1
|
Marchi E, Craig JW, Kalac M. Current and upcoming treatment approaches to uncommon subtypes of PTCL (EATL, MEITL, SPTCL, and HSTCL). Blood 2024; 144:1898-1909. [PMID: 38657272 DOI: 10.1182/blood.2023021788] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2024] [Revised: 04/19/2024] [Accepted: 04/19/2024] [Indexed: 04/26/2024] Open
Abstract
ABSTRACT Rare subtypes of peripheral T-cell lymphoma (PTCL) including enteropathy-associated T-cell lymphoma (EATL), monomorphic epitheliotropic intestinal T-cell lymphoma (MEITL), subcutaneous panniculitis-like T-cell lymphoma (SPTCL), and hepatosplenic T-cell lymphoma (HSTCL) are underrepresented in most registries and clinical studies. Most of the literature is obtained from small case series, single-institution retrospective studies, and subgroup analyses of the largest studies with few recent and ongoing exceptions. Although the pathogenesis and biology of these entities have yet to be fully elucidated, global efforts by the scientific community have started to shed some light on the most frequently deregulated pathways. In this review, we highlight the most pertinent clinical and pathologic features of rare subtypes of PTCL including EATL/MEITL, SPTCL, and HSTCL. We also summarize the results of recent developments identifying potential targets for novel therapeutic strategies based on molecular studies. Finally, we highlight the underrepresentation of these rare subtypes in most clinical trials, making evidence-based therapeutic decisions extremely challenging.
Collapse
Affiliation(s)
- Enrica Marchi
- Division of Hematology and Oncology, Department of Medicine, University of Virginia Cancer Center, Charlottesville, VA
| | - Jeffrey W Craig
- Department of Pathology, University of Virginia Cancer Center, Charlottesville, VA
| | - Matko Kalac
- Department of Hematology and Oncology, University of California, Irvine, CA
| |
Collapse
|
2
|
Noori E, Hashemi N, Rezaee D, Maleki R, Shams F, Kazemi B, Bandepour M, Rahimi F. Potential therapeutic options for celiac Disease: An update on Current evidence from Gluten-Free diet to cell therapy. Int Immunopharmacol 2024; 133:112020. [PMID: 38608449 DOI: 10.1016/j.intimp.2024.112020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2024] [Revised: 04/01/2024] [Accepted: 04/03/2024] [Indexed: 04/14/2024]
Abstract
Celiac disease (CD) is a chronic autoimmune enteropathy and multifactorial disease caused by inappropriate immune responses to gluten in the small intestine. Weight loss, anemia, osteoporosis, arthritis, and hepatitis are among the extraintestinal manifestations of active CD. Currently, a strict lifelong gluten-free diet (GFD) is the only safe, effective, and available treatment. Despite the social burden, high expenses, and challenges of following a GFD, 2 to 5 percent of patients do not demonstrate clinical or pathophysiological improvement. Therefore, we need novel and alternative therapeutic approaches for patients. Innovative approaches encompass a broad spectrum of strategies, including enzymatic degradation of gluten, inhibition of intestinal permeability, modulation of the immune response, inhibition of the transglutaminase 2 (TG2) enzyme, blocking antigen presentation by HLA-DQ2/8, and induction of tolerance. Hence, this review is focused on comprehensive therapeutic strategies ranging from dietary approaches to novel methods such as antigen-based immunotherapy, cell and gene therapy, and the usage of nanoparticles for CD treatment.
Collapse
Affiliation(s)
- Effat Noori
- Department of Biotechnology, Faculty of Medicine, Shahed University, Tehran, Iran.
