1
|
Hetta HF, Ahmed R, Ramadan YN, Fathy H, Khorshid M, Mabrouk MM, Hashem M. Gut virome: New key players in the pathogenesis of inflammatory bowel disease. World J Methodol 2025; 15:92592. [DOI: 10.5662/wjm.v15.i2.92592] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/30/2024] [Revised: 05/28/2024] [Accepted: 07/23/2024] [Indexed: 11/27/2024] Open
Abstract
Inflammatory bowel disease (IBD) is a chronic inflammatory illness of the intestine. While the mechanism underlying the pathogenesis of IBD is not fully understood, it is believed that a complex combination of host immunological response, environmental exposure, particularly the gut microbiota, and genetic susceptibility represents the major determinants. The gut virome is a group of viruses found in great frequency in the gastrointestinal tract of humans. The gut virome varies greatly among individuals and is influenced by factors including lifestyle, diet, health and disease conditions, geography, and urbanization. The majority of research has focused on the significance of gut bacteria in the progression of IBD, although viral populations represent an important component of the microbiome. We conducted this review to highlight the viral communities in the gut and their expected roles in the etiopathogenesis of IBD regarding published research to date.
Collapse
Affiliation(s)
- Helal F Hetta
- Department of Medical Microbiology and Immunology, Faculty of Medicine, Assiut University, Assiut 71515, Egypt
- Division of Microbiology, Immunology and Biotechnology, Faculty of pharmacy, University of Tabuk, Tabuk 71491, Saudi Arabia
| | - Rehab Ahmed
- Division of Microbiology, Immunology and Biotechnology, Department of Natural Products and Alternative Medicine, Faculty of Pharmacy, University of Tabuk, Tabuk 71491, Saudi Arabia
| | - Yasmin N Ramadan
- Department of Medical Microbiology and Immunology, Faculty of Medicine, Assiut University, Assiut 71515, Egypt
| | - Hayam Fathy
- Department of Internal Medicine, Division Hepatogastroenterology, Assiut University, Assiut 71515, Egypt
| | - Mohammed Khorshid
- Department of Clinical Research, Egyptian Developers of Gastroenterology and Endoscopy Foundation, Cairo 11936, Egypt
| | - Mohamed M Mabrouk
- Department of Internal Medicine, Faculty of Medicine. Tanta University, Tanta 31527, Egypt
| | - Mai Hashem
- Department of Tropical Medicine, Gastroenterology and Hepatology, Assiut University Hospital, Assiut 71515, Egypt
| |
Collapse
|
2
|
Jin Y, Liu H, Wang Y, Zhang R, Wang Q, Wang Y, Cui H, Wang X, Bian Y. Pathogenesis and treatment of colitis-associated colorectal cancer: Insights from Traditional Chinese Medicine. JOURNAL OF ETHNOPHARMACOLOGY 2025; 338:119096. [PMID: 39532222 DOI: 10.1016/j.jep.2024.119096] [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: 06/18/2024] [Revised: 10/11/2024] [Accepted: 11/09/2024] [Indexed: 11/16/2024]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Inflammatory Bowel Disease (IBD) is an inflammatory intestinal disease, and with prolonged illness duration, the annual risk of IBD progressing to colitis-associated colorectal cancer (CAC) gradually increases. In recent years, there has been a noticeable trend towards the application of traditional Chinese medicine (TCM) in the treatment of CAC. AIM OF THIS REVIEW This comprehensive review summarizes the pathogenesis of CAC and details the therapeutic benefits of TCM in treating CAC, including various TCM prescriptions and ingredients, establishing the theoretical foundation for the application of TCM in CAC treatment. METHODS We assessed literature published before March 24, 2024, from several databases, including Web of Science, PubMed, Scopus and Google Scholar. The keywords used include "traditional Chinese medicine", "traditional Chinese medicine prescriptions", "traditional Chinese medicine ingredients", "herbal medicine", "colitis-associated colorectal cancer", "inflammatory bowel disease", "colorectal cancer" and "colitis-cancer transformation". We conducted a comprehensive collection and collation of pertinent scientific articles from various databases, focusing on the efficacy of TCM in the prevention and treatment of "colitis-cancer transformation". RESULTS This paper provides a concise summary and thorough analysis of twenty-eight prescriptions and ingredients of TCM for the prevention and treatment of CAC, based on existing experimental and clinical research. There are positive signs that TCM can effectively prevent and treat the "colitis-cancer transformation" through repairing the intestinal mucosal barrier, correcting intestinal flora imbalance, and regulating intestinal immune responses. CONCLUSION TCM possesses comprehensive regulatory advantages that are multifaceted, multilevel, and multitarget. It has a definite curative effect in the prevention and treatment of CAC. It is essential to enhance the clinical efficacy of TCM in the prevention and treatment of CAC based on syndrome differentiation and treatment, with the assistance of modern medicine.
Collapse
Affiliation(s)
- Yutong Jin
- School of Integrative Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China
| | - Haizhao Liu
- Department of Integrated Traditional Chinese and Western Medicine, Tianjin First Central Hospital, Nankai University, Tianjin, 300192, China
| | - Yuhui Wang
- School of Integrative Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China
| | - Ruixuan Zhang
- School of Integrative Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China
| | - Qiaochu Wang
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, 300073, China
| | - Yao Wang
- School of Integrative Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China; School of Basic Medical Sciences, Heilongjiang University of Chinese Medicine, Harbin, 150040, China
| | - Huantian Cui
- First School of Clinical Medicine, Yunnan University of Chinese Medicine, Kunming, 650500, China.
| | - Xiangling Wang
- School of Integrative Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China.
| | - Yuhong Bian
- School of Integrative Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China.
| |
Collapse
|
3
|
Cao H, Liu H, Dai X, Shi B, Yuan J, Shan J, Lin J. Qingchang suppository ameliorates mucosal inflammation in ulcerative colitis by inhibiting the differentiation and effector functions of Th1 and Th17 cells. JOURNAL OF ETHNOPHARMACOLOGY 2025; 337:118865. [PMID: 39343108 DOI: 10.1016/j.jep.2024.118865] [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: 07/26/2024] [Revised: 09/23/2024] [Accepted: 09/25/2024] [Indexed: 10/01/2024]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Qing Chang Suppository (QCS), a traditional Chinese medicine formula, has been shown to effectively alleviate mucosal inflammation in patients with ulcerative colitis (UC). While the mechanism of QCS appears to be related to the regulation of CD4+T cell subset responses, direct evidence demonstrating that QCS inhibits Th1 and Th17 cell activation in UC (particularly based on human data) remains lacking. Additionally, the precise mechanisms through which QCS affects these cells have yet to be fully elucidated. AIM OF STUDY This study aimed to investigate the effects of QCS on Th1 and Th17 cell responses in UC and to explore the underlying mechanisms. MATERIALS AND METHODS Twenty-eight patients with mild-to-moderate UC were recruited and treated with QCS for 12 weeks. Symptoms were assessed every two weeks, with sigmoidoscopies performed at baseline and at week 12. Intestinal mucosal biopsies and peripheral blood (PB) were collected at these time points. At the end of the trial, patients were categorized into responder and non-responder groups based on a modified Mayo disease activity index score. Healthy controls (HCs) were defined as subjects without IBD or colorectal carcinoma but with colon polyps. The frequencies of IFN-γ+CD4+T cells and IL-17A+CD4+T cells in PB and colonic mucosa were measured using flow cytometry. The expression levels and localization of T-bet, RORγT, IFN-γ, TNF-α, and IL-17A were determined via immunofluorescence, and JNK signaling activation was assessed through immunoblotting and immunohistochemistry. All parameters were compared across the three groups. RESULTS At week 12, responders showed a significant reduction in colonic mucosal inflammation compared to baseline, accompanied by decreased frequencies of IFN-γ+CD4+T and IL-17A+CD4+ T cells in both PB and the colonic epithelial layer. Notably, Th1 and Th17 cell activity around intestinal epithelial cells (IECs) was nearly undetectable, as evidenced by the diminished expression of T-bet, RORγT, IFN-γ, TNF-α, and IL-17A. Additionally, JNK phosphorylation in these cells was significantly reduced. In contrast, non-responders exhibited no meaningful improvement; colonic pathology remained unchanged, and elevated levels of IFN-γ+CD4+T and IL-17A+CD 4+T cells persisted in both the PB and colonic epithelial layer. The presence of Th1 and Th17 cells and their associated cytokines around IECs remained substantial, and there was no significant change in JNK activation. CONCLUSION QCS attenuates mucosal inflammation in UC patients by inhibiting the differentiation and effector functions of Th1 and Th17 cells, primarily through the regulation of the JNK signaling pathway.
Collapse
Affiliation(s)
- Hui Cao
- Department of Gastroenterology, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China; Department of Spleen and Stomach Diseases, Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, China
| | - Huosheng Liu
- Department of Acupuncture and Moxibustion, Shanghai Jiading Hospital of Traditional Chinese Medicine, Shanghai, China
| | - Xiaoling Dai
- Department of Gastroenterology, Shanghai Putuo Traditional Chinese Medicine Hospital, Shanghai 200063, China
| | - Bei Shi
- Department of Gastroenterology, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Jianye Yuan
- Clinical Research Unit, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Jingyi Shan
- Department of Gastroenterology, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China.
| | - Jiang Lin
- Department of Gastroenterology, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China.
| |
Collapse
|
4
|
Mansouri P, Mansouri P, Behmard E, Najafipour S, Kouhpayeh A, Farjadfar A. Novel targets for mucosal healing in inflammatory bowel disease therapy. Int Immunopharmacol 2025; 144:113544. [PMID: 39571265 DOI: 10.1016/j.intimp.2024.113544] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2024] [Revised: 10/13/2024] [Accepted: 10/28/2024] [Indexed: 12/15/2024]
Abstract
Inflammatory bowel disease (IBD) is a chronic condition affecting the gastrointestinal tract, primarily manifesting as ulcerative colitis (UC) or Crohn's disease (CD). Both inflammation and disruption of the intestinal epithelial barrier are key factors in IBD pathogenesis. Substantial evidence has revealed a significant association between aberrant immune responses and impairment of the intestinal epithelial barrier in IBD pathogenesis. The components of the intestinal epithelium, particularly goblet cells and Paneth cells, are crucial to gut homeostasis, as they secrete mucin, antimicrobial peptides (AMPs), and cytokines. Furthermore, impairment of epithelial integrity, which is regulated by tight junctions, is a hallmark of IBD pathology. While common treatments for IBD, such as anti-inflammatory drugs, target various signaling pathways with varying efficacies, therapeutic approaches focused on mucosal and epithelial barrier healing have been largely neglected. Moreover, high costs, side effects, and insufficient or inconsistent therapeutic outcomes remain major drawbacks of conventional anti-IBD drugs. Recent studies on epithelial barrier regeneration and permeability reduction have introduced promising therapeutic targets, including farnesoid X receptor (FXR), urokinase-type plasminogen activator (uPA)-urokinase-type plasminogen activator receptor (uPAR) interaction, fecal microbiota transplantation (FMT), and insulin receptor (INSR). Notably, the simultaneous targeting of intestinal inflammation and promotion of epithelial barrier healing shows promise for efficient IBD treatment. Future research should explore targeted therapies and combination treatments, including natural remedies, microbiota colonization, stem cell approaches, and computer-aided drug design. It is also crucial to focus on accurate prognosis and developing a thorough understanding of IBD development mechanisms.
Collapse
Affiliation(s)
- Pardis Mansouri
- Student Research Committee, Fasa University of Medical Sciences, Fasa, Iran; Department of Medical Biotechnology, Fasa University of Medical Sciences, Fasa, Iran
| | - Pegah Mansouri
- Student Research Committee, Fasa University of Medical Sciences, Fasa, Iran; Department of Medical Biotechnology, Fasa University of Medical Sciences, Fasa, Iran
| | - Esmaeil Behmard
- School of Advanced Technologies in Medicine, Fasa University of Medical Sciences, Fasa, Iran; Zarrin Avaye Kowsar Salamat (ZAX Company), Fasa, Iran
| | - Sohrab Najafipour
- School of Advanced Technologies in Medicine, Fasa University of Medical Sciences, Fasa, Iran; Zarrin Avaye Kowsar Salamat (ZAX Company), Fasa, Iran
| | - Amin Kouhpayeh
- Department of Pharmacology, Faculty of Medicine, Fasa University of Medical Sciences, Fasa, Iran; Zarrin Avaye Kowsar Salamat (ZAX Company), Fasa, Iran.
| | - Akbar Farjadfar
- Department of Medical Biotechnology, Fasa University of Medical Sciences, Fasa, Iran; Zarrin Avaye Kowsar Salamat (ZAX Company), Fasa, Iran.
| |
Collapse
|
5
|
Nicolò S, Faggiani I, Errico C, D'Amico F, Parigi TL, Danese S, Ungaro F. Translational characterization of immune pathways in inflammatory bowel disease: insights for targeted treatments. Expert Rev Clin Immunol 2025; 21:55-72. [PMID: 39313992 DOI: 10.1080/1744666x.2024.2400300] [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: 01/15/2024] [Accepted: 08/30/2024] [Indexed: 09/25/2024]
Abstract
INTRODUCTION The pathogenesis of inflammatory bowel disease (IBD) involves the dysregulation of multiple inflammatory pathways. The understanding of these mechanisms allows their selective targeting for therapeutic purposes. The discovery of Tumor Necrosis Factor-alpha's (TNF-α) role in mucosal inflammation ushered an exciting new era of drug development which now comprises agents targeting multiple pro-inflammatory signaling pathways, integrins, and leukocyte trafficking regulators. AREA COVERED This review provides an overview of the main molecular players of IBD, their translation into therapeutic targets and the successful development of the advanced agents modulating them. We combine basic science with clinical trials data to present a critical review of both the successful and failed drug development programs. A PubMed literature search was conducted to delve into the available literature and clinical trials. EXPERT OPINION The treatment landscape for IBD has rapidly expanded, particularly with the development of biologics targeting TNF-α, integrins, and S1P modulators, as well as newer agents such as IL-12/IL-23 inhibitors and JAK inhibitors, offering robust efficacy and safety profiles. However, challenges persist in understanding and effectively treating difficult-to-treat IBD, highlighting the need for continued research to uncover novel therapeutic targets and optimize patient outcomes.
Collapse
Affiliation(s)
- Sabrina Nicolò
- Department of Gastroenterology and Digestive Endoscopy, IRCCS Ospedale San Raffaele, Milan, Italy
- Faculty of Medicine, Università Vita-Salute San Raffaele, Milan, Italy
| | - Ilaria Faggiani
- Department of Gastroenterology and Digestive Endoscopy, IRCCS Ospedale San Raffaele, Milan, Italy
- Faculty of Medicine, Università Vita-Salute San Raffaele, Milan, Italy
| | - Carmela Errico
- Department of Gastroenterology and Digestive Endoscopy, IRCCS Ospedale San Raffaele, Milan, Italy
- Faculty of Medicine, Università Vita-Salute San Raffaele, Milan, Italy
| | - Ferdinando D'Amico
- Department of Gastroenterology and Digestive Endoscopy, IRCCS Ospedale San Raffaele, Milan, Italy
| | - Tommaso Lorenzo Parigi
- Department of Gastroenterology and Digestive Endoscopy, IRCCS Ospedale San Raffaele, Milan, Italy
- Faculty of Medicine, Università Vita-Salute San Raffaele, Milan, Italy
| | - Silvio Danese
- Department of Gastroenterology and Digestive Endoscopy, IRCCS Ospedale San Raffaele, Milan, Italy
- Faculty of Medicine, Università Vita-Salute San Raffaele, Milan, Italy
| | - Federica Ungaro
- Department of Gastroenterology and Digestive Endoscopy, IRCCS Ospedale San Raffaele, Milan, Italy
- Faculty of Medicine, Università Vita-Salute San Raffaele, Milan, Italy
| |
Collapse
|
6
|
Zhang J, Sun Z, Cheng L, Kang J, Liu Y, Zhao Y, Xiao M, Liu H, Zhu Q, Guo Q, Lin C. Structural Characterization of Water-Soluble Pectin from the Fruit of Diospyros lotus L. and Its Protective Effects against DSS-Induced Colitis in Mice. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2025. [PMID: 39742427 DOI: 10.1021/acs.jafc.4c07611] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
Abstract
Polysaccharides from Diospyros lotus L. were investigated for their structural characterization and anti-inflammatory activity. Four low polymer dispersity index (PDI) subfractions were obtained: DRP-1 (153.95 kDa), DRP-2 (61.22 kDa), DRP-3 (22.80 kDa), and DRP-4 (8.93 kDa), respectively. DRP-4 contained the highest number of RG-I domains (43.25%), while DRP-1 had the highest degree of methyl esterification (37.5%). Results from the dextran sulfate sodium (DSS) salt-induced ulcerative colitis (UC) mice model indicated that DCP promoted mucosal and tight junction protein (caudin-1 and occludin) expression. Moreover, DCP improved the microbial community composition through selective enrichment of beneficial bacteria such as Lachnospiraceae and Lactobacillaceae. The anti-inflammatory activity of DCP was speculated to be related to its neutral sugar side chain and low esterification degree. These results suggested that DCP could prevent DSS-induced colitis and inhibit colon inflammation by maintaining a balanced gut microbiome and protecting the colon mucosal barrier.
Collapse
Affiliation(s)
- Jixiang Zhang
- State Key Laboratory of Food Nutrition and Safety, College of Food Science and Engineering, Tianjin University of Science and Technology, Tianjin 300457, China
| | - Zhenou Sun
- State Key Laboratory of Food Nutrition and Safety, College of Food Science and Engineering, Tianjin University of Science and Technology, Tianjin 300457, China
| | - Liting Cheng
- State Key Laboratory of Food Nutrition and Safety, College of Food Science and Engineering, Tianjin University of Science and Technology, Tianjin 300457, China
| | - Ji Kang
- State Key Laboratory of Food Nutrition and Safety, College of Food Science and Engineering, Tianjin University of Science and Technology, Tianjin 300457, China
| | - Yan Liu
- College of Food and Health, Zhejiang Agriculture and Forestry University, No. 666 Wusu Road, Linan District, Hangzhou 311300 Zhejiang, China
| | - Yi Zhao
- State Key Laboratory of Food Nutrition and Safety, College of Food Science and Engineering, Tianjin University of Science and Technology, Tianjin 300457, China
| | - Meng Xiao
- State Key Laboratory of Food Nutrition and Safety, College of Food Science and Engineering, Tianjin University of Science and Technology, Tianjin 300457, China
| | - Huanhuan Liu
- State Key Laboratory of Food Nutrition and Safety, College of Food Science and Engineering, Tianjin University of Science and Technology, Tianjin 300457, China
| | - Qiaomei Zhu
- State Key Laboratory of Food Nutrition and Safety, College of Food Science and Engineering, Tianjin University of Science and Technology, Tianjin 300457, China
| | - Qingbin Guo
- State Key Laboratory of Food Nutrition and Safety, College of Food Science and Engineering, Tianjin University of Science and Technology, Tianjin 300457, China
| | - Changbin Lin
- Institute of Agro-Products Processing Science and Technology, Sichuan Academy of Agricultural Sciences, Chengdu 610066 Sichuan, China
| |
Collapse
|
7
|
Kalafateli M, Tourkochristou E, Tsounis EP, Aggeletopoulou I, Triantos C. New Insights into the Pathogenesis of Intestinal Fibrosis in Inflammatory Bowel Diseases: Focusing on Intestinal Smooth Muscle Cells. Inflamm Bowel Dis 2024:izae292. [PMID: 39680685 DOI: 10.1093/ibd/izae292] [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: 06/15/2024] [Indexed: 12/18/2024]
Abstract
Strictures in inflammatory bowel disease, especially Crohn's disease (CD), are characterized by increased intestinal wall thickness, which, according to recent accumulating data, is mainly attributed to the expansion of the intestinal smooth muscle layers and to a lesser extent to collagen deposition. In this review, we will discuss the role of intestinal smooth muscle cells (SMCs) as crucial orchestrators of stricture formation. Activated SMCs can synthesize extracellular matrix (ECM), thus contributing to intestinal fibrosis, as well as growth factors and cytokines that can further enhance ECM production, stimulate other surrounding mesenchymal and immune cells, and increase SMC proliferation via paracrine or autocrine signaling. There is also evidence that, in stricturing CD, a phenotypic modulation of SMC toward a myofibroblast-like synthetic phenotype takes place. Moreover, the molecular mechanisms and signaling pathways that regulate SMC hyperplasia/hypertrophy will be extensively reviewed. The understanding of the cellular network and the molecular background behind stricture formation is essential for the design of effective anti-fibrotic strategies, and SMCs might be a promising therapeutic target in the future.
Collapse
Affiliation(s)
- Maria Kalafateli
- Department of Gastroenterology, General Hospital of Patras, Patras, Greece
| | - Evanthia Tourkochristou
- Division of Hematology, Department of Internal Medicine, Medical School, University of Patras, Patras, Greece
| | - Efthymios P Tsounis
- Division of Hematology, Department of Internal Medicine, Medical School, University of Patras, Patras, Greece
| | - Ioanna Aggeletopoulou
- Division of Gastroenterology, Department of Internal Medicine, University Hospital of Patras, Patras, Greece
| | - Christos Triantos
- Division of Hematology, Department of Internal Medicine, Medical School, University of Patras, Patras, Greece
| |
Collapse
|
8
|
Neurath MF, Sands BE, Rieder F. Cellular immunotherapies and immune cell depleting therapies in inflammatory bowel diseases: the next magic bullet? Gut 2024; 74:9-14. [PMID: 39025492 DOI: 10.1136/gutjnl-2024-332919] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/13/2024] [Accepted: 07/08/2024] [Indexed: 07/20/2024]
Abstract
Despite significant advances in biologic and small molecule treatments and the emergence of combination therapies to treat inflammatory bowel diseases (IBD) a large unmet need remains to control intestinal inflammation. New approaches targeting several pathways simultaneously with a favorable safety profile and agents that trigger anti-inflammatory pathways to drive durable resolution of inflammation are needed. This article discusses novel cellular immunotherapies and immune cell depleting therapies in IBD, including CAR-T cell approaches, Tr1 and T regulatory (Treg) cells and cell depleting antibodies such as rosnilimab. These novel approaches have the potential to overcome current therapeutic limitations in the treatment of IBD.
