, Dalian Polytechnic University, Dalian 116034, Liaoning, China; Academy of Food Interdisciplinary Science, School of Food Science and Technology, Dalian Polytechnic University, Dalian 116034, Liaoning, China; National Engineering Research Center of Seafood, Dalian Polytechnic University, Dalian 116034, Liaoning, China; Collaborative Innovation Center of Seafood Deep Processing, Dalian Polytechnic University, Dalian 116034, Liaoning, China; Dalian Key Laboratory for Precision Nutrition, Dalian Polytechnic University, Dalian 116034, Liaoning, China
| | - Jiale Ma
- State Key Laboratory of Marine Food Processing and Safety Control, Dalian Polytechnic University, Dalian 116034, Liaoning, China; Academy of Food Interdisciplinary Science, School of Food Science and Technology, Dalian Polytechnic University, Dalian 116034, Liaoning, China; National Engineering Research Center of Seafood, Dalian Polytechnic University, Dalian 116034, Liaoning, China; Collaborative Innovation Center of Seafood Deep Processing, Dalian Polytechnic University, Dalian 116034, Liaoning, China; Dalian Key Laboratory for Precision Nutrition, Dalian Polytechnic University, Dalian 116034, Liaoning, China
| | - Wentao Su
- State Key Laboratory of Marine Food Processing and Safety Control, Dalian Polytechnic University, Dalian 116034, Liaoning, China; Academy of Food Interdisciplinary Science, School of Food Science and Technology, Dalian Polytechnic University, Dalian 116034, Liaoning, China; National Engineering Research Center of Seafood, Dalian Polytechnic University, Dalian 116034, Liaoning, China; Collaborative Innovation Center of Seafood Deep Processing, Dalian Polytechnic University, Dalian 116034, Liaoning, China; Dalian Key Laboratory for Precision Nutrition, Dalian Polytechnic University, Dalian 116034, Liaoning, China
| | - Shasha Cheng
- State Key Laboratory of Marine Food Processing and Safety Control, Dalian Polytechnic University, Dalian 116034, Liaoning, China; Academy of Food Interdisciplinary Science, School of Food Science and Technology, Dalian Polytechnic University, Dalian 116034, Liaoning, China; National Engineering Research Center of Seafood, Dalian Polytechnic University, Dalian 116034, Liaoning, China; Collaborative Innovation Center of Seafood Deep Processing, Dalian Polytechnic University, Dalian 116034, Liaoning, China; Dalian Key Laboratory for Precision Nutrition, Dalian Polytechnic University, Dalian 116034, Liaoning, China.
| |
Collapse
|
29
|
Kim SM, Shin HY, Sim SH, Rho Y, Yu KW, Shin KS. Effects of pectin-type polysaccharides derived from Houttuynia cordata on the intestinal immune system in native C3H/HeN mice. Int J Biol Macromol 2025; 309:142995. [PMID: 40210073 DOI: 10.1016/j.ijbiomac.2025.142995] [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: 12/26/2024] [Revised: 03/18/2025] [Accepted: 04/07/2025] [Indexed: 04/12/2025]
Abstract
Dietary polysaccharides have attracted considerable attention because of their species-specific chemical composition and diverse biological activities. Polysaccharides derived from Houttuynia cordata (HC) exhibit broad pharmacological activities, emphasizing their diverse roles in biological processes. This study investigated the effects of HC-derived polysaccharides on Peyer's patch (PP)-mediated intestinal immune response. The crude polysaccharide (HCP) isolated from HC extract (HCE) was identified as a pectin-type polysaccharide rich in the homogalacturonan (HG) domain, which was further hydrolyzed by endo-polygalacturonase to produce HCPE. In vitro, HCPE significantly enhanced PP-mediated cytokine secretion and bone marrow cell (BMC) proliferation compared with HCP. In vivo, a 4-week treatment with oral HCPE stimulated PP-mediated secretion of hematopoietic growth factors and promoted BMC proliferation. Additionally, HCPE upregulated IgA-associated factors, leading to increased IgA levels in the small intestine, serum, and feces, while also significantly elevating short-chain fatty acid levels, potentially improving the gut environment. To the best of our knowledge, this is the first study to systematically analyze PP-mediated intestinal immunostimulatory activity of HC-derived polysaccharides both in vitro and in vivo. These findings provide valuable insights into the potential of HCPE as an intestinal immunostimulatory agent and establish a foundation for future research on the immunological effects of HC-derived polysaccharides.
Collapse
Affiliation(s)
- So Min Kim
- Department of Food Science and Biotechnology, Kyonggi University, Suwon 16227, Republic of Korea
| | - Hyun Young Shin
- Transdisciplinary Major in Learning Health Systems, Department of Integrated Biomedical and Life Science, Graduate School, Korea University, Seoul 02841, Republic of Korea.
| | - Se Hyeon Sim
- Department of Food Science and Biotechnology, Kyonggi University, Suwon 16227, Republic of Korea
| | - YangKook Rho
- PharmacoBio Co. Ltd., Seongnam 13219, Republic of Korea.
| | - Kwang-Won Yu
- Major in Food & Nutrition, Korea National University of Transportation, Chungbuk 27909, Republic of Korea.
| | - Kwang-Soon Shin
- Department of Food Science and Biotechnology, Kyonggi University, Suwon 16227, Republic of Korea.
| |
Collapse
|
30
|
Chen Y, Qi W, Peng W, Fang W, Song G, Hao Y, Wang Y. Cyanidin-3-glucoside improves cognitive impairment in naturally aging mice by modulating the gut microbiota and activating the ERK/CREB/BDNF pathway. Food Res Int 2025; 208:116086. [PMID: 40263878 DOI: 10.1016/j.foodres.2025.116086] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2024] [Revised: 01/21/2025] [Accepted: 02/22/2025] [Indexed: 04/24/2025]
Abstract
Aging-related cognitive impairment has emerged as a major health-threatening factor among the elderly, and cyanidin-3-glucoside (C3G) is a prominent anthocyanin with biological activities, including antioxidant, anti-inflammatory, and alleviation of neurodegeneration. However, the role of C3G in alleviating natural aging-induced cognitive impairment and the underlying mechanisms thereof remain unclear. In this study, experimental methods mainly included biochemical analysis, pathological analysis, immunofluorescence staining, transmission electron microscopy analysis, western blot, as well as the determination of the gut microbiota composition and detection of metabolites. We found that C3G may exert neuroprotective effects and promote brain health by alleviating brain atrophy and neuroinflammation, enhancing brain antioxidant capacity, regulating neurotransmitter expression and hypothalamic-pituitary-adrenal axis activity, and attenuating blood-brain barrier and hippocampal synaptic damage. Furthermore, C3G also promotes gut health by decreasing inflammatory responses and intestinal tissue crypt damage, upregulating the expression of tight junction proteins, and attenuating intestinal damage. Notably, C3G regulated the microbiota composition in different intestinal segments and intestinal mucosa, as well as the metabolic homeostasis of gut microbiota metabolites, such as short-chain fatty acids (SCFAs), amino acids, and bile acids. Substantially increased levels of SCFAs could activate the extracellular signal-regulated kinase (ERK)/cAMP response element-binding protein (CREB)/brain-derived neurotrophic factor (BDNF) signaling pathway by acting on the G protein-coupled receptors. Correlation analysis indicated that increased gut microbiota, such as Faecalibaculum and Bifidobacterium, and elevated SCFAs were positively correlated with behavioral improvement and brain health. In conclusion, our findings reveal that C3G has the potential to improve natural aging-induced cognitive impairment by modulating the gut microbiota and its metabolite SCFAs, thereby activating the ERK/CREB/BDNF pathway.
Collapse
Affiliation(s)
- Yuyu Chen
- Academy of National Food and Strategic Reserves Administration, Beijing 100037, PR China; School of Health Science and Engineering, University of Shanghai for Science and Technology, Shanghai 200093, China
| | - Wentao Qi
- Academy of National Food and Strategic Reserves Administration, Beijing 100037, PR China
| | - Wenting Peng
- Academy of National Food and Strategic Reserves Administration, Beijing 100037, PR China
| | - Wei Fang
- Academy of National Food and Strategic Reserves Administration, Beijing 100037, PR China
| | - Ge Song
- Academy of National Food and Strategic Reserves Administration, Beijing 100037, PR China
| | - Yanling Hao
- Key Laboratory of Precision Nutrition and Food Quality, Department of Nutrition and Health, China Agricultural University, Beijing 100093, China
| | - Yong Wang
- Academy of National Food and Strategic Reserves Administration, Beijing 100037, PR China.
| |
Collapse
|
31
|
Tan T, Li J, Fan W, Shang K, Yang C, Liu X, Zhu S, Liu T, Wang J, Li Y, Lin Y. Tetrahedral Framework Nucleic Acid Relieves Sepsis-Induced Intestinal Injury by Regulating M2 Macrophages. Cell Prolif 2025; 58:e13803. [PMID: 39844345 PMCID: PMC12099223 DOI: 10.1111/cpr.13803] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2024] [Revised: 12/22/2024] [Accepted: 12/30/2024] [Indexed: 01/24/2025] Open
Abstract
This study aimed to clarify the role and mechanism of tetrahedral framework nucleic acids (tFNAs) in regulating M2 macrophages to reduce intestinal injury. An intestinal injury model was established by intraperitoneal injection of lipopolysaccharides (LPS) in mice to explore the alleviating effects of tFNAs on intestinal injury. Inflammatory factors were detected by quantitative polymerase chain reaction (qPCR) and enzyme-linked immunosorbent assay (ELISA). The intestinal barrier and permeability were assessed using western blotting and immunohistochemistry. Macrophages in the gut were localised and quantified using immunofluorescence. Western blotting was used to investigate the role and mechanism of tFNAs in regulating macrophages and alleviating inflammation in the injured intestines. These results show that tFNAs attenuated sepsis-induced intestinal injury. tFNAs can also promote the intestinal barrier reconstruction and reduce intestinal permeability. In vivo, tFNAs accelerated the aggregation of M2 macrophages at an early stage of injury and reduced the number of M1 macrophages in the intestine. In addition, tFNAs enhanced the clearance ability of intestinal macrophages. They activated the signalling and transcription activating factor 1(STAT1) and cytokine signalling inhibitory factor 1/3 (SOCS1/3) pathways by increasing the expression of the phagocytic receptor Mertk. These findings indicated that tFNAs can alleviate sepsis-induced intestinal injury by regulating M2 macrophages, providing a new option for treating intestinal injury.