| | - Nader Hashemi
- Department of Medical Biotechnology, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Delsuz Rezaee
- School of Allied Medical Sciences, Ilam University of Medical Sciences, Ilam, Iran; Department of Medical Biotechnology, School of Medicine, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Reza Maleki
- Adelaide Medical School, The University of Adelaide, Adelaide, SA 5005, Australia
| | - Forough Shams
- Department of Medical Biotechnology, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| | - Bahram Kazemi
- Department of Medical Biotechnology, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran; Cellular and Molecular Biology Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mojgan Bandepour
- Department of Medical Biotechnology, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran; Cellular and Molecular Biology Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Fardin Rahimi
- Department of Biotechnology, Faculty of Medicine, Shahed University, Tehran, Iran
| |
Collapse
|
3
|
Proteins from Modern and Ancient Wheat Cultivars: Impact on Immune Cells of Healthy Individuals and Patients with NCGS. Nutrients 2022; 14:nu14204257. [PMID: 36296938 PMCID: PMC9611902 DOI: 10.3390/nu14204257] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2022] [Revised: 09/30/2022] [Accepted: 10/06/2022] [Indexed: 11/19/2022] Open
Abstract
In non-celiac gluten sensitivity (NCGS), the elimination of wheat results in a clear symptom improvement, but gluten has still not been proven as (the sole) trigger. Due to the increase in the prevalence of gluten-related diseases, the breeding of high-performance wheat cultivars is discussed as a trigger. To analyze the immune stimulation and signal pathways, the immune cells of healthy subjects and patients with NCGS were stimulated with gliadins from wheat, and the expression and secretion of interleukin 1ß (IL1ß) and interleukin 6 (IL6) were studied. To determine the impact of wheat breeding, the monocyte cell line THP1 and human immune cells were stimulated with gliadin, glutenin, and albumin/globulin fractions of ancient and modern cereals, and expression of inflammatory molecules was checked. Immune cells of patients with NCGS showed an increased expression of IL1ß and IL6 after stimulation with gliadins compared to immune cells of healthy controls. Gliadins caused a strong activation of P-STAT3 in immune cells of healthy controls, and inhibitors of JAK and NFκB pathways considerably reduced this response. In addition to gliadins, we further showed that glutenins and albumin/globulins from all wheat cultivars from the last century, and especially from einkorn and spelt, also markedly induced the expression of inflammatory genes in THP1 and human immune cells. There was no correlation between enhanced immune stimulation and ancient or modern cultivars. This does not support the hypothesis that modern wheat breeding is responsible for the increase in gluten-related diseases. An altered immune situation is suggested in patients with NCGS.
Collapse
|
4
|
Bragde HG, Jansson U, Fredrikson M, Grodzinsky E, Söderman J. Characterisation of gene and pathway expression in stabilised blood from children with coeliac disease. BMJ Open Gastroenterol 2020; 7:bmjgast-2020-000536. [PMID: 33323471 PMCID: PMC7745333 DOI: 10.1136/bmjgast-2020-000536] [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: 09/04/2020] [Revised: 11/05/2020] [Accepted: 11/21/2020] [Indexed: 12/20/2022] Open
Abstract
Introduction A coeliac disease (CD) diagnosis is likely in children with levels of tissue transglutaminase autoantibodies (anti-TG2) >10 times the upper reference value, whereas children with lower anti-TG2 levels need an intestinal biopsy to confirm or rule out CD. A blood sample is easier to obtain than an intestinal biopsy sample, and stabilised blood is suitable for routine diagnostics because transcript levels are preserved at sampling. Therefore, we investigated gene expression in stabilised whole blood to explore the possibility of gene expression-based diagnostics for the diagnosis and follow-up of CD. Design We performed RNA sequencing of stabilised whole blood from active CD cases (n=10), non-CD cases (n=10), and treated CD cases on a gluten-free diet (n=10) to identify diagnostic CD biomarkers and pathways involved in CD pathogenesis. Results No single gene was differentially expressed between the sample groups. However, by using gene set enrichment analysis (GSEA), significantly differentially expressed pathways were identified in active CD, and these pathways involved the inflammatory response, negative regulation of viral replication, translation, as well as cell proliferation, differentiation, migration, and survival. The results indicate that there are differences in pathway regulation in CD, which could be used for diagnostic purposes. Comparison between GSEA results based on stabilised blood with GSEA results based on small intestinal biopsies revealed that type I interferon response, defence response to virus, and negative regulation of viral replication were identified as pathways common to both tissues. Conclusions Stabilised whole blood is not a suitable sample for clinical diagnostics of CD based on single genes. However, diagnostics based on a pathway-focused gene expression panel may be feasible, but requires further investigation.
