51
|
Van den Berghe N, Verstockt B, Gils A, Sabino J, Ferrante M, Vermeire S, Declerck P, Thomas D. Tissue Exposure does not Explain Non-Response in Ulcerative Colitis Patients with Adequate Serum Vedolizumab Concentrations. J Crohns Colitis 2021; 15:988-993. [PMID: 33245363 DOI: 10.1093/ecco-jcc/jjaa239] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
BACKGROUND AND AIMS Some patients with ulcerative colitis [UC] do not respond to vedolizumab treatment despite adequate drug exposure in serum. This study aimed to investigate vedolizumab in tissue and questioned whether insufficient tissue exposure could explain non-response in UC patients with adequate serum vedolizumab concentrations. METHODS A paired serum sample and colonic mucosal biopsy was collected from 40 UC patients [20 endoscopic responders, 20 non-responders] at week 14 of vedolizumab treatment. Vedolizumab, soluble [s]-mucosal addressin cell adhesion molecule-1 [MAdCAM-1], s-vascular cell adhesion molecule-1 [VCAM-1] and s-intercellular adhesion molecule-1 [ICAM-1] were measured in serum and/or tissue. Endoscopic response was defined as Mayo endoscopic sub-score ≤1. RESULTS A significant positive correlation was observed between vedolizumab serum and colonic tissue concentrations [ρ = 0.84, p < 0.0001], regardless of the macroscopic inflammatory state of the tissue. Vedolizumab tissue concentrations were lower in non-responders than in responders [0.07 vs 0.11 µg/mg, p = 0.04]. In the subgroup of patients with adequate vedolizumab serum concentrations [>14.6 µg/mL], tissue vedolizumab was not significantly different between responders and non-responders [0.15 vs 0.13 µg/mg; p = 0.92]. Serum sMAdCAM-1 concentrations, but not serum sICAM-1 or sVCAM-1 concentrations, were significantly higher in responders than in non-responders with adequate vedolizumab serum concentrations [1.04 vs 0.83 ng/mL, p = 0.03]. CONCLUSIONS Vedolizumab concentrations in colonic mucosal tissue of UC patients reflect the concentration in serum regardless of the macroscopic inflammatory state of the tissue. Our data show that insufficient tissue exposure does not explain non-response in UC patients with adequate serum vedolizumab concentrations.
Collapse
Affiliation(s)
- Nathalie Van den Berghe
- Laboratory for Therapeutic and Diagnostic Antibodies, Department of Pharmaceutical and Pharmacological Sciences, KU Leuven, Leuven, Belgium
| | - Bram Verstockt
- Department of Gastroenterology and Hepatology, University Hospitals Leuven, KU Leuven, Leuven, Belgium.,Translational Research in Gastrointestinal Disorders, Department of Chronic Diseases, Metabolism and Ageing, KU Leuven, Leuven, Belgium
| | - Ann Gils
- Laboratory for Therapeutic and Diagnostic Antibodies, Department of Pharmaceutical and Pharmacological Sciences, KU Leuven, Leuven, Belgium
| | - João Sabino
- Department of Gastroenterology and Hepatology, University Hospitals Leuven, KU Leuven, Leuven, Belgium.,Translational Research in Gastrointestinal Disorders, Department of Chronic Diseases, Metabolism and Ageing, KU Leuven, Leuven, Belgium
| | - Marc Ferrante
- Department of Gastroenterology and Hepatology, University Hospitals Leuven, KU Leuven, Leuven, Belgium.,Translational Research in Gastrointestinal Disorders, Department of Chronic Diseases, Metabolism and Ageing, KU Leuven, Leuven, Belgium
| | - Séverine Vermeire
- Department of Gastroenterology and Hepatology, University Hospitals Leuven, KU Leuven, Leuven, Belgium.,Translational Research in Gastrointestinal Disorders, Department of Chronic Diseases, Metabolism and Ageing, KU Leuven, Leuven, Belgium
| | - Paul Declerck
- Laboratory for Therapeutic and Diagnostic Antibodies, Department of Pharmaceutical and Pharmacological Sciences, KU Leuven, Leuven, Belgium
| | - Debby Thomas
- Laboratory for Therapeutic and Diagnostic Antibodies, Department of Pharmaceutical and Pharmacological Sciences, KU Leuven, Leuven, Belgium
| |
Collapse
|
52
|
Wiendl M, Becker E, Müller TM, Voskens CJ, Neurath MF, Zundler S. Targeting Immune Cell Trafficking - Insights From Research Models and Implications for Future IBD Therapy. Front Immunol 2021; 12:656452. [PMID: 34017333 PMCID: PMC8129496 DOI: 10.3389/fimmu.2021.656452] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2021] [Accepted: 04/16/2021] [Indexed: 12/12/2022] Open
Abstract
Inflammatory bowel diseases (IBDs), including Crohn's disease (CD) and ulcerative colitis (UC) are multifactorial diseases with still unknown aetiology and an increasing prevalence and incidence worldwide. Despite plentiful therapeutic options for IBDs, the lack or loss of response in certain patients demands the development of further treatments to tackle this unmet medical need. In recent years, the success of the anti-α4β7 antibody vedolizumab highlighted the potential of targeting the homing of immune cells, which is now an important pillar of IBD therapy. Due to its complexity, leukocyte trafficking and the involved molecules offer a largely untapped resource for a plethora of potential therapeutic interventions. In this review, we aim to summarise current and future directions of specifically interfering with immune cell trafficking. We will comment on concepts of homing, retention and recirculation and particularly focus on the role of tissue-derived chemokines. Moreover, we will give an overview of the mode of action of drugs currently in use or still in the pipeline, highlighting their mechanisms and potential to reduce disease burden.
Collapse
Affiliation(s)
- Maximilian Wiendl
- Department of Medicine 1, Deutsches Zentrum Immuntherapie (DZI), University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Emily Becker
- Department of Medicine 1, Deutsches Zentrum Immuntherapie (DZI), University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Tanja M. Müller
- Department of Medicine 1, Deutsches Zentrum Immuntherapie (DZI), University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Caroline J. Voskens
- Department of Dermatology, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Markus F. Neurath
- Department of Medicine 1, Deutsches Zentrum Immuntherapie (DZI), University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Sebastian Zundler
- Department of Medicine 1, Deutsches Zentrum Immuntherapie (DZI), University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| |
Collapse
|
53
|
Okamoto Y, Shikano S. Tyrosine sulfation and O-glycosylation of chemoattractant receptor GPR15 differentially regulate interaction with GPR15L. J Cell Sci 2021; 134:237784. [PMID: 33758080 DOI: 10.1242/jcs.247833] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2020] [Accepted: 03/15/2021] [Indexed: 12/23/2022] Open
Abstract
GPR15 is a G-protein-coupled receptor (GPCR) that directs lymphocyte homing to the colon and skin. Recent studies have identified a chemokine-like protein GPR15L (also known as C10orf99) as a functional ligand of GPR15. In this study, we examined the structural elements that regulate the GPR15-GPR15L interaction with primary focus on post-translational modifications (PTMs) of receptor N-terminus and on the C-terminus of the ligand. Our findings reveal that the GPR15 receptor is sulfated on the N-terminal tyrosine residue(s) and disruption of tyrosine sulfation inhibits binding of GPR15L. In contrast, the disruption of O-glycosylation on the N-terminal threonine or serine residues, or the removal of α2,3-linked sialic acids from O-glycans, enhances the GPR15L binding. Thus, GPR15 represents a unique chemoattractant receptor in which different N-terminal PTMs regulate its ligand binding in a contrasting manner. We further demonstrate that, unlike canonical chemokines, GPR15L activity critically requires its extreme C-terminal residue and that its hydrophobicity may be a key attribute that facilitates an optimal interaction with the receptor. Our results reveal novel insights into chemoattractant receptor-ligand interaction and provide a valid footing for potential intervention targeting the GPR15-GPR15L axis.
Collapse
Affiliation(s)
- Yukari Okamoto
- Department of Biochemistry and Molecular Genetics, University of Illinois at Chicago, Chicago, IL 60607-7170, USA
| | - Sojin Shikano
- Department of Biochemistry and Molecular Genetics, University of Illinois at Chicago, Chicago, IL 60607-7170, USA
| |
Collapse
|
54
|
Qaiyum Z, Lim M, Inman RD. The gut-joint axis in spondyloarthritis: immunological, microbial, and clinical insights. Semin Immunopathol 2021; 43:173-192. [PMID: 33625549 DOI: 10.1007/s00281-021-00845-0] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2020] [Accepted: 01/27/2021] [Indexed: 12/13/2022]
Abstract
The strong genetic and clinical overlaps between spondyloarthritis (SpA) and inflammatory bowel disease (IBD) have placed much needed focus on the gut-joint axis of inflammation in SpA, leading to three key hypotheses that attempt to unravel this complex relationship. The arthritogenic peptide hypothesis and the aberrant cellular trafficking hypothesis have been put forth to rationalize the manner by which the innate and adaptive immune systems cooperate and converge during SpA pathogenesis. The bacterial dysbiosis hypothesis discusses how changes in the microbiome lead to architectural and immunological consequences in SpA. These theories are not mutually exclusive, but can provide an explanation as to why subclinical gut inflammation may sometimes precede joint inflammation in SpA patients, thereby implying a causal relationship. Such investigations will be important in informing therapeutic decisions which may be common to both SpA and IBD. However, these hypotheses can also offer insights for a coincident inflammatory relationship between the gut and the joint, particularly when assessing the immunological players involved. Insights from understanding how these systems might affect the gut and joint differently will be equally imperative to address where the therapeutic differences lie between the two diseases. Collectively, this knowledge has practical implications in predicting the likelihood of IBD development in SpA or presence of coincident SpA-IBD, uncovering novel therapeutic targets, and redesigning currently approved treatments. It is evident that a multidisciplinary approach between the rheumatology and gastroenterology fields cannot be ignored, when it comes to the care of SpA patients at risk of IBD or vice versa.
Collapse
Affiliation(s)
- Zoya Qaiyum
- Schroeder Arthritis Institute, University Health Network, 60 Leonard Avenue, 5, Toronto, Ontario, KD-408, Canada
| | - Melissa Lim
- Schroeder Arthritis Institute, University Health Network, 60 Leonard Avenue, 5, Toronto, Ontario, KD-408, Canada
| | - Robert D Inman
- Schroeder Arthritis Institute, University Health Network, 60 Leonard Avenue, 5, Toronto, Ontario, KD-408, Canada.
- Department of Immunology, University of Toronto, Toronto, Ontario, Canada.
- Department of Medicine, University of Toronto, Toronto, Ontario, Canada.
| |
Collapse
|
55
|
Luzentales-Simpson M, Pang YCF, Zhang A, Sousa JA, Sly LM. Vedolizumab: Potential Mechanisms of Action for Reducing Pathological Inflammation in Inflammatory Bowel Diseases. Front Cell Dev Biol 2021; 9:612830. [PMID: 33614645 PMCID: PMC7887288 DOI: 10.3389/fcell.2021.612830] [Citation(s) in RCA: 44] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Accepted: 01/12/2021] [Indexed: 12/22/2022] Open
Abstract
Inflammatory bowel diseases (IBD), encompassing ulcerative colitis (UC), and Crohn’s disease (CD), are a group of disorders characterized by chronic, relapsing, and remitting, or progressive inflammation along the gastrointestinal tract. IBD is accompanied by massive infiltration of circulating leukocytes into the intestinal mucosa. Leukocytes such as neutrophils, monocytes, and T-cells are recruited to the affected site, exacerbating inflammation and causing tissue damage. Current treatments used to block inflammation in IBD include aminosalicylates, corticosteroids, immunosuppressants, and biologics. The first successful biologic, which revolutionized IBD treatment, targeted the pro-inflammatory cytokine, tumor necrosis factor alpha (TNFα). Infliximab, adalimumab, and other anti-TNF antibodies neutralize TNFα, preventing interactions with its receptors and reducing the inflammatory response. However, up to 40% of people with IBD become unresponsive to anti-TNFα therapy. Thus, more recent biologics have been designed to block leukocyte trafficking to the inflamed intestine by targeting integrins and adhesins. For example, natalizumab targets the α4 chain of integrin heterodimers, α4β1 and α4β7, on leukocytes. However, binding of α4β1 is associated with increased risk for developing progressive multifocal leukoencephalopathy, an often-fatal disease, and thus, it is not used to treat IBD. To target leukocyte infiltration without this life-threatening complication, vedolizumab was developed. Vedolizumab specifically targets the α4β7 integrin and was approved to treat IBD based on the presumption that it would block T-cell recruitment to the intestine. Though vedolizumab is an effective treatment for IBD, some studies suggest that it may not block T-cell recruitment to the intestine and its mechanism(s) of action remain unclear. Vedolizumab may reduce inflammation by blocking recruitment of T-cells, or pro-inflammatory monocytes and dendritic cells to the intestine, and/or vedolizumab may lead to changes in the programming of innate and acquired immune cells dampening down inflammation.
Collapse
Affiliation(s)
- Matthew Luzentales-Simpson
- Division of Gastroenterology, Department of Pediatrics, BC Children's Hospital and the University of British Columbia, Vancouver, BC, Canada
| | - Yvonne C F Pang
- Division of Gastroenterology, Department of Pediatrics, BC Children's Hospital and the University of British Columbia, Vancouver, BC, Canada
| | - Ada Zhang
- Division of Gastroenterology, Department of Pediatrics, BC Children's Hospital and the University of British Columbia, Vancouver, BC, Canada
| | - James A Sousa
- Division of Gastroenterology, Department of Pediatrics, BC Children's Hospital and the University of British Columbia, Vancouver, BC, Canada
| | - Laura M Sly
- Division of Gastroenterology, Department of Pediatrics, BC Children's Hospital and the University of British Columbia, Vancouver, BC, Canada
| |
Collapse
|
56
|
Jefremow A, Neurath MF. SARS-CoV-2 Virus Manifestations in the Gastrointestinal Tract: Therapeutic Implications. Visc Med 2021; 37:63-69. [PMID: 33693046 PMCID: PMC7802000 DOI: 10.1159/000513180] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2020] [Accepted: 11/18/2020] [Indexed: 12/22/2022] Open
Abstract
BACKGROUND About 1 year ago a novel virus - SARS-CoV-2 - began to spread around the world. It can lead to the disease COVID-19, which has caused more than 1 million deaths already. SUMMARY While it was first recognized as a disease leading to pneumonia and lung failure, we know by now that COVID-19 is more complex. COVID-19 is a systemic hyperinflammatory disease affecting not only the lungs, but also many other organs. Especially the gastrointestinal (GI) tract is often involved in COVID-19. KEY MESSAGES This review provides an overview of the different affected organs of the GI tract and offers information on how gastroenterologists should take care of their patients with different GI disorders.
Collapse
Affiliation(s)
- André Jefremow
- Department of Internal Medicine 1, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen, Germany
- Deutsches Zentrum Immuntherapie (DZI), Erlangen, Germany
| | - Markus F. Neurath
- Department of Internal Medicine 1, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen, Germany
- Deutsches Zentrum Immuntherapie (DZI), Erlangen, Germany
| |
Collapse
|
57
|
Andersen AM, Lei MK, Beach SR, Philibert RA. Inflammatory biomarker relationships with helper T cell GPR15 expression and cannabis and tobacco smoking. J Psychosom Res 2021; 141:110326. [PMID: 33310155 PMCID: PMC9045001 DOI: 10.1016/j.jpsychores.2020.110326] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/26/2020] [Revised: 11/30/2020] [Accepted: 12/01/2020] [Indexed: 01/08/2023]
Abstract
OBJECTIVE Smoking is associated with numerous inflammatory and autoimmune conditions. The goal of this study was to examine whether increased expression of G-protein-coupled receptor 15 (GPR15) on helper T cells in smokers could predispose to these conditions through its relationship with inflammatory biomarkers. METHODS We used flow cytometric measurement of GPR15+CD3+CD4+ helper T cells and serum assays for C-reactive protein (CRP) and 17 cytokines drawn from peripheral blood samples from a cohort of n = 62 primarily African American young adults (aged 27-35 years). These variables were examined cross-sectionally in conjunction with serum biomarkers of tobacco (cotinine) and cannabis (tetrahydrocannabinol) use and lifestyle factors potentially impacting immune function in correlational analyses and linear regression models. RESULTS Tobacco and cannabis smoking were strongly associated with increased GPR15 expression on helper T cells (p < 0.001), which was in turn was strongly associated with the ratio of pro-inflammatory to anti-inflammatory cytokines (p < 0.001). Mediation analyses indicated increased GPR15 expression accounted for roughly half of the relationship between smoking variables and pro-inflammatory to anti-inflammatory cytokine balance. CRP was not associated with cannabis or tobacco use or GPR15+ expression, but was associated with body mass index (p < 0.001). These relationships persisted after controlling for lifestyle and medical factors impacting immune function. CONCLUSIONS Increased expression of GPR15 by helper T cells in smokers may mediate some of the relationship between smoking and a pro-inflammatory cytokine milieu. Better understanding of this relationship may help uncover how smoking increases the risk of inflammatory diseases.
Collapse
Affiliation(s)
- Allan M. Andersen
- Department of Psychiatry, University of Iowa, Iowa City, IA 52242,Correspondence: Allan M. Andersen, MD, 500 Newton Road, Iowa City, IA 52246, Tel: (319) 384-4420, Fax: (319) 353-3003,
| | - Man-Kit Lei
- Department of Sociology, University of Georgia, Athens, GA 30602,Center for Family Research, University of Georgia, Athens, GA 30602
| | - Steven R.H. Beach
- Center for Family Research, University of Georgia, Athens, GA 30602,Department of Psychology, University of Georgia, Athens, GA 30602
| | - Robert A. Philibert
- Department of Psychiatry, University of Iowa, Iowa City, IA 52242,Behavioral Diagnostics, Coralville, Iowa 52241
| |
Collapse
|
58
|
Giannoudaki E, Gargan S, Hussey S, Long A, Walsh PT. Opportunities to Target T Cell Trafficking in Pediatric Inflammatory Bowel Disease. Front Pediatr 2021; 9:640497. [PMID: 33816403 PMCID: PMC8012547 DOI: 10.3389/fped.2021.640497] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/11/2020] [Accepted: 02/08/2021] [Indexed: 12/12/2022] Open
Abstract
T cell subsets are considered central orchestrators of inflammation and homeostasis in the intestine and are established targets for the treatment of inflammatory bowel disease. While approaches aimed at the neutralization of T cell effector cytokines have provided significant benefits for pediatric and adult patients, more recent strategies aimed at inhibiting the infiltration of pathogenic T cell subsets have also emerged. In this review, we describe current knowledge surrounding the function of T cell subsets in pediatric inflammatory bowel disease and outline approaches aimed at targeting T cell trafficking to the intestine which may represent a new treatment option for pediatric inflammatory bowel disease.