Collapse
Affiliation(s)
- Markus Friedrich Neurath
- Department of Medicine 1, Kussmaul Research Campus & Ludwig Demling Endoscopy Center of Excellence, Deutsches Zentrum Immuntherapie DZI, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Bruce Eric Sands
- Dr Henry D Janowitz Division of Gastroenterology, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Florian Rieder
- Department of Gastroenterology, Hepatology & Nutrition, Digestive Diseases Institute; Department of Inflammation and Immunity, Lerner Research Institute, Center for Global Translational Inflammatory Bowel Disease Research, Cleveland Clinic Foundation, Cleveland, Ohio, USA
| |
Collapse
|
9
|
Peng Y, Chen L, Chen X, Lin J, Wei J, Cheng J, Zhou F, Ge L, Zhou R, Ding F, Wang X. NPSR1 promotes chronic colitis through regulating CD4 + T cell effector function in inflammatory bowel disease. Int Immunopharmacol 2024; 142:113252. [PMID: 39332092 DOI: 10.1016/j.intimp.2024.113252] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2024] [Revised: 08/31/2024] [Accepted: 09/22/2024] [Indexed: 09/29/2024]
Abstract
BACKGROUND Neuropeptide S receptor 1 (NPSR1) has been implicated in the the onset of inflammatory bowel disease (IBD), though its exact mechanism remains unclear. This study investigates the role of NPSR1 in regulating CD4+ T cell effector function in IBD. METHODS Peripheral blood and colonic mucosal biopsies from IBD patients, as well as dextran sodium sulfate (DSS)-induced mouse colitis models, were analyzed to assess the effects of NPSR1 on colitis and CD4+ T cell-mediated immune responses. NPSR1 knockdown was conducted both in vitro and in vivo to elucidate underlying mechanisms. Expression of NPSR1 and CD4+ T cell-related factors was measured using quantitative real-time PCR, immunoblotting, cytometric bead array, immunofluorescence, and immunohistochemistry. CD4 + T cell effector functions were evaluated through flow cytometry, EdU incorporation assay, Annexin V-FITC/PI staining, and transwell assay. RESULTS NPSR1 expression was elevated in the intestinal tissues from IBD patients. Its downregulation provided protection in DSS-induced mouse colitis models. NPSR1 correlated positively with CD4 + T cell-mediated inflammation, and its knockdown reduced CD4+ T cell-mediated immune responses and inhibited CD4+ T cell differentiation. Additionally, NPSR1 knockdown decreased CD4+ T cell proliferation, increased apoptosis, and enhanced CCL2-induced migration in vitro, while significantly reducing Th1 cell chemotaxis in vivo. CONCLUSIONS This study demonstrates that NPSR1 promotes chronic colitis by regulating CD4 + T cell effector functions in IBD, offering potential new therapeutic strategies for IBD treatment.
Collapse
Affiliation(s)
- Yanan Peng
- Department of Gastroenterology, Zhongnan Hospital of Wuhan University, Wuhan, China; Hubei Provincial Clinical Research Center for Intestinal and Colorectal Diseases, Hubei Key Laboratory of Intestinal and Colorectal Diseases, Wuhan, China
| | - Liping Chen
- Department of Gastroenterology, Zhongnan Hospital of Wuhan University, Wuhan, China; Hubei Provincial Clinical Research Center for Intestinal and Colorectal Diseases, Hubei Key Laboratory of Intestinal and Colorectal Diseases, Wuhan, China
| | - Xiaojia Chen
- Department of Gastroenterology, Zhongnan Hospital of Wuhan University, Wuhan, China; Hubei Provincial Clinical Research Center for Intestinal and Colorectal Diseases, Hubei Key Laboratory of Intestinal and Colorectal Diseases, Wuhan, China
| | - Jun Lin
- Department of Gastroenterology, Zhongnan Hospital of Wuhan University, Wuhan, China; Hubei Provincial Clinical Research Center for Intestinal and Colorectal Diseases, Hubei Key Laboratory of Intestinal and Colorectal Diseases, Wuhan, China
| | - Jia Wei
- Department of Gastroenterology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shanxi, China
| | - Jie Cheng
- Department of Gastroenterology, Zhongnan Hospital of Wuhan University, Wuhan, China; Hubei Provincial Clinical Research Center for Intestinal and Colorectal Diseases, Hubei Key Laboratory of Intestinal and Colorectal Diseases, Wuhan, China
| | - Feng Zhou
- Department of Gastroenterology, Zhongnan Hospital of Wuhan University, Wuhan, China; Hubei Provincial Clinical Research Center for Intestinal and Colorectal Diseases, Hubei Key Laboratory of Intestinal and Colorectal Diseases, Wuhan, China
| | - Liuqing Ge
- Department of Gastroenterology, Zhongnan Hospital of Wuhan University, Wuhan, China; Hubei Provincial Clinical Research Center for Intestinal and Colorectal Diseases, Hubei Key Laboratory of Intestinal and Colorectal Diseases, Wuhan, China
| | - Rui Zhou
- Department of Gastroenterology, Zhongnan Hospital of Wuhan University, Wuhan, China; Hubei Provincial Clinical Research Center for Intestinal and Colorectal Diseases, Hubei Key Laboratory of Intestinal and Colorectal Diseases, Wuhan, China
| | - Feng Ding
- Department of Gastroenterology, Zhongnan Hospital of Wuhan University, Wuhan, China; Hubei Provincial Clinical Research Center for Intestinal and Colorectal Diseases, Hubei Key Laboratory of Intestinal and Colorectal Diseases, Wuhan, China
| | - Xiaobing Wang
- Department of Gastroenterology, Zhongnan Hospital of Wuhan University, Wuhan, China; Hubei Provincial Clinical Research Center for Intestinal and Colorectal Diseases, Hubei Key Laboratory of Intestinal and Colorectal Diseases, Wuhan, China.
| |
Collapse
|
10
|
Raftery AL, Pattaroni C, Harris NL, Tsantikos E, Hibbs ML. Environmental and inflammatory factors influencing concurrent gut and lung inflammation. Inflamm Res 2024; 73:2123-2139. [PMID: 39432107 PMCID: PMC11632041 DOI: 10.1007/s00011-024-01953-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2024] [Revised: 09/22/2024] [Accepted: 09/25/2024] [Indexed: 10/22/2024] Open
Abstract
BACKGROUND Crohn's disease and chronic obstructive pulmonary disease (COPD) are chronic inflammatory diseases that affect the gut and lung respectively and can occur comorbidly. METHODS Using the SHIP-1-/- model of Crohn's-like ileitis and chronic lung inflammation, the two diseases were co-investigated. RESULTS Contrary to prior literature, Crohn's-like ileitis was not fully penetrant in SHIP-1-/- mice, and housing in a specific pathogen-free facility was completely protective. Indeed, ileal tissue from SHIP-1-/- mice without overt ileitis was similar to control ilea. However, SHIP-1-/- mice with ileitis exhibited increased granulocytes in ileal tissue together with T cell lymphopenia and they lacked low abundance Bifidobacteria, suggesting this bacterium protects against ileitis. Lung disease, as defined by inflammation in lung washes, emphysema, and lung consolidation, was present in SHIP-1-/- mice regardless of ileitis phenotype; however, there was a shift in the nature of lung inflammation in animals with ileitis, with increased G-CSF and neutrophils, in addition to type 2 cytokines and eosinophils. Deficiency of G-CSF, which protects against lung disease, protected against the development of ileitis in SHIP-1-/- mice. CONCLUSIONS These studies have defined environmental, immune, and inflammatory factors that predispose to ileitis, and have identified that comorbid lung disease correlates with a granulocyte signature.
Collapse
Affiliation(s)
- April L Raftery
- Department of Immunology, School of Translational Medicine, Monash University, 89 Commercial Road, Melbourne, VIC, 3004, Australia
| | - Céline Pattaroni
- Department of Immunology, School of Translational Medicine, Monash University, 89 Commercial Road, Melbourne, VIC, 3004, Australia
| | - Nicola L Harris
- Department of Immunology, School of Translational Medicine, Monash University, 89 Commercial Road, Melbourne, VIC, 3004, Australia
| | - Evelyn Tsantikos
- Department of Immunology, School of Translational Medicine, Monash University, 89 Commercial Road, Melbourne, VIC, 3004, Australia
| | - Margaret L Hibbs
- Department of Immunology, School of Translational Medicine, Monash University, 89 Commercial Road, Melbourne, VIC, 3004, Australia.
| |
Collapse
|
11
|
Poli G, Argiento R, Amedei A, Stingo FC. High-Dimensional Bayesian Semiparametric Models for Small Samples: A Principled Approach to the Analysis of Cytokine Expression Data. Biom J 2024; 66:e70000. [PMID: 39470109 DOI: 10.1002/bimj.70000] [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: 11/29/2023] [Revised: 06/26/2024] [Accepted: 07/04/2024] [Indexed: 10/30/2024]
Abstract
In laboratory medicine, due to the lack of sample availability and resources, measurements of many quantities of interest are commonly collected over a few samples, making statistical inference particularly challenging. In this context, several hypotheses can be tested, and studies are not often powered accordingly. We present a semiparametric Bayesian approach to effectively test multiple hypotheses applied to an experiment that aims to identify cytokines involved in Crohn's disease (CD) infection that may be ongoing in multiple tissues. We assume that the positive correlation commonly observed between cytokines is caused by latent groups of effects, which in turn result from a common cause. These clusters are effectively modeled through a Dirichlet Process (DP) that is one of the most popular choices as nonparametric prior in Bayesian statistics and has been proven to be a powerful tool for model-based clustering. We use a spike-slab distribution as the base measure of the DP. The nonparametric part has been included in an additive model whose parametric component is a Bayesian hierarchical model. We include simulations that empirically demonstrate the effectiveness of the proposed testing procedure in settings that mimic our application's sample size and data structure. Our CD data analysis shows strong evidence of a cytokine gradient in the external intestinal tissue.
Collapse
Affiliation(s)
- Giovanni Poli
- Department of Statistics, Computer Science, Applications "G. Parenti", Università degli Studi di Firenze, Firenze, Italy
| | - Raffaele Argiento
- Department of Economics, Università degli Studi di Bergamo, Bergamo, Italy
- Department of Statistical Sciences, Università Cattolica del Sacro Cuore, Milano, Italy
| | - Amedeo Amedei
- Department of Experimental and Clinical Medicine, Università degli Studi di Firenze, Firenze, Italy
| | - Francesco C Stingo
- Department of Statistics, Computer Science, Applications "G. Parenti", Università degli Studi di Firenze, Firenze, Italy
| |
Collapse
|
12
|
Zhao W, Li S, Li Q, Li Q, Zheng Y, Lu H. Mendelian randomization reveals predictive, preventive, and personalized insights into inflammatory bowel disease: the role of gut microbiome and circulating inflammatory proteins. EPMA J 2024; 15:693-709. [PMID: 39635016 PMCID: PMC11612091 DOI: 10.1007/s13167-024-00384-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2024] [Accepted: 11/05/2024] [Indexed: 12/07/2024]
Abstract
Background A chronic illness with increasing global frequency, inflammatory bowel disease (IBD), which includes ulcerative colitis (UC) and Crohn's disease (CD), profoundly affects patients' quality of life and healthcare systems. IBD pathogenesis consists of changes in gut microbiota, immune system dysregulation, and genetic predisposition. Although emerging data suggests that gut microbiota and circulating inflammatory proteins play critical roles in IBD, their utility as biomarkers for predictive, preventive, and personalized medicine (PPPM) remains incompletely understood. Working hypothesis and methods We hypothesized that specific gut microbiota and inflammatory proteins causally influence IBD risk and mediate pathways between gut microbiota and IBD development. We employed Mendelian randomization (MR) using genome-wide association studies (GWAS) to explore these causal relationships, including further analyses on UC and CD subtypes. Results We identified eight gut microbiota species linked to IBD, with four protective and four increasing risk. Nine inflammatory proteins were also associated, six increasing risk and three protective. MMP-10 and IL-10Rα mediated the effects of Clostridiaceae1 on IBD risk. For UC, five microbiota species were protective, five were risk factors, and two proteins increased risk while three were protective. IL-10Rα mediated the effects of Clostridiaceae1 on UC risk. For CD, eight microbiota species were protective, four increased risk, and nine proteins were implicated. However, no mediation pathways were supported by multivariable MR. Conclusions This study highlights specific gut microbiota and inflammatory proteins that may serve as therapeutic targets for PPPM in IBD, UC, and CD. These findings offer new insights into IBD pathogenesis and suggest potential avenues for improved prevention, early detection, and personalized treatment strategies. Supplementary Information The online version contains supplementary material available at 10.1007/s13167-024-00384-2.
Collapse
Affiliation(s)
- Wuqing Zhao
- The First Clinical Medical College, Lanzhou University, Lanzhou, China
- Department of Gastroenterology, The First Hospital of Lanzhou University, Lanzhou, China
- Gansu Province Clinical Research Center for Digestive Diseases, The First Hospital of Lanzhou University, Lanzhou, China
| | - Shixiao Li
- The First Clinical Medical College, Lanzhou University, Lanzhou, China
- Department of Gastroenterology, The First Hospital of Lanzhou University, Lanzhou, China
- Gansu Province Clinical Research Center for Digestive Diseases, The First Hospital of Lanzhou University, Lanzhou, China
| | - Qianqian Li
- The First Clinical Medical College, Lanzhou University, Lanzhou, China
- Department of Gastroenterology, The First Hospital of Lanzhou University, Lanzhou, China
- Gansu Province Clinical Research Center for Digestive Diseases, The First Hospital of Lanzhou University, Lanzhou, China
| | - Qiang Li
- Department of Gastroenterology, The First Hospital of Lanzhou University, Lanzhou, China
- Gansu Province Clinical Research Center for Digestive Diseases, The First Hospital of Lanzhou University, Lanzhou, China
| | - Ya Zheng
- Department of Gastroenterology, The First Hospital of Lanzhou University, Lanzhou, China
- Gansu Province Clinical Research Center for Digestive Diseases, The First Hospital of Lanzhou University, Lanzhou, China
| | - Hong Lu
- Department of Gastroenterology, The First Hospital of Lanzhou University, Lanzhou, China
- Gansu Province Clinical Research Center for Digestive Diseases, The First Hospital of Lanzhou University, Lanzhou, China
| |
Collapse
|
13
|
Vitali R, Novelli F, Palone F, Cucchiara S, Stronati L, Pioli C. PARP1 inactivation increases regulatory T / Th17 cell proportion in intestinal inflammation. Role of HMGB1. Immunol Lett 2024; 270:106912. [PMID: 39237041 DOI: 10.1016/j.imlet.2024.106912] [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: 12/29/2023] [Revised: 08/12/2024] [Accepted: 09/01/2024] [Indexed: 09/07/2024]
Abstract
Inflammatory bowel diseases (IBD) are chronic relapsing disorders with increasing prevalence. Knowledge gaps still limit the possibility to develop more specific and effective therapies. Using a dextran sodium sulfate colitis mouse model, we found that inflammation increased the total number and altered the frequencies of leukocytes within colon mesenteric lymph nodes (cMLNs). Although the inflammation reduced the frequency of regulatory T (Treg) cells, their absolute numbers were increased. Increased frequency of colitogenic Th17 cells was also observed. Noteworthy, untreated mice lacking Poly(ADP-ribose)-Polimerase-1 functional gene (PARP-1KO) displayed higher frequency of Treg cells and lower percentage of Th17 cells in cMLNs. In colitic PARP-1KO mice the inflammation driven expansion of the Foxp3 Treg population was more pronounced than in WT mice. Conversely, colitis increased Th17 cells to a lower extent in PARP-1KO mice compared with WT mice, resulting in a more protective Treg/Th17 cell ratio. Consequently PARP-1KO mice developed less severe colitis with reduced expression of inflammatory cytokines. In ex vivo experiments PARP-1KO and WT CD11c dendritic cells (DCs) promoted naïve CD4 T cell differentiation differently, the former sustaining more efficiently the generation of Treg cells, the latter that of Th17 cells. Addition of HMGB1 B box or of dipotassium glycyrrhizate, which sequesters extracellular HMGB1, revealed a role for this alarmin in the regulation exerted by PARP-1 on the stimulating vs. tolerogenic function of DCs during colitis. Moreover, a higher percentage of CD11c DC from PARP-1KO mice expressed CD103, a marker associated with the ability of DC to induce Treg cells, compared with WT DC. Conversely, PARP-1KO DC were including a reduced percentage of CX3CR1+ DC, described to induce Th17 cells. These findings were observed in both splenic and colon lamina propria DC.
Collapse
Affiliation(s)
| | | | | | - Salvatore Cucchiara
- Department of Maternal Infantile and Urological Sciences, Sapienza University, Rome, Italy
| | - Laura Stronati
- Department of Molecular Medicine, Sapienza University, Rome, Italy
| | | |
Collapse
|
14
|
Zou ZP, Zhang XP, Zhang Q, Yin BC, Zhou Y, Ye BC. Genetically engineered bacteria as inflammatory bowel disease therapeutics. ENGINEERING MICROBIOLOGY 2024; 4:100167. [PMID: 39628589 PMCID: PMC11611042 DOI: 10.1016/j.engmic.2024.100167] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 06/18/2024] [Revised: 08/24/2024] [Accepted: 08/26/2024] [Indexed: 12/06/2024]
Abstract
Inflammatory bowel disease (IBD) is a chronic and recurrent disease caused by immune response disorders that disrupt the intestinal lumen symbiotic ecosystem and dysregulate mucosal immune functions. Current therapies available for IBD primarily focus on symptom management, making early diagnosis and prompt intervention challenging. The development of genetically engineered bacteria using synthetic biology presents a new strategy for addressing these challenges. In this review, we present recent breakthroughs in the field of engineered bacteria for the treatment and detection of IBD and describe how bacteria can be genetically modified to produce therapeutic molecules or execute diagnostic functions. In particular, we discuss the challenges faced in translating live bacterial therapeutics from bacterial design to delivery strategies for further clinical applications.
Collapse
Affiliation(s)
| | | | - Qian Zhang
- Laboratory of Biosystems and Microanalysis, State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, Shanghai, 200237, China
| | - Bin-Cheng Yin
- Laboratory of Biosystems and Microanalysis, State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, Shanghai, 200237, China
| | - Ying Zhou
- Laboratory of Biosystems and Microanalysis, State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, Shanghai, 200237, China
| | - Bang-Ce Ye
- Laboratory of Biosystems and Microanalysis, State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, Shanghai, 200237, China
| |
Collapse
|
15
|
Holm M, Stepanauskaitė L, Bäckström A, Birgersson M, Socciarelli F, Archer A, Stadler C, Williams C. Spatial profiling of the mouse colonic immune landscape associated with colitis and sex. Commun Biol 2024; 7:1595. [PMID: 39613949 DOI: 10.1038/s42003-024-07276-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2024] [Accepted: 11/15/2024] [Indexed: 12/01/2024] Open
Abstract
Inflammatory intestinal conditions are a major disease burden. Numerous factors shape the distribution of immune cells in the colon, but a spatial characterization of the homeostatic and inflamed colonic immune microenvironment is lacking. Here, we use the COMET platform for multiplex immunofluorescence to profile the infiltration of nine immune cell populations in mice of both sexes (N = 16) with full spatial context, including in regions of squamous metaplasia. Unsupervised clustering, neighborhood analysis, and manual quantification along the proximal-distal axis characterized the colonic immune landscape, quantified cell-cell interactions, and revealed sex differences. The distal colon was the most affected region during colitis, which was pronounced in males, who exhibited a sex-dependent increase of B cells and reduction of M2-like macrophages. Regions of squamous metaplasia exhibited strong infiltration of numerous immune cell populations, especially in males. Females exhibited more helper T cells and neutrophils at homeostasis and increased M2-like macrophage infiltration in the mid-colon upon colitis. Sex differences were corroborated by plasma cytokine profiles. Our results provide a foundation for future studies of inflammatory intestinal conditions.
Collapse
Affiliation(s)
- Matilda Holm
- SciLifeLab, Department of Protein Science, KTH Royal Institute of Technology, Solna, Sweden
- Division of Biosciences and Nutrition, Department of Medicine Huddinge, Karolinska Institutet, Huddinge, Sweden
| | - Lina Stepanauskaitė
- SciLifeLab, Department of Protein Science, KTH Royal Institute of Technology, Solna, Sweden
- Division of Biosciences and Nutrition, Department of Medicine Huddinge, Karolinska Institutet, Huddinge, Sweden
| | - Anna Bäckström
- SciLifeLab, Department of Protein Science, KTH Royal Institute of Technology, Solna, Sweden
| | - Madeleine Birgersson
- SciLifeLab, Department of Protein Science, KTH Royal Institute of Technology, Solna, Sweden
- Division of Biosciences and Nutrition, Department of Medicine Huddinge, Karolinska Institutet, Huddinge, Sweden
| | - Fabio Socciarelli
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, NY, USA
| | - Amena Archer
- SciLifeLab, Department of Protein Science, KTH Royal Institute of Technology, Solna, Sweden
- Division of Biosciences and Nutrition, Department of Medicine Huddinge, Karolinska Institutet, Huddinge, Sweden
| | - Charlotte Stadler
- SciLifeLab, Department of Protein Science, KTH Royal Institute of Technology, Solna, Sweden
| | - Cecilia Williams
- SciLifeLab, Department of Protein Science, KTH Royal Institute of Technology, Solna, Sweden.