Collapse
Affiliation(s)
- Hanna Gustafsson Bragde
- Laboratory Medicine, Region Jönköping County, Jönköping, Sweden .,Department of Biomedical and Clinical Sciences, Linköping University, Linköping, Sweden
| | - Ulf Jansson
- Department of Paediatrics, Region Jönköping County, Jönköping, Sweden
| | - Mats Fredrikson
- Department of Biomedical and Clinical Sciences, Linköping University, Linköping, Sweden
| | - Ewa Grodzinsky
- Department of Biomedical and Clinical Sciences, Linköping University, Linköping, Sweden
| | - Jan Söderman
- Laboratory Medicine, Region Jönköping County, Jönköping, Sweden.,Department of Biomedical and Clinical Sciences, Linköping University, Linköping, Sweden
| |
Collapse
|
5
|
A Driver Never Works Alone-Interplay Networks of Mutant p53, MYC, RAS, and Other Universal Oncogenic Drivers in Human Cancer. Cancers (Basel) 2020; 12:cancers12061532. [PMID: 32545208 PMCID: PMC7353041 DOI: 10.3390/cancers12061532] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2020] [Revised: 06/03/2020] [Accepted: 06/05/2020] [Indexed: 12/12/2022] Open
Abstract
The knowledge accumulating on the occurrence and mechanisms of the activation of oncogenes in human neoplasia necessitates an increasingly detailed understanding of their systemic interactions. None of the known oncogenic drivers work in isolation from the other oncogenic pathways. The cooperation between these pathways is an indispensable element of a multistep carcinogenesis, which apart from inactivation of tumor suppressors, always includes the activation of two or more proto-oncogenes. In this review we focus on representative examples of the interaction of major oncogenic drivers with one another. The drivers are selected according to the following criteria: (1) the highest frequency of known activation in human neoplasia (by mutations or otherwise), (2) activation in a wide range of neoplasia types (universality) and (3) as a part of a distinguishable pathway, (4) being a known cause of phenotypic addiction of neoplastic cells and thus a promising therapeutic target. Each of these universal oncogenic factors—mutant p53, KRAS and CMYC proteins, telomerase ribonucleoprotein, proteasome machinery, HSP molecular chaperones, NF-κB and WNT pathways, AP-1 and YAP/TAZ transcription factors and non-coding RNAs—has a vast network of molecular interrelations and common partners. Understanding this network allows for the hunt for novel therapeutic targets and protocols to counteract drug resistance in a clinical neoplasia treatment.
Collapse
|
6
|
Iervasi E, Auricchio R, Strangio A, Greco L, Saverino D. Serum IL-21 levels from celiac disease patients correlates with anti-tTG IgA autoantibodies and mucosal damage. Autoimmunity 2020; 53:225-230. [PMID: 32157915 DOI: 10.1080/08916934.2020.1736047] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Objectives: Coeliac disease is a multifactorial disorder influenced by environmental, genetic and immunological factors. Interleukin (IL)-21 has been linked to an increase disease risk and the serum level of IL-21 seems to be increased in CD compared to a healthy control population.Methods: Sera were collected from 160 CD patients, 120 untreated and 40 following a gluten-free diet, and form 45 healthy subjects. Serum IL-21 was evaluated by specific ELISA tests.Results: Our data show that patients with untreated CD display IL-21 concentrations significantly higher than both treated-CD patients (following a gluten-free diet) and controls. In addition, serum IL-21 correlates with serum titres of anti-tTG IgA autoantibodies. Finally, our results show a correlation of this cytokine with duodenal mucosal damage.Conclusions: A role of gluten, as antigen with stimulatory function on IL-21 production, seems to be confirmed by the longitudinal analyses showing that the gluten-free diet decreases to a nearly undetectable amount this cytokine. In addition, the finding of a positive correlation between the serum amount of IL-21 and the grade of duodenal mucosa damage suggests a strong immunomodulatory effect of this cytokine on cytotoxic T lymphocyte functions. This study provides an extra evidence to emerging data on the potential role IL-21 in CD pathogenesis, suggesting its involvement in the development and progression of CD. Significance statement: In untreated CD, serum IL-21 shows higher levels compared with treated CD and healthy subjects. Serum amounts of IL-21 correlate with anti-tTG IgA autoantibodies and with duodenal mucosa damage.
