1
|
Bamias G, Menghini P, Pizarro TT, Cominelli F. Targeting TL1A and DR3: the new frontier of anti-cytokine therapy in IBD. Gut 2025; 74:652-668. [PMID: 39266053 PMCID: PMC11885054 DOI: 10.1136/gutjnl-2024-332504] [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: 06/26/2024] [Accepted: 08/20/2024] [Indexed: 09/14/2024]
Abstract
TNF-like cytokine 1A (TL1A) and its functional receptor, death-domain receptor 3 (DR3), are members of the TNF and TNFR superfamilies, respectively, with recognised roles in regulating innate and adaptive immune responses; additional existence of a decoy receptor, DcR3, indicates a tightly regulated cytokine system. The significance of TL1A:DR3 signalling in the pathogenesis of inflammatory bowel disease (IBD) is supported by several converging lines of evidence. Herein, we aim to provide a comprehensive understanding of what is currently known regarding the TL1A/DR3 system in the context of IBD. TL1A and DR3 are expressed by cellular subsets with important roles for the initiation and maintenance of intestinal inflammation, serving as potent universal costimulators of effector immune responses, indicating their participation in the pathogenesis of IBD. Recent evidence also supports a homoeostatic role for TL1A:DR3 via regulation of Tregs and innate lymphoid cells. TL1A and DR3 are also expressed by stromal cells and may contribute to inflammation-induced or inflammation-independent intestinal fibrogenesis. Finally, discovery of genetic polymorphisms with functional consequences may allow for patient stratification, including differential responses to TL1A-targeted therapeutics. In conclusion, TL1A:DR3 signalling plays a central and multifaceted role in the immunological pathways that underlie intestinal inflammation, such as that observed in IBD. Such evidence provides the foundation for developing pharmaceutical approaches targeting this ligand-receptor pair in IBD.
Collapse
Affiliation(s)
- Giorgos Bamias
- GI Unit, 3rd Academic Department of Internal Medicine, National and Kapodistrian University of Athens, Sotiria Hospital, Athens, Greece
| | - Paola Menghini
- Department of Medicine, Case Western Reserve University School of Medicine, Cleveland, Ohio, USA
- Digestive Health Research Institute, Case Western Reserve University School of Medicine, Cleveland, Ohio, USA
| | - Theresa T Pizarro
- Department of Pathology, Case Western Reserve University School of Medicine, Cleveland, Ohio, USA
| | - Fabio Cominelli
- Department of Medicine, Case Western Reserve University School of Medicine, Cleveland, Ohio, USA
- Digestive Health Research Institute, Case Western Reserve University School of Medicine, Cleveland, Ohio, USA
| |
Collapse
|
2
|
Solitano V, Estevinho MM, Ungaro F, Magro F, Danese S, Jairath V. TL1A Inhibition in Inflammatory Bowel Disease: A Pipeline Review. BioDrugs 2025; 39:171-183. [PMID: 39907869 DOI: 10.1007/s40259-025-00706-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/15/2025] [Indexed: 02/06/2025]
Abstract
Inflammatory bowel disease (IBD), encompassing Crohn's disease (CD) and ulcerative colitis (UC), remains challenging to manage, with a substantial proportion of patients not responding to conventional therapies or developing complications. The tumor necrosis factor (TNF) superfamily member TL1A has emerged as an important player in the pathogenesis of IBD, influencing pathways of inflammation and fibrosis. This leading article reviews the role of TL1A in IBD, evaluates the efficacy of anti-TL1A therapies in clinical trials, and discusses future directions for research and treatment. TL1A is implicated in IBD through its interaction with death domain receptor 3 (DR3), promoting T-cell activation and contributing to both inflammatory responses and fibrotic changes. Phase 2 clinical trials of anti-TL1A agents have demonstrated promising results, showing improvements in endoscopic and histologic outcomes for both UC and CD. Phase 2 and 3 clinical trials are ongoing, which are expected to provide further clarity on the efficacy and safety of TL1A-targeting agents in treating IBD.
Collapse
Affiliation(s)
- Virginia Solitano
- Division of Gastroenterology, Department of Medicine, Western University Schulich School of Medicine, London, ON, Canada.
- Division of Gastroenterology and Gastrointestinal Endoscopy, IRCCS Ospedale San Raffaele, Università Vita-Salute San Raffaele, Milan, Italy.
- Department of Epidemiology and Biostatistics, Western University, London, ON, Canada.
| | - Maria Manuela Estevinho
- Department of Gastroenterology, Unidade Local de Saúde Gaia e Espinho (ULSGE), Vila Nova de Gaia, Portugal
- Department of Biomedicine, Unit of Pharmacology and Therapeutics, Faculty of Medicine, University of Porto, Porto, Portugal
| | - Federica Ungaro
- Division of Gastroenterology, Department of Medicine, Western University Schulich School of Medicine, London, ON, Canada
| | - Fernando Magro
- CINTESIS@RISE, Department of Community Medicine, Information and Health Decision Sciences (MEDCIDS), Faculty of Medicine of the University of Porto (FMUP), Porto, Portugal
- Department of Gastroenterology, Unidade Local de Saúde São João (ULSSJ), Porto, Portugal
| | - Silvio Danese
- Division of Gastroenterology, Department of Medicine, Western University Schulich School of Medicine, London, ON, Canada
| | - Vipul Jairath
- Division of Gastroenterology, Department of Medicine, Western University Schulich School of Medicine, London, ON, Canada
- Department of Epidemiology and Biostatistics, Western University, London, ON, Canada
| |
Collapse
|
3
|
He F, Huang X, Wang Z, Qin M, Chen C, Huang Z, Wu Y, Huang Y, Tang B, Long C, Mo X, Tang W, Liu J. The Effect of Gender on the Intestinal Flora of Colorectal Cancer Under Different Stages. Mol Carcinog 2025; 64:526-542. [PMID: 39692233 DOI: 10.1002/mc.23863] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2024] [Revised: 11/23/2024] [Accepted: 11/26/2024] [Indexed: 12/19/2024]
Abstract
This study aims to determine whether gender is a factor in the interplay between the human intestinal flora and colorectal cancer (CRC), ultimately providing new evidence for the clinical prediction and management of CRC in different genders. In this study, we included 186 untreated CRC patients, and classified them into two groups based on pathological staging: Groups Ⅰ-Ⅱ and Groups Ⅲ-Ⅳ, with male and female groups within each group. We collected preoperative fecal samples from these patients and performed 16S rRNA gene sequencing to analyze their intestinal flora. In the CRC Stages I-II cohort, the gut microbiota of the female group exhibited greater diversity and abundance compared to the male group, with a total of 13 gut microbiota demonstrating significant disparities. Notably, s__Parabacteroides gordonii, s__Bacteroides faecis, and s__Bacteroides nordii were found to be more prevalent in the female group relative to the male group. Within the CRC Stages III-IV cohort, 51 gut microbiota exhibited significant differences between the genders. In the immunocyte composition of fecal samples from patients with CRC, a higher proportion of naive B cells is observed in the male group as compared to the female group. In female CRC patients within the CRC Stages III-IV cohort, Actinomyces exhibited a significant negative correlation with activated dendritic cells, CD4+ memory T cells, and eosinophils. In male CRC patients within the CRC Stages III-IV cohort, Actinomyces demonstrated a significant positive correlation with naive B cells and a significant positive correlation with immune activation genes TNFRSF25 and TMIGD2. In female CRC patients within the CRC Stages III-IV cohort, Actinomyces showed a significant negative correlation with activated dendritic cells, CD4+ memory T cells, and eosinophils, and a significant positive correlation with immune activation genes TNFSF13B, LTA, KLRK1, and CXCL12. In the CRC Stages I-II group, the female group's intestinal flora is more diverse and richer than the male group. In the CRC Stages III-IV group, there are a total of 51 different intestinal flora in both the male and female groups. We also found that Actinomyces affects the occurrence and development of CRC in the male and female groups through different pathways. The results show that the intestinal flora differs between male and female CRC patients and is closely associated with cancer development.
Collapse
Affiliation(s)
- Fuhai He
- Department of Gastrointestinal Surgery, Division of Colorectal and Anal Surgery, Guangxi Medical University Cancer Hospital, Nanning, People's Republic of China
| | - Xiaoliang Huang
- Department of Gastrointestinal Surgery, Division of Colorectal and Anal Surgery, Guangxi Medical University Cancer Hospital, Nanning, People's Republic of China
| | - Zhen Wang
- Department of Gastrointestinal Surgery, Division of Colorectal and Anal Surgery, Guangxi Medical University Cancer Hospital, Nanning, People's Republic of China
| | - Mingjian Qin
- Department of Gastrointestinal Surgery, Division of Colorectal and Anal Surgery, Guangxi Medical University Cancer Hospital, Nanning, People's Republic of China
| | - Chuanbin Chen
- Department of Gastrointestinal Surgery, Division of Colorectal and Anal Surgery, Guangxi Medical University Cancer Hospital, Nanning, People's Republic of China
| | - Zigui Huang
- Department of Gastrointestinal Surgery, Division of Colorectal and Anal Surgery, Guangxi Medical University Cancer Hospital, Nanning, People's Republic of China
| | - Yongzhi Wu
- Department of Gastrointestinal Surgery, Division of Colorectal and Anal Surgery, Guangxi Medical University Cancer Hospital, Nanning, People's Republic of China
| | - Yongqi Huang
- Department of Gastrointestinal Surgery, Division of Colorectal and Anal Surgery, Guangxi Medical University Cancer Hospital, Nanning, People's Republic of China
| | - Binzhe Tang
- Department of Gastrointestinal Surgery, Division of Colorectal and Anal Surgery, Guangxi Medical University Cancer Hospital, Nanning, People's Republic of China
| | - Chenyan Long
- Department of Gastrointestinal Surgery, Division of Colorectal and Anal Surgery, Guangxi Medical University Cancer Hospital, Nanning, People's Republic of China
| | - Xianwei Mo
- Department of Gastrointestinal Surgery, Division of Colorectal and Anal Surgery, Guangxi Medical University Cancer Hospital, Nanning, People's Republic of China
| | - Weizhong Tang
- Department of Gastrointestinal Surgery, Division of Colorectal and Anal Surgery, Guangxi Medical University Cancer Hospital, Nanning, People's Republic of China
| | - Jungang Liu
- Department of Gastrointestinal Surgery, Division of Colorectal and Anal Surgery, Guangxi Medical University Cancer Hospital, Nanning, People's Republic of China
| |
Collapse
|
4
|
Von Ruff ZD, Miller MJ, Moro T, Reidy PT, Ebert SM, Volpi E, Adams CM, Rasmussen BB. Resistance exercise training in older men reduces ATF4-activated and senescence-associated mRNAs in skeletal muscle. GeroScience 2025:10.1007/s11357-025-01564-2. [PMID: 40011348 DOI: 10.1007/s11357-025-01564-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2024] [Accepted: 02/10/2025] [Indexed: 02/28/2025] Open
Abstract
Sarcopenia increases the risk of frailty, morbidity, and mortality in older adults. Resistance exercise training improves muscle size and function; however, the response to exercise training is variable in older adults. The objective of our study was to determine both the age-independent and age-dependent changes to the transcriptome following progressive resistance exercise training. Skeletal muscle biopsies were obtained before and after 12 weeks of resistance exercise training in 8 young (24 ± 3.3 years) and 10 older (72 ± 4.9 years) men. RNA was extracted from each biopsy and prepared for analysis via RNA sequencing. We performed differential mRNA expression, gene ontology, and gene set enrichment analyses. We report that when comparing post-training vs pre-training 226 mRNAs and 959 mRNAs were differentially expressed in the skeletal muscle of young and older men, respectively. Additionally, 94 mRNAs increased, and 17 mRNAs decreased in both young and old, indicating limited overlap in response to resistance exercise training. Furthermore, the differential gene expression was larger in older skeletal muscle. Finally, we report three novel findings: 1) resistance exercise training decreased the abundance of ATF4-activated and senescence-associated skeletal muscle mRNAs in older men; 2) resistance exercise-induced increases in lean mass correlate with increased mRNAs encoding mitochondrial proteins; and 3) increases in muscle strength following resistance exercise positively correlate with increased mRNAs involved in translation, rRNA processing, and polyamine metabolism. We conclude that resistance exercise training elicits a differential gene expression response in young and old skeletal muscle, including reduced ATF-4 activated and senescence-associated gene expression.
Collapse
Affiliation(s)
| | - Matthew J Miller
- Division of Endocrinology, Diabetes, Metabolism and Nutrition, Mayo Clinic, Rochester, MN, USA
- University of Iowa, Iowa City, IA, USA
| | - Tatiana Moro
- Department of Biomedical Sciences, University of Padova, Padua, Italy
| | - Paul T Reidy
- Department of Kinesiology, Nutrition, and Health, Miami University, Oxford, OH, USA
| | - Scott M Ebert
- Division of Endocrinology, Diabetes, Metabolism and Nutrition, Mayo Clinic, Rochester, MN, USA
| | - Elena Volpi
- Barshop Institute for Longevity & Aging Studies, University of Texas Health Science Center at San Antonio, 7703 Floyd Curl Drive, MC 7756, San Antonio, TX, 78229, USA
| | - Christopher M Adams
- Division of Endocrinology, Diabetes, Metabolism and Nutrition, Mayo Clinic, Rochester, MN, USA
| | - Blake B Rasmussen
- Barshop Institute for Longevity & Aging Studies, University of Texas Health Science Center at San Antonio, 7703 Floyd Curl Drive, MC 7756, San Antonio, TX, 78229, USA.
- Department of Cellular & Integrative Physiology, University of Texas Health Science Center at San Antonio, 7703 Floyd Curl Drive, MC 7756, San Antonio, TX, 78229, USA.
| |
Collapse
|
5
|
Varricchi G, Poto R, Criscuolo G, Strisciuglio C, Nair P, Marone G. TL1A, a novel alarmin in airway, intestinal, and autoimmune disorders. J Allergy Clin Immunol 2025:S0091-6749(25)00214-3. [PMID: 40010414 DOI: 10.1016/j.jaci.2025.02.018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2024] [Revised: 02/13/2025] [Accepted: 02/14/2025] [Indexed: 02/28/2025]
Abstract
The term alarmin denotes a broad class of molecules rapidly released to alert the immune system through the engagement of specific receptors on immune cells. Three alarmin cytokines-thymic stromal lymphopoietin, IL-33, and IL-25-are released from epithelial and certain stromal cells. TNF-like cytokine 1A (TL1A) is a member of the TNF cytokine superfamily, first identified in human endothelial cells. TL1A is now considered a novel alarmin expressed by human and mouse bronchial and intestinal epithelial cells. TL1A exerts its biological activities by binding to a trimeric receptor DR3 (death receptor 3), expressed on a wide spectrum of immune and structural cells, including lung fibroblasts, endothelial cells, and bronchial epithelial cells. TL1A has been implicated in experimental and human inflammatory bowel diseases as well as in airway inflammation and remodeling in severe asthma. A monoclonal antibody anti-TL1A (tulisokibart) is effective in inducing clinical remission in ulcerative colitis patients. Increasing evidence suggests that TL1A is also involved in certain autoimmune disorders, such as rheumatoid arthritis and psoriasis. These emerging findings broaden the role of TL1A in various human inflammatory conditions. Several clinical trials are currently evaluating the safety and efficacy of monoclonal antibodies targeting TL1A in asthma or inflammatory bowel disease patients.
Collapse
Affiliation(s)
- Gilda Varricchi
- Department of Translational Medical Sciences, University of Naples Federico II, Naples, Italy; Center for Basic and Clinical Immunology Research (CISI), University of Naples Federico II, Naples, Italy; World Allergy Organization (WAO) Center of Excellence (CoE), Naples, Italy; Istituto Endotipi in Oncologia, Metabolismo e Immunologia "G. Salvatore" (IEOMI), National Research Council (CNR), Naples, Italy.
| | - Remo Poto
- Department of Translational Medical Sciences, University of Naples Federico II, Naples, Italy; World Allergy Organization (WAO) Center of Excellence (CoE), Naples, Italy; Istituti Clinici Scientifici Maugeri-IRCCS Scientific Institute of Telese Terme, Benevento, Italy
| | - Gjada Criscuolo
- Department of Translational Medical Sciences, University of Naples Federico II, Naples, Italy; Center for Basic and Clinical Immunology Research (CISI), University of Naples Federico II, Naples, Italy; World Allergy Organization (WAO) Center of Excellence (CoE), Naples, Italy
| | - Caterina Strisciuglio
- Department of Woman, Child and General and Specialistic Surgery, University of Campania "L. Vanvitelli," Naples, Italy
| | - Parameswaran Nair
- Department of Medicine, McMaster University, Hamilton, Ontario, Canada; Research Institute of St Joe's Hamilton, St Joseph's Healthcare Hamilton, Hamilton, Ontario, Canada
| | - Gianni Marone
- Department of Translational Medical Sciences, University of Naples Federico II, Naples, Italy; Center for Basic and Clinical Immunology Research (CISI), University of Naples Federico II, Naples, Italy; World Allergy Organization (WAO) Center of Excellence (CoE), Naples, Italy
| |
Collapse
|
6
|
Calvez V, Puca P, Di Vincenzo F, Del Gaudio A, Bartocci B, Murgiano M, Iaccarino J, Parand E, Napolitano D, Pugliese D, Gasbarrini A, Scaldaferri F. Novel Insights into the Pathogenesis of Inflammatory Bowel Diseases. Biomedicines 2025; 13:305. [PMID: 40002718 PMCID: PMC11853239 DOI: 10.3390/biomedicines13020305] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2024] [Revised: 01/17/2025] [Accepted: 01/21/2025] [Indexed: 02/27/2025] Open
Abstract
Inflammatory bowel diseases (IBDs), encompassing Crohn's disease and ulcerative colitis, are complex chronic disorders characterized by an intricate interplay between genetic predisposition, immune dysregulation, gut microbiota alterations, and environmental exposures. This review aims to synthesize recent advances in IBD pathogenesis, exploring key mechanisms and potential avenues for prevention and personalized therapy. A comprehensive literature search was conducted across major bibliographic databases, selecting the most recent and impactful studies on IBD pathogenesis. The review integrates findings from multi-omics analyses, single-cell transcriptomics, and longitudinal cohort studies, focusing on immune regulation, gut microbiota dynamics, and environmental factors influencing disease onset and progression. Immune dysregulation, including macrophage polarization (M1 vs. M2) and Th17 activation, emerges as a cornerstone of IBD pathogenesis. Dysbiosis, as a result of reduced alpha and beta diversity and overgrowth of harmful taxa, is one of the main contributing factors in causing inflammation in IBD. Environmental factors, including air and water pollutants, maternal smoking, and antibiotic exposure during pregnancy and infancy, significantly modulate IBD risk through epigenetic and microbiota-mediated mechanisms. While recent advances have supported the development of new therapeutic strategies, deeply understanding the complex dynamics of IBD pathogenesis remains challenging. Future efforts should aim to reduce the burden of disease with precise, personalized treatments and lower the incidence of IBD through early-life prevention and targeted interventions addressing modifiable risk factors.
Collapse
Affiliation(s)
- Valentin Calvez
- IBD Unit, UOC CEMAD Medicina Interna e Gastroenterologia, Centro Malattie dell’Apparato Digerente, Dipartimento di Scienze Mediche e Chirurgiche Addominali ed Endocrino Metaboliche, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, 00168 Rome, Italy; (V.C.); (P.P.); (D.N.); (D.P.)
- Dipartimento di Medicina e Chirurgia Traslazionale, Università Cattolica del Sacro Cuore, 00168 Rome, Italy; (F.D.V.); (A.D.G.); (B.B.); (M.M.); (J.I.); (E.P.); (A.G.)
| | - Pierluigi Puca
- IBD Unit, UOC CEMAD Medicina Interna e Gastroenterologia, Centro Malattie dell’Apparato Digerente, Dipartimento di Scienze Mediche e Chirurgiche Addominali ed Endocrino Metaboliche, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, 00168 Rome, Italy; (V.C.); (P.P.); (D.N.); (D.P.)
- Dipartimento di Medicina e Chirurgia Traslazionale, Università Cattolica del Sacro Cuore, 00168 Rome, Italy; (F.D.V.); (A.D.G.); (B.B.); (M.M.); (J.I.); (E.P.); (A.G.)
| | - Federica Di Vincenzo
- Dipartimento di Medicina e Chirurgia Traslazionale, Università Cattolica del Sacro Cuore, 00168 Rome, Italy; (F.D.V.); (A.D.G.); (B.B.); (M.M.); (J.I.); (E.P.); (A.G.)
| | - Angelo Del Gaudio
- Dipartimento di Medicina e Chirurgia Traslazionale, Università Cattolica del Sacro Cuore, 00168 Rome, Italy; (F.D.V.); (A.D.G.); (B.B.); (M.M.); (J.I.); (E.P.); (A.G.)
| | - Bianca Bartocci
- Dipartimento di Medicina e Chirurgia Traslazionale, Università Cattolica del Sacro Cuore, 00168 Rome, Italy; (F.D.V.); (A.D.G.); (B.B.); (M.M.); (J.I.); (E.P.); (A.G.)
| | - Marco Murgiano
- Dipartimento di Medicina e Chirurgia Traslazionale, Università Cattolica del Sacro Cuore, 00168 Rome, Italy; (F.D.V.); (A.D.G.); (B.B.); (M.M.); (J.I.); (E.P.); (A.G.)
| | - Jacopo Iaccarino
- Dipartimento di Medicina e Chirurgia Traslazionale, Università Cattolica del Sacro Cuore, 00168 Rome, Italy; (F.D.V.); (A.D.G.); (B.B.); (M.M.); (J.I.); (E.P.); (A.G.)
| | - Erfan Parand
- Dipartimento di Medicina e Chirurgia Traslazionale, Università Cattolica del Sacro Cuore, 00168 Rome, Italy; (F.D.V.); (A.D.G.); (B.B.); (M.M.); (J.I.); (E.P.); (A.G.)
| | - Daniele Napolitano
- IBD Unit, UOC CEMAD Medicina Interna e Gastroenterologia, Centro Malattie dell’Apparato Digerente, Dipartimento di Scienze Mediche e Chirurgiche Addominali ed Endocrino Metaboliche, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, 00168 Rome, Italy; (V.C.); (P.P.); (D.N.); (D.P.)
| | - Daniela Pugliese
- IBD Unit, UOC CEMAD Medicina Interna e Gastroenterologia, Centro Malattie dell’Apparato Digerente, Dipartimento di Scienze Mediche e Chirurgiche Addominali ed Endocrino Metaboliche, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, 00168 Rome, Italy; (V.C.); (P.P.); (D.N.); (D.P.)