Collapse
Affiliation(s)
- Eirini Giannoudaki
- National Children's Research Center, Children's Health Ireland (CHI) Crumlin, Dublin, Ireland.,Trinity Translational Medicine Institute, Trinity College Dublin, Dublin, Ireland
| | - Siobhan Gargan
- National Children's Research Center, Children's Health Ireland (CHI) Crumlin, Dublin, Ireland.,Trinity Translational Medicine Institute, Trinity College Dublin, Dublin, Ireland
| | - Seamus Hussey
- National Children's Research Center, Children's Health Ireland (CHI) Crumlin, Dublin, Ireland.,Department of Paediatrics, Royal College of Surgeons of Ireland, Dublin, Ireland
| | - Aideen Long
- National Children's Research Center, Children's Health Ireland (CHI) Crumlin, Dublin, Ireland.,Trinity Translational Medicine Institute, Trinity College Dublin, Dublin, Ireland
| | - Patrick T Walsh
- National Children's Research Center, Children's Health Ireland (CHI) Crumlin, Dublin, Ireland.,Trinity Translational Medicine Institute, Trinity College Dublin, Dublin, Ireland
| |
Collapse
|
59
|
Abstract
The coronavirus disease 2019 (COVID-19) pandemic is an ongoing global health crisis causing major challenges for clinical care in patients with gastrointestinal diseases. Although triggering of anti-viral immune responses is essential for clearance of infection, some patients have severe lung inflammation and multiorgan failure due to marked immune cell dysregulation and cytokine storm syndrome. Importantly, the activation of cytotoxic follicular helper T cells and a reduction of regulatory T cells have a crucial, negative prognostic role. These findings lead to the question of whether immunosuppressive and biologic therapies for gastrointestinal diseases affect the incidence or prognosis of COVID-19 and, thus, whether they should be adjusted to prevent or affect the course of the disease. In this Review, data on the use of such therapies are discussed with a primary focus on inflammatory bowel disease, autoimmune hepatitis and liver transplantation. In particular, the roles of corticosteroids, classic immunosuppressive agents (such as thiopurines and mycophenolate mofetil), small molecules (such as Janus kinase (JAK) inhibitors), and biologic agents (such as tumour necrosis factor (TNF) blockers, vedolizumab and ustekinumab) are reviewed. Finally, the use of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) vaccines for the prevention of infection in patients with gastrointestinal diseases and concomitant immunosuppressive or biologic therapy will be discussed.
Collapse
|
60
|
Kirby C, Baig A, Avlasevich SL, Torous DK, Tian S, Singh P, Bemis JC, Saubermann LJ, Dertinger SD. Dextran sulfate sodium mouse model of inflammatory bowel disease evaluated for systemic genotoxicity via blood micronucleus and Pig-a gene mutation assays. Mutagenesis 2020; 35:161-167. [PMID: 32050029 DOI: 10.1093/mutage/geaa006] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2019] [Accepted: 01/24/2020] [Indexed: 12/31/2022] Open
Abstract
Inflammatory bowel disease (IBD) is an important risk factor for gastrointestinal cancers. Inflammation and other carcinogenesis-related effects at distal, tissue-specific sites require further study. In order to better understand if systemic genotoxicity is associated with IBD, we exposed mice to dextran sulfate sodium salt (DSS) and measured the incidence of micronucleated cells (MN) and Pig-a mutant phenotype cells in blood erythrocyte populations. In one study, 8-week-old male CD-1 mice were exposed to 0, 1, 2, 3 or 4% w/v DSS in drinking water. The 4-week in-life period was divided into four 1-week intervals-alternately on then off DSS treatment. Low volume blood samples were collected for MN analysis at the end of each week, and cardiac blood samples were collected at the end of the 4-week period for Pig-a analyses. The two highest doses of DSS were observed to induce significant increases in reticulocyte frequencies. Even so, no statistically significant treatment-related effects on the genotoxicity biomarkers were evident. While one high-dose mouse showed modestly elevated MN frequencies during the DSS treatment cycles, it also exhibited exceptionally high reticulocyte frequencies (e.g. 18.7% at the end of the second DSS cycle). In a second study, mice were treated with 0 or 4% DSS for 9-18 consecutive days. Exposure was continued until rectal bleeding or morbidity was evident, at which point the treatment was terminated and blood was collected for MN analysis. The Pig-a assay was conducted on samples collected 29 days after the start of treatment. The initial blood specimens showed highly elevated reticulocyte frequencies in DSS-exposed mice (mean ± SEM = 1.75 ± 0.10% vs. 13.04 ± 3.66% for 0 vs. 4% mice, respectively). Statistical analyses showed no treatment-related effect on MN or Pig-a mutant frequencies. Even so, the incidence of MN versus reticulocytes in the DSS-exposed mice were positively correlated (linear fit R2 = 0.657, P = 0.0044). Collectively, these results suggest that in the case of the DSS CD-1 mouse model, systemic effects include stress erythropoiesis but not remarkable genotoxicity. To the extent MN may have been slightly elevated in a minority of individual mice, these effects appear to be secondary, likely attributable to stimulated erythropoiesis.
Collapse
Affiliation(s)
| | - Ayesha Baig
- Department of Pediatrics, University of Rochester Medical Center, Rochester, NY
| | | | | | | | | | | | | | | |
Collapse
|
61
|
Recent advances in inflammatory bowel disease therapy. Eur J Pharm Sci 2020; 155:105550. [DOI: 10.1016/j.ejps.2020.105550] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2020] [Revised: 07/29/2020] [Accepted: 09/11/2020] [Indexed: 02/06/2023]
|
62
|
|
63
|
Kobayashi T, Siegmund B, Le Berre C, Wei SC, Ferrante M, Shen B, Bernstein CN, Danese S, Peyrin-Biroulet L, Hibi T. Ulcerative colitis. Nat Rev Dis Primers 2020; 6:74. [PMID: 32913180 DOI: 10.1038/s41572-020-0205-x] [Citation(s) in RCA: 885] [Impact Index Per Article: 177.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 07/21/2020] [Indexed: 02/07/2023]
Abstract
Ulcerative colitis (UC) is a chronic inflammatory bowel disease of unknown aetiology affecting the colon and rectum. Multiple factors, such as genetic background, environmental and luminal factors, and mucosal immune dysregulation, have been suggested to contribute to UC pathogenesis. UC has evolved into a global burden given its high incidence in developed countries and the substantial increase in incidence in developing countries. An improved understanding of the mechanisms underlying UC has led to the emergence of new treatments. Since the early 2000s, anti-tumour necrosis factor (TNF) treatment has significantly improved treatment outcomes. Advances in medical treatments have enabled a paradigm shift in treatment goals from symptomatic relief to endoscopic and histological healing to achieve better long-term outcomes and, consequently, diagnostic modalities have also been improved to monitor disease activity more tightly. Despite these improvements in patient care, a substantial proportion of patients, for example, those who are refractory to medical treatment or those who develop colitis-associated colorectal dysplasia or cancer, still require restorative proctocolectomy. The development of novel drugs and improvement of the treatment strategy by implementing personalized medicine are warranted to achieve optimal disease control. However, delineating the aetiology of UC is necessary to ultimately achieve disease cure.
Collapse
Affiliation(s)
- Taku Kobayashi
- Center for Advanced IBD Research and Treatment, Kitasato University Kitasato Institute Hospital, Tokyo, Japan.
| | - Britta Siegmund
- Division of Gastroenterology, Infectiology and Rheumatology, Charite-Universitatsmedizin, Berlin, Germany
| | - Catherine Le Berre
- Department of Gastroenterology, Nancy University Hospital, Inserm U1256 NGERE, Lorraine University, Lorraine, France
| | - Shu Chen Wei
- Department of Internal Medicine, National Taiwan University Hospital, Taipei, Taiwan
| | - Marc Ferrante
- Department of Gastroenterology and Hepatology, University Hospitals Leuven, KU Leuven, Leuven, Belgium
| | - Bo Shen
- Center for Inflammatory Bowel Diseases, Columbia University Irving Medical Center-New York Presbyterian Hospital, New York, NY, USA
| | - Charles N Bernstein
- University of Manitoba IBD Clinical and Research Centre and Department of Internal Medicine, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, Canada
| | - Silvio Danese
- Humanitas Clinical and Research Center - IRCCS - and Humanitas University, Department of Biomedical Sciences, Milan, Italy
| | - Laurent Peyrin-Biroulet
- Department of Gastroenterology, Nancy University Hospital, Inserm U1256 NGERE, Lorraine University, Lorraine, France
| | - Toshifumi Hibi
- Center for Advanced IBD Research and Treatment, Kitasato University Kitasato Institute Hospital, Tokyo, Japan.
| |
Collapse
|
64
|
Revisiting the gut-joint axis: links between gut inflammation and spondyloarthritis. Nat Rev Rheumatol 2020; 16:415-433. [PMID: 32661321 DOI: 10.1038/s41584-020-0454-9] [Citation(s) in RCA: 113] [Impact Index Per Article: 22.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/05/2020] [Indexed: 02/07/2023]
Abstract
Gut inflammation is strongly associated with spondyloarthritis (SpA), as exemplified by the high prevalence of inflammatory bowel disease (IBD) and the even higher occurrence of subclinical gut inflammation in patients with SpA. The gut-joint axis of inflammation in SpA is further reinforced by similarities in immunopathogenesis at both anatomical sites and by the clinical success of therapies blocking TNF and IL-23 in IBD and in some forms of SpA. Many genetic risk factors are shared between SpA and IBD, and changes in the composition of gut microbiota are seen in both diseases. Current dogma is that inflammation in SpA initiates in the gut and leads to joint inflammation; however, although conceptually attractive, some research does not support this causal relationship. For example, therapies targeting IL-17A are efficacious in the joint but not the gut, and interfering with gut trafficking by targeting molecules such as α4β7 in IBD can lead to onset or flares of SpA. Several important knowledge gaps remain that must be addressed in future studies. Determining the true nature of the gut-joint axis has real-world implications for the treatment of patients with co-incident IBD and SpA and for the repurposing of therapeutics from one disease to the other.
Collapse
|
65
|
Abstract
The current coronavirus pandemic is an ongoing global health crisis due to COVID-19, caused by severe acute respiratory syndrome coronavirus 2. Although COVID-19 leads to little or mild flu-like symptoms in the majority of affected patients, the disease may cause severe, frequently lethal complications such as progressive pneumonia, acute respiratory distress syndrome and organ failure driven by hyperinflammation and a cytokine storm syndrome. This situation causes various major challenges for gastroenterology. In the context of IBD, several key questions arise. For instance, it is an important question to understand whether patients with IBD (eg, due to intestinal ACE2 expression) might be particularly susceptible to COVID-19 and the cytokine release syndrome associated with lung injury and fatal outcomes. Another highly relevant question is how to deal with immunosuppression and immunomodulation during the current pandemic in patients with IBD and whether immunosuppression affects the progress of COVID-19. Here, the current understanding of the pathophysiology of COVID-19 is reviewed with special reference to immune cell activation. Moreover, the potential implications of these new insights for immunomodulation and biological therapy in IBD are discussed.
Collapse
Affiliation(s)
- Markus F Neurath
- First Department of Medicine and Deutsches Zentrum Immuntherapie DZI, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen 91052, Germany
- Deutsches Zentrum Immuntherapie (DZI), Erlangen, Germany
| |
Collapse
|
66
|
Controlling leukocyte trafficking in IBD. Pharmacol Res 2020; 159:105050. [PMID: 32598943 DOI: 10.1016/j.phrs.2020.105050] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/17/2020] [Revised: 06/24/2020] [Accepted: 06/24/2020] [Indexed: 12/11/2022]
Abstract
Inflammatory bowel disease (IBD) is characterized by the accumulation of immune cells, myeloid cells and lymphocytes in the inflamed intestine. The presence and persistence of these cells, together with the production of pro-inflammatory mediators, perpetuate intestinal inflammation in both ulcerative colitis and Crohn's disease. Thus, blockade of leukocyte migration to the intestine is a main strategy used to control the disease and alleviate symptoms. Vedolizumab is the only anti-integrin drug approved for the treatment of IBD but several other drugs also targeting integrins, chemokines or receptors involved in leukocyte intestinal trafficking are under development and investigated for their efficacy and safety in IBD. The challenge now is to better understand the specific mechanism of action underlying each drug and to identify biomarkers that would guide drug selection in the individual patient.
Collapse
|
67
|
Zhang H, Zheng Y, Pan Y, Lin C, Wang S, Yan Z, Lu L, Ge G, Li J, Zeng YA, Chen J. A mutation that blocks integrin α 4β 7 activation prevents adaptive immune-mediated colitis without increasing susceptibility to innate colitis. BMC Biol 2020; 18:64. [PMID: 32522281 PMCID: PMC7288534 DOI: 10.1186/s12915-020-00784-6] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2020] [Accepted: 04/21/2020] [Indexed: 01/08/2023] Open
Abstract
Background β7 integrins are responsible for the efficient recruitment of lymphocytes from the blood and their retention in gut-associated lymphoid tissues. Integrin α4β7 binds MAdCAM-1, mediating rolling adhesion of lymphocytes on blood vessel walls when inactive and firm adhesion when activated, thereby controlling two critical steps of lymphocyte homing to the gut. By contrast, integrin αEβ7 mediates the adhesion of lymphocytes to gut epithelial cells by interacting with E-cadherin. Integrin β7 blocking antibodies have shown efficacy in clinical management of inflammatory bowel disease (IBD); however, fully blocking β7 function leads to the depletion of colonic regulatory T (Treg) cells and exacerbates dextran sulfate sodium (DSS)-induced colitis by evoking aberrant innate immunity, implying its potential adverse effect for IBD management. Thus, a better therapeutic strategy targeting integrin β7 is required to avoid this adverse effect. Results Herein, we inhibited integrin α4β7 activation in vivo by creating mice that carry in their integrin β7 gene a mutation (F185A) which from structural studies is known to lock α4β7 in its resting state. Lymphocytes from β7-F185A knock-in (KI) mice expressed α4β7 integrins that could not be activated by chemokines and showed significantly impaired homing to the gut. The β7-F185A mutation did not inhibit αEβ7 activation, but led to the depletion of αEβ7+ lymphocytes in the spleen and a significantly reduced population of αEβ7+ lymphocytes in the gut of KI mice. β7-F185A KI mice were resistant to T cell transfer-induced chronic colitis, but did not show an increased susceptibility to DSS-induced innate colitis, the adverse effect of fully blocking β7 function. Conclusions Our findings demonstrate that specific inhibition of integrin α4β7 activation is a potentially better strategy than fully blocking α4β7 function for IBD treatment.
Collapse
Affiliation(s)
- Hailong Zhang
- State Key Laboratory of Cell Biology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences; University of Chinese Academy of Sciences, 320 YueYang Road, Shanghai, 200031, China
| | - Yajuan Zheng
- State Key Laboratory of Cell Biology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences; University of Chinese Academy of Sciences, 320 YueYang Road, Shanghai, 200031, China.
| | - Youdong Pan
- State Key Laboratory of Cell Biology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences; University of Chinese Academy of Sciences, 320 YueYang Road, Shanghai, 200031, China
| | - Changdong Lin
- State Key Laboratory of Cell Biology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences; University of Chinese Academy of Sciences, 320 YueYang Road, Shanghai, 200031, China
| | - Shihui Wang
- State Key Laboratory of Cell Biology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences; University of Chinese Academy of Sciences, 320 YueYang Road, Shanghai, 200031, China
| | - Zhanjun Yan
- Department of Orthopedics, the First People's Hospital of Wujiang District, 169 GongYuan Road, Suzhou, 215200, China
| | - Ling Lu
- State Key Laboratory of Cell Biology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences; University of Chinese Academy of Sciences, 320 YueYang Road, Shanghai, 200031, China
| | - Gaoxiang Ge
- State Key Laboratory of Cell Biology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences; University of Chinese Academy of Sciences, 320 YueYang Road, Shanghai, 200031, China.,School of Life Science, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou, 310024, China
| | - Jinsong Li
- State Key Laboratory of Cell Biology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences; University of Chinese Academy of Sciences, 320 YueYang Road, Shanghai, 200031, China.,School of Life Science, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou, 310024, China
| | - Yi Arial Zeng
- State Key Laboratory of Cell Biology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences; University of Chinese Academy of Sciences, 320 YueYang Road, Shanghai, 200031, China.,School of Life Science, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou, 310024, China
| | - Jianfeng Chen
- State Key Laboratory of Cell Biology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences; University of Chinese Academy of Sciences, 320 YueYang Road, Shanghai, 200031, China. .,School of Life Science, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou, 310024, China.
| |
Collapse
|
68
|
Allner C, Melde M, Becker E, Fuchs F, Mühl L, Klenske E, Müller L, Morgenstern N, Fietkau K, Hirschmann S, Atreya R, Atreya I, Neurath MF, Zundler S. Baseline levels of dynamic CD4 + T cell adhesion to MAdCAM-1 correlate with clinical response to vedolizumab treatment in ulcerative colitis: a cohort study. BMC Gastroenterol 2020; 20:103. [PMID: 32293299 PMCID: PMC7158080 DOI: 10.1186/s12876-020-01253-8] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/19/2019] [Accepted: 03/30/2020] [Indexed: 12/16/2022] Open
Abstract
Background While the number of therapeutic options for treating inflammatory bowel diseases (IBD) is increasing, evidence for rational treatment decisions is scarce in many cases. In particular, appropriate biomarkers to predict the response to the anti-α4β7 integrin antibody vedolizumab are currently lacking. Methods We performed a cohort study with 21 patients suffering from ulcerative colitis (UC), in which first-time treatment with vedolizumab was initiated. CD4+ T cells were isolated from the peripheral blood and dynamic adhesion to recombinant mucosal vascular addressin cell adhesion molecule (MAdCAM-)1 in vitro as well as the effect of vedolizumab on such adhesion in vitro was determined. The expression of α4β1 integrin on peripheral blood CD4+ T cells was quantified by flow cytometry. Electronic patient records were reviewed to determine clinical response to vedolizumab. Results Dynamic adhesion of peripheral blood CD4+ T cells to MAdCAM-1 and the reduction of adhesion following vedolizumab treatment in vitro were higher and the change in α4β1 expression on CD4+ T cells was different in vedolizumab responders and non-responders. Responders could be identified with high specificity and positive-predictive value. Conclusions Determining dynamic adhesion of CD4+ T cells to MAdCAM-1 and the in vitro response to vedolizumab before treatment initiation or dynamic integrin regulation in the early course of treatment seem to be promising tools to predict the clinical response to vedolizumab therapy. Larger prospective studies are warranted.
