1
|
Xu W, Golovchenko NB, Martínez-Vargas IU, Fong A, Rout P, Achi S, Bucar EB, Hsieh JJ, Vidmar KJ, Zhang L, Polydorides AD, Prinz I, Kollias G, Frey MR, Pizarro TT, Verzi MP, Edelblum KL. Dysregulation of γδ intraepithelial lymphocytes precedes Crohn's disease-like ileitis. Sci Immunol 2025; 10:eadk7429. [PMID: 40117343 DOI: 10.1126/sciimmunol.adk7429] [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: 09/08/2023] [Revised: 11/07/2024] [Accepted: 02/14/2025] [Indexed: 03/23/2025]
Abstract
Intraepithelial lymphocytes expressing the γδ T cell receptor (γδ IELs) provide immunosurveillance of the intestinal barrier but are reduced in patients with active Crohn's disease (CD). Here, we report an underappreciated role for γδ IELs in maintaining immunological tolerance during the onset and progression of CD-like ileitis using TNFΔARE/+ mice. We found that TNF-induced down-regulation of epithelial hepatocyte nuclear factor 4-gamma/butyrophilin is followed by a loss of ileal Vγ7 IELs and impaired barrier surveillance before the histological onset of disease. A reduction of immunoregulatory CD39+ γδ IELs coincided with the influx of immature, peripheral CD39neg γδ T cells into the epithelium, leading to an expansion of induced IELs, whereas an earlier depletion of γδ IELs correlated with accelerated onset of ileal inflammation. Our findings identify multiple layers of γδ IEL dysregulation before ileitis development, indicating that the loss of steady-state immunoregulatory γδ IELs may contribute to the initiation of ileal CD.
Collapse
Affiliation(s)
- Weili Xu
- Center for Immunity and Inflammation, Department of Pathology, Immunology and Laboratory Medicine, Rutgers New Jersey Medical School, Newark, NJ, USA
| | - Natasha B Golovchenko
- Center for Immunity and Inflammation, Department of Pathology, Immunology and Laboratory Medicine, Rutgers New Jersey Medical School, Newark, NJ, USA
- Department of Pathology, Molecular and Cell-Based Medicine and Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Irving U Martínez-Vargas
- Department of Pathology, Molecular and Cell-Based Medicine and Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Andrew Fong
- Center for Immunity and Inflammation, Department of Pathology, Immunology and Laboratory Medicine, Rutgers New Jersey Medical School, Newark, NJ, USA
- Department of Pathology, Molecular and Cell-Based Medicine and Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Prateeksha Rout
- Department of Genetics, Human Genetics Institute of New Jersey, Rutgers, the State University of New Jersey, Piscataway, NJ, USA
- Cancer Institute of New Jersey, Rutgers, the State University of New Jersey, New Brunswick, NJ, USA
| | - Sajan Achi
- Department of Pathology, Molecular and Cell-Based Medicine and Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Edie B Bucar
- Saban Research Institute, Children's Hospital Los Angeles, Los Angeles, CA, USA
- Departments of Pediatrics and Biochemistry and Molecular Medicine, University of Southern California Keck School of Medicine, Los Angeles, CA, USA
| | - Jonathan J Hsieh
- Saban Research Institute, Children's Hospital Los Angeles, Los Angeles, CA, USA
- Departments of Pediatrics and Biochemistry and Molecular Medicine, University of Southern California Keck School of Medicine, Los Angeles, CA, USA
| | - Kaylynn J Vidmar
- Department of Pathology, Case Western Reserve University School of Medicine, Cleveland, OH, USA
| | - Lanjing Zhang
- Department of Chemical Biology, Ernest Mario School of Pharmacy, Rutgers University, Piscataway, NJ, USA
- Department of Pathology, Princeton Medical Center, Plainsboro, NJ, USA
| | - Alexandros D Polydorides
- Department of Pathology, Molecular and Cell-Based Medicine and Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Immo Prinz
- Institute of Immunology, Hannover Medical School, Hanover, Germany
- Institute of Systems Immunology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - George Kollias
- Institute for Fundamental Biomedical Research, Biomedical Sciences Research Center "Alexander Fleming," Vari, Greece
- Department of Physiology, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - Mark R Frey
- Saban Research Institute, Children's Hospital Los Angeles, Los Angeles, CA, USA
- Departments of Pediatrics and Biochemistry and Molecular Medicine, University of Southern California Keck School of Medicine, Los Angeles, CA, USA
| | - Theresa T Pizarro
- Department of Pathology, Case Western Reserve University School of Medicine, Cleveland, OH, USA
| | - Michael P Verzi
- Department of Genetics, Human Genetics Institute of New Jersey, Rutgers, the State University of New Jersey, Piscataway, NJ, USA
- Cancer Institute of New Jersey, Rutgers, the State University of New Jersey, New Brunswick, NJ, USA
| | - Karen L Edelblum
- Center for Immunity and Inflammation, Department of Pathology, Immunology and Laboratory Medicine, Rutgers New Jersey Medical School, Newark, NJ, USA
- Department of Pathology, Molecular and Cell-Based Medicine and Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| |
Collapse
|
2
|
Kelly C, Sartor RB, Rawls JF. Early subclinical stages of the inflammatory bowel diseases: insights from human and animal studies. Am J Physiol Gastrointest Liver Physiol 2025; 328:G17-G31. [PMID: 39499254 PMCID: PMC11901386 DOI: 10.1152/ajpgi.00252.2024] [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: 08/16/2024] [Revised: 10/30/2024] [Accepted: 10/31/2024] [Indexed: 11/07/2024]
Abstract
The inflammatory bowel diseases (IBD) occur in genetically susceptible individuals that mount inappropriate immune responses to their microbiota leading to chronic intestinal inflammation. The natural history of IBD progression begins with early subclinical stages of disease that occur before clinical diagnosis. Improved understanding of those early subclinical stages could lead to new or improved strategies for IBD diagnosis, prognostication, or prevention. Here, we review our current understanding of the early subclinical stages of IBD in humans including studies from first-degree relatives of patients with IBD and members of the general population who go on to develop IBD. We also discuss representative mouse models of IBD that can be used to investigate disease dynamics and host-microbiota relationships during these early stages. In particular, we underscore how mouse models of IBD that develop disease later in life with variable penetrance may present valuable opportunities to discern early subclinical mechanisms of disease before histological inflammation and other severe symptoms become apparent.
Collapse
Affiliation(s)
- Cecelia Kelly
- Department of Molecular Genetics and Microbiology, Duke Microbiome Center, Duke University School of Medicine, Durham, North Carolina, United States
| | - R Balfour Sartor
- Division of Gastroenterology and Hepatology, Department of Medicine, University of North Carolina, Chapel Hill, North Carolina, United States
- Department of Microbiology and Immunology, University of North Carolina, Chapel Hill, North Carolina, United States
| | - John F Rawls
- Department of Molecular Genetics and Microbiology, Duke Microbiome Center, Duke University School of Medicine, Durham, North Carolina, United States
| |
Collapse
|
3
|
Walraven T, Busch M, Wang J, Donkers JM, Duijvestein M, van de Steeg E, Kramer NI, Bouwmeester H. Elevated risk of adverse effects from foodborne contaminants and drugs in inflammatory bowel disease: a review. Arch Toxicol 2024; 98:3519-3541. [PMID: 39249550 PMCID: PMC11489187 DOI: 10.1007/s00204-024-03844-w] [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: 06/04/2024] [Accepted: 08/19/2024] [Indexed: 09/10/2024]
Abstract
The global burden of Inflammatory bowel disease (IBD) has been rising over the last decades. IBD is an intestinal disorder with a complex and largely unknown etiology. The disease is characterized by a chronically inflamed gastrointestinal tract, with intermittent phases of exacerbation and remission. This compromised intestinal barrier can contribute to, enhance, or even enable the toxicity of drugs, food-borne chemicals and particulate matter. This review discusses whether the rising prevalence of IBD in our society warrants the consideration of IBD patients as a specific population group in toxicological safety assessment. Various in vivo, ex vivo and in vitro models are discussed that can simulate hallmarks of IBD and may be used to study the effects of prevalent intestinal inflammation on the hazards of these various toxicants. In conclusion, risk assessments based on healthy individuals may not sufficiently cover IBD patient safety and it is suggested to consider this susceptible subgroup of the population in future toxicological assessments.
Collapse
Affiliation(s)
- Tom Walraven
- Division of Toxicology, Wageningen University and Research, Wageningen, The Netherlands.
| | - Mathias Busch
- Division of Toxicology, Wageningen University and Research, Wageningen, The Netherlands
| | - Jingxuan Wang
- Division of Toxicology, Wageningen University and Research, Wageningen, The Netherlands
| | - Joanne M Donkers
- Department of Metabolic Health Research, Netherlands Organization for Applied Scientific Research (TNO), Leiden, The Netherlands
| | - Marjolijn Duijvestein
- Department of Gastroenterology and Hepatology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Evita van de Steeg
- Department of Metabolic Health Research, Netherlands Organization for Applied Scientific Research (TNO), Leiden, The Netherlands
| | - Nynke I Kramer
- Division of Toxicology, Wageningen University and Research, Wageningen, The Netherlands
| | - Hans Bouwmeester
- Division of Toxicology, Wageningen University and Research, Wageningen, The Netherlands
| |
Collapse
|
4
|
Veisman I, Massey WJ, Goren I, Liu W, Chauhan G, Rieder F. Muscular hyperplasia in Crohn's disease strictures: through thick and thin. Am J Physiol Cell Physiol 2024; 327:C671-C683. [PMID: 38912732 PMCID: PMC11427014 DOI: 10.1152/ajpcell.00307.2024] [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: 05/06/2024] [Revised: 06/18/2024] [Accepted: 06/18/2024] [Indexed: 06/25/2024]
Abstract
Fibrostenosing Crohn's disease (CD) represents a challenging clinical condition characterized by the development of symptomatic strictures within the gastrointestinal tract. Despite therapeutic advancements in managing inflammation, the progression of fibrostenotic complications remains a significant concern, often necessitating surgical intervention. Recent investigations have unveiled the pivotal role of smooth muscle cell hyperplasia in driving luminal narrowing and clinical symptomatology. Drawing parallels to analogous inflammatory conditions affecting other organs, such as the airways and blood vessels, sheds light on common underlying mechanisms of muscular hyperplasia. This review synthesizes current evidence to elucidate the mechanisms underlying smooth muscle cell proliferation in CD-associated strictures, offering insights into potential therapeutic targets. By highlighting the emerging significance of muscle thickening as a novel therapeutic target, this review aims to inform future research endeavors and clinical strategies with the goal to mitigate the burden of fibrostenotic complications in CD and other conditions.
Collapse
Affiliation(s)
- Ido Veisman
- Department of Inflammation and Immunity, Lerner Research Institute, Cleveland Clinic Foundation, Cleveland, Ohio, United States
| | - William J Massey
- Department of Inflammation and Immunity, Lerner Research Institute, Cleveland Clinic Foundation, Cleveland, Ohio, United States
| | - Idan Goren
- Department of Inflammation and Immunity, Lerner Research Institute, Cleveland Clinic Foundation, Cleveland, Ohio, United States
| | - Weiwei Liu
- Department of Inflammation and Immunity, Lerner Research Institute, Cleveland Clinic Foundation, Cleveland, Ohio, United States
| | - Gaurav Chauhan
- Department of Inflammation and Immunity, Lerner Research Institute, Cleveland Clinic Foundation, Cleveland, Ohio, United States
| | - Florian Rieder
- Department of Inflammation and Immunity, Lerner Research Institute, Cleveland Clinic Foundation, Cleveland, Ohio, United States
- Department of Gastroenterology, Hepatology and Nutrition, Digestive Diseases and Surgery Institute, Cleveland Clinic Foundation, Cleveland, Ohio, United States
- Cleveland Clinic Program for Global Translational Inflammatory Bowel Diseases (GRID), Cleveland, Ohio, United States
| |
Collapse
|
5
|
Karout I, Salhab Z, Sherri N, Bitar ER, Borghol AH, Sabra H, Kassem A, Osman O, Alam C, Znait S, Assaf R, Fadlallah S, Jurjus A, Hashash JG, Rahal EA. The Effects of Endosomal Toll-like Receptor Inhibitors in an EBV DNA-Exacerbated Inflammatory Bowel Disease Mouse Model. Viruses 2024; 16:624. [PMID: 38675965 PMCID: PMC11054613 DOI: 10.3390/v16040624] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2024] [Revised: 04/09/2024] [Accepted: 04/12/2024] [Indexed: 04/28/2024] Open
Abstract
Epstein-Barr virus (EBV), a Herpesviridae family member, is associated with an increased risk of autoimmune disease development in the host. We previously demonstrated that EBV DNA elevates levels of the pro-inflammatory cytokine IL-17A and that inhibiting Toll-like receptor (TLR) 3, 7, or 9 reduces its levels. Moreover, this DNA exacerbated colitis in a mouse model of inflammatory bowel disease (IBD). In the study at hand, we examined whether inhibition of TLR3, 7, or 9 alleviates this exacerbation. Mice were fed 1.5% dextran sulfate sodium (DSS) water and administered EBV DNA. Then, they were treated with a TLR3, 7, or 9 inhibitor or left untreated. We also assessed the additive impact of combined inhibition of all three receptors. Mice that received DSS, EBV DNA, and each inhibitor alone, or a combination of inhibitors, showed significant improvement. They also had a decrease in the numbers of the pathogenic colonic IL-17A+IFN-γ+ foci. Inhibition of all three endosomal TLR receptors offered no additive benefit over administering a single inhibitor. Therefore, inhibition of endosomal TLRs reduces EBV DNA exacerbation of mouse colitis, offering a potential approach for managing IBD patients infected with EBV.
Collapse
Affiliation(s)
- Iman Karout
- Department of Experimental Pathology, Immunology, and Microbiology, American University of Beirut, Beirut 1107-2020, Lebanon; (I.K.); (Z.S.); (N.S.); (E.R.B.); (A.H.B.); (H.S.); (A.K.); (O.O.); (C.A.); (S.Z.); (R.A.); (S.F.)
| | - Zahraa Salhab
- Department of Experimental Pathology, Immunology, and Microbiology, American University of Beirut, Beirut 1107-2020, Lebanon; (I.K.); (Z.S.); (N.S.); (E.R.B.); (A.H.B.); (H.S.); (A.K.); (O.O.); (C.A.); (S.Z.); (R.A.); (S.F.)
| | - Nour Sherri
- Department of Experimental Pathology, Immunology, and Microbiology, American University of Beirut, Beirut 1107-2020, Lebanon; (I.K.); (Z.S.); (N.S.); (E.R.B.); (A.H.B.); (H.S.); (A.K.); (O.O.); (C.A.); (S.Z.); (R.A.); (S.F.)
| | - Elio R. Bitar
- Department of Experimental Pathology, Immunology, and Microbiology, American University of Beirut, Beirut 1107-2020, Lebanon; (I.K.); (Z.S.); (N.S.); (E.R.B.); (A.H.B.); (H.S.); (A.K.); (O.O.); (C.A.); (S.Z.); (R.A.); (S.F.)
| | - Abdul Hamid Borghol
- Department of Experimental Pathology, Immunology, and Microbiology, American University of Beirut, Beirut 1107-2020, Lebanon; (I.K.); (Z.S.); (N.S.); (E.R.B.); (A.H.B.); (H.S.); (A.K.); (O.O.); (C.A.); (S.Z.); (R.A.); (S.F.)
| | - Hady Sabra
- Department of Experimental Pathology, Immunology, and Microbiology, American University of Beirut, Beirut 1107-2020, Lebanon; (I.K.); (Z.S.); (N.S.); (E.R.B.); (A.H.B.); (H.S.); (A.K.); (O.O.); (C.A.); (S.Z.); (R.A.); (S.F.)
| | - Aya Kassem
- Department of Experimental Pathology, Immunology, and Microbiology, American University of Beirut, Beirut 1107-2020, Lebanon; (I.K.); (Z.S.); (N.S.); (E.R.B.); (A.H.B.); (H.S.); (A.K.); (O.O.); (C.A.); (S.Z.); (R.A.); (S.F.)
| | - Omar Osman
- Department of Experimental Pathology, Immunology, and Microbiology, American University of Beirut, Beirut 1107-2020, Lebanon; (I.K.); (Z.S.); (N.S.); (E.R.B.); (A.H.B.); (H.S.); (A.K.); (O.O.); (C.A.); (S.Z.); (R.A.); (S.F.)
| | - Charbel Alam
- Department of Experimental Pathology, Immunology, and Microbiology, American University of Beirut, Beirut 1107-2020, Lebanon; (I.K.); (Z.S.); (N.S.); (E.R.B.); (A.H.B.); (H.S.); (A.K.); (O.O.); (C.A.); (S.Z.); (R.A.); (S.F.)
| | - Sabah Znait
- Department of Experimental Pathology, Immunology, and Microbiology, American University of Beirut, Beirut 1107-2020, Lebanon; (I.K.); (Z.S.); (N.S.); (E.R.B.); (A.H.B.); (H.S.); (A.K.); (O.O.); (C.A.); (S.Z.); (R.A.); (S.F.)
| | - Rayan Assaf
- Department of Experimental Pathology, Immunology, and Microbiology, American University of Beirut, Beirut 1107-2020, Lebanon; (I.K.); (Z.S.); (N.S.); (E.R.B.); (A.H.B.); (H.S.); (A.K.); (O.O.); (C.A.); (S.Z.); (R.A.); (S.F.)
| | - Sukayna Fadlallah
- Department of Experimental Pathology, Immunology, and Microbiology, American University of Beirut, Beirut 1107-2020, Lebanon; (I.K.); (Z.S.); (N.S.); (E.R.B.); (A.H.B.); (H.S.); (A.K.); (O.O.); (C.A.); (S.Z.); (R.A.); (S.F.)
| | - Abdo Jurjus
- Department of Anatomy, Cell Biology and Physiological Sciences, American University of Beirut, Beirut 1107-2020, Lebanon;
| | - Jana G. Hashash
- Division of Gastroenterology and Hepatology, Mayo Clinic, Jacksonville, FL 32224, USA;
| | - Elias A. Rahal
- Department of Experimental Pathology, Immunology, and Microbiology, American University of Beirut, Beirut 1107-2020, Lebanon; (I.K.); (Z.S.); (N.S.); (E.R.B.); (A.H.B.); (H.S.); (A.K.); (O.O.); (C.A.); (S.Z.); (R.A.); (S.F.)
| |
Collapse
|
6
|
Rieder F, Mukherjee PK, Massey WJ, Wang Y, Fiocchi C. Fibrosis in IBD: from pathogenesis to therapeutic targets. Gut 2024; 73:854-866. [PMID: 38233198 PMCID: PMC10997492 DOI: 10.1136/gutjnl-2023-329963] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/24/2023] [Accepted: 12/29/2023] [Indexed: 01/19/2024]
Abstract
BACKGROUND Intestinal fibrosis resulting in stricture formation and obstruction in Crohn's disease (CD) and increased wall stiffness leading to symptoms in ulcerative colitis (UC) is among the largest unmet needs in inflammatory bowel disease (IBD). Fibrosis is caused by a multifactorial and complex process involving immune and non-immune cells, their soluble mediators and exposure to luminal contents, such as microbiota and environmental factors. To date, no antifibrotic therapy is available. Some progress has been made in creating consensus definitions and measurements to quantify stricture morphology for clinical practice and trials, but approaches to determine the degree of fibrosis within a stricture are still lacking. OBJECTIVE We herein describe the current state of stricture pathogenesis, measuring tools and clinical trial endpoints development. DESIGN Data presented and discussed in this review derive from the past and recent literature and the authors' own research and experience. RESULTS AND CONCLUSIONS Significant progress has been made in better understanding the pathogenesis of fibrosis, but additional studies and preclinical developments are needed to define specific therapeutic targets.