- Division of Biosciences and Nutrition, Department of Medicine Huddinge, Karolinska Institutet, Huddinge, Sweden.
| |
Collapse
|
16
|
Heredia M, Barendregt DMH, Tindemans I, Klomberg RCW, Aardoom MA, Calado B, Costes LMM, Joosse ME, Hulleman-van Haaften DH, Tuk B, van Berkel LA, Kemos P, Ruemmele FM, Croft NM, Escher JC, de Ridder L, Samsom JN. Gut-homing and intestinal TIGIT negCD38 + memory T cells acquire an IL-12-induced, ex-Th17 pathogenic phenotype in a subgroup of Crohn's disease patients with a severe disease course. Mucosal Immunol 2024:S1933-0219(24)00116-8. [PMID: 39586377 DOI: 10.1016/j.mucimm.2024.11.008] [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: 06/01/2024] [Revised: 11/18/2024] [Accepted: 11/19/2024] [Indexed: 11/27/2024]
Abstract
CD4+ memory T cell (TM) reactivation drives chronicity in inflammatory bowel disease (IBD), including Crohn's disease (CD) and ulcerative colitis. Defects driving loss of TM regulation likely differ between patients but remain undefined. In health, approximately 40 % of circulating gut-homing CD38+TM express co-inhibitory receptor T-cell immunoreceptor with immunoglobulin and ITIM domains (TIGIT). TIGIT+CD38+TM have regulatory function while TIGITnegCD38+TM are enriched in IFN-γ-producing cells. We hypothesized TIGITnegCD38+TM are inflammatory and drive disease in a subgroup of IBD patients. We characterized TIGIT+CD38+TM in a uniquely large cohort of pediatric IBD patients from time of diagnosis into adulthood. Circulating TIGITnegCD38+TM frequencies were higher in a subgroup of therapy-naïve CD patients with high plasma IFN-γ and a more severe disease course. TIGITnegCD38+TM were highly enriched in HLA-DR+ and ex-Th17/Th1-like cells, high producers of IFN-γ. Cultures of healthy-adult-stimulated TM identified IL-12 as the only IBD-related inflammatory cytokine to drive the pathogenic ex-Th17-TIGITnegCD38+ phenotype. Moreover, IL12RB2 mRNA expression was higher in TIGITnegCD38+TM than TIGIT+CD38+TM, elevated in CD biopsies compared to controls, and correlated with severity of intestinal inflammation. Overall, we argue that in a subgroup of pediatric CD, increased IL-12 signaling drives reprogramming of Th17 to inflammatory Th1-like TIGITnegCD38+TM and causes more severe disease.
Collapse
Affiliation(s)
- Maud Heredia
- Laboratory of Pediatrics, Division Gastroenterology and Nutrition, Erasmus University Medical Center-Sophia Children's Hospital, Rotterdam, The Netherlands
| | - Daniëlle M H Barendregt
- Laboratory of Pediatrics, Division Gastroenterology and Nutrition, Erasmus University Medical Center-Sophia Children's Hospital, Rotterdam, The Netherlands
| | - Irma Tindemans
- Laboratory of Pediatrics, Division Gastroenterology and Nutrition, Erasmus University Medical Center-Sophia Children's Hospital, Rotterdam, The Netherlands
| | - Renz C W Klomberg
- Department of Pediatric Gastroenterology, Erasmus University Medical Center-Sophia Children's Hospital, Rotterdam, The Netherlands
| | - Martine A Aardoom
- Department of Pediatric Gastroenterology, Erasmus University Medical Center-Sophia Children's Hospital, Rotterdam, The Netherlands
| | - Beatriz Calado
- Laboratory of Pediatrics, Division Gastroenterology and Nutrition, Erasmus University Medical Center-Sophia Children's Hospital, Rotterdam, The Netherlands
| | - Léa M M Costes
- Laboratory of Pediatrics, Division Gastroenterology and Nutrition, Erasmus University Medical Center-Sophia Children's Hospital, Rotterdam, The Netherlands
| | - Maria E Joosse
- Laboratory of Pediatrics, Division Gastroenterology and Nutrition, Erasmus University Medical Center-Sophia Children's Hospital, Rotterdam, The Netherlands
| | - Daniëlle H Hulleman-van Haaften
- Laboratory of Pediatrics, Division Gastroenterology and Nutrition, Erasmus University Medical Center-Sophia Children's Hospital, Rotterdam, The Netherlands
| | - Bastiaan Tuk
- Laboratory of Pediatrics, Division Gastroenterology and Nutrition, Erasmus University Medical Center-Sophia Children's Hospital, Rotterdam, The Netherlands
| | - Lisette A van Berkel
- Laboratory of Pediatrics, Division Gastroenterology and Nutrition, Erasmus University Medical Center-Sophia Children's Hospital, Rotterdam, The Netherlands
| | - Polychronis Kemos
- Centre for Immunobiology, Blizard Institute, Queen Mary University of London, London, UK
| | - Frank M Ruemmele
- Department of Pediatric Gastroenterology, Necker-Enfants Malades University Hospital, Institut Imagine, AP-HP, Université de Paris, Paris, France
| | - Nicholas M Croft
- Centre for Immunobiology, Blizard Institute, Queen Mary University of London, London, UK
| | - Johanna C Escher
- Department of Pediatric Gastroenterology, Erasmus University Medical Center-Sophia Children's Hospital, Rotterdam, The Netherlands
| | - Lissy de Ridder
- Department of Pediatric Gastroenterology, Erasmus University Medical Center-Sophia Children's Hospital, Rotterdam, The Netherlands
| | - Janneke N Samsom
- Laboratory of Pediatrics, Division Gastroenterology and Nutrition, Erasmus University Medical Center-Sophia Children's Hospital, Rotterdam, The Netherlands.
| |
Collapse
|
17
|
Kojima F, Hioki Y, Sekiya H, Kashiwagi H, Iizuka Y, Eto K, Maehana S, Kawakami F, Kubo M, Ishibashi H, Ichikawa T. Microsomal Prostaglandin E Synthase-1 Controls Colonic Prostaglandin E 2 Production and Exerts a Protective Effect on Colitis Induced by Trinitrobenzene Sulfonic Acid in Mice. Int J Mol Sci 2024; 25:12326. [PMID: 39596393 PMCID: PMC11594388 DOI: 10.3390/ijms252212326] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2024] [Accepted: 11/15/2024] [Indexed: 11/28/2024] Open
Abstract
Microsomal prostaglandin E synthase-1 (mPGES-1) is an isozyme of the prostaglandin (PG) E synthase that acts downstream of cyclooxygenase and catalyzes the conversion of PGH2 to PGE2. The impact of genetic deletion of mPGES-1 on the development of 2,4,6-trinitrobenzene sulfonic acid (TNBS)-induced colitis, a well-established model of inflammatory bowel disease (IBD), was investigated in this study. After administration of TNBS, mice deficient in mPGES-1 (mPGES-1-/- mice) showed more severe colitis than did wild-type (WT) mice. Histological examination revealed that mPGES-1-/- mice had markedly exacerbated symptoms of colitis. mPGES-1 expression was detectable in the colons of WT mice at both the mRNA and protein levels. Lack of mPGES-1 resulted in marked reduction of colonic PGE2 production. Our study also showed a significant increase in colonic expression of interleukin-17A (IL-17A), as well as interferon γ (IFNγ) and tumor necrosis factor α, during colitis in mPGES-1-/- mice compared with that in WT mice. Furthermore, loss of mPGES-1 increased the populations of IL-17A-producing T-helper (Th) 17 and IFNγ-producing Th1 cells in mesenteric lymph nodes. These results suggest that mPGES-1 is the main enzyme responsible for colonic PGE2 production and deficiency of mPGES-1 facilitates the development of colitis and T-cell-mediated immunity. mPGES-1 might, therefore, impact T-cell-related immune response associated with IBD.
Collapse
Affiliation(s)
- Fumiaki Kojima
- Department of Pharmacology, Kitasato University School of Allied Health Sciences, Sagamihara 252-0373, Japan
- Department of Regulation Biochemistry, Kitasato University Graduate School of Medical Sciences, Sagamihara 252-0373, Japan; (F.K.); (T.I.)
- Regenerative Medicine and Cell Design Research Facility, Sagamihara 252-0373, Japan; (Y.I.); (K.E.); (S.M.); (M.K.); (H.I.)
| | - Yuka Hioki
- Department of Pharmacology, Kitasato University School of Allied Health Sciences, Sagamihara 252-0373, Japan
- Department of Regulation Biochemistry, Kitasato University Graduate School of Medical Sciences, Sagamihara 252-0373, Japan; (F.K.); (T.I.)
| | - Hiroki Sekiya
- Department of Pharmacology, Kitasato University School of Allied Health Sciences, Sagamihara 252-0373, Japan
- Department of Regulation Biochemistry, Kitasato University Graduate School of Medical Sciences, Sagamihara 252-0373, Japan; (F.K.); (T.I.)
| | - Hitoshi Kashiwagi
- Faculty of Pharmaceutical Sciences, Hokkaido University, Sapporo 060-0812, Japan;
| | - Yoshiko Iizuka
- Regenerative Medicine and Cell Design Research Facility, Sagamihara 252-0373, Japan; (Y.I.); (K.E.); (S.M.); (M.K.); (H.I.)
- Department of Public Health, Kitasato University Graduate School of Medical Sciences, Sagamihara 252-0373, Japan
| | - Kei Eto
- Regenerative Medicine and Cell Design Research Facility, Sagamihara 252-0373, Japan; (Y.I.); (K.E.); (S.M.); (M.K.); (H.I.)
- Department of Physiology, Kitasato University School of Allied Health Sciences, Sagamihara 252-0373, Japan
| | - Shotaro Maehana
- Regenerative Medicine and Cell Design Research Facility, Sagamihara 252-0373, Japan; (Y.I.); (K.E.); (S.M.); (M.K.); (H.I.)
- Department of Environmental Microbiology, Kitasato University Graduate School of Medical Sciences, Sagamihara 252-0373, Japan
| | - Fumitaka Kawakami
- Department of Regulation Biochemistry, Kitasato University Graduate School of Medical Sciences, Sagamihara 252-0373, Japan; (F.K.); (T.I.)
- Regenerative Medicine and Cell Design Research Facility, Sagamihara 252-0373, Japan; (Y.I.); (K.E.); (S.M.); (M.K.); (H.I.)
| | - Makoto Kubo
- Regenerative Medicine and Cell Design Research Facility, Sagamihara 252-0373, Japan; (Y.I.); (K.E.); (S.M.); (M.K.); (H.I.)
- Department of Environmental Microbiology, Kitasato University Graduate School of Medical Sciences, Sagamihara 252-0373, Japan
| | - Hitoshi Ishibashi
- Regenerative Medicine and Cell Design Research Facility, Sagamihara 252-0373, Japan; (Y.I.); (K.E.); (S.M.); (M.K.); (H.I.)
- Department of Physiology, Kitasato University School of Allied Health Sciences, Sagamihara 252-0373, Japan
| | - Takafumi Ichikawa
- Department of Regulation Biochemistry, Kitasato University Graduate School of Medical Sciences, Sagamihara 252-0373, Japan; (F.K.); (T.I.)
- Regenerative Medicine and Cell Design Research Facility, Sagamihara 252-0373, Japan; (Y.I.); (K.E.); (S.M.); (M.K.); (H.I.)
| |
Collapse
|
18
|
Xu S, Peng Y, Yang K, Liu S, He Z, Huang J, Xiao R, Liu J, Yan Z, Lian Z, Pan H, Chen J, Shi J, Yao X, Deng H. PROTAC based STING degrader attenuates acute colitis by inhibiting macrophage M1 polarization and intestinal epithelial cells pyroptosis mediated by STING-NLRP3 axis. Int Immunopharmacol 2024; 141:112990. [PMID: 39223062 DOI: 10.1016/j.intimp.2024.112990] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2024] [Revised: 08/17/2024] [Accepted: 08/18/2024] [Indexed: 09/04/2024]
Abstract
Inflammatory bowel diseases (IBDs) are chronic, relapsing, and inflammatory disorders of the gastrointestinal tract characterized by abnormal immune responses. Recently, STING has emerged as a promising therapeutic target for various autoinflammatory diseases. However, few STING-selective small molecules have been investigated as novel strategies for IBD. In this study, we sought to examine the effects of PROTAC-based STING degrader SP23 on acute colitis and explore its underlying mechanism. SP23 treatment notably alleviates dextran sulfate sodium (DSS)-induced colitis. Pharmacological degradation of STING significantly reduced the production of inflammatory cytokines, such as TNF-α, IL-1β, and IL-6, and inhibited macrophage polarization towards the M1 type. Furthermore, SP23 administration decreased the loss of tight junction proteins, including ZO-1, occludin, and claudin-1, and downregulated STING and NLRP3 signaling pathways in intestinal inflammation. In vitro, STING activated NLRP3 inflammasome-mediated pyroptosis in intestinal epithelial cells, which could be abrogated by SP23 and STING siRNA intervention. In conclusion, these findings provide new evidence for STING as a novel therapeutic target for IBD, and reveal that hyperactivation of STING could exaggerate colitis by inducing NLRP3/Caspase-1/GSDMD axis mediated intestinal epithelial cells pyroptosis.
Collapse
Affiliation(s)
- Shuai Xu
- Department of General Surgery, Nanfang Hospital, Southern Medical University, Guangdong Provincial Key Laboratory of Precision Medicine for Gastrointestinal Tumor, Guangzhou, Guangdong, China
| | - Yifeng Peng
- Department of General Surgery, Nanfang Hospital, Southern Medical University, Guangdong Provincial Key Laboratory of Precision Medicine for Gastrointestinal Tumor, Guangzhou, Guangdong, China
| | - Kai Yang
- Department of General Surgery, Nanfang Hospital, Southern Medical University, Guangdong Provincial Key Laboratory of Precision Medicine for Gastrointestinal Tumor, Guangzhou, Guangdong, China
| | - Sheng Liu
- Department of General Surgery, Nanfang Hospital, Southern Medical University, Guangdong Provincial Key Laboratory of Precision Medicine for Gastrointestinal Tumor, Guangzhou, Guangdong, China
| | - Zhanke He
- Department of General Surgery, Nanfang Hospital, Southern Medical University, Guangdong Provincial Key Laboratory of Precision Medicine for Gastrointestinal Tumor, Guangzhou, Guangdong, China
| | - Junli Huang
- Department of Pharmacy, The People's Hospital of Guangxi Zhuang Autonomous Region & Guangxi Academy of Medical Sciences, Nanning, Guangxi 530021, China
| | - Ruipei Xiao
- Department of General Surgery, Nanfang Hospital, Southern Medical University, Guangdong Provincial Key Laboratory of Precision Medicine for Gastrointestinal Tumor, Guangzhou, Guangdong, China
| | - Jin Liu
- Laboratory of Tropical Biological Resources of Ministry of Education, School of Pharmaceutical Sciences, Hainan University, Haikou 570228, China
| | - Ziyan Yan
- Department of General Surgery, Nanfang Hospital, Southern Medical University, Guangdong Provincial Key Laboratory of Precision Medicine for Gastrointestinal Tumor, Guangzhou, Guangdong, China
| | - Zhiying Lian
- Department of General Surgery, Nanfang Hospital, Southern Medical University, Guangdong Provincial Key Laboratory of Precision Medicine for Gastrointestinal Tumor, Guangzhou, Guangdong, China
| | - Huayang Pan
- Department of General Surgery, Nanfang Hospital, Southern Medical University, Guangdong Provincial Key Laboratory of Precision Medicine for Gastrointestinal Tumor, Guangzhou, Guangdong, China
| | - Jianjun Chen
- School of Pharmaceutical Sciences, Guangdong Provincial Key Laboratory of New Drug Screening, Southern Medical University, Guangzhou 510515, China
| | - Jiaolong Shi
- Department of General Surgery, Nanfang Hospital, Southern Medical University, Guangdong Provincial Key Laboratory of Precision Medicine for Gastrointestinal Tumor, Guangzhou, Guangdong, China.
| | - Xingxing Yao
- Department of General Surgery, Nanfang Hospital, Southern Medical University, Guangdong Provincial Key Laboratory of Precision Medicine for Gastrointestinal Tumor, Guangzhou, Guangdong, China.
| | - Haijun Deng
- Department of General Surgery, Nanfang Hospital, Southern Medical University, Guangdong Provincial Key Laboratory of Precision Medicine for Gastrointestinal Tumor, Guangzhou, Guangdong, China.
| |
Collapse
|
19
|
Li X, Yao X, Wen J, Chen Q, Zhu Z, Zhang X, Wang S, Lan W, Huang Y, Tang S, Zhou X, Han X, Zhang T. The application of sphingomyelin in mediating the causal role of the T-cell surface glycoprotein CD5 in Crohn's disease: A two-step Mendelian randomization study. Medicine (Baltimore) 2024; 103:e40513. [PMID: 39560554 PMCID: PMC11576039 DOI: 10.1097/md.0000000000040513] [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] [Indexed: 11/20/2024] Open
Abstract
To examine the possible causative association between Crohn disease (CD) and the T-cell surface glycoprotein CD5 and to ascertain whether sphingomyelin (SM) functions as a mediator. We conducted a two-step Mendelian randomization (MR) study to further explore the pathogenesis of Crohn and its related targets. MR study was performed on CD5 and CD using summary-level data from a genome-wide association study. Additionally, by employing a two-step MR study method, we determined that SM might mediate the causal effect of CD5 on CD. There was a favorable correlation between the surface glycoprotein CD5 on T cells and vulnerability to CD, and SM mediated the causal effect of CD5 on CD (the mediating effect accounts for 9.2%). Our study revealed that CD5 and CD are causally related, with SM mediating a small fraction of the impact (approximately 9.2%). The mediating function of SM in the link between CD5 and CD is anticipated to be realized through the regulation of immune cell transportation, apoptosis of intestinal barrier cells, and maintenance of the intestinal microenvironment.
Collapse
Affiliation(s)
- Xiao Li
- The graduate School of Guangxi University of Chinese Medicine, Nanning, Guangxi, China
| | - Xin Yao
- The graduate School of Guangxi University of Chinese Medicine, Nanning, Guangxi, China
| | - Jieying Wen
- The graduate School of Guangxi University of Chinese Medicine, Nanning, Guangxi, China
| | - Qiaoling Chen
- The graduate School of Guangxi University of Chinese Medicine, Nanning, Guangxi, China
| | - Ziming Zhu
- The graduate School of Guangxi University of Chinese Medicine, Nanning, Guangxi, China
| | - Xinyue Zhang
- Ruikang Hospital Affiliated to Guangxi University of Chinese Medicine, Nanning, Guangxi, China
| | - Song Wang
- The graduate School of Guangxi University of Chinese Medicine, Nanning, Guangxi, China
| | - Weixuan Lan
- The graduate School of Guangxi University of Chinese Medicine, Nanning, Guangxi, China
| | - Yunsi Huang
- The graduate School of Guangxi University of Chinese Medicine, Nanning, Guangxi, China
| | - Shanneng Tang
- The graduate School of Guangxi University of Chinese Medicine, Nanning, Guangxi, China
| | - Xuan Zhou
- The graduate School of Guangxi University of Chinese Medicine, Nanning, Guangxi, China
| | - Xuedong Han
- The graduate School of Guangxi University of Chinese Medicine, Nanning, Guangxi, China
| | - Tao Zhang
- Ruikang Hospital Affiliated to Guangxi University of Chinese Medicine, Nanning, Guangxi, China
| |
Collapse
|
20
|
Li J, Wu Z, Wu Y, Hu X, Yang J, Zhu D, Wu M, Li X, Bentum-Ennin L, Wanglai H. IL-22, a vital cytokine in autoimmune diseases. Clin Exp Immunol 2024; 218:242-263. [PMID: 38651179 PMCID: PMC11557150 DOI: 10.1093/cei/uxae035] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2024] [Revised: 03/05/2024] [Accepted: 04/19/2024] [Indexed: 04/25/2024] Open
Abstract
Interleukin-22 (IL-22) is a vital cytokine that is dysregulated in various autoimmune conditions including rheumatoid arthritis (RA), multiple sclerosis (MS), and Alzheimer's disease (AD). As the starting point for the activation of numerous signaling pathways, IL-22 plays an important role in the initiation and development of autoimmune diseases. Specifically, imbalances in IL-22 signaling can interfere with other signaling pathways, causing cross-regulation of target genes which ultimately leads to the development of immune disorders. This review delineates the various connections between the IL-22 signaling pathway and autoimmune disease, focusing on the latest understanding of the cellular sources of IL-22 and its effects on various cell types. We further explore progress with pharmacological interventions related to targeting IL-22, describing how such therapeutic strategies promise to usher in a new era in the treatment of autoimmune disease.
