1
|
Risnes LF, Reims HM, Doyle RM, Qiao SW, Sollid LM, Lundin KEA, Christophersen A. Gluten-Free Diet Induces Rapid Changes in Phenotype and Survival Properties of Gluten-Specific T Cells in Celiac Disease. Gastroenterology 2024; 167:250-263. [PMID: 38552723 DOI: 10.1053/j.gastro.2024.03.027] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/26/2023] [Revised: 03/16/2024] [Accepted: 03/18/2024] [Indexed: 04/29/2024]
Abstract
BACKGROUND & AIMS The treatment of celiac disease (CeD) with gluten-free diet (GFD) normalizes gut inflammation and disease-specific antibodies. CeD patients have HLA-restricted, gluten-specific T cells persisting in the blood and gut even after decades of GFD, which are reactivated and disease driving upon gluten exposure. Our aim was to examine the transition of activated gluten-specific T cells into a pool of persisting memory T cells concurrent with normalization of clinically relevant biomarkers during the first year of treatment. METHODS We followed 17 CeD patients during their initial GFD year, leading to disease remission. We assessed activation and frequency of gluten-specific CD4+ blood and gut T cells with HLA-DQ2.5:gluten tetramers and flow cytometry, disease-specific serology, histology, and symptom scores. We assessed gluten-specific blood T cells within the first 3 weeks of GFD in 6 patients and serology in an additional 9 patients. RESULTS Gluten-specific CD4+ T cells peaked in blood at day 14 while up-regulating Bcl-2 and down-regulating Ki-67 and then decreased in frequency within 10 weeks of GFD. CD38, ICOS, HLA-DR, and Ki-67 decreased in gluten-specific cells within 3 days. PD-1, CD39, and OX40 expression persisted even after 12 months. IgA-transglutaminase 2 decreased significantly within 4 weeks. CONCLUSIONS GFD induces rapid changes in the phenotype and number of gluten-specific CD4+ blood T cells, including a peak of nonproliferating, nonapoptotic cells at day 14. Subsequent alterations in T-cell phenotype associate with the quiescent but chronic nature of treated CeD. The rapid changes affecting gluten-specific T cells and disease-specific antibodies offer opportunities for clinical trials aiming at developing nondietary treatments for patients with newly diagnosed CeD.
Collapse
Affiliation(s)
- Louise F Risnes
- KG Jebsen Coeliac Disease Research Centre, University of Oslo, Oslo, Norway; Faculty of Medicine, University of Oslo, Oslo, Norway; Department of Immunology, Oslo University Hospital, Oslo, Norway
| | - Henrik M Reims
- Department of Pathology, Oslo University Hospital, Oslo, Norway
| | - Ronan M Doyle
- Department of Pathology, Oslo University Hospital, Oslo, Norway
| | - Shuo-Wang Qiao
- Faculty of Medicine, University of Oslo, Oslo, Norway; Department of Immunology, Oslo University Hospital, Oslo, Norway
| | - Ludvig M Sollid
- KG Jebsen Coeliac Disease Research Centre, University of Oslo, Oslo, Norway; Faculty of Medicine, University of Oslo, Oslo, Norway; Department of Immunology, Oslo University Hospital, Oslo, Norway
| | - Knut E A Lundin
- KG Jebsen Coeliac Disease Research Centre, University of Oslo, Oslo, Norway; Faculty of Medicine, University of Oslo, Oslo, Norway; Department of Gastroenterology, Oslo University Hospital, Oslo, Norway
| | - Asbjørn Christophersen
- KG Jebsen Coeliac Disease Research Centre, University of Oslo, Oslo, Norway; Faculty of Medicine, University of Oslo, Oslo, Norway; Department of Immunology, Oslo University Hospital, Oslo, Norway; Institute of Basic Medical Sciences, University of Oslo, Oslo, Norway.
| |
Collapse
|
2
|
Functional implications of the CpG island methylation in the pathogenesis of celiac disease. Mol Biol Rep 2022; 49:10051-10064. [PMID: 35633417 DOI: 10.1007/s11033-022-07585-w] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2022] [Accepted: 05/09/2022] [Indexed: 10/18/2022]
Abstract
Investigation of gene-environment cross talk through epigenetic modifications led to better understanding of the number of complex diseases. Clinical heterogeneity and differential treatment response often contributed by the epigenetic signatures which could be personal. DNA methylation at CpG islands presents a critical nuclear process as a result of gene-environment interactions. These CpG islands are frequently present near the promoter sequence of genes and get differentially methylated under specific environmental conditions. Technical advancements facilitate in high throughput screening of differentially methylated CpG islands. Recent epigenetic studies unraveled several CD susceptibility genes expressed in peripheral blood lymphocytes (PBLs), duodenal mucosa, lamina and epithelial cells that are influenced by differentially methylated CpG islands. Here we highlighted these susceptibility genes; classify these genes based on cellular functions and tissue of expression. We further discussed how these genes interacts with each other to influence critical pathways like NF-κB signaling pathway, IL-17 signaling cascade, RIG-I like receptor signaling pathway, NOD-like receptor pathways among several others. This review also shed light on how gut microbiota may lead to the differential methylation of CpG islands of CD susceptibility genes. Large scale epigenetic studies followed by estimation of heritability of these CpG methylation and polygenic risk score estimation of these genes would prioritize potentially druggable targets for better therapeutics. In vivo studies are warranted to unravel further cellular responses to CpG methylation.