Collapse
Affiliation(s)
- Florian Rieder
- Department of Inflammation and Immunity, Cleveland Clinic Foundation, Cleveland, Ohio, USA
| | - Pranab K Mukherjee
- Department of Inflammation and Immunity, Cleveland Clinic Foundation, Cleveland, Ohio, USA
| | - William J Massey
- Department of Inflammation and Immunity, Cleveland Clinic Foundation, Cleveland, Ohio, USA
| | - Yan Wang
- Department of Inflammation and Immunity, Cleveland Clinic Foundation, Cleveland, Ohio, USA
| | - Claudio Fiocchi
- Department of Inflammation and Immunity, Cleveland Clinic Foundation, Cleveland, Ohio, USA
| |
Collapse
|
7
|
Du R, Chen S, Han C, He Z, Pei H, Yang Y. M1 intestinal macrophages-derived exosomes promote colitis progression and mucosal barrier injury. Aging (Albany NY) 2024; 16:5703-5710. [PMID: 38535999 PMCID: PMC11006457 DOI: 10.18632/aging.205672] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2023] [Accepted: 12/07/2023] [Indexed: 04/06/2024]
Abstract
AIM This work aimed to investigate the role of M1 intestinal macrophages-derived exosomes (M1-Exo) in colitis and its mechanism. METHODS M1 polarization of intestinal macrophages was induced in vitro, and their exosomes were extracted and identified. Thereafter, the DSS-induced colitis mouse model was built. Each mouse was given intraperitoneal injection of exosomes, and then mouse weight and DAI were dynamically monitored. In addition, the levels of cytokines were detected by ELISA. After treatment with the TLR4 inhibitor Resatorvid, the effects of M1 macrophages-derived exosomes were observed. Besides, the mouse intestinal epithelial cells were cultured in vitro for observing function of M1-Exo. RESULTS M1-exo aggravated the colitis and tissue inflammation in mice, activated the TLR4 signal, and destroyed the mucosal barrier. But M0 macrophages-derived exosomes (M0-Exo) did not have the above effects. Resatorvid treatment antagonized the roles of M1-exo. Moreover, as confirmed by cellular experiments in vitro, M1-exo destroyed mucosal barrier. CONCLUSION M1-exo serve as the pro-inflammatory mediator, which can promote mouse colitis progression by activating TLR4 signal.
Collapse
Affiliation(s)
- Rui Du
- College of Chinese Medicinal Materials, Jilin Agricultural University, Changchun 130118, China
| | - Sihan Chen
- College of Chinese Medicinal Materials, Jilin Agricultural University, Changchun 130118, China
| | - Chenyang Han
- Department of Pharmacy, The Second Affiliated Hospital of Jiaxing University, Jiaxing 314001, China
| | - Zhongmei He
- College of Chinese Medicinal Materials, Jilin Agricultural University, Changchun 130118, China
| | - Hongyan Pei
- College of Chinese Medicinal Materials, Jilin Agricultural University, Changchun 130118, China
| | - Yang Yang
- Department of Cardiology, Shenyang Medical College Affiliated Second Hospital, Shenyang 314005, China
| |
Collapse
|
8
|
Li W, Sheng Y, Wang J, Wu S, Han C. MicroRNA-9a-5p-NOX4 inhibits intestinal inflammatory injury by regulating the M1 polarization of intestinal macrophages. J Biochem Mol Toxicol 2023; 37:e23245. [PMID: 36281492 DOI: 10.1002/jbt.23245] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2022] [Revised: 08/31/2022] [Accepted: 10/12/2022] [Indexed: 11/08/2022]
Abstract
We found that the expression of microRNA (miRNA)-9a-5p decreased in inflammatory bowel diseases (IBD; ulcerative colitis and Crohn's disease). Further, we revealed the effects and mechanisms of miRNA-9a-5p for regulating IBD progression. In C57BL/6N mice, IBD was induced with dextran sodium sulfate (DSS), and the effects of endogenous miRNA-9a-5p were mimicked/antagonized through intraperitoneal injection of miRNA-9a-5p agomir and antagomir. In animal experimentation, agomir could inhibit intestinal inflammation and tissue damage, and reduce the mucosal barrier permeability. Antagomir, on the other hand, could promote barrier damage, whose effect was associated with the M1 macrophage polarization. This study finds that miRNA-9a-5p targets NOX4 to suppress ROS production, which plays an important role in mucosal barrier damage in IBD.
Collapse
Affiliation(s)
- Wenyan Li
- Department of Pharmacy, The Second Affiliated Hospital of Jiaxing University, Jiaxing, China
| | - Yongjia Sheng
- Department of Pharmacy, The Second Affiliated Hospital of Jiaxing University, Jiaxing, China
| | - Jin Wang
- Department of Pharmacy, The Second Affiliated Hospital of Jiaxing University, Jiaxing, China
| | - Shasha Wu
- Department of Pharmacy, The Second Affiliated Hospital of Jiaxing University, Jiaxing, China
| | - Chenyang Han
- Department of Pharmacy, The Second Affiliated Hospital of Jiaxing University, Jiaxing, China
| |
Collapse
|
9
|
Szczepanski HE, Flannigan KL, Mainoli B, Alston L, Baruta GM, Lee JW, Venu VKP, Shearer J, Dufour A, Hirota SA. NR4A1 modulates intestinal smooth muscle cell phenotype and dampens inflammation-associated intestinal remodeling. FASEB J 2022; 36:e22609. [PMID: 36250380 DOI: 10.1096/fj.202101817rr] [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: 12/01/2021] [Revised: 09/16/2022] [Accepted: 09/30/2022] [Indexed: 11/11/2022]
Abstract
Stricture formation is a common complication of Crohn's disease (CD), driven by enhanced deposition of extracellular matrix (ECM) and expansion of the intestinal smooth muscle layers. Nuclear receptor subfamily 4 group A member 1 (NR4A1) is an orphan nuclear receptor that exhibits anti-proliferative effects in smooth muscle cells (SMCs). We hypothesized that NR4A1 regulates intestinal SMC proliferation and muscle thickening in the context of inflammation. Intestinal SMCs isolated from Nr4a1+/+ and Nr4a1-/- littermates were subjected to shotgun proteomic analysis, proliferation, and bioenergetic assays. Proliferation was assessed in the presence and absence of NR4A1 agonists, cytosporone-B (Csn-B) and 6-mercaptopurine (6-MP). In vivo, we compared colonic smooth muscle thickening in Nr4a1+/+ and Nr4a1-/- mice using the chronic dextran sulfate sodium (DSS) model of colitis. Second, SAMP1/YitFc mice (a model of spontaneous ileitis) were treated with Csn-B and small intestinal smooth muscle thickening was assessed. SMCs isolated from Nr4a1-/- mice exhibited increased abundance of proteins related to cell proliferation, metabolism, and ECM production, whereas Nr4a1+/+ SMCs highly expressed proteins related to the regulation of the actin cytoskeleton and contractile processes. SMCs isolated from Nr4a1-/- mice exhibited increased proliferation and alterations in cellular metabolism, whereas activation of NR4A1 attenuated proliferation. In vivo, Nr4a1-/- mice exhibited increased colonic smooth muscle thickness following repeated cycles of DSS. Activating NR4A1 with Csn-B, in the context of established inflammation, reduced ileal smooth muscle thickening in SAMP1/YitFc mice. Targeting NR4A1 may provide a novel approach to regulate intestinal SMC phenotype, limiting excessive proliferation that contributes to stricture development in CD.
Collapse
Affiliation(s)
- Holly E Szczepanski
- Department of Physiology & Pharmacology, University of Calgary, Calgary, Alberta, Canada.,Snyder Institute for Chronic Diseases, University of Calgary, Calgary, Alberta, Canada
| | - Kyle L Flannigan
- Department of Physiology & Pharmacology, University of Calgary, Calgary, Alberta, Canada.,Snyder Institute for Chronic Diseases, University of Calgary, Calgary, Alberta, Canada
| | - Barbara Mainoli
- Department of Physiology & Pharmacology, University of Calgary, Calgary, Alberta, Canada.,Snyder Institute for Chronic Diseases, University of Calgary, Calgary, Alberta, Canada.,Department of Biochemistry & Molecular Biology, University of Calgary, Calgary, Alberta, Canada
| | - Laurie Alston
- Department of Physiology & Pharmacology, University of Calgary, Calgary, Alberta, Canada.,Snyder Institute for Chronic Diseases, University of Calgary, Calgary, Alberta, Canada
| | - Grace M Baruta
- Department of Physiology & Pharmacology, University of Calgary, Calgary, Alberta, Canada.,Snyder Institute for Chronic Diseases, University of Calgary, Calgary, Alberta, Canada
| | - Joshua W Lee
- Department of Physiology & Pharmacology, University of Calgary, Calgary, Alberta, Canada.,Snyder Institute for Chronic Diseases, University of Calgary, Calgary, Alberta, Canada
| | - Vivek Krishna Pulakazhi Venu
- Department of Physiology & Pharmacology, University of Calgary, Calgary, Alberta, Canada.,Snyder Institute for Chronic Diseases, University of Calgary, Calgary, Alberta, Canada
| | - Jane Shearer
- Snyder Institute for Chronic Diseases, University of Calgary, Calgary, Alberta, Canada.,Department of Biochemistry & Molecular Biology, University of Calgary, Calgary, Alberta, Canada.,Alberta Children's Hospital Research Institute, University of Calgary, Calgary, Alberta, Canada
| | - Antoine Dufour
- Department of Physiology & Pharmacology, University of Calgary, Calgary, Alberta, Canada.,Snyder Institute for Chronic Diseases, University of Calgary, Calgary, Alberta, Canada.,Department of Biochemistry & Molecular Biology, University of Calgary, Calgary, Alberta, Canada.,McCaig Institute for Bone and Joint Health, University of Calgary, Calgary, Alberta, Canada
| | - Simon A Hirota
- Department of Physiology & Pharmacology, University of Calgary, Calgary, Alberta, Canada.,Snyder Institute for Chronic Diseases, University of Calgary, Calgary, Alberta, Canada.,Alberta Children's Hospital Research Institute, University of Calgary, Calgary, Alberta, Canada.,Department of Immunology, Microbiology & Infectious Diseases, University of Calgary, Calgary, Alberta, Canada
| |
Collapse
|
10
|
Nonnecke EB, Castillo PA, Akahoshi DT, Goley SM, Bevins CL, Lönnerdal B. Characterization of an intelectin-1 ( Itln1) knockout mouse model. Front Immunol 2022; 13:894649. [PMID: 36072603 PMCID: PMC9441953 DOI: 10.3389/fimmu.2022.894649] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2022] [Accepted: 07/28/2022] [Indexed: 01/26/2023] Open
Abstract
Intelectins are carbohydrate-binding proteins implicated in innate immunity and highly conserved across chordate evolution, including both ascidians and humans. Human intelectin-1 (ITLN1) is highly abundant within the intestinal mucosa and binds microbial but not host glycans. Genome-wide association studies identified SNPs in ITLN1 that are linked to susceptibility for Crohn's disease. Moreover, ITLN1 has been implicated in the pathophysiology of obesity and associated metabolic disease. To gain insight on biological activities of human ITLN1 in vivo, we developed a C57BL/6 mouse model genetically targeting the gene encoding the functional mouse ortholog. In wild-type C57BL/6 mice, both mRNA and protein analysis showed high expression of Itln1 in the small intestine, but manifold lower levels in colon and other extraintestinal tissues. Whereas intestinal expression of human ITLN1 localizes to goblet cells, our data confirm that mouse Itln1 is expressed in Paneth cells. Compared to wild-type littermate controls, mice homozygous for the Itln1 hypomorphic trapping allele had reduced expression levels of Itln1 expression (~10,000-fold). The knockout mice exhibited increased susceptibility in an acute model of experimentally induced colitis with 2% w/v dextran sulfate sodium (DSS). In a model of chronic colitis using a lower dose of DSS (1.5% w/v), which enabled a detailed view of disease activity across a protracted period, no differences were observed in body weight, fecal texture, hemoccult scores, food/water intake, or colon length at necropsy, but there was a statistically significant genotype over time effect for the combined fecal scores of disease activity. In model of diet-induced obesity, using two western-style diets, which varied in amounts of sugar (as sucrose) and saturated fat (as lard), mice with Itln1 expression ablated showed no increased susceptibility, in terms of weight gain, food intake, plasma markers of obesity compared to wildtype littermates. While the mouse genetic knockout model for Itln1 holds promise for elucidating physiological function(s) for mammalian intelectins, results reported here suggest that Itln1, a Paneth cell product in C57BL/6 mice, likely plays a minor role in the pathophysiology of chemically induced colitis or diet-induced obesity.
Collapse
Affiliation(s)
- Eric B. Nonnecke
- Department of Microbiology and Immunology, School of Medicine, University of California, Davis, Davis, CA, United States,*Correspondence: Eric B. Nonnecke, ; Charles L. Bevins,
| | - Patricia A. Castillo
- Department of Microbiology and Immunology, School of Medicine, University of California, Davis, Davis, CA, United States
| | - Douglas T. Akahoshi
- Department of Microbiology and Immunology, School of Medicine, University of California, Davis, Davis, CA, United States
| | - Stephanie M. Goley
- Department of Nutrition, University of California, Davis, Davis, CA, United States
| | - Charles L. Bevins
- Department of Microbiology and Immunology, School of Medicine, University of California, Davis, Davis, CA, United States,*Correspondence: Eric B. Nonnecke, ; Charles L. Bevins,
| | - Bo Lönnerdal
- Department of Nutrition, University of California, Davis, Davis, CA, United States
| |
Collapse
|
11
|
Xie Y, Fontenot L, Chupina Estrada A, Nelson B, Wang J, Shih DQ, Ho W, Mattai SA, Rieder F, Jensen DD, Bunnett NW, Koon HW. Elafin Reverses Intestinal Fibrosis by Inhibiting Cathepsin S-Mediated Protease-Activated Receptor 2. Cell Mol Gastroenterol Hepatol 2022; 14:841-876. [PMID: 35840034 PMCID: PMC9425040 DOI: 10.1016/j.jcmgh.2022.06.011] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/23/2021] [Revised: 06/29/2022] [Accepted: 06/30/2022] [Indexed: 12/10/2022]
Abstract
BACKGROUND & AIMS More than half of Crohn's disease patients develop intestinal fibrosis-induced intestinal strictures. Elafin is a human protease inhibitor that is down-regulated in the stricturing intestine of Crohn's disease patients. We investigated the efficacy of elafin in reversing intestinal fibrosis and elucidated its mechanism of action. METHODS We developed a new method to mimic a stricturing Crohn's disease environment and induce fibrogenesis using stricturing Crohn's disease patient-derived serum exosomes to condition fresh human intestinal tissues and primary stricturing Crohn's disease patient-derived intestinal fibroblasts. Three mouse models of intestinal fibrosis, including SAMP1/YitFc mice, Salmonella-infected mice, and trinitrobenzene sulfonic acid-treated mice, were also studied. Elafin-Eudragit FS30D formulation and elafin-overexpressing construct and lentivirus were used. RESULTS Elafin reversed collagen synthesis in human intestinal tissues and fibroblasts pretreated with Crohn's disease patient-derived serum exosomes. Proteome arrays identified cathepsin S as a novel fibroblast-derived pro-fibrogenic protease. Elafin directly suppressed cathepsin S activity to inhibit protease-activated receptor 2 activity and Zinc finger E-box-binding homeobox 1 expression, leading to reduced collagen expression in intestinal fibroblasts. Elafin overexpression reversed ileal fibrosis in SAMP1/YitFc mice, cecal fibrosis in Salmonella-infected mice, and colonic fibrosis in trinitrobenzene sulfonic acid-treated mice. Cathepsin S, protease-activated receptor 2 agonist, and zinc finger E-box-binding homeobox 1 overexpression abolished the anti-fibrogenic effect of elafin in fibroblasts and all 3 mouse models of intestinal fibrosis. Oral elafin-Eudragit FS30D treatment abolished colonic fibrosis in trinitrobenzene sulfonic acid-treated mice. CONCLUSIONS Elafin suppresses collagen synthesis in intestinal fibroblasts via cathepsin S-dependent protease-activated receptor 2 inhibition and decreases zinc finger E-box-binding homeobox 1 expression. The reduced collagen synthesis leads to the reversal of intestinal fibrosis. Thus, modified elafin may be a therapeutic approach for intestinal fibrosis.