Collapse
Affiliation(s)
- Jiajin Li
- The Second Clinical School of Medicine, Anhui Medical University, Hefei, China
| | - Zhen Wu
- The First Clinical School of Medicine, Anhui Medical University, Hefei, China
| | - Yuxin Wu
- The First Clinical School of Medicine, Anhui Medical University, Hefei, China
| | - XinYu Hu
- The Second Clinical School of Medicine, Anhui Medical University, Hefei, China
| | - Jun Yang
- The Second Clinical School of Medicine, Anhui Medical University, Hefei, China
| | - Dacheng Zhu
- The First Clinical School of Medicine, Anhui Medical University, Hefei, China
| | - Mingyue Wu
- The School of pharmacy, Anhui Medical University, Hefei, China
| | - Xin Li
- The School of pharmacy, Anhui Medical University, Hefei, China
| | | | - Hu Wanglai
- The School of Basic Medical Sciences, Anhui Medical University, Hefei, China
| |
Collapse
|
21
|
Wu Q, Liu K, Hou R, Wu X, Ruan X, Wang M, Sun Z, Meng L, Dai G, Li C, Wu J, Mu G. Integrative Lipid Pseudotargeted Metabolomics and Amino Acids Targeted Metabolomics Unravel the Therapeutic Mechanism of Rhizoma Paridis Saponins on Experimental Colitis of Damp-Heat Type. Drug Des Devel Ther 2024; 18:5087-5108. [PMID: 39554759 PMCID: PMC11568858 DOI: 10.2147/dddt.s476494] [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: 07/19/2024] [Accepted: 10/14/2024] [Indexed: 11/19/2024] Open
Abstract
Purpose Inflammatory bowel disease (IBD) is a serious disease that affects the metabolism and inflammatory responses of human beings. From the perspective of traditional Chinese medicine, damp-heat syndrome is one of the main syndromes of IBD. Rhizoma Paridis, also known as the root of Paris polyphylla, a well-known herbal medicine used in China, is used to treat IBD with damp-heat syndrome (IBD-DH). However, uncertainty still exists regarding the underlying mechanisms and the impact of Rhizoma Paridis on IBD-DH. Methods The rats in the model (DAT) and medication administration (Rhizoma Paridis total saponins (RPTS) and Pennogenin (PN)) groups were given a high temperature and high humidity environment, high fat and high sugar diet combined with 2,4,6-trinitrobenzene sulfonic acid (TNBS) to establish the model of experimental colitis of damp-heat type, and the normal control group (RNC) rats were given a normal diet at normal temperature and humidity. Damp-heat control group (DNC) was set with the same condition as DAT without TNBS. Hematoxylin-Eosin (HE) staining was used to observe the histopathological morphology of the rat colorectum. The expression of the metabolism-related genes (Phospholipase A2 (sPLA2, cPLA2), and phosphatidylethanolamine N-methyltransferase (PEMT)) was assessed by using real-time quantitative PCR analysis (RT-qPCR). And the levels of the metabolism-related proteins (sPLA2, cPLA2), S100A8/9, Arg-1, and cytokines were detected by enzyme-linked immunosorbent assay (ELISA) kit. To investigate lipids and amino acids which closely associated with the IBD and IBD-DH, lipid pseudotargeted metabolomics with UHPLC-TQ/MS analysis method, as well as targeted quantitative amino acid analysis were performed. Results Our data showed that RPTS (50 mg/kg) and PN (20 mg/kg) significantly ameliorated the severity of TNBS-induced colitis and downregulated the levels of circulating proinflammatory cytokines. Compared with RNC group, lipid pseudotargeted metabolomics demonstrated that glycerophospholipids, sphingolipids, carnitine, and glycerolipids were the four most perturbed lipid classes, and amino acids targeted metabolomics demonstrated that serine, N-acetylneuraminic acid, histidine, proline, taurine, and kynurenine changed significantly in DAT group . Correlation analyses showed tight associations between most of differential metabolites and proinflammatory cytokines. RPTS and PN both regulated glycerophospholipid metabolism and sphingolipid metabolism. However, both of them did not have a significant effect on amino acid modulation. RPTS and PN potentially regulated sPLA2, cPLA2, and PEMT. Conclusion These results showed that RPTS (50 mg/kg) and PN (20 mg/kg) effectively alleviated rats' colitis of damp-heat type, affected cytokines, and altered lipid metabolism without significant modulation on amino acid metabolism.
Collapse
Affiliation(s)
- Qi Wu
- Department of Traditional Chinese Medicine, Nanjing Drum Tower Hospital, Nanjing Drum Tower Hospital Clinical College of Nanjing University of Chinese Medicine, Nanjing, 210008, People’s Republic of China
| | - Kexin Liu
- Department of Gastroenterology, The 981th Hospital of the Joint Logistics Support Force of the Chinese People’s Liberation Army, Chengde, Hebei, 067000, People’s Republic of China
| | - Ruijuan Hou
- Department of Traditional Chinese Medicine, Nanjing Drum Tower Hospital, Nanjing Drum Tower Hospital Clinical College of Nanjing University of Chinese Medicine, Nanjing, 210008, People’s Republic of China
| | - Xingxing Wu
- Department of Gastroenterology, Henan Province Hospital of Traditional Chinese Medicine, Zhengzhou, 450053, People’s Republic of China
| | - Xiaoyu Ruan
- Internal Medicine Department, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, 210029, People’s Republic of China
| | - Mao Wang
- Ethics Committee, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, 210029, People’s Republic of China
| | - Zhiting Sun
- Institute of Chinese Medicine of Nanjing University, Nanjing Drum Tower Hospital, Drum Tower Clinical Medicine College of Nanjing University of Chinese Medicine, Nanjing, 210008, People’s Republic of China
| | - Lingchang Meng
- Institute of Chinese Medicine of Nanjing University, Nanjing Drum Tower Hospital, Drum Tower Clinical Medicine College of Nanjing University of Chinese Medicine, Nanjing, 210008, People’s Republic of China
| | - Guoliang Dai
- Clinical Pharmacology Laboratory, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, 210029, People’s Republic of China
| | - Changyin Li
- Clinical Pharmacology Laboratory, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, 210029, People’s Republic of China
| | - Jing Wu
- Department of Traditional Chinese Medicine, Nanjing Drum Tower Hospital, Nanjing Drum Tower Hospital Clinical College of Nanjing University of Chinese Medicine, Nanjing, 210008, People’s Republic of China
- Institute of Chinese Medicine of Nanjing University, Nanjing Drum Tower Hospital, Drum Tower Clinical Medicine College of Nanjing University of Chinese Medicine, Nanjing, 210008, People’s Republic of China
| | - Genglin Mu
- Institute of Chinese Medicine of Nanjing University, Nanjing Drum Tower Hospital, Drum Tower Clinical Medicine College of Nanjing University of Chinese Medicine, Nanjing, 210008, People’s Republic of China
| |
Collapse
|
22
|
Ouboter LF, Lindelauf C, Jiang Q, Schreurs M, Abdelaal TR, Luk SJ, Barnhoorn MC, Hueting WE, Han-Geurts IJ, Peeters KCMJ, Holman FA, Koning F, van der Meulen-de Jong AE, Pascutti MF. Activated HLA-DR+CD38+ Effector Th1/17 Cells Distinguish Crohn's Disease-associated Perianal Fistulas from Cryptoglandular Fistulas. Inflamm Bowel Dis 2024; 30:2146-2161. [PMID: 38776553 DOI: 10.1093/ibd/izae103] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/05/2023] [Indexed: 05/25/2024]
Abstract
BACKGROUND Perianal fistulas are a debilitating complication of Crohn's disease (CD). Due to unknown reasons, CD-associated fistulas are in general more difficult to treat than cryptoglandular fistulas (non-CD-associated). Understanding the immune cell landscape is a first step towards the development of more effective therapies for CD-associated fistulas. In this work, we characterized the composition and spatial localization of disease-associated immune cells in both types of perianal fistulas by high-dimensional analyses. METHODS We applied single-cell mass cytometry (scMC), spectral flow cytometry (SFC), and imaging mass cytometry (IMC) to profile the immune compartment in CD-associated perianal fistulas and cryptoglandular fistulas. An exploratory cohort (CD fistula, n = 10; non-CD fistula, n = 5) was analyzed by scMC to unravel disease-associated immune cell types. SFC was performed on a second fistula cohort (CD, n = 10; non-CD, n = 11) to comprehensively phenotype disease-associated T helper (Th) cells. IMC was used on a third cohort (CD, n = 5) to investigate the spatial distribution/interaction of relevant immune cell subsets. RESULTS Our analyses revealed that activated HLA-DR+CD38+ effector CD4+ T cells with a Th1/17 phenotype were significantly enriched in CD-associated compared with cryptoglandular fistulas. These cells, displaying features of proliferation, regulation, and differentiation, were also present in blood, and colocalized with other CD4+ T cells, CCR6+ B cells, and macrophages in the fistula tracts. CONCLUSIONS Overall, proliferating activated HLA-DR+CD38+ effector Th1/17 cells distinguish CD-associated from cryptoglandular perianal fistulas and are a promising biomarker in blood to discriminate between these 2 fistula types. Targeting HLA-DR and CD38-expressing CD4+ T cells may offer a potential new therapeutic strategy for CD-related fistulas.
Collapse
Affiliation(s)
- Laura F Ouboter
- Department of Gastroenterology and Hepatology, Leiden University Medical Center, Leiden, the Netherlands
- Department of Immunology, Leiden University Medical Center, Leiden, the Netherlands
| | - Ciska Lindelauf
- Department of Immunology, Leiden University Medical Center, Leiden, the Netherlands
| | - Qinyue Jiang
- Department of Immunology, Leiden University Medical Center, Leiden, the Netherlands
| | - Mette Schreurs
- Department of Immunology, Leiden University Medical Center, Leiden, the Netherlands
| | - Tamim R Abdelaal
- Department of Radiology, Leiden University Medical Center, Leiden, the Netherlands
- Bioinformatics Lab, Delft University of Technology, Delft, the Netherlands
- Systems and Biomedical Engineering Department, Faculty of Engineering Cairo University, Giza, Egypt
| | - Sietse J Luk
- Department of Hematology, Leiden University Medical Center, Leiden, the Netherlands
| | - Marieke C Barnhoorn
- Department of Gastroenterology and Hepatology, Leiden University Medical Center, Leiden, the Netherlands
| | - Willem E Hueting
- Department of Surgery, Alrijne hospital, Leiderdorp, the Netherlands
| | | | - Koen C M J Peeters
- Department of Surgery, Leiden University Medical Center, Leiden, the Netherlands
| | - Fabian A Holman
- Department of Surgery, Leiden University Medical Center, Leiden, the Netherlands
| | - Frits Koning
- Department of Immunology, Leiden University Medical Center, Leiden, the Netherlands
| | | | | |
Collapse
|
23
|
Kwong AC, Ordovas-Montanes J. Deconstructing inflammatory memory across tissue set points using cell circuit motifs. J Allergy Clin Immunol 2024; 154:1095-1105. [PMID: 39341577 DOI: 10.1016/j.jaci.2024.09.014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2024] [Revised: 09/22/2024] [Accepted: 09/23/2024] [Indexed: 10/01/2024]
Abstract
Tissue ecosystems are cellular communities that maintain set points through a network of intercellular interactions. We position health and chronic inflammatory disease as alternative stable set points that are (1) robust to perturbation and (2) capable of adaptation and memory. Inflammatory memory, which is the storage of prior experience to durably influence future responsiveness, is central to how tissue ecosystems may be pushed past tipping points that stabilize disease over health. Here, we develop a reductionist framework of circuit motifs that recur in tissue set points. In type 2 immunity, we distinctly find the emergence of 2-cell positive feedback motifs. In contrast, directional motif relays and 3-cell networks feature more prominently in type 1 and 17 responses. We propose that these differences guide the ecologic networks established after surpassing tipping points and associate closely with therapeutic responsiveness. We highlight opportunities to improve our current knowledge of how circuit motifs interact when building toward tissue-level networks across adaptation and memory. By developing new tools for circuit motif nomination and applying them to temporal profiling of tissue ecosystems, we hope to dissect the stability of the chronic inflammatory set point and open therapeutic avenues for rewriting memory to restore health.
Collapse
Affiliation(s)
- Andrew C Kwong
- Division of Gastroenterology, Hepatology, and Nutrition, Boston Children's Hospital, Boston, Mass; Broad Institute of MIT and Harvard, Cambridge, Mass; Program in Biological and Biomedical Sciences, Harvard Medical School, Boston, Mass
| | - Jose Ordovas-Montanes
- Division of Gastroenterology, Hepatology, and Nutrition, Boston Children's Hospital, Boston, Mass; Broad Institute of MIT and Harvard, Cambridge, Mass; Ragon Institute of Massachusetts General Hospital, MIT and Harvard, Boston; Program in Immunology, Harvard Medical School, Boston, Mass; Harvard Stem Cell Institute, Harvard University, Cambridge, Mass.
| |
Collapse
|
24
|
Menescal-de-Oliveira L, Brentegani MR, Teixeira FP, Giusti H, Saia RS. Immune-mediated impairment of tonic immobility defensive behavior in an experimental model of colonic inflammation. Pflugers Arch 2024; 476:1743-1760. [PMID: 39218820 DOI: 10.1007/s00424-024-03011-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2024] [Revised: 08/05/2024] [Accepted: 08/12/2024] [Indexed: 09/04/2024]
Abstract
Ulcerative colitis has been associated with psychological distress and an aberrant immune response. The immunomodulatory role of systemic cytokines produced during experimental intestinal inflammation in tonic immobility (TI) defensive behavior remains unknown. The present study characterized the TI defensive behavior of guinea pigs subjected to colitis induction at the acute stage and after recovery from intestinal mucosa injury. Moreover, we investigated whether inflammatory mediators (tumor necrosis factor (TNF)-α, interleukin (IL)-1β, IL-8, IL-10, and prostaglandins) act on the mesencephalic nucleus, periaqueductal gray matter (PAG). Colitis was induced in guinea pigs by intrarectal administration of acetic acid. The TI defensive behavior, histology, cytokine production, and expression of c-FOS, IBA-1, and cyclooxygenase (COX)-2 in PAG were evaluated. Colitis reduced the duration of TI episodes from the first day, persisting throughout the 7-day experimental period. Neuronal c-FOS immunoreactivity was augmented in both columns of the PAG (ventrolateral (vlPAG) and dorsal), but there were no changes in IBA-1 expression. Dexamethasone, infliximab, and parecoxib treatments increased the duration of TI episodes, suggesting a modulatory role of peripheral inflammatory mediators in this behavior. Immunoneutralization of TNF-α, IL-1β, and IL-8 in the vlPAG reversed all effects produced by colitis. In contrast, IL-10 neutralization further reduced the duration of TI episodes. Our results reveal that peripherally produced inflammatory mediators during colitis may modulate neuronal functioning in mesencephalic structures such as vlPAG.
Collapse
Affiliation(s)
- Leda Menescal-de-Oliveira
- Department of Physiology, Ribeirão Preto Medical School, University of São Paulo, Avenida Dos Bandeirantes, 3900, Ribeirão Preto, São Paulo, 14049-900, Brazil
| | - Mariulza Rocha Brentegani
- Department of Physiology, Ribeirão Preto Medical School, University of São Paulo, Avenida Dos Bandeirantes, 3900, Ribeirão Preto, São Paulo, 14049-900, Brazil
| | - Fernanda Pincelli Teixeira
- Department of Physiology, Ribeirão Preto Medical School, University of São Paulo, Avenida Dos Bandeirantes, 3900, Ribeirão Preto, São Paulo, 14049-900, Brazil
| | - Humberto Giusti
- Department of Physiology, Ribeirão Preto Medical School, University of São Paulo, Avenida Dos Bandeirantes, 3900, Ribeirão Preto, São Paulo, 14049-900, Brazil
| | - Rafael Simone Saia
- Department of Physiology, Ribeirão Preto Medical School, University of São Paulo, Avenida Dos Bandeirantes, 3900, Ribeirão Preto, São Paulo, 14049-900, Brazil.
| |
Collapse
|
25
|
Li S, Peng H, Sun Y, Yang J, Wang J, Bai F, Peng C, Fang S, Cai H, Chen G. Yeast β-glucan attenuates dextran sulfate sodium-induced colitis: Involvement of gut microbiota and short-chain fatty acids. Int J Biol Macromol 2024; 280:135846. [PMID: 39307486 DOI: 10.1016/j.ijbiomac.2024.135846] [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: 03/25/2024] [Revised: 09/10/2024] [Accepted: 09/18/2024] [Indexed: 09/29/2024]
Abstract
Yeast β-glucan intervention offers a promising strategy for managing colitis; however, the mechanisms remain unknown. In the present work, the protective effects of yeast β-glucan on DSS-induced colitis in mice was evaluated, focusing on its interaction with gut microbiota. The result showed yeast β-glucan significantly alleviated colitis symptoms, evidenced by reduced weight loss, lower disease activity index (DAI) scores, and minimized intestinal damage. It enhanced intestinal barrier integrity via upregulation of tight junction proteins, suppressed lipopolysaccharide (LPS) release, and decreased pro-inflammatory cytokines production. Additionally, yeast β-glucan boosted short-chain fatty acids (SCFAs) production, and activated their receptors, increased the relative abundances of beneficial microbes like Lactobacillus and Lachnospiraceae_UCG-006. Transcriptomic analyses suggest that yeast β-glucan mitigates inflammation by downregulating gene expression related to IL-17 pathway. Our findings highlight potential of yeast β-glucan as a therapeutic agent for colitis through modulation of gut microbiota and inflammatory responses.
Collapse
Affiliation(s)
- Sichen Li
- State Key Laboratory of Tea Plant Biology and Utilization, Anhui Provincial Key Laboratory of Food Safety Monitoring and Quality Control, School of Tea & Food Science and Technology, Anhui Agricultural University, Hefei 230036, Anhui, PR China; Joint Research Center for Food Nutrition and Health of IHM, Anhui Agricultural University, Hefei 230036, Anhui, PR China
| | - Huihui Peng
- State Key Laboratory of Tea Plant Biology and Utilization, Anhui Provincial Key Laboratory of Food Safety Monitoring and Quality Control, School of Tea & Food Science and Technology, Anhui Agricultural University, Hefei 230036, Anhui, PR China; Joint Research Center for Food Nutrition and Health of IHM, Anhui Agricultural University, Hefei 230036, Anhui, PR China
| | - Yuning Sun
- State Key Laboratory of Tea Plant Biology and Utilization, Anhui Provincial Key Laboratory of Food Safety Monitoring and Quality Control, School of Tea & Food Science and Technology, Anhui Agricultural University, Hefei 230036, Anhui, PR China; Joint Research Center for Food Nutrition and Health of IHM, Anhui Agricultural University, Hefei 230036, Anhui, PR China
| | - Jiali Yang
- State Key Laboratory of Tea Plant Biology and Utilization, Anhui Provincial Key Laboratory of Food Safety Monitoring and Quality Control, School of Tea & Food Science and Technology, Anhui Agricultural University, Hefei 230036, Anhui, PR China; Joint Research Center for Food Nutrition and Health of IHM, Anhui Agricultural University, Hefei 230036, Anhui, PR China
| | - Juan Wang
- State Key Laboratory of Tea Plant Biology and Utilization, Anhui Provincial Key Laboratory of Food Safety Monitoring and Quality Control, School of Tea & Food Science and Technology, Anhui Agricultural University, Hefei 230036, Anhui, PR China; Joint Research Center for Food Nutrition and Health of IHM, Anhui Agricultural University, Hefei 230036, Anhui, PR China
| | - Fuqing Bai
- State Key Laboratory of Tea Plant Biology and Utilization, Anhui Provincial Key Laboratory of Food Safety Monitoring and Quality Control, School of Tea & Food Science and Technology, Anhui Agricultural University, Hefei 230036, Anhui, PR China; Joint Research Center for Food Nutrition and Health of IHM, Anhui Agricultural University, Hefei 230036, Anhui, PR China
| | - Chuanyi Peng
- State Key Laboratory of Tea Plant Biology and Utilization, Anhui Provincial Key Laboratory of Food Safety Monitoring and Quality Control, School of Tea & Food Science and Technology, Anhui Agricultural University, Hefei 230036, Anhui, PR China; Joint Research Center for Food Nutrition and Health of IHM, Anhui Agricultural University, Hefei 230036, Anhui, PR China
| | - Shuzhen Fang
- The First Aliated Hospital of Anhui University of Chinese Medicine, Hefei, Anhui, China.
| | - Huimei Cai
- State Key Laboratory of Tea Plant Biology and Utilization, Anhui Provincial Key Laboratory of Food Safety Monitoring and Quality Control, School of Tea & Food Science and Technology, Anhui Agricultural University, Hefei 230036, Anhui, PR China; Joint Research Center for Food Nutrition and Health of IHM, Anhui Agricultural University, Hefei 230036, Anhui, PR China.
| | - Guijie Chen
- State Key Laboratory of Tea Plant Biology and Utilization, Anhui Provincial Key Laboratory of Food Safety Monitoring and Quality Control, School of Tea & Food Science and Technology, Anhui Agricultural University, Hefei 230036, Anhui, PR China; Joint Research Center for Food Nutrition and Health of IHM, Anhui Agricultural University, Hefei 230036, Anhui, PR China.
| |
Collapse
|
26
|
Yuan Y, Wu D, Chen H, Ma Z, Peng X, Li X, Zhao C, Jiang L, Liang J, Zhang W, Dai J. Farnesol ameliorates DSS-induced IBD by regulating inflammatory cytokines, repairing the intestinal barrier, reversing the gut microbiota imbalance, and influencing fecal metabolome in C57BL/6 mice. Biomed Pharmacother 2024; 180:117518. [PMID: 39405907 DOI: 10.1016/j.biopha.2024.117518] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2024] [Revised: 09/26/2024] [Accepted: 10/04/2024] [Indexed: 11/14/2024] Open
Abstract
The incidence of inflammatory bowel disease (IBD) is rising globally, increasing interest in food ingredients for its prevention and control. This study evaluated the effect of farnesol (FAR), a key component of pomelo flower volatile oil, on dextran sodium sulfate (DSS)-induced colitis in C57BL/6 mice. FAR significantly alleviated DSS-induced colitis and secondary liver injury, as shown by improved body weight, DAI, colon length, and pathology, as well as liver function and blood lipid indices. The mechanism involves FAR-mediated regulation of inflammatory cytokines, increased expression of tight junction protein genes, and decreased expression of lipid metabolism-related proteins. FAR also enhanced gut microbiota diversity, balancing harmful and probiotic bacteria. Fecal metabolome analysis indicated FAR's role in reversing metabolic disturbances related to inflammation and liver lipid metabolism. These findings support developing functional foods for IBD treatment using pomelo flower volatile oil.