Collapse
Affiliation(s)
- Clarissa Allner
- Department of Medicine 1, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Kussmaul Campus for Medical Research & Translational Research Center, Ulmenweg 18, 91054, Erlangen, Germany
| | - Michaela Melde
- Department of Medicine 1, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Kussmaul Campus for Medical Research & Translational Research Center, Ulmenweg 18, 91054, Erlangen, Germany
| | - Emily Becker
- Department of Medicine 1, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Kussmaul Campus for Medical Research & Translational Research Center, Ulmenweg 18, 91054, Erlangen, Germany
| | - Friederike Fuchs
- Department of Medicine 1, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Kussmaul Campus for Medical Research & Translational Research Center, Ulmenweg 18, 91054, Erlangen, Germany
| | - Laura Mühl
- Department of Medicine 1, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Kussmaul Campus for Medical Research & Translational Research Center, Ulmenweg 18, 91054, Erlangen, Germany
| | - Entcho Klenske
- Department of Medicine 1, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Kussmaul Campus for Medical Research & Translational Research Center, Ulmenweg 18, 91054, Erlangen, Germany
| | - Lisa Müller
- Department of Medicine 1, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Kussmaul Campus for Medical Research & Translational Research Center, Ulmenweg 18, 91054, Erlangen, Germany
| | - Nadine Morgenstern
- Department of Medicine 1, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Kussmaul Campus for Medical Research & Translational Research Center, Ulmenweg 18, 91054, Erlangen, Germany
| | - Konstantin Fietkau
- Department of Medicine 1, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Kussmaul Campus for Medical Research & Translational Research Center, Ulmenweg 18, 91054, Erlangen, Germany
| | - Simon Hirschmann
- Department of Medicine 1, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Kussmaul Campus for Medical Research & Translational Research Center, Ulmenweg 18, 91054, Erlangen, Germany
| | - Raja Atreya
- Department of Medicine 1, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Kussmaul Campus for Medical Research & Translational Research Center, Ulmenweg 18, 91054, Erlangen, Germany
| | - Imke Atreya
- Department of Medicine 1, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Kussmaul Campus for Medical Research & Translational Research Center, Ulmenweg 18, 91054, Erlangen, Germany
| | - Markus F Neurath
- Department of Medicine 1, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Kussmaul Campus for Medical Research & Translational Research Center, Ulmenweg 18, 91054, Erlangen, Germany
| | - Sebastian Zundler
- Department of Medicine 1, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Kussmaul Campus for Medical Research & Translational Research Center, Ulmenweg 18, 91054, Erlangen, Germany.
| |
Collapse
|
69
|
de Wolf ACMT, Herberts CA, Hoefnagel MHN. Dawn of Monitoring Regulatory T Cells in (Pre-)clinical Studies: Their Relevance Is Slowly Recognised. Front Med (Lausanne) 2020; 7:91. [PMID: 32300597 PMCID: PMC7142310 DOI: 10.3389/fmed.2020.00091] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2019] [Accepted: 03/03/2020] [Indexed: 12/14/2022] Open
Abstract
Regulatory T cells (Tregs) have a prominent role in the control of immune homeostasis. Pharmacological impact on their activity or balance with effector T cells could contribute to (impaired) clinical responses or adverse events. Monitoring treatment-related effects on T cell subsets may therefore be part of (pre-)clinical studies for medicinal products. However, the extent of immune monitoring performed in studies for marketing authorisation and the degree of correspondence with data available in the public domain is not known. We evaluated the presence of T cell immunomonitoring in 46 registration dossiers of monoclonal antibodies indicated for immune-related disorders and published scientific papers. We found that the depth of Treg analysis in registration dossiers was rather small. Nevertheless, data on treatment-related Treg effects are available in public academia-driven studies (post-registration) and suggest that Tregs may act as a biomarker for clinical responses. However, public data are fragmented and obtained with heterogeneity of experimental approaches from a diversity of species and tissues. To reveal the potential added value of T cell (and particular Treg) evaluation in (pre-)clinical studies, more cell-specific data should be acquired, at least for medicinal products with an immunomodulatory mechanism. Therefore, extensive analysis of T cell subset contribution to clinical responses and the relevance of treatment-induced changes in their levels is needed. Preferably, industry and academia should work together to obtain these data in a standardised manner and to enrich our knowledge about T cell activity in disease pathogenesis and therapies. This will ultimately elucidate the necessity of T cell subset monitoring in the therapeutic benefit-risk assessment.
Collapse
|
70
|
Zhang S, Liang W, Luo L, Sun S, Wang F. The role of T cell trafficking in CTLA-4 blockade-induced gut immunopathology. BMC Biol 2020; 18:29. [PMID: 32183814 PMCID: PMC7079427 DOI: 10.1186/s12915-020-00765-9] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2019] [Accepted: 03/05/2020] [Indexed: 12/18/2022] Open
Abstract
Background Immune checkpoint inhibitor (ICPI) can augment the anti-tumour response by blocking negative immunoregulators with monoclonal antibodies. The anti-cytotoxic T lymphocyte-associated antigen-4 (CTLA-4) antibody is the first ICPI which has shown remarkable benefits in the clinical treatment of cancers. However, the increased activity of the immune system also causes some side effects called immune-related adverse events (irAEs). Colitis is one of the most common irAEs related to anti-CTLA-4 immunotherapy. Results We identified that CD4+ T cells were the primary responders in CTLA-4 blockade and that the expansion of gut-homing CD4+ T cells by anti-CTLA-4 therapy was independent of CD103. We used dextran sulfate sodium (DSS)-induced colitis mice as our model and tested the possibility of using a trafficking-blocking antibody to treat anti-CTLA-4 antibody-induced irAEs. We found that blocking T cell homing increased colitis severity in the context of CTLA-4 blockade and that gut-trafficking blockade had different effects on different Th subsets and could facilitate the proliferation of Th17 cells in the lamina propria (LP). Conclusions Our data reveals the fundamental mechanism underlying trafficking-blocking antibody therapy for CTLA-4 blockade-induced colitis and provide a caution in regard to apply trafficking-blocking antibody treatment under CTLA-4 blockade condition.
Collapse
Affiliation(s)
- Shashuang Zhang
- Research Center of Translational Medicine, Shanghai Children's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,The Center for Microbiota and Immunological Diseases, Shanghai General Hospital, Shanghai Institute of Immunology, Department of Immunology and Microbiology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Wenhua Liang
- Research Center of Translational Medicine, Shanghai Children's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,The Center for Microbiota and Immunological Diseases, Shanghai General Hospital, Shanghai Institute of Immunology, Department of Immunology and Microbiology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Lingjie Luo
- Research Center of Translational Medicine, Shanghai Children's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,The Center for Microbiota and Immunological Diseases, Shanghai General Hospital, Shanghai Institute of Immunology, Department of Immunology and Microbiology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Shan Sun
- Research Center of Translational Medicine, Shanghai Children's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,The Center for Microbiota and Immunological Diseases, Shanghai General Hospital, Shanghai Institute of Immunology, Department of Immunology and Microbiology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Feng Wang
- Research Center of Translational Medicine, Shanghai Children's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China. .,The Center for Microbiota and Immunological Diseases, Shanghai General Hospital, Shanghai Institute of Immunology, Department of Immunology and Microbiology, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| |
Collapse
|
71
|
Schleier L, Wiendl M, Heidbreder K, Binder MT, Atreya R, Rath T, Becker E, Schulz-Kuhnt A, Stahl A, Schulze LL, Ullrich K, Merz SF, Bornemann L, Gunzer M, Watson AJM, Neufert C, Atreya I, Neurath MF, Zundler S. Non-classical monocyte homing to the gut via α4β7 integrin mediates macrophage-dependent intestinal wound healing. Gut 2020; 69:252-263. [PMID: 31092589 DOI: 10.1136/gutjnl-2018-316772] [Citation(s) in RCA: 81] [Impact Index Per Article: 16.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/09/2018] [Revised: 04/12/2019] [Accepted: 04/17/2019] [Indexed: 12/12/2022]
Abstract
OBJECTIVE To study the role of α4β7 integrin for gut homing of monocytes and to explore the biological consequences of therapeutic α4β7 inhibition with regard to intestinal wound healing. DESIGN We studied the expression of homing markers on monocyte subsets in the peripheral blood and on macrophage subsets in the gut of patients with IBD and controls with flow cytometry and immunohistochemistry. Integrin function was addressed with dynamic adhesion assays and in vivo gut homing assays. In vivo wound healing was studied in mice deficient for or depleted of α4β7 integrin. RESULTS Classical and non-classical monocytes were clearly dichotomous regarding homing marker expression including relevant expression of α4β7 integrin on human and mouse non-classical monocytes but not on classical monocytes. Monocyte-expressed α4β7 integrin was functionally important for dynamic adhesion to mucosal vascular addressin cell adhesion molecule 1 and in vivo gut homing. Impaired α4β7-dependent gut homing was associated with reduced (effect size about 20%) and delayed wound healing and suppressed perilesional presence of wound healing macrophages. Non-classical monocytes in the peripheral blood were increased in patients with IBD under clinical treatment with vedolizumab. CONCLUSION In addition to reported effects on lymphocytes, anti-α4β7 therapy in IBD also targets non-classical monocytes. Impaired gut homing of such monocytes might lead to a reduction of wound healing macrophages and could potentially explain increased rates of postoperative complications in vedolizumab-treated patients, which have been observed in some studies.
Collapse
Affiliation(s)
- Lena Schleier
- Department of Medicine 1, Kussmaul Campus for Medical Research & Translational Research Center, University of Erlangen-Nuremberg, Erlangen, Germany
| | - Maximilian Wiendl
- Department of Medicine 1, Kussmaul Campus for Medical Research & Translational Research Center, University of Erlangen-Nuremberg, Erlangen, Germany
| | - Karin Heidbreder
- Department of Medicine 1, Kussmaul Campus for Medical Research & Translational Research Center, University of Erlangen-Nuremberg, Erlangen, Germany
| | - Marie-Theres Binder
- Department of Medicine 1, Kussmaul Campus for Medical Research & Translational Research Center, University of Erlangen-Nuremberg, Erlangen, Germany
| | - Raja Atreya
- Department of Medicine 1, Kussmaul Campus for Medical Research & Translational Research Center, University of Erlangen-Nuremberg, Erlangen, Germany
| | - Timo Rath
- Department of Medicine 1, Kussmaul Campus for Medical Research & Translational Research Center, University of Erlangen-Nuremberg, Erlangen, Germany
| | - Emily Becker
- Department of Medicine 1, Kussmaul Campus for Medical Research & Translational Research Center, University of Erlangen-Nuremberg, Erlangen, Germany
| | - Anja Schulz-Kuhnt
- Department of Medicine 1, Kussmaul Campus for Medical Research & Translational Research Center, University of Erlangen-Nuremberg, Erlangen, Germany
| | - Annette Stahl
- Department of Medicine 1, Kussmaul Campus for Medical Research & Translational Research Center, University of Erlangen-Nuremberg, Erlangen, Germany
| | - Lisa Lou Schulze
- Department of Medicine 1, Kussmaul Campus for Medical Research & Translational Research Center, University of Erlangen-Nuremberg, Erlangen, Germany
| | - Karen Ullrich
- Department of Medicine 1, Kussmaul Campus for Medical Research & Translational Research Center, University of Erlangen-Nuremberg, Erlangen, Germany
| | - Simon F Merz
- Institute of Experimental Immunology and Imaging, University Duisburg-Essen and University Hospital Essen, Essen, Germany
| | - Lea Bornemann
- Institute of Experimental Immunology and Imaging, University Duisburg-Essen and University Hospital Essen, Essen, Germany
| | - Matthias Gunzer
- Institute of Experimental Immunology and Imaging, University Duisburg-Essen and University Hospital Essen, Essen, Germany
| | - Alastair J M Watson
- Norwich Medical School, Norwich Research Park, University of East Anglia, Norwich, UK
| | - Clemens Neufert
- Department of Medicine 1, Kussmaul Campus for Medical Research & Translational Research Center, University of Erlangen-Nuremberg, Erlangen, Germany
| | - Imke Atreya
- Department of Medicine 1, Kussmaul Campus for Medical Research & Translational Research Center, University of Erlangen-Nuremberg, Erlangen, Germany
| | - Markus F Neurath
- Department of Medicine 1, Kussmaul Campus for Medical Research & Translational Research Center, University of Erlangen-Nuremberg, Erlangen, Germany
| | - Sebastian Zundler
- Department of Medicine 1, Kussmaul Campus for Medical Research & Translational Research Center, University of Erlangen-Nuremberg, Erlangen, Germany
| |
Collapse
|
72
|
Cigarette and Cannabis Smoking Effects on GPR15+ Helper T Cell Levels in Peripheral Blood: Relationships with Epigenetic Biomarkers. Genes (Basel) 2020; 11:genes11020149. [PMID: 32019074 PMCID: PMC7074551 DOI: 10.3390/genes11020149] [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: 01/15/2020] [Accepted: 01/27/2020] [Indexed: 12/20/2022] Open
Abstract
Background: Smoking causes widespread epigenetic changes that have been linked with an increased risk of smoking-associated diseases and elevated mortality. Of particular interest are changes in the level of T cells expressing G-protein-coupled receptor 15 (GPR15), a chemokine receptor linked with multiple autoimmune diseases, including inflammatory bowel disease, multiple sclerosis and psoriasis. Accordingly, a better understanding of the mechanisms by which smoking influences variation in the GPR15+ helper T cell subpopulation is of potential interest. Methods: In the current study, we used flow cytometry and digital PCR assays to measure the GPR15+CD3+CD4+ populations in peripheral blood from a cohort of n = 62 primarily African American young adults (aged 27–35 years) with a high rate of tobacco and cannabis use. Results: We demonstrated that self-reported tobacco and cannabis smoking predict GPR15+CD3+CD4+ helper T cell levels using linear regression models. Further, we demonstrated that methylation of two candidate CpGs, cg19859270, located in GPR15, and cg05575921, located in the gene Aryl Hydrocarbon Receptor Repressor (AHRR), were both significant predictors of GPR15+CD3+CD4+ cell levels, mediating the relationship between smoking habits and increases in GPR15+CD3+CD4+ cells. As hypothesized, the interaction between cg05575921 and cg19859270 was also significant, indicating that low cg05575921 methylation was more strongly predictive of GPR15+CD3+CD4+ cell levels for those who also had lower cg19859270 methylation. Conclusions: Smoking leads changes in two CpGs, cg05575921 and cg19859270, that mediate 38.5% of the relationship between tobacco and cannabis smoking and increased GPR15+ Th levels in this sample. The impact of cg19859270 in amplifying the association between cg05575921 and increased GPR15+ Th levels is of potential theoretical interest given the possibility that it reflects a permissive interaction between different parts of the adaptive immune system.
Collapse
|
73
|
Cox SR, Lindsay JO, Fromentin S, Stagg AJ, McCarthy NE, Galleron N, Ibraim SB, Roume H, Levenez F, Pons N, Maziers N, Lomer MC, Ehrlich SD, Irving PM, Whelan K. Effects of Low FODMAP Diet on Symptoms, Fecal Microbiome, and Markers of Inflammation in Patients With Quiescent Inflammatory Bowel Disease in a Randomized Trial. Gastroenterology 2020; 158:176-188.e7. [PMID: 31586453 DOI: 10.1053/j.gastro.2019.09.024] [Citation(s) in RCA: 224] [Impact Index Per Article: 44.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/02/2019] [Revised: 09/17/2019] [Accepted: 09/19/2019] [Indexed: 02/07/2023]
Abstract
BACKGROUND & AIMS There is limited evidence that a diet low in fermentable oligosaccharides, disaccharides, monosaccharides, and polyols (FODMAPs) reduces gut symptoms in quiescent inflammatory bowel disease (IBD). We performed a randomized, controlled trial to investigate the effects of a low FODMAP diet on persistent gut symptoms, the intestinal microbiome, and circulating markers of inflammation in patients with quiescent IBD. METHODS We performed a single-blind trial of 52 patients with quiescent Crohn's disease or ulcerative colitis and persistent gut symptoms at 2 large gastroenterology clinics in the United Kingdom. Patients were randomly assigned to groups that followed a diet low in FODMAPs (n = 27) or a control diet (n = 25), with dietary advice, for 4 weeks. Gut symptoms and health-related quality of life were measured using validated questionnaires. Stool and blood samples were collected at baseline and end of trial. We assessed fecal microbiome composition and function using shotgun metagenomic sequencing and phenotypes of T cells in blood using flow cytometry. RESULTS A higher proportion of patients reported adequate relief of gut symptoms following the low FODMAP diet (14/27, 52%) than the control diet (4/25, 16%, P=.007). Patients had a greater reduction in irritable bowel syndrome severity scores following the low FODMAP diet (mean reduction of 67; standard error, 78) than the control diet (mean reduction of 34; standard error, 50), although this difference was not statistically significant (P = .075). Following the low FODMAP diet, patients had higher health-related quality of life scores (81.9 ± 1.2) than patients on the control diet (78.3 ± 1.2, P = .042). A targeted analysis revealed that in stool samples collected at the end of the study period, patients on the low FODMAP diet had significantly lower abundance of Bifidobacterium adolescentis, Bifidobacterium longum, and Faecalibacterium prausnitzii than patients on control diet. However, microbiome diversity and markers of inflammation did not differ significantly between groups. CONCLUSIONS In a trial of the low FODMAP diet vs a control diet in patients with quiescent IBD, we found no significant difference after 4 weeks in change in irritable bowel syndrome severity scores, but significant improvements in specific symptom scores and numbers reporting adequate symptom relief. The low FODMAP diet reduced fecal abundance of microbes believed to regulate the immune response, compared with the control diet, but had no significant effect on markers of inflammation. We conclude that a 4-week diet low in FODMAPs is safe and effective for managing persistent gut symptoms in patients with quiescent IBD. www.isrctn.com no.: ISRCTN17061468.