Collapse
|
3
|
Klonarakis M, Andrews CN, Raman M, Panaccione R, Ma C. Review article: therapeutic targets for the pharmacologic management of coeliac disease-the future beyond a gluten-free diet. Aliment Pharmacol Ther 2022; 55:1277-1296. [PMID: 35229332 DOI: 10.1111/apt.16846] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/04/2021] [Revised: 11/03/2021] [Accepted: 02/13/2022] [Indexed: 12/15/2022]
Abstract
BACKGROUND Coeliac disease (CeD) is an immune-mediated small bowel enteropathy resulting from dietary gluten exposure. Presently, the only effective treatment is adoption of a gluten-free diet (GFD), although strict adherence is challenging to maintain, and inadvertent gluten exposures are inevitable for most patients. Hence, there is substantial interest in drug development in CeD and multiple novel therapies are under investigation. AIMS To review existing and upcoming clinical trial programmes for pharmacologic agents for CeD. METHODS A narrative review was performed, informed by a search of MEDLINE, Embase, the Cochrane CENTRAL Library and clinicaltrials.gov. RESULTS We summarise the pathophysiology of CeD and the specific steps that are potentially amenable to pharmacologic treatment. We evaluate the evidence supporting existing and future drug targets, including trials of peptidases, gluten sequestrants, tight junction regulators, anti-transglutaminase 2 therapies, immune tolerizing agents, advanced biologics and small molecules, and microbiome-targeted strategies. We highlight unique considerations for conducting CeD trials, including identifying appropriate study populations, assessing results in the context of a gluten challenge, and interpreting CeD-specific clinical and histologic outcomes. Understanding these factors is crucial for accurately appraising the evidence. Finally, we outline what the future of CeD therapy may hold with the introduction of pharmacotherapies. CONCLUSIONS There is a need for pharmacologic options for CeD, either used adjunctively with a GFD for accidental or intentional gluten exposures or for refractory disease. Multiple promising agents are in development, and these trials are likely to lead to approvals for the first generation of pharmacologic agents for CeD within the next 5 years.
Collapse
Affiliation(s)
| | - Christopher N Andrews
- Division of Gastroenterology & Hepatology, University of Calgary, Calgary, Alberta, Canada
| | - Maitreyi Raman
- Division of Gastroenterology & Hepatology, University of Calgary, Calgary, Alberta, Canada.,Alberta's Collaboration of Excellence for Nutrition in Digestive Diseases, Calgary, Alberta, Canada
| | - Remo Panaccione
- Division of Gastroenterology & Hepatology, University of Calgary, Calgary, Alberta, Canada
| | - Christopher Ma
- Division of Gastroenterology & Hepatology, University of Calgary, Calgary, Alberta, Canada.,Department of Community Health Sciences, University of Calgary, Calgary, Alberta, Canada
| |
Collapse
|
4
|
Garlatti V, Lovisa S, Danese S, Vetrano S. The Multiple Faces of Integrin-ECM Interactions in Inflammatory Bowel Disease. Int J Mol Sci 2021; 22:10439. [PMID: 34638778 PMCID: PMC8508809 DOI: 10.3390/ijms221910439] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2021] [Revised: 09/20/2021] [Accepted: 09/21/2021] [Indexed: 12/03/2022] Open
Abstract
Inflammatory Bowel Disease (IBD) comprises a series of chronic and relapsing intestinal diseases, with Crohn's disease and ulcerative colitis being the most common. The abundant and uncontrolled deposition of extracellular matrix, namely fibrosis, is one of the major hallmarks of IBD and is responsible for the progressive narrowing and closure of the intestine, defined as stenosis. Although fibrosis is usually considered the product of chronic inflammation, the substantial failure of anti-inflammatory therapies to target and reduce fibrosis in IBD suggests that fibrosis might be sustained in an inflammation-independent manner. Pharmacological therapies targeting integrins have recently shown great promise in the treatment of IBD. The efficacy of these therapies mainly relies on their capacity to target the integrin-mediated recruitment and functionality of the immune cells at the damage site. However, by nature, integrins also act as mechanosensitive molecules involved in the intracellular transduction of signals and modifications originating from the extracellular matrix. Therefore, understanding integrin signaling in the context of IBD may offer important insights into mechanisms of matrix remodeling, which are uncoupled from inflammation and could underlie the onset and persistency of intestinal fibrosis. In this review, we present the currently available knowledge on the role of integrins in the etiopathogenesis of IBD, highlighting their role in the context of immune-dependent and independent mechanisms.
Collapse
Affiliation(s)
- Valentina Garlatti
- IRCCS Humanitas Research Hospital, Rozzano, 20089 Milan, Italy; (V.G.); (S.L.); (S.D.)
- Department of Pharmaceutical Sciences, University of Piemonte Orientale ‘A. Avogadro’, 28100 Novara, Italy
| | - Sara Lovisa
- IRCCS Humanitas Research Hospital, Rozzano, 20089 Milan, Italy; (V.G.); (S.L.); (S.D.)
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, 20072 Milan, Italy
| | - Silvio Danese
- IRCCS Humanitas Research Hospital, Rozzano, 20089 Milan, Italy; (V.G.); (S.L.); (S.D.)
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, 20072 Milan, Italy
| | - Stefania Vetrano
- IRCCS Humanitas Research Hospital, Rozzano, 20089 Milan, Italy; (V.G.); (S.L.); (S.D.)