- Dipartimento di Medicina e Chirurgia Traslazionale, Università Cattolica del Sacro Cuore, 00168 Rome, Italy; (F.D.V.); (A.D.G.); (B.B.); (M.M.); (J.I.); (E.P.); (A.G.)
| | - Antonio Gasbarrini
- Dipartimento di Medicina e Chirurgia Traslazionale, Università Cattolica del Sacro Cuore, 00168 Rome, Italy; (F.D.V.); (A.D.G.); (B.B.); (M.M.); (J.I.); (E.P.); (A.G.)
| | - Franco Scaldaferri
- IBD Unit, UOC CEMAD Medicina Interna e Gastroenterologia, Centro Malattie dell’Apparato Digerente, Dipartimento di Scienze Mediche e Chirurgiche Addominali ed Endocrino Metaboliche, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, 00168 Rome, Italy; (V.C.); (P.P.); (D.N.); (D.P.)
- Dipartimento di Medicina e Chirurgia Traslazionale, Università Cattolica del Sacro Cuore, 00168 Rome, Italy; (F.D.V.); (A.D.G.); (B.B.); (M.M.); (J.I.); (E.P.); (A.G.)
| |
Collapse
|
7
|
Jarmakiewicz-Czaja S, Sokal-Dembowska A, Ferenc K, Filip R. Mechanisms of Insulin Signaling as a Potential Therapeutic Method in Intestinal Diseases. Cells 2024; 13:1879. [PMID: 39594627 PMCID: PMC11593555 DOI: 10.3390/cells13221879] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2024] [Revised: 11/04/2024] [Accepted: 11/12/2024] [Indexed: 11/28/2024] Open
Abstract
Gastrointestinal diseases are becoming a growing public health problem. One of them is inflammatory bowel disease (IBD), which includes ulcerative colitis (UC) and Crohn's disease (CD). The incidence of IBD is increasing in developing countries and declining in developed countries, affecting people of all ages. Researchers have been exploring new treatment options including insulin signaling pathways in the inflammation of the gastrointestinal tract. It seems that a better understanding of the mechanism of IGF-1, GLP-1 and TL1A on the gut microbiota and inflammation may provide new advances in future therapeutic strategies for patients with IBD, but also other intestinal diseases. This review aims to synthesize insights into the effects of GLP, IGF and anti-TL1A on inflammation and the gut microbiota, which may enable their future use in therapy for people with intestinal diseases.
Collapse
Affiliation(s)
- Sara Jarmakiewicz-Czaja
- Institute of Health Sciences, Medical College of Rzeszow University, 35-959 Rzeszow, Poland; (S.J.-C.); (A.S.-D.)
| | - Aneta Sokal-Dembowska
- Institute of Health Sciences, Medical College of Rzeszow University, 35-959 Rzeszow, Poland; (S.J.-C.); (A.S.-D.)
| | - Katarzyna Ferenc
- Institute of Medicine, Medical College of Rzeszow University, 35-959 Rzeszow, Poland;
| | - Rafał Filip
- Institute of Medicine, Medical College of Rzeszow University, 35-959 Rzeszow, Poland;
- Department of Gastroenterology with IBD Unit, Clinical Hospital No. 2, 35-301 Rzeszow, Poland
| |
Collapse
|
8
|
Bilsborough JM, Targan SR. TL1A: A model for a precision medicine approach in the treatment of Crohn's disease and ulcerative colitis. ADVANCES IN PHARMACOLOGY (SAN DIEGO, CALIF.) 2024; 101:287-299. [PMID: 39521604 DOI: 10.1016/bs.apha.2024.10.014] [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
Inflammatory bowel disease (IBD) is a collective term for chronic inflammatory diseases of the intestinal tract. The term IBD encompasses two main forms, Crohn's disease (CD) and Ulcerative colitis (UC). CD is characterized by inflammation throughout the length of the gut, especially the ileum and colon, and is often complicated with fistulae and/or intestinal strictures. Ulcerative colitis (UC) is inflammatory disease restricted to the colon and rectum. In practice however, IBD is a heterogenous disease with CD and UC representing the extremes of a continuum of diseases with varied clinical presentation, including disease location, severity, and manifestation of extraintestinal diseases. This disease heterogeneity poses a challenge to successful and efficacious therapeutic treatment as the etiology driving disease in individual patients is unknown and likely to be multifactorial, including genetic predisposition, environmental factors such as the microbiota, as well as social behaviors such as smoking and diet. Precision medicine provides a strategy to account for disease heterogeneity and diverse etiology to select for patients most likely to respond to a given therapeutic. In this chapter we present an example of the development of a novel antibody therapeutic, Tulisokibart, as a model for a Precision Medicine approach to the successful treatment of patients with IBD.
Collapse
Affiliation(s)
- Janine M Bilsborough
- F. Widjaja Inflammatory Bowel Disease Institute, Los Angeles, CA, United States.
| | - Stephan R Targan
- F. Widjaja Inflammatory Bowel Disease Institute, Los Angeles, CA, United States
| |
Collapse
|
9
|
Sands BE, Feagan BG, Peyrin-Biroulet L, Danese S, Rubin DT, Laurent O, Luo A, Nguyen DD, Lu J, Yen M, Leszczyszyn J, Kempiński R, McGovern DPB, Ma C, Ritter TE, Targan S. Phase 2 Trial of Anti-TL1A Monoclonal Antibody Tulisokibart for Ulcerative Colitis. N Engl J Med 2024; 391:1119-1129. [PMID: 39321363 DOI: 10.1056/nejmoa2314076] [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: 09/27/2024]
Abstract
BACKGROUND Tulisokibart is a tumor necrosis factor-like cytokine 1A (TL1A) monoclonal antibody in development for the treatment of moderately to severely active ulcerative colitis. A genetic-based diagnostic test was designed to identify patients with an increased likelihood of response. METHODS We randomly assigned patients with glucocorticoid dependence or failure of conventional or advanced therapies for ulcerative colitis to receive intravenous tulisokibart (1000 mg on day 1 and 500 mg at weeks 2, 6, and 10) or placebo. Cohort 1 included patients regardless of status with respect to the test for likelihood of response. Cohort 2 included only patients with a positive test for likelihood of response. The primary analysis was performed in cohort 1; the primary end point was clinical remission at week 12. Patients with a positive test for likelihood of response from cohorts 1 and 2 were combined in prespecified analyses. RESULTS In cohort 1, a total of 135 patients underwent randomization. A significantly higher percentage of patients who received tulisokibart had clinical remission than those who received placebo (26% vs. 1%; difference, 25 percentage points; 95% confidence interval [CI], 14 to 37; P<0.001). In cohort 2, a total of 43 patients underwent randomization. A total of 75 patients with a positive test for likelihood of response underwent randomization across both cohorts. Among patients with a positive test for likelihood of response (cohorts 1 and 2 combined), clinical remission occurred in a higher percentage of patients who received tulisokibart than in those who received placebo (32% vs. 11%; difference, 21 percentage points; 95% CI, 2 to 38; P = 0.02). Among all the enrolled patients, the incidence of adverse events was similar in the tulisokibart and placebo groups; most adverse events were mild to moderate in severity. CONCLUSIONS In this short-term trial, tulisokibart was more effective than placebo in inducing clinical remission in patients with moderately to severely active ulcerative colitis. (Funded by Prometheus Biosciences, a subsidiary of Merck; ARTEMIS-UC ClinicalTrials.gov number, NCT04996797.).
Collapse
Affiliation(s)
- Bruce E Sands
- From the Dr. Henry D. Janowitz Division of Gastroenterology, Icahn School of Medicine at Mount Sinai, New York (B.E.S.); the Departments of Medicine, Epidemiology, and Biostatistics, Western University, London, ON (B.G.F.), and the Division of Gastroenterology and Hepatology, Departments of Medicine and Community Health Sciences, University of Calgary, Calgary, AB (C.M.) - both in Canada; the Department of Gastroenterology, INFINY Institute, INSERM NGERE, Centre Hospitalier Régional Universitaire de Nancy, Vandœuvre-lès-Nancy, France (L.P.-B.); the Gastroenterology and Gastrointestinal Endoscopy Unit, IRCCS San Raffaele Scientific Institute, Vita-Salute San Raffaele University, Milan (S.D.); the University of Chicago Medicine Inflammatory Bowel Disease Center, Chicago (D.T.R.); Prometheus Biosciences, a subsidiary of Merck, Rahway, NJ (O.L., A.L., D.D.N., J. Lu, M.Y.); the Department of Gastroenterology, Melita Medical (J. Leszczyszyn), and the Department of Gastroenterology and Hepatology, Wroclaw Medical University (R.K.) - both in Wroclaw, Poland; F. Widjaja Inflammatory Bowel Disease Institute, Cedars-Sinai Medical Center, Los Angeles (D.P.B.M., S.T.); and GI Alliance, Southlake, TX (T.E.R.)
| | - Brian G Feagan
- From the Dr. Henry D. Janowitz Division of Gastroenterology, Icahn School of Medicine at Mount Sinai, New York (B.E.S.); the Departments of Medicine, Epidemiology, and Biostatistics, Western University, London, ON (B.G.F.), and the Division of Gastroenterology and Hepatology, Departments of Medicine and Community Health Sciences, University of Calgary, Calgary, AB (C.M.) - both in Canada; the Department of Gastroenterology, INFINY Institute, INSERM NGERE, Centre Hospitalier Régional Universitaire de Nancy, Vandœuvre-lès-Nancy, France (L.P.-B.); the Gastroenterology and Gastrointestinal Endoscopy Unit, IRCCS San Raffaele Scientific Institute, Vita-Salute San Raffaele University, Milan (S.D.); the University of Chicago Medicine Inflammatory Bowel Disease Center, Chicago (D.T.R.); Prometheus Biosciences, a subsidiary of Merck, Rahway, NJ (O.L., A.L., D.D.N., J. Lu, M.Y.); the Department of Gastroenterology, Melita Medical (J. Leszczyszyn), and the Department of Gastroenterology and Hepatology, Wroclaw Medical University (R.K.) - both in Wroclaw, Poland; F. Widjaja Inflammatory Bowel Disease Institute, Cedars-Sinai Medical Center, Los Angeles (D.P.B.M., S.T.); and GI Alliance, Southlake, TX (T.E.R.)
| | - Laurent Peyrin-Biroulet
- From the Dr. Henry D. Janowitz Division of Gastroenterology, Icahn School of Medicine at Mount Sinai, New York (B.E.S.); the Departments of Medicine, Epidemiology, and Biostatistics, Western University, London, ON (B.G.F.), and the Division of Gastroenterology and Hepatology, Departments of Medicine and Community Health Sciences, University of Calgary, Calgary, AB (C.M.) - both in Canada; the Department of Gastroenterology, INFINY Institute, INSERM NGERE, Centre Hospitalier Régional Universitaire de Nancy, Vandœuvre-lès-Nancy, France (L.P.-B.); the Gastroenterology and Gastrointestinal Endoscopy Unit, IRCCS San Raffaele Scientific Institute, Vita-Salute San Raffaele University, Milan (S.D.); the University of Chicago Medicine Inflammatory Bowel Disease Center, Chicago (D.T.R.); Prometheus Biosciences, a subsidiary of Merck, Rahway, NJ (O.L., A.L., D.D.N., J. Lu, M.Y.); the Department of Gastroenterology, Melita Medical (J. Leszczyszyn), and the Department of Gastroenterology and Hepatology, Wroclaw Medical University (R.K.) - both in Wroclaw, Poland; F. Widjaja Inflammatory Bowel Disease Institute, Cedars-Sinai Medical Center, Los Angeles (D.P.B.M., S.T.); and GI Alliance, Southlake, TX (T.E.R.)
| | - Silvio Danese
- From the Dr. Henry D. Janowitz Division of Gastroenterology, Icahn School of Medicine at Mount Sinai, New York (B.E.S.); the Departments of Medicine, Epidemiology, and Biostatistics, Western University, London, ON (B.G.F.), and the Division of Gastroenterology and Hepatology, Departments of Medicine and Community Health Sciences, University of Calgary, Calgary, AB (C.M.) - both in Canada; the Department of Gastroenterology, INFINY Institute, INSERM NGERE, Centre Hospitalier Régional Universitaire de Nancy, Vandœuvre-lès-Nancy, France (L.P.-B.); the Gastroenterology and Gastrointestinal Endoscopy Unit, IRCCS San Raffaele Scientific Institute, Vita-Salute San Raffaele University, Milan (S.D.); the University of Chicago Medicine Inflammatory Bowel Disease Center, Chicago (D.T.R.); Prometheus Biosciences, a subsidiary of Merck, Rahway, NJ (O.L., A.L., D.D.N., J. Lu, M.Y.); the Department of Gastroenterology, Melita Medical (J. Leszczyszyn), and the Department of Gastroenterology and Hepatology, Wroclaw Medical University (R.K.) - both in Wroclaw, Poland; F. Widjaja Inflammatory Bowel Disease Institute, Cedars-Sinai Medical Center, Los Angeles (D.P.B.M., S.T.); and GI Alliance, Southlake, TX (T.E.R.)
| | - David T Rubin
- From the Dr. Henry D. Janowitz Division of Gastroenterology, Icahn School of Medicine at Mount Sinai, New York (B.E.S.); the Departments of Medicine, Epidemiology, and Biostatistics, Western University, London, ON (B.G.F.), and the Division of Gastroenterology and Hepatology, Departments of Medicine and Community Health Sciences, University of Calgary, Calgary, AB (C.M.) - both in Canada; the Department of Gastroenterology, INFINY Institute, INSERM NGERE, Centre Hospitalier Régional Universitaire de Nancy, Vandœuvre-lès-Nancy, France (L.P.-B.); the Gastroenterology and Gastrointestinal Endoscopy Unit, IRCCS San Raffaele Scientific Institute, Vita-Salute San Raffaele University, Milan (S.D.); the University of Chicago Medicine Inflammatory Bowel Disease Center, Chicago (D.T.R.); Prometheus Biosciences, a subsidiary of Merck, Rahway, NJ (O.L., A.L., D.D.N., J. Lu, M.Y.); the Department of Gastroenterology, Melita Medical (J. Leszczyszyn), and the Department of Gastroenterology and Hepatology, Wroclaw Medical University (R.K.) - both in Wroclaw, Poland; F. Widjaja Inflammatory Bowel Disease Institute, Cedars-Sinai Medical Center, Los Angeles (D.P.B.M., S.T.); and GI Alliance, Southlake, TX (T.E.R.)
| | - Olivier Laurent
- From the Dr. Henry D. Janowitz Division of Gastroenterology, Icahn School of Medicine at Mount Sinai, New York (B.E.S.); the Departments of Medicine, Epidemiology, and Biostatistics, Western University, London, ON (B.G.F.), and the Division of Gastroenterology and Hepatology, Departments of Medicine and Community Health Sciences, University of Calgary, Calgary, AB (C.M.) - both in Canada; the Department of Gastroenterology, INFINY Institute, INSERM NGERE, Centre Hospitalier Régional Universitaire de Nancy, Vandœuvre-lès-Nancy, France (L.P.-B.); the Gastroenterology and Gastrointestinal Endoscopy Unit, IRCCS San Raffaele Scientific Institute, Vita-Salute San Raffaele University, Milan (S.D.); the University of Chicago Medicine Inflammatory Bowel Disease Center, Chicago (D.T.R.); Prometheus Biosciences, a subsidiary of Merck, Rahway, NJ (O.L., A.L., D.D.N., J. Lu, M.Y.); the Department of Gastroenterology, Melita Medical (J. Leszczyszyn), and the Department of Gastroenterology and Hepatology, Wroclaw Medical University (R.K.) - both in Wroclaw, Poland; F. Widjaja Inflammatory Bowel Disease Institute, Cedars-Sinai Medical Center, Los Angeles (D.P.B.M., S.T.); and GI Alliance, Southlake, TX (T.E.R.)
| | - Allison Luo
- From the Dr. Henry D. Janowitz Division of Gastroenterology, Icahn School of Medicine at Mount Sinai, New York (B.E.S.); the Departments of Medicine, Epidemiology, and Biostatistics, Western University, London, ON (B.G.F.), and the Division of Gastroenterology and Hepatology, Departments of Medicine and Community Health Sciences, University of Calgary, Calgary, AB (C.M.) - both in Canada; the Department of Gastroenterology, INFINY Institute, INSERM NGERE, Centre Hospitalier Régional Universitaire de Nancy, Vandœuvre-lès-Nancy, France (L.P.-B.); the Gastroenterology and Gastrointestinal Endoscopy Unit, IRCCS San Raffaele Scientific Institute, Vita-Salute San Raffaele University, Milan (S.D.); the University of Chicago Medicine Inflammatory Bowel Disease Center, Chicago (D.T.R.); Prometheus Biosciences, a subsidiary of Merck, Rahway, NJ (O.L., A.L., D.D.N., J. Lu, M.Y.); the Department of Gastroenterology, Melita Medical (J. Leszczyszyn), and the Department of Gastroenterology and Hepatology, Wroclaw Medical University (R.K.) - both in Wroclaw, Poland; F. Widjaja Inflammatory Bowel Disease Institute, Cedars-Sinai Medical Center, Los Angeles (D.P.B.M., S.T.); and GI Alliance, Southlake, TX (T.E.R.)
| | - Deanna D Nguyen
- From the Dr. Henry D. Janowitz Division of Gastroenterology, Icahn School of Medicine at Mount Sinai, New York (B.E.S.); the Departments of Medicine, Epidemiology, and Biostatistics, Western University, London, ON (B.G.F.), and the Division of Gastroenterology and Hepatology, Departments of Medicine and Community Health Sciences, University of Calgary, Calgary, AB (C.M.) - both in Canada; the Department of Gastroenterology, INFINY Institute, INSERM NGERE, Centre Hospitalier Régional Universitaire de Nancy, Vandœuvre-lès-Nancy, France (L.P.-B.); the Gastroenterology and Gastrointestinal Endoscopy Unit, IRCCS San Raffaele Scientific Institute, Vita-Salute San Raffaele University, Milan (S.D.); the University of Chicago Medicine Inflammatory Bowel Disease Center, Chicago (D.T.R.); Prometheus Biosciences, a subsidiary of Merck, Rahway, NJ (O.L., A.L., D.D.N., J. Lu, M.Y.); the Department of Gastroenterology, Melita Medical (J. Leszczyszyn), and the Department of Gastroenterology and Hepatology, Wroclaw Medical University (R.K.) - both in Wroclaw, Poland; F. Widjaja Inflammatory Bowel Disease Institute, Cedars-Sinai Medical Center, Los Angeles (D.P.B.M., S.T.); and GI Alliance, Southlake, TX (T.E.R.)
| | - Jiandong Lu
- From the Dr. Henry D. Janowitz Division of Gastroenterology, Icahn School of Medicine at Mount Sinai, New York (B.E.S.); the Departments of Medicine, Epidemiology, and Biostatistics, Western University, London, ON (B.G.F.), and the Division of Gastroenterology and Hepatology, Departments of Medicine and Community Health Sciences, University of Calgary, Calgary, AB (C.M.) - both in Canada; the Department of Gastroenterology, INFINY Institute, INSERM NGERE, Centre Hospitalier Régional Universitaire de Nancy, Vandœuvre-lès-Nancy, France (L.P.-B.); the Gastroenterology and Gastrointestinal Endoscopy Unit, IRCCS San Raffaele Scientific Institute, Vita-Salute San Raffaele University, Milan (S.D.); the University of Chicago Medicine Inflammatory Bowel Disease Center, Chicago (D.T.R.); Prometheus Biosciences, a subsidiary of Merck, Rahway, NJ (O.L., A.L., D.D.N., J. Lu, M.Y.); the Department of Gastroenterology, Melita Medical (J. Leszczyszyn), and the Department of Gastroenterology and Hepatology, Wroclaw Medical University (R.K.) - both in Wroclaw, Poland; F. Widjaja Inflammatory Bowel Disease Institute, Cedars-Sinai Medical Center, Los Angeles (D.P.B.M., S.T.); and GI Alliance, Southlake, TX (T.E.R.)
| | - Mark Yen
- From the Dr. Henry D. Janowitz Division of Gastroenterology, Icahn School of Medicine at Mount Sinai, New York (B.E.S.); the Departments of Medicine, Epidemiology, and Biostatistics, Western University, London, ON (B.G.F.), and the Division of Gastroenterology and Hepatology, Departments of Medicine and Community Health Sciences, University of Calgary, Calgary, AB (C.M.) - both in Canada; the Department of Gastroenterology, INFINY Institute, INSERM NGERE, Centre Hospitalier Régional Universitaire de Nancy, Vandœuvre-lès-Nancy, France (L.P.-B.); the Gastroenterology and Gastrointestinal Endoscopy Unit, IRCCS San Raffaele Scientific Institute, Vita-Salute San Raffaele University, Milan (S.D.); the University of Chicago Medicine Inflammatory Bowel Disease Center, Chicago (D.T.R.); Prometheus Biosciences, a subsidiary of Merck, Rahway, NJ (O.L., A.L., D.D.N., J. Lu, M.Y.); the Department of Gastroenterology, Melita Medical (J. Leszczyszyn), and the Department of Gastroenterology and Hepatology, Wroclaw Medical University (R.K.) - both in Wroclaw, Poland; F. Widjaja Inflammatory Bowel Disease Institute, Cedars-Sinai Medical Center, Los Angeles (D.P.B.M., S.T.); and GI Alliance, Southlake, TX (T.E.R.)