Collapse
Affiliation(s)
- Tingting Tan
- Department of Critical Care Medicine, Shanghai Tenth People's HospitalTongji University School of MedicineShanghaiPeople's Republic of China
- Key Laboratory of Pathogen‐Host InteractionMinistry of EducationBeijingPeople's Republic of China
- School of Basic Medicine and Clinical PharmacyChina Pharmaceutical UniversityNanjingPeople's Republic of China
| | - Jiajie Li
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of StomatologySichuan University, Chengdu, Department of Burns and Plastic SurgeryChengduPeople's Republic of China
| | - Wensi Fan
- Department of Critical Care Medicine, Shanghai Tenth People's HospitalTongji University School of MedicineShanghaiPeople's Republic of China
| | - Kangni Shang
- Department of Critical Care Medicine, Shanghai Tenth People's HospitalTongji University School of MedicineShanghaiPeople's Republic of China
- School of Basic Medicine and Clinical PharmacyChina Pharmaceutical UniversityNanjingPeople's Republic of China
| | - Chujun Yang
- Department of Critical Care Medicine, Shanghai Tenth People's HospitalTongji University School of MedicineShanghaiPeople's Republic of China
| | - Xiaohao Liu
- Department of Critical Care Medicine, Shanghai Tenth People's HospitalTongji University School of MedicineShanghaiPeople's Republic of China
| | - Shihui Zhu
- Shanghai Children's Medical CenterShanghai Jiao Tong University School of MedicineShanghaiPeople's Republic of China
| | - Tong Liu
- Department of Critical Care Medicine, Zhongshan HospitalFudan UniversityShanghaiPeople's Republic of China
| | - Junjie Wang
- Department of Critical Care Medicine, Shanghai Tenth People's HospitalTongji University School of MedicineShanghaiPeople's Republic of China
- Key Laboratory of Pathogen‐Host InteractionMinistry of EducationBeijingPeople's Republic of China
| | - Yingchuan Li
- Department of Critical Care Medicine, Shanghai Tenth People's HospitalTongji University School of MedicineShanghaiPeople's Republic of China
- Key Laboratory of Pathogen‐Host InteractionMinistry of EducationBeijingPeople's Republic of China
| | - Yunfeng Lin
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of StomatologySichuan University, Chengdu, Department of Burns and Plastic SurgeryChengduPeople's Republic of China
| |
Collapse
|
32
|
Tang R, Jiang L, Ji Q, Kang P, Liu Y, Miao P, Xu X, Tang M. Resveratrol targeting MDM2/P53/PUMA axis to inhibit colonocyte apoptosis in DSS-induced ulcerative colitis mice. Front Pharmacol 2025; 16:1572906. [PMID: 40371345 PMCID: PMC12075554 DOI: 10.3389/fphar.2025.1572906] [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: 02/07/2025] [Accepted: 04/07/2025] [Indexed: 05/16/2025] Open
Abstract
Background Resveratrol, a naturally occurring polyphenolic compound found in grapes, berries, and traditional medicinal plants like Polygonum cuspidatum, has been used for centuries in traditional medicine systems for its anti-inflammatory, antioxidant, and cardioprotective properties. Ulcerative colitis (UC), a chronic inflammatory bowel disease, is characterized by intestinal barrier disruption due to excessive colonocyte apoptosis, leading to increased permeability and inflammation. Targeting apoptosis is a critical therapeutic strategy for UC. Aim of the study This study aims to investigate the therapeutic potential of Resveratrol in ulcerative colitis (UC) by targeting excessive colonocyte apoptosis and intestinal barrier dysfunction. Specifically, we seek to elucidate the mechanisms through which Resveratrol modulates apoptosis-related pathways and evaluate its efficacy in restoring intestinal homeostasis and mitigating UC progression in both in vivo and in vitro models. Materials and Methods We used dextran sulfate sodium (DSS) to induce UC in a mouse model. Colonic damage was assessed through colonic length measurement, histological examination, and immunofluorescence staining. Single-cell sequencing was employed to explore changes in the colonic immune microenvironment and cellular signaling pathways after Resveratrol treatment. In vitro, colonocytes isolated from healthy mouse colonic tissue were exposed to TGF-β to induce apoptosis. DNA fragmentation, mitochondrial membrane potential, and annexin V/propidium iodide staining were used to assess apoptosis. Additionally, we employed an Adeno-Associated Virus system to overexpress MDM2 in the colon and evaluate its protective role in DSS-induced UC. Results Resveratrol treatment effectively repaired colonic damage in the UC mouse model by significantly increasing colon length, reducing inflammatory cell infiltration, and mitigating mucosal injury. Single-cell sequencing revealed that Resveratrol primarily targeted colonocytes, decreasing genes related to apoptosis and the P53 pathway. In vitro, Resveratrol reduced DNA fragmentation, apoptotic cell populations, and increased mitochondrial membrane potential in a dose-dependent manner. Furthermore, Resveratrol increased MDM2 expression, inhibiting P53 and downstream pro-apoptotic signaling. Nutlin-3a, an MDM2 inhibitor, reversed the anti-apoptotic effects of Resveratrol. Overexpression of MDM2 in the colon protected against DSS-induced damage. Conclusion Resveratrol is an effective treatment for DSS-induced UC, primarily by inhibiting excessive apoptosis in colonocytes through the MDM2/P53/PUMA axis. MDM2 presents a promising therapeutic target for UC treatment.
Collapse
Affiliation(s)
- Rui Tang
- Department of Pathology, Yaan People’s Hospital, Yaan, China
- School of Basic Medical Sciences, Southwest Medical University, Luzhou, China
| | - Ling Jiang
- School of Basic Medical Sciences, Southwest Medical University, Luzhou, China
| | - Quan Ji
- Department of Anesthesiology Management, the Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Pengyuan Kang
- Department of Microbiology and Infectious Disease Center, School of Basic Medical Sciences, Peking University, Beijing, China
| | - Yuan Liu
- School of Basic Medical Sciences, Southwest Medical University, Luzhou, China
| | - Pengyu Miao
- School of Basic Medical Sciences, Southwest Medical University, Luzhou, China
| | - Xiaofan Xu
- Department of Health Technology and Informatics, The Hong Kong Polytechnic University, Kowloon, China
| | - Mingxi Tang
- Department of Pathology, Yaan People’s Hospital, Yaan, China
- Department of Pathology, The Affiliated Hospital, Southwest Medical University, Luzhou, China
- Precision Medicine Center, Yaan People’s Hospital, Yaan, China
| |
Collapse
|
33
|
Zhao Q, Shao M, Ma L, Zhou R. Insights into Modeling Inflammatory Bowel Disease from Stem Cell Derived Intestinal Organoids. Stem Cell Rev Rep 2025:10.1007/s12015-025-10887-8. [PMID: 40299197 DOI: 10.1007/s12015-025-10887-8] [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] [Accepted: 04/13/2025] [Indexed: 04/30/2025]
Abstract
Inflammatory bowel disease (IBD), encompassing Crohn's disease (CD) and ulcerative colitis (UC), is a multifactorial, immune-mediated condition marked by chronic gastrointestinal inflammation. This condition significantly impairs patients' quality of life and represents a major public health challenge globally. Pathogenesis of IBD arises from complex interplay among genetic predisposition, environmental factors, immune dysregulation, and microbial dysbiosis. Although significant strides have been made in unraveling these mechanisms, existing therapeutic options remain inadequate in addressing the full spectrum of clinical needs, underscoring the urgent demand for innovative strategies. Regenerative medicine has emerged as a promising frontier, offering novel tools for therapeutic development. We briefly consolidated current knowledge on IBD pathogenesis and treatments, emphasized the pivotal potential of human intestinal organoids (including adult stem cell-derived organoids and pluripotent stem cell- derived organoids) as a robust platform for mechanistic studies and treatment exploration. Leveraging this technology, we aim to advance personalized and next-generation therapies for IBD.
Collapse
Affiliation(s)
- Qi Zhao
- The Affiliated Wenling Hospital of Wenzhou Medical University (The First People's Hospital of Wenling), Taizhou, Zhejiang Province, China
| | - Miaoli Shao
- The Affiliated Wenling Hospital of Wenzhou Medical University (The First People's Hospital of Wenling), Taizhou, Zhejiang Province, China
| | - Lisha Ma
- The Affiliated Wenling Hospital of Wenzhou Medical University (The First People's Hospital of Wenling), Taizhou, Zhejiang Province, China
| | - Renfang Zhou
- The Affiliated Wenling Hospital of Wenzhou Medical University (The First People's Hospital of Wenling), Taizhou, Zhejiang Province, China.
| |
Collapse
|
34
|
Yang S, Li Y, Ruan R, Yu J, Zhu B, Lou H, Zhang X, Wang S. Exogenous TSG-6 treatment alleviates DSS-induced colitis in mice by modulating Pou2f3 and promoting tuft cells differentiation. Mol Med 2025; 31:157. [PMID: 40301757 PMCID: PMC12042439 DOI: 10.1186/s10020-025-01230-5] [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: 11/08/2024] [Accepted: 04/24/2025] [Indexed: 05/01/2025] Open
Abstract
BACKGROUND Whereas intestinal epithelial barrier dysfunction is implicated in inflammatory bowel disease (IBD), the underlying mechanisms remain elusive. Tumor necrosis factor α stimulated gene 6 (TSG-6) is a secretory protein with anti-inflammatory properties. Our previous research demonstrated TSG-6 can relieve intestinal inflammation and mucosal damage. However, the underlying mechanism and targets remain unclear. This research sought to explore how TSG-6 regulates the intestinal epithelial barrier and its mechanistic role in experimental colitis. METHODS IBD mouse model was generated using dextran sodium sulfate (DSS), with or without intraperitoneal injection of TSG-6(100 µg/kg or 200 µg/kg). The effects of TSG-6 on colonic inflammation and intestinal barrier function were investigated. Label-free quantitative proteomic analysis was performed on intestinal samples to explore the mechanism and therapeutic target of TSG-6. Molecular interactions were determined by co-immunoprecipitation (Co-IP) and immunofluorescence colocalization. RESULTS TSG-6 treatment significantly attenuated DSS-induced colitis symptoms and inflammatory cell infiltration. Microarray analysis revealed that TSG-6 decreased pro-inflammatory cytokine levels in colon tissue. TSG-6 restored the intestinal epithelial barrier through the promotion of intestinal epithelial cells (IECs) proliferation and mitigation of tight junctions (TJs) damage. Mechanistically, TSG-6 promoted tuft cells differentiation and increased interleukin-25 (IL-25) levels by directly binding to Pou class 2 homeobox 3(Pou2f3) and up-regulating its expression in the gut. CONCLUSIONS This study demonstrated TSG-6 as a positive regulator of tuft cells differentiation by interacting with Pou2f3, and the effectiveness of exogenous TSG-6 treatment on maintaining intestinal barrier integrity showed a promising potential for its clinical application.
Collapse
Affiliation(s)
- Shaopeng Yang
- Department of Endoscopy, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang, 310022, China
| | - Yuqi Li
- Department of Endoscopy, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang, 310022, China
| | - Rongwei Ruan
- Department of Endoscopy, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang, 310022, China
| | - Jiangping Yu
- Department of Endoscopy, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang, 310022, China
| | - Bo Zhu
- Department of Endoscopy, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang, 310022, China
| | - Haibin Lou
- Department of Endoscopy, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang, 310022, China
| | - Xiaolan Zhang
- Department of Gastroenterology, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei, 050000, China.
| | - Shi Wang
- Department of Endoscopy, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang, 310022, China.
| |
Collapse
|
35
|
Yu W, Huang P, Jin Y, Wu F, Zhang C, Jing L, Chen Y, Xu H, Xiong J, Zhang R, Zhao K, Li X. Vitamin D enhances the therapeutic effect of TNF-α antibodies through lipid metabolism in overweight IBD patients. Cell Mol Life Sci 2025; 82:176. [PMID: 40285831 PMCID: PMC12033164 DOI: 10.1007/s00018-025-05626-8] [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: 12/22/2024] [Revised: 02/03/2025] [Accepted: 02/15/2025] [Indexed: 04/29/2025]
Abstract
The inhibitory effects of the tumor necrosis factor-α (TNF-α) antibody infliximab (IFX) on colitis are well established. Since IFX dosing is weight-based and associated with various side effects, there is a growing interest in identifying combination therapies that can enhance its efficacy, particularly in overweight inflammatory bowel disease (IBD) patients, to maximize the anti-inflammatory effect while minimizing the required dose. Our research revealed that overweight IBD patients present decreased vitamin D levels in the intestinal epithelium alongside elevated TNF-α levels. In mice fed a high-fat diet (HFD) for four weeks, treatment with the vitamin D analog palicalcitol (PAL) reduced lipid synthesis and TNF-α production in intestinal epithelial cells (IECs). In a 2,4,6-trinitrobenzenesulfonic acid (TNBS)-induced experimental colitis model, PAL treatment mitigated TNF-α-induced damage to the intestinal epithelial barrier and reduced the activation of Th1 and Th17 cells in the lamina propria, thereby reducing colitis development in HFD-fed mice. Notably, the combination of IFX and PAL was more effective than IFX alone in treating colitis in these mice. Overall, our findings suggest that vitamin D inhibits TNF-α production by reducing lipid synthesis in IECs, thereby enhancing IFX therapy in overweight IBD patients.