Collapse
Affiliation(s)
- Erika Iervasi
- Department of Experimental Medicine, University of Genova, Genova, Italy.,Laboratory of Autoimmunology, Ospedale Policlinico San Martino, Largo Rosanna Benzi, Genova, Italy
| | - Renata Auricchio
- Department of Translational Medical Science, University of Naples Federico II, Napoli, Italy.,European Laboratory for Food-Induced disease (ELFID), University of Naples Federico II, Napoli, Italy
| | - Antonella Strangio
- Department of Experimental Medicine, University of Genova, Genova, Italy
| | - Luigi Greco
- Department of Translational Medical Science, University of Naples Federico II, Napoli, Italy.,European Laboratory for Food-Induced disease (ELFID), University of Naples Federico II, Napoli, Italy
| | - Daniele Saverino
- Department of Experimental Medicine, University of Genova, Genova, Italy.,Laboratory of Autoimmunology, Ospedale Policlinico San Martino, Largo Rosanna Benzi, Genova, Italy
| |
Collapse
|
7
|
Soderquist CR, Bhagat G. Gastrointestinal T- and NK-cell lymphomas and indolent lymphoproliferative disorders. Semin Diagn Pathol 2020; 37:11-23. [DOI: 10.1053/j.semdp.2019.08.001] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
|
8
|
Castellanos-Rubio A, Santin I, Olazagoitia-Garmendia A, Romero-Garmendia I, Jauregi-Miguel A, Legarda M, Bilbao JR. A novel RT-QPCR-based assay for the relative quantification of residue specific m6A RNA methylation. Sci Rep 2019; 9:4220. [PMID: 30862814 PMCID: PMC6414506 DOI: 10.1038/s41598-019-40018-6] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2018] [Accepted: 02/06/2019] [Indexed: 11/25/2022] Open
Abstract
N6-methyladenosine (m6A) is the most common and abundant RNA modification. Recent studies have shown its importance in the regulation of several biological processes, including the immune response, and different approaches have been developed in order to map and quantify m6A marks. However, site specific detection of m6A methylation has been technically challenging, and existing protocols are long and tedious and often involve next-generation sequencing. Here, we describe a simple RT-QPCR based approach for the relative quantification of candidate m6A regions that takes advantage of the diminished capacity of BstI enzyme to retrotranscribe m6A residues. Using this technique, we have been able to confirm the recently described m6A methylation in the 3′UTR of SOCS1 and SOCS3 transcripts. Moreover, using the method presented here, we have also observed alterations in the relative levels of m6A in specific motifs of SOCS genes in celiac disease patients and in pancreatic β-cells exposed to inflammatory stimuli.
Collapse
Affiliation(s)
- Ainara Castellanos-Rubio
- Department of Genetics, Physical Anthropology and Animal Physiology, University of the Basque Country (UPV-EHU), Biocruces-Bizkaia Health Research Institute, Leioa, Spain. .,IKERBASQUE, Basque Foundation for Science, Bilbao, Spain. .,Spanish Biomedical Research Center in Diabetes and Associated Metabolic Disorders (CIBERDEM), Madrid, Spain.
| | - Izortze Santin
- Department of Biochemistry and Molecular Biology, University of the Basque Country (UPV-EHU), Endocrinology and Diabetes Research Group, Biocruces-Bizkaia Health Research Institute, Leioa, Spain.,Spanish Biomedical Research Center in Diabetes and Associated Metabolic Disorders (CIBERDEM), Madrid, Spain
| | - Ane Olazagoitia-Garmendia
- Department of Genetics, Physical Anthropology and Animal Physiology, University of the Basque Country (UPV-EHU), Biocruces-Bizkaia Health Research Institute, Leioa, Spain
| | - Irati Romero-Garmendia
- Department of Genetics, Physical Anthropology and Animal Physiology, University of the Basque Country (UPV-EHU), Biocruces-Bizkaia Health Research Institute, Leioa, Spain
| | - Amaia Jauregi-Miguel
- Department of Genetics, Physical Anthropology and Animal Physiology, University of the Basque Country (UPV-EHU), Biocruces-Bizkaia Health Research Institute, Leioa, Spain
| | - Maria Legarda
- Pediatric Gastroenterology Unit, Cruces University Hospital, University of the Basque Country (UPV-EHU), Barakaldo, Spain
| | - Jose Ramon Bilbao
- Department of Genetics, Physical Anthropology and Animal Physiology, University of the Basque Country (UPV-EHU), Biocruces-Bizkaia Health Research Institute, Leioa, Spain. .,Spanish Biomedical Research Center in Diabetes and Associated Metabolic Disorders (CIBERDEM), Madrid, Spain.