Collapse
Affiliation(s)
- Ying Xie
- Vatche and Tamar Manoukian Division of Digestive Diseases, David Geffen School of Medicine at the University of California Los Angeles, Los Angeles, California,Department of Gastroenterology, The First Hospital of China Medical University, Shenyang City, Liaoning Province, China
| | - Lindsey Fontenot
- Vatche and Tamar Manoukian Division of Digestive Diseases, David Geffen School of Medicine at the University of California Los Angeles, Los Angeles, California
| | - Andrea Chupina Estrada
- Vatche and Tamar Manoukian Division of Digestive Diseases, David Geffen School of Medicine at the University of California Los Angeles, Los Angeles, California
| | - Becca Nelson
- Vatche and Tamar Manoukian Division of Digestive Diseases, David Geffen School of Medicine at the University of California Los Angeles, Los Angeles, California
| | - Jiani Wang
- Vatche and Tamar Manoukian Division of Digestive Diseases, David Geffen School of Medicine at the University of California Los Angeles, Los Angeles, California,Department of Gastroenterology, The First Hospital of China Medical University, Shenyang City, Liaoning Province, China
| | - David Q. Shih
- F. Widjaja Foundation, Inflammatory Bowel & Immunobiology Research Institute, Cedars-Sinai Medical Center, Los Angeles, California
| | - Wendy Ho
- Vatche and Tamar Manoukian Division of Digestive Diseases, David Geffen School of Medicine at the University of California Los Angeles, Los Angeles, California
| | - S. Anjani Mattai
- Department of Medicine, David Geffen School of Medicine at the University of California Los Angeles, Los Angeles, California
| | - Florian Rieder
- Department of Gastroenterology, Hepatology, and Nutrition, Digestive Diseases and Surgery Institute, Cleveland Clinic Foundation, Cleveland, Ohio
| | - Dane D. Jensen
- Bluestone Center for Clinical Research, New York University College of Dentistry, New York, New York
| | - Nigel W. Bunnett
- Department of Molecular Pathobiology, Department of Neuroscience and Physiology, Neuroscience Institute, New York University, New York, New York
| | - Hon Wai Koon
- Vatche and Tamar Manoukian Division of Digestive Diseases, David Geffen School of Medicine at the University of California Los Angeles, Los Angeles, California.
| |
Collapse
|
12
|
Garabatos N, Santamaria P. Gut Microbial Antigenic Mimicry in Autoimmunity. Front Immunol 2022; 13:873607. [PMID: 35572569 PMCID: PMC9094498 DOI: 10.3389/fimmu.2022.873607] [Citation(s) in RCA: 37] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2022] [Accepted: 03/14/2022] [Indexed: 12/12/2022] Open
Abstract
The gut microbiota plays a major role in the developmental biology and homeostasis of cells belonging to the adaptive and innate arms of the immune system. Alterations in its composition, which are known to be regulated by both genetic and environmental factors, can either promote or suppress the pathogenic processes underlying the development of various autoimmune diseases, including inflammatory bowel disease, multiple sclerosis, systemic lupus erythematosus, type 1 diabetes and rheumatoid arthritis, to just name a few. Cross-recognition of gut microbial antigens by autoreactive T cells as well as gut microbe-driven alterations in the activation and homeostasis of effector and regulatory T cells have been implicated in this process. Here, we summarize our current understanding of the positive and negative associations between alterations in the composition of the gut microbiota and the development of various autoimmune disorders, with a special emphasis on antigenic mimicry.
Collapse
Affiliation(s)
- Nahir Garabatos
- Institut D'Investigacions Biomèdiques August Pi i Sunyer, Barcelona, Spain
| | - Pere Santamaria
- Institut D'Investigacions Biomèdiques August Pi i Sunyer, Barcelona, Spain
- Julia McFarlane Diabetes Research Centre (JMDRC), Snyder Institute for Chronic Diseases, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
- Department of Microbiology, Immunology and Infectious Diseases, Snyder Institute for Chronic Diseases, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| |
Collapse
|
13
|
Harnessing murine models of Crohn's disease ileitis to advance concepts of pathophysiology and treatment. Mucosal Immunol 2022; 15:10-26. [PMID: 34316007 DOI: 10.1038/s41385-021-00433-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2021] [Revised: 07/09/2021] [Accepted: 07/12/2021] [Indexed: 02/04/2023]
Abstract
Crohn's disease (CD) and ulcerative colitis (UC) are both characterized by chronic inflammation and severe dysfunction of the gastrointestinal tract. These two forms of inflammatory bowel disease (IBD) represent distinct clinical disorders with diverse driving mechanisms; however, this divergence is not reflected in currently approved therapeutics that commonly target general proinflammatory pathways. A compelling need therefore remains to understand factors that differentiate the topology and the distinct clinical manifestations of CD versus UC, in order to develop more effective and specialized therapies. Animal models provide valuable platforms for studying IBD heterogeneity and deciphering disease-specific mechanisms. Both the established and the newly developed ileitis mouse models are characterized by various disease initiating mechanisms and diverse phenotypic outcomes that reflect the complexity of human CD-ileitis. Microbial dysbiosis, destruction of epithelial barrier integrity, immune cell deregulation, as well as the recently described genome instability and stromal cell activation have all been proposed as the triggering factors for the development of ileitis-associated pathology. In this review, we aim to critically evaluate the mechanistic underpinnings of murine models of CD-ileitis, discuss their phenotypic similarities to human disease, and envisage their further exploitation for the development of novel targeted and personalized therapeutics.
Collapse
|
14
|
An Update of Research Animal Models of Inflammatory Bowel Disease. ScientificWorldJournal 2021; 2021:7479540. [PMID: 34938152 PMCID: PMC8687830 DOI: 10.1155/2021/7479540] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2021] [Accepted: 11/28/2021] [Indexed: 12/14/2022] Open
Abstract
Inflammatory bowel disease (IBD) is a group of chronic disorders that includes two main disease forms, Crohn's disease, and ulcerative colitis. The understanding of the intestinal inflammation occurring in IBD has been immeasurably advanced by the development of the now numerous murine models of intestinal inflammation. The usefulness of this research tool in IBD arises from a convergence of underlying genetic susceptibility, immune system dysfunction, environmental factors, and shifts in gut microbiota. Due to the multifactorial feature of these diseases, different animal models have been used to investigate the underlying mechanisms and develop potential therapeutic strategies. The results of preclinical efficacy studies often inform the progression of therapeutic strategies. This review describes the distinct feature and limitations of each murine IBD model and discusses the previous and current lessons from the IBD models.
Collapse
|
15
|
Verma A, Sharda S, Rathi B, Somvanshi P, Pandey BD. Elucidating potential molecular signatures through host-microbe interactions for reactive arthritis and inflammatory bowel disease using combinatorial approach. Sci Rep 2020; 10:15131. [PMID: 32934294 PMCID: PMC7492238 DOI: 10.1038/s41598-020-71674-8] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2019] [Accepted: 07/06/2020] [Indexed: 02/08/2023] Open
Abstract
Reactive Arthritis (ReA), a rare seronegative inflammatory arthritis, lacks exquisite classification under rheumatic autoimmunity. ReA is solely established using differential clinical diagnosis of the patient cohorts, where pathogenic triggers linked to enteric and urogenital microorganisms e.g. Salmonella, Shigella, Yersinia, Campylobacter, Chlamydia have been reported. Inflammatory Bowel Disease (IBD), an idiopathic enteric disorder co-evolved and attuned to present gut microbiome dysbiosis, can be correlated to the genesis of enteropathic arthropathies like ReA. Gut microbes symbolically modulate immune system homeostasis and are elementary for varied disease patterns in autoimmune disorders. The gut-microbiota axis structured on the core host-microbe interactions execute an imperative role in discerning the etiopathogenesis of ReA and IBD. This study predicts the molecular signatures for ReA with co-evolved IBD through the enveloped host-microbe interactions and microbe-microbe 'interspecies communication', using synonymous gene expression data for selective microbes. We have utilized a combinatorial approach that have concomitant in-silico work-pipeline and experimental validation to corroborate the findings. In-silico analysis involving text mining, metabolic network reconstruction, simulation, filtering, host-microbe interaction, docking and molecular mimicry studies results in robust drug target/s and biomarker/s for co-evolved IBD and ReA. Cross validation of the target/s or biomarker/s was done by targeted gene expression analysis following a non-probabilistic convenience sampling. Studies were performed to substantiate the host-microbe disease network consisting of protein-marker-symptom/disease-pathway-drug associations resulting in possible identification of vital drug targets, biomarkers, pathways and inhibitors for IBD and ReA.Our study identified Na(+)/H(+) anti-porter (NHAA) and Kynureninase (KYNU) to be robust early and essential host-microbe interacting targets for IBD co-evolved ReA. Other vital host-microbe interacting genes, proteins, pathways and drugs include Adenosine Deaminase (ADA), Superoxide Dismutase 2 (SOD2), Catalase (CAT), Angiotensin I Converting Enzyme (ACE), carbon metabolism (folate biosynthesis) and methotrexate. These can serve as potential prognostic/theranostic biomarkers and signatures that can be extrapolated to stratify ReA and related autoimmunity patient cohorts for further pilot studies.
Collapse
Affiliation(s)
- Anukriti Verma
- Amity Institute of Biotechnology, J-3 Block, Amity University Campus, Sector-125, Noida, UP, 201313, India
| | - Shivani Sharda
- Amity Institute of Biotechnology, J-3 Block, Amity University Campus, Sector-125, Noida, UP, 201313, India.
| | - Bhawna Rathi
- Amity Institute of Biotechnology, J-3 Block, Amity University Campus, Sector-125, Noida, UP, 201313, India
| | - Pallavi Somvanshi
- Department of Biotechnology, TERI School of Advanced Studies, 10, Institutional Area, Vasant Kunj, New Delhi, 110070, India
| | - Bimlesh Dhar Pandey
- Fortis Hospital, B-22, Sector 62, Gautam Buddh Nagar, Noida, Uttar Pradesh, 201301, India
| |
Collapse
|
16
|
Hu WC. A Framework of All Discovered Immunological Pathways and Their Roles for Four Specific Types of Pathogens and Hypersensitivities. Front Immunol 2020; 11:1992. [PMID: 32849663 PMCID: PMC7426516 DOI: 10.3389/fimmu.2020.01992] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2020] [Accepted: 07/23/2020] [Indexed: 01/05/2023] Open
Affiliation(s)
- Wan-Chung Hu
- Department of Clinical Pathology, Taipei Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, New Taipei City, Taiwan
| |
Collapse
|
17
|
Buttó LF, Pelletier A, More SK, Zhao N, Osme A, Hager CL, Ghannoum MA, Sekaly RP, Cominelli F, Dave M. Intestinal Stem Cell Niche Defects Result in Impaired 3D Organoid Formation in Mouse Models of Crohn's Disease-like Ileitis. Stem Cell Reports 2020; 15:389-407. [PMID: 32679063 PMCID: PMC7419719 DOI: 10.1016/j.stemcr.2020.06.017] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2019] [Revised: 06/16/2020] [Accepted: 06/18/2020] [Indexed: 02/08/2023] Open
Abstract
Intestinal epithelial barrier dysfunction is a risk factor in the pathogenesis of Crohn’s disease (CD); however, no corrective FDA-approved therapies exist. We used an enteroid (EnO)-based system in two murine models of experimental CD, SAMP1/YitFc (SAMP) and TNFΔARE/+ (TNF). While severely inflamed SAMP mice do not generate EnOs, “inflammation-free” SAMP mice form EnO structures with impaired morphology and reduced intestinal stem cell (ISC) and Paneth cell viability. We validated these findings in TNF mice concluding that inflammation in intestinal tissues impedes EnO generation and suppressing inflammation by steroid administration partially rescues impaired formation in SAMP mice. We generated the first high-resolution transcriptional profile of the SAMP ISC niche demonstrating that alterations in multiple key pathways contribute to niche defect and targeting them may partially rescue the phenotype. Furthermore, we correlated the defects in formation and the rescue of EnO formation to reduced viability of ISCs and Paneth cells. Enteroid (EnO) formation is impaired in inflammation-free SAMP mice SAMP EnOs maintain impaired functions ex vivo recapitulating epithelial CD defect Inflammation impedes EnO formation, which is partially restored by steroid treatment Reduced number of viable intestinal stem and Paneth cells correlate with EnO defect
Collapse
Affiliation(s)
- Ludovica F Buttó
- Department of Medicine, Case Western Reserve University School of Medicine, University Hospitals Cleveland Medical Center, 11100 Euclid Avenue, Cleveland, OH 44106-5066, USA; Department of Digestive Health Research Institute, Case Western Reserve University School of Medicine, Cleveland, OH 44106, USA
| | - Adam Pelletier
- Department of Pathology, Case Western Reserve University School of Medicine, Cleveland, OH 44106, USA
| | - Shyam K More
- Division of Gastroenterology & Hepatology University of California Davis, School of Medicine, Institute for Regenerative Cures, 2921 Stockton Boulevard, Suite 1615, Sacramento, CA 95817, USA
| | - Nan Zhao
- Department of Medicine, Case Western Reserve University School of Medicine, University Hospitals Cleveland Medical Center, 11100 Euclid Avenue, Cleveland, OH 44106-5066, USA
| | - Abdullah Osme
- Department of Medicine, Case Western Reserve University School of Medicine, University Hospitals Cleveland Medical Center, 11100 Euclid Avenue, Cleveland, OH 44106-5066, USA; Department of Pathology, Case Western Reserve University School of Medicine, Cleveland, OH 44106, USA
| | - Christopher L Hager
- Center for Medical Mycology, Case Western Reserve University School of Medicine, Cleveland, OH 44106, USA
| | - Mahmoud A Ghannoum
- Center for Medical Mycology, Case Western Reserve University School of Medicine, Cleveland, OH 44106, USA
| | - Rafick-Pierre Sekaly
- Department of Pathology, Case Western Reserve University School of Medicine, Cleveland, OH 44106, USA
| | - Fabio Cominelli
- Department of Medicine, Case Western Reserve University School of Medicine, University Hospitals Cleveland Medical Center, 11100 Euclid Avenue, Cleveland, OH 44106-5066, USA; Department of Pathology, Case Western Reserve University School of Medicine, Cleveland, OH 44106, USA; Department of Digestive Health Research Institute, Case Western Reserve University School of Medicine, Cleveland, OH 44106, USA.
| | - Maneesh Dave
- Department of Medicine, Case Western Reserve University School of Medicine, University Hospitals Cleveland Medical Center, 11100 Euclid Avenue, Cleveland, OH 44106-5066, USA; Department of Digestive Health Research Institute, Case Western Reserve University School of Medicine, Cleveland, OH 44106, USA; Division of Gastroenterology & Hepatology University of California Davis, School of Medicine, Institute for Regenerative Cures, 2921 Stockton Boulevard, Suite 1615, Sacramento, CA 95817, USA.
| |
Collapse
|
18
|
Mizoguchi E, Low D, Ezaki Y, Okada T. Recent updates on the basic mechanisms and pathogenesis of inflammatory bowel diseases in experimental animal models. Intest Res 2020; 18:151-167. [PMID: 32326669 PMCID: PMC7206339 DOI: 10.5217/ir.2019.09154] [Citation(s) in RCA: 72] [Impact Index Per Article: 14.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/21/2019] [Accepted: 02/10/2020] [Indexed: 12/19/2022] Open
Abstract
The specific pathogenesis underlining inflammatory bowel disease (IBD) is very complicated, and it is further more difficult to clearly explain the pathophysiology of 2 major forms of IBD, Crohn’s disease (CD) and ulcerative colitis (UC), and both disorders affect individuals throughout life. Despite every extensive effort, the interplay among genetic factors, immunological factors, environmental factors and intestinal microbes is still completely unrevealed. Animal models are indispensable to find out mechanistic details that will facilitate better preclinical setting to target specific components involved in the pathogenesis of IBD. Based on many recent reports, dysbiosis of the commensal microbiota is implicated in the pathogenesis of several diseases, not only IBD but also colon cancer, obesity, psoriasis as well as allergic disorders, in both human and animal models. Advanced technologies including cell-specific and inducible knockout systems, which are recently employed to mouse IBD models, have further enhanced the ability of developing new therapeutic strategies for IBD. Furthermore, data from these mouse models highlight the critical involvement of dysregulated immune responses and impaired colonic epithelial defense system in the pathogenesis of IBD. In this review, we will explain from the history of animal models of IBD to the recent reports of the latest compounds, therapeutic strategies, and approaches tested on IBD animal models.
Collapse
Affiliation(s)
- Emiko Mizoguchi
- Department of Immunology, Kurume University School of Medicine, Kurume, Japan.,Department of Molecular Microbiology and Immunology, Brown University Warren Alpert Medical School, Providence, RI, USA
| | - Daren Low
- Crohn's & Colitis Society of Singapore, Singapore
| | - Yui Ezaki
- Department of Immunology, Kurume University School of Medicine, Kurume, Japan
| | - Toshiyuki Okada
- Department of Immunology, Kurume University School of Medicine, Kurume, Japan
| |
Collapse
|
19
|
Singh S, Arthur S, Sundaram U. Mechanisms of Regulation of Transporters of Amino Acid Absorption in Inflammatory Bowel Diseases. Compr Physiol 2020; 10:673-686. [PMID: 32163200 DOI: 10.1002/cphy.c190016] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Intestinal absorption of dietary amino acids/peptides is essential for protein homeostasis, which in turn is crucial for maintaining health as well as restoration of health from significant diseases. Dietary amino acids/peptides are absorbed by unique transporter processes present in the brush border membrane of absorptive villus cells, which line the entire length of the intestine. To date, the only nutrient absorptive system described in the secretory crypt cells in the mammalian intestine is the one that absorbs the amino acid glutamine. Majority of the amino acid transporters are sodium dependent and therefore require basolateral membrane Na-K-ATPase to maintain an efficient transcellular Na gradient for their activity. These transport processes are tightly regulated by various cellular and molecular mechanisms that facilitate their optimal activity during normal physiological processes. Malabsorption of amino acids, recently described in pathophysiological states such as in inflammatory bowel disease (IBD), is undoubtedly responsible for the debilitating symptoms of IBD such as malnutrition, weight loss and ultimately a failure to thrive. Also recently, in vivo models of IBD and in vitro studies have demonstrated that specific immune-inflammatory mediators/pathways regulate specific amino acid transporters. This provides possibilities to derive novel nutrition and immune-based treatment options for conditions such as IBD. © 2020 American Physiological Society. Compr Physiol 10:673-686, 2020.