Collapse
Affiliation(s)
- Selina R Cox
- Department of Nutritional Sciences, King's College London, London, United Kingdom
| | - James O Lindsay
- Barts Health NHS Trust, Department of Gastroenterology, Royal London Hospital, London, United Kingdom; Blizard Institute, Queen Mary University of London, Centre for Immunobiology, London, United Kingdom
| | - Sébastien Fromentin
- Metagénopolis, Institut National de la Recherche Agronomique, Université Paris-Saclay, Paris, France
| | - Andrew J Stagg
- Blizard Institute, Queen Mary University of London, Centre for Immunobiology, London, United Kingdom
| | - Neil E McCarthy
- Blizard Institute, Queen Mary University of London, Centre for Immunobiology, London, United Kingdom
| | - Nathalie Galleron
- Metagénopolis, Institut National de la Recherche Agronomique, Université Paris-Saclay, Paris, France
| | - Samar B Ibraim
- Metagénopolis, Institut National de la Recherche Agronomique, Université Paris-Saclay, Paris, France
| | - Hugo Roume
- Metagénopolis, Institut National de la Recherche Agronomique, Université Paris-Saclay, Paris, France
| | - Florence Levenez
- Metagénopolis, Institut National de la Recherche Agronomique, Université Paris-Saclay, Paris, France
| | - Nicolas Pons
- Metagénopolis, Institut National de la Recherche Agronomique, Université Paris-Saclay, Paris, France
| | - Nicolas Maziers
- Metagénopolis, Institut National de la Recherche Agronomique, Université Paris-Saclay, Paris, France
| | - Miranda C Lomer
- Department of Nutritional Sciences, King's College London, London, United Kingdom; Department of Nutrition and Dietetics, Guy's and St Thomas' NHS Foundation Trust, London, United Kingdom
| | - S Dusko Ehrlich
- Metagénopolis, Institut National de la Recherche Agronomique, Université Paris-Saclay, Paris, France
| | - Peter M Irving
- Department of Gastroenterology, Guy's and St Thomas' NHS Foundation Trust, London, United Kingdom
| | - Kevin Whelan
- Department of Nutritional Sciences, King's College London, London, United Kingdom.
| |
Collapse
|
74
|
Long Y, Wang C, Xia C, Li X, Fan C, Zhao X, Liu C. Recovery of CD226-TIGIT +FoxP3 + and CD226-TIGIT-FoxP3 + regulatory T cells contributes to clinical remission from active stage in ulcerative colitis patients. Immunol Lett 2019; 218:30-39. [PMID: 31883787 DOI: 10.1016/j.imlet.2019.12.007] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2019] [Revised: 12/16/2019] [Accepted: 12/24/2019] [Indexed: 12/31/2022]
Abstract
PURPOSE Ulcerative colitis (UC) is a chronic inflammatory immune-related disease. Imbalance between pathogenic cells and immunosuppressive cells is associated with disease activity of UC. Regulatory T cells (Tregs) are critical for this immune homeostasis. However, the clinical significance of CD226 and TIGIT expressions on FoxP3+Tregs in UC remains unclear. METHODS Comprehensive analyses of CD226 and TIGIT expressions on FoxP3+Tregs were performed by flow cytometry in 72 UC patients and 35 healthy controls, and ten active UC patients achieving remission after treatment with 5-aminosalicylic acid were followed up. Expressions of β7, α4 and αE on FoxP3+Tregs were analyzed. Clinical indicators were retrospectively acquired and serum cytokines were detected using ELISA, and their correlations with FoxP3+Treg subsets were conducted. RESULTS In active UC, levels of FoxP3+Tregs and CD226-FoxP3+Tregs regardless of TIGIT expression were significantly decreased while percentages of CD226+TIGIT-FoxP3+Tregs and CD226+TIGIT+FoxP3+Tregs were obviously increased. The expressions of β7, α4β7 and αEβ7 in FoxP3+Tregs, CD226-TIGIT+FoxP3+Tregs and CD226-TIGIT-FoxP3+Tregs were significantly elevated in active UC. Furthermore, inverse correlations were found between FoxP3+Tregs, CD226-TIGIT+FoxP3+Tregs, CD226-TIGIT-FoxP3+Tregs and serum CRP, as well as Mayo scores. IL-10 was reduced and positively correlated with CD226-TIGIT+FoxP3+Tregs and CD226-TIGIT-FoxP3+Tregs while IL-12 was increased and negatively correlated with CD226-TIGIT+FoxP3+Tregs and CD226-TIGIT-FoxP3+Tregs in active UC. In follow-up patients, the levels of FoxP3+Tregs, CD226-TIGIT+FoxP3+Tregs and CD226-TIGIT-FoxP3+Tregs and serum IL-10 levels were significantly recovered when achieving remission after treatment. CONCLUSIONS Lack of CD226 expression on FoxP3+Tregs regardless of TIGIT expression may play an important role in exhibiting their suppressive function and preventing from disease activity in UC.
Collapse
Affiliation(s)
- Yan Long
- Department of Clinical Laboratory, Peking University People's Hospital, Beijing, China
| | - Chengbin Wang
- Medical School of Chinese PLA & Department of Clinical Laboratory Medicine, Chinese People's Liberation Army General Hospital, Beijing, China
| | - Changsheng Xia
- Department of Clinical Laboratory, Peking University People's Hospital, Beijing, China
| | - Xiaoxu Li
- Department of Gastroenterology, Peking University People's Hospital, Beijing, China
| | - Chunhong Fan
- Department of Clinical Laboratory, Peking University People's Hospital, Beijing, China
| | - Xiaotao Zhao
- Department of Clinical Laboratory, Peking University People's Hospital, Beijing, China
| | - Chen Liu
- Department of Clinical Laboratory, Peking University People's Hospital, Beijing, China.
| |
Collapse
|
75
|
Sun H, Kuk W, Rivera-Nieves J, Lopez-Ramirez MA, Eckmann L, Ginsberg MH. β7 Integrin Inhibition Can Increase Intestinal Inflammation by Impairing Homing of CD25 hiFoxP3 + Regulatory T Cells. Cell Mol Gastroenterol Hepatol 2019; 9:369-385. [PMID: 31707128 PMCID: PMC7016000 DOI: 10.1016/j.jcmgh.2019.10.012] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/05/2019] [Revised: 10/28/2019] [Accepted: 10/30/2019] [Indexed: 12/11/2022]
Abstract
BACKGROUND & AIMS Integrin α4β7 mediates lymphocyte trafficking to the gut and gut-associated lymphoid tissues, a process critical for recruitment of effector lymphocytes from the circulation to the gut mucosa in inflammatory bowel disease (IBD) and murine models of intestinal inflammation. Antibody blockade of β7 integrins generally is efficacious in IBD; however, some patients fail to respond, and a few patients can experience exacerbations. This study examined the effects of loss of β7 integrin function in murine models of IBD. METHODS In a mouse IBD model caused by lack of interleukin 10, a cytokine important in CD25hiFoxP3+ regulatory T cell (Treg) function, genetic deletion of β7 integrin or antibody blockade of α4β7-mucosal addressin cell adhesion molecule-1 interaction paradoxically exacerbated colitis. RESULTS Loss of β7 impaired the capacity of Tregs homing to the gut and therefore suppress intestinal inflammation in an adoptive T-cell transfer model; however, the intrinsic suppressive function of β7-deficient Tregs remained intact, indicating that the β7 deficiency selectively impacts gut homing. Deletion of β7 integrin did not worsen colitis in an acute dextran sodium sulfate model in which Treg number and function were normal. CONCLUSIONS In Integrin subunit beta (Itgb)7-/-Il10-/- mice, loss of β7-dependent Treg homing to gut-associated lymphoid tissues combined with loss of intrinsic Treg function exacerbated intestinal inflammation. These results suggest that IBD patients with reduced CD25hiFoxP3+ Treg numbers or function or lack of interleukin 10 could be at risk for failure of α4β7 blocking therapy.
Collapse
Affiliation(s)
- Hao Sun
- Department of Medicine, University of California San Diego, La Jolla, California
| | - Wun Kuk
- Department of Medicine, University of California San Diego, La Jolla, California
| | - Jesús Rivera-Nieves
- Inflammatory Bowel Disease Center, University of California San Diego, La Jolla, California
| | | | - Lars Eckmann
- Division of Gastroenterology, University of California San Diego, La Jolla, California
| | - Mark H Ginsberg
- Department of Medicine, University of California San Diego, La Jolla, California.
| |
Collapse
|
76
|
López-Posadas R, Fastancz P, Martínez-Sánchez LDC, Panteleev-Ivlev J, Thonn V, Kisseleva T, Becker LS, Schulz-Kuhnt A, Zundler S, Wirtz S, Atreya R, Carlé B, Friedrich O, Schürmann S, Waldner MJ, Neufert C, Brakebusch CH, Bergö MO, Neurath MF, Atreya I. Inhibiting PGGT1B Disrupts Function of RHOA, Resulting in T-cell Expression of Integrin α4β7 and Development of Colitis in Mice. Gastroenterology 2019; 157:1293-1309. [PMID: 31302143 DOI: 10.1053/j.gastro.2019.07.007] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/23/2018] [Revised: 07/01/2019] [Accepted: 07/07/2019] [Indexed: 01/07/2023]
Abstract
BACKGROUND & AIMS It is not clear how regulation of T-cell function is altered during development of inflammatory bowel diseases (IBD). We studied the mechanisms by which geranylgeranyltransferase-mediated prenylation controls T-cell localization to the intestine and chronic inflammation. METHODS We generated mice with T-cell-specific disruption of the geranylgeranyltransferase type I, beta subunit gene (Pggt1b), called Pggt1bΔCD4 mice, or the ras homolog family member A gene (Rhoa), called RhoaΔCD4 mice. We also studied mice with knockout of CDC42 or RAC1 and wild-type mice (controls). Intestinal tissues were analyzed by histology, multiphoton and confocal microscopy, and real-time polymerase chain reaction. Activation of CDC42, RAC1, and RHOA were measured with G-LISA, cell fractionation, and immunoblots. T cells and lamina propria mononuclear cells from mice were analyzed by flow cytometry or transferred to Rag1-/- mice. Mice were given injections of antibodies against integrin alpha4beta7 or gavaged with the RORC antagonist GSK805. We obtained peripheral blood and intestinal tissue samples from patients with and without IBD and analyzed them by flow cytometry. RESULTS Pggt1bΔCD4 mice developed spontaneous colitis, characterized by thickening of the intestinal wall, edema, fibrosis, accumulation of T cells in the colon, and increased expression of inflammatory cytokines. Compared with control CD4+ T cells, PGGT1B-deficient CD4+ T cells expressed significantly higher levels of integrin alpha4beta7, which regulates their localization to the intestine. Inflammation induced by transfer of PGGT1B-deficient CD4+ T cells to Rag1-/- mice was blocked by injection of an antibody against integrin alpha4beta7. Lamina propria of Pggt1bΔCD4 mice had increased numbers of CD4+ T cells that expressed RORC and higher levels of cytokines produced by T-helper 17 cells (granulocyte-macrophage colony-stimulating factor, interleukin [IL]17A, IL17F, IL22, and tumor necrosis factor [TNF]). The RORC inverse agonist GSK805, but not antibodies against IL17A or IL17F, prevented colitis in Pggt1bΔCD4 mice. PGGT1B-deficient CD4+ T cells had decreased activation of RHOA. RhoAΔCD4 mice had a similar phenotype to Pggt1bΔCD4 mice, including development of colitis, increased numbers of CD4+ T cells in colon, increased expression of integrin alpha4beta7 by CD4+ T cells, and increased levels of IL17A and other inflammatory cytokines in lamina propria. T cells isolated from intestinal tissues from patients with IBD had significantly lower levels of PGGT1B than tissues from individuals without IBD. CONCLUSION Loss of PGGT1B from T cells in mice impairs RHOA function, increasing CD4+ T-cell expression of integrin alpha4beta7 and localization to colon, resulting in increased expression of inflammatory cytokines and colitis. T cells isolated from gut tissues from patients with IBD have lower levels of PGGT1B than tissues from patients without IBD.
Collapse
Affiliation(s)
- Rocío López-Posadas
- Department of Medicine 1, Friedrich-Alexander-University Erlangen-Nuremberg, Erlangen, Germany.
| | - Petra Fastancz
- Department of Medicine 1, Friedrich-Alexander-University Erlangen-Nuremberg, Erlangen, Germany
| | | | - Julia Panteleev-Ivlev
- Department of Medicine 1, Friedrich-Alexander-University Erlangen-Nuremberg, Erlangen, Germany
| | - Veronika Thonn
- Department of Medicine 1, Friedrich-Alexander-University Erlangen-Nuremberg, Erlangen, Germany
| | - Tatyana Kisseleva
- Department of Medicine 1, Friedrich-Alexander-University Erlangen-Nuremberg, Erlangen, Germany
| | - Lukas S Becker
- Department of Medicine 1, Friedrich-Alexander-University Erlangen-Nuremberg, Erlangen, Germany
| | - Anja Schulz-Kuhnt
- Department of Medicine 1, Friedrich-Alexander-University Erlangen-Nuremberg, Erlangen, Germany
| | - Sebastian Zundler
- Department of Medicine 1, Friedrich-Alexander-University Erlangen-Nuremberg, Erlangen, Germany
| | - Stefan Wirtz
- Department of Medicine 1, Friedrich-Alexander-University Erlangen-Nuremberg, Erlangen, Germany
| | - Raja Atreya
- Department of Medicine 1, Friedrich-Alexander-University Erlangen-Nuremberg, Erlangen, Germany
| | - Birgitta Carlé
- Department of Chemical and Biological Engineering, Institute of Medical Biotechnology, Friedrich-Alexander-University Erlangen-Nuremberg, Erlangen, Germany
| | - Oliver Friedrich
- Department of Chemical and Biological Engineering, Institute of Medical Biotechnology, Friedrich-Alexander-University Erlangen-Nuremberg, Erlangen, Germany
| | - Sebastian Schürmann
- Department of Chemical and Biological Engineering, Institute of Medical Biotechnology, Friedrich-Alexander-University Erlangen-Nuremberg, Erlangen, Germany
| | - Maximilian J Waldner
- Department of Medicine 1, Friedrich-Alexander-University Erlangen-Nuremberg, Erlangen, Germany
| | - Clemens Neufert
- Department of Medicine 1, Friedrich-Alexander-University Erlangen-Nuremberg, Erlangen, Germany
| | - Cord H Brakebusch
- Biotec Research and Innovation Center, University of Copenhagen, Copenhagen, Denmark
| | - Martin O Bergö
- Sahlgrenska Cancer Center, University of Gothenburg, Gothenburg, Sweden
| | - Markus F Neurath
- Department of Medicine 1, Friedrich-Alexander-University Erlangen-Nuremberg, Erlangen, Germany
| | - Imke Atreya
- Department of Medicine 1, Friedrich-Alexander-University Erlangen-Nuremberg, Erlangen, Germany
| |
Collapse
|
77
|
Abstract
PURPOSE OF REVIEW To describe the latest developments in the field of anti-trafficking agents (ATAs), a class of therapeutics with growing importance in the field of inflammatory bowel diseases (IBDs) that specifically inhibit steps of immune cell trafficking. RECENT FINDINGS Several translational and clinical studies have further shaped the knowledge about the mechanisms and effects of the anti-α4β7 integrin antibody vedolizumab. In parallel, new ATAs like the anti-β7 integrin antibody etrolizumab and the anti-MAdCAM-1 antibody ontamalimab are investigated in phase III clinical trials and might soon increase the therapeutic armamentarium in IBD. SUMMARY ATAs have unique mechanisms of action and can meanwhile be considered an indispensable column of IBD therapy. Further efforts are necessary to elucidate complex mechanistic aspects, to exactly define their role in relation to other therapeutic approaches and to identify novel treatment targets as well as biomarkers for personalized medicine.
Collapse
|
78
|
Gill PA, van Zelm MC, Ffrench RA, Muir JG, Gibson PR. Successful elevation of circulating acetate and propionate by dietary modulation does not alter T-regulatory cell or cytokine profiles in healthy humans: a pilot study. Eur J Nutr 2019; 59:2651-2661. [PMID: 31650328 DOI: 10.1007/s00394-019-02113-2] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2019] [Accepted: 10/04/2019] [Indexed: 12/13/2022]
Abstract
PURPOSE Increased circulating concentrations of short-chain fatty acids (SCFA) achieved by ingestion of high-fibre diets is associated with anti-inflammatory effects through promotion of FoxP3+ regulatory T(reg) cells in mouse models. This study aimed to determine whether similar increments in blood SCFA levels can be achieved in humans and whether these are associated with similar immune modulatory effects. METHODS In a pilot single-blinded, randomised, controlled cross-over study in ten healthy subjects, the effects were determined of high- (39 g/day) and low-fibre (18 g/day) intake (all food provided) on SCFA (gas chromatography), proportions of Treg cells (flow cytometry) and a panel of cytokines (multiplex methodology) measured in peripheral blood at day 5 of each diet. RESULTS Actual fibre intake differed between the diets by 19 [16-21] g/day (P< 0.001). Median [range] total plasma SCFA levels with high-fibre intake were 174.5 [104.8-249.5] µmol/L, which were greater than those associated with low-fibre intake at 59.0 [26.5-79.9] (P < 0.001). Differences were significantly different for both acetate and propionate. The frequencies of total CD4 T cells and T-regulatory cells, and concentrations of inflammatory and anti-inflammatory cytokines were not significantly different between the dietary interventions. CONCLUSIONS Plasma SCFA levels can be modulated by altering dietary fibre consumption in healthy individuals with increments similar to those achieved in murine studies. Five days of diet intervention did not result in changes in regulatory T-cell proportions and cytokine concentrations in peripheral blood, and may require longer duration of dietary change.
Collapse
Affiliation(s)
- Paul A Gill
- Department of Gastroenterology, Central Clinical School, Monash University and Alfred Hospital, Melbourne, VIC, Australia.,Department of Immunology and Pathology, Central Clinical School, Monash University and Alfred Hospital, Melbourne, VIC, Australia
| | - Menno C van Zelm
- Department of Immunology and Pathology, Central Clinical School, Monash University and Alfred Hospital, Melbourne, VIC, Australia
| | | | - Jane G Muir
- Department of Gastroenterology, Central Clinical School, Monash University and Alfred Hospital, Melbourne, VIC, Australia
| | - Peter R Gibson
- Department of Gastroenterology, Central Clinical School, Monash University and Alfred Hospital, Melbourne, VIC, Australia.
| |
Collapse
|
79
|
Zundler S, Becker E, Schulze LL, Neurath MF. Immune cell trafficking and retention in inflammatory bowel disease: mechanistic insights and therapeutic advances. Gut 2019; 68:1688-1700. [PMID: 31127023 DOI: 10.1136/gutjnl-2018-317977] [Citation(s) in RCA: 108] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/13/2019] [Revised: 05/01/2019] [Accepted: 05/03/2019] [Indexed: 12/12/2022]
Abstract
Intestinal immune cell trafficking has been identified as a central event in the pathogenesis of inflammatory bowel diseases (IBD). Intensive research on different aspects of the immune mechanisms controlling and controlled by T cell trafficking and retention has led to the approval of the anti-α4β7 antibody vedolizumab, the ongoing development of a number of further anti-trafficking agents (ATAs) such as the anti-β7 antibody etrolizumab or the anti-MAdCAM-1 antibody ontamalimab and the identification of potential future targets like G-protein coupled receptor 15. However, several aspects of the biology of immune cell trafficking and regarding the mechanism of action of ATAs are still unclear, for example, which impact these compounds have on the trafficking of non-lymphocyte populations like monocytes and how precisely these therapies differ with regard to their effect on immune cell subpopulations. This review will summarise recent advances of basic science in the field of intestinal immune cell trafficking and discuss these findings with regard to different pharmacological approaches from a translational perspective.