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, 20072 Milan, Italy
| |
Collapse
|
5
|
Rustgi S, Shewry P, Brouns F. Health Hazards Associated with Wheat and Gluten Consumption in Susceptible Individuals and Status of Research on Dietary Therapies. WHEAT QUALITY FOR IMPROVING PROCESSING AND HUMAN HEALTH 2020:471-515. [DOI: 10.1007/978-3-030-34163-3_20] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
|
6
|
Alriyami M, Marchand L, Li Q, Du X, Olivier M, Polychronakos C. Clonal copy-number mosaicism in autoreactive T lymphocytes in diabetic NOD mice. Genome Res 2019; 29:1951-1961. [PMID: 31694869 PMCID: PMC6886509 DOI: 10.1101/gr.247882.118] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2019] [Accepted: 11/02/2019] [Indexed: 01/10/2023]
Abstract
Concordance for type 1 diabetes (T1D) is far from 100% in monozygotic twins and in inbred nonobese diabetic (NOD) mice, despite genetic identity and shared environment during incidence peak years. This points to stochastic determinants, such as postzygotic mutations (PZMs) in the expanding antigen-specific autoreactive T cell lineages, by analogy to their role in the expanding tumor lineage in cancer. Using comparative genomic hybridization of DNA from pancreatic lymph-node memory CD4+ T cells of 25 diabetic NOD mice, we found lymphocyte-exclusive mosaic somatic copy-number aberrations (CNAs) with highly nonrandom independent involvement of the same gene(s) across different mice, some with an autoimmunity association (e.g., Ilf3 and Dgka). We confirmed genes of interest using the gold standard approach for CNA quantification, multiplex ligation-dependent probe amplification (MLPA), as an independent method. As controls, we examined lymphocytes expanded during normal host defense (17 NOD and BALB/c mice infected with Leishmania major parasite). Here, CNAs found were fewer and significantly smaller compared to those in autoreactive cells (P = 0.0019). We determined a low T cell clonality for our samples suggesting a prethymic formation of these CNAs. In this study, we describe a novel, unexplored phenomenon of a potential causal contribution of PZMs in autoreactive T cells in T1D pathogenesis. We expect that exploration of point mutations and studies in human T cells will enable the further delineation of driver genes to target for functional studies. Our findings challenge the classical notions of autoimmunity and open conceptual avenues toward individualized prevention and therapeutics.
Collapse
Affiliation(s)
- Maha Alriyami
- The Endocrine Genetics Laboratory, Child Health and Human Development Program and Department of Pediatrics, McGill University Health Centre Research Institute, Montreal, Quebec H3H 1P3, Canada.,Department of Biochemistry, College of Medicine and Health Sciences, Sultan Qaboos University, 123, Muscat, Oman
| | - Luc Marchand
- The Endocrine Genetics Laboratory, Child Health and Human Development Program and Department of Pediatrics, McGill University Health Centre Research Institute, Montreal, Quebec H3H 1P3, Canada
| | - Quan Li
- The Endocrine Genetics Laboratory, Child Health and Human Development Program and Department of Pediatrics, McGill University Health Centre Research Institute, Montreal, Quebec H3H 1P3, Canada.,Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, ON M5G 2C1, Canada
| | - Xiaoyu Du
- The Endocrine Genetics Laboratory, Child Health and Human Development Program and Department of Pediatrics, McGill University Health Centre Research Institute, Montreal, Quebec H3H 1P3, Canada
| | - Martin Olivier
- Departments of Medicine, Microbiology, and Immunology, McGill University Health Centre Research Institute, Montreal, Quebec H3H 1P3, Canada
| | - Constantin Polychronakos
- The Endocrine Genetics Laboratory, Child Health and Human Development Program and Department of Pediatrics, McGill University Health Centre Research Institute, Montreal, Quebec H3H 1P3, Canada
| |
Collapse
|
7
|
Kustrimovic N, Marino F, Cosentino M. Peripheral Immunity, Immunoaging and Neuroinflammation in Parkinson's Disease. Curr Med Chem 2019; 26:3719-3753. [PMID: 30306855 DOI: 10.2174/0929867325666181009161048] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2017] [Revised: 06/26/2018] [Accepted: 09/12/2018] [Indexed: 12/20/2022]
Abstract
Parkinson's disease (PD) is the second most common neurodegenerative disorder among elderly population, characterized by the progressive degeneration of dopaminergic neurons in the midbrain. To date, exact cause remains unknown and the mechanism of neurons death uncertain. It is typically considered as a disease of central nervous system (CNS). Nevertheless, numerous evidence has been accumulated in several past years testifying undoubtedly about the principal role of neuroinflammation in progression of PD. Neuroinflammation is mainly associated with presence of activated microglia in brain and elevated levels of cytokine levels in CNS. Nevertheless, active participation of immune system as well has been noted, such as, elevated levels of cytokine levels in blood, the presence of auto antibodies, and the infiltration of T cell in CNS. Moreover, infiltration and reactivation of those T cells could exacerbate neuroinflammation to greater neurotoxic levels. Hence, peripheral inflammation is able to prime microglia into pro-inflammatory phenotype, which can trigger stronger response in CNS further perpetuating the on-going neurodegenerative process. In the present review, the interplay between neuroinflammation and the peripheral immune response in the pathobiology of PD will be discussed. First of all, an overview of regulation of microglial activation and neuroinflammation is summarized and discussed. Afterwards, we try to collectively analyze changes that occurs in peripheral immune system of PD patients, suggesting that these peripheral immune challenges can exacerbate the process of neuroinflammation and hence the symptoms of the disease. In the end, we summarize some of proposed immunotherapies for treatment of PD.