Collapse
Affiliation(s)
- Soudamani Singh
- Department of Clinical and Translational Sciences and Appalachian Clinical and Translational Science Institute, Joan C. Edwards School of Medicine, Marshall University, Huntington, West Virginia, USA
| | - Subha Arthur
- Department of Clinical and Translational Sciences and Appalachian Clinical and Translational Science Institute, Joan C. Edwards School of Medicine, Marshall University, Huntington, West Virginia, USA
| | - Uma Sundaram
- Department of Clinical and Translational Sciences and Appalachian Clinical and Translational Science Institute, Joan C. Edwards School of Medicine, Marshall University, Huntington, West Virginia, USA
| |
Collapse
|
20
|
Toribio‐Fernández R, Herrero‐Fernandez B, Zorita V, López JA, Vázquez J, Criado G, Pablos JL, Collas P, Sánchez‐Madrid F, Andrés V, Gonzalez‐Granado JM. Lamin A/C deficiency in CD4
+
T‐cells enhances regulatory T‐cells and prevents inflammatory bowel disease. J Pathol 2019; 249:509-522. [DOI: 10.1002/path.5332] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2019] [Revised: 07/15/2019] [Accepted: 07/29/2019] [Indexed: 12/19/2022]
Affiliation(s)
| | | | - Virginia Zorita
- Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC) Madrid Spain
| | - Juan A López
- Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC) Madrid Spain
- CIBER de Enfermedades Cardiovasculares Madrid Spain
| | - Jesús Vázquez
- Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC) Madrid Spain
- CIBER de Enfermedades Cardiovasculares Madrid Spain
| | - Gabriel Criado
- Instituto de Investigación Hospital 12 de Octubre (imas12) Madrid Spain
| | - Jose L Pablos
- Instituto de Investigación Hospital 12 de Octubre (imas12) Madrid Spain
| | - Philippe Collas
- Institute of Basic Medical SciencesUniversity of Oslo Oslo Norway
| | - Francisco Sánchez‐Madrid
- Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC) Madrid Spain
- CIBER de Enfermedades Cardiovasculares Madrid Spain
- Servicio de Inmunología, Hospital de la PrincesaInstituto de Investigación Sanitaria La Princesa (IIS Princesa) Madrid Spain
| | - Vicente Andrés
- Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC) Madrid Spain
- CIBER de Enfermedades Cardiovasculares Madrid Spain
| | - Jose M Gonzalez‐Granado
- Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC) Madrid Spain
- Instituto de Investigación Hospital 12 de Octubre (imas12) Madrid Spain
- CIBER de Enfermedades Cardiovasculares Madrid Spain
- Departamento de Fisiología, Facultad de MedicinaUniversidad Autónoma de Madrid (UAM) Madrid Spain
| |
Collapse
|
21
|
Abstract
Inflammatory bowel disease (IBD) defines a spectrum of complex disorders. Understanding how environmental risk factors, alterations of the intestinal microbiota, and polygenetic and epigenetic susceptibility impact on immune pathways is key for developing targeted therapies. Mechanistic understanding of polygenic IBD is complemented by Mendelian disorders that present with IBD, pharmacological interventions that cause colitis, autoimmunity, and multiple animal models. Collectively, this multifactorial pathogenesis supports a concept of immune checkpoints that control microbial-host interactions in the gut by modulating innate and adaptive immunity, as well as epithelial and mesenchymal cell responses. In addition to classical immunosuppressive strategies, we discuss how resetting the microbiota and restoring innate immune responses, in particular autophagy and epithelial barrier function, might be key for maintaining remission or preventing IBD. Targeting checkpoints in genetically stratified subgroups of patients with Mendelian disorder-associated IBD increasingly directs treatment strategies as part of personalized medicine.
Collapse
Affiliation(s)
- Holm H Uhlig
- Department of Pediatrics, University of Oxford, Oxford OX3 9DU, United Kingdom; .,Translational Gastroenterology Unit, University of Oxford, Oxford OX3 9DU, United Kingdom
| | - Fiona Powrie
- Kennedy Institute of Rheumatology, University of Oxford, Oxford OX3 7FY, United Kingdom; .,Translational Gastroenterology Unit, University of Oxford, Oxford OX3 9DU, United Kingdom
| |
Collapse
|
22
|
Dillon A, Lo DD. M Cells: Intelligent Engineering of Mucosal Immune Surveillance. Front Immunol 2019; 10:1499. [PMID: 31312204 PMCID: PMC6614372 DOI: 10.3389/fimmu.2019.01499] [Citation(s) in RCA: 112] [Impact Index Per Article: 18.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2019] [Accepted: 06/14/2019] [Indexed: 12/25/2022] Open
Abstract
M cells are specialized intestinal epithelial cells that provide the main machinery for sampling luminal microbes for mucosal immune surveillance. M cells are usually found in the epithelium overlying organized mucosal lymphoid tissues, but studies have identified multiple distinct lineages of M cells that are produced under different conditions, including intestinal inflammation. Among these lineages there is a common morphology that helps explain the efficiency of M cells in capturing luminal bacteria and viruses; in addition, M cells recruit novel cellular mechanisms to transport the particles across the mucosal barrier into the lamina propria, a process known as transcytosis. These specializations used by M cells point to a novel engineering of cellular machinery to selectively capture and transport microbial particles of interest. Because of the ability of M cells to effectively violate the mucosal barrier, the circumstances of M cell induction have important consequences. Normal immune surveillance insures that transcytosed bacteria are captured by underlying myeloid/dendritic cells; in contrast, inflammation can induce development of new M cells not accompanied by organized lymphoid tissues, resulting in bacterial transcytosis with the potential to amplify inflammatory disease. In this review, we will discuss our own perspectives on the life history of M cells and also raise a few questions regarding unique aspects of their biology among epithelia.
Collapse
Affiliation(s)
- Andrea Dillon
- Division of Biomedical Sciences, School of Medicine, University of California, Riverside, Riverside, CA, United States
| | - David D Lo
- Division of Biomedical Sciences, School of Medicine, University of California, Riverside, Riverside, CA, United States
| |
Collapse
|
23
|
Li J, Zuo L, Tian Y, He Y, Zhang Z, Guo P, Ge Y, Hu J. Spontaneous colitis in IL-10-deficient mice was ameliorated via inhibiting glutaminase1. J Cell Mol Med 2019; 23:5632-5641. [PMID: 31211512 PMCID: PMC6653008 DOI: 10.1111/jcmm.14471] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2019] [Revised: 04/09/2019] [Accepted: 04/26/2019] [Indexed: 12/30/2022] Open
Abstract
Immunity imbalance and barrier damage in the intestinal mucosa are the main pathogenic factors of Crohn's disease (CD). Bis‐2‐(5‐phenylacetamido‐1,2,4‐thiadiazol‐2‐yl) ethyl sulfide (BPTES) is a glutaminase 1 (Gls1) inhibitor with the dual functions of increasing glutamine levels and immune regulation. In this study, we focused on the role of BPTES in CD‐like enteritis and the possible mechanisms. We found that Gls1 expression was significantly increased in CD intestinal tissue compared with control tissue. Bis‐2‐(5‐phenylacetamido‐1,2,4‐thiadiazol‐2‐yl) ethyl sulfide treatment significantly ameliorated chronic colitis in the IL‐10−/−, as manifested by decreased disease activity index, body weight change, histological inflammatory degree and inflammatory cytokine expression. Bis‐2‐(5‐phenylacetamido‐1,2,4‐thiadiazol‐2‐yl) ethyl sulfide treatment exerted protective effects on CD that were associated with the maintenance of intestinal barrier integrity and the Th/Treg balance. Bis‐2‐(5‐phenylacetamido‐1,2,4‐thiadiazol‐2‐yl) ethyl sulfide treatment may act in part through TCR‐mediated mammalian target of rapamycin complex 1 (mTORC1) signalling activation. In conclusion, inhibition of Gls1 expression attenuated chronic colitis by maintaining intestinal barrier integrity and the Th/Treg balance, thereby ameliorating CD‐like colitis.
Collapse
Affiliation(s)
- Jing Li
- Department of Clinical Laboratory, First Affiliated Hospital of Bengbu Medical College, Bengbu, Anhui, China.,Anhui Key Laboratory of Tissue Transplantation, Bengbu Medical College, Bengbu, China
| | - Lugen Zuo
- Anhui Key Laboratory of Tissue Transplantation, Bengbu Medical College, Bengbu, China.,Department of Gastrointestinal Surgery, First Affiliated Hospital of Bengbu Medical College, Bengbu, Anhui, China
| | - Yun Tian
- Department of Oncology, Shanghai Dermatology Hospital, Tongji University, Shanghai, China.,Tongji University Cancer Center, The Shanghai Tenth People's Hospital, Tongji University, Shanghai, China
| | - Yifan He
- Clinical Medicine of Bengbu Medical College, Bengbu, Anhui, China
| | - Zhichao Zhang
- Clinical Medicine of Bengbu Medical College, Bengbu, Anhui, China
| | - Pu Guo
- Department of Clinical Laboratory, First Affiliated Hospital of Bengbu Medical College, Bengbu, Anhui, China
| | - Yuanyuan Ge
- Department of Colorectal Surgery, The Third Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
| | - Jianguo Hu
- Department of Clinical Laboratory, First Affiliated Hospital of Bengbu Medical College, Bengbu, Anhui, China.,Department of Gastrointestinal Surgery, First Affiliated Hospital of Bengbu Medical College, Bengbu, Anhui, China
| |
Collapse
|
24
|
Di Martino L, Osme A, Kossak-Gupta S, Pizarro TT, Cominelli F. TWEAK/Fn14 Is Overexpressed in Crohn's Disease and Mediates Experimental Ileitis by Regulating Critical Innate and Adaptive Immune Pathways. Cell Mol Gastroenterol Hepatol 2019; 8:427-446. [PMID: 31181286 PMCID: PMC6718944 DOI: 10.1016/j.jcmgh.2019.05.009] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/14/2018] [Revised: 05/24/2019] [Accepted: 05/28/2019] [Indexed: 12/14/2022]
Abstract
BACKGROUND & AIMS Crohn's disease (CD) is a debilitating inflammatory disorder that affects more than 1.6 million people in North America alone. Members of the tumor necrosis factor superfamily are key regulators of intestinal inflammation; specifically, tumor necrosis factor-like weak inducer of apoptosis (TWEAK) and its receptor, fibroblast growth factor-inducible 14 (Fn14), are involved in normal and pathologic tissue remodeling. Our aim was to determine the role of TWEAK/Fn14 in CD and a murine model of CD-like ileitis (ie, SAMP1/YitFc [SAMP] strain). METHODS SAMP mice deficient in Fn14 (SAMP × Fn14-/-) were developed and a detailed time-course study was performed evaluating ileal tissues by histology and stereomicroscopy, as well as quantitative polymerase chain reaction and NanoString technology (Seattle, WA). Reciprocal bone marrow chimeras were generated to assess the relevance of Fn14 in hematopoietic vs nonhematopoietic compartments. Surgically resected intestinal tissues and mucosal biopsy specimens from patients with CD, ulcerative colitis, and healthy controls were analyzed for the expression of TWEAK/Fn14 by quantitative polymerase chain reaction, Western blot, immunohistochemistry, and immunofluorescence. RESULTS SAMP × Fn14-/- showed a marked decrease in ileitis severity at 20 weeks of age compared with SAMP WT controls. Bone marrow chimeras showed that Fn14 was required in both hematopoietic and nonhematopoietic compartments for ileitis to develop. Transcriptome data showed multiple cellular pathways regulated by Fn14 signaling. Finally, increased expression of TWEAK and Fn14 was observed in tissue lesions from CD patients compared with ulcerative colitis and healthy controls. CONCLUSIONS TWEAK/Fn14 are up-regulated in CD, and also mediate experimental CD-like ileitis, by regulation of multiple innate and adaptive cellular pathways. Therefore, TWEAK/Fn14 may represent a novel therapeutic target for the treatment of small intestinal inflammation in CD.
Collapse
Affiliation(s)
- Luca Di Martino
- Division of Gastroenterology and Liver Disease, Case Western University School of Medicine, Cleveland, Ohio; Department of Medicine, Case Western University School of Medicine, Cleveland, Ohio
| | - Abdullah Osme
- Division of Gastroenterology and Liver Disease, Case Western University School of Medicine, Cleveland, Ohio; Department of Medicine, Case Western University School of Medicine, Cleveland, Ohio
| | - Sarah Kossak-Gupta
- Division of Gastroenterology and Liver Disease, Case Western University School of Medicine, Cleveland, Ohio; Department of Medicine, Case Western University School of Medicine, Cleveland, Ohio
| | - Theresa T Pizarro
- Division of Gastroenterology and Liver Disease, Case Western University School of Medicine, Cleveland, Ohio; Department of Pathology, Case Western University School of Medicine, Cleveland, Ohio
| | - Fabio Cominelli
- Division of Gastroenterology and Liver Disease, Case Western University School of Medicine, Cleveland, Ohio; Department of Medicine, Case Western University School of Medicine, Cleveland, Ohio; Department of Pathology, Case Western University School of Medicine, Cleveland, Ohio.
| |
Collapse
|
25
|
Dosh RH, Jordan-Mahy N, Sammon C, Le Maitre C. Interleukin 1 is a key driver of inflammatory bowel disease-demonstration in a murine IL-1Ra knockout model. Oncotarget 2019; 10:3559-3575. [PMID: 31191826 PMCID: PMC6544399 DOI: 10.18632/oncotarget.26894] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2018] [Accepted: 04/03/2019] [Indexed: 02/07/2023] Open
Abstract
Interleukin 1 (IL-1) is an important mediator of inflammation and tissue damage in inflammatory bowel disease (IBD). The balance between IL-1 and IL-1Ra as a natural inhibitor plays a vital role in a variety of diseases. Here, we investigated whether changes seen during IBD are induced spontaneously in mice lacking a functional IL-1rn gene. Histological staining was performed on the jejunum and ileum of BALB/c IL-1rn+/+ and IL-1rn-/- mice to characterize crypt-villus height, villus width, and number of goblet cells per villus. Pro-inflammatory cytokines, immune cell infiltration and matrix-degrading enzymes, together with the production of intestinal enzymes and the integrity of tight and adherent junction proteins were determined using immunohistochemistry. In the small intestine of BALB/c IL-1rn-/- mice the villus heights were significantly reduced; and in the ileum this was accompanied by a decrease in villi width. There was also an increase in goblet cell number and mucin production compared to wild-type mice. IL-1α and IL-1β immunopositivity were increased, whilst IL-1R1 expression was decreased in IL-1rn-/- mice. IL-15 and TNFα were also increased in older IL-1rn-/- mice. Increased polymorphonuclear and macrophage infiltration were seen in IL-1rn-/- mice, whilst expression of matrix-degrading enzymes and digestive enzymes were unchanged, except for dipeptidyl peptidase IV which was increased in younger IL-1rn-/- mice compared to wild type mice. The expression of tight and adhesion junctions were also dramatically decreased in IL-1rn-/- mice. In conclusion, IL-1rn-/- mice developed spontaneous abnormalities which displayed features associated with IBD, demonstrating a clear role for IL-1 in IBD.
Collapse
Affiliation(s)
- Rasha H. Dosh
- Biomolecular Sciences Research Centre, Sheffield Hallam University, Sheffield, UK
- Department of Anatomy and Histology, Faculty of Medicine, University of Kufa, Kufa, Iraq
| | - Nicola Jordan-Mahy
- Biomolecular Sciences Research Centre, Sheffield Hallam University, Sheffield, UK
| | - Christopher Sammon
- Materials and Engineering Research Institute, Sheffield Hallam University, Sheffield, UK
| | - Christine Le Maitre
- Biomolecular Sciences Research Centre, Sheffield Hallam University, Sheffield, UK
| |
Collapse
|
26
|
Intestinal Organoids as a Novel Complementary Model to Dissect Inflammatory Bowel Disease. Stem Cells Int 2019; 2019:8010645. [PMID: 31015842 PMCID: PMC6444246 DOI: 10.1155/2019/8010645] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2018] [Accepted: 02/04/2019] [Indexed: 12/13/2022] Open
Abstract
Inflammatory bowel diseases (IBDs) include colitis ulcerosa and Crohn's disease, besides the rare microscopic colitis. Both diseases show a long-lasting, relapsing-remitting, or even chronic active course with tremendous impact on quality of life. IBDs frequently cause disability, surgical interventions, and high costs; as in other autoimmune diseases, their prevalent occurrence at an early phase of life raises the burden on health care systems. Unfortunately, our understanding of the pathogenesis is still incomplete and treatment therefore largely focuses on suppressing the resulting excessive inflammation. One obstacle for deciphering the causative processes is the scarcity of models that parallel the development of the disease, since intestinal inflammation is mostly induced artificially; moreover, the intestinal epithelium, which strongly contributes to IBD pathogenesis, is difficult to assess. Recently, the development of intestinal epithelial organoids has overcome many of those problems. Here, we give an overview on the current understanding of the pathogenesis of IBDs with reference to the limitations of previous well-established experimental models. We highlight the advantages and detriments of recent organoid-based experimental setups within the IBD field and suggest possible future applications.