| |
Collapse
|
9
|
Tocilizumab Promotes Regulatory T-cell Alleviation in STAT3 Gain-of-function−associated Multi-organ Autoimmune Syndrome. Clin Ther 2017; 39:444-449. [DOI: 10.1016/j.clinthera.2017.01.004] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2016] [Revised: 12/06/2016] [Accepted: 01/03/2017] [Indexed: 11/19/2022]
|
10
|
Goswami R, Kaplan M. STAT Transcription Factors in T Cell Control of Health and Disease. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2017; 331:123-180. [DOI: 10.1016/bs.ircmb.2016.09.012] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
|
11
|
Borrelli M, Gianfrani C, Lania G, Aitoro R, Ferrara K, Nanayakkara M, Ponticelli D, Zanzi D, Discepolo V, Vitale S, Barone MV, Troncone R, Auricchio R, Maglio M. In the Intestinal Mucosa of Children With Potential Celiac Disease IL-21 and IL-17A are Less Expressed than in the Active Disease. Am J Gastroenterol 2016; 111:134-44. [PMID: 26753888 DOI: 10.1038/ajg.2015.390] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/05/2014] [Accepted: 11/11/2015] [Indexed: 12/11/2022]
Abstract
OBJECTIVES Potential celiac disease (CD) patients are at an increased risk to developing CD as indicated by positive CD-associated serology. We investigated in duodenal mucosa of such patients the presence of both IL-21 and IL-17A and the role of gliadin peptides and IL-15 in their expression. METHODS Duodenal biopsies from 76 active CD, 90 potential CD, and 58 control patients were analyzed for IL-21 and/or IL-17A production by quantitative real-time PCR, immunohistochemistry, flow cytometry, and ELISA. The presence of IL-21 receptor was investigated by western blot. Potential CD duodenal fragments were cultured with gliadin peptides (PTG) and/or IL-15 and the expression/production of IL-21 and IL-17A assessed by quantitative real-time PCR and by immunohistochemistry. RESULTS In potential CD, IL-21 was lower than in active CD, in terms of RNA expression (P<0.01), density of lamina propria (LP) IL-21(+) cells (P<0.05), and protein secretion (P<0.05). Also, IL-21R was weakly detectable in potential CD. Several LP cell types produced IL-21 in CD. In potential CD, CD4(+)IL-21(+) cells increased after PMA-ionomycin stimulation and co-produced IFN-γ but not IL-17A. After 24 hours of culture stimulation with PTG, IL-21-producing cells increased but not the ones producing IL-17A. This increase was further enhanced by the addition of IL-15 to culture medium. CONCLUSIONS In potential CD, IL-21 is less expressed than in active CD; however, IL-21-producing cells are present and prone to respond after specific stimuli. This suggests a key role of IL-21 in the progression of mucosal damage in CD.
Collapse
Affiliation(s)
- Melissa Borrelli
- Department of Pediatrics, European Laboratory for the Investigation of Food-Induced Diseases, University Federico II, Naples, Italy
| | | | - Giuliana Lania
- Department of Pediatrics, European Laboratory for the Investigation of Food-Induced Diseases, University Federico II, Naples, Italy
| | - Rosita Aitoro
- Department of Pediatrics, European Laboratory for the Investigation of Food-Induced Diseases, University Federico II, Naples, Italy
| | - Katia Ferrara
- Department of Pediatrics, European Laboratory for the Investigation of Food-Induced Diseases, University Federico II, Naples, Italy
| | - Merlin Nanayakkara
- Department of Pediatrics, European Laboratory for the Investigation of Food-Induced Diseases, University Federico II, Naples, Italy
| | - Domenico Ponticelli
- Department of Pediatrics, European Laboratory for the Investigation of Food-Induced Diseases, University Federico II, Naples, Italy
| | - Delia Zanzi
- Department of Pediatrics, European Laboratory for the Investigation of Food-Induced Diseases, University Federico II, Naples, Italy
| | - Valentina Discepolo
- Department of Pediatrics, European Laboratory for the Investigation of Food-Induced Diseases, University Federico II, Naples, Italy
| | - Serena Vitale
- Institute of Protein Biochemistry, CNR, Naples, Italy
| | - Maria Vittoria Barone
- Department of Pediatrics, European Laboratory for the Investigation of Food-Induced Diseases, University Federico II, Naples, Italy
| | - Riccardo Troncone
- Department of Pediatrics, European Laboratory for the Investigation of Food-Induced Diseases, University Federico II, Naples, Italy
| | - Renata Auricchio
- Department of Pediatrics, European Laboratory for the Investigation of Food-Induced Diseases, University Federico II, Naples, Italy
| | - Mariantonia Maglio
- Department of Pediatrics, European Laboratory for the Investigation of Food-Induced Diseases, University Federico II, Naples, Italy
| |
Collapse
|
12
|
Diosdado B, Wijmenga C. Molecular mechanisms of the adaptive, innate and regulatory immune responses in the intestinal mucosa of celiac disease patients. Expert Rev Mol Diagn 2014; 5:681-700. [PMID: 16149872 DOI: 10.1586/14737159.5.5.681] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Celiac disease is a complex genetic disorder that affects the small intestine of genetically predisposed individuals when they ingest gluten, a dietary protein. Although several genome screens have been successful in identifying susceptibility loci in celiac disease, the only genetic contributors identified so far are the human leukocyte antigen (HLA)-DQ2/DQ8 molecules. One of the most important aspects in the pathogenesis of celiac disease is the activation of a T-helper 1 immune response, when the antigen-presenting cells that express HLA-DQ2/DQ8 molecules present the toxic gluten peptides to reactive CD4(+) T-cells. Recently, new insights into the activation of an innate immune response have also been described. It is generally accepted that the immune response triggers destruction of the mucosa in the small intestine of celiac disease patients. Hence, the activation of a detrimental immune response in the intestine of celiac disease patients appears to be key in the initiation and progression of the disease. This review summarizes the immunologic pathways that have been studied in celiac disease thus far, and will point to new potential candidate genes and pathways involved in the etiopathogenesis of celiac disease, which should lead to novel alternatives for diagnosis and treatment.