Collapse
Affiliation(s)
- Sebastian Zundler
- Department of Medicine 1, Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, Kussmaul Campus for Medical Research & Translational Research Center, Erlangen, Germany
| | - Emily Becker
- Department of Medicine 1, Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, Kussmaul Campus for Medical Research & Translational Research Center, Erlangen, Germany
| | - Lisa Lou Schulze
- Department of Medicine 1, Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, Kussmaul Campus for Medical Research & Translational Research Center, Erlangen, Germany
| | - Markus F Neurath
- Department of Medicine 1, Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, Kussmaul Campus for Medical Research & Translational Research Center, Erlangen, Germany
| |
Collapse
|
80
|
Fu Z, Ye J, Dean JW, Bostick JW, Weinberg SE, Xiong L, Oliff KN, Chen ZE, Avram D, Chandel NS, Zhou L. Requirement of Mitochondrial Transcription Factor A in Tissue-Resident Regulatory T Cell Maintenance and Function. Cell Rep 2019; 28:159-171.e4. [PMID: 31269437 PMCID: PMC6679941 DOI: 10.1016/j.celrep.2019.06.024] [Citation(s) in RCA: 60] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2018] [Revised: 04/19/2019] [Accepted: 06/04/2019] [Indexed: 01/09/2023] Open
Abstract
Regulatory T cells (Tregs) are pivotal for immune suppression. Cellular metabolism is important for Treg homeostasis and function. However, the exact role of mitochondrial respiration in Tregs remains elusive. Mitochondrial transcription factor A (Tfam) is essential for mitochondrial respiration and controls mitochondrial DNA replication, transcription, and packaging. Here, we show that genetic ablation of Tfam in Tregs impairs Treg maintenance in non-lymphoid tissues in the steady state and in tumors. Tfam-deficient Tregs have reduced proliferation and Foxp3 expression upon glucose deprivation in vitro. Tfam deficiency preferentially affects gene activation in Tregs through regulation of DNA methylation, with enhanced methylation in the TSDR of the Foxp3 locus. Deletion of Tfam in Tregs affects Treg homing and stability, resulting in tissue inflammation in colitis, but enhances tumor rejection. Thus, our work reveals a critical role of Tfam-mediated mitochondrial respiration in Tregs to regulate inflammation and anti-tumor immunity.
Collapse
Affiliation(s)
- Zheng Fu
- Department of Infectious Diseases and Immunology, College of Veterinary Medicine, University of Florida, Gainesville, FL 32608, USA
| | - Jian Ye
- Department of Infectious Diseases and Immunology, College of Veterinary Medicine, University of Florida, Gainesville, FL 32608, USA
| | - Joseph W Dean
- Department of Infectious Diseases and Immunology, College of Veterinary Medicine, University of Florida, Gainesville, FL 32608, USA
| | - John W Bostick
- Department of Infectious Diseases and Immunology, College of Veterinary Medicine, University of Florida, Gainesville, FL 32608, USA; Department of Chemical and Biological Engineering, Northwestern University, Evanston, IL 60208, USA
| | - Samuel E Weinberg
- Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Lifeng Xiong
- Department of Infectious Diseases and Immunology, College of Veterinary Medicine, University of Florida, Gainesville, FL 32608, USA
| | - Kristen N Oliff
- Department of Infectious Diseases and Immunology, College of Veterinary Medicine, University of Florida, Gainesville, FL 32608, USA
| | - Zongming E Chen
- Geisinger Medical Center, Laboratory Medicine, 01-31, 100 North Academy Avenue, Danville, PA 17822, USA
| | - Dorina Avram
- Department of Anatomy and Cell Biology, College of Medicine, University of Florida, Gainesville, FL 32608, USA
| | - Navdeep S Chandel
- Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Liang Zhou
- Department of Infectious Diseases and Immunology, College of Veterinary Medicine, University of Florida, Gainesville, FL 32608, USA.
| |
Collapse
|
81
|
Dotan I, Allez M, Danese S, Keir M, Tole S, McBride J. The role of integrins in the pathogenesis of inflammatory bowel disease: Approved and investigational anti-integrin therapies. Med Res Rev 2019; 40:245-262. [PMID: 31215680 PMCID: PMC6973243 DOI: 10.1002/med.21601] [Citation(s) in RCA: 65] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2019] [Revised: 04/12/2019] [Accepted: 05/13/2019] [Indexed: 12/12/2022]
Abstract
Inflammatory bowel disease (IBD) is characterized by uncontrolled inflammation in the gastrointestinal tract. The underlying pathobiology of IBD includes an increase in infiltrating gut-homing lymphocytes. Although lymphocyte homing is typically a tightly regulated and stepwise process involving multiple integrins and adhesion molecules expressed on endothelial cells, the distinct roles of integrin-expressing immune cells is not fully understood in the pathology of IBD. In this review, we detail the involvement of integrins expressed on specific lymphocyte subsets in the pathogenesis of IBD and discuss the current status of approved and investigational integrin-targeted therapies.
Collapse
Affiliation(s)
- Iris Dotan
- Division of Gastroenterology, Rabin Medical Center, Petah Tikva, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Matthieu Allez
- Department of Gastroenterology, Hôpital Saint-Louis, AP-HP, INSERM U1160, University Denis Diderot, Paris, France
| | - Silvio Danese
- Gastrointestinal Immunopathology Laboratory and IBD Unit, Humanitas Clinical and Research Center, Milan, Italy
| | - Mary Keir
- Department of Research and Early Development, Genentech, South San Francisco, California
| | - Swati Tole
- Department of Product Development, Genentech, South San Francisco, California
| | - Jacqueline McBride
- Department of Research and Early Development, Genentech, South San Francisco, California
| |
Collapse
|
82
|
Xu Y, Cheng Y, Baylink DJ, Wasnik S, Goel G, Huang M, Cao H, Qin X, Lau KHW, Chan C, Koch A, Pham LH, Zhang J, Li CH, Wang X, Berumen EC, Smith J, Tang X. In Vivo Generation of Gut-Homing Regulatory T Cells for the Suppression of Colitis. THE JOURNAL OF IMMUNOLOGY 2019; 202:3447-3457. [PMID: 31053627 PMCID: PMC10234421 DOI: 10.4049/jimmunol.1800018] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/04/2018] [Accepted: 04/15/2019] [Indexed: 12/18/2022]
Abstract
Current therapies for gut inflammation have not reached the desired specificity and are attended by unintended immune suppression. This study aimed to provide evidence for supporting a hypothesis that direct in vivo augmentation of the induction of gut-homing regulatory T (Treg) cells is a strategy of expected specificity for the treatment of chronic intestinal inflammation (e.g., inflammatory bowel disease). We showed that dendritic cells (DCs), engineered to de novo produce high concentrations of both 1,25-dihydroxyvitamin D, the active vitamin D metabolite, and retinoic acid, an active vitamin A metabolite, augmented the induction of T cells that express both the regulatory molecule Foxp3 and the gut-homing receptor CCR9 in vitro and in vivo. In vivo, the newly generated Ag-specific Foxp3+ T cells homed to intestines. Additionally, transfer of such engineered DCs robustly suppressed ongoing experimental colitis. Moreover, CD4+ T cells from spleens of the mice transferred with the engineered DCs suppressed experimental colitis in syngeneic hosts. The data suggest that the engineered DCs enhance regulatory function in CD4+ T cell population in peripheral lymphoid tissues. Finally, we showed that colitis suppression following in vivo transfer of the engineered DCs was significantly reduced when Foxp3+ Treg cells were depleted. The data indicate that maximal colitis suppression mediated by the engineered DCs requires Treg cells. Collectively, our data support that DCs de novo overproducing both 1,25-dihydroxyvitamin D and retinoic acid are a promising novel therapy for chronic intestinal inflammation.
Collapse
Affiliation(s)
- Yi Xu
- Division of Regenerative Medicine, Department of Medicine, Loma Linda University, Loma Linda, CA 92354.,Department of Hematology and Oncology, Loma Linda University Cancer Center, Loma Linda, CA 92354
| | - Yanmei Cheng
- Division of Regenerative Medicine, Department of Medicine, Loma Linda University, Loma Linda, CA 92354.,Gastroenterology Department, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 200437, China
| | - David J Baylink
- Division of Regenerative Medicine, Department of Medicine, Loma Linda University, Loma Linda, CA 92354
| | - Samiksha Wasnik
- Division of Regenerative Medicine, Department of Medicine, Loma Linda University, Loma Linda, CA 92354
| | - Gati Goel
- Division of Gastroenterology, Hepatology and Nutrition, Department of Medicine, Loma Linda University, Loma Linda, CA 92354
| | - Mei Huang
- Division of Regenerative Medicine, Department of Medicine, Loma Linda University, Loma Linda, CA 92354.,College of Pharmacy, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian 350122, China
| | - Huynh Cao
- Department of Hematology and Oncology, Loma Linda University Cancer Center, Loma Linda, CA 92354
| | - Xuezhong Qin
- Division of Regenerative Medicine, Department of Medicine, Loma Linda University, Loma Linda, CA 92354.,Musculoskeletal Disease Center, Jerry L. Pettis Memorial Veterans Affairs Medical Center, Loma Linda, CA 92357
| | - Kin-Hing William Lau
- Division of Regenerative Medicine, Department of Medicine, Loma Linda University, Loma Linda, CA 92354.,Musculoskeletal Disease Center, Jerry L. Pettis Memorial Veterans Affairs Medical Center, Loma Linda, CA 92357
| | - Christian Chan
- Division of Regenerative Medicine, Department of Medicine, Loma Linda University, Loma Linda, CA 92354
| | - Adam Koch
- Division of Regenerative Medicine, Department of Medicine, Loma Linda University, Loma Linda, CA 92354
| | - Linh H Pham
- Division of Regenerative Medicine, Department of Medicine, Loma Linda University, Loma Linda, CA 92354
| | - Jintao Zhang
- Division of Regenerative Medicine, Department of Medicine, Loma Linda University, Loma Linda, CA 92354.,Institute of Medical and Pharmaceutical Sciences, Zhengzhou University, Henan 450052, China
| | - Chih-Huang Li
- Division of Regenerative Medicine, Department of Medicine, Loma Linda University, Loma Linda, CA 92354.,Department of Emergency Medicine, Chang-Gung Memorial Hospital, Linkou Medical Center, Taoyuan, Taiwan.,Graduate Institute of Clinical Medical Sciences, School of Medicine, Chang-Gung University, Taoyuan 333, Taiwan
| | - Xiaohua Wang
- Division of Regenerative Medicine, Department of Medicine, Loma Linda University, Loma Linda, CA 92354.,Jinan Infectious Disease Hospital, Shandong University, Shandong 250014, China; and
| | - Edmundo Carreon Berumen
- Division of Regenerative Medicine, Department of Medicine, Loma Linda University, Loma Linda, CA 92354
| | - James Smith
- X Cell Laboratories Inc., Redlands, CA 92373
| | - Xiaolei Tang
- Division of Regenerative Medicine, Department of Medicine, Loma Linda University, Loma Linda, CA 92354;
| |
Collapse
|
83
|
Goldberg R, Scotta C, Cooper D, Nissim-Eliraz E, Nir E, Tasker S, Irving PM, Sanderson J, Lavender P, Ibrahim F, Corcoran J, Prevost T, Shpigel NY, Marelli-Berg F, Lombardi G, Lord GM. Correction of Defective T-Regulatory Cells From Patients With Crohn's Disease by Ex Vivo Ligation of Retinoic Acid Receptor-α. Gastroenterology 2019; 156:1775-1787. [PMID: 30710527 DOI: 10.1053/j.gastro.2019.01.025] [Citation(s) in RCA: 44] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/30/2018] [Revised: 01/07/2019] [Accepted: 01/11/2019] [Indexed: 12/19/2022]
Abstract
BACKGROUND & AIMS Crohn's disease (CD) is characterized by an imbalance of effector and regulatory T cells in the intestinal mucosa. The efficacy of anti-adhesion therapies led us to investigate whether impaired trafficking of T-regulatory (Treg) cells contributes to the pathogenesis of CD. We also investigated whether proper function could be restored to Treg cells by ex vivo expansion in the presence of factors that activate their regulatory activities. METHODS We measured levels of the integrin α4β7 on Treg cells isolated from peripheral blood or lamina propria of patients with CD and healthy individuals (controls). Treg cells were expanded ex vivo and incubated with rapamycin with or without agonists of the retinoic acid receptor-α (RARA), and their gene expression profiles were analyzed. We also studied the cells in cytokine challenge, suppression, and flow chamber assays and in SCID mice with human intestinal xenografts. RESULTS We found that Treg cells from patients with CD express lower levels of the integrin α4β7 than Treg cells from control patients. The pathway that regulates the expression of integrin subunit α is induced by retinoic acid (RA). Treg cells from patients with CD incubated with rapamycin and an agonist of RARA (RAR568) expressed high levels of integrin α4β7, as well as CD62L and FOXP3, compared with cells incubated with rapamycin or rapamycin and all-trans retinoic acid. These Treg cells had increased suppressive activities in assays and migrated under conditions of shear flow; they did not produce inflammatory cytokines, and RAR568 had no effect on cell stability or lineage commitment. Fluorescently labeled Treg cells incubated with RAR568 were significantly more likely to traffic to intestinal xenografts than Treg cells expanded in control medium. CONCLUSIONS Treg cells from patients with CD express lower levels of the integrin α4β7 than Treg cells from control patients. Incubation of patients' ex vivo expanded Treg cells with rapamycin and an RARA agonist induced expression of α4β7 and had suppressive and migratory activities in culture and in intestinal xenografts in mice. These cells might be developed for treatment of CD. ClinicalTrials.gov, Number: NCT03185000.
Collapse
Affiliation(s)
- Rimma Goldberg
- Inflammatory Bowel Disease Unit, Department of Gastroenterology, Guy's and St Thomas' NHS Foundation Trust, London, UK; School of Immunology and Microbial Sciences, King's College London, London, UK; National Institute for Health Research Biomedical Research Centre, Guy's and St Thomas' NHS Trust and King's College London, London, UK
| | - Cristiano Scotta
- School of Immunology and Microbial Sciences, King's College London, London, UK
| | - Dianne Cooper
- William Harvey Research Institute, Queen Mary University of London, London, UK
| | - Einat Nissim-Eliraz
- Department of Respiratory Medicine and Allergy, King's College London, London, UK
| | - Eilam Nir
- Department of Respiratory Medicine and Allergy, King's College London, London, UK
| | - Scott Tasker
- School of Immunology and Microbial Sciences, King's College London, London, UK; National Institute for Health Research Biomedical Research Centre, Guy's and St Thomas' NHS Trust and King's College London, London, UK
| | - Peter M Irving
- Inflammatory Bowel Disease Unit, Department of Gastroenterology, Guy's and St Thomas' NHS Foundation Trust, London, UK
| | - Jeremy Sanderson
- Inflammatory Bowel Disease Unit, Department of Gastroenterology, Guy's and St Thomas' NHS Foundation Trust, London, UK
| | - Paul Lavender
- Department of Respiratory Medicine and Allergy, King's College London, London, UK
| | - Fowzia Ibrahim
- Department of Rheumatology, King's College London School of Medicine, Weston Education Centre, King's College London, London, UK
| | - Jonathan Corcoran
- Wolfson Centre for Age Related Diseases, King's College London, London, UK
| | - Toby Prevost
- Imperial Clinical Trials Unit, Imperial College London, London, UK
| | - Nahum Y Shpigel
- Koret School of Veterinary Medicine, Hebrew University of Jerusalem, Rehovot, Israel
| | | | - Giovanna Lombardi
- School of Immunology and Microbial Sciences, King's College London, London, UK
| | - Graham M Lord
- School of Immunology and Microbial Sciences, King's College London, London, UK; National Institute for Health Research Biomedical Research Centre, Guy's and St Thomas' NHS Trust and King's College London, London, UK.
| |
Collapse
|
84
|
Atreya R, Neurath MF. Mechanisms of molecular resistance and predictors of response to biological therapy in inflammatory bowel disease. Lancet Gastroenterol Hepatol 2019; 3:790-802. [PMID: 30353856 DOI: 10.1016/s2468-1253(18)30265-6] [Citation(s) in RCA: 64] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/02/2018] [Revised: 07/25/2018] [Accepted: 07/27/2018] [Indexed: 02/07/2023]
Abstract
Biological therapy has led to marked improvements in treatment of patients with inflammatory bowel disease, and an increasing number of drugs has been approved for treatment. However, only a subgroup of patients responds to therapy, highlighting the need to identify biomarkers for therapeutic response to allow personalised medicine in inflammatory bowel disease. Potential markers of response to biological therapy have been identified; however, studies also suggest that changes in the composition of immune cell infiltrates in response to therapeutic pressure lead to molecular resistance to these drugs. For instance, the cytokine interleukin 23 has been identified as a driver of evasion of apoptosis in response to anti-tumour necrosis factor drugs in patients with Crohn's disease, leading to expansion of apoptosis-resistant T cells and drug resistance. In this Review, we examine the concept of molecular resistance to biological therapy and discuss implications for future therapy.