| | - Jaroslaw Leszczyszyn
- From the Dr. Henry D. Janowitz Division of Gastroenterology, Icahn School of Medicine at Mount Sinai, New York (B.E.S.); the Departments of Medicine, Epidemiology, and Biostatistics, Western University, London, ON (B.G.F.), and the Division of Gastroenterology and Hepatology, Departments of Medicine and Community Health Sciences, University of Calgary, Calgary, AB (C.M.) - both in Canada; the Department of Gastroenterology, INFINY Institute, INSERM NGERE, Centre Hospitalier Régional Universitaire de Nancy, Vandœuvre-lès-Nancy, France (L.P.-B.); the Gastroenterology and Gastrointestinal Endoscopy Unit, IRCCS San Raffaele Scientific Institute, Vita-Salute San Raffaele University, Milan (S.D.); the University of Chicago Medicine Inflammatory Bowel Disease Center, Chicago (D.T.R.); Prometheus Biosciences, a subsidiary of Merck, Rahway, NJ (O.L., A.L., D.D.N., J. Lu, M.Y.); the Department of Gastroenterology, Melita Medical (J. Leszczyszyn), and the Department of Gastroenterology and Hepatology, Wroclaw Medical University (R.K.) - both in Wroclaw, Poland; F. Widjaja Inflammatory Bowel Disease Institute, Cedars-Sinai Medical Center, Los Angeles (D.P.B.M., S.T.); and GI Alliance, Southlake, TX (T.E.R.)
| | - Radosław Kempiński
- From the Dr. Henry D. Janowitz Division of Gastroenterology, Icahn School of Medicine at Mount Sinai, New York (B.E.S.); the Departments of Medicine, Epidemiology, and Biostatistics, Western University, London, ON (B.G.F.), and the Division of Gastroenterology and Hepatology, Departments of Medicine and Community Health Sciences, University of Calgary, Calgary, AB (C.M.) - both in Canada; the Department of Gastroenterology, INFINY Institute, INSERM NGERE, Centre Hospitalier Régional Universitaire de Nancy, Vandœuvre-lès-Nancy, France (L.P.-B.); the Gastroenterology and Gastrointestinal Endoscopy Unit, IRCCS San Raffaele Scientific Institute, Vita-Salute San Raffaele University, Milan (S.D.); the University of Chicago Medicine Inflammatory Bowel Disease Center, Chicago (D.T.R.); Prometheus Biosciences, a subsidiary of Merck, Rahway, NJ (O.L., A.L., D.D.N., J. Lu, M.Y.); the Department of Gastroenterology, Melita Medical (J. Leszczyszyn), and the Department of Gastroenterology and Hepatology, Wroclaw Medical University (R.K.) - both in Wroclaw, Poland; F. Widjaja Inflammatory Bowel Disease Institute, Cedars-Sinai Medical Center, Los Angeles (D.P.B.M., S.T.); and GI Alliance, Southlake, TX (T.E.R.)
| | - Dermot P B McGovern
- From the Dr. Henry D. Janowitz Division of Gastroenterology, Icahn School of Medicine at Mount Sinai, New York (B.E.S.); the Departments of Medicine, Epidemiology, and Biostatistics, Western University, London, ON (B.G.F.), and the Division of Gastroenterology and Hepatology, Departments of Medicine and Community Health Sciences, University of Calgary, Calgary, AB (C.M.) - both in Canada; the Department of Gastroenterology, INFINY Institute, INSERM NGERE, Centre Hospitalier Régional Universitaire de Nancy, Vandœuvre-lès-Nancy, France (L.P.-B.); the Gastroenterology and Gastrointestinal Endoscopy Unit, IRCCS San Raffaele Scientific Institute, Vita-Salute San Raffaele University, Milan (S.D.); the University of Chicago Medicine Inflammatory Bowel Disease Center, Chicago (D.T.R.); Prometheus Biosciences, a subsidiary of Merck, Rahway, NJ (O.L., A.L., D.D.N., J. Lu, M.Y.); the Department of Gastroenterology, Melita Medical (J. Leszczyszyn), and the Department of Gastroenterology and Hepatology, Wroclaw Medical University (R.K.) - both in Wroclaw, Poland; F. Widjaja Inflammatory Bowel Disease Institute, Cedars-Sinai Medical Center, Los Angeles (D.P.B.M., S.T.); and GI Alliance, Southlake, TX (T.E.R.)
| | - Christopher Ma
- From the Dr. Henry D. Janowitz Division of Gastroenterology, Icahn School of Medicine at Mount Sinai, New York (B.E.S.); the Departments of Medicine, Epidemiology, and Biostatistics, Western University, London, ON (B.G.F.), and the Division of Gastroenterology and Hepatology, Departments of Medicine and Community Health Sciences, University of Calgary, Calgary, AB (C.M.) - both in Canada; the Department of Gastroenterology, INFINY Institute, INSERM NGERE, Centre Hospitalier Régional Universitaire de Nancy, Vandœuvre-lès-Nancy, France (L.P.-B.); the Gastroenterology and Gastrointestinal Endoscopy Unit, IRCCS San Raffaele Scientific Institute, Vita-Salute San Raffaele University, Milan (S.D.); the University of Chicago Medicine Inflammatory Bowel Disease Center, Chicago (D.T.R.); Prometheus Biosciences, a subsidiary of Merck, Rahway, NJ (O.L., A.L., D.D.N., J. Lu, M.Y.); the Department of Gastroenterology, Melita Medical (J. Leszczyszyn), and the Department of Gastroenterology and Hepatology, Wroclaw Medical University (R.K.) - both in Wroclaw, Poland; F. Widjaja Inflammatory Bowel Disease Institute, Cedars-Sinai Medical Center, Los Angeles (D.P.B.M., S.T.); and GI Alliance, Southlake, TX (T.E.R.)
| | - Timothy E Ritter
- From the Dr. Henry D. Janowitz Division of Gastroenterology, Icahn School of Medicine at Mount Sinai, New York (B.E.S.); the Departments of Medicine, Epidemiology, and Biostatistics, Western University, London, ON (B.G.F.), and the Division of Gastroenterology and Hepatology, Departments of Medicine and Community Health Sciences, University of Calgary, Calgary, AB (C.M.) - both in Canada; the Department of Gastroenterology, INFINY Institute, INSERM NGERE, Centre Hospitalier Régional Universitaire de Nancy, Vandœuvre-lès-Nancy, France (L.P.-B.); the Gastroenterology and Gastrointestinal Endoscopy Unit, IRCCS San Raffaele Scientific Institute, Vita-Salute San Raffaele University, Milan (S.D.); the University of Chicago Medicine Inflammatory Bowel Disease Center, Chicago (D.T.R.); Prometheus Biosciences, a subsidiary of Merck, Rahway, NJ (O.L., A.L., D.D.N., J. Lu, M.Y.); the Department of Gastroenterology, Melita Medical (J. Leszczyszyn), and the Department of Gastroenterology and Hepatology, Wroclaw Medical University (R.K.) - both in Wroclaw, Poland; F. Widjaja Inflammatory Bowel Disease Institute, Cedars-Sinai Medical Center, Los Angeles (D.P.B.M., S.T.); and GI Alliance, Southlake, TX (T.E.R.)
| | - Stephan Targan
- From the Dr. Henry D. Janowitz Division of Gastroenterology, Icahn School of Medicine at Mount Sinai, New York (B.E.S.); the Departments of Medicine, Epidemiology, and Biostatistics, Western University, London, ON (B.G.F.), and the Division of Gastroenterology and Hepatology, Departments of Medicine and Community Health Sciences, University of Calgary, Calgary, AB (C.M.) - both in Canada; the Department of Gastroenterology, INFINY Institute, INSERM NGERE, Centre Hospitalier Régional Universitaire de Nancy, Vandœuvre-lès-Nancy, France (L.P.-B.); the Gastroenterology and Gastrointestinal Endoscopy Unit, IRCCS San Raffaele Scientific Institute, Vita-Salute San Raffaele University, Milan (S.D.); the University of Chicago Medicine Inflammatory Bowel Disease Center, Chicago (D.T.R.); Prometheus Biosciences, a subsidiary of Merck, Rahway, NJ (O.L., A.L., D.D.N., J. Lu, M.Y.); the Department of Gastroenterology, Melita Medical (J. Leszczyszyn), and the Department of Gastroenterology and Hepatology, Wroclaw Medical University (R.K.) - both in Wroclaw, Poland; F. Widjaja Inflammatory Bowel Disease Institute, Cedars-Sinai Medical Center, Los Angeles (D.P.B.M., S.T.); and GI Alliance, Southlake, TX (T.E.R.)
| |
Collapse
|
10
|
Ma C, Huang J, Zheng Y, Na Y, Wei J, Shan J, Meng K, Zhang X, Zhang S, Wen Y, Ding J. Anti-TL1A monoclonal antibody modulates the dysregulation of Th1/Th17 cells and attenuates granuloma formation in sarcoidosis by inhibiting the PI3K/AKT signaling pathway. Int Immunopharmacol 2024; 137:112360. [PMID: 38852524 DOI: 10.1016/j.intimp.2024.112360] [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: 03/19/2024] [Revised: 05/26/2024] [Accepted: 05/26/2024] [Indexed: 06/11/2024]
Abstract
Sarcoidosis is a systemic granulomatous disease characterized by non-caseating epithelioid cell granulomas. One of its immunological hallmarks is the differentiation of CD4 + naïve T cells into Th1/Th17 cells, accompanied by the release of numerous pro-inflammatory cytokines. The TL1A/DR3 signaling pathway plays a crucial role in activating effector lymphocytes, thereby triggering pro-inflammatory responses. The primary aim of this investigation was to scrutinize the impact of anti-TL1A monoclonal antibody on the dysregulation of Th1/Th17 cells and granuloma formation in sarcoidosis. Initially, the abnormal activation of the TL1A/DR3 signaling pathway in pulmonary tissues of sarcoidosis patients was confirmed using qPCR and immunohistochemistry techniques. Subsequently, employing a murine model of sarcoidosis, the inhibitory effects of anti-TL1A monoclonal antibody on the TL1A/DR3 signaling pathway in sarcoidosis were investigated through qPCR, immunohistochemistry, and Western blot experiments. The influence of anti-TL1A monoclonal antibody on granulomas was assessed through HE staining, while their effects on sarcoidosis Th1/Th17 cells and associated cytokine mRNA levels were evaluated using flow cytometry and qPCR, respectively. Immunofluorescence and Western blot experiments corroborated the inhibitory effects of anti-TL1A monoclonal antibody on the aberrant activation of the PI3K/AKT signaling pathway in sarcoidosis. The findings of this study indicate that the TL1A/DR3 signaling pathway is excessively activated in sarcoidosis. Anti-TL1A monoclonal antibody effectively inhibit this abnormal activation in sarcoidosis, thereby alleviating the dysregulation of Th1/Th17 cells and reducing the formation of pulmonary granulomas. This effect may be associated with the inhibition of the downstream PI3K/AKT signaling pathway. Anti-TL1A monoclonal antibody hold promise as a potential novel therapeutic intervention for sarcoidosis.
Collapse
Affiliation(s)
- Chengxing Ma
- Department of Respiratory and Critical Care Medicine, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, China
| | - Jiayi Huang
- Department of Respiratory and Critical Care Medicine, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, China
| | - Yin Zheng
- Department of Respiratory and Critical Care Medicine, Nanjing Drum Tower Hospital Clinical College of Nanjing Medical University, Nanjing, China
| | - Yuqi Na
- Department of Respiratory and Critical Care Medicine, Nanjing Drum Tower Hospital Clinical College of Nanjing Medical University, Nanjing, China
| | - Jia Wei
- Department of Respiratory and Critical Care Medicine, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, China
| | - Jiajia Shan
- Department of Basic Medicine, Center of Translational Medicine, Jiangsu Key Laboratory of Molecular Medicine, Nanjing University Medical School, Nanjing, China
| | - Kaifang Meng
- Department of Respiratory and Critical Care Medicine, Nanjing Drum Tower Hospital Clinical College of Nanjing Medical University, Nanjing, China
| | - Xian Zhang
- Department of Basic Medicine, Center of Translational Medicine, Jiangsu Key Laboratory of Molecular Medicine, Nanjing University Medical School, Nanjing, China
| | - Shiyun Zhang
- Department of Basic Medicine, Center of Translational Medicine, Jiangsu Key Laboratory of Molecular Medicine, Nanjing University Medical School, Nanjing, China
| | - Yanting Wen
- Department of Basic Medicine, Center of Translational Medicine, Jiangsu Key Laboratory of Molecular Medicine, Nanjing University Medical School, Nanjing, China.
| | - Jingjing Ding
- Department of Respiratory and Critical Care Medicine, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, China.
| |
Collapse
|
11
|
Niese ML, Pajulas AL, Rostron CR, Cheung CCL, Krishnan MS, Zhang J, Cannon AM, Kaplan MH. TL1A priming induces a multi-cytokine Th9 cell phenotype that promotes robust allergic inflammation in murine models of asthma. Mucosal Immunol 2024; 17:537-553. [PMID: 38493956 PMCID: PMC11354665 DOI: 10.1016/j.mucimm.2024.03.006] [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: 10/18/2023] [Revised: 02/22/2024] [Accepted: 03/11/2024] [Indexed: 03/19/2024]
Abstract
Multi-cytokine-producing Th9 cells secrete IL-9 and type 2 cytokines and mediate mouse and human allergic inflammation. However, the cytokines that promote a multi-cytokine secreting phenotype have not been defined. Tumor necrosis factor superfamily member TL1A signals through its receptor DR3 to increase IL-9. Here we demonstrate that TL1A increases expression of IL-9 and IL-13 co-expressing cells in murine Th9 cell cultures, inducing a multi-cytokine phenotype. Mechanistically, this is linked to histone modifications allowing for increased accessibility at the Il9 and Il13 loci. We further show that TL1A alters the transcription factor network underlying expression of IL-9 and IL-13 in Th9 cells and increases binding of transcription factors to Il9 and Il13 loci. TL1A-priming enhances the pathogenicity of Th9 cells in murine models of allergic airway disease through the increased expression of IL-9 and IL-13. Lastly, in both chronic and memory-recall models of allergic airway disease, blockade of TL1A signaling decreases the multi-cytokine Th9 cell population and attenuates the allergic phenotype. Taken together, these data demonstrate that TL1A promotes the development of multi-cytokine Th9 cells that drive allergic airway diseases and that targeting pathogenic T helper cell-promoting cytokines could be an effective approach for modifying disease.
Collapse
Affiliation(s)
- Michelle L Niese
- Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Abigail L Pajulas
- Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Cameron R Rostron
- Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Cherry C L Cheung
- Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Maya S Krishnan
- Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Jilu Zhang
- Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Anthony M Cannon
- Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Mark H Kaplan
- Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis, IN, USA.
| |
Collapse
|
12
|
Schweckendiek D, Rogler G. Antibodies Targeting the Tumor Necrosis Factor-Like Ligand 1A in Inflammatory Bowel Disease: A New Kid on the (Biologics) Block? Digestion 2024; 105:411-418. [PMID: 39068930 PMCID: PMC11633870 DOI: 10.1159/000540421] [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: 05/03/2023] [Accepted: 07/16/2024] [Indexed: 07/30/2024]
Abstract
BACKGROUND The treatment options for inflammatory bowel disease (IBD) have grown over the last years. However, a significant fraction of patients either do not respond to their treatment or lose response over time. SUMMARY Future treatment options could include antibodies that target the tumor necrosis factor-like ligand 1A (TL1A). TL1A is a key cytokine involved in the pathogenesis of a variety of autoimmune diseases including IBD. Studies have shown that IBD disease severity correlates well with serum levels of TL1A. Phase 2 data from two agents currently in clinical testing have been released. In line with requirements for modern therapeutics, companion diagnostic was part of these trials. This aims to identify those patients that are more likely to respond to the agents tested. KEY MESSAGES With regard to the available data the risk/benefit profile of TL1A inhibitors seems to be promising. This article gives a short update and overview, where we are at this point in time with antibodies targeting the TL1A protein in IBD.
Collapse
Affiliation(s)
- Daniel Schweckendiek
- Department of Gastroenterology and Hepatology, University Hospital Zurich, Zurich, Switzerland
| | - Gerhard Rogler
- Department of Gastroenterology and Hepatology, University Hospital Zurich, Zurich, Switzerland
| |
Collapse
|
13
|
Chang SR, Chou CH, Tu HF, Liu CJ, Chang KW, Lin SC. The expression of immune co-stimulators as a prognostic predictor of head and neck squamous cell carcinomas and oral squamous cell carcinomas. J Dent Sci 2024; 19:1380-1388. [PMID: 39035328 PMCID: PMC11259670 DOI: 10.1016/j.jds.2024.05.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2024] [Revised: 05/06/2024] [Indexed: 07/23/2024] Open
Abstract
Background/purpose T cells require second immune checkpoint molecules for activation and immune memory after antigen presentation. We found that inducible co-stimulator (ICOS) has been a favorable prognostic factor amongst B7 immune checkpoint co-stimulators (ICSs) families in head and neck squamous cell carcinoma (HNSCC) and oral SCC (OSCC). Materials and methods This study analyzed the expression of non-B7 tumor necrosis factor (TNF) superfamily ICSs in the Cancer Genome Atlas (TCGA) HNSCC cohort, our OSCC cohort, and TCGA pan-cancer datasets. The correlation in expression, prognosis, and immune status was assessed. Results The higher expression of CD27, CD30, CD40L, death domain 3 (DR3), and OX40, presumably on the T cell surface, defined better overall survival of HNSCC patients. Besides, CD27, CD30, CD40L, and OX40 were highly correlated with ICOS expression in tumors. CD27, CD40L, and DR3 expression are higher in HPV+ HNSCC tumors than in HPV- tumors. The combined expression level of CD27/OX40 or CD27/CD40L/OX40 enables the potent survival prediction of small, less nodal involvement, early stage, and HPV + tumor subsets. Tumors expressing high CD27, CD30, CD40L, ICOS, and OX40 exhibited enhanced immune cell infiltration. The high correlation in the expression of these ICSs was also noted in the vast majority of tumor types in TCGA datasets. Conclusion The findings of this study not only confirm the potential of the concordant stimulation of CD27, CD30, CD40L, ICOS, and OX40 as a crucial strategy in cancer immunotherapy but also inspire further exploration into the field, highlighting the promising future of cancer treatment.
Collapse
Affiliation(s)
- Shi-Rou Chang
- Institute of Oral Biology, College of Dentistry, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Chung-Hsien Chou
- Institute of Oral Biology, College of Dentistry, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Hsi-Feng Tu
- Institute of Oral Biology, College of Dentistry, National Yang Ming Chiao Tung University, Taipei, Taiwan
- Department of Dentistry, College of Dentistry, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Chung-Ji Liu
- Department of Dentistry, College of Dentistry, National Yang Ming Chiao Tung University, Taipei, Taiwan
- Department of Dentistry, Taipei Mackay Memorial Hospital, Taipei, Taiwan
| | - Kuo-Wei Chang
- Institute of Oral Biology, College of Dentistry, National Yang Ming Chiao Tung University, Taipei, Taiwan
- Department of Dentistry, College of Dentistry, National Yang Ming Chiao Tung University, Taipei, Taiwan
- Department of Stomatology, Taipei Veterans General Hospital, Taipei, Taiwan
| | - Shu-Chun Lin
- Institute of Oral Biology, College of Dentistry, National Yang Ming Chiao Tung University, Taipei, Taiwan
- Department of Dentistry, College of Dentistry, National Yang Ming Chiao Tung University, Taipei, Taiwan
- Department of Stomatology, Taipei Veterans General Hospital, Taipei, Taiwan
| |
Collapse
|
14
|
Pires S, Longman RS. Sounding the alarm in the lung with TL1A. J Exp Med 2024; 221:e20240389. [PMID: 38597953 PMCID: PMC11010314 DOI: 10.1084/jem.20240389] [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: 04/11/2024] Open
Abstract
Environmental airborne antigens are central to the development of allergic asthma, but the cellular processes that trigger disease remain incompletely understood. In this report, Schmitt et al. (https://doi.org/10.1084/jem.20231236) identify TNF-like protein 1A (TL1A) as an epithelial alarmin constitutively expressed by a subset of lung epithelial cells, which is released in response to airborne microbial challenge and synergizes with IL-33 to drive allergic disease.
Collapse
Affiliation(s)
- Silvia Pires
- Division of Gastroenterology and Hepatology, Department of Medicine, Jill Roberts Center and Institute for Research in Inflammatory Bowel Disease, Weill Cornell Medicine, New York, NY, USA
| | - Randy S. Longman
- Division of Gastroenterology and Hepatology, Department of Medicine, Jill Roberts Center and Institute for Research in Inflammatory Bowel Disease, Weill Cornell Medicine, New York, NY, USA
| |
Collapse
|
15
|
Solitano V, Jairath V, Ungaro F, Peyrin-Biroulet L, Danese S. TL1A inhibition for inflammatory bowel disease treatment: From inflammation to fibrosis. MED 2024; 5:386-400. [PMID: 38574740 DOI: 10.1016/j.medj.2024.03.010] [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: 12/15/2023] [Revised: 02/09/2024] [Accepted: 03/13/2024] [Indexed: 04/06/2024]
Abstract
The pivotal role of TL1A in modulating immune pathways crucial for inflammatory bowel disease (IBD) and intestinal fibrosis offers a promising therapeutic target. Phase 2 trials (TUSCANY and ARTEMIS-UC) evaluating an anti-TL1A antibody show progress in expanding IBD therapeutic options. First-in-human data reveal reduced expression of genes associated with extracellular matrix remodeling and fibrosis post-anti-TL1A treatment. Investigational drug TEV-48574, potentially exerting dual antifibrotic and anti-inflammatory effects, is undergoing a phase 2 basket study in both ulcerative colitis (UC) and Crohn disease (CD). Results are eagerly awaited, marking advancements in IBD therapeutics. This critical review comprehensively examines the existing literature, illuminating TL1A and the intricate role of DR3 in IBD, emphasizing the evolving therapeutic landscape and ongoing clinical trials, with potential implications for more effective IBD management.