Collapse
Affiliation(s)
- Wei Yu
- Institute of Clinical Medicine Research, Zhejiang Provincial People'S Hospital(Affiliated People'S Hospital), Hangzhou Medical College, Hangzhou, 310013, Zhejiang, China
| | - Pengpeng Huang
- Institute of Clinical Medicine Research, Zhejiang Provincial People'S Hospital(Affiliated People'S Hospital), Hangzhou Medical College, Hangzhou, 310013, Zhejiang, China
| | - Yanling Jin
- Institute of Clinical Medicine Research, Zhejiang Provincial People'S Hospital(Affiliated People'S Hospital), Hangzhou Medical College, Hangzhou, 310013, Zhejiang, China
| | - Fang Wu
- Department of General, Cancer Center, Division of Gastrointestinal and Pancreatic Surgery, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, 310013, Zhejiang, China
| | - Cuiping Zhang
- Department of Pathology, Yantai Affiliated Hospital of Binzhou Medical University, Yantai, 264100, Shandong, China
| | - Lili Jing
- Immunology and Pathology Teaching and Research Office, Shandong College of Traditional Chinese Medicine, Yantai, 264199, Shandong, China
| | - Ying Chen
- Institute of Clinical Medicine Research, Zhejiang Provincial People'S Hospital(Affiliated People'S Hospital), Hangzhou Medical College, Hangzhou, 310013, Zhejiang, China
| | - Han Xu
- Institute of Clinical Medicine Research, Zhejiang Provincial People'S Hospital(Affiliated People'S Hospital), Hangzhou Medical College, Hangzhou, 310013, Zhejiang, China
| | - Jiapin Xiong
- Institute of Clinical Medicine Research, Zhejiang Provincial People'S Hospital(Affiliated People'S Hospital), Hangzhou Medical College, Hangzhou, 310013, Zhejiang, China
| | - Rong Zhang
- Institute of Clinical Medicine Research, Zhejiang Provincial People'S Hospital(Affiliated People'S Hospital), Hangzhou Medical College, Hangzhou, 310013, Zhejiang, China
| | - Ke Zhao
- Institute of Clinical Medicine Research, Zhejiang Provincial People'S Hospital(Affiliated People'S Hospital), Hangzhou Medical College, Hangzhou, 310013, Zhejiang, China
| | - Xue Li
- Institute of Clinical Medicine Research, Zhejiang Provincial People'S Hospital(Affiliated People'S Hospital), Hangzhou Medical College, Hangzhou, 310013, Zhejiang, China.
- Department of Urology, Zhejiang Provincial People'S Hospital (Affiliated People'S Hospital), Hangzhou Medical College, Hangzhou, 310013, Zhejiang, China.
| |
Collapse
|
36
|
Buchynskyi M, Kamyshna I, Halabitska I, Petakh P, Kunduzova O, Oksenych V, Kamyshnyi O. Unlocking the gut-liver axis: microbial contributions to the pathogenesis of metabolic-associated fatty liver disease. Front Microbiol 2025; 16:1577724. [PMID: 40351307 PMCID: PMC12061941 DOI: 10.3389/fmicb.2025.1577724] [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: 02/16/2025] [Accepted: 04/07/2025] [Indexed: 05/14/2025] Open
Abstract
Metabolic dysfunction-associated fatty liver disease (MAFLD) is a complex metabolic disorder characterized by hepatic lipid accumulation and subsequent inflammation. This condition is closely linked to metabolic syndrome and obesity, with its prevalence rising due to sedentary lifestyles and high-calorie diets. The pathogenesis of MAFLD involves multiple factors, including insulin resistance, lipotoxicity, oxidative stress, and inflammatory responses. The gut microbiota plays a crucial role in MAFLD development, with dysbiosis contributing to liver inflammation through various mechanisms, such as enhanced intestinal permeability and the translocation of bacterial products like lipopolysaccharide (LPS). Microbial metabolites, including short-chain fatty acids (SCFAs) and bile acids, influence hepatic function and immune responses, with potential implications for disease progression. Specific gut microbiome signatures have been identified in MAFLD patients, offering potential diagnostic and therapeutic targets. Moreover, gut-derived toxins, such as endotoxins, lipopolysaccharides, trimethylamine-N-oxide and bacterial metabolites, significantly influence liver damage and inflammation, highlighting the complex interplay between the gut microbiome and hepatic health. This review comprehensively examines the complex interplay between the gut microbiota and MAFLD, focusing on underlying pathogenic mechanisms, potential biomarkers, and emerging microbiome-targeted therapeutic strategies for disease management.
Collapse
Affiliation(s)
- Mykhailo Buchynskyi
- Department of Microbiology, Virology, and Immunology, I. Horbachevsky Ternopil National Medical University, Ternopil, Ukraine
| | - Iryna Kamyshna
- Department of Medical Rehabilitation, I. Horbachevsky Ternopil National Medical University, Ternopil, Ukraine
| | - Iryna Halabitska
- Department of Therapy and Family Medicine, I. Horbachevsky Ternopil National Medical University, Ternopil, Ukraine
| | - Pavlo Petakh
- Department of Biochemistry and Pharmacology, Uzhhorod National University, Uzhhorod, Ukraine
| | - Oksana Kunduzova
- Institute of Metabolic and Cardiovascular Diseases (I2MC), National Institute of Health and Medical Research (INSERM) 1297, Toulouse III University, Toulouse, France
| | - Valentyn Oksenych
- Department of Clinical Science, University of Bergen, Bergen, Norway
| | - Oleksandr Kamyshnyi
- Department of Microbiology, Virology, and Immunology, I. Horbachevsky Ternopil National Medical University, Ternopil, Ukraine
| |
Collapse
|
37
|
Xu Y, Chen L, Hu X, Lai Z, Chen B, Wu M, Mai L, Su Z, Chen J, Lai Z, Ai W, Xie J, Liao H, Xie Y. Brusatol ameliorates intestinal mucosal injury in ulcerative colitis via activating IL-22/STAT3 pathway. Int Immunopharmacol 2025; 153:114482. [PMID: 40101416 DOI: 10.1016/j.intimp.2025.114482] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2025] [Revised: 02/23/2025] [Accepted: 03/10/2025] [Indexed: 03/20/2025]
Abstract
Brusatol (BR) is an active compounds isolated from Brucea javanica, a Chinese herbal medicine that is famous for its anti-diarrheal effect. We have previously reported that BR mitigated inflammation in murine ulcerative colitis (UC) models. However, BR's role in intestinal mucosal healing, which is recently established as central strategy for the prevention and treatment of UC, remains unknown. In this study, the ameliorative effect of BR on intestinal mucosal damage was investigated in DSS-induced UC mice. BR significantly alleviated colitis symptoms, improved intestinal barrier function by preventing loss of goblet cells and downregulation of mucins and tight junction proteins, as well as maintained proliferative and apoptotic homeostasis in the colonic epithelium of UC mice. Mechanistically, BR enhanced the level and secretion of IL-22, but inhibited IL-22BP, an inhibitory protein of IL-22, in the blood serum and intestinal tissues of UC mice, as well as in MNK3 cells which is an effective cell model for studying ILC3s. Additionally, BR elevated the expressions of receptors for IL-22 (IL-10R2 and IL-22R1), and activated its downstream STAT3 signaling pathway. Furthermore, the involvement of IL-22 was further investigated by using recombinant IL-22 (rIL-22) and IL-22 antibody (anti-IL-22). BR demonstrated comparable effects with rIL-22 on alleviating intestinal inflammation and repairing intestinal mucosal injury. Treatment with anti-IL-22 abrogated the mucosal protective effects of BR. The present findings shed novel insights into the role of BR in intestinal mucosal healing via activating IL-22/STAT3 signaling pathway in UC.
Collapse
Affiliation(s)
- Ying Xu
- Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, PR China
| | - Li Chen
- Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, PR China; Pharmacy Center, Shenzhen Nanshan Medical Group Headquarters, Shenzhen, PR China
| | - Xiaoxia Hu
- Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, PR China
| | - Zixuan Lai
- Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, PR China
| | - Baoyi Chen
- Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, PR China
| | - Minghui Wu
- Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, PR China
| | - Liting Mai
- Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, PR China
| | - Ziren Su
- Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, PR China
| | - Jiannan Chen
- Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, PR China
| | - Zhengquan Lai
- Department of Pharmacy, Shenzhen University General Hospital/Shenzhen University Clinical Medical Academy, Shenzhen University, Shenzhen, PR China
| | - Weipeng Ai
- Department of Pharmacy, Shenzhen University General Hospital/Shenzhen University Clinical Medical Academy, Shenzhen University, Shenzhen, PR China
| | - Jianhui Xie
- Guangdong Provincial Key Laboratory of Clinical Research on TCM Syndrome, Guangzhou, PR China
| | - Huijun Liao
- Pharmacy Center, Shenzhen Nanshan Medical Group Headquarters, Shenzhen, PR China; Department of Clinical Pharmacy and Pharmaceutical Services, Shenzhen Nanshan People's Hospital, Shenzhen, PR China.
| | - Youliang Xie
- Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, PR China; Guangdong Provincial Key Laboratory of Clinical Research on TCM Syndrome, Guangzhou, PR China.
| |
Collapse
|
38
|
Costa M, Pottier M, Jacob M, Zarnitzky P, Segain B, Figeac M, Sebda S, Leprêtre F, Meresse B, Demaret J, Foligné B, Standaert A, Bertin B. Relevance of mouse and human IBD patient-derived colon organoids to investigate intestinal macrophage differentiation. J Leukoc Biol 2025; 117:qiaf004. [PMID: 39832522 DOI: 10.1093/jleuko/qiaf004] [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: 07/04/2024] [Revised: 10/09/2024] [Accepted: 01/15/2025] [Indexed: 01/22/2025] Open
Abstract
The gastrointestinal tract is a remarkable example of complex biology, with a constant dialogue between the intestinal epithelium, in close contact with the microbiota, and the immune cells that protect the gut from infection. Organoids have revolutionized our approach to modeling the intestinal cellular compartment and have opened new avenues for unraveling the mechanisms involved in intestinal homeostasis and chronic pathogenesis, such as inflammatory bowel disease. To date, few models have been established to explore the role of the colon, which is, however, the main site of inflammation in ulcerative colitis. Here, we used conditioned media produced by colon organoids from mice or humans (control patients and patients with ulcerative colitis) to investigate the relationship between macrophages and the colon epithelium. We addressed transcriptomic profiles of organoid conditioned media-stimulated bone marrow-derived macrophages and found that these cells exhibited a unique anti-inflammatory signature distinct from that of conventional in vitro IL-4/IL-13 M2-differentiated macrophages. In addition, organoid conditioned media induced a clear CD5 antigen-like-mediated immunoregulatory effect characterized by a significant reduction in lipopolysaccharide-induced inducible nitric oxide synthase expression. In line, organoid conditioned media from human colons inhibited lipopolysaccharide-dependent inflammatory cytokine expression in human monocyte-derived macrophages. Interestingly, the inflammatory marker CD68 was reduced by organoid conditioned media from control patients but not from patients with ulcerative colitis, suggesting epithelial dysfunction in patients with ulcerative colitis. Our results report new regulatory mechanisms in the colon and highlight the importance of developing new in vitro models to better characterize the relationship between the intestinal epithelium and immune mucosal cells.