Collapse
Affiliation(s)
- Raja Atreya
- Department of Medicine 1, University Hospital Erlangen, and Ludwig Demling Endoscopy Center of Excellence, University of Erlangen-Nuremberg, Erlangen, Germany
| | - Markus F Neurath
- Department of Medicine 1, University Hospital Erlangen, and Ludwig Demling Endoscopy Center of Excellence, University of Erlangen-Nuremberg, Erlangen, Germany.
| |
Collapse
|
85
|
Belarif L, Danger R, Kermarrec L, Nerrière-Daguin V, Pengam S, Durand T, Mary C, Kerdreux E, Gauttier V, Kucik A, Thepenier V, Martin JC, Chang C, Rahman A, Guen NSL, Braudeau C, Abidi A, David G, Malard F, Takoudju C, Martinet B, Gérard N, Neveu I, Neunlist M, Coron E, MacDonald TT, Desreumaux P, Mai HL, Le Bas-Bernardet S, Mosnier JF, Merad M, Josien R, Brouard S, Soulillou JP, Blancho G, Bourreille A, Naveilhan P, Vanhove B, Poirier N. IL-7 receptor influences anti-TNF responsiveness and T cell gut homing in inflammatory bowel disease. J Clin Invest 2019; 129:1910-1925. [PMID: 30939120 DOI: 10.1172/jci121668] [Citation(s) in RCA: 86] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2018] [Accepted: 02/21/2019] [Indexed: 12/16/2022] Open
Abstract
It remains unknown what causes inflammatory bowel disease (IBD), including signaling networks perpetuating chronic gastrointestinal inflammation in Crohn's disease (CD) and ulcerative colitis (UC), in humans. According to an analysis of up to 500 patients with IBD and 100 controls, we report that key transcripts of the IL-7 receptor (IL-7R) pathway are accumulated in inflamed colon tissues of severe CD and UC patients not responding to either immunosuppressive/corticosteroid, anti-TNF, or anti-α4β7 therapies. High expression of both IL7R and IL-7R signaling signature in the colon before treatment is strongly associated with nonresponsiveness to anti-TNF therapy. While in mice IL-7 is known to play a role in systemic inflammation, we found that in humans IL-7 also controlled α4β7 integrin expression and imprinted gut-homing specificity on T cells. IL-7R blockade reduced human T cell homing to the gut and colonic inflammation in vivo in humanized mouse models, and altered effector T cells in colon explants from UC patients grown ex vivo. Our findings show that failure of current treatments for CD and UC is strongly associated with an overexpressed IL-7R signaling pathway and point to IL-7R as a relevant therapeutic target and potential biomarker to fill an unmet need in clinical IBD detection and treatment.
Collapse
Affiliation(s)
| | - Richard Danger
- Centre de Recherche en Transplantation et Immunologie (CRTI), UMR 1064, Inserm, Université de Nantes, Nantes, France.,Institut de Transplantation Urologie Néphrologie (ITUN), Centre Hospitalier Universitaire de Nantes (CHU Nantes), Nantes, France
| | - Laetitia Kermarrec
- Institut des Maladies de l'Appareil Digestif (IMAD), The Enteric Nervous System in Gut and Brain Disorders, Université de Nantes, INSERM, Nantes, France
| | - Véronique Nerrière-Daguin
- Centre de Recherche en Transplantation et Immunologie (CRTI), UMR 1064, Inserm, Université de Nantes, Nantes, France.,Institut de Transplantation Urologie Néphrologie (ITUN), Centre Hospitalier Universitaire de Nantes (CHU Nantes), Nantes, France
| | | | - Tony Durand
- Institut des Maladies de l'Appareil Digestif (IMAD), The Enteric Nervous System in Gut and Brain Disorders, Université de Nantes, INSERM, Nantes, France
| | | | | | | | - Aneta Kucik
- Blizard Institute, Barts and The London School of Medicine and Dentistry, London, United Kingdom
| | | | - Jerome C Martin
- Precision Immunology Institute.,Tisch Cancer Institute.,Department of Oncological Sciences
| | - Christie Chang
- Precision Immunology Institute.,Tisch Cancer Institute.,Department of Oncological Sciences
| | - Adeeb Rahman
- Precision Immunology Institute.,Charles Bronfman Institute for Personalized Medicine, and.,Human Immune Monitoring Center, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Nina Salabert-Le Guen
- Centre de Recherche en Transplantation et Immunologie (CRTI), UMR 1064, Inserm, Université de Nantes, Nantes, France.,CHU Nantes, Laboratoire d'Immunologie, Center for Immuno Monitoring Nantes-Atlantique (CIMNA), Nantes, France.,LabEx Immunograft Oncology (IGO), Nantes, France.,Université de Nantes, Faculté de Médecine, Nantes, France
| | - Cécile Braudeau
- Centre de Recherche en Transplantation et Immunologie (CRTI), UMR 1064, Inserm, Université de Nantes, Nantes, France.,CHU Nantes, Laboratoire d'Immunologie, Center for Immuno Monitoring Nantes-Atlantique (CIMNA), Nantes, France.,LabEx Immunograft Oncology (IGO), Nantes, France
| | - Ahmed Abidi
- Centre de Recherche en Transplantation et Immunologie (CRTI), UMR 1064, Inserm, Université de Nantes, Nantes, France.,Université de Tunis El Manar, Laboratoire de génétique, immunologie et pathologies humaines, Faculté des sciences de Tunis, Tunis, Tunisia
| | - Grégoire David
- Institut des Maladies de l'Appareil Digestif (IMAD), The Enteric Nervous System in Gut and Brain Disorders, Université de Nantes, INSERM, Nantes, France
| | - Florent Malard
- Centre de Recherche en Transplantation et Immunologie (CRTI), UMR 1064, Inserm, Université de Nantes, Nantes, France
| | - Celine Takoudju
- Institut des Maladies de l'Appareil Digestif (IMAD), The Enteric Nervous System in Gut and Brain Disorders, Université de Nantes, INSERM, Nantes, France
| | - Bernard Martinet
- Centre de Recherche en Transplantation et Immunologie (CRTI), UMR 1064, Inserm, Université de Nantes, Nantes, France.,Institut de Transplantation Urologie Néphrologie (ITUN), Centre Hospitalier Universitaire de Nantes (CHU Nantes), Nantes, France
| | - Nathalie Gérard
- Centre de Recherche en Transplantation et Immunologie (CRTI), UMR 1064, Inserm, Université de Nantes, Nantes, France.,Institut de Transplantation Urologie Néphrologie (ITUN), Centre Hospitalier Universitaire de Nantes (CHU Nantes), Nantes, France
| | - Isabelle Neveu
- Institut des Maladies de l'Appareil Digestif (IMAD), The Enteric Nervous System in Gut and Brain Disorders, Université de Nantes, INSERM, Nantes, France.,CHU Nantes, IMAD, Nantes, France
| | - Michel Neunlist
- Institut des Maladies de l'Appareil Digestif (IMAD), The Enteric Nervous System in Gut and Brain Disorders, Université de Nantes, INSERM, Nantes, France.,CHU Nantes, IMAD, Nantes, France
| | - Emmanuel Coron
- Institut des Maladies de l'Appareil Digestif (IMAD), The Enteric Nervous System in Gut and Brain Disorders, Université de Nantes, INSERM, Nantes, France.,CHU Nantes, IMAD, Nantes, France
| | - Thomas T MacDonald
- Blizard Institute, Barts and The London School of Medicine and Dentistry, London, United Kingdom
| | - Pierre Desreumaux
- Hepato-Gastroenterology Department, Claude Huriez Hospital, University of Lille 2, Lille, France
| | - Hoa-Le Mai
- Centre de Recherche en Transplantation et Immunologie (CRTI), UMR 1064, Inserm, Université de Nantes, Nantes, France.,Institut de Transplantation Urologie Néphrologie (ITUN), Centre Hospitalier Universitaire de Nantes (CHU Nantes), Nantes, France
| | - Stephanie Le Bas-Bernardet
- Centre de Recherche en Transplantation et Immunologie (CRTI), UMR 1064, Inserm, Université de Nantes, Nantes, France.,Institut de Transplantation Urologie Néphrologie (ITUN), Centre Hospitalier Universitaire de Nantes (CHU Nantes), Nantes, France
| | - Jean-François Mosnier
- Centre de Recherche en Transplantation et Immunologie (CRTI), UMR 1064, Inserm, Université de Nantes, Nantes, France.,CHU Nantes, Service d'Anatomie et Cytologie Pathologiques, Nantes, France
| | - Miriam Merad
- Precision Immunology Institute.,Tisch Cancer Institute.,Department of Oncological Sciences.,Human Immune Monitoring Center, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Régis Josien
- Centre de Recherche en Transplantation et Immunologie (CRTI), UMR 1064, Inserm, Université de Nantes, Nantes, France.,Institut de Transplantation Urologie Néphrologie (ITUN), Centre Hospitalier Universitaire de Nantes (CHU Nantes), Nantes, France.,CHU Nantes, Laboratoire d'Immunologie, Center for Immuno Monitoring Nantes-Atlantique (CIMNA), Nantes, France.,Université de Nantes, Faculté de Médecine, Nantes, France
| | - Sophie Brouard
- Centre de Recherche en Transplantation et Immunologie (CRTI), UMR 1064, Inserm, Université de Nantes, Nantes, France.,Institut de Transplantation Urologie Néphrologie (ITUN), Centre Hospitalier Universitaire de Nantes (CHU Nantes), Nantes, France
| | - Jean-Paul Soulillou
- Centre de Recherche en Transplantation et Immunologie (CRTI), UMR 1064, Inserm, Université de Nantes, Nantes, France
| | - Gilles Blancho
- Centre de Recherche en Transplantation et Immunologie (CRTI), UMR 1064, Inserm, Université de Nantes, Nantes, France.,Institut de Transplantation Urologie Néphrologie (ITUN), Centre Hospitalier Universitaire de Nantes (CHU Nantes), Nantes, France
| | - Arnaud Bourreille
- Institut des Maladies de l'Appareil Digestif (IMAD), The Enteric Nervous System in Gut and Brain Disorders, Université de Nantes, INSERM, Nantes, France.,CHU Nantes, IMAD, Nantes, France
| | - Philippe Naveilhan
- Institut des Maladies de l'Appareil Digestif (IMAD), The Enteric Nervous System in Gut and Brain Disorders, Université de Nantes, INSERM, Nantes, France.,CHU Nantes, IMAD, Nantes, France
| | | | | |
Collapse
|
86
|
Duijvestein M, D’Haens GR. Rational and clinical development of the anti-MAdCAM monoclonal antibody for the treatment of IBD. Expert Opin Biol Ther 2019; 19:361-366. [DOI: 10.1080/14712598.2019.1576631] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Affiliation(s)
- Marjolijn Duijvestein
- Inflammatory Bowel Disease Center and Amsterdam Gastroenterology and Metabolism, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Geert R. D’Haens
- Inflammatory Bowel Disease Center and Amsterdam Gastroenterology and Metabolism, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| |
Collapse
|
87
|
Zundler S, Becker E, Spocinska M, Slawik M, Parga-Vidal L, Stark R, Wiendl M, Atreya R, Rath T, Leppkes M, Hildner K, López-Posadas R, Lukassen S, Ekici AB, Neufert C, Atreya I, van Gisbergen KPJM, Neurath MF. Hobit- and Blimp-1-driven CD4 + tissue-resident memory T cells control chronic intestinal inflammation. Nat Immunol 2019; 20:288-300. [PMID: 30692620 DOI: 10.1038/s41590-018-0298-5] [Citation(s) in RCA: 160] [Impact Index Per Article: 26.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2018] [Accepted: 12/12/2018] [Indexed: 12/12/2022]
Abstract
Although tissue-resident memory T cells (TRM cells) have been shown to regulate host protection in infectious disorders, their function in inflammatory bowel disease (IBD) remains to be investigated. Here we characterized TRM cells in human IBD and in experimental models of intestinal inflammation. Pro-inflammatory TRM cells accumulated in the mucosa of patients with IBD, and the presence of CD4+CD69+CD103+ TRM cells was predictive of the development of flares. In vivo, functional impairment of TRM cells in mice with double knockout of the TRM-cell-associated transcription factors Hobit and Blimp-1 attenuated disease in several models of colitis, due to impaired cross-talk between the adaptive and innate immune system. Finally, depletion of TRM cells led to a suppression of colitis activity. Together, our data demonstrate a central role for TRM cells in the pathogenesis of chronic intestinal inflammation and suggest that these cells could be targets for future therapeutic approaches in IBD.
Collapse
Affiliation(s)
- Sebastian Zundler
- Department of Medicine 1, Kussmaul Campus for Medical Research and Translational Research Center, Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, Erlangen, Germany.,Department of Hematopoiesis, Sanquin Research and Landsteiner Laboratory, Amsterdam UMC, The University of Amsterdam, Amsterdam, Netherlands
| | - Emily Becker
- Department of Medicine 1, Kussmaul Campus for Medical Research and Translational Research Center, Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, Erlangen, Germany
| | - Marta Spocinska
- Department of Medicine 1, Kussmaul Campus for Medical Research and Translational Research Center, Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, Erlangen, Germany
| | - Monique Slawik
- Department of Medicine 1, Kussmaul Campus for Medical Research and Translational Research Center, Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, Erlangen, Germany
| | - Loreto Parga-Vidal
- Department of Hematopoiesis, Sanquin Research and Landsteiner Laboratory, Amsterdam UMC, The University of Amsterdam, Amsterdam, Netherlands
| | - Regina Stark
- Department of Hematopoiesis, Sanquin Research and Landsteiner Laboratory, Amsterdam UMC, The University of Amsterdam, Amsterdam, Netherlands
| | - Maximilian Wiendl
- Department of Medicine 1, Kussmaul Campus for Medical Research and Translational Research Center, Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, Erlangen, Germany
| | - Raja Atreya
- Department of Medicine 1, Kussmaul Campus for Medical Research and Translational Research Center, Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, Erlangen, Germany
| | - Timo Rath
- Department of Medicine 1, Kussmaul Campus for Medical Research and Translational Research Center, Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, Erlangen, Germany
| | - Moritz Leppkes
- Department of Medicine 1, Kussmaul Campus for Medical Research and Translational Research Center, Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, Erlangen, Germany
| | - Kai Hildner
- Department of Medicine 1, Kussmaul Campus for Medical Research and Translational Research Center, Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, Erlangen, Germany
| | - Rocío López-Posadas
- Department of Medicine 1, Kussmaul Campus for Medical Research and Translational Research Center, Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, Erlangen, Germany
| | - Sören Lukassen
- Institute of Human Genetics, Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, Erlangen, Germany
| | - Arif B Ekici
- Institute of Human Genetics, Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, Erlangen, Germany
| | - Clemens Neufert
- Department of Medicine 1, Kussmaul Campus for Medical Research and Translational Research Center, Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, Erlangen, Germany
| | - Imke Atreya
- Department of Medicine 1, Kussmaul Campus for Medical Research and Translational Research Center, Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, Erlangen, Germany
| | - Klaas P J M van Gisbergen
- Department of Hematopoiesis, Sanquin Research and Landsteiner Laboratory, Amsterdam UMC, The University of Amsterdam, Amsterdam, Netherlands.,Department of Experimental Immunology, Amsterdam UMC, The University of Amsterdam, Amsterdam, Netherlands
| | - Markus F Neurath
- Department of Medicine 1, Kussmaul Campus for Medical Research and Translational Research Center, Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, Erlangen, Germany.
| |
Collapse
|
88
|
Hoeppli RE, MacDonald KN, Leclair P, Fung VCW, Mojibian M, Gillies J, Rahavi SMR, Campbell AIM, Gandhi SK, Pesenacker AM, Reid G, Lim CJ, Levings MK. Tailoring the homing capacity of human Tregs for directed migration to sites of Th1-inflammation or intestinal regions. Am J Transplant 2019; 19:62-76. [PMID: 29766641 DOI: 10.1111/ajt.14936] [Citation(s) in RCA: 57] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2017] [Revised: 04/17/2018] [Accepted: 05/06/2018] [Indexed: 01/25/2023]
Abstract
Cell-based therapy with CD4+ FOXP3+ regulatory T cells (Tregs) is a promising strategy to limit organ rejection and graft-vs-host disease. Ongoing clinical applications have yet to consider how human Tregs could be modified to direct their migration to specific inflammation sites and/or tissues for more targeted immunosuppression. We show here that stable, homing-receptor-tailored human Tregs can be generated from thymic Tregs isolated from pediatric thymus or adult blood. To direct migration to Th1-inflammatory sites, addition of interferon-γ and IL-12 during Treg expansion produced suppressive, epigenetically stable CXCR3+ TBET+ FOXP3+ T helper (Th)1-Tregs. CXCR3 remained expressed after injection in vivo and Th1-Tregs migrated efficiently towards CXCL10 in vitro. To induce tissue-specific migration, addition of retinoic acid (RA) during Treg expansion induced expression of the gut-homing receptors α4β7-integrin and CCR9. FOXP3+ RA-Tregs had elevated expression of the functional markers latency-associated peptide and glycoprotein A repetitions predominant, increased suppressive capacity in vitro and migrated efficiently to healthy and inflamed intestine after injection into mice. Homing-receptor-tailored Tregs were epigenetically stable even after long-term exposure to inflammatory conditions, suppressive in vivo and characterized by Th1- or gut-homing-specific transcriptomes. Tailoring human thymic Treg homing during in vitro expansion offers a new and clinically applicable approach to improving the potency and specificity of Treg therapy.
Collapse
Affiliation(s)
- R E Hoeppli
- Department of Surgery, University of British Columbia & British Columbia Children's Hospital Research Institute, Vancouver, BC, Canada
| | - K N MacDonald
- School of Biomedical Engineering, University of British Columbia, Vancouver, BC, Canada.,Michael Smith Laboratories, University of British Columbia, Vancouver, BC, Canada
| | - P Leclair
- Department of Pediatrics, University of British Columbia & British Columbia Children's Hospital Research Institute, Vancouver, BC, Canada
| | - V C W Fung
- Department of Surgery, University of British Columbia & British Columbia Children's Hospital Research Institute, Vancouver, BC, Canada
| | - M Mojibian
- Department of Surgery, University of British Columbia & British Columbia Children's Hospital Research Institute, Vancouver, BC, Canada
| | - J Gillies
- Department of Surgery, University of British Columbia & British Columbia Children's Hospital Research Institute, Vancouver, BC, Canada
| | - S M R Rahavi
- Department of Pediatrics, University of British Columbia & British Columbia Children's Hospital Research Institute, Vancouver, BC, Canada
| | - A I M Campbell
- Department of Surgery, University of British Columbia & British Columbia Children's Hospital Research Institute, Vancouver, BC, Canada
| | - S K Gandhi
- Department of Surgery, University of British Columbia & British Columbia Children's Hospital Research Institute, Vancouver, BC, Canada
| | - A M Pesenacker
- Department of Surgery, University of British Columbia & British Columbia Children's Hospital Research Institute, Vancouver, BC, Canada
| | - G Reid
- Department of Pediatrics, University of British Columbia & British Columbia Children's Hospital Research Institute, Vancouver, BC, Canada
| | - C J Lim
- Department of Pediatrics, University of British Columbia & British Columbia Children's Hospital Research Institute, Vancouver, BC, Canada
| | - M K Levings
- Department of Surgery, University of British Columbia & British Columbia Children's Hospital Research Institute, Vancouver, BC, Canada.,School of Biomedical Engineering, University of British Columbia, Vancouver, BC, Canada
| |
Collapse
|
89
|
Forkel M, van Tol S, Höög C, Michaëlsson J, Almer S, Mjösberg J. Distinct Alterations in the Composition of Mucosal Innate Lymphoid Cells in Newly Diagnosed and Established Crohn's Disease and Ulcerative Colitis. J Crohns Colitis 2019; 13:67-78. [PMID: 30496425 DOI: 10.1093/ecco-jcc/jjy119] [Citation(s) in RCA: 92] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
BACKGROUND AND AIMS Innate lymphoid cells [ILC] have been suggested to play a role in inflammatory bowel disease [IBD]. Here, we investigated the ILC compartment in intestinal biopsies and blood from distinct patient groups with Crohn's disease [CD] and ulcerative colitis [UC], either newly diagnosed or with disease established for at least 1 year. This approach allowed us to simultaneously investigate temporal, disease-specific, and tissue-specific changes in ILC composition in IBD. METHODS ILC subset frequencies, phenotype, and transcription factor profile in blood and intestinal biopsies were investigated by multi-parameter flow cytometry analysis. Endoscopic disease severity was judged using the ulcerative colitis endoscopic index of severity and the simple endoscopic score for Crohn's disease. RESULTS The frequency of NKp44+ILC3 was decreased in inflamed tissue, both in patients with CD and those with UC, already at the time of diagnosis, and correlated with disease severity. Simultaneously, the frequency of ILC1 was increased in patients with CD, whereas the frequency of ILC2 was increased in patients with UC. However, in patients with established UC or CD, both ILC1 and ILC2 were increased. In contrast to the ILC composition in inflamed tissue, ILC in non-inflamed tissue or blood were unchanged compared with non-IBD controls. Finally, in patients undergoing treatment with an anti-α4β7 antibody the frequencies of ILC in peripheral blood remained unchanged. CONCLUSIONS We report both shared and distinct changes in ILC composition depending on diagnosis and disease duration. The alterations in ILC composition in IBD occur selectively at inflamed sites in the gut.