Collapse
Affiliation(s)
- Begoña Diosdado
- University Medical Centre, Complex Genetics Section, Stratenum 2.117, Department of Biomedical Genetics, PO Box 85060, 3508 AB Utrecht, The Netherlands.
| | | |
Collapse
|
13
|
Rauch I, Müller M, Decker T. The regulation of inflammation by interferons and their STATs. JAKSTAT 2013; 2:e23820. [PMID: 24058799 PMCID: PMC3670275 DOI: 10.4161/jkst.23820] [Citation(s) in RCA: 173] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2013] [Revised: 01/29/2013] [Accepted: 01/29/2013] [Indexed: 12/30/2022] Open
Abstract
Interferons (IFN) are subdivided into type I IFN (IFN-I, here synonymous with IFN-α/β), type II (IFN-γ) and type III IFN (IFN-III/IFN-λ) that reprogram nuclear gene expression through STATs 1 and 2 by forming STAT1 dimers (mainly IFN-γ) or the ISGF3 complex, a STAT1-STAT2-IRF9 heterotrimer (IFN-I and IFN-III). Dominant IFN activities in the immune system are to protect cells from viral replication and to activate macrophages for enhanced effector function. However, the impact of IFN and their STATs on the immune system stretches far beyond these activities and includes the control of inflammation. The goal of this review is to give an overview of the different facets of the inflammatory process that show regulatory input by IFN/STAT.
Collapse
Affiliation(s)
- Isabella Rauch
- Max F. Perutz Laboratories; University of Vienna; Vienna, Austria
| | - Mathias Müller
- Institute of Animal Breeding and Genetics and Biomodels Austria; University of Veterinary Medicine Vienna; Vienna, Austria
| | - Thomas Decker
- Max F. Perutz Laboratories; University of Vienna; Vienna, Austria
| |
Collapse
|
14
|
Santin I, Moore F, Colli ML, Gurzov EN, Marselli L, Marchetti P, Eizirik DL. PTPN2, a candidate gene for type 1 diabetes, modulates pancreatic β-cell apoptosis via regulation of the BH3-only protein Bim. Diabetes 2011; 60:3279-88. [PMID: 21984578 PMCID: PMC3219938 DOI: 10.2337/db11-0758] [Citation(s) in RCA: 112] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
OBJECTIVE Genome-wide association studies allowed the identification of several associations between specific loci and type 1 diabetes (T1D). However, the mechanisms by which most candidate genes predispose to T1D remain unclear. We presently evaluated the mechanisms by which PTPN2, a candidate gene for T1D, modulates β-cell apoptosis after exposure to type I and II interferons (IFNs), cytokines that contribute to β-cell loss in early T1D. RESEARCH DESIGN AND METHODS Small interfering RNAs were used to inhibit PTPN2, STAT1, Bim, and Jun NH(2)-terminal kinase 1 (JNK1) expression. Cell death was assessed by Hoechst and propidium iodide staining. BAX translocation, Bim phosphorylation, cytochrome c release, and caspases 9 and 3 activation were measured by Western blot or immunofluorescence. RESULTS PTPN2 knockdown exacerbated type I IFN-induced apoptosis in INS-1E, primary rat, and human β-cells. PTPN2 silencing and exposure to type I and II IFNs induced BAX translocation to the mitochondria, cytochrome c release, and caspase 3 activation. There was also an increase in Bim phosphorylation that was at least in part regulated by JNK1. Of note, both Bim and JNK1 knockdown protected β-cells against IFN-induced apoptosis in PTPN2-silenced cells. CONCLUSIONS The present findings suggest that local IFN production may interact with a genetic factor (PTPN2) to induce aberrant proapoptotic activity of the BH3-only protein Bim, resulting in increased β-cell apoptosis via JNK activation and the intrinsic apoptotic pathway. This is the first indication of a direct interaction between a candidate gene for T1D and the activation of a specific downstream proapoptotic pathway in β-cells.