Collapse
Affiliation(s)
- Natasa Kustrimovic
- Center of Research in Medical Pharmacology, University of Insubria, Varese, Italy
| | - Franca Marino
- Center of Research in Medical Pharmacology, University of Insubria, Varese, Italy
| | - Marco Cosentino
- Center of Research in Medical Pharmacology, University of Insubria, Varese, Italy
| |
Collapse
|
8
|
Abstract
Celiac disease (CD) is an autoimmune enteropathy triggered by gluten. Gluten-free diets can be challenging because of their restrictive nature, inadvertent cross-contaminations, and the high cost of gluten-free food. Novel nondietary therapies are at the preclinical stage, clinical trial phase, or have already been developed for other indications and are now being applied to CD. These therapies include enzymatic gluten degradation, binding and sequestration of gluten, restoration of epithelial tight junction barrier function, inhibition of tissue transglutaminase-mediated potentiation of gliadin oligopeptide immunogenicity or of human leukocyte antigen-mediated gliadin presentation, induction of tolerance to gluten, and antiinflammatory interventions.
Collapse
Affiliation(s)
- Gloria Serena
- Division of Pediatric Gastroenterology and Nutrition, Mucosal Immunology and Biology Research Center, MassGeneral Hospital for Children, 175 Cambridge Street, CPZS - 574, Boston, MA 02114, USA; Celiac Research Program, Harvard Medical School, 25 Shattuck Street, Boston, MA 02115, USA
| | - Ciaran P Kelly
- Celiac Research Program, Harvard Medical School, 25 Shattuck Street, Boston, MA 02115, USA; Department of Medicine, Division of Gastroenterology, Beth Israel Deaconess Medical Center, 330 Brookline Avenue, Boston, MA 02215, USA
| | - Alessio Fasano
- Division of Pediatric Gastroenterology and Nutrition, Mucosal Immunology and Biology Research Center, MassGeneral Hospital for Children, 175 Cambridge Street, CPZS - 574, Boston, MA 02114, USA; Celiac Research Program, Harvard Medical School, 25 Shattuck Street, Boston, MA 02115, USA.
| |
Collapse
|
9
|
Di Sabatino A, Giuffrida P, Fornasa G, Salvatore C, Vanoli A, Naviglio S, De Leo L, Pasini A, De Amici M, Alvisi C, Not T, Rescigno M, Corazza GR. Innate and adaptive immunity in self-reported nonceliac gluten sensitivity versus celiac disease. Dig Liver Dis 2016; 48:745-52. [PMID: 27130911 DOI: 10.1016/j.dld.2016.03.024] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/13/2015] [Revised: 03/09/2016] [Accepted: 03/30/2016] [Indexed: 12/11/2022]
Abstract
BACKGROUND Immune mechanisms have been implicated in nonceliac gluten sensitivity (NCGS), a condition characterized by intestinal and/or extraintestinal symptoms caused by the ingestion of gluten in non-celiac/non-wheat allergic individuals. AIMS We investigated innate and adaptive immunity in self-reported NCGS versus celiac disease (CD). METHODS In the supernatants of ex vivo-cultured duodenal biopsies from 14 self-reported NCGS patients, 9 untreated and 10 treated CD patients, and 12 controls we detected innate cytokines - interleukin (IL)-15, tumor necrosis factor-α, IL-1β, IL-6, IL-12p70, IL-23, IL-27, IL-32α, thymic stromal lymphopoietin (TSLP), IFN-α-, adaptive cytokines - interferon (IFN)-γ, IL-17A, IL-4, IL-5, IL-10, IL-13-, chemokines - IL-8, CCL1, CCL2, CCL3, CCL4, CCL5, CXCL1, CXCL10-, granulocyte colony stimulating factor (G-CSF) and granulocyte-macrophage colony stimulating factor (GM-CSF). RESULTS Mucosal innate and adaptive cytokines, chemokines and growth factors did not differ between self-reported NCGS, treated CD and controls. On the contrary, IL-6, IL-15, IL-27, IFN-α, IFN-γ, IL-17A, IL-23, G-CSF, GM-CSF, IL-8, CCL1 and CCL4 were significantly higher in untreated CD than in self-reported NCGS, treated CD and controls, while TSLP was significantly lower in untreated CD than in self-reported NCGS, treated CD and controls. CONCLUSION In our hands, patients with self-reported NCGS showed no abnormalities of the mucosal immune response.