Collapse
Affiliation(s)
- Marianne Forkel
- Center for Infectious Medicine, Department of Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Sophie van Tol
- Center for Infectious Medicine, Department of Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Charlotte Höög
- Unit for Inflammation, Gastroenterology and Rheumatology, Department of Medicine, Huddinge, Karolinska Institutet, Stockholm, Sweden
| | - Jakob Michaëlsson
- Center for Infectious Medicine, Department of Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Sven Almer
- Department of Medicine, Solna, Karolinska Institutet, and IBD-Center, Gastroenterology, Karolinska University Hospital, Stockholm, Sweden
| | - Jenny Mjösberg
- Center for Infectious Medicine, Department of Medicine, Karolinska Institutet, Stockholm, Sweden.,Department of Clinical and Experimental Medicine, Linköping University, Linköping, Sweden
| |
Collapse
|
90
|
Zeissig S, Rosati E, Dowds CM, Aden K, Bethge J, Schulte B, Pan WH, Mishra N, Zuhayra M, Marx M, Paulsen M, Strigli A, Conrad C, Schuldt D, Sinha A, Ebsen H, Kornell SC, Nikolaus S, Arlt A, Kabelitz D, Ellrichmann M, Lützen U, Rosenstiel PC, Franke A, Schreiber S. Vedolizumab is associated with changes in innate rather than adaptive immunity in patients with inflammatory bowel disease. Gut 2019; 68:25-39. [PMID: 29730603 DOI: 10.1136/gutjnl-2018-316023] [Citation(s) in RCA: 160] [Impact Index Per Article: 26.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/13/2018] [Revised: 04/03/2018] [Accepted: 04/17/2018] [Indexed: 01/10/2023]
Abstract
OBJECTIVE Vedolizumab, a monoclonal antibody directed against the integrin heterodimer α4β7, is approved for the treatment of Crohn's disease and ulcerative colitis. The efficacy of vedolizumab has been suggested to result from inhibition of intestinal T cell trafficking although human data to support this conclusion are scarce. We therefore performed a comprehensive analysis of vedolizumab-induced alterations in mucosal and systemic immunity in patients with inflammatory bowel disease (IBD), using anti-inflammatory therapy with the TNFα antibody infliximab as control. DESIGN Immunophenotyping, immunohistochemistry, T cell receptor profiling and RNA sequencing were performed using blood and colonic biopsies from patients with IBD before and during treatment with vedolizumab (n=18) or, as control, the anti-TNFα antibody infliximab (n=20). Leucocyte trafficking in vivo was assessed using single photon emission computed tomography and endomicroscopy. RESULTS Vedolizumab was not associated with alterations in the abundance or phenotype of lamina propria T cells and did not affect the mucosal T cell repertoire or leucocyte trafficking in vivo. Surprisingly, however, α4β7 antibody treatment was associated with substantial effects on innate immunity including changes in macrophage populations and pronounced alterations in the expression of molecules involved in microbial sensing, chemoattraction and regulation of the innate effector response. These effects were specific to vedolizumab, not observed in response to the TNFα antibody infliximab, and associated with inhibition of intestinal inflammation. CONCLUSION Our findings suggest that modulation of innate immunity contributes to the therapeutic efficacy of vedolizumab in IBD. TRIAL REGISTRATION NUMBER NCT02694588.
Collapse
Affiliation(s)
- Sebastian Zeissig
- Department of Medicine I, Universitätsklinikum Carl Gustav Carus Dresden, Technische Universität (TU) Dresden, Dresden, Germany.,Center for Regenerative Therapies Dresden, Technische Universität (TU) Dresden, Dresden, Germany.,Department of Internal Medicine I, Universitätsklinikum Schleswig-Holstein, Campus Kiel, Kiel, Germany
| | - Elisa Rosati
- Institute of Clinical Molecular Biology, Universitätsklinikum Schleswig-Holstein, Campus Kiel, Kiel, Germany
| | - C Marie Dowds
- Department of Internal Medicine I, Universitätsklinikum Schleswig-Holstein, Campus Kiel, Kiel, Germany.,Institute of Clinical Molecular Biology, Universitätsklinikum Schleswig-Holstein, Campus Kiel, Kiel, Germany
| | - Konrad Aden
- Department of Internal Medicine I, Universitätsklinikum Schleswig-Holstein, Campus Kiel, Kiel, Germany.,Institute of Clinical Molecular Biology, Universitätsklinikum Schleswig-Holstein, Campus Kiel, Kiel, Germany
| | - Johannes Bethge
- Department of Internal Medicine I, Universitätsklinikum Schleswig-Holstein, Campus Kiel, Kiel, Germany
| | - Berenice Schulte
- Department of Internal Medicine I, Universitätsklinikum Schleswig-Holstein, Campus Kiel, Kiel, Germany
| | - Wei Hung Pan
- Institute of Clinical Molecular Biology, Universitätsklinikum Schleswig-Holstein, Campus Kiel, Kiel, Germany
| | - Neha Mishra
- Institute of Clinical Molecular Biology, Universitätsklinikum Schleswig-Holstein, Campus Kiel, Kiel, Germany
| | - Maaz Zuhayra
- Department of Nuclear Medicine, Molecular Diagnostic Imaging and Therapy, Universitätsklinikum Schleswig-Holstein, Campus Kiel, Kiel, Germany
| | - Marlies Marx
- Department of Nuclear Medicine, Molecular Diagnostic Imaging and Therapy, Universitätsklinikum Schleswig-Holstein, Campus Kiel, Kiel, Germany
| | - Maren Paulsen
- Institute of Clinical Molecular Biology, Universitätsklinikum Schleswig-Holstein, Campus Kiel, Kiel, Germany
| | - Anne Strigli
- Department of Medicine I, Universitätsklinikum Carl Gustav Carus Dresden, Technische Universität (TU) Dresden, Dresden, Germany.,Center for Regenerative Therapies Dresden, Technische Universität (TU) Dresden, Dresden, Germany
| | - Claudio Conrad
- Department of Internal Medicine I, Universitätsklinikum Schleswig-Holstein, Campus Kiel, Kiel, Germany
| | - Dörthe Schuldt
- Department of Internal Medicine I, Universitätsklinikum Schleswig-Holstein, Campus Kiel, Kiel, Germany
| | - Anupam Sinha
- Institute of Clinical Molecular Biology, Universitätsklinikum Schleswig-Holstein, Campus Kiel, Kiel, Germany
| | - Henriette Ebsen
- Institute of Immunology, Christian-Albrechts-Universität zu Kiel, Kiel, Germany
| | - Sabin-Christin Kornell
- Department of Internal Medicine I, Universitätsklinikum Schleswig-Holstein, Campus Kiel, Kiel, Germany.,Institute of Clinical Molecular Biology, Universitätsklinikum Schleswig-Holstein, Campus Kiel, Kiel, Germany
| | - Susanna Nikolaus
- Department of Internal Medicine I, Universitätsklinikum Schleswig-Holstein, Campus Kiel, Kiel, Germany
| | - Alexander Arlt
- Department of Internal Medicine I, Universitätsklinikum Schleswig-Holstein, Campus Kiel, Kiel, Germany
| | - Dieter Kabelitz
- Institute of Immunology, Christian-Albrechts-Universität zu Kiel, Kiel, Germany
| | - Mark Ellrichmann
- Department of Internal Medicine I, Universitätsklinikum Schleswig-Holstein, Campus Kiel, Kiel, Germany
| | - Ulf Lützen
- Department of Nuclear Medicine, Molecular Diagnostic Imaging and Therapy, Universitätsklinikum Schleswig-Holstein, Campus Kiel, Kiel, Germany
| | - Philip C Rosenstiel
- Institute of Clinical Molecular Biology, Universitätsklinikum Schleswig-Holstein, Campus Kiel, Kiel, Germany
| | - Andre Franke
- Institute of Clinical Molecular Biology, Universitätsklinikum Schleswig-Holstein, Campus Kiel, Kiel, Germany
| | - Stefan Schreiber
- Department of Internal Medicine I, Universitätsklinikum Schleswig-Holstein, Campus Kiel, Kiel, Germany.,Institute of Clinical Molecular Biology, Universitätsklinikum Schleswig-Holstein, Campus Kiel, Kiel, Germany
| |
Collapse
|
91
|
Stagg AJ. Intestinal Dendritic Cells in Health and Gut Inflammation. Front Immunol 2018; 9:2883. [PMID: 30574151 PMCID: PMC6291504 DOI: 10.3389/fimmu.2018.02883] [Citation(s) in RCA: 143] [Impact Index Per Article: 20.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2018] [Accepted: 11/23/2018] [Indexed: 12/19/2022] Open
Abstract
Dendritic cells (DCs) mediate tolerance to food antigens, limit reactivity to the gut microbiota and are required for optimal response to intestinal pathogens. Intestinal DCs are heterogeneous but collectively generate both regulatory and effector T cell responses. The balance of outcomes is determined by the activity of functionally distinct DC subsets and their modulation by environmental cues. DCs constantly sample luminal content to monitor for pathogens; the significance of the various pathways by which this occurs is incompletely understood. Intestinal DC have distinctive properties shaped by local host, dietary and microbial signals. These properties include the ability to produce all-trans retinoic acid (RA) and imprint gut tropism on T cells they activate. In the steady-state, subsets of intestinal DC are potent generators of inducible Treg, aided by their ability to activate TGFβ and produce RA. However, responses induced by steady-state intestinal DCs are not exclusively regulatory in nature; effector T cells with specificity for commensal bacterial can be found in the healthy mucosa and these can be locally controlled to prevent inflammation. The ability of intestinal DCs to enhance effector responses in infection or sustain inflammation in disease is likely to involve both modulation of the local DC population and recruitment of additional populations. Immune pathways in the pathogenesis of inflammatory bowel disease can be mapped to DCs and in inflamed intestinal tissue, DCs show increased expression of microbial recognition machinery, activation, and production of key immunological mediators. Intestinal DCs may be targeted for disease therapy or to improve vaccine responses.
Collapse
Affiliation(s)
- Andrew J Stagg
- Centre for Immunobiology, Blizard Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom
| |
Collapse
|
92
|
Iborra M, Beltrán B, Maroto N, Navarro-Cortés P, Boscá-Watts M, Ferrer-Bradley I, García-Morales N, Sáez-González E, Hinojosa J, Mínguez M, Nos P. Vedolizumab, una opción en pacientes con enfermedad inflamatoria intestinal intolerantes a tiopurinas y refractarios a biológicos. GASTROENTEROLOGIA Y HEPATOLOGIA 2018; 41:535-543. [DOI: 10.1016/j.gastrohep.2018.06.001] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/01/2018] [Revised: 05/23/2018] [Accepted: 06/10/2018] [Indexed: 02/07/2023]
|
93
|
Lutter L, Hoytema van Konijnenburg DP, Brand EC, Oldenburg B, van Wijk F. The elusive case of human intraepithelial T cells in gut homeostasis and inflammation. Nat Rev Gastroenterol Hepatol 2018; 15:637-649. [PMID: 29973676 DOI: 10.1038/s41575-018-0039-0] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
The epithelial barrier of the gastrointestinal tract is home to numerous intraepithelial T cells (IETs). IETs are functionally adapted to the mucosal environment and are among the first adaptive immune cells to encounter microbial and dietary antigens. They possess hallmark features of tissue-resident T cells: they are long-lived nonmigratory cells capable of rapidly responding to antigen challenges independent of T cell recruitment from the periphery. Gut-resident T cells have been implicated in the relapsing and remitting course and persisting low-grade inflammation of chronic gastrointestinal diseases, including IBD and coeliac disease. So far, most data IETs have been derived from experimental animal models; however, IETs and the environmental makeup differ between mice and humans. With advances in techniques, the number of human studies has grown exponentially in the past 5 years. Here, we review the literature on the involvement of human IETs in gut homeostasis and inflammation, and how these cells are influenced by the microbiota and dietary antigens. Finally, targeting of IETs in therapeutic interventions is discussed. Broad insight into the function and role of human IETs in gut homeostasis and inflammation is essential to identify future diagnostic, prognostic and therapeutic strategies.
Collapse
Affiliation(s)
- Lisanne Lutter
- Laboratory of Translational Immunology, Department of Pediatric Immunology, University Medical Center Utrecht, Utrecht, Netherlands.,Department of Gastroenterology and Hepatology, University Medical Center Utrecht, Utrecht, Netherlands
| | - David P Hoytema van Konijnenburg
- Laboratory of Translational Immunology, Department of Pediatric Immunology, University Medical Center Utrecht, Utrecht, Netherlands.,Laboratory of Mucosal Immunology, The Rockefeller University, New York, NY, USA
| | - Eelco C Brand
- Laboratory of Translational Immunology, Department of Pediatric Immunology, University Medical Center Utrecht, Utrecht, Netherlands.,Department of Gastroenterology and Hepatology, University Medical Center Utrecht, Utrecht, Netherlands
| | - Bas Oldenburg
- Department of Gastroenterology and Hepatology, University Medical Center Utrecht, Utrecht, Netherlands
| | - Femke van Wijk
- Laboratory of Translational Immunology, Department of Pediatric Immunology, University Medical Center Utrecht, Utrecht, Netherlands.
| |
Collapse
|
94
|
Ammitzbøll C, von Essen MR, Börnsen L, Petersen ER, McWilliam O, Ratzer R, Romme Christensen J, Oturai AB, Søndergaard HB, Sellebjerg F. GPR15 + T cells are Th17 like, increased in smokers and associated with multiple sclerosis. J Autoimmun 2018; 97:114-121. [PMID: 30245027 DOI: 10.1016/j.jaut.2018.09.005] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2018] [Revised: 09/07/2018] [Accepted: 09/12/2018] [Indexed: 01/15/2023]
Abstract
Smoking is a risk factor for the development and progression of multiple sclerosis (MS); however, the pathogenic effects of smoking are poorly understood. We studied the smoking-associated chemokine receptor-like molecule GPR15 in relation to relapsing-remitting MS (RRMS). Using microarray analyses and qPCR we found elevated GPR15 in blood cells from smokers, and increased GPR15 expression in RRMS. By flow cytometry we detected increased frequencies of GPR15 expressing T and B cells in smokers, but no difference between patients with RRMS and healthy controls. However, after cell culture with the autoantigens myelin basic protein (MBP) and myelin oligodendrocyte glycoprotein, frequencies of MBP-reactive and non-proliferating GPR15+CD4+ T cells were increased in patients with RRMS compared with healthy controls. GPR15+CD4+ T cells produced IL-17 and were enriched in the cerebrospinal fluid (CSF). Furthermore, in the CSF of patients with RRMS, GPR15+ T cells were associated with CCR6+CXCR3+/CCR6-CXCR3+ phenotypes and correlated positively with concentrations of the newly identified GPR15-ligand (GPR15L), myelin degradation and disability. In conclusion, we have identified a proinflammatory cell type linking smoking with pathogenic immune cell functions in RRMS.
Collapse
Affiliation(s)
- Cecilie Ammitzbøll
- Danish Multiple Sclerosis Center, Department of Neurology, Rigshospitalet, University of Copenhagen, Blegdamsvej 9, DK-2100, Denmark.
| | - Marina R von Essen
- Danish Multiple Sclerosis Center, Department of Neurology, Rigshospitalet, University of Copenhagen, Blegdamsvej 9, DK-2100, Denmark.
| | - Lars Börnsen
- Danish Multiple Sclerosis Center, Department of Neurology, Rigshospitalet, University of Copenhagen, Blegdamsvej 9, DK-2100, Denmark.
| | - Eva Rosa Petersen
- Danish Multiple Sclerosis Center, Department of Neurology, Rigshospitalet, University of Copenhagen, Blegdamsvej 9, DK-2100, Denmark.
| | - Oskar McWilliam
- Danish Multiple Sclerosis Center, Department of Neurology, Rigshospitalet, University of Copenhagen, Blegdamsvej 9, DK-2100, Denmark.
| | - Rikke Ratzer
- Danish Multiple Sclerosis Center, Department of Neurology, Rigshospitalet, University of Copenhagen, Blegdamsvej 9, DK-2100, Denmark.
| | - Jeppe Romme Christensen
- Danish Multiple Sclerosis Center, Department of Neurology, Rigshospitalet, University of Copenhagen, Blegdamsvej 9, DK-2100, Denmark.
| | - Annette B Oturai
- Danish Multiple Sclerosis Center, Department of Neurology, Rigshospitalet, University of Copenhagen, Blegdamsvej 9, DK-2100, Denmark.
| | - Helle B Søndergaard
- Danish Multiple Sclerosis Center, Department of Neurology, Rigshospitalet, University of Copenhagen, Blegdamsvej 9, DK-2100, Denmark.
| | - Finn Sellebjerg
- Danish Multiple Sclerosis Center, Department of Neurology, Rigshospitalet, University of Copenhagen, Blegdamsvej 9, DK-2100, Denmark.
| |
Collapse
|
95
|
Becker E, Schramm S, Binder MT, Allner C, Wiendl M, Neufert C, Atreya I, Neurath M, Zundler S. Dynamic Adhesion Assay for the Functional Analysis of Anti-adhesion Therapies in Inflammatory Bowel Disease. J Vis Exp 2018. [PMID: 30295649 DOI: 10.3791/58210] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Gut homing of immune cells is important for the pathogenesis of inflammatory bowel diseases (IBD). Integrin-dependent cell adhesion to addressins is a crucial step in this process and therapeutic strategies interfering with adhesion have been successfully established. The anti-α4β7 integrin antibody, vedolizumab, is used for the clinical treatment of Crohn's disease (CD) and ulcerative colitis (UC) and further compounds are likely to follow. The details of the adhesion procedure and the action mechanisms of anti-integrin antibodies are still unclear in many regards due to the limited available techniques for the functional research in this field. Here, we present a dynamic adhesion assay for the functional analysis of human cell adhesion under flow conditions and the impact of anti-integrin therapies in the context of IBD. It is based on the perfusion of primary human cells through addressin-coated ultrathin glass capillaries with real-time microscopic analysis. The assay offers a variety of opportunities for refinements and modifications and holds potentials for mechanistic discoveries and translational applications.