Collapse
Affiliation(s)
- Maxime Costa
- Univ. Lille, Inserm, CHU Lille, U1286-INFINITE-Institute for Translational Research in Inflammation, F-59000 Lille, France
| | - Muriel Pottier
- Univ. Lille, Inserm, CHU Lille, U1286-INFINITE-Institute for Translational Research in Inflammation, F-59000 Lille, France
| | - Marie Jacob
- Univ. Lille, Inserm, CHU Lille, U1286-INFINITE-Institute for Translational Research in Inflammation, F-59000 Lille, France
| | - Pauline Zarnitzky
- Univ. Lille, Inserm, CHU Lille, U1286-INFINITE-Institute for Translational Research in Inflammation, F-59000 Lille, France
| | - Benjamin Segain
- Univ. Lille, Inserm, CHU Lille, U1286-INFINITE-Institute for Translational Research in Inflammation, F-59000 Lille, France
| | - Martin Figeac
- Plateau de Génomique Fonctionnelle et Structurale, CHU Lille, Univ. Lille, Lille France
| | - Shéhérazade Sebda
- Plateau de Génomique Fonctionnelle et Structurale, CHU Lille, Univ. Lille, Lille France
| | - Frédéric Leprêtre
- Plateau de Génomique Fonctionnelle et Structurale, CHU Lille, Univ. Lille, Lille France
| | - Bertrand Meresse
- Univ. Lille, Inserm, CHU Lille, U1286-INFINITE-Institute for Translational Research in Inflammation, F-59000 Lille, France
| | - Julie Demaret
- Univ. Lille, Inserm, CHU Lille, U1286-INFINITE-Institute for Translational Research in Inflammation, F-59000 Lille, France
| | - Benoit Foligné
- Univ. Lille, Inserm, CHU Lille, U1286-INFINITE-Institute for Translational Research in Inflammation, F-59000 Lille, France
| | - Annie Standaert
- Univ. Lille, Inserm, CHU Lille, U1286-INFINITE-Institute for Translational Research in Inflammation, F-59000 Lille, France
| | - Benjamin Bertin
- Univ. Lille, Inserm, CHU Lille, U1286-INFINITE-Institute for Translational Research in Inflammation, F-59000 Lille, France
| |
Collapse
|
39
|
Lan X, Ma L, Ma J, Huang Z, Liu L, Li F, Wang M, Hu Y. Tas2r105 ameliorates gut inflammation, possibly through influencing the gut microbiota and metabolites. mSystems 2025; 10:e0155624. [PMID: 40079578 PMCID: PMC12013267 DOI: 10.1128/msystems.01556-24] [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: 11/20/2024] [Accepted: 02/24/2025] [Indexed: 03/15/2025] Open
Abstract
Inflammatory bowel disease (IBD) is an immune-mediated gastrointestinal disorder that significantly impacts the life quality of people worldwide. Genetic factors play crucial roles in the development of IBD. Tas2rs, members of the G protein-coupled receptor (GPCR) superfamily, are known for their roles in bitter taste perception. However, Tas2rs have also been identified in the gut, where they help sense luminal contents and regulate gastrointestinal hormones. Periodontal Tas2r105 has been shown to modulate innate immunity by interacting with metabolites produced by oral bacteria. In this study, we observed increased Tas2r105 in the inflammatory colons induced by dextran sulfate sodium salt (DSS). We also noted that α-gustducin, the α-subunit of GPCRs, is present in the intestine, and that α-gustducin knockout mice exhibit aggravated colitis. Based on these findings, we hypothesize that Tas2r105 may play a role in immune regulation during IBD pathogenesis. To test this hypothesis, we used Tas2r105 knockout (KO) mice in a colitis model. Our results show that the KO mice had significantly shorter colon length, more severe colon inflammation, and greater destruction of the gut barrier compared with control mice. We also observed increased recruitment of macrophages to the lamina propria mucosa in the KO mice. Microbiological analysis revealed a significant increase in Proteobacteria and Bacteroidota, with a concomitant decrease in Firmicutes after Tas2r105 knockout. Metabolomic analysis showed a significant reduction in lysophosphatidylethanolamine (LPE) levels in the KO mice, which is known to have anti-inflammatory effects. Based on these findings, we speculate that Tas2r105 may help protect the intestine from inflammation by influencing the gut microbiota composition and LPE production.IMPORTANCEIncreased Tas2r105 was detected in the inflamed colon of mice outside the tongue. Tas2r105 deletion aggravated mice colon colitis. Tas2r105 might alleviate mice colitis by downregulating the Proteobacteria and the Bacteroidota abundance in the colon. Lysophosphatidylethanolamine (LPE) might be the key metabolite that mediated the intestinal protection of Tas2r105.
Collapse
Affiliation(s)
- Xiucai Lan
- Department of Geriatrics, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, China
| | - Liang Ma
- Department of Radiology, Children's Hospital of Fudan University, National Children's Medical Center, Shanghai, China
| | - Jiaming Ma
- Department of Health-Related Product Assessment, Shanghai Municipal Center for Disease Control and Prevention, Shanghai, China
| | - Zhipeng Huang
- Departments of Gastroenterology, First Hospital of Quanzhou affiliated to Fujian Medical University, Quanzhou, China
| | - Lingling Liu
- Department of Laboratory Animal Science, Shanghai Public Health Clinical Center, Shanghai, China
| | - Feng Li
- Department of Laboratory Animal Science, Shanghai Public Health Clinical Center, Shanghai, China
| | - Mingbang Wang
- Department of Neonatology, Affiliated Shenzhen Women and Children's Hospital (Longgang) of Shantou University Medical College (Longgang District Maternity & Child Healthcare Hospital of Shenzhen City, Shenzhen, Guangdong, China
- Department of Experiment & Research, South China Hospital, Medical School, Shenzhen University, Shenzhen, Guangdong, China
| | - Yaomin Hu
- Department of Geriatrics, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, China
| |
Collapse
|
40
|
Jin S, Wang H, Gong H, Guo L, Zhang H, Zhang J, Chang Q, Li J, Zhang R, Bao J. Music intervention mitigates LPS-induced gut barrier disruption and immune stress in broilers via TLR4/NF-κB regulation. Poult Sci 2025; 104:105189. [PMID: 40294553 PMCID: PMC12059385 DOI: 10.1016/j.psj.2025.105189] [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: 11/11/2024] [Revised: 04/02/2025] [Accepted: 04/18/2025] [Indexed: 04/30/2025] Open
Abstract
Immune stress induced by harsh environment in intensive farming can impair broiler intestinal health. Although music as an environmental intervention can alleviate short-term stress injury, its long-term regulatory mechanism on intestinal inflammation has not been clarified. In this study, we investigated the effects of a music-enriched environment on growth performance, intestinal barrier function, and inflammatory responses in lipopolysaccharide (LPS)-induced immunostressed broilers. AA broilers were randomly divided into four groups: control group (CON), music-enriched environment group (MUC), LPS-induced immune stress group (LPS) and music-enriched environment + LPS group (MUC+LPS). On the 14th, 16th and 18th days, the LPS and MUC+LPS groups were injected intraperitoneally with 500 μg of LPS to construct an immune stress model, and the CON and MUC groups were injected with an equal amount of saline. On day 28, the birds were sacrificed to detect the indicators associated with intestinal barrier and inflammation. The LPS group showed a significant decrease in performance from 14 to 28 days, with elevated serum levels of CORT, ACTH, DAO, and d-LA, and a decrease in the activity of intestinal mucosal SOD/GSH-Px, and impaired gut morphology. impaired; music remission significantly alleviated the decline in production performance, reduced the levels of stress hormones and markers of intestinal barrier damage, while elevating jejuno-ileal GSH-Px activity and improving intestinal morphology. Significant inflammatory gene expression characteristics were observed in jejunum and ileum tissues after LPS injection: upregulation of TLR4, NF-κB, TNF-α, IL-1β, and IL-6, and significant suppression of jejunal IL-10 expression. Notably, IL-10 and IFN-γ expression in the ileum did not show statistical differences. Inflammation-related gene expression showed an overall down-regulation trend after the music intervention, but was still significantly different from the control group. Music intervention on the regulation of jejunal MYD88 and ileal TNF-α - the LPS group did not show statistically significant differences in the expression of these two key inflammatory nodes with the LPS+MUS group. Mechanistic studies have shown that LPS triggers an oxidative stress cascade through activation of the TLR4/NF-κB signaling axis, leading to disruption of intestinal barrier integrity. In contrast, music exposure exerts a protective effect through a dual mechanism: on the one hand, it helps to enhance the expression of the tight junction protein ZO-1/Occludin to repair the physical barrier; on the other hand, it inhibits the activation of the TLR4/NF-κB pathway, which can effectively alleviate LPS-induced immunopathological damage.
Collapse
Affiliation(s)
- Shengzi Jin
- College of Animal Science and Technology, Northeast Agricultural University, Harbin 150030, PR China
| | - Haowen Wang
- College of Animal Science and Technology, Northeast Agricultural University, Harbin 150030, PR China
| | - Haiyue Gong
- College of Animal Science and Technology, Northeast Agricultural University, Harbin 150030, PR China
| | - Lu Guo
- Department of Basic Medical Sciences, Heilongjiang Nursing College, Harbin, Heilongjiang 150086, China
| | - Haoran Zhang
- College of Animal Science and Technology, Northeast Agricultural University, Harbin 150030, PR China
| | - Jiaqi Zhang
- College of Animal Science and Technology, Northeast Agricultural University, Harbin 150030, PR China
| | - Qingqing Chang
- College of Animal Science and Technology, Northeast Agricultural University, Harbin 150030, PR China
| | - Jianhong Li
- College of Life Science, Northeast Agricultural University, Harbin 150030, PR China
| | - Runxiang Zhang
- College of Animal Science and Technology, Northeast Agricultural University, Harbin 150030, PR China.
| | - Jun Bao
- College of Animal Science and Technology, Northeast Agricultural University, Harbin 150030, PR China
| |
Collapse
|
41
|
Garg G, Trisal A, Singh AK. Unlocking the therapeutic potential of gut microbiota for preventing and treating aging-related neurological disorders. Neuroscience 2025; 572:190-203. [PMID: 40073931 DOI: 10.1016/j.neuroscience.2025.03.020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2024] [Revised: 03/03/2025] [Accepted: 03/08/2025] [Indexed: 03/14/2025]
Abstract
Billions of microorganisms inhabit the human gut and maintain overall health. Recent research has revealed the intricate interaction between the brain and gut microbiota through the microbiota-gut-brain axis (MGBA) and its effect on neurodegenerative disorders (NDDs). Alterations in the gut microbiota, known as gut dysbiosis, are linked to the development and progression of several NDDs. Studies suggest that the gut microbiota may be a viable target for improving cognitive health and reducing hallmarks of brain aging. Numerous pathways including hypothalamic-pituitary-adrenal axis stimulation, neurotransmitter release disruption, system-wide inflammation, and increased intestinal and blood-brain barrier permeability connect gut dysbiosis to neurological conditions. Metabolites produced by the gut microbiota influence neural processes that affect brain function. Clinical interventions depend on the capacity to understand the equilibrium between beneficial and detrimental gut microbiota, as it affects both neurodegeneration and neuroprotection. The importance of the gut microbiota and its metabolites during brain aging and the development of neurological disorders is summarized in this review. Moreover, we explored the possible therapeutic effects of the gut microbiota on age-related NDDs. Highlighting various pathways that connect the gut and the brain, this review identifies several important domains where gut microbiota-based interventions could offer possible solutions for age-related NDDs. Furthermore, prebiotics and probiotics are discussed as effective alternatives for mitigating indirect causes of gut dysbiosis. These therapeutic interventions are poised to play a significant role in improving dysbiosis and NDDs, paving the way for further research.