Collapse
Affiliation(s)
- Antonio Di Sabatino
- Department of Internal Medicine, San Matteo Hospital, University of Pavia, Pavia, Italy.
| | - Paolo Giuffrida
- Department of Internal Medicine, San Matteo Hospital, University of Pavia, Pavia, Italy
| | - Giulia Fornasa
- Department of Experimental Oncology, European Institute of Oncology, Milan, Italy
| | - Chiara Salvatore
- Department of Internal Medicine, San Matteo Hospital, University of Pavia, Pavia, Italy
| | - Alessandro Vanoli
- Department of Molecular Medicine, San Matteo Hospital, University of Pavia, Pavia, Italy
| | - Samuele Naviglio
- Department of Medicine, Surgery, and Health Sciences, University of Trieste, Trieste, Italy
| | - Luigina De Leo
- Institute for Maternal and Child Health IRCCS "Burlo Garofolo", Trieste, Italy
| | - Alessandra Pasini
- Department of Internal Medicine, San Matteo Hospital, University of Pavia, Pavia, Italy
| | - Mara De Amici
- Department of Pediatrics, San Matteo Hospital, University of Pavia, Pavia, Italy
| | - Costanza Alvisi
- Department of Internal Medicine, San Matteo Hospital, University of Pavia, Pavia, Italy
| | - Tarcisio Not
- Department of Medicine, Surgery, and Health Sciences, University of Trieste, Trieste, Italy; Institute for Maternal and Child Health IRCCS "Burlo Garofolo", Trieste, Italy
| | - Maria Rescigno
- Department of Experimental Oncology, European Institute of Oncology, Milan, Italy
| | - Gino Roberto Corazza
- Department of Internal Medicine, San Matteo Hospital, University of Pavia, Pavia, Italy
| |
Collapse
|
10
|
Plugis NM, Khosla C. Therapeutic approaches for celiac disease. Best Pract Res Clin Gastroenterol 2015; 29:503-21. [PMID: 26060114 PMCID: PMC4465084 DOI: 10.1016/j.bpg.2015.04.005] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/24/2015] [Revised: 04/23/2015] [Accepted: 04/26/2015] [Indexed: 02/06/2023]
Abstract
Celiac disease is a common, lifelong autoimmune disorder for which dietary control is the only accepted form of therapy. A strict gluten-free diet is burdensome to patients and can be limited in efficacy, indicating there is an unmet need for novel therapeutic approaches to supplement or supplant dietary therapy. Many molecular events required for disease pathogenesis have been recently characterized and inspire most current and emerging drug-discovery efforts. Genome-wide association studies (GWAS) confirm the importance of human leukocyte antigen genes in our pathogenic model and identify a number of new risk loci in this complex disease. Here, we review the status of both emerging and potential therapeutic strategies in the context of disease pathophysiology. We conclude with a discussion of how genes identified during GWAS and follow-up studies that enhance susceptibility may offer insight into developing novel therapies.
Collapse
|
11
|
Makharia GK. Current and emerging therapy for celiac disease. Front Med (Lausanne) 2014; 1:6. [PMID: 25705619 PMCID: PMC4335393 DOI: 10.3389/fmed.2014.00006] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2013] [Accepted: 03/13/2014] [Indexed: 12/12/2022] Open
Abstract
At present, strict and lifelong gluten-free diet is the only effective treatment for celiac disease. Even small amounts of gluten (50 mg/day) can be immunogenic; therefore all food and food items and drugs that contain gluten and its derivatives must be eliminated completely from the diet. While prescribing gluten-free diet is easy; the key to the success is the dietary counseling by a nutrition specialist and maintenance of adherence to GFD by the patient. In recent times, a number of targets to halt the process of immunological injury have been explored to find out alternative treatment for celiac disease. These targets include exploration of ancient wheat if they are less immunogenic, intra-luminal digestion of gluten using prolylendopeptidases, pretreatment of whole gluten with bacterial-derived peptidase before ingestion; prevention of passage of immunogenic peptides through the tight junctions such as zonulin antagonists, Blocking of HLA-DQ2 to prevent binding of immunogenic peptides, inhibition of transglutaminase 2, immune-modulation, and induction of tolerance to gluten using gluten tolerizing vaccines, use of gluten-sequestering polymers, use of anti-inflammatory drugs (glucocorticoids, budesonides) and anti-cytokines such as anti TNF-α, and anti-interleukin-15. While many of these targets are still in the pre-clinical phase, some of them including zonulin antagonist and endopeptidases have already reached phase II and phase III clinical trials. Furthermore, while these targets appear very exciting; they at best are likely to be used as adjunctive therapy rather than a complete replacement for gluten-free diet.
Collapse
Affiliation(s)
- Govind K. Makharia
- Department of Gastroenterology and Human Nutrition, All India Institute of Medical Sciences, New Delhi, India
| |
Collapse
|
12
|
Abstract
Coeliac disease is a common and fairly well-characterized systemic disorder that mainly affects the small intestine, but also has extraintestinal manifestations. The environmental trigger (gluten derived from wheat, rye and barley), the genetic predisposition conferred by the HLA-DQ2 and HLA-DQ8 haplotypes and many steps in the disease pathogenesis are known. This knowledge has enabled researchers to suggest novel alternative treatments or adjunctive therapies to the gluten-free diet, which is currently the only available and effective treatment for the condition. This Review focuses on emerging and potential treatment strategies that are based on the current concept of the disease pathophysiology. The search for novel future treatment modes, including nonpharmacological and pharmacological approaches, is also outlined. The potential pitfalls associated with the various research avenues are also discussed.