Collapse
Affiliation(s)
- Ya Yuan
- School of Laboratory Medicine, Chengdu Medical College, Chengdu 610500, PR China
| | - Dazuo Wu
- School of Laboratory Medicine, Chengdu Medical College, Chengdu 610500, PR China
| | - Heping Chen
- The 3rd Affiliated Hospital of Chengdu Medical College, Pidu District People's Hospital,Chengdu 611730, PR China
| | - Zheng Ma
- School of Laboratory Medicine, Chengdu Medical College, Chengdu 610500, PR China
| | - Xinyue Peng
- School of Laboratory Medicine, Chengdu Medical College, Chengdu 610500, PR China
| | - Xiaodie Li
- School of Laboratory Medicine, Chengdu Medical College, Chengdu 610500, PR China
| | - Chuchu Zhao
- School of Laboratory Medicine, Chengdu Medical College, Chengdu 610500, PR China
| | - Linping Jiang
- School of Laboratory Medicine, Chengdu Medical College, Chengdu 610500, PR China
| | - Jinping Liang
- School of Laboratory Medicine, Chengdu Medical College, Chengdu 610500, PR China
| | - Weiwei Zhang
- Department of Public Health, Chengdu Medical College, Chengdu 610500, PR China.
| | - Juan Dai
- School of Laboratory Medicine, Chengdu Medical College, Chengdu 610500, PR China.
| |
Collapse
|
27
|
La Vecchia M, Sala G, Sculco M, Aspesi A, Dianzani I. Genetics, diet, microbiota, and metabolome: partners in crime for colon carcinogenesis. Clin Exp Med 2024; 24:248. [PMID: 39470880 PMCID: PMC11522171 DOI: 10.1007/s10238-024-01505-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2024] [Accepted: 10/15/2024] [Indexed: 11/01/2024]
Abstract
Colorectal cancer (CRC) ranks among the most prevalent malignant tumors worldwide, with a multifactorial etiology encompassing genetic, environmental, and life-style factors, as well as the intestinal microbiota and its metabolome. These risk factors often work together in specific groups of patients, influencing how CRC develops and progresses. Importantly, alterations in the gut microbiota act as a critical nexus in this interplay, significantly affecting susceptibility to CRC. This review highlights recent insights into unmodifiable and modifiable risk factors for CRC and how they might interact with the gut microbiota and its metabolome. Understanding the mechanisms of these interactions will help us develop targeted, precision-medicine strategies that can adjust the composition of the gut microbiota to meet individual health needs, preventing or treating CRC more effectively.
Collapse
Affiliation(s)
- Marta La Vecchia
- Department of Health Sciences, Università del Piemonte Orientale, 28100, Novara, Italy
| | - Gloria Sala
- Department of Health Sciences, Università del Piemonte Orientale, 28100, Novara, Italy
| | - Marika Sculco
- Department of Health Sciences, Università del Piemonte Orientale, 28100, Novara, Italy
| | - Anna Aspesi
- Department of Health Sciences, Università del Piemonte Orientale, 28100, Novara, Italy
| | - Irma Dianzani
- Department of Health Sciences, Università del Piemonte Orientale, 28100, Novara, Italy.
| |
Collapse
|
28
|
Chauhan G, Massey WJ, Veisman I, Rieder F. Anti-fibrotics in inflammatory bowel diseases: Challenges and successes. ADVANCES IN PHARMACOLOGY (SAN DIEGO, CALIF.) 2024; 101:85-106. [PMID: 39521606 DOI: 10.1016/bs.apha.2024.10.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/16/2024]
Abstract
Stricture formation leading to obstruction in Crohn's disease (CD) remains one of the largest unmet needs in the field of inflammatory bowel diseases (IBD). Despite this need no selective anti-stricture drug has been approved for use in CD patients. This contrasts with other fibrotic diseases, such as in the lung, liver or kidney, where multiple drug development programs crossed the starting line and two anti-fibrotics are now being approved for pulmonary fibrosis. Strictures are composed of a mix of inflammation, excessive deposition of extracellular matrix (ECM) and smooth muscle hyperplasia, likely all ultimately being responsible for the luminal narrowing driving patient symptoms. Our understanding of the pathogenesis of stricturing CD has evolved and indicates a multifactorial process involving immune and non-immune cells and their soluble mediators. This understanding has rendered target pathways for anti-stricture drug development. Significant progress was made in creating consensus definitions and tools to enable clinical trials with two clinical development programs having been conceived to date. In this chapter, we discuss stricture pathogenesis with a focus on the pathways being tested in clinical trials, and clinical trial endpoints developed for this indication.
Collapse
Affiliation(s)
- Gaurav Chauhan
- Department of Inflammation and Immunity, Lerner Research Institute, Cleveland Clinic Foundation, Cleveland, OH, United States
| | - William J Massey
- Department of Inflammation and Immunity, Lerner Research Institute, Cleveland Clinic Foundation, Cleveland, OH, United States
| | - Ido Veisman
- Department of Inflammation and Immunity, Lerner Research Institute, Cleveland Clinic Foundation, Cleveland, OH, United States
| | - Florian Rieder
- Department of Inflammation and Immunity, Lerner Research Institute, Cleveland Clinic Foundation, Cleveland, OH, United States; Department of Gastroenterology, Hepatology and Nutrition, Digestive Disease Institute, Cleveland Clinic Foundation, Cleveland, OH, United States.
| |
Collapse
|
29
|
Wang H, Chen Y, Wang Z, Yuan Y, Yue T. Novel selenium-enriched Pichia kudriavzevii as a dietary supplement to alleviate dextran sulfate sodium-induced colitis in mice by modulating the gut microbiota and host metabolism. Food Funct 2024; 15:10698-10716. [PMID: 39378068 DOI: 10.1039/d4fo02598a] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/29/2024]
Abstract
Inflammatory bowel disease (IBD) poses persistent challenges due to its chronic and recurrent nature, exacerbated by the unsatisfactory outcomes of the traditional treatment approaches. In this study, we developed a dietary supplement, selenium-enriched Pichia kudriavzevii (SeY), to alleviate dextran sulfate sodium-induced colitis in mice. The newly developed functional food shows dual-functional activity, acting both as a probiotic and a reliable source of organic selenium. This study aimed to investigate the preventive effects of SeY against dextran sulfate sodium-induced colitis in mice and elucidate the underlying mechanisms. Results showed that SeY, especially at high doses (HSeY), significantly ameliorated colitis symptoms, reduced colonic damage, attenuated inflammatory responses, and mitigated oxidative stress. Furthermore, HSeY strengthened intestinal barrier function by increasing goblet cell numbers, upregulating MUC2 expression, and enhancing tight junction proteins (ZO-1, claudin-1, and occludin). Additionally, HSeY alleviated gut microbiota dysbiosis by promoting the colonization of beneficial bacteria such as norank-f-Muribaculaceae and Bacteroides, while suppressing harmful microorganisms such as norank-f-norank-o-Clostridia-UCG-014. The altered gut microbiota also affected gut metabolism, with differential metabolites primarily associated with amino acids, such as tryptophan metabolism, contributing to the mitigation of oxidative stress and inflammatory responses. Further studies involving antibiotic-mediated depletion of gut flora and fecal microbiota transfer trials corroborated that the preventive effect of HSeY against IBD relied on the gut microbiota. This study provides vital insights into colitis prevention and advances selenium-enriched fortified food-targeted nutritional interventions.
Collapse
Affiliation(s)
- Huijuan Wang
- College of Food Science and Engineering, Northwest A&F University, Yangling, Shaanxi, 712100, China.
| | - Yue Chen
- College of Food Science and Engineering, Northwest A&F University, Yangling, Shaanxi, 712100, China.
| | - Zhouli Wang
- College of Food Science and Engineering, Northwest A&F University, Yangling, Shaanxi, 712100, China.
| | - Yahong Yuan
- College of Food Science and Technology, Northwest University, Xi'an, 710069, China
| | - Tianli Yue
- College of Food Science and Engineering, Northwest A&F University, Yangling, Shaanxi, 712100, China.
- College of Food Science and Technology, Northwest University, Xi'an, 710069, China
| |
Collapse
|
30
|
Chen S, Gao W, Ge P, Chang S, Wang T, Zhao Q, He B. Negatively Charged Thermosensitive Hydrogel Loaded with Pectin Microspheres to Recover the Mucosal Barrier for Ulcerative Colitis Therapy. Biomacromolecules 2024; 25:6801-6813. [PMID: 39311442 DOI: 10.1021/acs.biomac.4c01019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/15/2024]
Abstract
Ulcerative colitis (UC), a chronic inflammatory bowel disease, poses a heightened colorectal cancer risk due to persistent mucosal inflammation and barrier dysfunction. In this article, a negatively charged thermosensitive hydrogel loaded with pectin microspheres was used as the enema for UC treatment. Succinic acid was immobilized on poly(ε-caprolactone-co-glycolide)-poly(ethylene glycol)-poly(ε-caprolactone-co-glycolide) (PCLGA-PEG-PCLGA) triblock copolymers to preferentially coat on cationic-inflamed sites via electrostatic interaction for reconstructing the mucosal barrier. Anti-inflammation drug 5-aminosalicylic acid (5-ASA) and curcumin-loaded pectin microspheres (Pec@Cur) were dispersed in the hydrogel for the inflammatory treatment of UC. The thermally sensitive hydrogels were rectally injected into UC model mice. The hydrogel effectively adhered to ulcers and prolonged colon retention, enabling sustained drug release and remarkably relieving the symptoms of colitis. The negatively charged hydrogel exhibited excellent significance in the UC treatment.
Collapse
Affiliation(s)
- Shuqi Chen
- School of Pharmacy, Chengdu University, Chengdu 610106, China
| | - Wenxia Gao
- School of Pharmacy, Chengdu University, Chengdu 610106, China
| | - Pengjin Ge
- Chengdu Baili-Biopharm. Ltd., Chengdu 610041, China
| | - Shuhua Chang
- National Engineering Research Center for Biomaterials, College of Biomedical Engineering, Sichuan University, Chengdu 610065, China
| | - Ting Wang
- Department of Ophthalmology, West China Hospital, Sichuan University, No. 37 Guoxue Alley, Chengdu 610041, China
| | - Quan Zhao
- National Engineering Research Center for Biomaterials, College of Biomedical Engineering, Sichuan University, Chengdu 610065, China
| | - Bin He
- National Engineering Research Center for Biomaterials, College of Biomedical Engineering, Sichuan University, Chengdu 610065, China
| |
Collapse
|
31
|
Kuenstner JT, Zhang P, Potula R, Galarneau JM, Bach H. Human antibodies against Mycobacterium avium ssp. paratuberculosis combined with cytokine levels for the diagnosis and selection of Crohn's disease patients for anti-mycobacterial therapy-A pilot study. PLoS One 2024; 19:e0308911. [PMID: 39365776 PMCID: PMC11451984 DOI: 10.1371/journal.pone.0308911] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2024] [Accepted: 07/23/2024] [Indexed: 10/06/2024] Open
Abstract
Increasing evidence links a worldwide bacterial infection of cattle and other animal species by Mycobacterium avium ssp. paratuberculosis (MAP) to Crohn's disease (CD). A large, FDA phase 2/3 controlled clinical trial of combination antimycobacterial antibiotic therapy for CD has been completed, and the report describing the trial is pending publication. The identification of MAP infection in CD patients will become increasingly important. Thus, it is desirable to develop MAP-based tests that accurately predict which CD patients have a MAP infection. A prospective, case-control laboratory test study of 199 subjects (61 CD patients and 138 non-CD controls) was performed using a panel of MAP antigens, including Hsp65, PknG, PtpA, CL1, and MAP IDEXX, which were measured under blind conditions in the plasma of the 199 subjects. Results showed that compared to any individual MAP antigen, combinations of antigens showed improved CD classification performance. For the Hsp65 antigen, the sensitivity (SEN), specificity (SPE), positive predictive value (PPV), negative predictive value (NPV), correct classification (CC), and area under the curve (AUC) were 59.02%, 58.70%, 38.71%, 76.42%, 59.3% and 0.606, respectively. For the best combination of MAP antibodies (Hsp65 and PknG), the SEN, SPE, PPV, NPV, CC, and AUC were 59.02%, 60.87%, 40.00%, 77.06%, 60.30%, and 0.631, respectively. Further improvement of the CD classification performance was achieved by combining IFN-γ, IL-8, and IL-17 cytokines with antibodies against MAP antigens, yielding SEN, SPE, PPV, NPV, CC, and AUC of 62.3%, 62.32%, 42.22%, 78.9%, 62.31% and 0.708, respectively. Thus, combinations of antibodies against MAP antigens and cytokine levels yield better CD diagnostic predictive performance than any individual antibodies against MAP antigens.
Collapse
Affiliation(s)
- J. Todd Kuenstner
- Biology Department, Abilene Christian University, Abilene, Texas, United States of America
| | - Peilin Zhang
- PZM Diagnostics, Charleston, West Virginia, United States of America
| | - Raghava Potula
- Lewis Katz School of Medicine, Temple University, Philadelphia, Pennsylvania, United States of America
| | - Jean-Michel Galarneau
- Sport Injury Prevention Research Centre, University of Calgary, Calgary, Alberta, Canada
| | - Horacio Bach
- Division of Infectious Diseases, Department of Medicine, University of British Columbia, Vancouver, British Columbia, Canada
| |
Collapse
|
32
|
Shanthamallu US, Kilpatrick C, Jones A, Rubin J, Saleh A, Barabási AL, Akmaev VR, Ghiassian SD. A Network-Based Framework to Discover Treatment-Response-Predicting Biomarkers for Complex Diseases. J Mol Diagn 2024; 26:917-930. [PMID: 39067570 DOI: 10.1016/j.jmoldx.2024.06.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2023] [Revised: 06/10/2024] [Accepted: 06/26/2024] [Indexed: 07/30/2024] Open
Abstract
The potential of precision medicine to transform complex autoimmune disease treatment is often challenged by limited data availability and inadequate sample size when compared with the number of molecular features found in high-throughput multi-omics data sets. To address this issue, the novel framework PRoBeNet (Predictive Response Biomarkers using Network medicine) was developed. PRoBeNet operates under the hypothesis that the therapeutic effect of a drug propagates through a protein-protein interaction network to reverse disease states. PRoBeNet prioritizes biomarkers by considering i) therapy-targeted proteins, ii) disease-specific molecular signatures, and iii) an underlying network of interactions among cellular components (the human interactome). PRoBeNet helped discover biomarkers predicting patient responses to both an established autoimmune therapy (infliximab) and an investigational compound (a mitogen-activated protein kinase 3/1 inhibitor). The predictive power of PRoBeNet biomarkers was validated with retrospective gene-expression data from patients with ulcerative colitis and rheumatoid arthritis and prospective data from tissues from patients with ulcerative colitis and Crohn disease. Machine-learning models using PRoBeNet biomarkers significantly outperformed models using either all genes or randomly selected genes, especially when data were limited. These results illustrate the value of PRoBeNet in reducing features and for constructing robust machine-learning models when data are limited. PRoBeNet may be used to develop companion and complementary diagnostic assays, which may help stratify suitable patient subgroups in clinical trials and improve patient outcomes.
Collapse
Affiliation(s)
- Uday S Shanthamallu
- Department of Data Science and Network Medicine, Scipher Medicine, Waltham, Massachusetts
| | - Casey Kilpatrick
- Department of Therapeutics, Scipher Medicine, Waltham, Massachusetts
| | - Alex Jones
- Department of Data Science and Network Medicine, Scipher Medicine, Waltham, Massachusetts
| | | | - Alif Saleh
- Department of Data Science and Network Medicine, Scipher Medicine, Waltham, Massachusetts
| | - Albert-László Barabási
- Center for Complex Network Research, Northeastern University, Boston, Massachusetts; Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts; Department of Network and Data Science, Central European University, Budapest, Hungary
| | - Viatcheslav R Akmaev
- Department of Data Science and Network Medicine, Scipher Medicine, Waltham, Massachusetts
| | - Susan D Ghiassian
- Department of Data Science and Network Medicine, Scipher Medicine, Waltham, Massachusetts.
| |
Collapse
|
33
|
Freitas ADS, Barroso FAL, Campos GM, Américo MF, Viegas RCDS, Gomes GC, Vital KD, Fernandes SOA, Carvalho RDDO, Jardin J, Miranda APGDS, Ferreira E, Martins FS, Laguna JG, Jan G, Azevedo V, de Jesus LCL. Exploring the anti-inflammatory effects of postbiotic proteins from Lactobacillus delbrueckii CIDCA 133 on inflammatory bowel disease model. Int J Biol Macromol 2024; 277:134216. [PMID: 39069058 DOI: 10.1016/j.ijbiomac.2024.134216] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2024] [Revised: 07/22/2024] [Accepted: 07/25/2024] [Indexed: 07/30/2024]
Abstract
Lactobacillus delbrueckii CIDCA 133 is a promising health-promoting bacterium shown to alleviate intestinal inflammation. However, the specific bacterial components responsible for these effects remain largely unknown. Here, we demonstrated that consuming extractable proteins from the CIDCA 133 strain effectively relieved acute ulcerative colitis in mice. This postbiotic protein fraction reduced the disease activity index and prevented colon shortening in mice. Furthermore, histological analysis revealed colitis prevention with reduced inflammatory cell infiltration into the colon mucosa. Postbiotic consumption also induced an immunomodulatory profile in colitic mice, as evidenced by both mRNA transcript levels (Tlr2, Nfkb1, Nlpr3, Tnf, and Il6) and cytokines concentration (IL1β, TGFβ, and IL10). Additionally, it enhanced the levels of secretory IgA, upregulated the transcript levels of tight junction proteins (Hp and F11r), and improved paracellular intestinal permeability. More interestingly, the consumption of postbiotic proteins modulated the gut microbiota (Bacteroides, Arkkemansia, Dorea, and Oscillospira). Pearson correlation analysis indicated that IL10 and IL1β levels were positively associated with Bacteroides and Arkkemansia_Lactobacillus abundance. Our study reveals that CIDCA 133-derived proteins possess anti-inflammatory properties in colonic inflammation.
Collapse
Affiliation(s)
- Andria Dos Santos Freitas
- Federal University of Minas Gerais, Department of Genetics, Ecology, and Evolution, Belo Horizonte, Minas Gerais, Brazil
| | | | - Gabriela Munis Campos
- Federal University of Minas Gerais, Department of Genetics, Ecology, and Evolution, Belo Horizonte, Minas Gerais, Brazil
| | - Monique Ferrary Américo
- Federal University of Minas Gerais, Department of Genetics, Ecology, and Evolution, Belo Horizonte, Minas Gerais, Brazil
| | | | - Gabriel Camargos Gomes
- Federal University of Minas Gerais, Department of Genetics, Ecology, and Evolution, Belo Horizonte, Minas Gerais, Brazil
| | - Kátia Duarte Vital
- Federal University of Minas Gerais, Department of Clinical and Toxicological Analysis, Belo Horizonte, Minas Gerais, Brazil
| | | | | | | | | | - Enio Ferreira
- Federal University of Minas Gerais, Department of General Pathology, Belo Horizonte, Minas Gerais, Brazil
| | - Flaviano Santos Martins
- Federal University of Minas Gerais, Department of Microbiology, Belo Horizonte, Minas Gerais, Brazil
| | - Juliana Guimarães Laguna
- Federal University of Minas Gerais, Department of Genetics, Ecology, and Evolution, Belo Horizonte, Minas Gerais, Brazil
| | | | - Vasco Azevedo
- Federal University of Minas Gerais, Department of Genetics, Ecology, and Evolution, Belo Horizonte, Minas Gerais, Brazil.
| | - Luís Cláudio Lima de Jesus
- Federal University of Minas Gerais, Department of Genetics, Ecology, and Evolution, Belo Horizonte, Minas Gerais, Brazil.
| |
Collapse
|
34
|
Dai N, Yang X, Pan P, Zhang G, Sheng K, Wang J, Liang X, Wang Y. Bacillus paralicheniformis, an acetate-producing probiotic, alleviates ulcerative colitis via protecting the intestinal barrier and regulating the NLRP3 inflammasome. Microbiol Res 2024; 287:127856. [PMID: 39079268 DOI: 10.1016/j.micres.2024.127856] [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: 05/28/2024] [Revised: 07/11/2024] [Accepted: 07/22/2024] [Indexed: 08/22/2024]
Abstract
Ulcerative colitis (UC) presents a challenging scenario in digestive health, characterized by recurrent inflammation that is often hard to manage. Bacteria capable of producing short-chain fatty acids (SCFAs) play a pivotal role in mitigating UC symptoms, rendering them promising candidates for probiotic therapy. In this investigation, we assessed the impact of Bacillus paralicheniformis HMPM220325 on dextran sodium sulfate (DSS)-induced UC in mice. Genomic analysis of the strain revealed the presence of protease genes associated with acetate and butyrate synthesis, with acetic acid detected in its fermentation broth. Administration of B. paralicheniformis HMPM220325 to UC mice ameliorated pathological manifestations of the condition and restored intestinal barrier function. Furthermore, B. paralicheniformis HMPM220325 suppressed the activation of the NLRP3 inflammasome signaling pathway and modulated the composition of the intestinal microbiota. These findings shed significant light on the potential of B. paralicheniformis as a probiotic candidate, offering a novel avenue for the prevention and therapeutic intervention of colitis.