Collapse
Affiliation(s)
- Virginia Solitano
- Division of Gastroenterology, Western University, London, ON, Canada; Gastroenterology and Endoscopy, IRCCS Ospedale San Raffaele and Vita-Salute San Raffaele University, Milan, Italy
| | - Vipul Jairath
- Division of Gastroenterology, Western University, London, ON, Canada; Department of Epidemiology and Biostatistics, Western University, London, ON, Canada
| | - Federica Ungaro
- Gastroenterology and Endoscopy, IRCCS Ospedale San Raffaele and Vita-Salute San Raffaele University, Milan, Italy; Division of Immunology, Transplantation, and Infectious Disease, IRCCS Ospedale San Raffaele, Milan, Italy
| | - Laurent Peyrin-Biroulet
- Department of Gastroenterology and Inserm NGERE U1256, University Hospital of Nancy, University of Lorraine, Vandoeuvre-lès-Nancy, France; Department of Gastroenterology, Nancy University Hospital, Vandœuvre-lès-Nancy, France; INFINY Institute, Nancy University Hospital, Vandœuvre-lès-Nancy, France; FHU-CURE, Nancy University Hospital, Vandœuvre-lès-Nancy, France; Groupe Hospitalier privé Ambroise Paré - Hartmann, Paris IBD Center, Neuilly sur Seine, France; Division of Gastroenterology and Hepatology, McGill University Health Centre, Montreal, QC, Canada
| | - Silvio Danese
- Gastroenterology and Endoscopy, IRCCS Ospedale San Raffaele and Vita-Salute San Raffaele University, Milan, Italy.
| |
Collapse
|
16
|
Li J, Wang L, Ma Y, Liu Y. Inflammatory bowel disease and allergic diseases: A Mendelian randomization study. Pediatr Allergy Immunol 2024; 35:e14147. [PMID: 38773751 DOI: 10.1111/pai.14147] [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: 11/16/2023] [Revised: 04/25/2024] [Accepted: 05/06/2024] [Indexed: 05/24/2024]
Abstract
BACKGROUND Inflammatory bowel disease (IBD) and allergic diseases possess similar genetic backgrounds and pathogenesis. Observational studies have shown a correlation, but the exact direction of cause and effect remains unclear. The aim of this Mendelian randomization (MR) study is to assess bidirectional causality between inflammatory bowel disease and allergic diseases. METHOD We comprehensively analyzed the causal relationship between inflammatory bowel disease (IBD), Crohn's disease (CD), ulcerative colitis (UC) and allergic disease (asthma, Hay fever, and eczema) as a whole, allergic conjunctivitis (AC), atopic dermatitis (AD), allergic asthma (AAS), and allergic rhinitis (AR) by performing a bidirectional Mendelian randomization study using summary-level data from genome-wide association studies. The analysis results mainly came from the random-effects model of inverse variance weighted (IVW-RE). In addition, multivariate Mendelian randomization (MVMR) analysis was conducted to adjust the effect of body mass index (BMI) on the instrumental variables. RESULTS The IVW-RE method revealed that IBD genetically increased the risk of allergic disease as a whole (OR = 1.03, 95% CI = 1.01-1.04, fdr.p = .015), AC (OR = 1.04, 95% CI = 1.01-1.06, fdr.p = .011), and AD (OR = 1.06, 95% CI = 1.02-1.09, fdr.p = .004). Subgroup analysis further confirmed that CD increased the risk of allergic disease as a whole (OR = 1.02, 95% CI = 1.00-1.03, fdr.p = .031), AC (OR = 1.03, 95% CI = 1.01-1.05, fdr.p = .012), AD (OR = 1.06, 95% CI = 1.02-1.09, fdr.p = 2E-05), AAS (OR = 1.05, 95% CI = 1.02-1.08, fdr.p = .002) and AR (OR = 1.03, 95% CI = 1.00-1.07, fdr.p = .025), UC increased the risk of AAS (OR = 1.02, 95% CI = 0.98-1.07, fdr.p = .038). MVMR results showed that after taking BMI as secondary exposure, the causal effects of IBD on AC, IBD on AD, CD on allergic disease as a whole, CD on AC, CD on AD, CD on AAS, and CD on AR were still statistically significant. No significant association was observed in the reverse MR analysis. CONCLUSION This Mendelian randomized study demonstrated that IBD is a risk factor for allergic diseases, which is largely attributed to its subtype CD increasing the risk of AC, AD, ASS, and AR. Further investigations are needed to explore the causal relationship between allergic diseases and IBD.
Collapse
Affiliation(s)
- Jiawei Li
- School of Basic Medical Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Lijun Wang
- School of Basic Medical Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Yuqi Ma
- School of Basic Medical Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Yuan Liu
- School of Basic Medical Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| |
Collapse
|
17
|
Liman N, Lanasa D, Meylan F, Park JH. The ever-expanding role of cytokine receptor DR3 in T cells. Cytokine 2024; 176:156540. [PMID: 38359559 PMCID: PMC10895922 DOI: 10.1016/j.cyto.2024.156540] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2023] [Revised: 02/05/2024] [Accepted: 02/06/2024] [Indexed: 02/17/2024]
Abstract
Death Receptor 3 (DR3) is a cytokine receptor of the Tumor Necrosis Factor receptor superfamily that plays a multifaceted role in both innate and adaptive immunity. Based on the death domain motif in its cytosolic tail, DR3 had been proposed and functionally affirmed as a trigger of apoptosis. Further studies, however, also revealed roles of DR3 in other cellular pathways, including inflammation, survival, and proliferation. DR3 is expressed in various cell types, including T cells, B cells, innate lymphocytes, myeloid cells, fibroblasts, and even outside the immune system. Because DR3 is mainly expressed on T cells, DR3-mediated immune perturbations leading to autoimmunity and other diseases were mostly attributed to DR3 activation of T cells. However, which T cell subset and what T effector functions are controlled by DR3 to drive these processes remain incompletely understood. DR3 engagement was previously found to alter CD4 T helper subset differentiation, expand the Foxp3+ Treg cell pool, and maintain intraepithelial γδ T cells in the gut. Recent studies further unveiled a previously unacknowledged aspect of DR3 in regulating innate-like invariant NKT (iNKT) cell activation, expanding the scope of DR3-mediated immunity in T lineage cells. Importantly, in the context of iNKT cells, DR3 ligation exerted costimulatory effects in agonistic TCR signaling, unveiling a new regulatory framework in T cell activation and proliferation. The current review is aimed at summarizing such recent findings on the role of DR3 on conventional T cells and innate-like T cells and discussing them in the context of immunopathogenesis.
Collapse
Affiliation(s)
- Nurcin Liman
- Experimental Immunology Branch, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, MD 20892, United States
| | - Dominic Lanasa
- Experimental Immunology Branch, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, MD 20892, United States
| | - Françoise Meylan
- Office of Science and Technology, National Institute of Arthritis, Musculoskeletal, and Skin Diseases, NIH, Bethesda, MD 20892, United States
| | - Jung-Hyun Park
- Experimental Immunology Branch, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, MD 20892, United States.
| |
Collapse
|
18
|
Rai V, Mendoza-Mari Y, Radwan MM, Brazdzionis J, Connett DA, Miulli DE, Agrawal DK. Transcriptional and Translational Regulation of Differentially Expressed Genes in Yucatan Miniswine Brain Tissues following Traumatic Brain Injury. JOURNAL OF BIOINFORMATICS AND SYSTEMS BIOLOGY : OPEN ACCESS 2024; 7:81-91. [PMID: 38818113 PMCID: PMC11138201 DOI: 10.26502/jbsb.5107080] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/01/2024]
Abstract
Traumatic brain injury (TBI) is a leading cause of morbidity, disability, and mortality worldwide. Motor and cognitive deficits and emotional disturbances are long-term consequences of TBI. A lack of effective treatment for TBI-induced neural damage, functional impairments, and cognitive deficits makes it challenging in the recovery following TBI. One of the reasons may be the lack of knowledge underlying the complex pathophysiology of TBI and the regulatory factors involved in the cellular and molecular mechanisms of inflammation, neural regeneration, and injury repair. These mechanisms involve a change in the expression of various proteins encoded by genes whose expression is regulated by transcription factors (TFs) at the transcriptional level and microRNA (miRs) at the mRNA level. In this pilot study, we performed the RNA sequencing of injured tissues and non-injured tissues from the brain of Yucatan miniswine and analyzed the sequencing data for differentially expressed genes (DEGs) and the TFs and miRs regulating the expression of DEGs using in-silico analysis. We also compared the effect of the electromagnetic field (EMF) applied to the injured miniswine on the expression profile of various DEGs. The results of this pilot study revealed a few DEGs that were significantly upregulated in the injured brain tissue and the EMF stimulation showed effect on their expression profile.
Collapse
Affiliation(s)
- Vikrant Rai
- Department of Translational Research, College of Osteopathic Medicine of the Pacific, Western University of Health Sciences, Pomona CA 91766, USA
| | - Yssel Mendoza-Mari
- Department of Translational Research, College of Osteopathic Medicine of the Pacific, Western University of Health Sciences, Pomona CA 91766, USA
| | - Mohamed M Radwan
- Department of Translational Research, College of Osteopathic Medicine of the Pacific, Western University of Health Sciences, Pomona CA 91766, USA
| | - James Brazdzionis
- Department of Translational Research, College of Osteopathic Medicine of the Pacific, Western University of Health Sciences, Pomona CA 91766, USA
| | - David A Connett
- Department of Translational Research, College of Osteopathic Medicine of the Pacific, Western University of Health Sciences, Pomona CA 91766, USA
| | - Dan E Miulli
- Department of Translational Research, College of Osteopathic Medicine of the Pacific, Western University of Health Sciences, Pomona CA 91766, USA
| | - Devendra K Agrawal
- Department of Translational Research, College of Osteopathic Medicine of the Pacific, Western University of Health Sciences, Pomona CA 91766, USA
| |
Collapse
|
19
|
Naeem A, Noureen N, Al-Naemi SK, Al-Emadi JA, Khan MJ. Computational design of anti-cancer peptides tailored to target specific tumor markers. BMC Chem 2024; 18:39. [PMID: 38388460 PMCID: PMC10882887 DOI: 10.1186/s13065-024-01143-0] [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: 11/28/2023] [Accepted: 02/13/2024] [Indexed: 02/24/2024] Open
Abstract
Anti-cancer peptides (ACPs) are short peptides known for their ability to inhibit tumor cell proliferation, migration, and the formation of tumor blood vessels. In this study, we designed ACPs to target receptors often overexpressed in cancer using a systematic in silico approach. Three target receptors (CXCR1, DcR3, and OPG) were selected for their significant roles in cancer pathogenesis and tumor cell proliferation. Our peptide design strategy involved identifying interacting residues (IR) of these receptors, with their natural ligands serving as a reference for designing peptides specific to each receptor. The natural ligands of these receptors, including IL8 for CXCR1, TL1A for DcR3, and RANKL for OPG, were identified from the literature. Using the identified interacting residues (IR), we generated a peptide library through simple permutation and predicted the structure of each peptide. All peptides were analyzed using the web-based prediction server for Anticancer peptides, AntiCP. Docking simulations were then conducted to analyze the binding efficiencies of peptides with their respective target receptors, using VEGA ZZ and Chimera for interaction analysis. Our analysis identified HPKFIKELR as the interacting residues (IR) of CXCR-IL8. For DcR3, we utilized three domains from TL1A (TDSYPEP, TKEDKTF, LGLAFTK) as templates, along with two regions (SIKIPSS and PDQDATYP) from RANKL, to generate a library of peptide analogs. Subsequently, peptides for each receptor were shortlisted based on their predicted anticancer properties as determined by AntiCP and were subjected to docking analysis. After docking, peptides that exhibited the least binding energy were further analyzed for their detailed interaction with their respective receptors. Among these, peptides C9 (HPKFELY) and C7 (HPKFEWL) for CXCR1, peptides D6 (ADSYPQP) and D18 (AFSYPFP) for DcR3, and peptides P19 (PDTYPQDP) and p16 (PDQDATYP) for OPG, demonstrated the highest affinity and stronger interactions compared to the other peptides. Although in silico predictions indicated a favorable binding affinity of the designed peptides with target receptors, further experimental validation is essential to confirm their binding affinity, stability and pharmacokinetic characteristics.
Collapse
Affiliation(s)
- Aisha Naeem
- QU Health, Qatar University, P.O. Box 2713, Doha, Qatar.
| | - Nighat Noureen
- Cancer Center and Department of Pediatrics, School of Medicine, Texas Tech University Health Sciences Center School of Medicine, Lubbock, TX, 79430, USA.
| | | | | | - Muhammad Jawad Khan
- Department of Biosciences, COMSATS University Islamabad, Islamabad, 45550, Pakistan
| |
Collapse
|
20
|
Niu W, Liu Q, Huo X, Luo Y, Zhang X. TL1A promotes metastasis and EMT process of colorectal cancer. Heliyon 2024; 10:e24392. [PMID: 38312710 PMCID: PMC10835226 DOI: 10.1016/j.heliyon.2024.e24392] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2023] [Revised: 12/15/2023] [Accepted: 01/08/2024] [Indexed: 02/06/2024] Open
Abstract
BACKGROUND Metastasis is the major problem of colorectal cancer (CRC) and is correlated with the high mortality. Tumor necrosis factor-like cytokine 1A (TL1A) is a novel regulatory factor for inflammatory diseases. This work aimed to investigate the role of TL1A in CRC metastasis. METHOD AOM/DSS-induced mouse model, xenograft tumor model and metastasis murine model were established to mimic the colitis-associated CRC and investigate CRC growth and metastasis in vivo. Colon tissues were assessed by hematoxylin/eosin (HE) staining and immunohistochemistry (IHC). CRC cell metastasis in vivo was observed using in vivo imaging system (IVIS). Cell viability and proliferation were examined using cell counting kit 8 (CCK-8) and EdU experiments. The expression of tumor growth factor β (TGFβ) and metastatic biomarkers were detected using western blotting experiment. The in vitro cell metastasis was measured by Transwell. RESULTS Knockdown of TL1A notably suppressed the generation of colonic tumors in azoxymethane/dextran sodium sulfate (AOM/DSS) model, suppressed in vivo CRC cell growth, as well as lung and liver metastasis. The inflammation response and inflammatory cell infiltration in tumor sites were decreased by TL1A depletion. The in vitro CRC cell growth and metastasis was also suppressed by shTL1A, along with altered expression of epithelial mesenchymal transition (EMT) biomarkers. TL1A depletion suppressed the level of the TGF-β1 receptor (TβRI) and phosphorylation of Smad3 in CRC cells. Stimulation with TGF-β recovered the CRC cell migration and invasion that suppressed by shTL1A. CONCLUSION Our work implicated TL1A as a promoter of CRC generation and metastasis and defines TGF-β/Smad3 signaling as mediator of TL1A-regualated CRC cell metastasis.
Collapse
Affiliation(s)
- Weiwei Niu
- The Second Hospital of Hebei Medical University, Heping West Road No. 215, Shijiazhuang City, Hebei province, 050000, China
| | - Qian Liu
- The Second Hospital of Hebei Medical University, Heping West Road No. 215, Shijiazhuang City, Hebei province, 050000, China
| | - Xiaoxia Huo
- The Second Hospital of Hebei Medical University, Heping West Road No. 215, Shijiazhuang City, Hebei province, 050000, China
| | - Yuxin Luo
- The Second Hospital of Hebei Medical University, Heping West Road No. 215, Shijiazhuang City, Hebei province, 050000, China
| | - Xiaolan Zhang
- The Second Hospital of Hebei Medical University, Heping West Road No. 215, Shijiazhuang City, Hebei province, 050000, China
| |
Collapse
|
21
|
Wang S, Kozai M, Hiraishi M, Rubel MZU, Ichii O, Inaba M, Matsuo K, Takada K. Roles of tumor necrosis factor-like ligand 1A in γδT-cell activation and psoriasis pathogenesis. Front Immunol 2024; 15:1340467. [PMID: 38348035 PMCID: PMC10859483 DOI: 10.3389/fimmu.2024.1340467] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2023] [Accepted: 01/15/2024] [Indexed: 02/15/2024] Open
Abstract
Background Interleukin (IL)-17-producing γδT (γδT17) cells mediate inflammatory responses in barrier tissues. Dysregulated γδT17 cell activation can lead to the overproduction of IL-17 and IL-22 and the development of inflammatory diseases, including psoriasis. IL-23 and IL-1β are known to synergistically activate γδT17 cells, but the regulatory mechanisms of γδT17 cells have not been fully elucidated. This study aimed to reveal the contribution of the inflammatory cytokine tumor necrosis factor-like ligand 1A (TL1A) to γδT17 cell activation and psoriasis development. Methods Anti-TL1A antibody was injected into an imiquimod (IMQ)-induced murine psoriasis model. TL1A receptor expression was analyzed in splenic and dermal γδT cells. γδT cells were tested for cytokine production in vitro and in vivo under stimulation with IL-23, IL-1β, and TL1A. TL1A was applied to a psoriasis model induced by intradermal IL-23 injection. Mice deficient in γδT cells were intradermally injected with IL-23 plus TL1A to verify the contribution of TL1A-dependent γδT-cell activation to psoriasis development. Results Neutralization of TL1A attenuated γδT17 cell activation in IMQ-treated skin. TL1A induced cytokine production by splenic γδT17 cells in synergy with IL-23. Dermal γδT17 cells constitutively expressed a TL1A receptor at high levels and vigorously produced IL-22 upon intradermal IL-23 and TL1A injection but not IL-23 alone. TL1A exacerbated the dermal symptoms induced by IL-23 injection in wild-type but not in γδT cell-deficient mice. Conclusion These findings suggest a novel regulatory mechanism of γδT cells through TL1A and its involvement in psoriasis pathogenesis as a possible therapeutic target.
Collapse
Affiliation(s)
- Shangyi Wang
- Laboratory of Molecular Medicine, Faculty of Veterinary Medicine, Hokkaido University, Sapporo, Japan
| | - Mina Kozai
- Division of Vaccinology for Clinical Development, Institute for Vaccine Research and Development (IVReD), Hokkaido University, Sapporo, Japan
| | - Masaya Hiraishi
- Laboratory of Anatomy, Faculty of Veterinary Medicine, Hokkaido University, Sapporo, Japan
| | - Md. Zahir Uddin Rubel
- Laboratory of Anatomy, Faculty of Veterinary Medicine, Hokkaido University, Sapporo, Japan
| | - Osamu Ichii
- Laboratory of Anatomy, Faculty of Veterinary Medicine, Hokkaido University, Sapporo, Japan
- Laboratory of Agrobiomedical Science, Faculty of Agriculture, Hokkaido University, Sapporo, Japan
| | - Mutsumi Inaba
- Laboratory of Molecular Medicine, Faculty of Veterinary Medicine, Hokkaido University, Sapporo, Japan
| | - Kazuhiro Matsuo
- Division of Vaccinology for Clinical Development, Institute for Vaccine Research and Development (IVReD), Hokkaido University, Sapporo, Japan
| | - Kensuke Takada
- Division of Vaccinology for Clinical Development, Institute for Vaccine Research and Development (IVReD), Hokkaido University, Sapporo, Japan
| |
Collapse
|
22
|
Li S, Cai Y, Guan T, Zhang Y, Huang K, Zhang Z, Cao W, Guan X. Quinic acid alleviates high-fat diet-induced neuroinflammation by inhibiting DR3/IKK/NF-κB signaling via gut microbial tryptophan metabolites. Gut Microbes 2024; 16:2374608. [PMID: 38972055 PMCID: PMC11229714 DOI: 10.1080/19490976.2024.2374608] [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: 02/04/2024] [Accepted: 06/26/2024] [Indexed: 07/09/2024] Open
Abstract
With the increasing of aging population and the consumption of high-fat diets (HFD), the incidence of Alzheimer's disease (AD) has skyrocketed. Natural antioxidants show promising potential in the prevention of AD, as oxidative stress and neuroinflammation are two hallmarks of AD pathogenesis. Here, we showed that quinic acid (QA), a polyphenol derived from millet, significantly decreased HFD-induced brain oxidative stress and neuroinflammation and the levels of Aβ and p-Tau. Examination of gut microbiota suggested the improvement of the composition of gut microbiota in HFD mice after QA treatment. Metabolomic analysis showed significant increase of gut microbial tryptophan metabolites indole-3-acetic acid (IAA) and kynurenic acid (KYNA) by QA. In addition, IAA and KYNA showed negative correlation with pro-inflammatory factors and AD indicators. Further experiments on HFD mice proved that IAA and KYNA could reproduce the effects of QA that suppress brain oxidative stress and inflammation and decrease the levels of of Aβ and p-Tau. Transcriptomics analysis of brain after IAA administration revealed the inhibition of DR3/IKK/NF-κB signaling pathway by IAA. In conclusion, this study demonstrated that QA could counteract HFD-induced brain oxidative stress and neuroinflammation by regulating inflammatory DR3/IKK/NF-κB signaling pathway via gut microbial tryptophan metabolites.