Collapse
|
13
|
Effect of tumor necrosis factor-α blockade on mucosal addressin cell-adhesion molecule-1 in Crohn's disease. Inflamm Bowel Dis 2013; 19:259-64. [PMID: 23328772 DOI: 10.1097/mib.0b013e31828100a4] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
BACKGROUND Mucosal addressin cell-adhesion molecule (MAdCAM)-1, which is overexpressed on gut endothelium in active Crohn's disease (CD), promotes intestinal recruitment of integrin α(4)β(7)(*) T cells that sustain chronic inflammation. As tumor necrosis factor alpha (TNF)-α, a cytokine centrally involved in CD, modulates gut endothelial adhesion molecules, we here explored the in vivo and ex vivo effects of TNF-α blockade on MAdCAM-1 expression in CD. METHODS MAdCAM-1 was determined by immunoblotting in colonic biopsies collected before and 10 weeks after either infliximab or adalimumab treatment in CD patients, and in CD biopsies incubated with either infliximab or adalimumab or control IgG(1). Integrin β(7)(*) circulating T cells were analyzed by flow cytometry. RESULTS MAdCAM-1 significantly decreased after either infliximab or adalimumab treatment in responder but not in nonresponder patients. In parallel, an increase of circulating β(7)(*) T cells was found in responder patients only. A marked downregulation of MAdCAM-1 was observed in CD biopsies cultured with either infliximab or adalimumab in comparison to IgG(1)-treated biopsies. CONCLUSIONS Our findings showing that MAdCAM-1 is downregulated by TNF-α blockade point to a novel mechanism of action of anti-TNF-α antibodies in CD.
Collapse
|
14
|
Saunders JAH, Estes KA, Kosloski LM, Allen HE, Dempsey KM, Torres-Russotto DR, Meza JL, Santamaria PM, Bertoni JM, Murman DL, Ali HH, Standaert DG, Mosley RL, Gendelman HE. CD4+ regulatory and effector/memory T cell subsets profile motor dysfunction in Parkinson's disease. J Neuroimmune Pharmacol 2012; 7:927-38. [PMID: 23054369 PMCID: PMC3515774 DOI: 10.1007/s11481-012-9402-z] [Citation(s) in RCA: 232] [Impact Index Per Article: 17.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2012] [Accepted: 08/27/2012] [Indexed: 02/06/2023]
Abstract
Animal models and clinical studies have linked the innate and adaptive immune system to the pathology of Parkinson's disease (PD). Despite such progress, the specific immune responses that influence disease progression have eluded investigators. Herein, we assessed relationships between T cell phenotype and function with PD progression. Peripheral blood lymphocytes from two separate cohorts, a discovery cohort and a validation cohort, totaling 113 PD patients and 96 age- and environment-matched caregivers were examined by flow cytometric analysis and T cell proliferation assays. Increased effector/memory T cells (Tem), defined as CD45RO+ and FAS+ CD4+ T cells and decreased CD31+ and α4β7+ CD4+ T cells were associated with progressive Unified Parkinson's Disease Rating Scale III scores. However, no associations were seen between immune biomarkers and increased age or disease duration. Impaired abilities of regulatory T cells (Treg) from PD patients to suppress effector T cell function was observed. These data support the concept that chronic immune stimulation, notably Tem activation and Treg dysfunction is linked to PD pathobiology and disease severity, but not disease duration. The association of T cell phenotypes with motor symptoms provides fresh avenues for novel biomarkers and therapeutic designs.
Collapse
Affiliation(s)
- Jessica A. Hutter Saunders
- Department of Pharmacology and Experimental Neuroscience, Center for Neurodegenerative Disorders, University of Nebraska Medical Center, Omaha, NE 68198-5880 USA
| | - Katherine A. Estes
- Department of Pharmacology and Experimental Neuroscience, Center for Neurodegenerative Disorders, University of Nebraska Medical Center, Omaha, NE 68198-5880 USA
| | - Lisa M. Kosloski
- Department of Pharmacology and Experimental Neuroscience, Center for Neurodegenerative Disorders, University of Nebraska Medical Center, Omaha, NE 68198-5880 USA
| | - Heather E. Allen
- Department of Neurology, University of Alabama at Birmingham, Birmingham, AL USA
| | - Kathryn M. Dempsey
- College of Information Science and Technology, University of Nebraska at Omaha, Omaha, NE USA
| | | | - Jane L. Meza
- Department of Biostatistics, University of Nebraska Medical Center, Omaha, NE USA
| | | | - John M. Bertoni
- Department of Neurological Sciences, University of Nebraska Medical Center, Omaha, NE USA
| | - Daniel L. Murman
- Department of Neurological Sciences, University of Nebraska Medical Center, Omaha, NE USA
| | - Hesham H. Ali
- College of Information Science and Technology, University of Nebraska at Omaha, Omaha, NE USA
| | - David G. Standaert
- Department of Neurology, University of Alabama at Birmingham, Birmingham, AL USA
| | - R. Lee Mosley
- Department of Pharmacology and Experimental Neuroscience, Center for Neurodegenerative Disorders, University of Nebraska Medical Center, Omaha, NE 68198-5880 USA
- Department of Pharmacology and Experimental Neuroscience Movement Disorders Program, University of Nebraska Medical Center, Omaha, NE 68198-5880 USA
| | - Howard E. Gendelman
- Department of Pharmacology and Experimental Neuroscience, Center for Neurodegenerative Disorders, University of Nebraska Medical Center, Omaha, NE 68198-5880 USA
| |
Collapse
|
15
|
Abstract
Integrins are a family of α/β heterodimeric adhesion metalloprotein receptors and their functions are highly dependent on and regulated by different divalent cations. Recently advanced studies have revolutionized our perception of integrin metal ion-binding sites and their specific functions. Ligand binding to integrins is bridged by a divalent cation bound at the MIDAS motif on top of either α I domain in I domain-containing integrins or β I domain in α I domain-less integrins. The MIDAS motif in β I domain is flanked by ADMIDAS and SyMBS, the other two crucial metal ion binding sites playing pivotal roles in the regulation of integrin affinity and bidirectional signaling across the plasma membrane. The β-propeller domain of α subunit contains three or four β-hairpin loop-like Ca(2+)-binding motifs that have essential roles in integrin biogenesis. The function of another Ca(2+)-binding motif located at the genu of α subunit remains elusive. Here, we provide an overview of the integrin metal ion-binding sites and discuss their roles in the regulation of integrin functions.