Collapse
|
27
|
Buttó LF, Jia LG, Arseneau KO, Tamagawa H, Rodriguez-Palacios A, Li Z, De Salvo C, Pizarro TT, Bamias G, Cominelli F. Death-Domain-Receptor 3 Deletion Normalizes Inflammatory Gene Expression and Prevents Ileitis in Experimental Crohn's Disease. Inflamm Bowel Dis 2019; 25:14-26. [PMID: 30295722 PMCID: PMC6290789 DOI: 10.1093/ibd/izy305] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/02/2018] [Indexed: 12/11/2022]
Abstract
Background TNF-like cytokine 1A (TL1A) and its functional receptor, death-domain-receptor-3 (DR3), are multifunctional mediators of effector and regulatory immunity. We aimed to evaluate the functional role and therapeutic potential of TL1A/DR3 signaling in Crohn's disease-like ileitis. Methods Ileitis-prone SAMP1/YitFc (SAMP) and TNFΔARE/+ mice were rendered deficient for DR3 or TL1A by microsatellite marker-assisted backcrossing. Pathological and immunological characteristics were compared between control and knockout mice, and mucosal immunophenotype was analyzed by Nanostring microarray assay. The therapeutic effect of pharmacological TL1A neutralization was also investigated. Results DR3 deficiency was associated with restoration of a homeostatic mucosal immunostat in SAMP mice through the regulation of several pro- and anti-inflammatory genes. This led to suppression of effector immunity, amelioration of ileitis severity, and compromised ability of either unfractionated CD4+ or CD4+CD45RBhi mucosal lymphocytes to transfer ileitis to severe combined immunodeficient mice recipients. TNF-driven ileitis was also prevented in TNFΔARE/+xDR3-/- mice, in association with decreased expression of the pro-inflammatory cytokines TNF and IFN-γ. In contrast to DR3, TL1A was dispensable for the development of ileitis although it affected the kinetics of inflammation, as TNFΔARE/+xTL1A-/- demonstrated delayed onset of inflammation, whereas administration of a neutralizing, anti-TL1A antibody ameliorated early but not late TNFΔARE/+ ileitis. Conclusion We found a prominent pro-inflammatory role of DR3 in chronic ileitis, which is only partially mediated via interaction with TL1A, raising the possibility for additional DR3 ligands. Death-domain-receptor-3 appears to be a master regulator of mucosal homeostasis and inflammation and may represent a candidate therapeutic target for chronic inflammatory conditions of the bowel.
Collapse
Affiliation(s)
- Ludovica F Buttó
- 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
| | - Li-Guo Jia
- 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
| | - Kristen O Arseneau
- 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
| | | | - Alex Rodriguez-Palacios
- 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
| | - Zhaodong Li
- 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
| | - Carlo De Salvo
- Department of Pathology, 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
- Digestive Health Research Institute, Case Western Reserve University School of Medicine, Cleveland, Ohio, USA
| | - Giorgos Bamias
- GI-Unit, 3rd Academic Department of Internal Medicine, National and Kapodistrian University of Athens, Sotiria Hospital, Athens, Greece
| | - Fabio Cominelli
- Department of Medicine, Case Western Reserve University School of Medicine, Cleveland, Ohio, USA
- Department of Pathology, 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
|
28
|
Wang K, Zhang Z, Liu K, Yang X, Zou H, Zhou J, Miao X, Chen W, Xiong L, Wen Y. Neat1-miRNA204-5p-PI3K-AKT axis as a potential mechanism for photodynamic therapy treated colitis in mice. Photodiagnosis Photodyn Ther 2018; 24:349-357. [PMID: 30385297 DOI: 10.1016/j.pdpdt.2018.10.020] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2018] [Revised: 10/25/2018] [Accepted: 10/26/2018] [Indexed: 02/08/2023]
Abstract
BACKGROUND Inflammatory bowel disease (IBD) is a group of inflammatory conditions characterized by chronic inflammation of the gastrointestinal (GI) tract. Recent preclinical and clinical studies have shown the therapeutic value of photodynamic therapy with low doses of photosensitizer and/or light in colitis treatment, but the detailed mechanism is unclear. METHODS We examined 5-aminolevulinic acid (5-ALA) mediated photodynamic therapy (PDT) in dextran sulfate sodium (DSS)-induced colitis in mice. Endoscopic and macroscopic observations as well as the Disease Activity Index score (DAI) was used to monitor the disease. Cytokine expression was measured by quantitative RT-PCR analysis to evaluate the efficacy of PDT. We applied the Clariom™ D bioinformatics analysis to investigate the differentially expressed genes after PDT. Finally, we used quantitative RT-PCR analysis and Western blotting to validate the targets of differentially expressed genes. RESULTS 5-ALA-PDT improved the DAI score and decreased the expression of various inflammatory cytokines. The Clariom™ D bioinformatics analysis identified 2043 diff ;erentially expressed genes (702 up-regulated and 1341 down-regulated). We confirmed the down-regulation of lncRNA-Neat1, lncRNA-Snhg5 and lncRNA-Gm9926 and the up-regulation of miRNA-204-5p, miRNA-218-5p, miRNA-200a-3p and miRNA-466h-5p. Additionally, the decreased expression level of the AKT3, p-AKT3, PI3K, p-PI3K protein were confirmed. CONCLUSIONS This study provides in vivo comprehensive lncRNA and miRNA microarray analysis after 5-ALA-PDT treatment in DSS-induced colitis, and it may help to develop novel lncRNA-miRNA-related therapeutic approaches, such as the Neat1-miRNA204-5p-PI3K-AKT axis, to further increase the efficiency of PDT in IBD.
Collapse
Affiliation(s)
- Kunpeng Wang
- Department of General Surgery, Second Xiangya Hospital, Central South University, Changsha, 410011, Hunan, China
| | - Zijian Zhang
- Department of General Surgery, Second Xiangya Hospital, Central South University, Changsha, 410011, Hunan, China
| | - Kai Liu
- Department of General Surgery, Second Xiangya Hospital, Central South University, Changsha, 410011, Hunan, China
| | - Xin Yang
- Department of General Surgery, Second Xiangya Hospital, Central South University, Changsha, 410011, Hunan, China
| | - Heng Zou
- Department of General Surgery, Second Xiangya Hospital, Central South University, Changsha, 410011, Hunan, China
| | - Jiangjiao Zhou
- Department of General Surgery, Second Xiangya Hospital, Central South University, Changsha, 410011, Hunan, China
| | - Xiongying Miao
- Department of General Surgery, Second Xiangya Hospital, Central South University, Changsha, 410011, Hunan, China
| | - Wei Chen
- Department of Physics and the SAVANT Center, University of Texas at Arlington, Arlington, 76019-0059, USA
| | - Li Xiong
- Department of General Surgery, Second Xiangya Hospital, Central South University, Changsha, 410011, Hunan, China.
| | - Yu Wen
- Department of General Surgery, Second Xiangya Hospital, Central South University, Changsha, 410011, Hunan, China.
| |
Collapse
|
29
|
He P, Haque A, Lin S, Cominelli F, Yun CC. Inhibition of autotaxin alleviates inflammation and increases the expression of sodium-dependent glucose cotransporter 1 and Na +/H + exchanger 3 in SAMP1/Fc mice. Am J Physiol Gastrointest Liver Physiol 2018; 315:G762-G771. [PMID: 30118349 PMCID: PMC6293258 DOI: 10.1152/ajpgi.00215.2018] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Crohn's disease (CD) is a chronic, relapsing, inflammatory disease that is often associated with malnutrition because of inflammation in the small intestine. Autotaxin (ATX) is a secreted enzyme that produces extracellular lysophosphatidic acid. Increasing evidence suggests that ATX is upregulated during inflammation, and inhibition of ATX has been effective in attenuating chronic inflammatory conditions, such as arthritis and pulmonary fibrosis. This study aims to determine whether inhibition of ATX alleviates CD-associated inflammation and malnutrition by using SAMP1/Fc mice, a model of CD-like ileitis. SAMP1/Fc mice were treated the ATX inhibitor PF-8380 for 4 wk. Inhibition of ATX led to increased weight gain in SAMP1/Fc mice, decreased T helper 2 cytokine expression, including IL-4, IL-5, and IL-13, and attenuated immune cell migration. SAMP1/Fc mice have low expression of Na+-dependent glucose transporter 1 (SGLT1), suggesting impaired nutrient absorption associated with ileitis. PF-8380 treatment significantly enhanced SGLT1 expression in SAMP1/Fc mice, which could reflect the increased weight changes. However, IL-4 or IL-13 did not alter SGLT1 expression in Caco-2 cells, ruling out their direct effects on SGLT1 expression. Immunofluorescence analysis showed that the expression of sucrase-isomaltase, a marker for intestinal epithelial cell (IEC) differentiation, was decreased in inflamed regions of SAMP1/Fc mice, which was partially restored by PF-8380. Moreover, expression of Na+/H+ exchanger 3 was also improved by PF-8380, suggesting that suppression of inflammation by PF-8380 enhanced IEC differentiation. Our study therefore suggests that ATX is a potential target for treating intestinal inflammation and restoration of the absorptive function of the intestine. NEW & NOTEWORTHY This study is the first, to our knowledge, to determine whether autotoxin (ATX) inhibition improves inflammation and body weights in SAMP1/Fc mice, a mouse model of ileitis. ATX inhibition increased body weights of SAMP1/Fc mice and increased Na+-dependent glucose transporter 1 (SGLT1) expression. Increased SGLT1 expression in the inflamed regions was not a direct effect of cytokines but an indirect effect of increased epithelial cell differentiation upon ATX inhibition.
Collapse
Affiliation(s)
- Peijian He
- 1Division of Digestive Diseases, Department of Medicine, Emory University School of Medicine, Atlanta, Georgia,2Atlanta Veterans Affairs Medical Center, Decatur, Georgia
| | - Abedul Haque
- 1Division of Digestive Diseases, Department of Medicine, Emory University School of Medicine, Atlanta, Georgia,2Atlanta Veterans Affairs Medical Center, Decatur, Georgia
| | - Songbai Lin
- 1Division of Digestive Diseases, Department of Medicine, Emory University School of Medicine, Atlanta, Georgia,2Atlanta Veterans Affairs Medical Center, Decatur, Georgia
| | - Fabio Cominelli
- 3Department of Medicine, Case Digestive Health Research Institute, Case Western University School of Medicine, Cleveland, Ohio
| | - C. Chris Yun
- 1Division of Digestive Diseases, Department of Medicine, Emory University School of Medicine, Atlanta, Georgia,2Atlanta Veterans Affairs Medical Center, Decatur, Georgia,4Winship Cancer Institute, Emory University School of Medicine, Atlanta, Georgia
| |
Collapse
|
30
|
Li Z, Buttó LF, Buela KA, Jia LG, Lam M, Ward JD, Pizarro TT, Cominelli F. Death Receptor 3 Signaling Controls the Balance between Regulatory and Effector Lymphocytes in SAMP1/YitFc Mice with Crohn's Disease-Like Ileitis. Front Immunol 2018; 9:362. [PMID: 29545797 PMCID: PMC5837992 DOI: 10.3389/fimmu.2018.00362] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2017] [Accepted: 02/08/2018] [Indexed: 12/17/2022] Open
Abstract
Death receptor 3 (DR3), a member of the tumor necrosis factor receptor (TNFR) superfamily, has been implicated in regulating T-helper type-1 (TH1), type-2 (TH2), and type-17 (TH17) responses as well as regulatory T cell (Treg) and innate lymphoid cell (ILC) functions during immune-mediated diseases. However, the role of DR3 in controlling lymphocyte functions in inflammatory bowel disease (IBD) is not fully understood. Recent studies have shown that activation of DR3 signaling modulates Treg expansion suggesting that stimulation of DR3 represents a potential therapeutic target in human inflammatory diseases, including Crohn's disease (CD). In this study, we tested a specific DR3 agonistic antibody (4C12) in SAMP1/YitFc (SAMP) mice with CD-like ileitis. Interestingly, treatment with 4C12 prior to disease manifestation markedly worsened the severity of ileitis in SAMP mice despite an increase in FoxP3+ lymphocytes in mesenteric lymph node (MLN) and small-intestinal lamina propria (LP) cells. Disease exacerbation was dominated by overproduction of both TH1 and TH2 cytokines and associated with expansion of dysfunctional CD25-FoxP3+ and ILC group 1 (ILC1) cells. These effects were accompanied by a reduction in CD25+FoxP3+ and ILC group 3 (ILC3) cells. By comparison, genetic deletion of DR3 effectively reversed the inflammatory phenotype in SAMP mice by promoting the expansion of CD25+FoxP3+ over CD25-FoxP3+ cells and the production of IL-10 protein. Collectively, our data demonstrate that DR3 signaling modulates a multicellular network, encompassing Tregs, T effectors, and ILCs, governing disease development and progression in SAMP mice with CD-like ileitis. Manipulating DR3 signaling toward the restoration of the balance between protective and inflammatory lymphocytes may represent a novel and targeted therapeutic modality for patients with CD.
Collapse
MESH Headings
- Animals
- Antibodies, Monoclonal/metabolism
- Cells, Cultured
- Crohn Disease/immunology
- Cytokines/metabolism
- Disease Models, Animal
- Female
- Forkhead Transcription Factors/metabolism
- Humans
- Ileitis/genetics
- Ileitis/immunology
- Male
- Membrane Proteins/genetics
- Mice
- Mice, Inbred AKR
- Mice, Knockout
- Mice, Transgenic
- Nuclear Proteins/genetics
- Receptors, Tumor Necrosis Factor, Member 25/agonists
- Receptors, Tumor Necrosis Factor, Member 25/genetics
- Receptors, Tumor Necrosis Factor, Member 25/immunology
- Signal Transduction
- T-Lymphocytes, Helper-Inducer/immunology
- T-Lymphocytes, Regulatory/immunology
Collapse
Affiliation(s)
- Zhaodong Li
- BRB-5, Digestive Health Research Institute, Case Western Reserve University, Cleveland, OH, United States
| | - Ludovica F. Buttó
- BRB-5, Digestive Health Research Institute, Case Western Reserve University, Cleveland, OH, United States
| | - Kristine-Anne Buela
- Department of Pathology, Case Western Reserve University, Cleveland, OH, United States
| | - Li-Guo Jia
- BRB-5, Digestive Health Research Institute, Case Western Reserve University, Cleveland, OH, United States
| | - Minh Lam
- BRB-5, Digestive Health Research Institute, Case Western Reserve University, Cleveland, OH, United States
| | - John D. Ward
- BRB-5, Digestive Health Research Institute, Case Western Reserve University, Cleveland, OH, United States
| | - Theresa T. Pizarro
- Department of Pathology, Case Western Reserve University, Cleveland, OH, United States
| | - Fabio Cominelli
- BRB-5, Digestive Health Research Institute, Case Western Reserve University, Cleveland, OH, United States
| |
Collapse
|
31
|
Cao W, Kayama H, Chen ML, Delmas A, Sun A, Kim SY, Rangarajan ES, McKevitt K, Beck AP, Jackson CB, Crynen G, Oikonomopoulos A, Lacey PN, Martinez GJ, Izard T, Lorenz RG, Rodriguez-Palacios A, Cominelli F, Abreu MT, Hommes DW, Koralov SB, Takeda K, Sundrud MS. The Xenobiotic Transporter Mdr1 Enforces T Cell Homeostasis in the Presence of Intestinal Bile Acids. Immunity 2017; 47:1182-1196.e10. [PMID: 29262351 PMCID: PMC5741099 DOI: 10.1016/j.immuni.2017.11.012] [Citation(s) in RCA: 68] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2016] [Revised: 07/27/2017] [Accepted: 11/07/2017] [Indexed: 12/13/2022]
Abstract
CD4+ T cells are tightly regulated by microbiota in the intestine, but whether intestinal T cells interface with host-derived metabolites is less clear. Here, we show that CD4+ T effector (Teff) cells upregulated the xenobiotic transporter, Mdr1, in the ileum to maintain homeostasis in the presence of bile acids. Whereas wild-type Teff cells upregulated Mdr1 in the ileum, those lacking Mdr1 displayed mucosal dysfunction and induced Crohn's disease-like ileitis following transfer into Rag1-/- hosts. Mdr1 mitigated oxidative stress and enforced homeostasis in Teff cells exposed to conjugated bile acids (CBAs), a class of liver-derived emulsifying agents that actively circulate through the ileal mucosa. Blocking ileal CBA reabsorption in transferred Rag1-/- mice restored Mdr1-deficient Teff cell homeostasis and attenuated ileitis. Further, a subset of ileal Crohn's disease patients displayed MDR1 loss of function. Together, these results suggest that coordinated interaction between mucosal Teff cells and CBAs in the ileum regulate intestinal immune homeostasis.