Collapse
Affiliation(s)
- Sen Li
- School of Health Science and Engineering, University of Shanghai for Science and Technology, Shanghai, China
- National Grain Industry (Urban Grain and Oil Security) Technology Innovation Center, University of Shanghai for Science and Technology, Shanghai, China
| | - Yuwei Cai
- School of Health Science and Engineering, University of Shanghai for Science and Technology, Shanghai, China
- National Grain Industry (Urban Grain and Oil Security) Technology Innovation Center, University of Shanghai for Science and Technology, Shanghai, China
| | - Tong Guan
- School of Health Science and Engineering, University of Shanghai for Science and Technology, Shanghai, China
- National Grain Industry (Urban Grain and Oil Security) Technology Innovation Center, University of Shanghai for Science and Technology, Shanghai, China
| | - Yu Zhang
- School of Health Science and Engineering, University of Shanghai for Science and Technology, Shanghai, China
- National Grain Industry (Urban Grain and Oil Security) Technology Innovation Center, University of Shanghai for Science and Technology, Shanghai, China
| | - Kai Huang
- School of Health Science and Engineering, University of Shanghai for Science and Technology, Shanghai, China
- National Grain Industry (Urban Grain and Oil Security) Technology Innovation Center, University of Shanghai for Science and Technology, Shanghai, China
| | - Ze Zhang
- School of Health Science and Engineering, University of Shanghai for Science and Technology, Shanghai, China
- National Grain Industry (Urban Grain and Oil Security) Technology Innovation Center, University of Shanghai for Science and Technology, Shanghai, China
| | - Wangqing Cao
- School of Health Science and Engineering, University of Shanghai for Science and Technology, Shanghai, China
- National Grain Industry (Urban Grain and Oil Security) Technology Innovation Center, University of Shanghai for Science and Technology, Shanghai, China
| | - Xiao Guan
- School of Health Science and Engineering, University of Shanghai for Science and Technology, Shanghai, China
- National Grain Industry (Urban Grain and Oil Security) Technology Innovation Center, University of Shanghai for Science and Technology, Shanghai, China
| |
Collapse
|
23
|
Zhou QM, Zheng L. Research progress on the relationship between Paneth cells-susceptibility genes, intestinal microecology and inflammatory bowel disease. World J Clin Cases 2023; 11:8111-8125. [PMID: 38130785 PMCID: PMC10731169 DOI: 10.12998/wjcc.v11.i34.8111] [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: 07/27/2023] [Revised: 09/26/2023] [Accepted: 11/27/2023] [Indexed: 12/06/2023] Open
Abstract
Inflammatory bowel disease (IBD) is a disorder of the immune system and intestinal microecosystem caused by environmental factors in genetically susceptible people. Paneth cells (PCs) play a central role in IBD pathogenesis, especially in Crohn's disease development, and their morphology, number and function are regulated by susceptibility genes. In the intestine, PCs participate in the formation of the stem cell microenvironment by secreting antibacterial particles and play a role in helping maintain the intestinal microecology and intestinal mucosal homeostasis. Moreover, PC proliferation and maturation depend on symbiotic flora in the intestine. This paper describes the interactions among susceptibility genes, PCs and intestinal microecology and their effects on IBD occurrence and development.
Collapse
Affiliation(s)
- Qi-Ming Zhou
- Department of Nephrology, Lanxi Hospital of Traditional Chinese Medicine, Lanxi 321100, Zhejiang Province, China
| | - Lie Zheng
- Department of Gastroenterology, Shaanxi Hospital of Traditional Chinese Medicine, Xi’an 710003, Shaanxi Province, China
| |
Collapse
|
24
|
Zou C, Zan X, Jia Z, Zheng L, Gu Y, Liu F, Han Y, Xu C, Wu A, Zhi Q. Crosstalk between alternative splicing and inflammatory bowel disease: Basic mechanisms, biotechnological progresses and future perspectives. Clin Transl Med 2023; 13:e1479. [PMID: 37983927 PMCID: PMC10659771 DOI: 10.1002/ctm2.1479] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2023] [Revised: 10/07/2023] [Accepted: 11/03/2023] [Indexed: 11/22/2023] Open
Abstract
BACKGROUND Alternative splicing (AS) is an omnipresent regulatory mechanism of gene expression that enables the generation of diverse splice isoforms from a single gene. Recently, AS events have gained considerable momentum in the pathogenesis of inflammatory bowel disease (IBD). METHODS Our review has summarized the complex process of RNA splicing, and firstly highlighted the potential involved molecules that target aberrant splicing events in IBD. The quantitative transcriptome analyses such as microarrays, next-generation sequencing (NGS) for AS events in IBD have been also discussed. RESULTS Available evidence suggests that some abnormal splicing RNAs can lead to multiple intestinal disorders during the onset of IBD as well as the progression to colitis-associated cancer (CAC), including gut microbiota perturbations, intestinal barrier dysfunctions, innate/adaptive immune dysregulations, pro-fibrosis activation and some other risk factors. Moreover, current data show that the advanced technologies, including microarrays and NGS, have been pioneeringly employed to screen the AS candidates and elucidate the potential regulatory mechanisms of IBD. Besides, other biotechnological progresses such as the applications of third-generation sequencing (TGS), single-cell RNA sequencing (scRNA-seq) and spatial transcriptomics (ST), will be desired with great expectations. CONCLUSIONS To our knowledge, the current review is the first one to evaluate the potential regulatory mechanisms of AS events in IBD. The expanding list of aberrantly spliced genes in IBD along with the developed technologies provide us new clues to how IBD develops, and how these important AS events can be explored for future treatment.
Collapse
Affiliation(s)
- Chentao Zou
- Department of GastroenterologyThe First Affiliated Hospital of Soochow UniversitySuzhouChina
| | - Xinquan Zan
- Department of General SurgeryThe First Affiliated Hospital of Soochow UniversitySuzhouChina
| | - Zhenyu Jia
- Department of GastroenterologyThe First Affiliated Hospital of Soochow UniversitySuzhouChina
| | - Lu Zheng
- Department of GastroenterologyThe First Affiliated Hospital of Soochow UniversitySuzhouChina
| | - Yijie Gu
- Department of GastroenterologyThe First Affiliated Hospital of Soochow UniversitySuzhouChina
| | - Fei Liu
- Department of GastroenterologyThe First Affiliated Hospital of Soochow UniversitySuzhouChina
| | - Ye Han
- Department of General SurgeryThe First Affiliated Hospital of Soochow UniversitySuzhouChina
| | - Chunfang Xu
- Department of GastroenterologyThe First Affiliated Hospital of Soochow UniversitySuzhouChina
| | - Airong Wu
- Department of GastroenterologyThe First Affiliated Hospital of Soochow UniversitySuzhouChina
| | - Qiaoming Zhi
- Department of General SurgeryThe First Affiliated Hospital of Soochow UniversitySuzhouChina
| |
Collapse
|
25
|
Lee BT, Baker LA, Momen M, Terhaar H, Binversie EE, Sample SJ, Muir P. Identification of genetic variants associated with anterior cruciate ligament rupture and AKC standard coat color in the Labrador Retriever. BMC Genom Data 2023; 24:60. [PMID: 37884875 PMCID: PMC10605342 DOI: 10.1186/s12863-023-01164-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: 09/28/2022] [Accepted: 10/09/2023] [Indexed: 10/28/2023] Open
Abstract
Canine anterior cruciate ligament (ACL) rupture is a common complex disease. Prevalence of ACL rupture is breed dependent. In an epidemiological study, yellow coat color was associated with increased risk of ACL rupture in the Labrador Retriever. ACL rupture risk variants may be linked to coat color through genetic selection or through linkage with coat color genes. To investigate these associations, Labrador Retrievers were phenotyped as ACL rupture case or controls and for coat color and were single nucleotide polymorphism (SNP) genotyped. After filtering, ~ 697 K SNPs were analyzed using GEMMA and mvBIMBAM for multivariate association. Functional annotation clustering analysis with DAVID was performed on candidate genes. A large 8 Mb region on chromosome 5 that included ACSF3, as well as 32 additional SNPs, met genome-wide significance at P < 6.07E-7 or Log10(BF) = 3.0 for GEMMA and mvBIMBAM, respectively. On chromosome 23, SNPs were located within or near PCCB and MSL2. On chromosome 30, a SNP was located within IGDCC3. SNPs associated with coat color were also located within ADAM9, FAM109B, SULT1C4, RTDR1, BCR, and RGS7. DZIP1L was associated with ACL rupture. Several significant SNPs on chromosomes 2, 3, 7, 24, and 26 were located within uncharacterized regions or long non-coding RNA sequences. This study validates associations with the previous ACL rupture candidate genes ACSF3 and DZIP1L and identifies novel candidate genes. These variants could act as targets for treatment or as factors in disease prediction modeling. The study highlighted the importance of regulatory SNPs in the disease, as several significant SNPs were located within non-coding regions.
Collapse
Affiliation(s)
- B T Lee
- Department of Surgical Sciences, University of Wisconsin-Madison, School of Veterinary Medicine, 2015 Linden Drive, Madison, WI, 53706, United States of America
| | - L A Baker
- Department of Surgical Sciences, University of Wisconsin-Madison, School of Veterinary Medicine, 2015 Linden Drive, Madison, WI, 53706, United States of America
| | - M Momen
- Department of Surgical Sciences, University of Wisconsin-Madison, School of Veterinary Medicine, 2015 Linden Drive, Madison, WI, 53706, United States of America
| | - H Terhaar
- Department of Surgical Sciences, University of Wisconsin-Madison, School of Veterinary Medicine, 2015 Linden Drive, Madison, WI, 53706, United States of America
| | - E E Binversie
- Department of Surgical Sciences, University of Wisconsin-Madison, School of Veterinary Medicine, 2015 Linden Drive, Madison, WI, 53706, United States of America
| | - S J Sample
- Department of Surgical Sciences, University of Wisconsin-Madison, School of Veterinary Medicine, 2015 Linden Drive, Madison, WI, 53706, United States of America
| | - Peter Muir
- Department of Surgical Sciences, University of Wisconsin-Madison, School of Veterinary Medicine, 2015 Linden Drive, Madison, WI, 53706, United States of America.
| |
Collapse
|
26
|
Eigenschink M, Wessely I, Dijmarescu M, Förster-Waldl E, Farr A, Kiss H, Berger A, Wisgrill L. Transcriptomic analysis identifies lactoferrin-induced quiescent circuits in neonatal macrophages. Front Immunol 2023; 14:1276173. [PMID: 37868991 PMCID: PMC10590118 DOI: 10.3389/fimmu.2023.1276173] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2023] [Accepted: 09/18/2023] [Indexed: 10/24/2023] Open
Abstract
Introduction Upon birth, a hitherto naïve immune system is confronted with a plethora of microbial antigens due to intestinal bacterial colonization. To prevent excessive inflammation and disruption of the epithelial barrier, physiological mechanisms must promote immune-anergy within the neonatal gut. As high concentrations of human lactoferrin (hLF), a transferrin glycoprotein shown to modulate macrophage function, are frequently encountered in colostrum, its direct interaction with intestinal macrophages may satisfy this physiological need. Thus, the primary objective of this study was to investigate transcriptional changes induced by human lactoferrin in neonatal monocyte-derived macrophages. Methods Cord blood-derived monocytes were differentiated with M-CSF in presence or absence of 500 µg/mL hLF for 7 days and afterwards stimulated with 1 ng/mL LPS or left untreated. RNA was then isolated and subjected to microarray analysis. Results Differentiation of cord blood-derived monocytes in presence of hLF induced a distinct transcriptional program defined by cell cycle arrest in the G2/M phase, induction of IL-4/IL-13-like signaling, altered extracellular matrix interaction, and enhanced propensity for cell-cell interaction. Moreover, near-complete abrogation of transcriptional changes induced by TLR4 engagement with LPS was observed in hLF-treated samples. Discussion The global transition towards an M2-like homeostatic phenotype and the acquisition of quiescence elegantly demonstrate the ontogenetical relevance of hLF in attenuating pro-inflammatory signaling within the developing neonatal intestine. The marked anergy towards proinflammatory stimuli such as LPS further underlines the glycoprotein's potential therapeutic relevance.
Collapse
Affiliation(s)
- Michael Eigenschink
- Division of Neonatology, Pediatric Intensive Care and Neuropaediatrics, Department of Pediatrics and Adolescent Medicine, Comprehensive Center for Pediatrics, Medical University of Vienna, Vienna, Austria
| | - Isabelle Wessely
- Division of Neonatology, Pediatric Intensive Care and Neuropaediatrics, Department of Pediatrics and Adolescent Medicine, Comprehensive Center for Pediatrics, Medical University of Vienna, Vienna, Austria
| | - Marco Dijmarescu
- Division of Neonatology, Pediatric Intensive Care and Neuropaediatrics, Department of Pediatrics and Adolescent Medicine, Comprehensive Center for Pediatrics, Medical University of Vienna, Vienna, Austria
| | - Elisabeth Förster-Waldl
- Division of Neonatology, Pediatric Intensive Care and Neuropaediatrics, Department of Pediatrics and Adolescent Medicine, Comprehensive Center for Pediatrics, Medical University of Vienna, Vienna, Austria
| | - Alex Farr
- Department of Obstetrics and Gynecology, Division of Obstetrics and Feto-Maternal Medicine, Comprehensive Center for Pediatrics, Medical University of Vienna, Vienna, Austria
| | - Herbert Kiss
- Department of Obstetrics and Gynecology, Division of Obstetrics and Feto-Maternal Medicine, Comprehensive Center for Pediatrics, Medical University of Vienna, Vienna, Austria
| | - Angelika Berger
- Division of Neonatology, Pediatric Intensive Care and Neuropaediatrics, Department of Pediatrics and Adolescent Medicine, Comprehensive Center for Pediatrics, Medical University of Vienna, Vienna, Austria
| | - Lukas Wisgrill
- Division of Neonatology, Pediatric Intensive Care and Neuropaediatrics, Department of Pediatrics and Adolescent Medicine, Comprehensive Center for Pediatrics, Medical University of Vienna, Vienna, Austria
| |
Collapse
|
27
|
Cason RK, Chambers E, Tu T, Chryst-Stangl M, Huggins K, Lane BM, Ochoa A, Jackson AM, Gbadegesin RA. Genetic risk variants for childhood nephrotic syndrome and corticosteroid response. Front Pediatr 2023; 11:1248733. [PMID: 37868272 PMCID: PMC10588181 DOI: 10.3389/fped.2023.1248733] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/27/2023] [Accepted: 09/22/2023] [Indexed: 10/24/2023] Open
Abstract
Introduction The etiology of most cases of nephrotic syndrome (NS) remains unknown, therefore patients are phenotypically categorized based on response to corticosteroid therapy as steroid sensitive NS (SSNS), or steroid resistant NS (SRNS). Genetic risk factors have been identified for SSNS from unbiased genome-wide association studies (GWAS), however it is unclear if these loci are disease risk loci in other forms of NS such as SRNS. Additionally, it remains unknown if these risk loci are associated with response to therapy. Thus, we investigated the association between SSNS risk loci and therapy response in a large, multi-race cohort of children along the entire spectrum of childhood-onset NS. Methods We enrolled 1,000 patients with childhood-onset NS comprised of SSNS and SRNS. Genotyping was done using TaqMan and Direct Sanger Sequencing for 9 previously reported childhood SSNS risk loci. We compared the allele frequencies (AF) and variant burden between NS vs. controls and SRNS vs. SSNS. Results All 9 risk loci were associated with NS compared with healthy controls (p = 3.5 × 10-3-<2.2 × 10-16). Variant burden greater than 7 was associated with risk of SRNS (OR 7.4, 95% CI 4.6-12.0, p = 8.2 × 10-16). Conclusion Our study showed that genetic risk loci for childhood SSNS are associated with pattern of therapy response, may help predict disease outcome, and set the stage for individualized treatment of NS.
Collapse
Affiliation(s)
- Rachel K. Cason
- Department of Pediatrics, Division of Nephrology, Duke University Medical Center, Durham, NC, United States
| | - Eileen Chambers
- Department of Pediatrics, Division of Nephrology, Duke University Medical Center, Durham, NC, United States
| | - Tiffany Tu
- Computational Biology and Bioinformatics Program, Duke Center for Statistical Genetics and Genomics, and Department of Biostatistics and Bioinformatics, Duke University, Durham, NC, United States
| | - Megan Chryst-Stangl
- Department of Pediatrics, Division of Nephrology, Duke University Medical Center, Durham, NC, United States
| | - Kinsie Huggins
- Department of Pediatrics, Division of Nephrology, Duke University Medical Center, Durham, NC, United States
| | - Brandon M. Lane
- Department of Pediatrics, Division of Nephrology, Duke University Medical Center, Durham, NC, United States
| | - Alejandro Ochoa
- Computational Biology and Bioinformatics Program, Duke Center for Statistical Genetics and Genomics, and Department of Biostatistics and Bioinformatics, Duke University, Durham, NC, United States
| | - Annette M. Jackson
- Department of Surgery, Duke University Medical Center, Durham, NC, United States
| | - Rasheed A. Gbadegesin
- Department of Pediatrics, Division of Nephrology, Duke University Medical Center, Durham, NC, United States
| |
Collapse
|
28
|
Dean EC, Ditoro DF, Pham D, Gao M, Zindl CL, Frey B, Harbour SN, Figge DA, Miller AT, Glassman CR, Garcia KC, Hatton RD, Weaver CT. IL-2-induced Stat3 Signaling is Critical for Effector Treg Cell Programming. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.09.26.559434. [PMID: 37808649 PMCID: PMC10557704 DOI: 10.1101/2023.09.26.559434] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/10/2023]
Abstract
Maintenance of immune homeostasis to the intestinal mictrobiota is dependent on a population of effector regulatory T (eTreg) cells that develop from microbiota-reactive induced (i)Treg cells. A cardinal feature of eTreg cells is their production of IL-10, which plays a non-redundant role in immune tolerance of commensal microbes. Here, we identify an unexpected role for IL-2-induced Stat3 signaling to program iTreg cells for eTreg cell differentiation and Il10 transcriptional competency. IL-2 proved to be both necessary and sufficient for eTreg cell development - contingent on Stat3 output of the IL-2 receptor coordinate with IL-2 signaling during early Treg cell commitment. Induction of iTreg cell programming in absence of IL-2-induced Stat3 signaling resulted in impaired eTreg cell differentiation and a failure to produce IL-10. An IL-2 mutein with reduced affinity for the IL-2Rγ (γ c ) chain was found to have blunted IL-2R Stat3 output, resulting in a deficiency of Il10 transcriptional programming that could not be fully rescued by Stat3 signaling subsequent to an initial window of iTreg cell differentiation. These findings expose a heretofore unappreciated role of IL-2 signaling that acts early to program subsequent production of IL-10 by developing eTreg cells, with broad implications for IL-2-based therapeutic interventions in immune-mediated diseases.
Collapse
|
29
|
Tang S, Wu Z, Chen L, She L, Zuo W, Luo W, Zhang Y, Liang S, Liu G, He B, He J, Zhang N. Multi-omics analysis reveals the association between elevated KIF18B expression and unfavorable prognosis, immune evasion, and regulatory T cell activation in nasopharyngeal carcinoma. Front Immunol 2023; 14:1258344. [PMID: 37744335 PMCID: PMC10514916 DOI: 10.3389/fimmu.2023.1258344] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2023] [Accepted: 08/25/2023] [Indexed: 09/26/2023] Open
Abstract
Background Nasopharyngeal carcinoma (NPC) is prevalent in Southern China. The expression profile and functions of kinesin family member 18B (KIF18B) remain unclear in NPC. Methods Bulk and single-cell transcriptome data for NPC were downloaded. KIF18B expression differences in NPC and normal tissues and its prognostic value were validated by immunohistochemistry and Cox model. We performed multi-faceted functional enrichment analysis on KIF18B. Immune infiltration was analyzed comprehensively by the CIBERSORT, EPIC, and quanTIseq algorithms and the BisqueRNA package and confirmed by immunofluorescence assay. The intercellular communication were investigated by the CellChat package. We explored the dynamics of KIF18B expression by pseudotime trajectory. M6A modification analysis rely on SRAMP platform. The treatment response were evaluated by Tumor Immune Dysfunction and Exclusion (TIDE) score, immunophenoscore and IC50 value. Results KIF18B overexpression in NPC led to unfavorable prognosis, and significantly associated with advanced T, N, and stage classifications. Functional analysis demonstrated that KIF18B was involved in immune suppression, epithelial-mesenchymal transition (EMT), N6-methyladenosine (m6A) modification and therapeutic responses. The deconvolution algorithm indicated that activated regulatory T cells (Tregs) had the strongest positive correlation with KIF18B among immune cells (R = 0.631). Validated by immunofluorescence assay, the high KIF18B expression group displayed a notable rise in Tregs infiltration, accompanied by a substantial decrease in the infiltration of CD8+ T cells and macrophages. In the intercellular communication network, malignant cells with high KIF18B expression implicated in more interactions, and activated and recruited Tregs by modulating cytokines, chemokines, and immune checkpoints. KIF18B was upregulated in more advanced malignant cells and influenced EMT by regulating ITGA6, VIM, and ZEB1/2. KIF18B expression was positively related to m6A "writer" and "reader" genes, and negatively related to "eraser" genes. The KIF18B high expression group exhibited a higher TIDE score and elevated IC50 values for the commonly used chemotherapy drugs, gemcitabine, oxaliplatin, and 5-fluorouracil. Conclusion KIF18B is a significant prognostic marker in NPC, and may modulate immune evasion and EMT. M6A modification may account for the aberrant overexpression of KIF18B in NPC. Furthermore, KIF18B may predict response to immunotherapy and chemotherapy.