Collapse
Affiliation(s)
- Emily Becker
- Department of Medicine 1, University of Erlangen-Nuremberg
| | | | | | | | | | | | - Imke Atreya
- Department of Medicine 1, University of Erlangen-Nuremberg
| | - Markus Neurath
- Department of Medicine 1, University of Erlangen-Nuremberg
| | | |
Collapse
|
96
|
Lamb CA, O'Byrne S, Keir ME, Butcher EC. Gut-Selective Integrin-Targeted Therapies for Inflammatory Bowel Disease. J Crohns Colitis 2018; 12:S653-S668. [PMID: 29767705 DOI: 10.1093/ecco-jcc/jjy060] [Citation(s) in RCA: 59] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Integrins are cell surface receptors with bidirectional signalling capabilities that can bind to adhesion molecules in order to mediate homing of leukocytes to peripheral tissues. Gut-selective leukocyte homing is facilitated by interactions between α4β7 and its ligand, mucosal addressin cellular adhesion molecule-1 [MAdCAM-1], while retention of lymphocytes in mucosal tissues is mediated by αEβ7 binding to its ligand E-cadherin. Therapies targeting gut-selective trafficking have shown efficacy in inflammatory bowel disease [IBD], confirming the importance of leukocyte trafficking in disease pathobiology. This review will provide an overview of integrin structure, function and signalling, and highlight the role that these molecules play in leukocyte homing and retention. Anti-integrin therapeutics, including gut-selective antibodies against the β7 integrin subunit [etrolizumab] and the α4β7 integrin heterodimer [vedolizumab and abrilumab], and the non-gut selective anti-α4 integrin [natalizumab], will be discussed, as well as novel targeting approaches using small molecules.
Collapse
Affiliation(s)
- Christopher A Lamb
- Institute of Cellular Medicine, Newcastle University, Newcastle upon Tyne, UK.,Department of Gastroenterology, Newcastle upon Tyne Hospitals NHS Foundation Trust, Newcastle upon Tyne, UK
| | - Sharon O'Byrne
- Global Medical Affairs, Takeda Pharmaceuticals International AG, Zurich, Switzerland
| | - Mary E Keir
- Genentech Research & Early Development, South San Francisco, CA, USA
| | - Eugene C Butcher
- Laboratory of Immunology and Vascular Biology, Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA.,Veterans Affairs Palo Alto Health Care System and The Palo Alto Veterans Institute for Research, Palo Alto, CA, USA
| |
Collapse
|
97
|
Haase T, Müller C, Krause J, Röthemeier C, Stenzig J, Kunze S, Waldenberger M, Münzel T, Pfeiffer N, Wild PS, Michal M, Marini F, Karakas M, Lackner KJ, Blankenberg S, Zeller T. Novel DNA Methylation Sites Influence GPR15 Expression in Relation to Smoking. Biomolecules 2018; 8:biom8030074. [PMID: 30127295 PMCID: PMC6163736 DOI: 10.3390/biom8030074] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2018] [Revised: 08/06/2018] [Accepted: 08/09/2018] [Indexed: 11/24/2022] Open
Abstract
Smoking is a major risk factor for cardiovascular diseases and has been implicated in the regulation of the G protein-coupled receptor 15 (GPR15) by affecting CpG methylation. The G protein-coupled receptor 15 is involved in angiogenesis and inflammation. An effect on GPR15 gene regulation has been shown for the CpG site CpG3.98251294. We aimed to analyze the effect of smoking on GPR15 expression and methylation sites spanning the GPR15 locus. DNA methylation of nine GPR15 CpG sites was measured in leukocytes from 1291 population-based individuals using the EpiTYPER. Monocytic GPR15 expression was measured by qPCR at baseline and five-years follow up. GPR15 gene expression was upregulated in smokers (beta (ß) = −2.699, p-value (p) = 1.02 × 10−77) and strongly correlated with smoking exposure (ß = −0.063, p = 2.95 × 10−34). Smoking cessation within five years reduced GPR15 expression about 19% (p = 9.65 × 10−5) with decreasing GPR15 expression over time (ß = 0.031, p = 3.81 × 10−6). Additionally, three novel CpG sites within GPR15 affected by smoking were identified. For CpG3.98251047, DNA methylation increased steadily after smoking cessation (ß = 0.123, p = 1.67 × 10−3) and strongly correlated with changes in GPR15 expression (ß = 0.036, p = 4.86 × 10−5). Three novel GPR15 CpG sites were identified in relation to smoking and GPR15 expression. Our results provide novel insights in the regulation of GPR15, which possibly linked smoking to inflammation and disease progression.
Collapse
Affiliation(s)
- Tina Haase
- Clinic for General and Interventional Cardiology, University Heart Center Hamburg, 20246 Hamburg, Germany.
- German Centre for Cardiovascular Research (DZHK), 13316 Berlin, Germany.
| | - Christian Müller
- Clinic for General and Interventional Cardiology, University Heart Center Hamburg, 20246 Hamburg, Germany.
- German Centre for Cardiovascular Research (DZHK), 13316 Berlin, Germany.
| | - Julia Krause
- Clinic for General and Interventional Cardiology, University Heart Center Hamburg, 20246 Hamburg, Germany.
- German Centre for Cardiovascular Research (DZHK), 13316 Berlin, Germany.
| | - Caroline Röthemeier
- Clinic for General and Interventional Cardiology, University Heart Center Hamburg, 20246 Hamburg, Germany.
| | - Justus Stenzig
- German Centre for Cardiovascular Research (DZHK), 13316 Berlin, Germany.
- Institute of Experimental Pharmacology and Toxicology, University Medical Center Hamburg-Eppendorf (UKE), 20246 Hamburg, Germany.
| | - Sonja Kunze
- Research Unit of Molecular Epidemiology, Helmholtz Zentrum München, German Research Center for Environmental Health, 85764 Neuherberg, Germany.
- Institute of Epidemiology II, Helmholtz Zentrum München, German Research Center for Environmental Health, 85764 Neuherberg, Germany.
| | - Melanie Waldenberger
- German Centre for Cardiovascular Research (DZHK), 13316 Berlin, Germany.
- Research Unit of Molecular Epidemiology, Helmholtz Zentrum München, German Research Center for Environmental Health, 85764 Neuherberg, Germany.
- Institute of Epidemiology II, Helmholtz Zentrum München, German Research Center for Environmental Health, 85764 Neuherberg, Germany.
| | - Thomas Münzel
- German Centre for Cardiovascular Research (DZHK), 13316 Berlin, Germany.
- Center for Cardiology, Cardiology I, University Medical Center Mainz, Johannes Gutenberg University-Mainz, 55131 Mainz, Germany.
- Center for Thrombosis and Hemostasis, University Medical Center Mainz, Johannes Gutenberg-University Mainz, 55131 Mainz, Germany.
- Center for Translational Vascular Biology (CTVB), University Medical Center Mainz, Johannes Gutenberg-University Mainz, 55131 Mainz, Germany.
| | - Norbert Pfeiffer
- Department of Ophthalmology, University Medical Center of the Johannes Gutenberg-University Mainz, 55131 Mainz, Germany.
| | - Philipp S Wild
- German Centre for Cardiovascular Research (DZHK), 13316 Berlin, Germany.
- Center for Translational Vascular Biology (CTVB), University Medical Center Mainz, Johannes Gutenberg-University Mainz, 55131 Mainz, Germany.
- Preventive Cardiology and Preventive Medicine, Center for Cardiology, University Medical Center of the Johannes Gutenberg-University Mainz, 55131 Mainz, Germany.
- Center for Thrombosis and Hemostasis, University Medical Center of the Johannes Gutenberg-University Mainz, 55131 Mainz, Germany.
| | - Matthias Michal
- Department of Psychosomatic Medicine and Psychotherapy, University Medical Center of the Johannes Gutenberg-University Mainz, 55131 Mainz, Germany.
| | - Federico Marini
- University Medical Center, Institute of Medical Biostatistics, Epidemiology and Informatics (IMBEI), 55131 Mainz, Germany.
| | - Mahir Karakas
- Clinic for General and Interventional Cardiology, University Heart Center Hamburg, 20246 Hamburg, Germany.
- German Centre for Cardiovascular Research (DZHK), 13316 Berlin, Germany.
| | - Karl J Lackner
- German Centre for Cardiovascular Research (DZHK), 13316 Berlin, Germany.
- Institute of Clinical Chemistry and Laboratory Medicine, University Medical Center, Johannes Gutenberg University Mainz, 55131 Mainz, Germany.
| | - Stefan Blankenberg
- Clinic for General and Interventional Cardiology, University Heart Center Hamburg, 20246 Hamburg, Germany.
- German Centre for Cardiovascular Research (DZHK), 13316 Berlin, Germany.
| | - Tanja Zeller
- Clinic for General and Interventional Cardiology, University Heart Center Hamburg, 20246 Hamburg, Germany.
- German Centre for Cardiovascular Research (DZHK), 13316 Berlin, Germany.
| |
Collapse
|
98
|
Rath T, Billmeier U, Ferrazzi F, Vieth M, Ekici A, Neurath MF, Atreya R. Effects of Anti-Integrin Treatment With Vedolizumab on Immune Pathways and Cytokines in Inflammatory Bowel Diseases. Front Immunol 2018; 9:1700. [PMID: 30131801 PMCID: PMC6090141 DOI: 10.3389/fimmu.2018.01700] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2018] [Accepted: 07/10/2018] [Indexed: 12/16/2022] Open
Abstract
Background and aims Despite proven clinical efficacy of vedolizumab (VDZ) for inducing and maintaining remission in patients with Crohn’s disease (CD) and ulcerative colitis (UC), subgroups of patients have no therapeutic benefit from anti-α4β7 integrin therapy with VDZ. Within this study, we aimed to identify genetic, cellular, and immunological mechanisms that define response and failure to VDZ treatment. Methods Intestinal RNA sequencing was performed in UC and CD patients before and at week 14 of VDZ therapy. α4β7 expression on peripheral and mucosal immune cells was assessed by flow cytometry and immunohistochemistry. Cellular modes of VDZ-mediated action were analyzed ex vivo and in VDZ-treated inflammatory bowel disease patients. Results Transcriptome analysis showed an impairment of signaling cascades associated with adhesion, diapedesis, and migration of granulocytes and agranulocytes upon VDZ therapy. In non-remitters to VDZ therapy, a tissue destructive and leukocyte-mediated inflammatory activity with activation of TNF-dependent pathways was present, all of which were inhibited in remitters to VDZ. Clinical remission was associated with a significant reduction of α4β7 expression on Th2 and Th17 polarized mucosal CD4+ T cells at week 14 of VDZ therapy and with significantly higher numbers of α4β7-expressing mucosal cells prior to the initiation of VDZ therapy compared with non-remitters. Conclusion Intestinal α4β7 expression prior to VDZ therapy might represent a biomarker that predicts therapeutic response to subsequent VDZ treatment. Due to high activation of TNF signaling in VDZ non-remitters, anti-TNF treatment might represent a promising therapeutic strategy in VDZ refractory patients.
Collapse
Affiliation(s)
- Timo Rath
- Department of Medicine 1, Division of Gastroenterology, Pneumology and Endocrinology, Ludwig Demling Endoscopy Center of Excellence, University of Erlangen-Nuremberg, Erlangen, Germany
| | - Ulrike Billmeier
- Department of Medicine 1, Division of Gastroenterology, Pneumology and Endocrinology, Ludwig Demling Endoscopy Center of Excellence, University of Erlangen-Nuremberg, Erlangen, Germany
| | - Fulvia Ferrazzi
- Institute of Human Genetics, Friedrich-Alexander-Universität Erlangen-Nuremberg, Erlangen, Germany
| | - Michael Vieth
- Institute of Pathology, Klinikum Bayreuth, Bayreuth, Germany
| | - Arif Ekici
- Institute of Human Genetics, Friedrich-Alexander-Universität Erlangen-Nuremberg, Erlangen, Germany
| | - Markus F Neurath
- Department of Medicine 1, Division of Gastroenterology, Pneumology and Endocrinology, Ludwig Demling Endoscopy Center of Excellence, University of Erlangen-Nuremberg, Erlangen, Germany
| | - Raja Atreya
- Department of Medicine 1, Division of Gastroenterology, Pneumology and Endocrinology, Ludwig Demling Endoscopy Center of Excellence, University of Erlangen-Nuremberg, Erlangen, Germany
| |
Collapse
|
99
|
McAleer JP, Fan J, Roar B, Primerano DA, Denvir J. Cytokine Regulation in Human CD4 T Cells by the Aryl Hydrocarbon Receptor and Gq-Coupled Receptors. Sci Rep 2018; 8:10954. [PMID: 30026493 PMCID: PMC6053392 DOI: 10.1038/s41598-018-29262-4] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2017] [Accepted: 07/09/2018] [Indexed: 12/11/2022] Open
Abstract
Th17 cells contribute to host defense on mucosal surfaces but also provoke autoimmune diseases when directed against self-antigens. Identifying therapeutic targets that regulate Th17 cell differentiation and/or cytokine production has considerable value. Here, we study the aryl hydrocarbon receptor (AhR)-dependent transcriptome in human CD4 T cells treated with Th17-inducing cytokines. We show that the AhR reciprocally regulates IL-17 and IL-22 production in human CD4 T cells. Global gene expression analysis revealed that AhR ligation decreased IL21 expression, correlating with delayed upregulation of RORC during culture with Th17-inducing cytokines. Several of the AhR-dependent genes have known roles in cellular assembly, organization, development, growth and proliferation. We further show that expression of GPR15, GPR55 and GPR68 positively correlates with IL-22 production in the presence of the AhR agonist FICZ. Activation of GPR68 with the lorazepam derivative ogerin resulted in suppression of IL-22 and IL-10 secretion by T cells, with no effect on IL-17. Under neutral Th0 conditions, ogerin and the Gq/11 receptor inhibitor YM254890 blunted IL-22 induction by FICZ. These data reveal the AhR-dependent transcriptome in human CD4 T cells and suggest the mechanism through which the AhR regulates T cell function may be partially dependent on Gq-coupled receptors including GPR68.
Collapse
Affiliation(s)
- Jeremy P McAleer
- Department of Pharmaceutical Science and Research, Marshall University School of Pharmacy, Huntington, WV, 25755, USA.
| | - Jun Fan
- Department of Biomedical Sciences, Joan C. Edwards School of Medicine, Marshall University, Huntington, WV, 25755, USA
| | - Bryanna Roar
- Department of Pharmaceutical Science and Research, Marshall University School of Pharmacy, Huntington, WV, 25755, USA
| | - Donald A Primerano
- Department of Biomedical Sciences, Joan C. Edwards School of Medicine, Marshall University, Huntington, WV, 25755, USA
| | - James Denvir
- Department of Biomedical Sciences, Joan C. Edwards School of Medicine, Marshall University, Huntington, WV, 25755, USA
| |
Collapse
|
100
|
Gill PA, van Zelm MC, Muir JG, Gibson PR. Review article: short chain fatty acids as potential therapeutic agents in human gastrointestinal and inflammatory disorders. Aliment Pharmacol Ther 2018; 48:15-34. [PMID: 29722430 DOI: 10.1111/apt.14689] [Citation(s) in RCA: 330] [Impact Index Per Article: 47.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/20/2017] [Revised: 02/09/2018] [Accepted: 04/06/2018] [Indexed: 12/12/2022]
Abstract
BACKGROUND Butyrate, propionate and acetate are short chain fatty acids (SCFA), important for maintaining a healthy colon and are considered as protective in colorectal carcinogenesis. However, they may also regulate immune responses and the composition of the intestinal microbiota. Consequently, their importance in a variety of chronic inflammatory diseases is emerging. AIMS To review the physiology and metabolism of SCFA in humans, cellular and molecular mechanisms by which SCFA may act in health and disease, and approaches for therapeutic delivery of SCFA. METHODS A PubMed literature search was conducted for clinical and pre-clinical studies using search terms: 'dietary fibre', short-chain fatty acids', 'acetate', 'propionate', 'butyrate', 'inflammation', 'immune', 'gastrointestinal', 'metabolism'. RESULTS A wide range of pre-clinical evidence supports roles for SCFA as modulators of not only colonic function, but also multiple inflammatory and metabolic processes. SCFA are implicated in many autoimmune, allergic and metabolic diseases. However, translating effects of SCFA from animal studies to human disease is limited by physiological and dietary differences and by the challenge of delivering sufficient amounts of SCFA to the target sites that include the colon and the systemic circulation. Development of novel targeted approaches for colonic delivery, combined with postbiotic supplementation, may represent desirable strategies to achieve adequate targeted SCFA delivery. CONCLUSIONS There is a large array of potential disease-modulating effects of SCFA. Adequate targeted delivery to the sites of action is the main limitation of such application. The ongoing development and evaluation of novel delivery techniques offer potential for translating promise to therapeutic benefit.
Collapse
Affiliation(s)
- P A Gill
- Department of Gastroenterology, Central Clinical School, Monash University and Alfred Hospital, Melbourne, Vic., Australia.,Department of Immunology and Pathology, Central Clinical School, Monash University and Alfred Hospital, Melbourne, Vic, Australia
| | - M C van Zelm
- Department of Immunology and Pathology, Central Clinical School, Monash University and Alfred Hospital, Melbourne, Vic, Australia
| | - J G Muir
- Department of Gastroenterology, Central Clinical School, Monash University and Alfred Hospital, Melbourne, Vic., Australia
| | - P R Gibson
- Department of Gastroenterology, Central Clinical School, Monash University and Alfred Hospital, Melbourne, Vic., Australia
| |
Collapse
|