Collapse
Affiliation(s)
- Izortze Santin
- Laboratory of Experimental Medicine, Université Libre de Bruxelles, Brussels, Belgium
| | - Fabrice Moore
- Laboratory of Experimental Medicine, Université Libre de Bruxelles, Brussels, Belgium
| | - Maikel L. Colli
- Laboratory of Experimental Medicine, Université Libre de Bruxelles, Brussels, Belgium
| | - Esteban N. Gurzov
- Laboratory of Experimental Medicine, Université Libre de Bruxelles, Brussels, Belgium
| | - Lorella Marselli
- Department of Endocrinology and Metabolism, Metabolic Unit, University of Pisa, Pisa, Italy
| | - Piero Marchetti
- Department of Endocrinology and Metabolism, Metabolic Unit, University of Pisa, Pisa, Italy
| | - Decio L. Eizirik
- Laboratory of Experimental Medicine, Université Libre de Bruxelles, Brussels, Belgium
- Corresponding author: Decio L. Eizirik,
| |
Collapse
|
15
|
Lahdenperä A, Ludvigsson J, Fälth-Magnusson K, Högberg L, Vaarala O. The effect of gluten-free diet on Th1-Th2-Th3-associated intestinal immune responses in celiac disease. Scand J Gastroenterol 2011; 46:538-49. [PMID: 21288140 DOI: 10.3109/00365521.2011.551888] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
OBJECTIVE To study T-helper (Th)1-Th2-Th3 gene activation profile in the small intestine and peripheral blood of children with celiac disease (CD) with special interest in the response to the gluten-free diet (GFD) treatment in order to elucidate an immune dysregulation not triggered by gluten. MATERIAL AND METHODS Small intestinal biopsies and venous blood were taken from seven children with CD (mean age: 8 years, four girls) at presentation and after 1 year of strict GFD. The Th1-Th2-Th3 gene expression profile was examined by real-time PCR arrays. The findings were compared with the corresponding expressions in peripheral blood and small intestinal biopsies from six reference children without CD (mean age: 6 years, four girls). RESULTS The Th1 gene expression profile including interferon (IFN)-γ, signal transducer and activator of transcription (STAT) 1 and interferon regulatory factor (IRF) 1 together with reduced interleukin (IL)-2 expression was pronounced in small intestinal biopsies from children with untreated CD. A downregulation of IFN-γ transcripts was seen after 1 year of GFD, but there was still increased expression of STAT1 and IRF1 in association with low IL-2 expression in spite of eliminated exposure to wheat gluten. By contrast, the decreased intestinal expression of Th2 gene markers observed at presentation was normalized with GFD. The alterations in the mucosal gene expression profile were not reflected in peripheral blood. CONCLUSION The GFD did not correct the increased activation of the IFN-γ signaling pathway related markers and reduced IL-2 expression, suggesting that they represent an immune dysregulation not dependent on gluten exposure.
Collapse
Affiliation(s)
- Anne Lahdenperä
- Division of Paediatrics, Department of Clinical and Experimental Medicine, Faculty of Health Sciences, Linköping University, Linköping, Sweden.