Collapse
Affiliation(s)
- Siqi Tang
- Department of Radiation Oncology, First People’s Hospital of Foshan, Foshan, China
| | - Zhenyu Wu
- Department of Urology, First People’s Hospital of Foshan, Foshan, China
| | - Lusi Chen
- Department of Radiation Oncology, First People’s Hospital of Foshan, Foshan, China
| | - Longjiang She
- Department of Radiation Oncology, First People’s Hospital of Foshan, Foshan, China
| | - Weihan Zuo
- Department of Radiation Oncology, First People’s Hospital of Foshan, Foshan, China
| | - Weijun Luo
- Department of Radiation Oncology, First People’s Hospital of Foshan, Foshan, China
| | - Yang Zhang
- Department of Radiation Oncology, First People’s Hospital of Foshan, Foshan, China
| | - Shaoqiang Liang
- Department of Comprehensive (Head and Neck) Oncology and Hospice Ward, First People’s Hospital of Foshan, Foshan, China
| | - Guichao Liu
- Department of Radiation Oncology, First People’s Hospital of Foshan, Foshan, China
| | - Biyi He
- Department of Rhinology, First People’s Hospital of Foshan, Foshan, China
| | - Jinfeng He
- Department of Rhinology, First People’s Hospital of Foshan, Foshan, China
| | - Ning Zhang
- Department of Radiation Oncology, First People’s Hospital of Foshan, Foshan, China
| |
Collapse
|
30
|
Nishihara K, van Niekerk J, He Z, Innes D, Guan LL, Steele M. Reduction in mucosa thickness is associated with changes in immune function in the colon mucosa during the weaning transition in Holstein bull dairy calves. Genomics 2023; 115:110680. [PMID: 37454938 DOI: 10.1016/j.ygeno.2023.110680] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2023] [Revised: 07/06/2023] [Accepted: 07/12/2023] [Indexed: 07/18/2023]
Abstract
This study aims to characterize changes in the structure and the molecules related to immune function in the colon mucosa in dairy calves during the weaning transition (weaned at week 6 of age). Colon mucosa thickness, measured at week 5 to 8 and 12 of age, decreased for 2 weeks after weaning, but then recovered. Colon mucosa's transcriptome profiling at week 5, 7, and 12 of age was obtained using RNA-sequencing. Functional analysis showed that pathways related to immune function were up-regulated postweaning. A weighted gene co-expression network analysis identified 17 immune function related genes, expressed higher postweaning, which were negatively correlated with colon mucosa thickness, suggesting that these genes may be involved in colon mucosa inflammation and recovery from mucosa thickness decrement during the weaning transition. As such, it is important to determine the function of immune cells in the colon mucosa during the weaning transition in dairy calves.
Collapse
Affiliation(s)
- Koki Nishihara
- Department of Animal Biosciences, Animal Science and Nutrition, University of Guelph, Guelph, ON N1G 1Y2, Canada
| | - Jolet van Niekerk
- Department of Agricultural, Food and Nutritional Science, University of Alberta, Edmonton, AB T6G 2P5, Canada
| | - Zhixiong He
- CAS Key Laboratory for Agro-Ecological Processes in Subtropical Region, Institute of Subtropical Agriculture, The Chinese Academy of Sciences, Changsha 410125, China
| | - David Innes
- Department of Animal Biosciences, Animal Science and Nutrition, University of Guelph, Guelph, ON N1G 1Y2, Canada
| | - Le Luo Guan
- Department of Agricultural, Food and Nutritional Science, University of Alberta, Edmonton, AB T6G 2P5, Canada
| | - Michael Steele
- Department of Animal Biosciences, Animal Science and Nutrition, University of Guelph, Guelph, ON N1G 1Y2, Canada.
| |
Collapse
|
31
|
Ryu S, Lim M, Kim J, Kim HY. Versatile roles of innate lymphoid cells at the mucosal barrier: from homeostasis to pathological inflammation. Exp Mol Med 2023; 55:1845-1857. [PMID: 37696896 PMCID: PMC10545731 DOI: 10.1038/s12276-023-01022-z] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2023] [Revised: 03/31/2023] [Accepted: 04/04/2023] [Indexed: 09/13/2023] Open
Abstract
Innate lymphoid cells (ILCs) are innate lymphocytes that do not express antigen-specific receptors and largely reside and self-renew in mucosal tissues. ILCs can be categorized into three groups (ILC1-3) based on the transcription factors that direct their functions and the cytokines they produce. Their signature transcription factors and cytokines closely mirror those of their Th1, Th2, and Th17 cell counterparts. Accumulating studies show that ILCs are involved in not only the pathogenesis of mucosal tissue diseases, especially respiratory diseases, and colitis, but also the resolution of such diseases. Here, we discuss recent advances regarding our understanding of the biology of ILCs in mucosal tissue health and disease. In addition, we describe the current research on the immune checkpoints by which other cells regulate ILC activities: for example, checkpoint molecules are potential new targets for therapies that aim to control ILCs in mucosal diseases. In addition, we review approved and clinically- trialed drugs and drugs in clinical trials that can target ILCs and therefore have therapeutic potential in ILC-mediated diseases. Finally, since ILCs also play important roles in mucosal tissue homeostasis, we explore the hitherto sparse research on cell therapy with regulatory ILCs. This review highlights various therapeutic approaches that could be used to treat ILC-mediated mucosal diseases and areas of research that could benefit from further investigation.
Collapse
Affiliation(s)
- Seungwon Ryu
- Department of Microbiology, Gachon University College of Medicine, Incheon, 21999, South Korea
| | - MinYeong Lim
- Laboratory of Mucosal Immunology, Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul, 03080, South Korea
- Institute of Allergy and Clinical Immunology, Seoul National University Medical Research Center, Seoul, South Korea
- CIRNO, Sungkyunkwan University, Suwon, South Korea
| | - Jinwoo Kim
- Laboratory of Mucosal Immunology, Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul, 03080, South Korea
- Institute of Allergy and Clinical Immunology, Seoul National University Medical Research Center, Seoul, South Korea
- CIRNO, Sungkyunkwan University, Suwon, South Korea
| | - Hye Young Kim
- Laboratory of Mucosal Immunology, Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul, 03080, South Korea.
- Institute of Allergy and Clinical Immunology, Seoul National University Medical Research Center, Seoul, South Korea.
- CIRNO, Sungkyunkwan University, Suwon, South Korea.
| |
Collapse
|
32
|
Park JS, Gazzaniga FS, Kasper DL, Sharpe AH. Microbiota-dependent regulation of costimulatory and coinhibitory pathways via innate immune sensors and implications for immunotherapy. Exp Mol Med 2023; 55:1913-1921. [PMID: 37696895 PMCID: PMC10545783 DOI: 10.1038/s12276-023-01075-0] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2023] [Revised: 07/11/2023] [Accepted: 07/19/2023] [Indexed: 09/13/2023] Open
Abstract
Our bodies are inhabited by trillions of microorganisms. The host immune system constantly interacts with the microbiota in barrier organs, including the intestines. Over decades, numerous studies have shown that our mucosal immune system is dynamically shaped by a variety of microbiota-derived signals. Elucidating the mediators of these interactions is an important step for understanding how the microbiota is linked to mucosal immune homeostasis and gut-associated diseases. Interestingly, the efficacy of cancer immunotherapies that manipulate costimulatory and coinhibitory pathways has been correlated with the gut microbiota. Moreover, adverse effects of these therapies in the gut are linked to dysregulation of the intestinal immune system. These findings suggest that costimulatory pathways in the immune system might serve as a bridge between the host immune system and the gut microbiota. Here, we review mechanisms by which commensal microorganisms signal immune cells and their potential impact on costimulation. We highlight how costimulatory pathways modulate the mucosal immune system through not only classical antigen-presenting cells but also innate lymphocytes, which are highly enriched in barrier organs. Finally, we discuss the adverse effects of immune checkpoint inhibitors in the gut and the possible relationship with the gut microbiota.
Collapse
Affiliation(s)
- Joon Seok Park
- Department of Immunology, Blavatnik Institute, Harvard Medical School, Boston, MA, 02115, USA
| | - Francesca S Gazzaniga
- Department of Pathology and Center for Cancer Research, Massachusetts General Hospital, Charlestown, MA, 02129, USA
- Department of Pathology, Harvard Medical School, Boston, MA, 02115, USA
| | - Dennis L Kasper
- Department of Immunology, Blavatnik Institute, Harvard Medical School, Boston, MA, 02115, USA
| | - Arlene H Sharpe
- Department of Immunology, Blavatnik Institute, Harvard Medical School, Boston, MA, 02115, USA.
| |
Collapse
|
33
|
Meng F, Jiang X, Wang X, Zheng Q, Wang XN, Mei C, Yan S, He Y, Xue J, Zhang X, Fu W, You Y, Zhai J, Wang Y, Sun X. Tumor necrosis factor-like cytokine 1A plays a role in inflammatory bowel disease pathogenesis. Proc Natl Acad Sci U S A 2023; 120:e2120771120. [PMID: 37579137 PMCID: PMC10452302 DOI: 10.1073/pnas.2120771120] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2021] [Accepted: 07/03/2023] [Indexed: 08/16/2023] Open
Abstract
The binding of tumor necrosis factor-like cytokine 1A (TL1A) to death receptor 3 (DR3) plays an important role in the interaction between dendritic cells (DCs) and T cells and contributes to intestinal inflammation development. However, the mechanism by which DCs expressing TL1A mediate helper T (Th) cell differentiation in the intestinal lamina propria (LP) during the pathogenesis of inflammatory bowel disease remains unclear. In this study, we found that TL1A/DR3 promoted Th1 and Th17 cell differentiation in T-T and DC-T cell interaction-dependent manners. TL1A-deficient CD4+ T cells failed to polarize into Th1/Th17 cells and did not cause colonic inflammation in a T cell transfer colitis model. Notably, TL1A was located in the cytoplasm and nuclei of DCs, positively regulated the DC-specific ICAM-grabbing nonintegrin/RAF1/nuclear factor κB signaling pathway, enhanced the antigen uptake ability of DCs, and promoted TLR4-mediated DC activation, inducing naive CD4+ T cell differentiation into Th1 and Th17 cells. Our work reveals that TL1A plays a regulatory role in inflammatory bowel disease pathogenesis.
Collapse
Affiliation(s)
- Fanxiang Meng
- Department of Immunology, Basic Medicine College, China Medical University, Shenyang, Liaoning Province110122, China
- Department of Laboratory Medicine, The First Hospital of China Medical University, Shenyang, Liaoning Province110801, China
| | - Xuefeng Jiang
- Department of Immunology, Basic Medicine College, China Medical University, Shenyang, Liaoning Province110122, China
| | - Xiao Wang
- Department of Immunology, Basic Medicine College, China Medical University, Shenyang, Liaoning Province110122, China
| | - Qianqian Zheng
- Department of Pathophysiology, Basic Medicine College, China Medical University, Shenyang, Liaoning Province110122, China
| | - Xiaonan N. Wang
- Department of Immunology, Basic Medicine College, China Medical University, Shenyang, Liaoning Province110122, China
| | - Chenxue Mei
- Department of Immunology, Basic Medicine College, China Medical University, Shenyang, Liaoning Province110122, China
- Department of Gastroenterology, Shengjing Hospital of China Medical University, Shenyang, Liaoning Province110122, China
| | - Siqi Yan
- Department of Immunology, Basic Medicine College, China Medical University, Shenyang, Liaoning Province110122, China
| | - Yuting He
- Department of Immunology, Basic Medicine College, China Medical University, Shenyang, Liaoning Province110122, China
| | - Junxiu Xue
- Department of Immunology, Basic Medicine College, China Medical University, Shenyang, Liaoning Province110122, China
| | - Xiaoqing Zhang
- Department of Immunology, Basic Medicine College, China Medical University, Shenyang, Liaoning Province110122, China
- Teaching Center for Medical Experiment, China Medical University, Shenyang, Liaoning Province110122, China
| | - Wenda Fu
- Department of Immunology, Basic Medicine College, China Medical University, Shenyang, Liaoning Province110122, China
- Department of Blood Transfusion, Tangdu Hospital, The Fourth Military Medical University, Xi’an, Shaanxi Province710032, China
| | - Yong You
- Department of Immunology, Basic Medicine College, China Medical University, Shenyang, Liaoning Province110122, China
- Department of Immunology, Chengde medical university, Chengde, Hebei Province067000, China
| | - Jingbo Zhai
- Medical College, Inner Mongolia Minzu University, Tongliao028000, China
- Key Laboratory of Zoonose Prevention and Control at Universities of Inner Mongolia Autonomous Region, Tongliao028000, China
| | - Yuanyuan Wang
- Department of Anesthesiology, The Fourth Affiliated Hospital of China Medical University, Shenyang, Liaoning Province110032, China
| | - Xun Sun
- Department of Immunology, Basic Medicine College, China Medical University, Shenyang, Liaoning Province110122, China
| |
Collapse
|
34
|
Su J, Tong Z, Wu S, Zhou F, Chen Q. Research Progress of DcR3 in the Diagnosis and Treatment of Sepsis. Int J Mol Sci 2023; 24:12916. [PMID: 37629097 PMCID: PMC10454171 DOI: 10.3390/ijms241612916] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2023] [Revised: 08/10/2023] [Accepted: 08/14/2023] [Indexed: 08/27/2023] Open
Abstract
Decoy receptor 3 (DcR3), a soluble glycosylated protein in the tumor necrosis factor receptor superfamily, plays a role in tumor and inflammatory diseases. Sepsis is a life-threatening organ dysfunction caused by the dysregulation of the response to infection. Currently, no specific drug that can alleviate or even cure sepsis in a comprehensive and multi-level manner has been found. DcR3 is closely related to sepsis and considerably upregulated in the serum of those patients, and its upregulation is positively correlated with the severity of sepsis and can be a potential biomarker for diagnosis. DcR3 alone or in combination with other markers has shown promising results in the early diagnosis of sepsis. Furthermore, DcR3 is a multipotent immunomodulator that can bind FasL, LIGHT, and TL1A through decoy action, and block downstream apoptosis and inflammatory signaling. It also regulates T-cell and macrophage differentiation and modulates immune status through non-decoy action; therefore, DcR3 could be a potential drug for the treatment of sepsis. The application of DcR3 in the treatment of a mouse model of sepsis also achieved good efficacy. Here, we introduce and discuss the progress in, and suggest novel ideas for, research regarding DcR3 in the diagnosis and treatment of sepsis.
Collapse
Affiliation(s)
| | | | | | | | - Qi Chen
- Fujian Key Laboratory of Innate Immune Biology, Biomedical Research Center of South China, College of Life Science, Fujian Normal University, Fuzhou 350117, China; (Z.T.); (S.W.); (F.Z.)
| |
Collapse
|
35
|
Kang J, Kim M, Yoon DY, Kim WS, Choi SJ, Kwon YN, Kim WS, Park SH, Sung JJ, Park M, Lee JS, Park JE, Kim SM. AXL +SIGLEC6 + dendritic cells in cerebrospinal fluid and brain tissues of patients with autoimmune inflammatory demyelinating disease of CNS. Clin Immunol 2023; 253:109686. [PMID: 37414380 DOI: 10.1016/j.clim.2023.109686] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2023] [Revised: 05/26/2023] [Accepted: 06/12/2023] [Indexed: 07/08/2023]
Abstract
Inflammatory demyelinating disease of the CNS (IDD) is a heterogeneous group of autoimmune diseases, and multiple sclerosis is the most common type. Dendritic cells (DCs), major antigen-presenting cells, have been proposed to play a central role in the pathogenesis of IDD. The AXL+SIGLEC6+ DC (ASDC) has been only recently identified in humans and has a high capability of T cell activation. Nevertheless, its contribution to CNS autoimmunity remains still obscure. Here, we aimed to identify the ASDC in diverse sample types from IDD patients and experimental autoimmune encephalomyelitis (EAE). A detailed analysis of DC subpopulations using single-cell transcriptomics for the paired cerebrospinal fluid (CSF) and blood samples of IDD patients (total n = 9) revealed that three subtypes of DCs (ASDCs, ACY3+ DCs, and LAMP3+ DCs) were overrepresented in CSF compared with their paired blood. Among these DCs, ASDCs were also more abundant in CSF of IDD patients than in controls, manifesting poly-adhesional and stimulatory characteristics. In the brain biopsied tissues of IDD patients, obtained at the acute attack of disease, ASDC were also frequently found in close contact with T cells. Lastly, the frequency of ASDC was found to be temporally more abundant in acute attack of disease both in CSF samples of IDD patients and in tissues of EAE, an animal model for CNS autoimmunity. Our analysis suggests that the ASDC might be involved in the pathogenesis of CNS autoimmunity.
Collapse
Affiliation(s)
- Junho Kang
- Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology, Daejeon, Republic of Korea
| | - Moonhang Kim
- Biomedical Research Institute, Seoul National University Hospital, Seoul, Republic of Korea
| | - Da-Young Yoon
- Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Woo-Seok Kim
- Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology, Daejeon, Republic of Korea
| | - Seok-Jin Choi
- Department of Neurology, Seoul National University Hospital, Seoul National University of Medicine, Seoul, Republic of Korea
| | - Young-Nam Kwon
- Department of Neurology, Seoul National University Hospital, Seoul National University of Medicine, Seoul, Republic of Korea
| | - Won-Seok Kim
- Department of Neurology, Seoul National University Hospital, Seoul National University of Medicine, Seoul, Republic of Korea
| | - Sung-Hye Park
- Department of Pathology, Seoul National University Hospital, Seoul National University of Medicine, Seoul, Republic of Korea
| | - Jung-Joon Sung
- Department of Neurology, Seoul National University Hospital, Seoul National University of Medicine, Seoul, Republic of Korea
| | - Myungsun Park
- Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology, Daejeon, Republic of Korea
| | - Jung Seok Lee
- Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology, Daejeon, Republic of Korea
| | - Jong-Eun Park
- Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology, Daejeon, Republic of Korea.
| | - Sung-Min Kim
- Department of Neurology, Seoul National University Hospital, Seoul National University of Medicine, Seoul, Republic of Korea.
| |
Collapse
|
36
|
Zhang D, Zhang J, Zhang J, Ji X, Qi Q, Xu J, Pan Y, Liu X, Sun F, Zeng R, Dong L. Identification of a novel role for TL1A/DR3 deficiency in acute respiratory distress syndrome that exacerbates alveolar epithelial disruption. Respir Res 2023; 24:182. [PMID: 37434162 DOI: 10.1186/s12931-023-02488-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2023] [Accepted: 07/01/2023] [Indexed: 07/13/2023] Open
Abstract
Alveolar epithelial barrier is a potential therapeutic target for acute respiratory distress syndrome (ARDS). However, an effective intervention against alveolar epithelial barrier has not been developed. Here, based on single-cell RNA and mRNA sequencing results, death receptor 3 (DR3) and its only known ligand tumor necrosis factor ligand-associated molecule 1A (TL1A) were significantly reduced in epithelium from an ARDS mice and cell models. The apparent reduction in the TL1A/DR3 axis in lungs from septic-ARDS patients was correlated with the severity of the disease. The examination of knockout (KO) and alveolar epithelium conditional KO (CKO) mice showed that TL1A deficiency exacerbated alveolar inflammation and permeability in lipopolysaccharide (LPS)-induced ARDS. Mechanistically, TL1A deficiency decreased glycocalyx syndecan-1 and tight junction-associated zonula occludens 3 by increasing cathepsin E level for strengthening cell-to-cell permeability. Additionally, DR3 deletion aggravated barrier dysfunction and pulmonary edema in LPS-induced ARDS through the above mechanisms based on the analyses of DR3 CKO mice and DR3 overexpression cells. Therefore, the TL1A/DR3 axis has a potential value as a key therapeutic signaling for the protection of alveolar epithelial barrier.
Collapse
Affiliation(s)
- Dong Zhang
- Department of Respiratory, Shandong Provincial Qianfoshan Hospital, Shandong University, Jinan, 250021, Shandong, China
| | - Jianning Zhang
- Department of Respiratory and Intensive Care Unit, The First Affiliated Hospital of Shandong First Medical University and Shandong Provincial Qianfoshan Hospital, Shandong Institute of Respiratory Diseases, Jinan, China
| | - Jintao Zhang
- Department of Respiratory, Shandong Provincial Qianfoshan Hospital, Shandong University, Jinan, 250021, Shandong, China
| | - Xiang Ji
- Department of Respiratory and Intensive Care Unit, The First Affiliated Hospital of Shandong First Medical University and Shandong Provincial Qianfoshan Hospital, Shandong Institute of Respiratory Diseases, Jinan, China
| | - Qian Qi
- Department of Respiratory and Intensive Care Unit, The First Affiliated Hospital of Shandong First Medical University and Shandong Provincial Qianfoshan Hospital, Shandong Institute of Respiratory Diseases, Jinan, China
| | - Jiawei Xu
- Department of Respiratory and Intensive Care Unit, The First Affiliated Hospital of Shandong First Medical University and Shandong Provincial Qianfoshan Hospital, Shandong Institute of Respiratory Diseases, Jinan, China
| | - Yun Pan
- Department of Respiratory, Shandong Provincial Qianfoshan Hospital, Shandong University, Jinan, 250021, Shandong, China
| | - Xiaofei Liu
- Department of Respiratory and Intensive Care Unit, The First Affiliated Hospital of Shandong First Medical University and Shandong Provincial Qianfoshan Hospital, Shandong Institute of Respiratory Diseases, Jinan, China
| | - Fang Sun
- Department of Respiratory and Intensive Care Unit, The First Affiliated Hospital of Shandong First Medical University and Shandong Provincial Qianfoshan Hospital, Shandong Institute of Respiratory Diseases, Jinan, China
| | - Rong Zeng
- Department of Respiratory, Shandong Provincial Qianfoshan Hospital, Shandong University, Jinan, 250021, Shandong, China
| | - Liang Dong
- Department of Respiratory, Shandong Provincial Qianfoshan Hospital, Shandong University, Jinan, 250021, Shandong, China.