Collapse
Affiliation(s)
- Kun Zhang
- State Key Laboratory of Cell Biology, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China
| | | |
Collapse
|
16
|
Rashtak S, Murray JA. Review article: coeliac disease, new approaches to therapy. Aliment Pharmacol Ther 2012; 35:768-81. [PMID: 22324389 PMCID: PMC3912561 DOI: 10.1111/j.1365-2036.2012.05013.x] [Citation(s) in RCA: 63] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/10/2011] [Revised: 04/03/2011] [Accepted: 01/18/2012] [Indexed: 02/07/2023]
Abstract
BACKGROUND Coeliac disease is managed by life-long gluten withdrawal from the diet. However, strict adherence to a gluten-free diet is difficult and is not always effective. Novel therapeutic approaches are needed to supplement or even replace the dietary treatment. AIM To review recent advances in new therapeutic options for coeliac disease. METHODS A literature search was performed on MEDLINE, EMBASE, Web of Science, Scopus, DDW.org and ClinicalTrials.gov for English articles and abstracts. The search terms used included, but not limited to, 'Celiac disease', 'new', 'novel', 'Advances', 'alternatives' and 'Drug therapy'. The cited articles were selected based on the relevancy to the review objective. RESULTS Several new therapeutic approaches for coeliac disease are currently under development by targeting its underlying pathogenesis. Alternative therapies range from reproduction of harmless wheat strains to immunomodulatory approaches. Some of these therapies such as enzymatic cleavage of gluten and permeability inhibitors have shown promise in clinical studies. CONCLUSIONS Gluten-free diet is still the only practical treatment for patients with coeliac disease. Novel strategies provide promise of alternative adjunctive approaches to diet restriction alone for patients with this disorder.
Collapse
Affiliation(s)
- S Rashtak
- Celiac Disease Research Program, Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, MN, USA
| | | |
Collapse
|
17
|
Laroni A, Giacosa A, Uccelli A, Mancardi GL. Can we kill an extra bird with the same stone? Inflamm Bowel Dis 2011; 17:E124-5. [PMID: 21710537 DOI: 10.1002/ibd.21802] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/16/2011] [Accepted: 05/23/2011] [Indexed: 01/05/2023]
|
18
|
Stefanich EG, Danilenko DM, Wang H, O'Byrne S, Erickson R, Gelzleichter T, Hiraragi H, Chiu H, Ivelja S, Jeet S, Gadkari S, Hwang O, Fuh F, Looney C, Howell K, Albert V, Balazs M, Refino C, Fong S, Iyer S, Williams M. A humanized monoclonal antibody targeting the β7 integrin selectively blocks intestinal homing of T lymphocytes. Br J Pharmacol 2011; 162:1855-70. [PMID: 21232034 DOI: 10.1111/j.1476-5381.2011.01205.x] [Citation(s) in RCA: 70] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
BACKGROUND AND PURPOSE rhuMAb Beta7 is a humanized anti-human β7 monoclonal antibody currently in phase I in inflammatory bowel disease. rhuMAb Beta7 binds the β7 subunit of the integrins α4β7 and αEβ7, blocking interaction with their ligands. These integrins play key roles in immune cell homing to and retention in mucosal sites, and are associated with chronic inflammatory diseases of the gastrointestinal tract. The goal of this study was to evaluate the mucosal specificity of rhuMAb Beta7. EXPERIMENTAL APPROACH We assessed the effect of murine anti-Beta7 on lymphocyte homing in mouse models of autoimmune disease. We also compared the effect of rhuMAb Beta7 on circulating mucosal-homing versus peripheral-homing T cells in naïve non-human primates. KEY RESULTS In cynomolgus monkeys, occupancy of β7 integrin receptors by rhuMAb Beta7 correlated with an increase in circulating β7(+) mucosal-homing lymphocytes, with no apparent effect on levels of circulating β7(-) peripheral-homing lymphocytes. rhuMAb Beta7 also inhibited lymphocyte homing to the inflamed colons of severe combined immunodeficient mice in CD45RB(high) CD4(+) T-cell transfer models. Consistent with a lack of effect on peripheral homing, in a mouse model of experimental autoimmune encephalomyelitis, anti-β7 treatment resulted in no amelioration of CNS inflammation. CONCLUSIONS AND IMPLICATIONS The results presented here suggest that rhuMAb Beta7 selectively blocks lymphocyte homing to the gastrointestinal tract without affecting lymphocyte trafficking to non-mucosal tissues. rhuMAb Beta7 provides a targeted therapeutic approach with the potential for a more attractive benefit:risk ratio than currently available inflammatory bowel disease therapies.