Collapse
Affiliation(s)
- Nini Dai
- School of Life Sciences, Anhui University, Hefei, China; Key Laboratory of Human Microenvironment and Precision Medicine of Anhui Higher Education Institutes, Anhui University, Hefei, China; Anhui Province Joint Construction Discipline Key Laboratory of Nanobody Technology, Hefei, China
| | - Xinting Yang
- School of Life Sciences, Anhui University, Hefei, China; Key Laboratory of Human Microenvironment and Precision Medicine of Anhui Higher Education Institutes, Anhui University, Hefei, China; Anhui Province Joint Construction Discipline Key Laboratory of Nanobody Technology, Hefei, China
| | - Peilong Pan
- School of Life Sciences, Anhui University, Hefei, China; Key Laboratory of Human Microenvironment and Precision Medicine of Anhui Higher Education Institutes, Anhui University, Hefei, China; Anhui Province Joint Construction Discipline Key Laboratory of Nanobody Technology, Hefei, China
| | - Guanghui Zhang
- School of Life Sciences, Anhui University, Hefei, China; Key Laboratory of Human Microenvironment and Precision Medicine of Anhui Higher Education Institutes, Anhui University, Hefei, China; Anhui Province Joint Construction Discipline Key Laboratory of Nanobody Technology, Hefei, China
| | - Kangliang Sheng
- School of Life Sciences, Anhui University, Hefei, China; Key Laboratory of Human Microenvironment and Precision Medicine of Anhui Higher Education Institutes, Anhui University, Hefei, China; Anhui Province Joint Construction Discipline Key Laboratory of Nanobody Technology, Hefei, China
| | - Jingmin Wang
- School of Life Sciences, Anhui University, Hefei, China; Key Laboratory of Human Microenvironment and Precision Medicine of Anhui Higher Education Institutes, Anhui University, Hefei, China; Anhui Province Joint Construction Discipline Key Laboratory of Nanobody Technology, Hefei, China
| | - Xiao Liang
- School of Life Sciences, Anhui University, Hefei, China; Key Laboratory of Human Microenvironment and Precision Medicine of Anhui Higher Education Institutes, Anhui University, Hefei, China; Anhui Province Joint Construction Discipline Key Laboratory of Nanobody Technology, Hefei, China.
| | - Yongzhong Wang
- School of Life Sciences, Anhui University, Hefei, China; Key Laboratory of Human Microenvironment and Precision Medicine of Anhui Higher Education Institutes, Anhui University, Hefei, China; Anhui Province Joint Construction Discipline Key Laboratory of Nanobody Technology, Hefei, China.
| |
Collapse
|
35
|
Paula Sanchez Zanotti M, Cataldi de Alcântara C, Junko Inoue C, Piantoni Gonçalves B, Rabello Espinosa B, Luiz Cândido de Souza Cassela P, Lerner Trigo G, Mendes Ahrens T, Alysson Batisti Lozovoy M, Eduardo Coral de Oliveira C, Maria Vissoci Reiche E, Name Colado Simão A. Involvement of IL17A and IL17RA variants in interleukin-17A levels and disease activity in ulcerative colitis. Cytokine 2024; 182:156716. [PMID: 39111114 DOI: 10.1016/j.cyto.2024.156716] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2024] [Revised: 06/29/2024] [Accepted: 07/23/2024] [Indexed: 08/25/2024]
Abstract
Ulcerative colitis (UC) is characterized by chronic inflammation of the large intestine with involvement of Th17 cells and interleukin (IL)-17A. The role of IL17A and IL17A receptor (IL17RA) variants in pathophysiology of UC still remains inconclusive. The aim was to evaluate the association between IL17A and IL17RA variants with susceptibility, IL-17A plasma levels, and endoscopic activity in UC. The study included 104 patients with UC and 213 controls. Patients were divided according to endoscopic activity (remission/mild and moderate/severe). The IL17A rs3819024 A>G and rs3819025 G>A, and IL17RA rs2241043 C>T, rs2241049 A>G, and rs6518661 G>A variants were genotyped using real time polymerase chain reaction. IL-17A plasma levels were determined using immunofluorimetric assay. Neither IL17A nor IL17RA variants were associated with UC susceptibility. The IL17A rs3819024 AG genotype was associated to high levels of IL-17 only in patients. Patients with the G allele of IL17RA rs2241049 showed 2.944 more chance of developing moderate/severe disease. The haplotype analysis showed that IL17RA rs2241049 and rs6518661 was not associated with UC susceptibility and haplotypes constituted with G allele of these variants were not associated with disease severity (p = 0.09). In conclusion, the IL17A rs3819024 AG genotype was associated with elevated IL-17A plasma levels in patients with UC but not in controls and the IL17RA rs2241049 AG+GG genotypes were associated to severity of UC. These results suggest a possible hidden interaction between the IL17A rs3819024 variant and other genetic, environmental, and epigenetic factors in the IL-17A expression that is present only in patients with UC.
Collapse
Affiliation(s)
- Mariana Paula Sanchez Zanotti
- Laboratory of Research in Applied Immunology, University of Londrina, Londrina, PR, Brazil; Department of Gastroenterology, University of Londrina, Londrina, PR, Brazil
| | | | - Cláudia Junko Inoue
- Laboratory of Research in Applied Immunology, University of Londrina, Londrina, PR, Brazil; Department of Gastroenterology, University of Londrina, Londrina, PR, Brazil
| | - Beatriz Piantoni Gonçalves
- Laboratory of Research in Applied Immunology, University of Londrina, Londrina, PR, Brazil; Department of Gastroenterology, University of Londrina, Londrina, PR, Brazil
| | | | | | - Guilherme Lerner Trigo
- Laboratory of Research in Applied Immunology, University of Londrina, Londrina, PR, Brazil
| | - Tainah Mendes Ahrens
- Laboratory of Research in Applied Immunology, University of Londrina, Londrina, PR, Brazil
| | - Marcell Alysson Batisti Lozovoy
- Laboratory of Research in Applied Immunology, University of Londrina, Londrina, PR, Brazil; Department of Pathology, Clinical Analysis and Toxicology, Laboratory of Research in Applied Immunology, University of Londrina, Londrina, PR, Brazil
| | - Carlos Eduardo Coral de Oliveira
- Postgraduate Program of Clinical and Laboratory Pathophysiology, Health Sciences Center, Londrina State University, Londrina, Paraná, Brazil; Pontifical Catholic University of Paraná, School of Medicine, Campus Londrina, Londrina, Paraná, Brazil.
| | - Edna Maria Vissoci Reiche
- Postgraduate Program of Clinical and Laboratory Pathophysiology, Health Sciences Center, Londrina State University, Londrina, Paraná, Brazil; Pontifical Catholic University of Paraná, School of Medicine, Campus Londrina, Londrina, Paraná, Brazil.
| | - Andréa Name Colado Simão
- Laboratory of Research in Applied Immunology, University of Londrina, Londrina, PR, Brazil; Department of Pathology, Clinical Analysis and Toxicology, Laboratory of Research in Applied Immunology, University of Londrina, Londrina, PR, Brazil.
| |
Collapse
|
36
|
Adams CE, Rutherford DG, Jones GR, Ho GT. Immunometabolism and mitochondria in inflammatory bowel disease: a role for therapeutic intervention? Dis Model Mech 2024; 17:dmm050895. [PMID: 39415736 PMCID: PMC11512101 DOI: 10.1242/dmm.050895] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2024] Open
Abstract
Inflammatory bowel diseases (IBDs), incurable conditions characterised by recurrent episodes of immune-mediated gut inflammation and damage of unknown aetiology, are common. Current advanced therapies target key leukocyte-trafficking and cytokine-signalling hubs but are only effective in 50% of patients. With growing evidence of mitochondrial dysfunction in IBD and advances in our understanding of the role of metabolism in inflammation, we provide an overview of novel metabolic approaches to IBD therapy, challenging the current 'therapeutic ceiling', identifying critical pathways for intervention and re-imagining metabolic biomarkers for the 21st century.
Collapse
Affiliation(s)
- Claire E. Adams
- Gut Research Unit, Centre for Inflammation Research, Institute for Regeneration and Repair, University of Edinburgh, Edinburgh EH16 4UU, UK
| | - Duncan G. Rutherford
- Gut Research Unit, Centre for Inflammation Research, Institute for Regeneration and Repair, University of Edinburgh, Edinburgh EH16 4UU, UK
| | - Gareth R. Jones
- Gut Research Unit, Centre for Inflammation Research, Institute for Regeneration and Repair, University of Edinburgh, Edinburgh EH16 4UU, UK
| | - Gwo-tzer Ho
- Gut Research Unit, Centre for Inflammation Research, Institute for Regeneration and Repair, University of Edinburgh, Edinburgh EH16 4UU, UK
| |
Collapse
|
37
|
Lv Y, Peng J, Ma X, Liang Z, Salekdeh GH, Ke Q, Shen W, Yan Z, Li H, Wang S, Ding X. Network Analysis of Gut Microbial Communities Reveals Key Reason for Quercetin Protects against Colitis. Microorganisms 2024; 12:1973. [PMID: 39458282 PMCID: PMC11509604 DOI: 10.3390/microorganisms12101973] [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: 07/31/2024] [Revised: 09/19/2024] [Accepted: 09/19/2024] [Indexed: 10/28/2024] Open
Abstract
As one of the most representative natural products among flavonoids, quercetin (QUE) has been reported to exhibit beneficial effects on gut health in recent years. In this study, we utilized a dextran sulfate sodium (DSS)-induced colitis mice model to explore the protective effects and underlying mechanisms of QUE on colitis. Our data demonstrated that QUE oral gavage administration significantly ameliorates the symptoms and histopathological changes associated with colitis. Additionally, the concentration of mucin-2, the number of goblet cells, and the expression of tight junction proteins (such as ZO-1, Occludin, and Claudin-1) were all found to be increased. Furthermore, QUE treatment regulated the levels of inflammatory cytokines and macrophage polarization, as well as the oxidative stress-related pathway (Nrf2/HO-1) and associated enzymes. Additionally, 16S rDNA sequencing revealed that QUE treatment rebalances the alterations in colon microbiota composition (inlcuding Bacteroidaceae, Bacteroides, and Odoribacter) in DSS-induced colitis mice. The analysis of network dynamics reveals a significant correlation between gut microbial communities and microenvironmental factors associated with inflammation and oxidative stress, in conjunction with the previously mentioned findings. Collectively, our results suggest that QUE has the potential to treat colitis by maintaining the mucosal barrier, modulating inflammation, and reducing oxidation stress, which may depend on the reversal of gut microbiota dysbiosis.
Collapse
Affiliation(s)
- Yanan Lv
- Key Laboratory of Veterinary Pharmaceutical Development, Ministry of Agriculture and Rural Affairs, Lanzhou Institute of Husbandry and Pharmaceutical Sciences of Chinese Academy of Agricultural Science, Lanzhou 730050, China; (Y.L.); (J.P.); (X.M.); (Z.L.); (Q.K.); (W.S.); (Z.Y.); (H.L.)
| | - Jing Peng
- Key Laboratory of Veterinary Pharmaceutical Development, Ministry of Agriculture and Rural Affairs, Lanzhou Institute of Husbandry and Pharmaceutical Sciences of Chinese Academy of Agricultural Science, Lanzhou 730050, China; (Y.L.); (J.P.); (X.M.); (Z.L.); (Q.K.); (W.S.); (Z.Y.); (H.L.)
| | - Xiaoyu Ma
- Key Laboratory of Veterinary Pharmaceutical Development, Ministry of Agriculture and Rural Affairs, Lanzhou Institute of Husbandry and Pharmaceutical Sciences of Chinese Academy of Agricultural Science, Lanzhou 730050, China; (Y.L.); (J.P.); (X.M.); (Z.L.); (Q.K.); (W.S.); (Z.Y.); (H.L.)
- College of Veterinary Medicine, Inner Mongolia Agricultural University, Hohhot 010010, China
| | - Zeyi Liang
- Key Laboratory of Veterinary Pharmaceutical Development, Ministry of Agriculture and Rural Affairs, Lanzhou Institute of Husbandry and Pharmaceutical Sciences of Chinese Academy of Agricultural Science, Lanzhou 730050, China; (Y.L.); (J.P.); (X.M.); (Z.L.); (Q.K.); (W.S.); (Z.Y.); (H.L.)
| | - Ghasem Hosseini Salekdeh
- Department of Systems Biology, Agricultural Biotechnology Research Institute of Iran, Agricultural Research, Education, and Extension Organization, Karaj 3135933151, Iran;
- Department of Molecular Sciences, Macquarie University, North Ryde, NSW 2109, Australia
| | - Qunhua Ke
- Key Laboratory of Veterinary Pharmaceutical Development, Ministry of Agriculture and Rural Affairs, Lanzhou Institute of Husbandry and Pharmaceutical Sciences of Chinese Academy of Agricultural Science, Lanzhou 730050, China; (Y.L.); (J.P.); (X.M.); (Z.L.); (Q.K.); (W.S.); (Z.Y.); (H.L.)
| | - Wenxiang Shen
- Key Laboratory of Veterinary Pharmaceutical Development, Ministry of Agriculture and Rural Affairs, Lanzhou Institute of Husbandry and Pharmaceutical Sciences of Chinese Academy of Agricultural Science, Lanzhou 730050, China; (Y.L.); (J.P.); (X.M.); (Z.L.); (Q.K.); (W.S.); (Z.Y.); (H.L.)
- College of Veterinary Medicine, Northwest A&F University, Yangling 712100, China
| | - Zuoting Yan
- Key Laboratory of Veterinary Pharmaceutical Development, Ministry of Agriculture and Rural Affairs, Lanzhou Institute of Husbandry and Pharmaceutical Sciences of Chinese Academy of Agricultural Science, Lanzhou 730050, China; (Y.L.); (J.P.); (X.M.); (Z.L.); (Q.K.); (W.S.); (Z.Y.); (H.L.)
| | - Hongsheng Li
- Key Laboratory of Veterinary Pharmaceutical Development, Ministry of Agriculture and Rural Affairs, Lanzhou Institute of Husbandry and Pharmaceutical Sciences of Chinese Academy of Agricultural Science, Lanzhou 730050, China; (Y.L.); (J.P.); (X.M.); (Z.L.); (Q.K.); (W.S.); (Z.Y.); (H.L.)
| | - Shengyi Wang
- Key Laboratory of Veterinary Pharmaceutical Development, Ministry of Agriculture and Rural Affairs, Lanzhou Institute of Husbandry and Pharmaceutical Sciences of Chinese Academy of Agricultural Science, Lanzhou 730050, China; (Y.L.); (J.P.); (X.M.); (Z.L.); (Q.K.); (W.S.); (Z.Y.); (H.L.)
| | - Xuezhi Ding
- Key Laboratory of Veterinary Pharmaceutical Development, Ministry of Agriculture and Rural Affairs, Lanzhou Institute of Husbandry and Pharmaceutical Sciences of Chinese Academy of Agricultural Science, Lanzhou 730050, China; (Y.L.); (J.P.); (X.M.); (Z.L.); (Q.K.); (W.S.); (Z.Y.); (H.L.)
| |
Collapse
|
38
|
Górecka A, Jura-Półtorak A, Koźma EM, Szeremeta A, Olczyk K, Komosińska-Vassev K. Biochemical Modulators of Tight Junctions (TJs): Occludin, Claudin-2 and Zonulin as Biomarkers of Intestinal Barrier Leakage in the Diagnosis and Assessment of Inflammatory Bowel Disease Progression. Molecules 2024; 29:4577. [PMID: 39407507 PMCID: PMC11478261 DOI: 10.3390/molecules29194577] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2024] [Revised: 09/24/2024] [Accepted: 09/25/2024] [Indexed: 10/20/2024] Open
Abstract
BACKGROUND Considering the increasing worldwide prevalence of inflammatory bowel disease (IBD), the early diagnosis of this disease is extremely important. However, non-invasive diagnostic methods remain limited, while invasive techniques are the most commonly used in daily practice. Therefore, there is a serious need to find new non-invasive biomarkers of IBD. METHODS The serum profiles of occludin, claudin-2, and zonulin were assessed in IBD patients using the ELISA method. The levels of the analyzed biomarkers were measured before and after a year of anti-inflammatory treatment, which was a tumor necrosis factor α (TNF-α) inhibitor (adalimumab) in patients with ulcerative colitis (UC) and conventional therapy in patients with Crohn's disease (CD). RESULTS In IBD patients, the serum level of occludin (p < 0.001) decreased compared to healthy individuals, while the level of claudin-2 (p < 0.001) increased. Additionally, zonulin (p < 0.01) concentration increased in CD patients compared to the control group. The highest diagnostic ability was presented by occludin measurements with the area under the curve (AUC) of 0.959 (95% CI 0.907-1) in UC and 0.948 (95% CI 0.879-1) in CD. Claudin-2 also demonstrated very good ability in diagnosing UC and CD with AUC values of 0.864 (95% CI 0.776-0.952) and 0.896 (95% CI 0.792-0.999), respectively. The ability of zonulin to diagnose CD was estimated as good with an AUC of 0.74 (95% CI 0.598-0.881). Moreover, a significant correlation was identified between C-reactive protein (CRP), claudin-2 (r = -0.37; p < 0.05), and zonulin (r = -0.44; p < 0.05) in UC patients. Treatment with adalimumab improved the level of occludin, claudin-2, and zonulin in UC patients, while anti-inflammatory conventional therapy decreased the concentration of zonulin in CD. CONCLUSIONS Occludin and claudin-2 measurements present significant utility in diagnosing both UC and CD, while zonulin assessments may be useful in CD diagnosis. Additionally, claudin-2 and zonulin measurements may be helpful in evaluating the intensity of the inflammatory process. Anti-TNF-α treatment improved the value of occludin, claudin-2, and zonulin, indicating its beneficial effect on the integrity of tight junctions in UC.
Collapse
Affiliation(s)
- Aleksandra Górecka
- Department of Clinical Chemistry and Laboratory Diagnostics, Faculty of Pharmaceutical Sciences in Sosnowiec, Medical University of Silesia in Katowice, 41-200 Sosnowiec, Poland; (A.J.-P.); (E.M.K.); (A.S.); (K.O.); (K.K.-V.)
| | | | | | | | | | | |
Collapse
|
39
|
Byun S, Lee J, Choi YH, Ko H, Lee C, Park JC, Kim SW, Lee H, Sharma A, Kim KS, Rudra D, Kim JK, Im SH. Gut Microbiota Defines Functional Direction of Colonic Regulatory T Cells with Unique TCR Repertoires. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2024; 213:886-897. [PMID: 39101764 DOI: 10.4049/jimmunol.2300395] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/03/2023] [Accepted: 07/03/2024] [Indexed: 08/06/2024]
Abstract
Intestinal microbiota and selected strains of commensal bacteria influence regulatory T (Treg) cell functionality in the colon. Nevertheless, whether and how microbiota changes the transcriptome profile and TCR specificities of colonic Tregs remain to be precisely defined. In this study, we have employed single-cell RNA sequencing and comparatively analyzed colonic Tregs from specific pathogen-free and germ-free (GF) mice. We found that microbiota shifts the activation trajectory of colonic Tregs toward a distinct phenotypic subset enriched in specific pathogen-free but not in GF mice. Moreover, microbiota induced the expansion of specific Treg clonotypes with shared transcriptional specificities. The microbiota-induced subset of colonic Tregs, identified as PD-1- CXCR3+ Tregs, displayed enhanced suppressive capabilities compared with colonic Tregs derived from GF mice, enhanced production of IL-10, and were the primary regulators of enteric inflammation in dextran sodium sulfate-induced colitis. These findings identify a hitherto unknown gut microbiota and immune cell interaction module that could contribute to the development of a therapeutic modality for intestinal inflammatory diseases.