- Department of Respiratory and Intensive Care Unit, The First Affiliated Hospital of Shandong First Medical University and Shandong Provincial Qianfoshan Hospital, Shandong Institute of Respiratory Diseases, Jinan, China.
| |
Collapse
|
37
|
Tian L, Tao S, He C, Dong S, Chen Y, Chen L, Jiang S. Astragaloside IV regulates TL1A and NF-κB signal pathway to affect inflammation in necrotizing enterocolitis. Tissue Cell 2023; 83:102128. [PMID: 37413858 DOI: 10.1016/j.tice.2023.102128] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2023] [Revised: 04/24/2023] [Accepted: 06/06/2023] [Indexed: 07/08/2023]
Abstract
AIM This study aims to explore the possible effect of Astragaloside IV (AS-IV) on necrotizing enterocolitis (NEC) neonatal rat models and verify the possible implication of TNF-like ligand 1 A (TL1A) and NF-κB signal pathway. METHODS NEC neonatal rat models were established through formula feeding, cold/asphyxia stress and Lipopolysaccharide (LPS) gavage method. The appearance, activity and skin as well as the pathological status of rats subjected to NEC modeling were assessed. The intestinal tissues were observed after H&E staining. The expression of oxidative stress biomarkers (SOD, MDA and GSH-Px) and inflammatory cytokines (TNF-α, IL-1β and IL-6) were detected by ELISA and qRT-PCR. Western blotting and immunohistochemistry were applied to detect expressions of TL1A and NF-κB signal pathway-related proteins. Cell apoptosis was assessed by TUNEL. RESULTS NEC neonatal rat models were established successfully, in which TL1A was highly expressed and NF-κB signal pathway was activated, while TL1A and NF-κB signal pathway can be suppressed by AS-IV treatment in NEC rats. Meanwhile, inflammatory response in intestinal tissues was increased in NEC rat models and AS-IV can attenuate inflammatory response in NEC rats through inhibiting TL1A and NF-κb signal pathway. CONCLUSION AS-IV can inhibit TL1A expression and NF-κb signal pathway to attenuate the inflammatory response in NEC neonatal rat models.
Collapse
Affiliation(s)
- Lin Tian
- Department of Pediatrics, Wuhan Fourth Hospital, Wuhan, Hubei 430033, PR China
| | - Shuang Tao
- Department of Pediatrics, Wuhan Fourth Hospital, Wuhan, Hubei 430033, PR China.
| | - Chunzhi He
- Department of Pediatrics, Wuhan Fourth Hospital, Wuhan, Hubei 430033, PR China
| | - Shanwu Dong
- Department of Pediatrics, Wuhan Fourth Hospital, Wuhan, Hubei 430033, PR China
| | - Yongli Chen
- Department of Pediatrics, Wuhan Fourth Hospital, Wuhan, Hubei 430033, PR China
| | - Ling Chen
- Department of Pediatrics, Wuhan Fourth Hospital, Wuhan, Hubei 430033, PR China
| | - Shu Jiang
- Department of Pediatrics, Wuhan Fourth Hospital, Wuhan, Hubei 430033, PR China
| |
Collapse
|
38
|
Luo S, Liman N, Li C, Crossman A, Wang ECY, Meylan F, Park JH. The cytokine receptor DR3 identifies and promotes the activation of thymic NKT17 cells. Cell Mol Life Sci 2023; 80:76. [PMID: 36847849 PMCID: PMC10838626 DOI: 10.1007/s00018-023-04726-7] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2022] [Revised: 02/10/2023] [Accepted: 02/13/2023] [Indexed: 03/01/2023]
Abstract
Invariant natural killer T (iNKT) cells correspond to a population of thymus-generated T cells with innate-like characteristics and effector functions. Among the various iNKT subsets, NKT17 is the only subset that produces the proinflammatory cytokine IL-17. But, how NKT17 cells acquire this ability and what would selectively trigger their activation remain incompletely understood. Here, we identified the cytokine receptor DR3 being specifically expressed on thymic NKT17 cells and mostly absent on other thymic iNKT subsets. Moreover, DR3 ligation promoted the in vivo activation of thymic NKT17 cells and provided costimulatory effects upon agonistic α-GalCer stimulation. Thus, we identified a specific surface marker for thymic NKT17 cells that triggers their activation and augments their effector functions both in vivo and in vitro. These findings provide new insights for deciphering the role and function of murine NKT17 cells and for understanding the development and activation mechanisms of iNKT cells in general.
Collapse
Affiliation(s)
- Shunqun Luo
- Experimental Immunology Branch, Center for Cancer Research, National Cancer Institute, NIH, Building 10, Room 5B17, 10 Center Dr, Bethesda, MD, 20892, USA
| | - Nurcin Liman
- Experimental Immunology Branch, Center for Cancer Research, National Cancer Institute, NIH, Building 10, Room 5B17, 10 Center Dr, Bethesda, MD, 20892, USA
| | - Can Li
- Experimental Immunology Branch, Center for Cancer Research, National Cancer Institute, NIH, Building 10, Room 5B17, 10 Center Dr, Bethesda, MD, 20892, USA
| | - Assiatu Crossman
- Experimental Immunology Branch, Center for Cancer Research, National Cancer Institute, NIH, Building 10, Room 5B17, 10 Center Dr, Bethesda, MD, 20892, USA
| | - Eddie C Y Wang
- Division of Infection and Immunity, School of Medicine, Cardiff University, Cardiff, UK
| | - Françoise Meylan
- Translational Immunology Section, NIAMS, NIH, Bethesda, MD, 20892, USA
| | - Jung-Hyun Park
- Experimental Immunology Branch, Center for Cancer Research, National Cancer Institute, NIH, Building 10, Room 5B17, 10 Center Dr, Bethesda, MD, 20892, USA.
| |
Collapse
|
39
|
Gomez-Bris R, Saez A, Herrero-Fernandez B, Rius C, Sanchez-Martinez H, Gonzalez-Granado JM. CD4 T-Cell Subsets and the Pathophysiology of Inflammatory Bowel Disease. Int J Mol Sci 2023; 24:2696. [PMID: 36769019 PMCID: PMC9916759 DOI: 10.3390/ijms24032696] [Citation(s) in RCA: 86] [Impact Index Per Article: 43.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2023] [Revised: 01/24/2023] [Accepted: 01/28/2023] [Indexed: 02/04/2023] Open
Abstract
Inflammatory bowel disease (IBD) is an umbrella term for the chronic immune-mediated idiopathic inflammation of the gastrointestinal tract, manifesting as Crohn's disease (CD) or ulcerative colitis (UC). IBD is characterized by exacerbated innate and adaptive immunity in the gut in association with microbiota dysbiosis and the disruption of the intestinal barrier, resulting in increased bacterial exposure. In response to signals from microorganisms and damaged tissue, innate immune cells produce inflammatory cytokines and factors that stimulate T and B cells of the adaptive immune system, and a prominent characteristic of IBD patients is the accumulation of inflammatory T-cells and their proinflammatory-associated cytokines in intestinal tissue. Upon antigen recognition and activation, CD4 T-cells differentiate towards a range of distinct phenotypes: T helper(h)1, Th2, Th9, Th17, Th22, T follicular helper (Tfh), and several types of T-regulatory cells (Treg). T-cells are generated according to and adapt to microenvironmental conditions and participate in a complex network of interactions among other immune cells that modulate the further progression of IBD. This review examines the role of the CD4 T-cells most relevant to IBD, highlighting how these cells adapt to the environment and interact with other cell populations to promote or inhibit the development of IBD.
Collapse
Affiliation(s)
- Raquel Gomez-Bris
- LamImSys Lab, Instituto de Investigación Sanitaria Hospital 12 de Octubre (imas12), 28041 Madrid, Spain
- Departamento de Fisiología, Facultad de Medicina, Universidad Autónoma de Madrid (UAM), 28029 Madrid, Spain
| | - Angela Saez
- LamImSys Lab, Instituto de Investigación Sanitaria Hospital 12 de Octubre (imas12), 28041 Madrid, Spain
- Facultad de Ciencias Experimentales, Universidad Francisco de Vitoria (UFV), 28223 Pozuelo de Alarcón, Spain
| | - Beatriz Herrero-Fernandez
- LamImSys Lab, Instituto de Investigación Sanitaria Hospital 12 de Octubre (imas12), 28041 Madrid, Spain
- Departamento de Fisiología, Facultad de Medicina, Universidad Autónoma de Madrid (UAM), 28029 Madrid, Spain
| | - Cristina Rius
- Department of History of Science and Information Science, School of Medicine and Dentistry, University of Valencia, 46010 Valencia, Spain
- UISYS Research Unit, University of Valencia, 46010 Valencia, Spain
- CIBER de Enfermedades Cardiovasculares (CIBERCV), 28029 Madrid, Spain
| | - Hector Sanchez-Martinez
- LamImSys Lab, Instituto de Investigación Sanitaria Hospital 12 de Octubre (imas12), 28041 Madrid, Spain
| | - Jose M. Gonzalez-Granado
- LamImSys Lab, Instituto de Investigación Sanitaria Hospital 12 de Octubre (imas12), 28041 Madrid, Spain
- CIBER de Enfermedades Cardiovasculares (CIBERCV), 28029 Madrid, Spain
- Department of Immunology, Ophthalmology and ENT, School of Medicine, Universidad Complutense de Madrid, 28040 Madrid, Spain
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), 28029 Madrid, Spain
| |
Collapse
|
40
|
Wang Z, Yao C, Huang L, Liang J, Zhang X, Shi J, Wei W, Zhou J, Zhang Y, Wu G. Enhanced external counterpulsation improves dysfunction of forearm muscle caused by radial artery occlusion. Front Cardiovasc Med 2023; 10:1115494. [PMID: 36937941 PMCID: PMC10022471 DOI: 10.3389/fcvm.2023.1115494] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2022] [Accepted: 02/14/2023] [Indexed: 03/06/2023] Open
Abstract
Objective This study aimed to investigate the therapeutic effect of enhanced external counterpulsation (EECP) on radial artery occlusion (RAO) through the oscillatory shear (OS) and pulsatile shear (PS) models of human umbilical vein endothelial cells (HUVECs) and RAO dog models. Methods We used high-throughput sequencing data GSE92506 in GEO database to conduct time-series analysis of functional molecules on OS intervened HUVECs, and then compared the different molecules and their functions between PS and OS. Additionally, we studied the effect of EECP on the radial artery hemodynamics in Labrador dogs through multi-channel physiological monitor. Finally, we studied the therapeutic effect of EECP on RAO at the histological level through Hematoxylin-Eosin staining, Masson staining, ATPase staining and immunofluorescence in nine Labrador dogs. Results With the extension of OS intervention, the cell cycle decreased, blood vessel endothelial cell proliferation and angiogenesis responses of HUVECs were down-regulated. By contrast, the inflammation and oxidative stress responses and the related pathways of anaerobic metabolism of HUVECs were up-regulated. Additionally, we found that compared with OS, PS can significantly up-regulate muscle synthesis, angiogenesis, and NO production related molecules. Meanwhile, PS can significantly down-regulate inflammation and oxidative stress related molecules. The invasive arterial pressure monitoring showed that 30Kpa EECP treatment could significantly increase the radial artery peak pressure (p = 0.030, 95%CI, 7.236-82.524). Masson staining showed that RAO significantly increased muscle interstitial fibrosis (p = 0.002, 95%CI, 0.748-2.128), and EECP treatment can reduce this change (p = 0.011, 95%CI, -1.676 to -0.296). ATPase staining showed that RAO significantly increased the area of type II muscle fibers (p = 0.004, 95%CI, 7.181-25.326), and EECP treatment could reduce this change (p = 0.001, 95%CI, -29.213 to -11.069). In addition, immunofluorescence showed that EECP increased angiogenesis in muscle tissue (p = 0.035, 95%CI, 0.024-0.528). Conclusion EECP improves interstitial fibrosis and hypoxia, and increases angiogenesis of muscle tissue around radial artery induced by RAO.
Collapse
Affiliation(s)
- Zhenyu Wang
- Department of Cardiology, The Eighth Affiliated Hospital of Sun Yat-sen University, Shenzhen, Guangdong, China
| | - Chun Yao
- Department of Cardiology, The Eighth Affiliated Hospital of Sun Yat-sen University, Shenzhen, Guangdong, China
| | - Lihan Huang
- Department of Cardiology, The Eighth Affiliated Hospital of Sun Yat-sen University, Shenzhen, Guangdong, China
| | - Jianwen Liang
- Department of Cardiology, The Eighth Affiliated Hospital of Sun Yat-sen University, Shenzhen, Guangdong, China
| | - Xiaocong Zhang
- Department of Cardiology, Foshan Fosun Chancheng Hospital, Foshan, Guangdong, China
| | - Jian Shi
- Department of Cardiology, The Eighth Affiliated Hospital of Sun Yat-sen University, Shenzhen, Guangdong, China
| | - Wenbin Wei
- Department of Cardiology, The Eighth Affiliated Hospital of Sun Yat-sen University, Shenzhen, Guangdong, China
| | - Jing Zhou
- Department of Cardiology, Affiliated Hospital of Yan’an University, Yan’an, Shaanxi, China
| | - Yahui Zhang
- Department of Cardiology, The Eighth Affiliated Hospital of Sun Yat-sen University, Shenzhen, Guangdong, China
- School of Rehabilitation Sciences and Engineering, University of Health and Rehabilitation Sciences, Shandong, China
- Yahui Zhang,
| | - Guifu Wu
- Department of Cardiology, The Eighth Affiliated Hospital of Sun Yat-sen University, Shenzhen, Guangdong, China
- Guangdong Innovative Engineering and Technology Research Center for Assisted Circulation, Sun Yat-sen University, Shenzhen, Guangdong, China
- NHC Key Laboratory of Assisted Circulation, Sun Yat-sen University, Guangzhou, Guangdong, China
- *Correspondence: Guifu Wu,
| |
Collapse
|
41
|
Genetic and Epigenetic Etiology of Inflammatory Bowel Disease: An Update. Genes (Basel) 2022; 13:genes13122388. [PMID: 36553655 PMCID: PMC9778199 DOI: 10.3390/genes13122388] [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: 11/17/2022] [Revised: 12/06/2022] [Accepted: 12/09/2022] [Indexed: 12/23/2022] Open
Abstract
Inflammatory bowel disease (IBD) is a chronic disease with periods of exacerbation and remission of the disease. The etiology of IBD is not fully understood. Many studies point to the presence of genetic, immunological, environmental, and microbiological factors and the interactions between them in the occurrence of IBD. The review looks at genetic factors in the context of both IBD predisposition and pharmacogenetics.
Collapse
|
42
|
Lu HJ, Chuang CY, Su CW, Chen MK, Yang WE, Yeh CM, Tang CH, Lin CW, Yang SF. Role of TNFSF15 variants in oral cancer development and clinicopathologic characteristics. J Cell Mol Med 2022; 26:5452-5462. [PMID: 36226563 PMCID: PMC9639028 DOI: 10.1111/jcmm.17569] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2022] [Revised: 09/02/2022] [Accepted: 09/20/2022] [Indexed: 12/04/2022] Open
Abstract
Tumour necrosis family superfamily (TNFSF) member 15 (TNFSF15), encoded by TNFSF15, regulates immune responses and inflammation. However, the roles of TNFSF15 single‐nucleotide variants (SNVs; formerly SNPs) in oral cavity squamous cell carcinoma (OCSCC) remain unclear. This case–control study included 2523 participants (1324 patients with OCSCC [52.5%] and 1199 healthy controls [47.5%]). The effects of TNFSF15 rs3810936, rs6478108 and rs6478109 on cancer development and prognosis were analysed by real‐time PCR genotype assay. The Genotype‐Tissue Expression (GTEx) and The Cancer Genome Atlas (TCGA) databases were used to validate our findings. The results demonstrated that the patients with altered TNFSF15 SNVs had poorer histological differentiation than did those with wild‐type alleles. TNFSF15 SNVs were significantly associated with moderate‐to‐poor histological differentiation in univariate logistic regression. In the GTEx database, the expression of altered TNFSF15 SNVs in whole blood was lower than that of wild‐type alleles. However, the expression of altered SNVs in the upper aerodigestive mucosa was higher than that of wild‐type alleles. In the TCGA database, the patients with higher TNFSF15 expression had shorter overall survival than did those with lower TNFSF15 expression, especially for human papillomavirus‐negative and advanced staging groups. In conclusion, although TNFSF15 SNVs did not affect OCSCC development, the patients with altered TNFSF15 SNVs exhibited poorer histological differentiation. The patients with higher TNFSF15 expression had poorer prognosis than did those with lower TNFSF15 expression.
Collapse
Affiliation(s)
- Hsueh-Ju Lu
- Division of Hematology and Oncology, Department of Internal Medicine, Chung Shan Medical University Hospital, Taichung, Taiwan.,School of Medicine, Chung Shan Medical University, Taichung, Taiwan
| | - Chun-Yi Chuang
- School of Medicine, Chung Shan Medical University, Taichung, Taiwan.,Department of Otolaryngology, Chung Shan Medical University Hospital, Taichung, Taiwan
| | - Chun-Wen Su
- Department of Medical Research, Chung Shan Medical University Hospital, Taichung, Taiwan.,Institute of Medicine, Chung Shan Medical University, Taichung, Taiwan
| | - Mu-Kuan Chen
- Institute of Medicine, Chung Shan Medical University, Taichung, Taiwan.,Department of Otorhinolaryngology-Head and Neck Surgery, Changhua Christian Hospital, Changhua, Taiwan.,Oral cancer Research Center, Changhua Christian Hospital, Changhua, Taiwan
| | - Wei-En Yang
- Department of Medical Research, Chung Shan Medical University Hospital, Taichung, Taiwan.,Institute of Medicine, Chung Shan Medical University, Taichung, Taiwan
| | - Chia-Ming Yeh
- Institute of Medicine, Chung Shan Medical University, Taichung, Taiwan.,Oral cancer Research Center, Changhua Christian Hospital, Changhua, Taiwan
| | - Chih-Hsin Tang
- School of Medicine, China Medical University, Taichung, Taiwan.,Chinese Medicine Research Center, China Medical University, Taichung, Taiwan.,Department of Medical Laboratory Science and Biotechnology, College of Medical and Health Science, Asia University, Taichung, Taiwan
| | - Chiao-Wen Lin
- Institute of Oral Sciences, Chung Shan Medical University, Taichung, Taiwan.,Department of Dentistry, Chung Shan Medical University Hospital, Taichung, Taiwan
| | - Shun-Fa Yang
- Department of Medical Research, Chung Shan Medical University Hospital, Taichung, Taiwan.,Institute of Medicine, Chung Shan Medical University, Taichung, Taiwan
| |
Collapse
|
43
|
Xu M, Mu J, Wang J, Zhou Q, Wang J. Construction and validation of a cuproptosis-related lncRNA signature as a novel and robust prognostic model for colon adenocarcinoma. Front Oncol 2022; 12:961213. [PMID: 35965536 PMCID: PMC9367690 DOI: 10.3389/fonc.2022.961213] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2022] [Accepted: 07/05/2022] [Indexed: 12/22/2022] Open
Abstract
BackgroundCuproptosis, a newly identified form of programmed cell death, is thought to play a role in tumorigenesis. Long non-coding RNAs (lncRNAs) are reported to be associated with tumor progression and prognosis in colon adenocarcinoma (COAD). However, the role and prognostic value of cuproptosis-related lncRNAs in COAD remains unknown. This study is devoted to constructing and validating a cuproptosis-related lncRNA signature that can predict COAD patient outcomes using bioinformatics methods.MethodsThe COAD mRNA and lncRNA expression profiles and corresponding clinical data were downloaded from The Cancer Genome Atlas (TCGA) database and 2,567 cuproptosis-related lncRNAs were obtained. A 10 cuproptosis-related-lncRNA prognostic signature was then constructed using the least absolute shrinkage and selection operator (LASSO) algorithm and Cox regression model and patients were divided into high- and low-risk groups. Kaplan-Meier analysis, receiver operating characteristic (ROC) curve, and a nomogram were employed to evaluate the predictive power of the signature. The immune characteristics and drug sensitivity were also investigated based on the signature. Quantitative reverse transcription polymerase chain reaction (qRT-PCR) was performed to verify the risk model. In vitro experiments were conducted to validate the expression of the ten lncRNAs during cuproptosis.ResultsThe high-risk group was associated with shorter overall survival (OS) time in COAD patients (p<0.001). Multivariate Cox regression indicated that a high-risk score was an independent risk factor for poor prognosis (p<0.001). ROC curve analysis was performed to confirm the validity of the signature (area under the curve (AUC) at 3 years: 0.879). Gene Ontology (GO) enrichment analysis revealed that the signature was highly correlated with the immune response in biological processes. The immune function, the score of the immune cells, and the expression of immune checkpoints were significantly different between the two risk groups. Three drugs, LAQ824, FH535, YM155, were found to be more sensitive in the high-risk group. Finally, the expression levels of the ten lncRNAs comprising the signature were tested by qRT-PCR.ConclusionA ten-cuproptosis-related lncRNA signature was constructed that provided promising insights into personalized prognosis and drug selection among COAD patients.