Collapse
Affiliation(s)
- Geetika Garg
- Department of Zoology, Savitribai Phule Pune University, Pune 411007, India
| | - Anchal Trisal
- Department of Biosciences, Jamia Millia Islamia, New Delhi 110025, India
| | - Abhishek Kumar Singh
- Manipal Centre for Biotherapeutics Research, Manipal Academy of Higher Education, Karnataka, Manipal 576 104, India.
| |
Collapse
|
42
|
Dong Y, Zhang L, Qiu D, Yao R, Jia H, Wang H, Zhou L, Zhang J, Zhang N. Lactobacillus murinus ZNL-13 Modulates Intestinal Barrier Damage and Gut Microbiota in Cyclophosphamide-Induced Immunosuppressed Mice. Foods 2025; 14:1416. [PMID: 40282817 PMCID: PMC12026897 DOI: 10.3390/foods14081416] [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: 03/24/2025] [Revised: 04/14/2025] [Accepted: 04/17/2025] [Indexed: 04/29/2025] Open
Abstract
Cyclophosphamide (CTX) is a widely used anticancer drug in clinical practice; however, its administration can lead to gastrointestinal damage and immune suppression. Lactobacillus murinus (L. murinus) has been shown to regulate immune cell activity and protect the gastrointestinal system, showing potential application as a functional food. The objective of this study was to investigate the effects of L. murinus ZNL-13 on CTX-induced intestinal mucosal injury and gut microbiota in mice. The results demonstrated that L. murinus ZNL-13 significantly alleviated weight loss and intestinal pathological damage. Moreover, in CTX-induced intestinal injury mice, L. murinus ZNL-13 enhanced the release of immune factors, suppressed cell apoptosis, and protected the intestinal mucosal barrier. Additionally, it activated the TLR4/NF-κB pathway, thereby promoting immune cell activity. Furthermore, L. murinus ZNL-13 contributed to the restoration of gut microbial homeostasis by increasing the relative abundance of short-chain fatty acid-producing bacteria. Taken together, this investigation highlights the potential of L. murinus ZNL-13 in protecting the intestinal barrier and enhancing immune function while laying the groundwork for its development as a novel probiotic and functional food.
Collapse
Affiliation(s)
- Yihan Dong
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, China; (Y.D.); (L.Z.); (D.Q.); (R.Y.); (H.J.); (H.W.); (L.Z.)
- Heilongjiang Provincial Key Laboratory of Pathogenic Mechanism for Animal Disease and Comparative Medicine, Harbin 150030, China
| | - Luyao Zhang
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, China; (Y.D.); (L.Z.); (D.Q.); (R.Y.); (H.J.); (H.W.); (L.Z.)
| | - Di Qiu
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, China; (Y.D.); (L.Z.); (D.Q.); (R.Y.); (H.J.); (H.W.); (L.Z.)
| | - Renxin Yao
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, China; (Y.D.); (L.Z.); (D.Q.); (R.Y.); (H.J.); (H.W.); (L.Z.)
| | - Haitao Jia
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, China; (Y.D.); (L.Z.); (D.Q.); (R.Y.); (H.J.); (H.W.); (L.Z.)
| | - Haiyang Wang
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, China; (Y.D.); (L.Z.); (D.Q.); (R.Y.); (H.J.); (H.W.); (L.Z.)
| | - Luyao Zhou
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, China; (Y.D.); (L.Z.); (D.Q.); (R.Y.); (H.J.); (H.W.); (L.Z.)
| | - Jiantao Zhang
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, China; (Y.D.); (L.Z.); (D.Q.); (R.Y.); (H.J.); (H.W.); (L.Z.)
- Heilongjiang Provincial Key Laboratory of Pathogenic Mechanism for Animal Disease and Comparative Medicine, Harbin 150030, China
| | - Na Zhang
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, China; (Y.D.); (L.Z.); (D.Q.); (R.Y.); (H.J.); (H.W.); (L.Z.)
- Key Laboratory of Dairy Science, Ministry of Education, Northeast Agricultural University, Harbin 150030, China
| |
Collapse
|
43
|
Fang D, Zhao H, Pei L, Jiang K, Gan Y, Zhai X, Zhang L, Cheng Y, Liu C, Du J, Gao F. Diprovocim protects against the radiation-induced damage via the TLR2 signaling pathway. Mol Med 2025; 31:139. [PMID: 40247162 PMCID: PMC12004591 DOI: 10.1186/s10020-025-01198-2] [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: 09/03/2024] [Accepted: 04/03/2025] [Indexed: 04/19/2025] Open
Abstract
Severe ionizing radiation (IR) causes the acute lethal damage of hematopoietic system and gastrointestinal tract. By establishing a radiation injury model, we found that Diprovocim, a TLR2 agonist, protected mice against the lethal damage of hematopoietic system and gastrointestinal tract. Diprovocim inhibited the IR-induced damage, promoted erythrocyte differentiation and elevated the proportion of hematopoietic stem cells (HSCs) in irradiated mice, and promoted the proliferation and differentiation of intestinal stem cells (ISCs). In addition, the RNA seq results suggested that Diprovocim significantly upregulated the TLR2 signaling pathway, and Diprovocim had no radioprotective effect on TLR2 KO mice, suggesting that Diprovocim activated TLR2 signaling pathway to exert its radioprotective function. The RNA sequencing results also suggested that Diprovocim significantly up-regulated the expression of SOX9. Diprovocim had no radioprotective effect after SOX9 knockdown. In conclusion, we demonstrated that Diprovocim protected the radiation-induced damage and upregulated targeting TLR2-SOX9 axis and that Diprovocim might be a potential high-efficiency selective agent.
Collapse
Affiliation(s)
- Duo Fang
- School of Health Science and Engineering, University of Shanghai for Science and Technology, Shanghai, 200093, People's Republic of China
- Department of Radiation Medicine, Faculty of Naval Medicine, Naval Medical University, 800 Xiangyin Road, Shanghai, 200433, People's Republic of China
| | - Hainan Zhao
- Department of Radiology Intervention, Changhai Hospital Affiliated to the Naval Medical University, Shanghai, China
| | - Lu Pei
- Department of Radiation Medicine, Faculty of Naval Medicine, Naval Medical University, 800 Xiangyin Road, Shanghai, 200433, People's Republic of China
- Inner Mongolia Key Laboratory of Hypoxic Translational Medicine, Baotou Medical College, Baotou, 014040, China
| | - Kai Jiang
- Department of Radiation Medicine, Faculty of Naval Medicine, Naval Medical University, 800 Xiangyin Road, Shanghai, 200433, People's Republic of China
| | - Yuhan Gan
- Department of Radiation Medicine, Faculty of Naval Medicine, Naval Medical University, 800 Xiangyin Road, Shanghai, 200433, People's Republic of China
| | - Xuanlu Zhai
- Department of Radiation Medicine, Faculty of Naval Medicine, Naval Medical University, 800 Xiangyin Road, Shanghai, 200433, People's Republic of China
| | - Liao Zhang
- Department of Radiation Medicine, Faculty of Naval Medicine, Naval Medical University, 800 Xiangyin Road, Shanghai, 200433, People's Republic of China
| | - Ying Cheng
- Department of Radiation Medicine, Faculty of Naval Medicine, Naval Medical University, 800 Xiangyin Road, Shanghai, 200433, People's Republic of China
| | - Cong Liu
- Department of Radiation Medicine, Faculty of Naval Medicine, Naval Medical University, 800 Xiangyin Road, Shanghai, 200433, People's Republic of China
| | - Jicong Du
- Department of Radiation Medicine, Faculty of Naval Medicine, Naval Medical University, 800 Xiangyin Road, Shanghai, 200433, People's Republic of China.
| | - Fu Gao
- School of Health Science and Engineering, University of Shanghai for Science and Technology, Shanghai, 200093, People's Republic of China.
- Department of Radiation Medicine, Faculty of Naval Medicine, Naval Medical University, 800 Xiangyin Road, Shanghai, 200433, People's Republic of China.
| |
Collapse
|
44
|
Zhang C, Zhen Y, Weng Y, Lin J, Xu X, Ma J, Zhong Y, Wang M. Research progress on the microbial metabolism and transport of polyamines and their roles in animal gut homeostasis. J Anim Sci Biotechnol 2025; 16:57. [PMID: 40234982 PMCID: PMC11998418 DOI: 10.1186/s40104-025-01193-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2024] [Accepted: 03/09/2025] [Indexed: 04/17/2025] Open
Abstract
Polyamines (putrescine, spermidine, and spermine) are aliphatic compounds ubiquitous in prokaryotes and eukaryotes. Positively charged polyamines bind to negatively charged macromolecules, such as nucleic acids and acidic phospholipids, and are involved in physiological activities including cell proliferation, differentiation, apoptosis and gene regulation. Intracellular polyamine levels are regulated by biosynthesis, catabolism and transport. Polyamines in the body originate from two primary sources: dietary intake and intestinal microbial metabolism. These polyamines are then transported into the bloodstream, through which they are distributed to various tissues and organs to exert their biological functions. Polyamines synthesized by intestinal microorganisms serve dual critical roles. First, they are essential for maintaining polyamine concentrations within the digestive tract. Second, through transcriptional and post-transcriptional mechanisms, these microbial-derived polyamines modulate the expression of genes governing key processes in intestinal epithelial cells-including proliferation, migration, apoptosis, and cell-cell interactions. Collectively, these regulatory effects help maintain intestinal epithelial homeostasis and ensure the integrity of the gut barrier. In addition, polyamines interact with the gut microbiota to maintain intestinal homeostasis by promoting microbial growth, biofilm formation, swarming, and endocytosis vesicle production, etc. Supplementation with polyamines has been demonstrated to be important in regulating host intestinal microbial composition, enhancing nutrient absorption, and improving metabolism and immunity. In this review, we will focus on recent advances in the study of polyamine metabolism and transport in intestinal microbes and intestinal epithelial cells. We then summarize the scientific understanding of their roles in intestinal homeostasis, exploring the advances in cellular and molecular mechanisms of polyamines and their potential clinical applications, and providing a rationale for polyamine metabolism as an important target for the treatment of intestinal-based diseases.
Collapse
Affiliation(s)
- Chong Zhang
- Laboratory of Metabolic Manipulation of Herbivorous Animal Nutrition, College of Animal Science and Technology, Yangzhou University, Yangzhou, 225009, China
| | - Yongkang Zhen
- Laboratory of Metabolic Manipulation of Herbivorous Animal Nutrition, College of Animal Science and Technology, Yangzhou University, Yangzhou, 225009, China
| | - Yunan Weng
- Laboratory of Metabolic Manipulation of Herbivorous Animal Nutrition, College of Animal Science and Technology, Yangzhou University, Yangzhou, 225009, China
| | - Jiaqi Lin
- Laboratory of Metabolic Manipulation of Herbivorous Animal Nutrition, College of Animal Science and Technology, Yangzhou University, Yangzhou, 225009, China
| | - Xinru Xu
- Laboratory of Metabolic Manipulation of Herbivorous Animal Nutrition, College of Animal Science and Technology, Yangzhou University, Yangzhou, 225009, China
| | - Jianjun Ma
- Laboratory of Metabolic Manipulation of Herbivorous Animal Nutrition, College of Animal Science and Technology, Yangzhou University, Yangzhou, 225009, China
| | - Yuhong Zhong
- Laboratory of Metabolic Manipulation of Herbivorous Animal Nutrition, College of Animal Science and Technology, Yangzhou University, Yangzhou, 225009, China
| | - Mengzhi Wang
- Laboratory of Metabolic Manipulation of Herbivorous Animal Nutrition, College of Animal Science and Technology, Yangzhou University, Yangzhou, 225009, China.