Collapse
Affiliation(s)
- E G Stefanich
- Department of Pharmacodynamic Biomarkers, Genentech, Inc., South San Francisco, CA 94080, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
19
|
Abstract
PURPOSE OF REVIEW To summarize recent advances in celiac disease published between August 2008 and July 2009. RECENT FINDINGS Celiac disease affects nearly 1% of most populations but remains largely unrecognized. In the last year, work has shown that the prevalence of celiac disease has increased dramatically, not simply due to increased detection. Also, undiagnosed celiac disease may be associated with increased mortality. Significant progress has been made in understanding how gliadin peptides can cross the intestinal border and access the immune system. New genetic loci and candidate genes that may contribute to the risk of celiac disease and its overlap with type 1 diabetes mellitus have been identified. Novel deamidated gliadin peptides antibodies have better diagnostic accuracy over native gliadin-based tests. The inclusion of duodenal bulb biopsy specimens may increase the rate of celiac disease detection. The spectrum of celiac disease likely includes a minority of patients with mild enteropathy. A practical seven-item instrument may facilitate standardized evaluation of gluten-free diet adherence. Finally, refractory celiac disease, although rare, is associated with a poor prognosis. SUMMARY Celiac disease is a global health problem that requires a multidisciplinary and increasingly cooperative multinational research effort.
Collapse
|
20
|
Myrsky E, Caja S, Simon-Vecsei Z, Korponay-Szabo IR, Nadalutti C, Collighan R, Mongeot A, Griffin M, Mäki M, Kaukinen K, Lindfors K. Celiac disease IgA modulates vascular permeability in vitro through the activity of transglutaminase 2 and RhoA. Cell Mol Life Sci 2009; 66:3375-85. [PMID: 19680746 PMCID: PMC11115502 DOI: 10.1007/s00018-009-0116-1] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2009] [Revised: 07/02/2009] [Accepted: 07/20/2009] [Indexed: 02/07/2023]
Abstract
Celiac disease is characterized by the presence of specific autoantibodies targeted against transglutaminase 2 (TG2) in untreated patients' serum and at their production site in the small-bowel mucosa below the basement membrane and around the blood vessels. As these autoantibodies have biological activity in vitro, such as inhibition of angiogenesis, we studied if they might also modulate the endothelial barrier function. Our results show that celiac disease patient autoantibodies increase endothelial permeability for macromolecules, and enhance the binding of lymphocytes to the endothelium and their transendothelial migration when compared to control antibodies in an endothelial cell-based in vitro model. We also demonstrate that these effects are mediated by increased activities of TG2 and RhoA. Since the small bowel mucosal endothelium serves as a "gatekeeper" in inflammatory processes, the disease-specific autoantibodies targeted against TG2 could thus contribute to the pathogenic cascade of celiac disease by increasing blood vessel permeability.
Collapse
Affiliation(s)
- Essi Myrsky
- Pediatric Research Center, Medical School, University of Tampere, 33014 Tampere, Finland
| | - Sergio Caja
- Pediatric Research Center, Medical School, University of Tampere, 33014 Tampere, Finland
| | - Zsofi Simon-Vecsei
- Department of Biochemistry and Molecular Biology, Medical and Health Science Center, University of Debrecen, Debrecen, Hungary
| | - Ilma R. Korponay-Szabo
- Heim Pál Children’s Hospital, Budapest, and Department of Pediatrics, Medical and Health Science Center, University of Debrecen, Debrecen, Hungary
| | - Cristina Nadalutti
- Pediatric Research Center, Medical School, University of Tampere, 33014 Tampere, Finland
| | - Russell Collighan
- School of Life and Health Sciences, Aston University, Birmingham, UK
| | | | - Martin Griffin
- School of Life and Health Sciences, Aston University, Birmingham, UK
| | - Markku Mäki
- Pediatric Research Center, Medical School, University of Tampere, 33014 Tampere, Finland
- Department of Pediatrics, Tampere University Hospital, Medical School, University of Tampere, Tampere, Finland
| | - Katri Kaukinen
- Pediatric Research Center, Medical School, University of Tampere, 33014 Tampere, Finland
- Department of Gastroenterology and Alimentary Tract Surgery, Tampere University Hospital, Medical School, University of Tampere, Tampere, Finland
| | - Katri Lindfors
- Pediatric Research Center, Medical School, University of Tampere, 33014 Tampere, Finland
| |
Collapse
|
21
|
Rescigno M, Di Sabatino A. Dendritic cells in intestinal homeostasis and disease. J Clin Invest 2009; 119:2441-50. [PMID: 19729841 DOI: 10.1172/jci39134] [Citation(s) in RCA: 243] [Impact Index Per Article: 15.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
DCs are specialized APCs that orchestrate innate and adaptive immune responses. The intestinal mucosa contains numerous DCs, which induce either protective immunity to infectious agents or tolerance to innocuous antigens, including food and commensal bacteria. Several subsets of mucosal DCs have been described that display unique functions, dictated in part by the local microenvironment. In this review, we summarize the distinct subtypes of DCs and their distribution in the gut; examine how DC dysfunction contributes to intestinal disease development, including inflammatory bowel disease and celiac disease; and discuss manipulation of DCs for therapy.
Collapse
Affiliation(s)
- Maria Rescigno
- Department of Experimental Oncology, European Institute of Oncology, Milan, Italy.
| | | |
Collapse
|