Collapse
MESH Headings
- ATP Binding Cassette Transporter, Subfamily B, Member 1/deficiency
- ATP Binding Cassette Transporter, Subfamily B, Member 1/genetics
- ATP Binding Cassette Transporter, Subfamily B, Member 1/immunology
- Acridines/pharmacology
- Adult
- Animals
- Bile Acids and Salts/immunology
- Bile Acids and Salts/metabolism
- Bile Acids and Salts/pharmacology
- Biological Transport
- CD4-Positive T-Lymphocytes/drug effects
- CD4-Positive T-Lymphocytes/immunology
- CD4-Positive T-Lymphocytes/pathology
- Crohn Disease/genetics
- Crohn Disease/immunology
- Crohn Disease/pathology
- Disease Models, Animal
- Female
- Gene Expression Regulation
- Homeodomain Proteins/genetics
- Homeodomain Proteins/immunology
- Homeostasis/immunology
- Humans
- Ileitis/genetics
- Ileitis/immunology
- Ileitis/pathology
- Ileum/immunology
- Ileum/pathology
- Immunity, Mucosal
- Intestinal Mucosa/immunology
- Intestinal Mucosa/pathology
- Male
- Mice
- Mice, Inbred C57BL
- Mice, Transgenic
- Middle Aged
- Oxidative Stress
- Signal Transduction
- Tetrahydroisoquinolines/pharmacology
Collapse
Affiliation(s)
- Wei Cao
- Department of Immunology and Microbiology, The Scripps Research Institute, Jupiter, FL 33458, USA
| | - Hisako Kayama
- Department of Microbiology and Immunology, Graduate School of Medicine, Osaka University, Osaka 565-0871, Japan
| | - Mei Lan Chen
- Department of Immunology and Microbiology, The Scripps Research Institute, Jupiter, FL 33458, USA
| | - Amber Delmas
- Department of Immunology and Microbiology, The Scripps Research Institute, Jupiter, FL 33458, USA
| | - Amy Sun
- Department of Pathology , New York University School of Medicine, New York, NY 10016, USA
| | - Sang Yong Kim
- Rodent Genetic Engineering Core , New York University School of Medicine, New York, NY 10016, USA
| | - Erumbi S Rangarajan
- Department of Integrative Structural and Computational Biology, The Scripps Research Institute, Jupiter, FL 33458, USA
| | - Kelly McKevitt
- Department of Immunology and Microbiology, The Scripps Research Institute, Jupiter, FL 33458, USA
| | - Amanda P Beck
- Department of Veterinary Sciences, MD Anderson Cancer Center, Bastrop, TX 78602, USA
| | - Cody B Jackson
- Department of Immunology and Microbiology, The Scripps Research Institute, Jupiter, FL 33458, USA
| | - Gogce Crynen
- Bioinformatics Core Facility, The Scripps Research Institute, Jupiter, FL 33458, USA
| | - Angelos Oikonomopoulos
- Vatche and Tamar Manoukian Division of Digestive Diseases, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA 90095, USA
| | - Precious N Lacey
- Vatche and Tamar Manoukian Division of Digestive Diseases, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA 90095, USA
| | - Gustavo J Martinez
- Department of Microbiology and Immunology, Chicago Medical School, Rosalind Franklin University of Medicine and Science, North Chicago, IL 60064, USA
| | - Tina Izard
- Department of Integrative Structural and Computational Biology, The Scripps Research Institute, Jupiter, FL 33458, USA
| | - Robin G Lorenz
- Department of Pathology, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Alex Rodriguez-Palacios
- Digestive Health Research Institute, Case Western Reserve University School of Medicine, Cleveland, OH 44106, USA
| | - Fabio Cominelli
- Digestive Health Research Institute, Case Western Reserve University School of Medicine, Cleveland, OH 44106, USA
| | - Maria T Abreu
- Division of Gastroenterology, Department of Medicine, Miller School of Medicine, University of Miami, Miami, FL 33136, USA
| | - Daniel W Hommes
- Vatche and Tamar Manoukian Division of Digestive Diseases, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA 90095, USA
| | - Sergei B Koralov
- Department of Pathology , New York University School of Medicine, New York, NY 10016, USA
| | - Kiyoshi Takeda
- Department of Microbiology and Immunology, Graduate School of Medicine, Osaka University, Osaka 565-0871, Japan
| | - Mark S Sundrud
- Department of Immunology and Microbiology, The Scripps Research Institute, Jupiter, FL 33458, USA.
| |
Collapse
|
32
|
Dunkin D, Berin MC, Mondoulet L, Tobar S, Yeretssian G, Tordesillas L, Iuga A, Larcher T, Gillespie V, Benhamou PH, Colombel JF, Sampson HA. Epicutaneous Tolerance Induction to a Bystander Antigen Abrogates Colitis and Ileitis in Mice. Inflamm Bowel Dis 2017; 23:1972-1982. [PMID: 29019858 PMCID: PMC5659741 DOI: 10.1097/mib.0000000000001273] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
BACKGROUND Although inflammatory bowel disease (IBD) is a failure in maintaining tolerance to the intestinal microbiota, few studies have investigated the use of immunologic tolerance as a treatment approach for IBD. We hypothesized that induction of immune tolerance at a distal site could suppress intestinal inflammation through a process of bystander regulation. METHODS Epicutaneous tolerance was induced by topical application of ovalbumin (OVA) using a Viaskin patch for 48 hours. In some experiments, a single feed of ovalbumin was used to drive epicutaneous tolerance-induced regulatory T cells (Tregs) to the intestine. The mechanism of tolerance induction was tested using neutralizing antibodies against TGF-β, IL-10, and Treg depletion using Foxp3-DTR mice. The capacity of skin-draining Tregs, or epicutaneous tolerance, to prevent or treat experimental IBD was tested using T-cell transfer colitis, dextran sodium sulfate (DSS) colitis, and ileitis in SAMP-YITFc mice. Weight loss, colonic inflammatory cytokines and histology were assessed. RESULTS Epicutaneous exposure to ovalbumin induced systemic immune tolerance by a TGF-β-dependent, but IL-10 and iFoxp3 Treg-independent mechanism. Skin draining Tregs suppressed the development of colitis. Epicutaneous tolerance to a model antigen prevented intestinal inflammation in the dextran sodium sulfate and SAMP-YITFc models and importantly could halt disease in mice already experiencing weight loss in the T-cell transfer model of colitis. This was accompanied by a significant accumulation of LAP and Foxp3 Tregs in the colon. CONCLUSIONS This is the first demonstration that epicutaneous tolerance to a model antigen can lead to bystander suppression of inflammation and prevention of disease progression in preclinical models of IBD.
Collapse
Affiliation(s)
- David Dunkin
- *Division of Pediatric Gastroenterology, The Mindich Child Health and Development Institute, Icahn School of Medicine at Mount Sinai, New York, New York; †Division of Pediatric Allergy and Immunology, Precision Immunology Institute, The Mindich Child Health and Development Institute, Icahn School of Medicine at Mount Sinai, New York, New York; ‡DBV Technologies, Bagneux, France; §Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, New York; ‖Department of Pathology, Columbia University Medical School, New York, New York; ¶National Veterinary School, Nantes, France; **Department of Comparative Pathology, Icahn School of Medicine at Mount Sinai, New York, New York; and ††Division of Gastroenterology, Icahn School of Medicine at Mount Sinai, New York, New York
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
33
|
Cominelli F, Arseneau KO, Rodriguez-Palacios A, Pizarro TT. Uncovering Pathogenic Mechanisms of Inflammatory Bowel Disease Using Mouse Models of Crohn's Disease-Like Ileitis: What is the Right Model? Cell Mol Gastroenterol Hepatol 2017; 4:19-32. [PMID: 28560286 PMCID: PMC5439236 DOI: 10.1016/j.jcmgh.2017.02.010] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/06/2017] [Accepted: 02/15/2017] [Indexed: 02/06/2023]
Abstract
Crohn's disease and ulcerative colitis, together known as inflammatory bowel disease, are debilitating chronic disorders of unknown cause and cure. Our evolving understanding of these pathologies is enhanced greatly by the use of animal models of intestinal inflammation that allow in vivo mechanistic studies, preclinical evaluation of new therapies, and investigation into the causative factors that underlie disease pathogenesis. Several animal models, most commonly generated in mice, exist for the study of colitis. The appropriateness of their use often can be determined by their mode of generation (ie, chemical induction, T-cell transfer, targeted genetic manipulation, spontaneously occurring, and so forth), the type of investigation (mechanistic studies, pathogenic experiments, preclinical evaluations, and so forth), and the type of inflammation that occurs in the model (acute vs chronic colitis, tissue injury/repair, and so forth). Although most murine models of inflammatory bowel disease develop inflammation in the colon, Crohn's disease most commonly occurs in the terminal ileum, where a very limited number of mouse models manifest disease. This review discusses appropriate experimental applications for different mouse models of colitis, and highlights the particular utility of 2 highly relevant models of Crohn's-like ileitis-the spontaneous SAMP1/YitFc inbred mouse strain and the genetically engineered TnfΔAU-rich element/+ mouse model of tumor necrosis factor overexpression, both of which bear strong resemblance to the human condition. Similar to patients with Crohn's disease, SAMP1/YitFc ileitis develops spontaneously, without chemical, genetic, or immunologic manipulation, making this model particularly relevant for studies aimed at identifying the primary defect underlying the occurrence of Crohn's ileitis, as well as preclinical testing of novel treatment modalities.
Collapse
Affiliation(s)
- Fabio Cominelli
- Department of Medicine, Case Western Reserve University School of Medicine, Cleveland, Ohio,Department of Pathology, Case Western Reserve University School of Medicine, Cleveland, Ohio,Digestive Health Research Institute, Case Western Reserve University School of Medicine, Cleveland, Ohio,Correspondence Address correspondence to: Fabio Cominelli, MD, PhD, Division of Gastroenterology, Case Western Reserve University School of Medicine, 11100 Euclid Avenue, Cleveland, Ohio 44106-5066. fax: (216) 844-7371.Division of GastroenterologyCase Western Reserve University School of Medicine11100 Euclid AvenueClevelandOhio 44106-5066
| | - Kristen O. Arseneau
- Department of Medicine, Case Western Reserve University School of Medicine, Cleveland, Ohio,Digestive Health Research Institute, Case Western Reserve University School of Medicine, Cleveland, Ohio
| | - Alexander Rodriguez-Palacios
- Department of Medicine, Case Western Reserve University School of Medicine, Cleveland, Ohio,Digestive Health Research Institute, Case Western Reserve University School of Medicine, Cleveland, Ohio
| | - Theresa T. Pizarro
- Department of Medicine, Case Western Reserve University School of Medicine, Cleveland, Ohio,Department of Pathology, Case Western Reserve University School of Medicine, Cleveland, Ohio,Digestive Health Research Institute, Case Western Reserve University School of Medicine, Cleveland, Ohio
| |
Collapse
|
34
|
Epithelial-specific Toll-like Receptor (TLR)5 Activation Mediates Barrier Dysfunction in Experimental Ileitis. Inflamm Bowel Dis 2017; 23:392-403. [PMID: 28146004 DOI: 10.1097/mib.0000000000001035] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
BACKGROUND A large body of evidence supports a central role of TLR5 and its natural ligand, flagellin, in Crohn's disease (CD), with the precise mechanism(s) still unresolved. METHODS We investigated the role of flagellin/TLR5 in SAMP1/YitFc (SAMP) mice, a spontaneous model of Crohn's disease-like ileitis. RESULTS Ileal Tlr5 and serum antiflagellin IgG antibodies were increased in SAMP before the onset of inflammation and during established disease; these trends were abrogated in the absence of colonizing commensal bacteria. Irradiated SAMP receiving either wild-type (AKR) or SAMP bone marrow (BM) developed severe ileitis and displayed increased ileal Tlr5 compared with AKR recipients of either SAMP or AKR bone marrow, neither of which conferred ileitis, suggesting that elevated TLR5 in native SAMP is derived primarily from a nonhematopoietic (e.g., epithelial) source. Indeed, ileal epithelial TLR5 in preinflamed SAMP was increased compared with age-matched AKR and germ-free SAMP. TLR5-specific ex vivo activation of SAMP ileal tissues decreased epithelial barrier resistance, indicative of increased permeability, and was accompanied by altered expression of the tight junction proteins, claudin-3, occludin, and zonula occludens-1. CONCLUSIONS Our results provide evidence that aberrant, elevated TLR5 expression is present in the ileal epithelium of SAMP mice, is augmented in the presence of the gut microbiome, and that TLR5 activation in response to bacterial flagellin results in a deficiency to maintain appropriate epithelial barrier integrity. Together, these findings represent a potential mechanistic pathway leading to the exacerbation and perpetuation of chronic gut inflammation in experimental ileitis and possibly, in patients with Crohn's disease.
Collapse
|
35
|
Do JS, Kim S, Keslar K, Jang E, Huang E, Fairchild RL, Pizarro TT, Min B. γδ T Cells Coexpressing Gut Homing α4β7 and αE Integrins Define a Novel Subset Promoting Intestinal Inflammation. THE JOURNAL OF IMMUNOLOGY 2016; 198:908-915. [PMID: 27927968 DOI: 10.4049/jimmunol.1601060] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/17/2016] [Accepted: 11/10/2016] [Indexed: 02/07/2023]
Abstract
γδ T lymphocytes, dominant T cell subsets in the intestine, mediate both regulatory and pathogenic roles, yet the mechanisms underlying such opposing effects remain unclear. In this study, we identified a unique γδ T cell subset that coexpresses high levels of gut-homing integrins, CD103 and α4β7. They were exclusively found in the mesenteric lymph node after T cell-mediated colitis induction, and their appearance preceded the inflammation. Adoptive transfer of the CD103+α4β7high subsets enhanced Th1/Th17 T cell generation and accumulation in the intestine, and the disease severity. The level of generation correlated with the disease severity. Moreover, these cells were also found to be elevated in a spontaneous mouse model of ileitis. Based on the procolitogenic function, we referred to this subset as "inflammatory" γδ T cells. Targeting inflammatory γδ T cells may open a novel strategy to treat inflammatory diseases where γδ T cells play a pathogenic role including inflammatory bowel disease.
Collapse
Affiliation(s)
- Jeong-Su Do
- Department of Immunology, Lerner Research Institute, Cleveland Clinic Foundation, Cleveland, OH 44195
| | - Sohee Kim
- Department of Immunology, Lerner Research Institute, Cleveland Clinic Foundation, Cleveland, OH 44195
| | - Karen Keslar
- Department of Immunology, Lerner Research Institute, Cleveland Clinic Foundation, Cleveland, OH 44195
| | - Eunjung Jang
- Department of Immunology, Lerner Research Institute, Cleveland Clinic Foundation, Cleveland, OH 44195
| | - Emina Huang
- Department of Stem Cell Biology and Regenerative Medicine, Lerner Research Institute, Cleveland Clinic Foundation, Cleveland, OH 44195; and
| | - Robert L Fairchild
- Department of Immunology, Lerner Research Institute, Cleveland Clinic Foundation, Cleveland, OH 44195
| | - Theresa T Pizarro
- Department of Pathology, Case Western Reserve University, Cleveland, OH 44116
| | - Booki Min
- Department of Immunology, Lerner Research Institute, Cleveland Clinic Foundation, Cleveland, OH 44195;
| |
Collapse
|
36
|
Di Martino L, Dave M, Menghini P, Xin W, Arseneau KO, Pizarro TT, Cominelli F. Protective Role for TWEAK/Fn14 in Regulating Acute Intestinal Inflammation and Colitis-Associated Tumorigenesis. Cancer Res 2016; 76:6533-6542. [PMID: 27634763 PMCID: PMC5290134 DOI: 10.1158/0008-5472.can-16-0400] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2016] [Revised: 08/30/2016] [Accepted: 08/30/2016] [Indexed: 12/14/2022]
Abstract
Inflammatory bowel disease causes chronic, relapsing intestinal inflammation that can lead to the development of colorectal cancer. Members of the TNF superfamily are key regulators of intestinal inflammation. In particular, TNF-like weak inducer of apoptosis (TWEAK) and its receptor, Fn14, are involved in normal and pathologic intestinal tissue remodeling. In this study, we show that the TWEAK/Fn14 signaling complex plays a protective role during the acute stage of intestinal inflammation and contributes to the prevention of colitis-associated cancer during chronic inflammation through its proapoptotic effects. Colitis was induced in Fn14-/- and Fn14+/+ wild-type littermates by administering 3% dextran sodium sulfate (DSS) for 7 days followed by 2-week recovery; azoxymethane (AOM) administration followed by two cycles of DSS/recovery was used to induce tumors. Reciprocal bone marrow chimeric mice were generated to compare hematopoietic and nonhematopoietic-specific effector tissues. Fn14-/- mice had enhanced susceptibility to colitis compared with Fn14+/+ controls as assessed by endoscopic and histologic inflammatory scores, daily weight loss, and mortality rates during recovery after DSS administration. Bone marrow transfer experiments showed that both hematopoietic and nonhematopoietic components are involved in protection against colitis. Tumor lesions were found in the colons of most Fn14-/- mice, but not Fn14+/+ controls. AOM/DSS administration enhanced susceptibility to tumorigenesis in Fn14-/- mice. Overall, these findings show that Fn14 plays a protective role during the acute stages of intestinal inflammation, and its absence promotes the development of colitis-associated cancer. Cancer Res; 76(22); 6533-42. ©2016 AACR.
Collapse
Affiliation(s)
- Luca Di Martino
- Case Digestive Health Research Institute, Case Western University School of Medicine, Cleveland, Ohio
- Department of Medicine, Case Western University School of Medicine, Cleveland, Ohio
| | - Maneesh Dave
- Case Digestive Health Research Institute, Case Western University School of Medicine, Cleveland, Ohio
- Department of Medicine, Case Western University School of Medicine, Cleveland, Ohio
| | - Paola Menghini
- Case Digestive Health Research Institute, Case Western University School of Medicine, Cleveland, Ohio
- Case Digestive Health Research Institute, Case Western University School of Medicine, Cleveland, Ohio
| | - Wei Xin
- Case Digestive Health Research Institute, Case Western University School of Medicine, Cleveland, Ohio
- Department of Pathology, Case Western University School of Medicine, Cleveland, Ohio
| | - Kristen O Arseneau
- Case Digestive Health Research Institute, Case Western University School of Medicine, Cleveland, Ohio
- Department of Medicine, Case Western University School of Medicine, Cleveland, Ohio
| | - Theresa T Pizarro
- Case Digestive Health Research Institute, Case Western University School of Medicine, Cleveland, Ohio
- Department of Pathology, Case Western University School of Medicine, Cleveland, Ohio
| | - Fabio Cominelli
- Case Digestive Health Research Institute, Case Western University School of Medicine, Cleveland, Ohio.
- Department of Medicine, Case Western University School of Medicine, Cleveland, Ohio
- Department of Pathology, Case Western University School of Medicine, Cleveland, Ohio
| |
Collapse
|
37
|
Roulis M, Bongers G, Armaka M, Salviano T, He Z, Singh A, Seidler U, Becker C, Demengeot J, Furtado GC, Lira SA, Kollias G. Host and microbiota interactions are critical for development of murine Crohn's-like ileitis. Mucosal Immunol 2016; 9:787-97. [PMID: 26487367 PMCID: PMC5027991 DOI: 10.1038/mi.2015.102] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2015] [Accepted: 08/31/2015] [Indexed: 02/04/2023]
Abstract
Deregulation of host-microbiota interactions in the gut is a pivotal characteristic of Crohn's disease. It remains unclear, however, whether commensals and/or the dysbiotic microbiota associated with pathology in humans are causally involved in Crohn's pathogenesis. Here, we show that Crohn's-like ileitis in Tnf(ΔARE/+) mice is microbiota-dependent. Germ-free Tnf(ΔARE/+) mice are disease-free and the microbiota and its innate recognition through Myd88 are indispensable for tumor necrosis factor (TNF) overexpression and disease initiation in this model. The epithelium of diseased mice shows no major defects in mucus barrier and paracellular permeability. However, Tnf(ΔARE/+) ileitis associates with the reduction of lysozyme-expressing Paneth cells, mediated by adaptive immune effectors. Furthermore, we show that established but not early ileitis in Tnf(ΔARE/+) mice involves defective expression of antimicrobials and dysbiosis, characterized by Firmicutes expansion, including epithelial-attaching segmented filamentous bacteria, and decreased abundance of Bacteroidetes. Microbiota modulation by antibiotic treatment at an early disease stage rescues ileitis. Our results suggest that the indigenous microbiota is sufficient to drive TNF overexpression and Crohn's ileitis in the genetically susceptible Tnf(ΔARE/+) hosts, whereas dysbiosis in this model results from disease-associated alterations including loss of lysozyme-expressing Paneth cells.