- State Key Laboratory of Sheep Genetic Improvement and Healthy Production, Xinjiang Academy of Agricultural Reclamation Sciences, Shihezi, 832000, China.
| |
Collapse
|
45
|
Li J, Chen Y, Yu Q, Li S, Zhang X, Cheng Y, Fu X, Li J, Zhu L. Estrogen receptor β alleviates colitis in intestinal epithelial cells and activates HIF-1a and ATG-9a-mediated autophagy. Exp Cell Res 2025; 447:114520. [PMID: 40107441 DOI: 10.1016/j.yexcr.2025.114520] [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: 10/22/2024] [Revised: 03/10/2025] [Accepted: 03/15/2025] [Indexed: 03/22/2025]
Abstract
Estrogen receptor β (ERβ) plays a pivotal role in regulating intestinal epithelial function and inflammation. Its involvement in inflammatory bowel diseases (IBD), particularly in ulcerative colitis (UC), remains poorly understood, despite emerging evidence pointing to its anti-inflammatory properties. This study investigated ERβ expression in UC patients using quantitative PCR, Western blot, and immunofluorescence. To investigate the functional role of ERβ, a DSS-induced colitis mouse model and LPS-treated HT-29 cells were used. Autophagy activity was evaluated through Western blot, transmission electron microscopy (TEM), and autophagy inhibitors. Co-immunoprecipitation (Co-IP) and dual luciferase reporter assays were employed to explore the interaction between ERβ and hypoxia-inducible factor-1α (HIF-1α), as well as the regulation of ATG-9a expression. The results demonstrated that ERβ expression was significantly downregulated in the inflammatory colons of UC patients. In vivo, ERβ activation by ERB041 alleviated DSS-induced colitis in mice, reducing weight loss, histopathological damage, and inflammatory cytokine levels. In vitro, ERB041 enhanced autophagy in LPS-treated HT-29 cells, accompanied by a reduction in pro-inflammatory cytokines. Furthermore, ERβ activation promoted the expression of tight junction proteins and preserved epithelial barrier integrity. Co-IP and dual luciferase assays revealed that ERβ interacted with HIF-1α and modulated ATG-9a-mediated autophagy. These results indicate that ERβ alleviates intestinal inflammation and activates HIF-1a and ATG-9a-mediated autophagy, providing new insights into the therapeutic potential of targeting ERβ in UC and highlighting its role in maintaining intestinal homeostasis.
Collapse
Affiliation(s)
- Junrong Li
- Division of Gastroenterology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; Division of Gastroenterology, Chongqing Hospital Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Chongqing, China
| | - Yidong Chen
- Division of Gastroenterology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Qi Yu
- Division of Gastroenterology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Shuang Li
- Division of Gastroenterology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xiaopeng Zhang
- Division of Gastroenterology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yiyu Cheng
- Division of Gastroenterology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xiaoyu Fu
- Division of Gastroenterology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jiamin Li
- Division of Gastroenterology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Liangru Zhu
- Division of Gastroenterology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| |
Collapse
|
46
|
Jia R, Han Y, Zhu Q, Zhang J, Zhang H, Ka M, Ma Y, Gamah M, Zhang W. Activation of notch signaling pathway is a potential mechanism for mucin2 reduction and intestinal mucosal barrier dysfunction in high-altitude hypoxia. Sci Rep 2025; 15:12154. [PMID: 40204779 PMCID: PMC11982276 DOI: 10.1038/s41598-025-96176-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2024] [Accepted: 03/26/2025] [Indexed: 04/11/2025] Open
Abstract
High-altitude hypoxia can cause gastrointestinal issues and damage the intestinal mucosal barrier, which is crucial for digestion and nutrient absorption. The Notch signaling pathway affects this barrier's integrity. This study explores the Notch pathway's role in hypoxia-induced intestinal injury. C57BL/6 mice were used to model intestinal mucosal barrier injury through dextran sodium sulfate (DSS) and hypobaric hypoxia (simulating 5000 m altitude for 7 days). Mice were treated with Notch inhibitor Dibenzazepine (DBZ) and Mucin2 (MUC2) activator Prostaglandin E2 (PGE2). We evaluated weight, colon length, histology, Zonula occludens 1 (ZO-1) and Claudin-1 levels, MUC2 and Notch1 staining, serum diamine oxidase (DAO) and D-lactate (D-La), inflammatory markers, and Notch pathway proteins. DSS and hypoxia caused weight loss, colon shortening, ulcers, and inflammation, with fewer goblet cells and lower MUC2 levels. Elevated serum DAO, D-La, and inflammatory markers indicated severe intestinal damage. DBZ treatment post-DSS and hypoxia significantly reduced these symptoms. PGE2 activation of MUC2 also alleviated symptoms and mitigated intestinal damage. Hypoxia worsens DSS-induced mucosal barrier disruption by activating the Notch pathway, shifting stem cell differentiation towards absorptive cells instead of goblet cells, reducing MUC2 secretion, and intensifying damage. Targeting the Notch pathway and enhancing MUC2 expression could effectively treat hypoxia-induced intestinal injury.
Collapse
Affiliation(s)
- Ruhan Jia
- Research Center for High Altitude Medicine, Key Laboratory of High Altitude Medicine (Ministry of Education), Key Laboratory of Application and Foundation for High Altitude Medicine Research in Qinghai Province (Qinghai-Utah Joint Research Key Lab for High Altitude Medicine), Qinghai University, Xining, China
| | - Ying Han
- Research Center for High Altitude Medicine, Key Laboratory of High Altitude Medicine (Ministry of Education), Key Laboratory of Application and Foundation for High Altitude Medicine Research in Qinghai Province (Qinghai-Utah Joint Research Key Lab for High Altitude Medicine), Qinghai University, Xining, China
| | - Qinfang Zhu
- Research Center for High Altitude Medicine, Key Laboratory of High Altitude Medicine (Ministry of Education), Key Laboratory of Application and Foundation for High Altitude Medicine Research in Qinghai Province (Qinghai-Utah Joint Research Key Lab for High Altitude Medicine), Qinghai University, Xining, China
- Qinghai Provincial People's Hospital, Xining, Qinghai, China
| | - Jingxuan Zhang
- Research Center for High Altitude Medicine, Key Laboratory of High Altitude Medicine (Ministry of Education), Key Laboratory of Application and Foundation for High Altitude Medicine Research in Qinghai Province (Qinghai-Utah Joint Research Key Lab for High Altitude Medicine), Qinghai University, Xining, China
| | - Huan Zhang
- Research Center for High Altitude Medicine, Key Laboratory of High Altitude Medicine (Ministry of Education), Key Laboratory of Application and Foundation for High Altitude Medicine Research in Qinghai Province (Qinghai-Utah Joint Research Key Lab for High Altitude Medicine), Qinghai University, Xining, China
- Department of Pathology, The Second Affiliated Hospital of Xi'an Jiaotong University, 710004, Xi'an, China
| | - Maojia Ka
- Research Center for High Altitude Medicine, Key Laboratory of High Altitude Medicine (Ministry of Education), Key Laboratory of Application and Foundation for High Altitude Medicine Research in Qinghai Province (Qinghai-Utah Joint Research Key Lab for High Altitude Medicine), Qinghai University, Xining, China
| | - Yi Ma
- Research Center for High Altitude Medicine, Key Laboratory of High Altitude Medicine (Ministry of Education), Key Laboratory of Application and Foundation for High Altitude Medicine Research in Qinghai Province (Qinghai-Utah Joint Research Key Lab for High Altitude Medicine), Qinghai University, Xining, China
| | - Mohammed Gamah
- Research Center for High Altitude Medicine, Key Laboratory of High Altitude Medicine (Ministry of Education), Key Laboratory of Application and Foundation for High Altitude Medicine Research in Qinghai Province (Qinghai-Utah Joint Research Key Lab for High Altitude Medicine), Qinghai University, Xining, China
| | - Wei Zhang
- Research Center for High Altitude Medicine, Key Laboratory of High Altitude Medicine (Ministry of Education), Key Laboratory of Application and Foundation for High Altitude Medicine Research in Qinghai Province (Qinghai-Utah Joint Research Key Lab for High Altitude Medicine), Qinghai University, Xining, China.
| |
Collapse
|
47
|
Li ZY, Luo HY, Xu F, Xu Y, Ma CH, Zhang SL, Xu S, Ma YY, Li N, Miao CY. Metrnl protects intestinal barrier function by regulating tight junctions via the IKKβ/IκBα/NFκB/MLCK/MLC signaling pathway. Cell Death Discov 2025; 11:155. [PMID: 40199887 PMCID: PMC11979045 DOI: 10.1038/s41420-025-02457-1] [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: 06/06/2024] [Revised: 03/12/2025] [Accepted: 03/27/2025] [Indexed: 04/10/2025] Open
Abstract
Meteorin-like (Metrnl), also known as Subfatin, IL-41, or Cometin, is a secreted protein predominantly expressed in the intestinal epithelium. The intestinal barrier, primarily consisting of epithelial cells connected by tight junctions, is essential for maintaining gut homeostasis by preventing harmful substances from entering the body. Despite Metrnl's high expression in the intestine, its role in barrier function remains unclear. In this study, we investigated Metrnl's role in intestinal barrier function using both loss-of-function (using global and intestinal epithelium-specific knockout mice) and gain-of-function (using intestinal epithelium-specific overexpression mice) approaches. Our findings showed that Metrnl deficiency disrupted tight junctions between enterocytes and exacerbated endotoxin-induced barrier dysfunction. Mechanistically, Metrnl deficiency triggered activation of the IKKβ/IκBα/NFκB signaling pathway, leading to increased MLCK expression and MLC phosphorylation. The NFκB inhibitor PDTC reversed this effect both in vivo and in vitro. Macrophages played an essential role in Metrnl's intestinal barrier protective effects during endotoxemia, but were not necessary in burn-induced barrier injury, suggesting potential differences in mechanism between these conditions. Notably, recombinant Metrnl protein administration protected against barrier dysfunction, and genetic overexpression of Metrnl in enterocytes preserved barrier function and alleviated DSS-induced colitis. These findings establish Metrnl as a key regulator of intestinal barrier integrity through the IKKβ/IκBα/NFκB/MLCK/MLC pathway, highlighting its potential therapeutic value in treating barrier dysfunction disorders. Intestinal barrier dysfunction triggers, such as endotoxin and severe burns, may induce the release of Metrnl from vascular endothelium. This leads to an increase in circulating Metrnl. Both circulating Metrnl and local Metrnl inhibit inflammation and the IKKβ/IκBα/NFκB/MLCK/MLC signaling pathway in enterocytes, thereby protecting tight junctions from disruption caused by endotoxin or burns.
Collapse
Affiliation(s)
- Zhi-Yong Li
- Department of Pharmacology, Second Military Medical University/Naval Medical University, Shanghai, 200433, China.