Collapse
Affiliation(s)
- Seohyun Byun
- Department of Life Sciences, Pohang University of Science and Technology, Pohang, Republic of Korea
| | - Jusung Lee
- Department of Life Sciences, Pohang University of Science and Technology, Pohang, Republic of Korea
- Department of New Biology, Daegu Gyeongbuk Institute of Science & Technology, Daegu, Republic of Korea
| | - Yoon Ha Choi
- Department of Life Sciences, Pohang University of Science and Technology, Pohang, Republic of Korea
- Department of New Biology, Daegu Gyeongbuk Institute of Science & Technology, Daegu, Republic of Korea
| | - Haeun Ko
- Department of Life Sciences, Pohang University of Science and Technology, Pohang, Republic of Korea
| | - Changhon Lee
- Department of Life Sciences, Pohang University of Science and Technology, Pohang, Republic of Korea
| | - John Chulhoon Park
- Department of Life Sciences, Pohang University of Science and Technology, Pohang, Republic of Korea
| | - Seung Won Kim
- Department of Life Sciences, Pohang University of Science and Technology, Pohang, Republic of Korea
| | - Haena Lee
- Department of Life Sciences, Pohang University of Science and Technology, Pohang, Republic of Korea
| | - Amit Sharma
- Department of Life Sciences, Pohang University of Science and Technology, Pohang, Republic of Korea
| | - Kwang Soon Kim
- Department of Life Sciences, Pohang University of Science and Technology, Pohang, Republic of Korea
| | - Dipayan Rudra
- School of Life Science and Technology, ShanghaiTech University; Shanghai, People's Republic of China
| | - Jong Kyoung Kim
- Department of Life Sciences, Pohang University of Science and Technology, Pohang, Republic of Korea
- Department of New Biology, Daegu Gyeongbuk Institute of Science & Technology, Daegu, Republic of Korea
- Institute for Convergence Research and Education in Advanced Technology, Yonsei University, Seoul, Republic of Korea
| | - Sin-Hyeog Im
- Department of Life Sciences, Pohang University of Science and Technology, Pohang, Republic of Korea
- Institute for Convergence Research and Education in Advanced Technology, Yonsei University, Seoul, Republic of Korea
- ImmunoBiome Inc., Pohang, Republic of Korea
| |
Collapse
|
40
|
Sharma S, Gilberto VS, Levens CL, Chatterjee A, Kuhn KA, Nagpal P. Microbiome- and Host Inflammasome-Targeting Inhibitor Nanoligomers Are Therapeutic in the Murine Colitis Model. ACS Pharmacol Transl Sci 2024; 7:2677-2693. [PMID: 39296260 PMCID: PMC11406689 DOI: 10.1021/acsptsci.4c00102] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2024] [Revised: 07/30/2024] [Accepted: 08/20/2024] [Indexed: 09/21/2024]
Abstract
Autoimmune and autoinflammatory diseases account for more than 80 chronic conditions affecting more than 24 million people in the US. Among these autoinflammatory diseases, noninfectious chronic inflammation of the gastrointestinal (GI) tract causes inflammatory bowel diseases (IBDs), primarily Crohn's and ulcerative colitis (UC). IBD is a complex disease, and one hypothesis is that these are either caused or worsened by compounds produced by bacteria in the gut. While traditional approaches have focused on pan immunosuppressive techniques (e.g., steroids), low remission rates, prolonged illnesses, and an increased frequency of surgical procedures have prompted the search for more targeted and precision therapeutic approaches. IBD is a complex disease resulting from both genetic and environmental factors, but several recent studies have highlighted the potential pivotal contribution of gut microbiota dysbiosis. Gut microbiota are known to modulate the immune status of the gut by producing metabolites that are encoded in biosynthetic gene clusters (BGCs) of the bacterial genome. Here, we show a targeted and high-throughput screening of more than 90 biosynthetic genes in 41 gut anaerobes, through downselection using available bioinformatics tools, targeted gene manipulation in these genetically intractable organisms using the Nanoligomer platform, and identification and synthesis of top microbiome targets as a Nanoligomer BGC cocktail (SB_BGC_CK1, abbreviated as CK1) as a feasible precision therapeutic approach. Further, we used a host-directed immune target screening to identify the NF-κB and NLRP3 cocktail SB_NI_112 (or NI112 for short) as a targeted inflammasome inhibitor. We used these top two microbe- and host-targeted Nanoligomer cocktails in acute and chronic dextran sulfate sodium (DSS) mouse colitis and in TNFΔARE/+ transgenic mice that develop spontaneous Crohn's like ileitis. The mouse microbiome was humanized to replicate that in human IBD through antibiotic treatment, followed by mixed fecal gavage from 10 human donors and spiked with IBD-inducing microbial species. Following colonization, colitis was induced in mice using 1 week of 3% DSS (acute) or 6 weeks of 3 rounds of 2.5% DSS induction for a week followed by 1 week of no DSS (chronic colitis model). Both Nanoligomer cocktails (CK1 and NI112) showed a strong reduction in disease severity, significant improvement in disease histopathology, and profound downregulation of disease biomarkers in colon tissue, as assessed by multiplexed ELISA. Further, we used two different formulations of intraperitoneal injections (IP) and Nanoligomer pills in the chronic DSS colitis model. Although both formulations were highly effective, the oral pill formulation demonstrated a greater reduction in biochemical markers compared to IP. A similar therapeutic effect was observed in the TNFΔARE/+ model. Overall, these results point to the potential for further development and testing of this inflammasome-targeting host-directed therapy (NI112) and more personalized microbiome cocktails (CK1) for patients with recalcitrant IBD.
Collapse
Affiliation(s)
- Sadhana Sharma
- Sachi Bio, Colorado Technology Center, 685 S Arthur Avenue, Louisville, Colorado 8002, United States
| | - Vincenzo S Gilberto
- Sachi Bio, Colorado Technology Center, 685 S Arthur Avenue, Louisville, Colorado 8002, United States
| | - Cassandra L Levens
- Division of Rheumatology, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado 80045, United States
| | - Anushree Chatterjee
- Sachi Bio, Colorado Technology Center, 685 S Arthur Avenue, Louisville, Colorado 8002, United States
| | - Kristine A Kuhn
- Division of Rheumatology, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado 80045, United States
| | - Prashant Nagpal
- Sachi Bio, Colorado Technology Center, 685 S Arthur Avenue, Louisville, Colorado 8002, United States
| |
Collapse
|
41
|
Singh V, Choi SD, Mahra K, Son H, Lee H, Lee YJ, Kim ES, Shin JH. Cultured fecal microbial community and its impact as fecal microbiota transplantation treatment in mice gut inflammation. Appl Microbiol Biotechnol 2024; 108:463. [PMID: 39269473 PMCID: PMC11399162 DOI: 10.1007/s00253-024-13295-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: 06/19/2024] [Revised: 08/21/2024] [Accepted: 08/25/2024] [Indexed: 09/15/2024]
Abstract
The fecal microbiome is identical to the gut microbial communities and provides an easy access to the gut microbiome. Therefore, fecal microbial transplantation (FMT) strategies have been used to alter dysbiotic gut microbiomes with healthy fecal microbiota, successfully alleviating various metabolic disorders, such as obesity, type 2 diabetes, and inflammatory bowel disease (IBD). However, the success of FMT treatment is donor-dependent and variations in gut microbes cannot be avoided. This problem may be overcome by using a cultured fecal microbiome. In this study, a human fecal microbiome was cultured using five different media; growth in brain heart infusion (BHI) media resulted in the highest microbial community cell count. The microbiome (16S rRNA) data demonstrated that the cultured microbial communities were similar to that of the original fecal sample. Therefore, the BHI-cultured fecal microbiome was selected for cultured FMT (cFMT). Furthermore, a dextran sodium sulfate (DSS)-induced mice-IBD model was used to confirm the impact of cFMT. Results showed that cFMT effectively alleviated IBD-associated symptoms, including improved gut permeability, restoration of the inflamed gut epithelium, decreased expression of pro-inflammatory cytokines (IFN-γ, TNF-α, IL-1, IL-6, IL-12, and IL-17), and increased expression of anti-inflammatory cytokines (IL-4 and IL-10). Thus, study's findings suggest that cFMT can be a potential alternative to nFMT. KEY POINTS: • In vitro fecal microbial communities were grown in a batch culture using five different media. • Fecal microbial transplantation was performed on DSS-treated mice using cultured and normal fecal microbes. • Cultured fecal microbes effectively alleviated IBD-associated symptoms.
Collapse
Affiliation(s)
- Vineet Singh
- Department of Applied Biosciences, Kyungpook National University, Daegu, Republic of Korea
| | - Seung-Dae Choi
- Department of Applied Biosciences, Kyungpook National University, Daegu, Republic of Korea
| | - Kanika Mahra
- Department of Applied Biosciences, Kyungpook National University, Daegu, Republic of Korea
| | - HyunWoo Son
- Microbalance Co. Ltd, Daegu, Republic of Korea
| | - Hoyul Lee
- Research Institute of Aging and Metabolism, Kyungpook National University, Daegu, Republic of Korea
| | - Yu-Jeong Lee
- Cell & Matrix Research Institute, Kyungpook National University, Daegu, Republic of Korea
| | - Eun Soo Kim
- Department of Internal Medicine, School of Medicine, Kyungpook National University, Daegu, Republic of Korea
| | - Jae-Ho Shin
- Department of Applied Biosciences, Kyungpook National University, Daegu, Republic of Korea.
- Microbalance Co. Ltd, Daegu, Republic of Korea.
| |
Collapse
|
42
|
Mertens RT, Misra A, Xiao P, Baek S, Rone JM, Mangani D, Sivanathan KN, Arojojoye AS, Awuah SG, Lee I, Shi GP, Petrova B, Brook JR, Anderson AC, Flavell RA, Kanarek N, Hemberg M, Nowarski R. A metabolic switch orchestrated by IL-18 and the cyclic dinucleotide cGAMP programs intestinal tolerance. Immunity 2024; 57:2077-2094.e12. [PMID: 38906145 DOI: 10.1016/j.immuni.2024.06.001] [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: 10/19/2023] [Revised: 03/10/2024] [Accepted: 06/04/2024] [Indexed: 06/23/2024]
Abstract
Tissues are exposed to diverse inflammatory challenges that shape future inflammatory responses. While cellular metabolism regulates immune function, how metabolism programs and stabilizes immune states within tissues and tunes susceptibility to inflammation is poorly understood. Here, we describe an innate immune metabolic switch that programs long-term intestinal tolerance. Intestinal interleukin-18 (IL-18) stimulation elicited tolerogenic macrophages by preventing their proinflammatory glycolytic polarization via metabolic reprogramming to fatty acid oxidation (FAO). FAO reprogramming was triggered by IL-18 activation of SLC12A3 (NCC), leading to sodium influx, release of mitochondrial DNA, and activation of stimulator of interferon genes (STING). FAO was maintained in macrophages by a bistable switch that encoded memory of IL-18 stimulation and by intercellular positive feedback that sustained the production of macrophage-derived 2'3'-cyclic GMP-AMP (cGAMP) and epithelial-derived IL-18. Thus, a tissue-reinforced metabolic switch encodes durable immune tolerance in the gut and may enable reconstructing compromised immune tolerance in chronic inflammation.
Collapse
Affiliation(s)
- Randall T Mertens
- Gene Lay Institute of Immunology and Inflammation, Brigham and Women's Hospital, Mass General Hospital, and Harvard Medical School, Boston, MA 02115, USA; Ann Romney Center for Neurologic Diseases, Harvard Medical School and Brigham and Women's Hospital, Boston, MA 02115, USA; Department of Immunology, Blavatnik Institute, Harvard Medical School, Boston, MA 02115, USA
| | - Aditya Misra
- Gene Lay Institute of Immunology and Inflammation, Brigham and Women's Hospital, Mass General Hospital, and Harvard Medical School, Boston, MA 02115, USA; Ann Romney Center for Neurologic Diseases, Harvard Medical School and Brigham and Women's Hospital, Boston, MA 02115, USA; Department of Immunology, Blavatnik Institute, Harvard Medical School, Boston, MA 02115, USA; Harvard-MIT Program in Health Sciences and Technology, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Peng Xiao
- Gene Lay Institute of Immunology and Inflammation, Brigham and Women's Hospital, Mass General Hospital, and Harvard Medical School, Boston, MA 02115, USA; Ann Romney Center for Neurologic Diseases, Harvard Medical School and Brigham and Women's Hospital, Boston, MA 02115, USA
| | - Seungbyn Baek
- Department of Biotechnology, College of Life Science and Biotechnology, Yonsei University, Seoul 03722, Republic of Korea
| | - Joseph M Rone
- Gene Lay Institute of Immunology and Inflammation, Brigham and Women's Hospital, Mass General Hospital, and Harvard Medical School, Boston, MA 02115, USA; Ann Romney Center for Neurologic Diseases, Harvard Medical School and Brigham and Women's Hospital, Boston, MA 02115, USA; Department of Immunology, Blavatnik Institute, Harvard Medical School, Boston, MA 02115, USA
| | - Davide Mangani
- Gene Lay Institute of Immunology and Inflammation, Brigham and Women's Hospital, Mass General Hospital, and Harvard Medical School, Boston, MA 02115, USA; Ann Romney Center for Neurologic Diseases, Harvard Medical School and Brigham and Women's Hospital, Boston, MA 02115, USA
| | - Kisha N Sivanathan
- Gene Lay Institute of Immunology and Inflammation, Brigham and Women's Hospital, Mass General Hospital, and Harvard Medical School, Boston, MA 02115, USA; Ann Romney Center for Neurologic Diseases, Harvard Medical School and Brigham and Women's Hospital, Boston, MA 02115, USA; Department of Immunology, Blavatnik Institute, Harvard Medical School, Boston, MA 02115, USA
| | | | - Samuel G Awuah
- Department of Chemistry, University of Kentucky, Lexington, KY 40506, USA; Center for Pharmaceutical Research and Innovation, College of Pharmacy and Department of Pharmaceutical Sciences, College of Pharmacy, University of Kentucky, Lexington, KY, USA
| | - Insuk Lee
- Department of Biotechnology, College of Life Science and Biotechnology, Yonsei University, Seoul 03722, Republic of Korea; POSTECH Biotech Center, Pohang University of Science and Technology (POSTECH), Pohang 37673, Republic of Korea
| | - Guo-Ping Shi
- Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Boryana Petrova
- Department of Pathology, Boston Children's Hospital and Harvard Medical School, Boston, MA 02115, USA
| | - Jeannette R Brook
- Department of Pathology, Boston Children's Hospital and Harvard Medical School, Boston, MA 02115, USA
| | - Ana C Anderson
- Gene Lay Institute of Immunology and Inflammation, Brigham and Women's Hospital, Mass General Hospital, and Harvard Medical School, Boston, MA 02115, USA; Ann Romney Center for Neurologic Diseases, Harvard Medical School and Brigham and Women's Hospital, Boston, MA 02115, USA; Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Richard A Flavell
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT, USA; Howard Hughes Medical Institute, Yale University, New Haven, CT, USA
| | - Naama Kanarek
- Department of Pathology, Boston Children's Hospital and Harvard Medical School, Boston, MA 02115, USA; Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Martin Hemberg
- Gene Lay Institute of Immunology and Inflammation, Brigham and Women's Hospital, Mass General Hospital, and Harvard Medical School, Boston, MA 02115, USA; Ann Romney Center for Neurologic Diseases, Harvard Medical School and Brigham and Women's Hospital, Boston, MA 02115, USA; Department of Immunology, Blavatnik Institute, Harvard Medical School, Boston, MA 02115, USA; Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Roni Nowarski
- Gene Lay Institute of Immunology and Inflammation, Brigham and Women's Hospital, Mass General Hospital, and Harvard Medical School, Boston, MA 02115, USA; Ann Romney Center for Neurologic Diseases, Harvard Medical School and Brigham and Women's Hospital, Boston, MA 02115, USA; Department of Immunology, Blavatnik Institute, Harvard Medical School, Boston, MA 02115, USA; Broad Institute of MIT and Harvard, Cambridge, MA, USA.
| |
Collapse
|
43
|
Erfan R, Shaker OG, Khalil MAF, Mahmoud FAM, Gomaa MS, Abu-El-Azayem AK, Zaki OM, Ahmed AM, Samy A, Mohammed A. Circulating miR-199a and long noncoding-RNA ANRIL as Promising Diagnostic Biomarkers for Inflammatory Bowel Disease. Inflamm Bowel Dis 2024; 30:1500-1509. [PMID: 38190238 DOI: 10.1093/ibd/izad210] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/02/2022] [Indexed: 01/09/2024]
Abstract
BACKGROUND & AIMS Inflammatory bowel disease (IBD), involving both Crohn's disease (CD) and ulcerative colitis (UC), represents a chronic, immune-mediated inflammatory disease due to an uncontrolled, ongoing inflammatory response to intestinal bacteria in those with genetic susceptibility. MicroRNA (miRNA) extrusion from relevant remote organs or tissues is reflected in the expression of miRNAs in serum and plasma. Both UC and CD patients had higher blood levels of expressed miR-199a. Long noncoding RNA (lncRNA) ANRIL is a proinflammatory gene that mediates nuclear factor κB to play a role in inflammatory diseases, such as IBD. The aim of the current study is to investigate the potential role of both miR-199a and ANRIL in diagnosing IBD in adult patients. METHODS Sixty-seven IBD patients diagnosed clinically, radiologically, endoscopically, and histologically were included in this prospective cohort study. Participants were classified into 3 groups: the UC group (n = 35), the CD group (n = 32), and the control group (n = 30). Demographics, history taking, laboratory characteristics, and treatments were recorded. Tumor necrosis factor α, miR-199a, and ANRIL were measured. RESULTS The findings suggested that miR-199a and ANRIL might be associated with the occurrence or progression of IBD because both genes were substantially expressed in the peripheral blood of patients with this condition. Receiver-operating characteristic curve analysis indicated that the detection of miR-199a and ANRIL had a predictive sensitivity of 62.9% and 88.6% and a specificity of 70.7% and 96.7% for the occurrence of UC cases, respectively, and a predictive sensitivity of 72.4% and 46.9% and a specificity of 96.7% and 34.7% for the occurrence of CD cases, respectively. CONCLUSIONS Both miR-199a and ANRIL are abundant in the sera of IBD adult Egyptian patients (UC and CD). Both can represent a noninvasive marker for early disease diagnosis.
Collapse
Affiliation(s)
- Randa Erfan
- Department of Medical Biochemistry and Molecular Biology, Faculty of Medicine, Cairo University, Cairo, Egypt
| | - Olfat G Shaker
- Department of Medical Biochemistry and Molecular Biology, Faculty of Medicine, Cairo University, Cairo, Egypt
| | - Mahmoud A F Khalil
- Department of Microbiology and Immunology, Faculty of Pharmacy, Fayoum University, Fayoum, Egypt
| | - Fatma A M Mahmoud
- Department of Tropical Medicine, Faculty of Medicine, Fayoum University, Fayoum, Egypt
| | - Mohamed S Gomaa
- Department of General Medicine, Faculty of Medicine, Fayoum University, Fayoum, Egypt
| | - Abeer K Abu-El-Azayem
- Department of Medical Microbiology and Immunology, Faculty of Medicine, Cairo University, Cairo, Egypt
| | - Othman M Zaki
- Department of Clinical Pathology, Faculty of Medicine, Damietta University, Damietta, Egypt
| | - Azza M Ahmed
- Department of Rheumatology, Faculty of Medicine, Fayoum University, Fayoum, Egypt
| | - Amira Samy
- Department of Clinical and Chemical Pathology, Faculty of Medicine, Cairo University, Cairo, Egypt
| | - Asmaa Mohammed
- Department of Medical Biochemistry and Molecular Biology, Faculty of Medicine, Fayoum University, Fayoum, Egypt
| |
Collapse
|
44
|
Encarnacion-Garcia MR, De la Torre-Baez R, Hernandez-Cueto MA, Velázquez-Villegas LA, Candelario-Martinez A, Sánchez-Argáez AB, Horta-López PH, Montoya-García A, Jaimes-Ortega GA, Lopez-Bailon L, Piedra-Quintero Z, Carrasco-Torres G, De Ita M, Figueroa-Corona MDP, Muñoz-Medina JE, Sánchez-Uribe M, Ortiz-Fernández A, Meraz-Ríos MA, Silva-Olivares A, Betanzos A, Baay-Guzman GJ, Navarro-Garcia F, Villa-Treviño S, Garcia-Sierra F, Cisneros B, Schnoor M, Ortíz-Navarrete VF, Villegas-Sepúlveda N, Valle-Rios R, Medina-Contreras O, Noriega LG, Nava P. IFN-γ stimulates Paneth cell secretion through necroptosis mTORC1 dependent. Eur J Immunol 2024; 54:e2350716. [PMID: 38837757 DOI: 10.1002/eji.202350716] [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: 08/14/2023] [Revised: 05/22/2024] [Accepted: 05/23/2024] [Indexed: 06/07/2024]
Abstract
Immune mediators affect multiple biological functions of intestinal epithelial cells (IECs) and, like Paneth and Paneth-like cells, play an important role in intestinal epithelial homeostasis. IFN-γ a prototypical proinflammatory cytokine disrupts intestinal epithelial homeostasis. However, the mechanism underlying the process remains unknown. In this study, using in vivo and in vitro models we demonstrate that IFN-γ is spontaneously secreted in the small intestine. Furthermore, we observed that this cytokine stimulates mitochondrial activity, ROS production, and Paneth and Paneth-like cell secretion. Paneth and Paneth-like secretion downstream of IFN-γ, as identified here, is mTORC1 and necroptosis-dependent. Thus, our findings revealed that the pleiotropic function of IFN-γ also includes the regulation of Paneth cell function in the homeostatic gut.