Collapse
Affiliation(s)
- M Roulis
- Institute of Immunology, Biomedical Sciences Research Center “Alexander Fleming”, Vari 16672, Greece
| | - G Bongers
- Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - M Armaka
- Institute of Immunology, Biomedical Sciences Research Center “Alexander Fleming”, Vari 16672, Greece
| | - T Salviano
- Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Z He
- Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - A Singh
- Department of Gastroenterology, Hepatology and Endocrinology, Hannover Medical School, Hannover, Germany
| | - U Seidler
- Department of Gastroenterology, Hepatology and Endocrinology, Hannover Medical School, Hannover, Germany
| | - C Becker
- Department of Medicine 1, Universitätsklinikum der Friedrich-Alexander-Universität, Erlangen, Germany
| | - J Demengeot
- Instituto Gulbenkian de Ciência, Oeiras, Portugal
| | - GC Furtado
- Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - SA Lira
- Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA,Address correspondence to: Sergio Lira, Immunology Institute, Icahn School of Medicine at Mount Sinai, 1425 Madison Ave, Box 1630, New York, NY 10029-6574. Phone: 1-212-659-9404; Fax: 1-212-849-2525; and George Kollias, Biomedical Sciences Research Center “Alexander Fleming”, 34, Al. Fleming Street, 16672 Vari, Greece. Phone: +302109656507; Fax: +302109656563; and Department of Physiology, Medical School, National & Kapodistrian University of Athens, 75 Micras Asias, Goudi-Athens, 115 27, Greece. Phone: +302107462507; Fax: +30210-7462571;
| | - G Kollias
- Institute of Immunology, Biomedical Sciences Research Center “Alexander Fleming”, Vari 16672, Greece,Second address: Department of Physiology, Medical School, National & Kapodistrian University of Athens, Athens 11527, Greece,Address correspondence to: Sergio Lira, Immunology Institute, Icahn School of Medicine at Mount Sinai, 1425 Madison Ave, Box 1630, New York, NY 10029-6574. Phone: 1-212-659-9404; Fax: 1-212-849-2525; and George Kollias, Biomedical Sciences Research Center “Alexander Fleming”, 34, Al. Fleming Street, 16672 Vari, Greece. Phone: +302109656507; Fax: +302109656563; and Department of Physiology, Medical School, National & Kapodistrian University of Athens, 75 Micras Asias, Goudi-Athens, 115 27, Greece. Phone: +302107462507; Fax: +30210-7462571;
| |
Collapse
|
38
|
Abstract
Pathogenesis of the inflammatory bowel diseases (IBDs), such as ulcerative colitis (UC) and Crohn's disease (CD), involve proinflammatory changes within the microbiota, chronic immune-mediated inflammatory responses, and epithelial dysfunction. Converging data from genome-wide association studies, mouse models of IBD, and clinical trials indicate that cytokines are key effectors of both normal homeostasis and chronic inflammation in the gut. Yet many questions remain concerning the role of specific cytokines in different IBDs within distinct regions of the gut, and regarding cellular mechanisms of action. In this article, we review current and emerging concepts concerning the role of cytokines in IBD with a focus on immune regulation, T cell subsets, and potential clinical applications.
Collapse
Affiliation(s)
- Mei Lan Chen
- Department of Cancer Biology, The Scripps Research Institute, Jupiter, FL 33458 USA
| | - Mark S. Sundrud
- Department of Cancer Biology, The Scripps Research Institute, Jupiter, FL 33458 USA
| |
Collapse
|
39
|
Abstract
Pathogenesis of the inflammatory bowel diseases (IBDs), such as ulcerative colitis (UC) and Crohn's disease (CD), involve proinflammatory changes within the microbiota, chronic immune-mediated inflammatory responses, and epithelial dysfunction. Converging data from genome-wide association studies, mouse models of IBD, and clinical trials indicate that cytokines are key effectors of both normal homeostasis and chronic inflammation in the gut. Yet many questions remain concerning the role of specific cytokines in different IBDs within distinct regions of the gut, and regarding cellular mechanisms of action. In this article, we review current and emerging concepts concerning the role of cytokines in IBD with a focus on immune regulation, T cell subsets, and potential clinical applications.
Collapse
|
40
|
De Salvo C, Wang XM, Pastorelli L, Mattioli B, Omenetti S, Buela KA, Chowdhry S, Garg RR, Goodman WA, Rodriguez-Palacios A, Smith DE, Abbott DW, Cominelli F, Bamias G, Xin W, Lee JJ, Vecchi M, Pizarro TT. IL-33 Drives Eosinophil Infiltration and Pathogenic Type 2 Helper T-Cell Immune Responses Leading to Chronic Experimental Ileitis. THE AMERICAN JOURNAL OF PATHOLOGY 2016; 186:885-98. [PMID: 26908008 PMCID: PMC5807926 DOI: 10.1016/j.ajpath.2015.11.028] [Citation(s) in RCA: 61] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/07/2015] [Revised: 11/03/2015] [Accepted: 11/19/2015] [Indexed: 02/08/2023]
Abstract
Although a clear association has been established between IL-33 and inflammatory bowel disease, mechanistic studies to date, primarily using acute murine models of colitis, have yielded contradicting results, demonstrating both pathogenic and protective roles. We used a well-characterized, spontaneous model of inflammatory bowel disease [ie, SAMP1/YitFc (SAMP) mice] to investigate the role of IL-33 during chronic intestinal inflammation. Our results showed marked eosinophil infiltration into the gut mucosa with increased levels of eotaxins and type 2 helper T-cell (Th2) cytokines as disease progressed and became more severe, which could be reversed upon either eosinophil depletion or blockade of IL-33 signaling. Exogenous IL-33 administration recapitulated these effects in ilea of uninflamed (parental) control AKR/J mice. Human data supported these findings, showing colocalization and up-regulation of IL-33 and eosinophils in the colonic mucosa of inflammatory bowel disease patients versus noninflamed controls. Finally, colonization of commensal flora by fecal material transplantation into germ-free SAMP and the presence of the gut microbiome induced IL-33, subsequent eosinophil infiltration, and mounting of Th2 immune responses, leading to exacerbation of chronic intestinal inflammation characteristic of SAMP mice. These data demonstrate a pathogenic role for IL-33-mediated eosinophilia and activation of Th2 immunity in chronic intestinal inflammation that is dependent on the gut microbiome. Targeting IL-33 may represent a novel therapeutic approach to treat patients with inflammatory bowel disease.
Collapse
Affiliation(s)
- Carlo De Salvo
- Department of Pathology, Case Western Reserve University School of Medicine, Cleveland, Ohio; Gastroenterology and Gastrointestinal Endoscopy Unit, IRCCS Policlinico San Donato, San Donato Milanese and Department of Biomedical Sciences, University of Milan, Milan, Italy
| | - Xiao-Ming Wang
- Department of Pathology, Case Western Reserve University School of Medicine, Cleveland, Ohio
| | - Luca Pastorelli
- Department of Pathology, Case Western Reserve University School of Medicine, Cleveland, Ohio; Gastroenterology and Gastrointestinal Endoscopy Unit, IRCCS Policlinico San Donato, San Donato Milanese and Department of Biomedical Sciences, University of Milan, Milan, Italy
| | - Benedetta Mattioli
- Department of Pathology, Case Western Reserve University School of Medicine, Cleveland, Ohio
| | - Sara Omenetti
- Department of Pathology, Case Western Reserve University School of Medicine, Cleveland, Ohio
| | - Kristine A Buela
- Department of Pathology, Case Western Reserve University School of Medicine, Cleveland, Ohio
| | - Saleem Chowdhry
- Department of Pathology, Case Western Reserve University School of Medicine, Cleveland, Ohio; Department of Medicine, Case Western Reserve University School of Medicine, Cleveland, Ohio
| | - Rekha R Garg
- Department of Pathology, Case Western Reserve University School of Medicine, Cleveland, Ohio
| | - Wendy A Goodman
- Department of Pathology, Case Western Reserve University School of Medicine, Cleveland, Ohio
| | | | - Dirk E Smith
- Inflammation Research, Amgen, Seattle, Washington
| | - Derek W Abbott
- Department of Pathology, Case Western Reserve University School of Medicine, Cleveland, Ohio
| | - Fabio Cominelli
- Department of Medicine, Case Western Reserve University School of Medicine, Cleveland, Ohio
| | - Giorgos Bamias
- Academic Department of Gastroenterology, Kapodistrian University of Athens and Laikon Hospital, Athens, Greece
| | - Wei Xin
- Department of Pathology, Case Western Reserve University School of Medicine, Cleveland, Ohio
| | - James J Lee
- Division of Pulmonary Medicine, Department of Biochemistry and Molecular Biology, Mayo Clinic College of Medicine, Scottsdale, Arizona
| | - Maurizio Vecchi
- Gastroenterology and Gastrointestinal Endoscopy Unit, IRCCS Policlinico San Donato, San Donato Milanese and Department of Biomedical Sciences, University of Milan, Milan, Italy
| | - Theresa T Pizarro
- Department of Pathology, Case Western Reserve University School of Medicine, Cleveland, Ohio; Department of Medicine, Case Western Reserve University School of Medicine, Cleveland, Ohio.
| |
Collapse
|
41
|
Treatment with a Monoclonal Anti-IL-12p40 Antibody Induces Substantial Gut Microbiota Changes in an Experimental Colitis Model. Gastroenterol Res Pract 2016; 2016:4953120. [PMID: 26880890 PMCID: PMC4736578 DOI: 10.1155/2016/4953120] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/24/2015] [Accepted: 11/30/2015] [Indexed: 01/11/2023] Open
Abstract
Background and Aim. Crohn's disease is associated with gut microbiota (GM) dysbiosis. Treatment with the anti-IL-12p40 monoclonal antibody (12p40-mAb) has therapeutic effect in Crohn's disease patients. This study addresses whether a 12p40-mAb treatment influences gut microbiota (GM) composition in mice with adoptive transfer colitis (AdTr-colitis). Methods. AdTr-colitis mice were treated with 12p40-mAb or rat-IgG2a or NaCl from days 21 to 47. Disease was monitored by changes in body weight, stool, endoscopic and histopathology scores, immunohistochemistry, and colonic cytokine/chemokine profiles. GM was characterized through DGGE and 16S rRNA gene-amplicon high-throughput sequencing. Results. Following 12p40-mAb treatment, most clinical and pathological parameters associated with colitis were either reduced or absent. GM was shifted towards a higher Firmicutes-to-Bacteroidetes ratio compared to rat-IgG2a treated mice. Significant correlations between 17 bacterial genera and biological markers were found. The relative abundances of the RF32 order (Alphaproteobacteria) and Akkermansia muciniphila were positively correlated with damaged histopathology and colonic inflammation. Conclusions. Shifts in GM distribution were observed with clinical response to 12p40-mAb treatment, whereas specific GM members correlated with colitis symptoms. Our study implicates that specific changes in GM may be connected with positive clinical outcomes and suggests preventing or correcting GM dysbiosis as a treatment goal in inflammatory bowel disease.
Collapse
|
42
|
Rodriguez-Palacios A, Kodani T, Kaydo L, Pietropaoli D, Corridoni D, Howell S, Katz J, Xin W, Pizarro TT, Cominelli F. Stereomicroscopic 3D-pattern profiling of murine and human intestinal inflammation reveals unique structural phenotypes. Nat Commun 2015; 6:7577. [PMID: 26154811 PMCID: PMC4510646 DOI: 10.1038/ncomms8577] [Citation(s) in RCA: 57] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2014] [Accepted: 05/15/2015] [Indexed: 02/06/2023] Open
Abstract
Histology is fundamental to assess two-dimensional intestinal inflammation; however, inflammatory bowel diseases (IBDs) are often indistinguishable microscopically on the basis of mucosal biopsies. Here, we use stereomicroscopy (SM) to rapidly profile the entire intestinal topography and assess inflammation. We examine the mucosal surface of >700 mice (encompassing >16 strains and various IBD-models), create a profiling catalogue of 3D-stereomicroscopic abnormalities and demonstrate that mice with comparable histological scores display unique sub-clusters of 3D-structure-patterns of IBD pathology, which we call 3D-stereoenterotypes, and which are otherwise indiscernible histologically. We show that two ileal IBD-stereoenterotypes ('cobblestones' versus 'villous mini-aggregation') cluster separately within two distinct mouse lines of spontaneous ileitis, suggesting that host genetics drive unique and divergent inflammatory 3D-structural patterns in the gut. In humans, stereomicroscopy reveals 'liquefaction' lesions and hierarchical fistulous complexes, enriched with clostridia/segmented filamentous bacteria, running under healthy mucosa in Crohn's disease. We suggest that stereomicroscopic (3D-SMAPgut) profiling can be easily implemented and enable the comprehensive study of inflammatory 3D structures, genetics and flora in IBD.
Collapse
Affiliation(s)
- Alex Rodriguez-Palacios
- Division of Gastroenterology and Liver Disease, Department of Medicine, Case Western Reserve University School of Medicine, Cleveland, Ohio 44106, USA
| | - Tomohiro Kodani
- Division of Gastroenterology and Liver Disease, Department of Medicine, Case Western Reserve University School of Medicine, Cleveland, Ohio 44106, USA
| | - Lindsey Kaydo
- Division of Gastroenterology and Liver Disease, Department of Medicine, Case Western Reserve University School of Medicine, Cleveland, Ohio 44106, USA
| | - Davide Pietropaoli
- Division of Gastroenterology and Liver Disease, Department of Medicine, Case Western Reserve University School of Medicine, Cleveland, Ohio 44106, USA
| | - Daniele Corridoni
- Division of Gastroenterology and Liver Disease, Department of Medicine, Case Western Reserve University School of Medicine, Cleveland, Ohio 44106, USA
| | - Scott Howell
- Department of Visual Sciences, Case Western Reserve University School of Medicine, Cleveland, Ohio 44106, USA
| | - Jeffry Katz
- Division of Gastroenterology and Liver Disease, Department of Medicine, Case Western Reserve University School of Medicine, Cleveland, Ohio 44106, USA
- Department of Digestive Health Institute, University Hospitals Case Medical Center, Cleveland, Ohio 44106, USA
| | - Wei Xin
- Department of Digestive Health Institute, University Hospitals Case Medical Center, Cleveland, Ohio 44106, USA
- Department of Pathology, Case Western Reserve University School of Medicine, Cleveland, Ohio 44106, USA
| | - Theresa T. Pizarro
- Division of Gastroenterology and Liver Disease, Department of Medicine, Case Western Reserve University School of Medicine, Cleveland, Ohio 44106, USA
- Department of Pathology, Case Western Reserve University School of Medicine, Cleveland, Ohio 44106, USA
| | - Fabio Cominelli
- Division of Gastroenterology and Liver Disease, Department of Medicine, Case Western Reserve University School of Medicine, Cleveland, Ohio 44106, USA
- Department of Digestive Health Institute, University Hospitals Case Medical Center, Cleveland, Ohio 44106, USA
- Department of Pathology, Case Western Reserve University School of Medicine, Cleveland, Ohio 44106, USA
| |
Collapse
|
43
|
Valatas V, Bamias G, Kolios G. Experimental colitis models: Insights into the pathogenesis of inflammatory bowel disease and translational issues. Eur J Pharmacol 2015; 759:253-64. [PMID: 25814256 DOI: 10.1016/j.ejphar.2015.03.017] [Citation(s) in RCA: 68] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2014] [Revised: 02/03/2015] [Accepted: 03/12/2015] [Indexed: 02/06/2023]
|
44
|
Omenetti S, Brogi M, Goodman WA, Croniger CM, Eid S, Huang AY, Laffi G, Roskams T, Cominelli F, Pinzani M, Pizarro TT. Dysregulated intrahepatic CD4 + T-cell activation drives liver inflammation in ileitis-prone SAMP1/YitFc mice. Cell Mol Gastroenterol Hepatol 2015; 1. [PMID: 26213712 PMCID: PMC4511857 DOI: 10.1016/j.jcmgh.2015.05.007] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
BACKGROUND AND AIMS Liver inflammation is a common extraintestinal manifestation of inflammatory bowel disease (IBD); however, whether liver involvement is a consequence of a primary intestinal defect or results from alternative pathogenic processes remains unclear. Therefore, we sought to determine the potential pathogenic mechanism(s) of concomitant liver inflammation in an established murine model of IBD. METHODS Liver inflammation and immune cell subsets were characterized in ileitis-prone SAMP1/YitFc (SAMP) and AKR/J (AKR) control mice, lymphocyte-depleted SAMP (SAMPxRag-1-/-), and immunodeficient SCID recipient mice receiving SAMP or AKR donor CD4+ T-cells. Proliferation and suppressive capacity of CD4+ T-effector (Teff) and T-regulatory (Treg) cells from gut-associated lymphoid tissue (GALT) and livers of SAMP and AKR mice were measured. RESULTS Surprisingly, prominent inflammation was detected in 4-wk-old SAMP livers, prior to histologic evidence of ileitis, while both disease phenotypes were absent in age-matched AKRs. SAMP liver disease was characterized by abundant infiltration of lymphocytes, required for hepatic inflammation to occur, a Th1-skewed environment, and phenotypically-activated CD4+ T-cells. SAMP intrahepatic CD4+ T-cells also had the ability to induce liver and ileal inflammation when adoptively transferred into SCID recipients, whereas GALT-derived CD4+ T-cells produced milder ileitis, but not liver inflammation. Interestingly, SAMP intrahepatic CD4+ Teff cells showed increased proliferation compared to both SAMP GALT- and AKR liver-derived CD4+ Teff cells, while SAMP intrahepatic Tregs were decreased among CD4+ T-cells and impaired in in vitro suppressive function compared to AKR. CONCLUSIONS Activated intrahepatic CD4+ T-cells induce liver inflammation and contribute to experimental ileitis via locally-impaired hepatic immunosuppressive function.