- Department of Pathology, Faculty of Medical Imaging, Second Military Medical University/Naval Medical University, Shanghai, 200433, China.
| | - Heng-Yu Luo
- Department of Pharmacology, Second Military Medical University/Naval Medical University, Shanghai, 200433, China
| | - Fei Xu
- Department of Pharmacology, Second Military Medical University/Naval Medical University, Shanghai, 200433, China
| | - Yao Xu
- Department of Pharmacology, Second Military Medical University/Naval Medical University, Shanghai, 200433, China
| | - Chun-Hui Ma
- Department of Pathology, Faculty of Medical Imaging, Second Military Medical University/Naval Medical University, Shanghai, 200433, China
| | - Sai-Long Zhang
- Department of Pharmacology, Second Military Medical University/Naval Medical University, Shanghai, 200433, China
| | - Sheng Xu
- Department of Immunology, Second Military Medical University/Naval Medical University, Shanghai, 200433, China
| | - Yuan-Yuan Ma
- Senior Department of Hematology, The Fifth Medical Center of People's Liberation Army(PLA), General Hospital, Beijing, 100010, China
| | - Nan Li
- Department of Immunology, Second Military Medical University/Naval Medical University, Shanghai, 200433, China
| | - Chao-Yu Miao
- Department of Pharmacology, Second Military Medical University/Naval Medical University, Shanghai, 200433, China.
| |
Collapse
|
48
|
Wang T, Li L, Liu L, Tan R, Wu Q, Zhu X, Hua H, Dai Y, Li H, Mao J, Zhao J, Yin Z. Overview of pharmacodynamical research of traditional Chinese medicine on hyperuricemic nephropathy: from the perspective of dual-regulatory effect on the intestines and kidneys. Front Pharmacol 2025; 16:1517047. [PMID: 40264662 PMCID: PMC12011833 DOI: 10.3389/fphar.2025.1517047] [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: 10/25/2024] [Accepted: 03/28/2025] [Indexed: 04/24/2025] Open
Abstract
Uncontrolled hyperuricemia contributes to chronic kidney disease, characterized by renal inflammatory cell infiltration and tubulointerstitial fibrosis, eventually leading to renal failure. In addition to liver and kidney, the intestine tract plays a vital role in the development and progression of hyperuricemia and hyperuricemic nephropathy (HN) through various mechanisms. The conventional therapeutic strategy for HN is uric acid-lowering therapy (ULT) and renal protection; however, unsatisfactory results are often obtained in clinical practice. Growing evidence has demonstrated that traditional Chinese medicines (TCMs) achieve an anti-HN effect by modulating multiple targets and approaches with fewer side effects. Therefore, this paper reviews the pathogenesis of HN, including the role of soluble and insoluble urates in kidney and intestine, and the role of intestinal tract in the progression of HN. Meanwhile, the recent advancements in TCMs for the treatment of HN are summarized and analyzed, with a focus on their modulation of intestinal flora and metabolites, urate-related transporters, immuno-inflammation and barrier function in the intestines. Notably, for the first time, we propose the perspective that TCMs treat HN through a dual-regulatory effect on the intestines and kidneys. Additionally, the problems existing in current research and the feasible research strategies combined with emerging technologies such as fermentation and nanotechnology are discussed, thus providing novel ideas for HN management.
Collapse
Affiliation(s)
- Ting Wang
- Country School of Pharmacy, Southwest Medical University, Luzhou, China
- Sichuan Academy of Chinese Medicine Sciences, Institute of Pharmacology & Toxicology of Chinese Materia Medica, Translational Chinese Medicine Key Laboratory of Sichuan Province, Sichuan Engineering Technology Research Center of Genuine Regional Drug, Engineering Research Center for Formation Principle and Quality Evaluation of Genuine Medicinal Materials in Sichuan Province, Chengdu, China
| | - Li Li
- Sichuan Academy of Chinese Medicine Sciences, Institute of Pharmacology & Toxicology of Chinese Materia Medica, Translational Chinese Medicine Key Laboratory of Sichuan Province, Sichuan Engineering Technology Research Center of Genuine Regional Drug, Engineering Research Center for Formation Principle and Quality Evaluation of Genuine Medicinal Materials in Sichuan Province, Chengdu, China
| | - Li Liu
- Sichuan Institute for Translational Chinese Medicine, Translational Chinese Medicine Key Laboratory of Sichuan Province, Chengdu, China
| | - Ruirong Tan
- Sichuan Academy of Chinese Medicine Sciences, Institute of Pharmacology & Toxicology of Chinese Materia Medica, Translational Chinese Medicine Key Laboratory of Sichuan Province, Sichuan Engineering Technology Research Center of Genuine Regional Drug, Engineering Research Center for Formation Principle and Quality Evaluation of Genuine Medicinal Materials in Sichuan Province, Chengdu, China
| | - Qinxuan Wu
- Changsha Medical University, Hunan Provincial Key Laboratory of the Research and Development of Novel Pharmaceutical Preparations, The “Double-First Class” Application Characteristic Discipline of Hunan Province (Pharmaceutical Science), Changsha, China
| | - Xin Zhu
- Sichuan Academy of Chinese Medicine Sciences, Institute of Pharmacology & Toxicology of Chinese Materia Medica, Translational Chinese Medicine Key Laboratory of Sichuan Province, Sichuan Engineering Technology Research Center of Genuine Regional Drug, Engineering Research Center for Formation Principle and Quality Evaluation of Genuine Medicinal Materials in Sichuan Province, Chengdu, China
| | - Hua Hua
- Sichuan Institute for Translational Chinese Medicine, Translational Chinese Medicine Key Laboratory of Sichuan Province, Chengdu, China
| | - Ying Dai
- Sichuan Academy of Chinese Medicine Sciences, Institute of Pharmacology & Toxicology of Chinese Materia Medica, Translational Chinese Medicine Key Laboratory of Sichuan Province, Sichuan Engineering Technology Research Center of Genuine Regional Drug, Engineering Research Center for Formation Principle and Quality Evaluation of Genuine Medicinal Materials in Sichuan Province, Chengdu, China
| | - Huan Li
- Sichuan Acupuncture and Moxibustion School, Chengdu, China
| | - Jiuzhou Mao
- Sichuan Academy of Chinese Medicine Sciences, Institute of Pharmacology & Toxicology of Chinese Materia Medica, Translational Chinese Medicine Key Laboratory of Sichuan Province, Sichuan Engineering Technology Research Center of Genuine Regional Drug, Engineering Research Center for Formation Principle and Quality Evaluation of Genuine Medicinal Materials in Sichuan Province, Chengdu, China
| | - Junning Zhao
- Country School of Pharmacy, Southwest Medical University, Luzhou, China
- Sichuan Institute for Translational Chinese Medicine, Translational Chinese Medicine Key Laboratory of Sichuan Province, Chengdu, China
| | - Zhujun Yin
- Sichuan Academy of Chinese Medicine Sciences, Institute of Pharmacology & Toxicology of Chinese Materia Medica, Translational Chinese Medicine Key Laboratory of Sichuan Province, Sichuan Engineering Technology Research Center of Genuine Regional Drug, Engineering Research Center for Formation Principle and Quality Evaluation of Genuine Medicinal Materials in Sichuan Province, Chengdu, China
- Changsha Medical University, Hunan Provincial Key Laboratory of the Research and Development of Novel Pharmaceutical Preparations, The “Double-First Class” Application Characteristic Discipline of Hunan Province (Pharmaceutical Science), Changsha, China
| |
Collapse
|
49
|
Lin Q, Zhang S, Zhang J, Jin Y, Chen T, Lin R, Lv J, Xu W, Wu T, Tian S, Ying L, Li X, Huang Z, Niu J. Colonic epithelial-derived FGF1 drives intestinal stem cell commitment toward goblet cells to suppress inflammatory bowel disease. Nat Commun 2025; 16:3264. [PMID: 40188210 PMCID: PMC11972292 DOI: 10.1038/s41467-025-58644-2] [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: 10/11/2024] [Accepted: 03/31/2025] [Indexed: 04/07/2025] Open
Abstract
Understanding the molecular mechanisms that regulate intestinal epithelial cell (IEC) renewal provides potential targets for inflammatory bowel disease (IBD). Growing evidence has highlighted the importance of epithelial signals in regulating intestinal stem cell (ISC) differentiation. However, it remains unclear which IEC-derived cytokines can precisely regulate ISC commitment toward specific mature cells. Here we systematically analyze all fibroblast growth factors (FGFs) expression and find that colonic FGF1 levels are inversely correlated with the severity of IBD in mouse models and patients. IEC-specific Fgf1 deletion leads to impaired goblet cell differentiation and exacerbated colitis, while pharmacological administration of recombinant FGF1 (rFGF1) alleviates colitis by enhancing goblet cell differentiation and improving colonic epithelial integrity. Mechanistic studies reveal that rFGF1 directs ISC differentiation toward goblet cells via FGFR2-TCF4-ATOH1 signaling axis. In conclusion, our study identifies an epithelial niche-derived FGF1 that regulates ISC commitment toward goblet cells, shedding light on strategies for treating IBD.
Collapse
Affiliation(s)
- Qian Lin
- State Key Laboratory of Macromolecular Drugs and Large-scale Preparation, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, 325035, China
| | - Sudan Zhang
- State Key Laboratory of Macromolecular Drugs and Large-scale Preparation, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, 325035, China
| | - Jiaren Zhang
- State Key Laboratory of Macromolecular Drugs and Large-scale Preparation, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, 325035, China
| | - Yi Jin
- Department of Pathology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325035, Zhejiang, China
| | - Taoli Chen
- Department of Pharmacy, The Second Affiliated Hospital of Jiaxing University, Jiaxing, 314000, Zhejiang, China
| | - Ruoyu Lin
- State Key Laboratory of Macromolecular Drugs and Large-scale Preparation, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, 325035, China
| | - Jiaxuan Lv
- State Key Laboratory of Macromolecular Drugs and Large-scale Preparation, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, 325035, China
| | - Wenjing Xu
- State Key Laboratory of Macromolecular Drugs and Large-scale Preparation, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, 325035, China
| | - Tianzhen Wu
- State Key Laboratory of Macromolecular Drugs and Large-scale Preparation, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, 325035, China
| | - Shenyu Tian
- State Key Laboratory of Macromolecular Drugs and Large-scale Preparation, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, 325035, China
| | - Lei Ying
- School of Basic Medical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, 325035, China
| | - Xiaokun Li
- State Key Laboratory of Macromolecular Drugs and Large-scale Preparation, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, 325035, China
| | - Zhifeng Huang
- State Key Laboratory of Macromolecular Drugs and Large-scale Preparation, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, 325035, China.
- Translational Medicine Laboratory, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, 325035, China.
| | - Jianlou Niu
- State Key Laboratory of Macromolecular Drugs and Large-scale Preparation, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, 325035, China.
| |
Collapse
|
50
|
Xing J, Niu T, Yu T, Zou B, Shi C, Wang Y, Fan S, Li M, Bao M, Sun Y, Gao K, Qiu J, Zhang D, Wang N, Jiang Y, Huang H, Cao X, Zeng Y, Wang J, Zhang S, Hu J, Zhang D, Sun W, Yang G, Yang W, Wang C. Faecalibacterium prausnitzii-derived outer membrane vesicles reprogram gut microbiota metabolism to alleviate Porcine Epidemic Diarrhea Virus infection. MICROBIOME 2025; 13:90. [PMID: 40176190 PMCID: PMC11963522 DOI: 10.1186/s40168-025-02078-x] [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] [Subscribe] [Scholar Register] [Received: 10/28/2024] [Accepted: 03/04/2025] [Indexed: 04/04/2025]
Abstract
BACKGROUND The Porcine Epidemic Diarrhea Virus (PEDV) is one of the major challenges facing the global pig farming industry, and vaccines and treatments have proven difficult in controlling its spread. Faecalibacterium prausnitzii (F.prausnitzii), a key commensal bacterium in the gut, has been recognized as a promising candidate for next-generation probiotics due to its potential wide-ranging health benefits. A decrease in F.prausnitzii abundance has been associated with certain viral infections, suggesting its potential application in preventing intestinal viral infections. In this study, we utilized a piglet model to examine the potential role of F.prausnitzii in PEDV infections. RESULTS A piglet model of PEDV infection was established and supplemented with F.prausnitzii, revealing that F.prausnitzii mitigated PEDV infection. Further studies found that outer membrane vesicles (OMVs) are the main functional components of F.prausnitzii, and proteomics, untargeted metabolomics, and small RNA-seq were used to analyze the composition of OMVs. Exhaustion of the gut microbiota demonstrated that the function of Fp. OMVs relies on the presence of the gut microbiota. Additionally, metagenomic analysis indicated that Fp. OMVs altered the gut microbiota composition, enhancing the abundance of Faecalibacterium prausnitzii, Prevotellamassilia timonensis, and Limosilactobacillus reuteri. Untargeted metabolomics analysis showed that Fp. OMVs increased phosphatidylcholine (PC) levels, with PC identified as a key metabolite in alleviating PEDV infection. Single-cell sequencing revealed that PC altered the relative abundance of intestinal cells, increased the number of intestinal epithelial cells, and reduced necroptosis in target cells. PC treatment in infected IPEC-J2 and Vero cells alleviated necroptosis and reduced the activation of the RIPK1-RIPK3-MLKL signaling axis, thereby improving PEDV infection. CONCLUSION F.prausnitzii and its OMVs play a critical role in mitigating PEDV infections. These findings provide a promising strategy to ameliorate PEDV infection in piglets. Video Abstract.