Collapse
Affiliation(s)
- Miaorong Xu
- Department of Colorectal Surgery and Oncology, Key Laboratory of Cancer Prevention and Intervention, Ministry of Education, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Jiayi Mu
- Department of Colorectal Surgery and Oncology, Key Laboratory of Cancer Prevention and Intervention, Ministry of Education, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Jiaojiao Wang
- Department of Colorectal Surgery and Oncology, Key Laboratory of Cancer Prevention and Intervention, Ministry of Education, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Qin Zhou
- Department of Colorectal Surgery and Oncology, Key Laboratory of Cancer Prevention and Intervention, Ministry of Education, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Jianwei Wang
- Department of Colorectal Surgery and Oncology, Key Laboratory of Cancer Prevention and Intervention, Ministry of Education, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Department of Colorectal Surgery, 4th Affiliated Hospital, Zhejiang University School of Medicine, Yiwu, China
- *Correspondence: Jianwei Wang,
| |
Collapse
|
44
|
Zhu JJ, Stenfeldt C, Bishop EA, Canter JA, Eschbaumer M, Rodriguez LL, Arzt J. Inferred Causal Mechanisms of Persistent FMDV Infection in Cattle from Differential Gene Expression in the Nasopharyngeal Mucosa. Pathogens 2022; 11:pathogens11080822. [PMID: 35894045 PMCID: PMC9329776 DOI: 10.3390/pathogens11080822] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2022] [Revised: 07/14/2022] [Accepted: 07/18/2022] [Indexed: 02/05/2023] Open
Abstract
Foot-and-mouth disease virus (FMDV) can persistently infect pharyngeal epithelia in ruminants but not in pigs. Our previous studies demonstrated that persistent FMDV infection in cattle was associated with under-expression of several chemokines that recruit immune cells. This report focuses on the analysis of differentially expressed genes (DEG) identified during the transitional phase of infection, defined as the period when animals diverge between becoming carriers or terminators. During this phase, Th17-stimulating cytokines (IL6 and IL23A) and Th17-recruiting chemokines (CCL14 and CCL20) were upregulated in animals that were still infected (transitional carriers) compared to those that had recently cleared infection (terminators), whereas chemokines recruiting neutrophils and CD8+ T effector cells (CCL3 and ELR+CXCLs) were downregulated. Upregulated Th17-specific receptor, CCR6, and Th17-associated genes, CD146, MIR155, and ThPOK, suggested increased Th17 cell activity in transitional carriers. However, a complex interplay of the Th17 regulatory axis was indicated by non-significant upregulation of IL17A and downregulation of IL17F, two hallmarks of TH17 activity. Other DEG suggested that transitional carriers had upregulated aryl hydrocarbon receptor (AHR), non-canonical NFκB signaling, and downregulated canonical NFκB signaling. The results described herein provide novel insights into the mechanisms of establishment of FMDV persistence. Additionally, the fact that ruminants, unlike pigs, produce a large amount of AHR ligands suggests a plausible explanation of why FMDV persists in ruminants, but not in pigs.
Collapse
Affiliation(s)
- James J. Zhu
- Foreign Animal Disease Research Unit, Plum Island Animal Disease Center, Agricultural Research Service, United States Department of Agriculture, Orient, NY 11957, USA; (C.S.); (E.A.B.); (J.A.C.); (L.L.R.)
- Correspondence: (J.J.Z.); (J.A.); Tel.: +1-631-323-3340 (J.J.Z.); +1-631-323-4421 (J.A.); Fax: +1-631-323-3006 (J.A.)
| | - Carolina Stenfeldt
- Foreign Animal Disease Research Unit, Plum Island Animal Disease Center, Agricultural Research Service, United States Department of Agriculture, Orient, NY 11957, USA; (C.S.); (E.A.B.); (J.A.C.); (L.L.R.)
- Department of Diagnostic Medicine/Pathobiology, Kansas State University, Manhattan, KS 66506, USA
| | - Elizabeth A. Bishop
- Foreign Animal Disease Research Unit, Plum Island Animal Disease Center, Agricultural Research Service, United States Department of Agriculture, Orient, NY 11957, USA; (C.S.); (E.A.B.); (J.A.C.); (L.L.R.)
| | - Jessica A. Canter
- Foreign Animal Disease Research Unit, Plum Island Animal Disease Center, Agricultural Research Service, United States Department of Agriculture, Orient, NY 11957, USA; (C.S.); (E.A.B.); (J.A.C.); (L.L.R.)
- Plum Island Animal Disease Center Research Participation Program, Oak Ridge Institute for Science and Education (ORISE), Oak Ridge, TN 37830, USA
| | - Michael Eschbaumer
- Institute of Diagnostic Virology, Friedrich-Loeffler-Institut, 17493 Greifswald-Insel Riems, Germany;
| | - Luis L. Rodriguez
- Foreign Animal Disease Research Unit, Plum Island Animal Disease Center, Agricultural Research Service, United States Department of Agriculture, Orient, NY 11957, USA; (C.S.); (E.A.B.); (J.A.C.); (L.L.R.)
| | - Jonathan Arzt
- Foreign Animal Disease Research Unit, Plum Island Animal Disease Center, Agricultural Research Service, United States Department of Agriculture, Orient, NY 11957, USA; (C.S.); (E.A.B.); (J.A.C.); (L.L.R.)
- Correspondence: (J.J.Z.); (J.A.); Tel.: +1-631-323-3340 (J.J.Z.); +1-631-323-4421 (J.A.); Fax: +1-631-323-3006 (J.A.)
| |
Collapse
|
45
|
Guo H, Qian Y, Yu Y, Bi Y, Jiao J, Jiang H, Yu C, Wu H, Shi Y, Kong X. An Immunity-Related Gene Model Predicts Prognosis in Cholangiocarcinoma. Front Oncol 2022; 12:791867. [PMID: 35847907 PMCID: PMC9283581 DOI: 10.3389/fonc.2022.791867] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2021] [Accepted: 05/31/2022] [Indexed: 12/11/2022] Open
Abstract
The prognosis of patients with cholangiocarcinoma (CCA) is closely related to both immune cell infiltration and mRNA expression. Therefore, we aimed at conducting multi-immune-related gene analyses to improve the prediction of CCA recurrence. Immune-related genes were selected from the Gene Expression Omnibus (GEO), The Cancer Genome Atlas (TCGA), and the Immunology Database and Analysis Portal (ImmPort). The least absolute shrinkage and selection operator (LASSO) regression model was used to establish the multi-gene model that was significantly correlated with the recurrence-free survival (RFS) in two test series. Furthermore, compared with single genes, clinical characteristics, tumor immune dysfunction and exclusion (TIDE), and tumor inflammation signature (TIS), the 8-immune-related differentially expressed genes (8-IRDEGs) signature had a better prediction value. Moreover, the high-risk subgroup had a lower density of B-cell, plasma, B-cell naïve, CD8+ T-cell, CD8+ T-cell naïve, and CD8+ T-cell memory infiltration, as well as more severe immunosuppression and higher mutation counts. In conclusion, the 8-IRDEGs signature was a promising biomarker for distinguishing the prognosis and the molecular and immune features of CCA, and could be beneficial to the individualized immunotherapy for CCA patients.
Collapse
Affiliation(s)
- Han Guo
- Department of Oncology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
- Institute of Clinical Immunology, Department of Liver Diseases, Central Laboratory, Shuguang Hospital Affiliated to Shanghai University of Chinese Traditional Medicine, Shanghai, China
| | - Yihan Qian
- Institute of Clinical Immunology, Department of Liver Diseases, Central Laboratory, Shuguang Hospital Affiliated to Shanghai University of Chinese Traditional Medicine, Shanghai, China
| | - Yeping Yu
- Department of Liver Surgery, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Yuting Bi
- Institute of Clinical Immunology, Department of Liver Diseases, Central Laboratory, Shuguang Hospital Affiliated to Shanghai University of Chinese Traditional Medicine, Shanghai, China
| | - Junzhe Jiao
- Institute of Clinical Immunology, Department of Liver Diseases, Central Laboratory, Shuguang Hospital Affiliated to Shanghai University of Chinese Traditional Medicine, Shanghai, China
| | - Haocheng Jiang
- Institute of Clinical Immunology, Department of Liver Diseases, Central Laboratory, Shuguang Hospital Affiliated to Shanghai University of Chinese Traditional Medicine, Shanghai, China
| | - Chang Yu
- Institute of Clinical Immunology, Department of Liver Diseases, Central Laboratory, Shuguang Hospital Affiliated to Shanghai University of Chinese Traditional Medicine, Shanghai, China
| | - Hailong Wu
- Shanghai Key Laboratory for Molecular Imaging, Collaborative Research Center, Shanghai University of Medicine and Health Sciences, Shanghai, China
- *Correspondence: Xiaoni Kong, ; Yanjun Shi, ; Hailong Wu,
| | - Yanjun Shi
- Department of Hepatobiliary and Pancreas Surgery , The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
- *Correspondence: Xiaoni Kong, ; Yanjun Shi, ; Hailong Wu,
| | - Xiaoni Kong
- Institute of Clinical Immunology, Department of Liver Diseases, Central Laboratory, Shuguang Hospital Affiliated to Shanghai University of Chinese Traditional Medicine, Shanghai, China
- *Correspondence: Xiaoni Kong, ; Yanjun Shi, ; Hailong Wu,
| |
Collapse
|
46
|
Wang L, Sun X, He J, Liu Z. Identification and Validation of Prognostic Related Hallmark ATP-Binding Cassette Transporters Associated With Immune Cell Infiltration Patterns in Thyroid Carcinoma. Front Oncol 2022; 12:781686. [PMID: 35837087 PMCID: PMC9273952 DOI: 10.3389/fonc.2022.781686] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2021] [Accepted: 06/01/2022] [Indexed: 11/13/2022] Open
Abstract
ATP-binding cassette (ABC) transporters are a large superfamily of membrane proteins that facilitate the translocation of heterogeneous substrates. Studies indicate that ABC transporters may play important roles in various carcinomas. However, the correlation between ABC transporters and immunomodulation in thyroid carcinoma (TC), as well as the prognoses for this disease, is poorly understood.TC data from The Cancer Genome Atlas (TCGA) database were used to identify prognostic hallmark ABC transporters associated with immune cell infiltration patterns via multiple bioinformatic analyses. Thereafter, quantitative real-time polymerase chain reaction (qRT-PCR) was performed to validate the expression of these selected hallmark ABC transporters in both TC and para-cancerous thyroid tissues. Of a total of 49 ABC transporters, five (ABCA8, ABCA12, ABCB6, ABCB8, and ABCC10) were identified as hallmark ABC transporters. All five were differentially expressed in TC and associated with the relapse-free survival rates of patients with TC. Immunoregulation by these five hallmark ABC transporters involved the modulation of various aspects of immune cell infiltration, such as hot or cold tumor subsets and the abundances of infiltrating immune cells, as well as specific immunomodulators and chemokines. Besides the diverse significantly correlated factors, the five hallmark ABC transporters and correlated genes were most highly enriched in plasma membrane, transporter activity, and transmembrane transport of small molecules. In addition, many chemicals, namely bisphenol A and vincristine, affected the expression of these five transporters. The qRT-PCR results of collected TC and para-cancerous thyroid tissues were consistent with those of TCGA. The findings in this study may reveal the role played by these five hallmark ABC transporters in regulating immune cell infiltration patterns in TC as well as the molecular mechanisms underlying their functions, leading to a better understanding of their potential prognostic and immunotherapeutic values.
Collapse
Affiliation(s)
- Lidong Wang
- Department of General Surgery, Shengjing Hospital of China Medical University, Shenyang, China
| | - Xiaodan Sun
- Postdoctoral Research Workstation, Jilin Cancer Hospital, Changchun, China
- Department of 1st Gynecologic Oncology Surgery, Jilin Cancer Hospital, Changchun, China
| | - Jingni He
- Department of General Surgery, Shengjing Hospital of China Medical University, Shenyang, China
| | - Zhen Liu
- Department of General Surgery, Shengjing Hospital of China Medical University, Shenyang, China
- *Correspondence: Zhen Liu,
| |
Collapse
|
47
|
Kokkotis G, Bamias G. TL1A as a therapeutic target in inflammatory bowel disease. Expert Rev Clin Immunol 2022; 18:551-555. [PMID: 35507314 DOI: 10.1080/1744666x.2022.2074401] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2022] [Accepted: 05/03/2022] [Indexed: 11/04/2022]
Affiliation(s)
- Georgios Kokkotis
- GI Unit, 3 Department of Internal Medicine, Sotiria Hospital, National and Kapodistrian University of Athens, Athens, Greece
| | - Giorgos Bamias
- GI Unit, 3 Department of Internal Medicine, Sotiria Hospital, National and Kapodistrian University of Athens, Athens, Greece
| |
Collapse
|
48
|
Zhang S, Zhong R, Tang S, Han H, Chen L, Zhang H. Baicalin Alleviates Short-Term Lincomycin-Induced Intestinal and Liver Injury and Inflammation in Infant Mice. Int J Mol Sci 2022; 23:ijms23116072. [PMID: 35682750 PMCID: PMC9181170 DOI: 10.3390/ijms23116072] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2022] [Revised: 05/22/2022] [Accepted: 05/26/2022] [Indexed: 11/24/2022] Open
Abstract
The adverse effects of short-term megadose of antibiotics exposure on the gastrointestinal and liver tissue reactions in young children have been reported. Antibiotic-induced intestinal and liver reactions are usually unpredictable and present a poorly understood pathogenesis. It is, therefore, necessary to develop strategies for reducing the adverse effects of antibiotics. Studies on the harm and rescue measures of antibiotics from the perspective of the gut–liver system are lacking. Here, we demonstrate that lincomycin exposure reduced body weight, disrupted the composition of gut microbiota and intestinal morphology, triggered immune-mediated injury and inflammation, caused liver dysfunction, and affected lipid metabolism. However, baicalin administration attenuated the lincomycin-induced changes. Transcriptome analysis showed that baicalin improved immunity in mice, as evidenced by the decreased levels of intestinal inflammatory cytokines and expression of genes that regulate Th1, Th2, and Th17 cell differentiation, and inhibited mucin type O-glycan biosynthesis pathways. In addition, baicalin improved liver function by upregulating the expression of genes involved in bile acid secretion and lipid degradation, and downregulating genes involved in lipid synthesis in lincomycin-treated mice. Bile acids can regulate intestinal immunity and strengthen hepatoenteric circulation. In addition, baicalin also improved anti-inflammatory bacteria abundance (Blautia and Coprobacillus) and reduced pathogenic bacteria abundance (Proteobacteria, Klebsiella, and Citrobacter) in lincomycin-treated mice. Thus, baicalin can ameliorate antibiotic-induced injury and its associated complications such as liver disease.
Collapse
Affiliation(s)
| | | | | | | | - Liang Chen
- Correspondence: (L.C.); (H.Z.); Tel.: +86-10-6281-8910 (L.C.); Fax: +86-10-6281-6013 (H.Z.)
| | - Hongfu Zhang
- Correspondence: (L.C.); (H.Z.); Tel.: +86-10-6281-8910 (L.C.); Fax: +86-10-6281-6013 (H.Z.)
| |
Collapse
|
49
|
Zhang D, Yang H, Dong XL, Zhang JT, Liu XF, Pan Y, Zhang J, Xu JW, Wang ZH, Cui WJ, Dong L. TL1A/DR3 Axis, A Key Target of TNF-a, Augments the Epithelial–Mesenchymal Transformation of Epithelial Cells in OVA-Induced Asthma. Front Immunol 2022; 13:854995. [PMID: 35359966 PMCID: PMC8963920 DOI: 10.3389/fimmu.2022.854995] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2022] [Accepted: 02/16/2022] [Indexed: 01/16/2023] Open
Abstract
Tumor necrosis factor (TNF)-like cytokine 1A (TL1A), a member of the TNF family, exists in the form of membrane-bound (mTL1A) and soluble protein (sTL1A). TL1A binding its only known functional receptor death domain receptor 3 (DR3) affects the transmission of various signals. This study first proposed that the TL1A/DR3 axis was significantly upregulated in patients and mice with both asthma and high TNF-a expression and in TNF-a-stimulated epithelial Beas-2B cells. Two independent approaches were used to demonstrate that the TL1A/DR3 axis of mice was strongly correlated with TNF-a in terms of exacerbating asthmatic epithelial–mesenchymal transformation (EMT). First, high expression levels of EMT proteins (e.g., collagen I, fibronectin, N-cadherin, and vimentin) and TL1A/DR3 axis were observed when mice airways were stimulated by recombinant mouse TNF-a protein. Moreover, EMT protein and TL1A/DR3 axis expression synchronously decreased after mice with OVA-induced asthma were treated with infliximab by neutralizing TNF-a activity. Furthermore, the OVA-induced EMT of asthmatic mice was remarkably improved upon the deletion of the TL1A/DR3 axis by knocking out the TL1A gene. TL1A siRNA remarkably intervened EMT formation induced by TNF-a in the Beas-2B cells. In addition, EMT was induced by the addition of high concentrations of recombinant human sTL1A with the cell medium. The TL1A overexpression via pc-mTL1A in vitro remarkably increased the EMT formation induced by TNF-a. Overall, these findings indicate that the TL1A/DR3 axis may have a therapeutic role for asthmatic with high TNF-a level.
Collapse
Affiliation(s)
- Dong Zhang
- Department of Respiratory, Shandong Provincial Qianfoshan Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Hui Yang
- Department of Respiratory, Shandong Provincial Qianfoshan Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Xue-Li Dong
- Department of Respiratory, Shandong Provincial Qianfoshan Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Jin-Tao Zhang
- Department of Respiratory, Shandong Provincial Qianfoshan Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Xiao-Fei Liu
- Department of Respiratory, The First Affiliated Hospital of Shandong First Medical University and Shandong Provincial Qianfoshan Hospital, Shandong Institute of Respiratory Diseases, Jinan, China
| | - Yun Pan
- Department of Respiratory, Shandong Provincial Qianfoshan Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Jian Zhang
- Department of Respiratory, The First Affiliated Hospital of Shandong First Medical University and Shandong Provincial Qianfoshan Hospital, Shandong Institute of Respiratory Diseases, Jinan, China
| | - Jia-Wei Xu
- Department of Respiratory, The First Affiliated Hospital of Shandong First Medical University and Shandong Provincial Qianfoshan Hospital, Shandong Institute of Respiratory Diseases, Jinan, China
| | - Zi-Han Wang
- Department of Respiratory, Shandong Provincial Qianfoshan Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Wen-Jing Cui
- Department of Respiratory, Shandong Provincial Qianfoshan Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Liang Dong
- Department of Respiratory, Shandong Provincial Qianfoshan Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
- Department of Respiratory, The First Affiliated Hospital of Shandong First Medical University and Shandong Provincial Qianfoshan Hospital, Shandong Institute of Respiratory Diseases, Jinan, China
- *Correspondence: Liang Dong,
| |
Collapse
|
50
|
Hassan-Zahraee M, Ye Z, Xi L, Baniecki ML, Li X, Hyde CL, Zhang J, Raha N, Karlsson F, Quan J, Ziemek D, Neelakantan S, Lepsy C, Allegretti JR, Romatowski J, Scherl EJ, Klopocka M, Danese S, Chandra DE, Schoenbeck U, Vincent MS, Longman R, Hung KE. Antitumor Necrosis Factor-like Ligand 1A Therapy Targets Tissue Inflammation and Fibrosis Pathways and Reduces Gut Pathobionts in Ulcerative Colitis. Inflamm Bowel Dis 2022; 28:434-446. [PMID: 34427649 PMCID: PMC8889296 DOI: 10.1093/ibd/izab193] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Indexed: 12/21/2022]
Abstract
BACKGROUND The first-in-class treatment PF-06480605 targets the tumor necrosis factor-like ligand 1A (TL1A) molecule in humans. Results from the phase 2a TUSCANY trial highlighted the safety and efficacy of PF-06480605 in ulcerative colitis. Preclinical and in vitro models have identified a role for TL1A in both innate and adaptive immune responses, but the mechanisms underlying the efficacy of anti-TL1A treatment in inflammatory bowel disease (IBD) are not known. METHODS Here, we provide analysis of tissue transcriptomic, peripheral blood proteomic, and fecal metagenomic data from the recently completed phase 2a TUSCANY trial and demonstrate endoscopic improvement post-treatment with PF-06480605 in participants with ulcerative colitis. RESULTS Our results revealed robust TL1A target engagement in colonic tissue and a distinct colonic transcriptional response reflecting a reduction in inflammatory T helper 17 cell, macrophage, and fibrosis pathways in patients with endoscopic improvement. Proteomic analysis of peripheral blood revealed a corresponding decrease in inflammatory T-cell cytokines. Finally, microbiome analysis showed significant changes in IBD-associated pathobionts, Streptococcus salivarius, S. parasanguinis, and Haemophilus parainfluenzae post-therapy. CONCLUSIONS The ability of PF-06480605 to engage and inhibit colonic TL1A, targeting inflammatory T cell and fibrosis pathways, provides the first-in-human mechanistic data to guide anti-TL1A therapy for the treatment of IBD.
Collapse
Affiliation(s)
| | - Zhan Ye
- Pfizer Inc, Cambridge, MA, USA
| | - Li Xi
- Pfizer Inc, Cambridge, MA, USA
| | | | | | | | | | | | | | | | | | | | | | - Jessica R Allegretti
- Brigham and Women’s Hospital, Harvard Medical School, Division of Gastroenterology, Boston, MA, USA
| | - Jacek Romatowski
- J. Sniadecki’s Regional Hospital, Internal Medicine and Gastroenterology Department, Białystok, Poland
| | - Ellen J Scherl
- Jill Roberts Center for IBD, Weill Cornell Medicine, Division of Gastroenterology and Hepatology, New York, NY, USA
| | - Maria Klopocka
- Nicolaus Copernicus University in Toruń, Collegium Medicum, Department of Gastroenterology and Nutrition, Bydgoszcz, Poland
| | - Silvio Danese
- IBD Center, Humanitas Research Hospital, Department of Gastroenterology, Milan, Italy
- Humanitas University, Department of Biomedical Sciences, Milan, Italy
| | | | | | | | - Randy Longman
- Jill Roberts Center for IBD, Weill Cornell Medicine, Division of Gastroenterology and Hepatology, New York, NY, USA
| | | |
Collapse
|