Collapse
Affiliation(s)
- Sara Omenetti
- Department of Pathology, Case Western Reserve University School of Medicine, Cleveland, Ohio,“DENOThe” Center, University of Florence, Florence, Italy
| | - Marco Brogi
- Department of Pathology, Case Western Reserve University School of Medicine, Cleveland, Ohio,“DENOThe” Center, University of Florence, Florence, Italy
| | - Wendy A. Goodman
- Department of Pathology, Case Western Reserve University School of Medicine, Cleveland, Ohio
| | - Colleen M. Croniger
- Department of Nutrition, Case Western Reserve University School of Medicine, Cleveland, Ohio
| | - Saada Eid
- Department of Pediatrics, Case Western Reserve University School of Medicine, Cleveland, Ohio
| | - Alex Y. Huang
- Department of Pathology, Case Western Reserve University School of Medicine, Cleveland, Ohio,Department of Pediatrics, Case Western Reserve University School of Medicine, Cleveland, Ohio
| | - Giacomo Laffi
- “DENOThe” Center, University of Florence, Florence, Italy
| | - Tania Roskams
- Department of Morphology and Molecular Pathology, University of Leuven, Leuven, Belgium
| | - Fabio Cominelli
- Department of Pathology, Case Western Reserve University School of Medicine, Cleveland, Ohio,“DENOThe” Center, University of Florence, Florence, Italy,Department of Medicine, Case Western Reserve University School of Medicine, Cleveland, Ohio
| | - Massimo Pinzani
- “DENOThe” Center, University of Florence, Florence, Italy,Department of Medicine, Case Western Reserve University School of Medicine, Cleveland, Ohio,UCL Institute for Liver and Digestive Health, Royal Free Hospital, London, United Kingdom
| | - Theresa T. Pizarro
- Department of Pathology, Case Western Reserve University School of Medicine, Cleveland, Ohio,“DENOThe” Center, University of Florence, Florence, Italy,Department of Medicine, Case Western Reserve University School of Medicine, Cleveland, Ohio,Correspondence Address correspondence to: Theresa T. Pizarro, PhD, Department of Pathology, Case Western Reserve University School of Medicine, 2103 Cornell Road, WRB 5534, Cleveland, Ohio 44106. fax: (216) 368-0494.
| |
Collapse
|
45
|
Mikulski Z, Johnson R, Shaked I, Kim G, Nowyhed H, Goodman W, Chodaczek G, Pizarro TT, Cominelli F, Ley K. SAMP1/YitFc mice develop ileitis via loss of CCL21 and defects in dendritic cell migration. Gastroenterology 2015; 148:783-793.e5. [PMID: 25620669 PMCID: PMC4375031 DOI: 10.1053/j.gastro.2015.01.027] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/04/2014] [Revised: 01/07/2015] [Accepted: 01/08/2015] [Indexed: 12/16/2022]
Abstract
BACKGROUND & AIMS The lymphatic chemokine CCL21 is required for dendritic cell (DC) migration from tissues to lymph nodes, which helps establish tolerance to foreign yet harmless antigens. We demonstrate that CCL21 is almost completely absent from SAMP1/YitFc (SAMP) mice, which spontaneously develop chronic ileitis that resembles Crohn's disease, and that DC migration is severely impaired in these mice compared with AKR mice (controls). Toll-like receptor agonists like the Toll-like receptor 7 agonist R848 induce DC maturation and mobilization. METHODS We collected intestinal and other tissues and mesenteric lymph nodes (MLN) from SAMP mice. Expression of CCL21 was measured by quantitative reverse transcription polymerase chain reaction and immunofluorescence analyses; spontaneous and induced migration of DCs were assessed by flow cytometry. We analyzed production of retinoic acid by DCs and their ability to induce development of regulatory T cells. Mice were fed R848 to determine its effects on migration of DCs and development of ileitis in SAMP mice. RESULTS SAMP mice expressed almost no CCL21 in any tissue tested. Their CD11b(+)CD103(+) DCs were defective in migration from the ileal lamina propria to the MLN. DCs from SAMP mice also had a greatly reduced ability to produce retinoic acid and induce development of regulatory T cells compared with control mice. Young SAMP mice had reduced CCL21 expression and decreased DC migration before developing ileitis. Administration of R848 to adult SAMP mice increased migration of DC to the MLN and development of regulatory T cells there, and reduced the severity of ileitis. CONCLUSIONS Loss of CCL21 signaling and DC migration is required for development of ileitis in SAMP mice. Reagents such as R848, which activate DC migration to the MLN, may be developed as treatments for patients with Crohn's disease.
Collapse
Affiliation(s)
- Zbigniew Mikulski
- Division of Inflammation Biology, La Jolla Institute for Allergy and Immunology, La Jolla, CA, 92037
| | - Rebecca Johnson
- Department of Pathology, University of California - San Diego, San Diego, CA, USA
| | - Iftach Shaked
- Division of Inflammation Biology, La Jolla Institute for Allergy and Immunology, La Jolla, CA, 92037
| | - Gisen Kim
- Division of Developmental Immunology, La Jolla Institute for Allergy and Immunology, La Jolla, CA, 92037
| | - Heba Nowyhed
- Division of Inflammation Biology, La Jolla Institute for Allergy and Immunology, La Jolla, CA, 92037
| | - Wendy Goodman
- Department of Pathology, Case Western Reserve University School of Medicine, Cleveland, Ohio
| | - Grzegorz Chodaczek
- Division of Inflammation Biology, La Jolla Institute for Allergy and Immunology, La Jolla, CA, 92037
| | - Theresa T. Pizarro
- Department of Pathology, Case Western Reserve University School of Medicine, Cleveland, Ohio
| | - Fabio Cominelli
- Department of Medicine, Case Western Reserve University School of Medicine, Cleveland, Ohio
| | - Klaus Ley
- Division of Inflammation Biology, La Jolla Institute for Allergy and Immunology, La Jolla, California.
| |
Collapse
|
46
|
Dubé PE, Punit S, Polk DB. Redeeming an old foe: protective as well as pathophysiological roles for tumor necrosis factor in inflammatory bowel disease. Am J Physiol Gastrointest Liver Physiol 2015; 308:G161-70. [PMID: 25477373 PMCID: PMC4312954 DOI: 10.1152/ajpgi.00142.2014] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Tumor necrosis factor (TNF) and its receptors TNFR1 and TNFR2 are major therapeutic targets for inflammatory bowel disease. Research advances have demonstrated that TNF produces pleiotropic responses in the gastrointestinal (GI) tract. Although in excess TNF can contribute to GI pathology, TNF is also a critical protective factor to promote GI homeostasis following injury and inflammation. Genetic studies using candidate and genome-wide association study approaches have identified variants in TNF or its receptors that are associated with Crohn's disease or ulcerative colitis in multiple populations, although the basis for these associations remains unclear. This review considers the efficacy and mechanism of anti-TNF therapies for inflammatory bowel disease to reconcile the many disparate aspects of TNF research and to consider the potential protective effects of TNF signaling in GI health.
Collapse
Affiliation(s)
- Philip E. Dubé
- 1Saban Research Institute, Children's Hospital Los Angeles, Los Angeles, California; ,2Department of Pediatrics, Children's Hospital Los Angeles and University of Southern California Keck School of Medicine, Los Angeles, California; and
| | - Shivesh Punit
- 1Saban Research Institute, Children's Hospital Los Angeles, Los Angeles, California; ,2Department of Pediatrics, Children's Hospital Los Angeles and University of Southern California Keck School of Medicine, Los Angeles, California; and
| | - D. Brent Polk
- 1Saban Research Institute, Children's Hospital Los Angeles, Los Angeles, California; ,2Department of Pediatrics, Children's Hospital Los Angeles and University of Southern California Keck School of Medicine, Los Angeles, California; and ,3Department of Biochemistry and Molecular Biology, University of Southern California Keck School of Medicine, Los Angeles, California
| |
Collapse
|
47
|
Tahara M, Kondo Y, Yokosawa M, Tsuboi H, Takahashi S, Shibayama S, Matsumoto I, Sumida T. T-bet regulates differentiation of forkhead box protein 3+ regulatory T cells in programmed cell death-1-deficient mice. Clin Exp Immunol 2015; 179:197-209. [PMID: 25219397 PMCID: PMC4298397 DOI: 10.1111/cei.12455] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/09/2014] [Indexed: 01/22/2023] Open
Abstract
Programmed cell death-1 (PD-1) plays an important role in peripheral T cell tolerance, but whether or not it affects the differentiation of helper T cell subsets remains elusive. Here we describe the importance of PD-1 in the control of T helper type 1 (Th1) cell activation and development of forkhead box protein 3 (FoxP3(+)) regulatory T cells (Tr(egs)). PD-1-deficient T cell-specific T-bet transgenic (P/T) mice showed growth retardation, and the majority died within 10 weeks. P/T mice showed T-bet over-expression, increased interferon (IFN)-γ production by CD4(+) T cells and significantly low FoxP3(+) T(reg) cell percentage. P/T mice developed systemic inflammation, which was probably induced by augmented Th1 response and low FoxP3(+) T(reg) count. The study identified a unique, previously undescribed role for PD-1 in Th1 and T(reg) differentiation, with potential implication in the development of Th1 cell-targeted therapy.
Collapse
Affiliation(s)
- M Tahara
- Department of Internal Medicine, Faculty of Medicine, University of TsukubaTsukuba, Ibaraki, Japan
| | - Y Kondo
- Department of Internal Medicine, Faculty of Medicine, University of TsukubaTsukuba, Ibaraki, Japan
| | - M Yokosawa
- Department of Internal Medicine, Faculty of Medicine, University of TsukubaTsukuba, Ibaraki, Japan
| | - H Tsuboi
- Department of Internal Medicine, Faculty of Medicine, University of TsukubaTsukuba, Ibaraki, Japan
| | - S Takahashi
- Department of Anatomy and Embryology, Faculty of Medicine, University of TsukubaTsukuba, Ibaraki, Japan
- Laboratory Animal Resource Center, University of TsukubaTsukuba, Ibaraki, Japan
| | - S Shibayama
- Tsukuba Institute ONO Pharmaceutical Co., LTD.Tsukuba, Ibaraki, Japan
| | - I Matsumoto
- Department of Internal Medicine, Faculty of Medicine, University of TsukubaTsukuba, Ibaraki, Japan
| | - T Sumida
- Department of Internal Medicine, Faculty of Medicine, University of TsukubaTsukuba, Ibaraki, Japan
| |
Collapse
|
48
|
Abstract
Intestinal fibrosis is a common outcome in IBD leading to significant morbidity that, to date, has no effective medical treatment. Current knowledge regarding potential mechanism(s) of intestinal fibrogenesis and stricture formation is limited, due in large part to the lack of relevant animal models. Although conventional models possess aspects that are advantageous to study specific mechanisms involved in gut fibrosis, most lack the features of a spontaneously occurring process leading to the formation of intestinal fibrotic lesions following mucosal inflammatory events and the ability to investigate the natural course of disease over time. This review aims to discuss established and novel animal models of gut fibrosis, particularly focusing on the advantages and disadvantages of each model system and the insights they bring to our understanding of the mechanisms of fibrogenesis. In fact, recent enhancements to existing models and the expansion of novel animal models of gut fibrosis is opening up multiple avenues for investigation which should stimulate progress in our mechanistic understanding of intestinal fibrogenesis and facilitate the development of effective pharmacotherapy in an area of significant unmet need.
Collapse
Affiliation(s)
- Carlo De Salvo
- Department of Pathology, Case Western Reserve University School of Medicine, Cleveland, Ohio, USA
| | | | | |
Collapse
|
49
|
Zuo L, Li Y, Wang H, Wu R, Zhu W, Zhang W, Cao L, Gu L, Gong J, Li N, Li J. Cigarette smoking is associated with intestinal barrier dysfunction in the small intestine but not in the large intestine of mice. J Crohns Colitis 2014; 8:1710-22. [PMID: 25205553 DOI: 10.1016/j.crohns.2014.08.008] [Citation(s) in RCA: 58] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/23/2014] [Revised: 07/23/2014] [Accepted: 08/15/2014] [Indexed: 02/07/2023]
Abstract
AIMS To observe the effect of cigarette smoke (CS) on the small bowel and colon in mice and to attempt to explain the potential mechanisms that account for these effects. METHODS Male BALB/c mice age 6-8 weeks were randomly divided into a CS group and a control group (n=10 per group). CS mice were exposed to CS (five cigarettes each time, four times a day for 5 days a week using Hamburg II smoking machine and CS was diluted with air at a ratio of 1:6) for 10 weeks, and control mice were exposed to room air. After 10 weeks, mice were sacrificed for analysis (colon and small bowel). RESULTS CS exposure impaired the intestinal barrier of the small bowel, based on evidence that CS mice exhibited increased intestinal permeability, bacterial translocation, intestinal villi atrophy, damaged tight junctions and abnormal tight junction proteins. These changes were partly mediated through the activated NF-κB (p65) signalling pathway. However, no obvious changes associated with the intestinal barrier were identified in the small bowel of control mice or the colons of control or CS mice. CONCLUSIONS CS is associated with intestinal barrier dysfunction in the small intestine but not in the large intestine of mice.
Collapse
Affiliation(s)
- Lugen Zuo
- Department of General Surgery, Jinling Hospital, Medical School of Nanjing University, Nanjing, China
| | - Yi Li
- Department of General Surgery, Jinling Hospital, Medical School of Nanjing University, Nanjing, China
| | - Honggang Wang
- Department of General Surgery, Jinling Hospital, Medical School of Nanjing University, Nanjing, China
| | - Rong Wu
- Department of General Surgery, Jinling Hospital, Medical School of Nanjing University, Nanjing, China
| | - Weiming Zhu
- Department of General Surgery, Jinling Hospital, Medical School of Nanjing University, Nanjing, China.
| | - Wei Zhang
- Department of General Surgery, Jinling Hospital, Medical School of Nanjing University, Nanjing, China
| | - Lei Cao
- Department of General Surgery, Jinling Hospital, Medical School of Nanjing University, Nanjing, China
| | - Lili Gu
- Department of General Surgery, Jinling Hospital, Medical School of Nanjing University, Nanjing, China
| | - Jianfeng Gong
- Department of General Surgery, Jinling Hospital, Medical School of Nanjing University, Nanjing, China
| | - Ning Li
- Department of General Surgery, Jinling Hospital, Medical School of Nanjing University, Nanjing, China
| | - Jieshou Li
- Department of General Surgery, Jinling Hospital, Medical School of Nanjing University, Nanjing, China
| |
Collapse
|
50
|
Ansary MMU, Ishihara S, Oka A, Kusunoki R, Oshima N, Yuki T, Kawashima K, Maegawa H, Kashiwagi N, Kinoshita Y. Apoptotic cells ameliorate chronic intestinal inflammation by enhancing regulatory B-cell function. Inflamm Bowel Dis 2014; 20:2308-2320. [PMID: 25358066 DOI: 10.1097/mib.0000000000000240] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Apoptosis is a programmed physiological death of unwanted cells, and handling of apoptotic cells (ACs) is thought to have profound effects on immune-mediated disorders. However, there is scant information regarding the role of ACs in intestinal inflammation, in which immune homeostasis is a major concern. To investigate this, we injected ACs into a severe combined immunodeficiency adoptive transfer model of chronic colitis in the presence and absence of cotransferred whole B or regulatory B cell (Breg)-depleted B cells. We also injected syngeneic ACs into AKR/N mice as a control and into milk fat globule-epidermal growth factor 8 knockout mice deficient of phagocytic function. Chronic colitis severity was significantly reduced in the AC as opposed to the phosphate-buffered saline group with cotransferred whole B cells. The AC-mediated effect was lost in the absence of B cells or presence of Breg-depleted B cells. In addition, ACs induced splenic B cells to secrete significantly increased levels of interleukin 10 in AKR/N mice but not milk fat globule-epidermal growth factor 8 knockout mice. Apoptotic leukocytes were induced by reactive oxygen species during granulocyte/monocyte apheresis therapy in rabbits and H2O2-induced apoptotic neutrophils ameliorated mice colitis. Our results indicate that ACs are protective only in the presence of B cells and phagocytosis of ACs induced interleukin 10 producing Bregs. Thus, the ameliorative effect seen in this study might have been exerted by AC-induced Bregs through increased production of the immunosuppressive cytokine interleukin 10, whereas an AC-mediated effect may contribute to the anti-inflammatory effect of granulocyte/monocyte apheresis as a novel therapeutic mechanism for inflammatory bowel disease.
Collapse
MESH Headings
- Animals
- Apoptosis/immunology
- Arthritis, Experimental/immunology
- Arthritis, Experimental/metabolism
- Arthritis, Experimental/pathology
- B-Lymphocytes, Regulatory/immunology
- B-Lymphocytes, Regulatory/metabolism
- B-Lymphocytes, Regulatory/pathology
- Cells, Cultured
- Chronic Disease
- Colitis/immunology
- Colitis/metabolism
- Colitis/pathology
- Enzyme-Linked Immunosorbent Assay
- Flow Cytometry
- Immunoenzyme Techniques
- Inflammation/immunology
- Inflammation/metabolism
- Inflammation/pathology
- Intestinal Mucosa/immunology
- Intestinal Mucosa/metabolism
- Intestinal Mucosa/pathology
- Mice
- Mice, Inbred AKR
- Mice, Inbred C57BL
- Mice, Knockout
- Mice, SCID
- Neutrophils/immunology
- Neutrophils/metabolism
- Neutrophils/pathology
- RNA, Messenger/genetics
- Rabbits
- Reactive Oxygen Species/metabolism
- Real-Time Polymerase Chain Reaction
- Reverse Transcriptase Polymerase Chain Reaction
Collapse
Affiliation(s)
- Md Mesbah Uddin Ansary
- *Department of Internal Medicine II, School of Medicine, Shimane University, Izumo, Japan; †Department of Gastrointestinal Endoscopy, Shimane University Hospital, Izumo, Japan; and ‡Research Division, JIMRO Co. Ltd., Takasaki, Japan
| | | | | | | | | | | | | | | | | | | |
Collapse
|