Collapse
Affiliation(s)
- Maria R Encarnacion-Garcia
- Departament of Physiology, Biophysics, and Neurosciences, Center for Research and Advanced Studies of the National Polytechnic Institute (CINVESTAV-IPN), Mexico City, Mexico
| | - Raúl De la Torre-Baez
- Departament of Physiology, Biophysics, and Neurosciences, Center for Research and Advanced Studies of the National Polytechnic Institute (CINVESTAV-IPN), Mexico City, Mexico
| | - María A Hernandez-Cueto
- Clinical Laboratory of Infectology, National Hospital "La Raza" Medical Center, IMSS, Mexico City, Mexico
| | - Laura A Velázquez-Villegas
- Physiology of Nutrition Department, The National Institute of Health Sciences and Nutrition "Salvador Zubirán", Mexico City, Mexico
| | - Aurora Candelario-Martinez
- Departament of Physiology, Biophysics, and Neurosciences, Center for Research and Advanced Studies of the National Polytechnic Institute (CINVESTAV-IPN), Mexico City, Mexico
| | - Ana Beatriz Sánchez-Argáez
- Department of Molecular Biomedicine, Center for Research and Advanced Studies of the National Polytechnic Institute (CINVESTAV-IPN), Mexico City, Mexico
| | - Perla H Horta-López
- Departament of Physiology, Biophysics, and Neurosciences, Center for Research and Advanced Studies of the National Polytechnic Institute (CINVESTAV-IPN), Mexico City, Mexico
| | - Armando Montoya-García
- Department of Molecular Biomedicine, Center for Research and Advanced Studies of the National Polytechnic Institute (CINVESTAV-IPN), Mexico City, Mexico
| | - Gustavo Alberto Jaimes-Ortega
- Immunology and Proteomics Research Laboratory, Children's Hospital of Mexico "Federico Gómez" (HIMFG), Mexico City, Mexico
- Experimental Biology Postgraduate Program, Department of Biological and Health Sciences, Metropolitan Autonomous University (UAM), Mexico City, Mexico
| | - Luis Lopez-Bailon
- Immunology Department and Immunology Postgraduate Program, National School of Biological Sciences of the National Polytechnic Institute (ENCB-IPN), Mexico City, Mexico
| | - Zayda Piedra-Quintero
- Department of Molecular Biomedicine, Center for Research and Advanced Studies of the National Polytechnic Institute (CINVESTAV-IPN), Mexico City, Mexico
| | - Gabriela Carrasco-Torres
- CICATA Unidad Morelos, Instituto Politécnico Nacional, Boulevard de la Tecnología, 1036 Z-1, P 2/2, Atlacholoaya, 62790, México
| | - Marlon De Ita
- Department of Genetics and Molecular Biology, Center for Research and Advanced Studies of the National Polytechnic Institute (CINVESTAV-IPN), Mexico City, Mexico
- Medical Research Unit in Human Genetics, UMAE Children's Hospital, National Medical Center "Siglo XXI", IMSS, Ciudad de México, 06720, Mexico
| | - María Del Pilar Figueroa-Corona
- Department of Molecular Biomedicine, Center for Research and Advanced Studies of the National Polytechnic Institute (CINVESTAV-IPN), Mexico City, Mexico
| | - José Esteban Muñoz-Medina
- Clinical Laboratory of Infectology, National Hospital "La Raza" Medical Center, IMSS, Mexico City, Mexico
| | - Magdalena Sánchez-Uribe
- Pathological Anatomy, Specialized hospital "Dr. Antonio Fraga Mouret", National Hospital "La Raza" Medical Center, IMSS, Ciudad de México, México
| | - Arturo Ortiz-Fernández
- Department of Cell Biology, Center for Research and Advanced Studies of the National Polytechnic Institute (CINVESTAV-IPN), Mexico City, Mexico
| | - Marco Antonio Meraz-Ríos
- Department of Molecular Biomedicine, Center for Research and Advanced Studies of the National Polytechnic Institute (CINVESTAV-IPN), Mexico City, Mexico
| | - Angélica Silva-Olivares
- Departament of Infectomics and Molecular Pathogenesis, Center for Research and Advanced Studies of the National Polytechnic Institute (CINVESTAV-IPN), Mexico City, Mexico
| | - Abigail Betanzos
- Departament of Infectomics and Molecular Pathogenesis, Center for Research and Advanced Studies of the National Polytechnic Institute (CINVESTAV-IPN), Mexico City, Mexico
| | | | - Fernando Navarro-Garcia
- Department of Cell Biology, Center for Research and Advanced Studies of the National Polytechnic Institute (CINVESTAV-IPN), Mexico City, Mexico
| | - Saúl Villa-Treviño
- Department of Cell Biology, Center for Research and Advanced Studies of the National Polytechnic Institute (CINVESTAV-IPN), Mexico City, Mexico
| | - Francisco Garcia-Sierra
- Department of Cell Biology, Center for Research and Advanced Studies of the National Polytechnic Institute (CINVESTAV-IPN), Mexico City, Mexico
| | - Bulmaro Cisneros
- Department of Genetics and Molecular Biology, Center for Research and Advanced Studies of the National Polytechnic Institute (CINVESTAV-IPN), Mexico City, Mexico
| | - Michael Schnoor
- Department of Molecular Biomedicine, Center for Research and Advanced Studies of the National Polytechnic Institute (CINVESTAV-IPN), Mexico City, Mexico
| | - Vianney F Ortíz-Navarrete
- Department of Molecular Biomedicine, Center for Research and Advanced Studies of the National Polytechnic Institute (CINVESTAV-IPN), Mexico City, Mexico
| | - Nicolás Villegas-Sepúlveda
- Department of Molecular Biomedicine, Center for Research and Advanced Studies of the National Polytechnic Institute (CINVESTAV-IPN), Mexico City, Mexico
| | - Ricardo Valle-Rios
- Immunology and Proteomics Research Laboratory, Children's Hospital of Mexico "Federico Gómez" (HIMFG), Mexico City, Mexico
- University Research Unit, Research Division, Faculty of Medicine, National Autonomous University of Mexico-Children's Hospital of Mexico "Federico Gomez" (UNAM-HIMFG), Mexico City, Mexico
| | - Oscar Medina-Contreras
- Epidemiology, Endocrinology & Nutrition Research Unit, Children's Hospital of Mexico "Federico Gomez", Mexico City, Mexico
| | - Lilia G Noriega
- Physiology of Nutrition Department, The National Institute of Health Sciences and Nutrition "Salvador Zubirán", Mexico City, Mexico
| | - Porfirio Nava
- Departament of Physiology, Biophysics, and Neurosciences, Center for Research and Advanced Studies of the National Polytechnic Institute (CINVESTAV-IPN), Mexico City, Mexico
| |
Collapse
|
45
|
Privitera G, Bezzio C, Dal Buono A, Gabbiadini R, Loy L, Brandaleone L, Marcozzi G, Migliorisi G, Armuzzi A. How comparative studies can inform treatment decisions for Crohn's disease. Expert Opin Biol Ther 2024; 24:955-972. [PMID: 39132872 DOI: 10.1080/14712598.2024.2389985] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2024] [Accepted: 08/05/2024] [Indexed: 08/13/2024]
Abstract
INTRODUCTION As new therapies for the treatment of Crohn's disease (CD) are approved, there is an increasing need for evidence that clarifies their positioning and sequencing. AREAS COVERED Comparative effectiveness research (CER) aims to inform physicians' decisions when they choose which intervention (drug or treatment strategy) to administer to their patients. Pragmatic head-to-head trials represent the best tools for CER, but only a few have been published in the IBD field. Network meta-analyses can point toward the superiority of one drug over another, but they do not reflect everyday clinical practice. Finally, real-world evidence complements that coming from head-to-head trials and network meta-analyses, assessing the real-life effectiveness of therapeutic interventions. EXPERT OPINION There is insufficient evidence to create a definitive therapeutic algorithm for CD, but some general considerations can be made. Anti-TNF-α agents seemingly represent the most 'sustainable' first-line choice, considering benefit-harm ratio and costs; vedolizumab, ustekinumab, and risankizumab may be considered as first-line choice when safety issues become prominent. In the event of pharmacodynamic failure, out-of-class swap is to be preferred - possibly with anti-IL23p19 as the best option, with unclear data regarding upadacitinib positioning; a second anti-TNF-α could be considered, as a second choice, after pharmacokinetic failure.
Collapse
Affiliation(s)
- Giuseppe Privitera
- Dipartimento di Scienze della Salute, Università degli Studi di Milano, Milan, Italy
| | - Cristina Bezzio
- IBD Center, IRCCS Humanitas Research Hospital, Rozzano, Milan, Italy
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, Milan, Italy
| | - Arianna Dal Buono
- IBD Center, IRCCS Humanitas Research Hospital, Rozzano, Milan, Italy
| | | | - Laura Loy
- IBD Center, IRCCS Humanitas Research Hospital, Rozzano, Milan, Italy
| | - Luca Brandaleone
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, Milan, Italy
| | - Giacomo Marcozzi
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, Milan, Italy
| | - Giulia Migliorisi
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, Milan, Italy
| | - Alessandro Armuzzi
- IBD Center, IRCCS Humanitas Research Hospital, Rozzano, Milan, Italy
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, Milan, Italy
| |
Collapse
|
46
|
Wen J, Yang Y, Li L, Xie J, Yang J, Zhang F, Duan L, Hao J, Tong Y, He Y. Magnoflorine alleviates dextran sulfate sodium-induced ulcerative colitis via inhibiting JAK2/STAT3 signaling pathway. Phytother Res 2024; 38:4592-4613. [PMID: 39079890 DOI: 10.1002/ptr.8271] [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: 01/17/2024] [Revised: 05/14/2024] [Accepted: 06/05/2024] [Indexed: 10/25/2024]
Abstract
Magnoflorine (Mag), a natural alkaloid component originating from the Ranunculaceae Juss. Family, has a various of pharmacological activities. This study aimed to investigate the therapeutic effects and potential mechanism of Mag on dextran sulfate sodium (DSS)-induced ulcerative colitis (UC) based on comprehensive approaches. Therapeutic effects of Mag on 3% DSS-induced UC mice were analyzed. UHPLC-Q-TOF/MS was performed to investigate the potential metabolites and signaling pathway of Mag on DSS-induced UC. Furthermore, the predicted mRNA and protein levels of JAK2/STAT3 signaling pathway in colon tissue were verified and assessed by qRT-PCR and Western Blotting, respectively. Therapeutic effects of Mag on UC mice were presented in down-regulation serum biochemical indices, alleviating histological damage of colon tissue. Serum untargeted metabolomics analysis showed that the potential mechanism of Mag on UC is mainly associated with the regulation of six biomarkers and 11 pathways, which may be responsible for the therapeutic efficacy of UC. The "component-metabolites-targets" interactive network indicated that Mag exerts its anti-UC effect by regulating PTGS1 and PTGS2, thereby regulating arachidonic acid. Moreover, the results of qRT-PCR showed that Mag could substantially decrease the relative mRNA expression level of Hub genes. In addition, it was found that Mag could inhibit the relative mRNA and protein expression of JAK2/STAT3 signaling pathway. The present results highlighted the role of Mag ameliorated colon injury in DSS-induced UC mice by inhibiting the JAK2/STAT3 signaling pathway. These results suggest that Mag may be an effective agent for the treatment of UC.
Collapse
Affiliation(s)
- Jianxia Wen
- School of Food and Bioengineering, Food Microbiology Key Laboratory of Sichuan Province, Chongqing Key Laboratory of Sichuan Chongqing Joint Construction of Specialty Food, Xihua University, Chengdu, China
| | - Yi Yang
- School of Food and Bioengineering, Food Microbiology Key Laboratory of Sichuan Province, Chongqing Key Laboratory of Sichuan Chongqing Joint Construction of Specialty Food, Xihua University, Chengdu, China
| | - Lu Li
- School of Food and Bioengineering, Food Microbiology Key Laboratory of Sichuan Province, Chongqing Key Laboratory of Sichuan Chongqing Joint Construction of Specialty Food, Xihua University, Chengdu, China
| | - Jiachen Xie
- School of Food and Bioengineering, Food Microbiology Key Laboratory of Sichuan Province, Chongqing Key Laboratory of Sichuan Chongqing Joint Construction of Specialty Food, Xihua University, Chengdu, China
| | - Junjie Yang
- School of Food and Bioengineering, Food Microbiology Key Laboratory of Sichuan Province, Chongqing Key Laboratory of Sichuan Chongqing Joint Construction of Specialty Food, Xihua University, Chengdu, China
| | - Fangling Zhang
- School of Pharmacy, State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Liting Duan
- College of Traditional Chinese Medicine, Yunnan University of Chinese Medicine, Kunming, China
| | - Junjie Hao
- College of Traditional Chinese Medicine, Yunnan University of Chinese Medicine, Kunming, China
| | - Yuling Tong
- School of Medicine and Food, Sichuan Vocational College of Health and Rehabilitation, Zigong, China
| | - Yuxin He
- School of Food and Bioengineering, Food Microbiology Key Laboratory of Sichuan Province, Chongqing Key Laboratory of Sichuan Chongqing Joint Construction of Specialty Food, Xihua University, Chengdu, China
| |
Collapse
|
47
|
Watson S, Cabrera-Silva RI, Parkos CA, Nusrat A, Quiros M. Interferon-gamma signaling drives epithelial TNF-alpha receptor-2 expression during colonic tissue repair. FASEB J 2024; 38:e70001. [PMID: 39139033 DOI: 10.1096/fj.202401695] [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: 07/23/2024] [Accepted: 08/06/2024] [Indexed: 08/15/2024]
Abstract
Interferon-gamma (IFNγ) is traditionally recognized for its pro-inflammatory role during intestinal inflammation. Here, we demonstrate that IFNγ also functions as a pro-repair molecule by increasing TNFα receptor 2 (TNFR2 protein/TNFRSF1B gene) expression on intestinal epithelial cells (IEC) following injury in vitro and in vivo. In silico analyses identified binding sites for the IFNγ signaling transcription factor STAT1 in the promoter region of TNFRSF1B. Scratch-wounded IEC exposed to IFNγ exhibited a STAT1-dependent increase in TNFR2 expression. In situ hybridization revealed elevated Tnfrsf1b mRNA levels in biopsy-induced colonic mucosal wounds, while intraperitoneal administration of IFNγ neutralizing antibodies following mucosal injury resulted in impaired IEC Tnfrsf1b mRNA and inhibited colonic mucosal repair. These findings challenge conventional notions that "pro-inflammatory" mediators solely exacerbate damage by highlighting latent pro-repair functions. Moreover, these results emphasize the critical importance of timing and amount in the synthesis and release of IFNγ and TNFα during the inflammatory process, as they are pivotal in restoring tissue homeostasis.
Collapse
Grants
- DK055679 HHS | NIH | NIDDK | Division of Diabetes, Endocrinology, and Metabolic Diseases (DEM)
- DK059888 HHS | NIH | NIDDK | Division of Diabetes, Endocrinology, and Metabolic Diseases (DEM)
- DK129214 HHS | NIH | NIDDK | Division of Diabetes, Endocrinology, and Metabolic Diseases (DEM)
- DK61739 HHS | NIH | NIDDK | Division of Diabetes, Endocrinology, and Metabolic Diseases (DEM)
- DK72564 HHS | NIH | NIDDK | Division of Diabetes, Endocrinology, and Metabolic Diseases (DEM)
- DK79392 HHS | NIH | NIDDK | Division of Diabetes, Endocrinology, and Metabolic Diseases (DEM)
Collapse
Affiliation(s)
- Sean Watson
- Department of Pathology, School of Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - Rodolfo I Cabrera-Silva
- Department of Pathology, School of Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - Charles A Parkos
- Department of Pathology, School of Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - Asma Nusrat
- Department of Pathology, School of Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - Miguel Quiros
- Department of Pathology, School of Medicine, University of Michigan, Ann Arbor, Michigan, USA
| |
Collapse
|
48
|
Kou RW, Li ZQ, Wang JL, Jiang SQ, Zhang RJ, He YQ, Xia B, Gao JM. Ganoderic Acid A Mitigates Inflammatory Bowel Disease through Modulation of AhR Activity by Microbial Tryptophan Metabolism. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2024; 72:17912-17923. [PMID: 39078661 DOI: 10.1021/acs.jafc.4c01166] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/31/2024]
Abstract
Inflammatory bowel disease (IBD), including Crohn's disease and ulcerative colitis, is a complex gastrointestinal condition influenced by genetic, microbial, and environmental factors, among which the gut microbiota plays a crucial role and has emerged as a potential therapeutic target. Ganoderic acid A (GAA), which is a lanostane triterpenoid compound derived from edible mushroom Ganoderma lucidum, has demonstrated the ability to modulate gut dysbiosis. Thus, we investigated the impact of GAA on IBD using a dextran sodium sulfate (DSS)-induced colitis mouse model. GAA effectively prevented colitis, preserved epithelial and mucus layer integrity, and modulated the gut microbiota. In addition, GAA promoted tryptophan metabolism, especially 3-IAld generation, activated the aryl hydrocarbon receptor (AhR), and induced IL-22 production. Fecal microbiota transplantation validated the mediating role of the gut microbiota in the IBD protection conferred by GAA. Our study suggests that GAA holds potential as a nutritional intervention for ameliorating IBD by influencing the gut microbiota, thereby regulating tryptophan metabolism, enhancing AhR activity, and ultimately improving gut barrier function.
Collapse
Affiliation(s)
- Rong-Wei Kou
- School of Science, Xi'an University of Technology, Xi'an 710048, Shaanxi, People's Republic of China
| | - Zhi-Qing Li
- College of Food Science and Engineering, Northwest A&F University, Yangling 712100, Shaanxi, People's Republic of China
| | - Jia-Lin Wang
- College of Food Science and Engineering, Northwest A&F University, Yangling 712100, Shaanxi, People's Republic of China
| | - Shi-Qi Jiang
- College of Food Science and Engineering, Northwest A&F University, Yangling 712100, Shaanxi, People's Republic of China
| | - Rui-Jing Zhang
- College of Food Science and Engineering, Northwest A&F University, Yangling 712100, Shaanxi, People's Republic of China
| | - Yang-Qing He
- School of Science, Xi'an University of Technology, Xi'an 710048, Shaanxi, People's Republic of China
| | - Bing Xia
- College of Food Science and Engineering, Northwest A&F University, Yangling 712100, Shaanxi, People's Republic of China
| | - Jin-Ming Gao
- Shaanxi Key Laboratory of Natural Products & Chemical Biology, College of Chemistry & Pharmacy, Northwest A&F University, Yangling 712100, Shaanxi, People's Republic of China
| |
Collapse
|
49
|
Sun D, Zhang Z, Xue J. MiRNAs: a new target for Chinese medicine to repair the intestinal barrier in the treatment of ulcerative colitis. Front Pharmacol 2024; 15:1446554. [PMID: 39185319 PMCID: PMC11341499 DOI: 10.3389/fphar.2024.1446554] [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: 06/10/2024] [Accepted: 07/29/2024] [Indexed: 08/27/2024] Open
Abstract
Ulcerative colitis (UC) is a chronic nonspecific inflammatory bowel disease whose pathogenesis remains unclear. Dysfunction of the intestinal mucosal barrier is closely related to the pathogenesis of UC, which is characterised by damage to the colon epithelial barrier, disruption of immune homeostasis, and persistent inflammatory cell infiltration. MicroRNAs (miRNAs) exhibit specific or differential expression in both UC animal models and patients, implicating their involvement in the pathogenesis of UC. In recent years there has been progress in using Traditional Chinese medicine (TCM) to regulate miRNA expression for repairing the intestinal mucosal barrier in UC, as demonstrated in animal and cell experiments. However, it has not been applied in a clinical setting and its underlying molecular mechanisms require further investigation. Therefore, this study systematically described the role of miRNAs in UC-induced intestinal barrier damage and the application of TCM to repair this intestinal barrier by regulating miRNA expression, offering new therapeutic targets for UC treatment.
Collapse
Affiliation(s)
- Dajuan Sun
- The Affiliated Taian City Central Hospital of Qingdao University, Taian, China
- Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Zhongtao Zhang
- The Affiliated Taian City Central Hospital of Qingdao University, Taian, China
| | - Jingwei Xue
- The Affiliated Taian City Central Hospital of Qingdao University, Taian, China
| |
Collapse
|
50
|
Chulkina M, Rohmer C, McAninch S, Panganiban RP, Villéger R, Portolese A, Ciocirlan J, Yang W, Cohen C, Koltun W, Valentine JF, Cong Y, Yochum G, Beswick EJ, Pinchuk IV. Increased Activity of MAPKAPK2 within Mesenchymal Cells as a Target for Inflammation-Associated Fibrosis in Crohn's Disease. J Crohns Colitis 2024; 18:1147-1161. [PMID: 38224550 DOI: 10.1093/ecco-jcc/jjae009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/12/2023] [Revised: 12/14/2023] [Accepted: 01/12/2024] [Indexed: 01/17/2024]
Abstract
BACKGROUND Mesenchymal stromal cells are suggested to play a critical role in Crohn's disease [CD]-associated fibrosis. MAPKAPK2 [MK2] has emerged as a potential therapeutic target to reduce inflammation in CD. However, the cell-specific pattern of phospho-MK2 activation and its role in CD-associated fibrosis are unknown. The objectives of this study were to evaluate cell-specific changes in MK2 activity between predominantly inflammatory CD vs CD with fibrotic complications and define the role of stromal cell-specific MK2 activation in CD-associated fibrosis. METHODS CD tissue, CD tissue-derived mesenchymal stromal cells known as myo-/fibroblasts [CD-MFs], and fibroblast-specific MK2 conditional knockout [KO] mice were used. RESULTS In the inflamed area of predominantly inflammatory CD, high MK2 activity was equally distributed between mesenchymal and haematopoietic cells. By contrast, in CD with fibrotic complications, high MK2 activity was mostly associated with mesenchymal stromal cells. Using ex vivo CD tissue explants and an IL-10KO murine colitis model, we demonstrated that pro-fibrotic responses are significantly reduced by treatment with the MK2 inhibitor PF-3644022. Inhibition of MK2 activity in primary cultures of CD-MFs significantly reduced basal and TGF-β1-induced profibrotic responses. Using fibroblast-specific MK2 knockout mice in chronic dextran saline sulphate colitis, we demonstrated that fibroblast intrinsic MK2 signalling is among the key processes involved in the chronic inflammation-induced profibrotic responses. CONCLUSIONS Our data suggest that activation of MK2 within fibroblasts contributes to the chronic inflammation-induced fibrosis in CD and that targeting MK2 has potential for the development of novel therapeutic approaches for fibrosis in CD.
Collapse
Affiliation(s)
- Marina Chulkina
- Department of Medicine, Penn State College of Medicine, Hershey, PA, USA
| | - Christina Rohmer
- Department of Medicine, Penn State College of Medicine, Hershey, PA, USA
| | - Steven McAninch
- Department of Medicine, Penn State College of Medicine, Hershey, PA, USA
| | | | | | - Austin Portolese
- Department of Surgery, Division of Colon and Rectal Surgery, Penn State Milton S. Hershey Medical Center, Hershey, PA, USA
| | - Justin Ciocirlan
- Department of Medicine, Penn State College of Medicine, Hershey, PA, USA
| | - Wenjing Yang
- Department of Medicine, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Claire Cohen
- Department of Medicine, Penn State College of Medicine, Hershey, PA, USA
| | - Walter Koltun
- Department of Surgery, Division of Colon and Rectal Surgery, Penn State Milton S. Hershey Medical Center, Hershey, PA, USA
| | - John F Valentine
- Department of Internal Medicine, University of Utah School of Medicine, Salt Lake City, UT, USA
| | - Yingzi Cong
- Department of Medicine, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Gregory Yochum
- Department of Surgery, Division of Colon and Rectal Surgery, Penn State Milton S. Hershey Medical Center, Hershey, PA, USA
- Department of Biochemistry and Molecular Biology, Penn State College of Medicine, Hershey, PA, USA
| | - Ellen J Beswick
- Department of Internal Medicine, College of Medicine, University of Kentucky, Lexington, KY, USA
| | - Irina V Pinchuk
- Department of Medicine, Penn State College of Medicine, Hershey, PA, USA
| |
Collapse
|