Collapse
Affiliation(s)
- JunHong Xing
- College of Veterinary Medicine, Jilin Provincial Engineering Research Center of Animal Probiotics, Jilin Provincial Key Laboratory of Animal Microecology and Healthy Breeding, Engineering Research Center of Microecological Vaccines (Drugs) for Major Animal Diseases, Ministry of Education, Jilin Agricultural University, 2888 Xincheng Street, Changchun, 130118, China
| | - TianMing Niu
- College of Veterinary Medicine, Jilin Provincial Engineering Research Center of Animal Probiotics, Jilin Provincial Key Laboratory of Animal Microecology and Healthy Breeding, Engineering Research Center of Microecological Vaccines (Drugs) for Major Animal Diseases, Ministry of Education, Jilin Agricultural University, 2888 Xincheng Street, Changchun, 130118, China
| | - Tong Yu
- College of Veterinary Medicine, Jilin Provincial Engineering Research Center of Animal Probiotics, Jilin Provincial Key Laboratory of Animal Microecology and Healthy Breeding, Engineering Research Center of Microecological Vaccines (Drugs) for Major Animal Diseases, Ministry of Education, Jilin Agricultural University, 2888 Xincheng Street, Changchun, 130118, China
| | - BoShi Zou
- College of Veterinary Medicine, Jilin Provincial Engineering Research Center of Animal Probiotics, Jilin Provincial Key Laboratory of Animal Microecology and Healthy Breeding, Engineering Research Center of Microecological Vaccines (Drugs) for Major Animal Diseases, Ministry of Education, Jilin Agricultural University, 2888 Xincheng Street, Changchun, 130118, China
| | - ChunWei Shi
- College of Veterinary Medicine, Jilin Provincial Engineering Research Center of Animal Probiotics, Jilin Provincial Key Laboratory of Animal Microecology and Healthy Breeding, Engineering Research Center of Microecological Vaccines (Drugs) for Major Animal Diseases, Ministry of Education, Jilin Agricultural University, 2888 Xincheng Street, Changchun, 130118, China
| | - YingJie Wang
- College of Veterinary Medicine, Jilin Provincial Engineering Research Center of Animal Probiotics, Jilin Provincial Key Laboratory of Animal Microecology and Healthy Breeding, Engineering Research Center of Microecological Vaccines (Drugs) for Major Animal Diseases, Ministry of Education, Jilin Agricultural University, 2888 Xincheng Street, Changchun, 130118, China
| | - ShuHui Fan
- College of Veterinary Medicine, Jilin Provincial Engineering Research Center of Animal Probiotics, Jilin Provincial Key Laboratory of Animal Microecology and Healthy Breeding, Engineering Research Center of Microecological Vaccines (Drugs) for Major Animal Diseases, Ministry of Education, Jilin Agricultural University, 2888 Xincheng Street, Changchun, 130118, China
| | - MingHan Li
- College of Veterinary Medicine, Jilin Provincial Engineering Research Center of Animal Probiotics, Jilin Provincial Key Laboratory of Animal Microecology and Healthy Breeding, Engineering Research Center of Microecological Vaccines (Drugs) for Major Animal Diseases, Ministry of Education, Jilin Agricultural University, 2888 Xincheng Street, Changchun, 130118, China
| | - MeiYing Bao
- College of Veterinary Medicine, Jilin Provincial Engineering Research Center of Animal Probiotics, Jilin Provincial Key Laboratory of Animal Microecology and Healthy Breeding, Engineering Research Center of Microecological Vaccines (Drugs) for Major Animal Diseases, Ministry of Education, Jilin Agricultural University, 2888 Xincheng Street, Changchun, 130118, China
| | - Yu Sun
- College of Veterinary Medicine, Jilin Provincial Engineering Research Center of Animal Probiotics, Jilin Provincial Key Laboratory of Animal Microecology and Healthy Breeding, Engineering Research Center of Microecological Vaccines (Drugs) for Major Animal Diseases, Ministry of Education, Jilin Agricultural University, 2888 Xincheng Street, Changchun, 130118, China
| | - KuiPeng Gao
- College of Veterinary Medicine, Jilin Provincial Engineering Research Center of Animal Probiotics, Jilin Provincial Key Laboratory of Animal Microecology and Healthy Breeding, Engineering Research Center of Microecological Vaccines (Drugs) for Major Animal Diseases, Ministry of Education, Jilin Agricultural University, 2888 Xincheng Street, Changchun, 130118, China
| | - JingJing Qiu
- College of Veterinary Medicine, Jilin Provincial Engineering Research Center of Animal Probiotics, Jilin Provincial Key Laboratory of Animal Microecology and Healthy Breeding, Engineering Research Center of Microecological Vaccines (Drugs) for Major Animal Diseases, Ministry of Education, Jilin Agricultural University, 2888 Xincheng Street, Changchun, 130118, China
| | - DongXing Zhang
- College of Veterinary Medicine, Jilin Provincial Engineering Research Center of Animal Probiotics, Jilin Provincial Key Laboratory of Animal Microecology and Healthy Breeding, Engineering Research Center of Microecological Vaccines (Drugs) for Major Animal Diseases, Ministry of Education, Jilin Agricultural University, 2888 Xincheng Street, Changchun, 130118, China
| | - Nan Wang
- College of Veterinary Medicine, Jilin Provincial Engineering Research Center of Animal Probiotics, Jilin Provincial Key Laboratory of Animal Microecology and Healthy Breeding, Engineering Research Center of Microecological Vaccines (Drugs) for Major Animal Diseases, Ministry of Education, Jilin Agricultural University, 2888 Xincheng Street, Changchun, 130118, China
| | - YanLong Jiang
- College of Veterinary Medicine, Jilin Provincial Engineering Research Center of Animal Probiotics, Jilin Provincial Key Laboratory of Animal Microecology and Healthy Breeding, Engineering Research Center of Microecological Vaccines (Drugs) for Major Animal Diseases, Ministry of Education, Jilin Agricultural University, 2888 Xincheng Street, Changchun, 130118, China
| | - HaiBin Huang
- College of Veterinary Medicine, Jilin Provincial Engineering Research Center of Animal Probiotics, Jilin Provincial Key Laboratory of Animal Microecology and Healthy Breeding, Engineering Research Center of Microecological Vaccines (Drugs) for Major Animal Diseases, Ministry of Education, Jilin Agricultural University, 2888 Xincheng Street, Changchun, 130118, China
| | - Xin Cao
- College of Veterinary Medicine, Jilin Provincial Engineering Research Center of Animal Probiotics, Jilin Provincial Key Laboratory of Animal Microecology and Healthy Breeding, Engineering Research Center of Microecological Vaccines (Drugs) for Major Animal Diseases, Ministry of Education, Jilin Agricultural University, 2888 Xincheng Street, Changchun, 130118, China
| | - Yan Zeng
- College of Veterinary Medicine, Jilin Provincial Engineering Research Center of Animal Probiotics, Jilin Provincial Key Laboratory of Animal Microecology and Healthy Breeding, Engineering Research Center of Microecological Vaccines (Drugs) for Major Animal Diseases, Ministry of Education, Jilin Agricultural University, 2888 Xincheng Street, Changchun, 130118, China
| | - JianZhong Wang
- College of Veterinary Medicine, Jilin Provincial Engineering Research Center of Animal Probiotics, Jilin Provincial Key Laboratory of Animal Microecology and Healthy Breeding, Engineering Research Center of Microecological Vaccines (Drugs) for Major Animal Diseases, Ministry of Education, Jilin Agricultural University, 2888 Xincheng Street, Changchun, 130118, China
| | - ShuMin Zhang
- College of Veterinary Medicine, Jilin Provincial Engineering Research Center of Animal Probiotics, Jilin Provincial Key Laboratory of Animal Microecology and Healthy Breeding, Engineering Research Center of Microecological Vaccines (Drugs) for Major Animal Diseases, Ministry of Education, Jilin Agricultural University, 2888 Xincheng Street, Changchun, 130118, China
| | - JingTao Hu
- College of Veterinary Medicine, Jilin Provincial Engineering Research Center of Animal Probiotics, Jilin Provincial Key Laboratory of Animal Microecology and Healthy Breeding, Engineering Research Center of Microecological Vaccines (Drugs) for Major Animal Diseases, Ministry of Education, Jilin Agricultural University, 2888 Xincheng Street, Changchun, 130118, China
| | - Di Zhang
- College of Veterinary Medicine, Jilin Provincial Engineering Research Center of Animal Probiotics, Jilin Provincial Key Laboratory of Animal Microecology and Healthy Breeding, Engineering Research Center of Microecological Vaccines (Drugs) for Major Animal Diseases, Ministry of Education, Jilin Agricultural University, 2888 Xincheng Street, Changchun, 130118, China
| | - WuSheng Sun
- College of Veterinary Medicine, Jilin Provincial Engineering Research Center of Animal Probiotics, Jilin Provincial Key Laboratory of Animal Microecology and Healthy Breeding, Engineering Research Center of Microecological Vaccines (Drugs) for Major Animal Diseases, Ministry of Education, Jilin Agricultural University, 2888 Xincheng Street, Changchun, 130118, China
| | - GuiLian Yang
- College of Veterinary Medicine, Jilin Provincial Engineering Research Center of Animal Probiotics, Jilin Provincial Key Laboratory of Animal Microecology and Healthy Breeding, Engineering Research Center of Microecological Vaccines (Drugs) for Major Animal Diseases, Ministry of Education, Jilin Agricultural University, 2888 Xincheng Street, Changchun, 130118, China.
| | - WenTao Yang
- College of Veterinary Medicine, Jilin Provincial Engineering Research Center of Animal Probiotics, Jilin Provincial Key Laboratory of Animal Microecology and Healthy Breeding, Engineering Research Center of Microecological Vaccines (Drugs) for Major Animal Diseases, Ministry of Education, Jilin Agricultural University, 2888 Xincheng Street, Changchun, 130118, China.
| | - ChunFeng Wang
- College of Veterinary Medicine, Jilin Provincial Engineering Research Center of Animal Probiotics, Jilin Provincial Key Laboratory of Animal Microecology and Healthy Breeding, Engineering Research Center of Microecological Vaccines (Drugs) for Major Animal Diseases, Ministry of Education, Jilin Agricultural University, 2888 Xincheng Street, Changchun, 130118, China.
| |
Collapse
|