| | | | | | | | | |
Collapse
|
16
|
Han X, Mann E, Gilbert S, Guan Y, Steinbrecher KA, Montrose MH, Cohen MB. Loss of guanylyl cyclase C (GCC) signaling leads to dysfunctional intestinal barrier. PLoS One 2011; 6:e16139. [PMID: 21305056 PMCID: PMC3031533 DOI: 10.1371/journal.pone.0016139] [Citation(s) in RCA: 73] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2010] [Accepted: 12/11/2010] [Indexed: 12/12/2022] Open
Abstract
Background Guanylyl Cyclase C (GCC) signaling via uroguanylin (UGN) and guanylin activation is a critical mediator of intestinal fluid homeostasis, intestinal cell proliferation/apoptosis, and tumorigenesis. As a mechanism for some of these effects, we hypothesized that GCC signaling mediates regulation of intestinal barrier function. Methodology/Principal Findings Paracellular permeability of intestinal segments was assessed in wild type (WT) and GCC deficient (GCC−/−) mice with and without lipopolysaccharide (LPS) challenge, as well as in UGN deficient (UGN−/−) mice. IFNγ and myosin light chain kinase (MLCK) levels were determined by real time PCR. Expression of tight junction proteins (TJPs), phosphorylation of myosin II regulatory light chain (MLC), and STAT1 activation were examined in intestinal epithelial cells (IECs) and intestinal mucosa. The permeability of Caco-2 and HT-29 IEC monolayers, grown on Transwell filters was determined in the absence and presence of GCC RNA interference (RNAi). We found that intestinal permeability was increased in GCC−/− and UGN−/− mice compared to WT, accompanied by increased IFNγ levels, MLCK and STAT1 activation in IECs. LPS challenge promotes greater IFNγ and STAT1 activation in IECs of GCC−/− mice compared to WT mice. Claudin-2 and JAM-A expression were reduced in GCC deficient intestine; the level of phosphorylated MLC in IECs was significantly increased in GCC−/− and UGN−/− mice compared to WT. GCC knockdown induced MLC phosphorylation, increased permeability in IEC monolayers under basal conditions, and enhanced TNFα and IFNγ-induced monolayer hyperpermeability. Conclusions/Significance GCC signaling plays a protective role in the integrity of the intestinal mucosal barrier by regulating MLCK activation and TJ disassembly. GCC signaling activation may therefore represent a novel mechanism in maintaining the small bowel barrier in response to injury.
Collapse
Affiliation(s)
- Xiaonan Han
- Division of Gastroenterology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, United States of America.
| | | | | | | | | | | | | |
Collapse
|
17
|
Castellanos-Rubio A, Santin I, Martin-Pagola A, Irastorza I, Castaño L, Vitoria JC, Bilbao JR. Long-term and acute effects of gliadin on small intestine of patients on potentially pathogenic networks in celiac disease. Autoimmunity 2010; 43:131-9. [PMID: 19814655 DOI: 10.3109/08916930903225229] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Celiac disease (CD) is a complex, immune-mediated intolerance to gliadin that develops in genetically susceptible individuals. Although the main driving force of the disease is an aberrant autoimmune response, several other pathogenic mechanisms, many still unidentified, are also involved. In order to describe at a network level the alterations provoked by a gliadin insult on the intestinal mucosa of patients, we compared the expression profiles of biopsies from 9 active and 9 treated patients (long-term effects of gliadin), and of 10 biopsies from gluten-free diet treated patients that were incubated in vitro with or without gliadin (acute effects) and integrated significantly altered transcripts into potentially pathogenic biological processes. Using information on Kyoto Encyclopedia of Genes and Genomes pathways and Gene Ontology terms represented among the differentially expressed genes, we observed important dysfunction in several complex networks, including those related to cell-cell communication, intracellular signaling, ubiquitin-proteasome system, cell cycle/apoptosis and extracellular matrix. The reconstruction of the role of these biological networks in the development of the intestinal lesion in CD provides a comprehensive picture of key events that contribute to the disease, and could point towards novel functional candidates that might be potential therapeutic targets or responsible for genetic susceptibility.
Collapse
|
18
|
Osmond RIW, Das S, Crouch MF. Development of cell-based assays for cytokine receptor signaling, using an AlphaScreen SureFire assay format. Anal Biochem 2010; 403:94-101. [PMID: 20382104 DOI: 10.1016/j.ab.2010.04.007] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2010] [Revised: 03/21/2010] [Accepted: 04/05/2010] [Indexed: 11/27/2022]
Abstract
The signal transducers and activators of transcription (STAT) proteins are a small family of signaling proteins that are crucial for cytokine and growth factor receptor-mediated signaling in various blood cell types. Despite their central role in immune and hematopoietic cellular regulation, there are relatively few options for monitoring receptor-mediated JAK/STAT signaling events in a cell-based format, without the need for cellular transfections or labor intensive methodology. Indeed, traditional methods such as the Western blot or ELISA remain a standard method for determining the phosphorylation status of endogenous STAT proteins. Here we present data for the rapid detection of endogenous receptor-mediated phosphorylation of multiple STAT proteins using the bead-based AlphaScreen SureFire technology. With three different cell lines (human acute monocytic leukemia THP-1 cells, human erythroleukemic TF-1 cells, and human T lymphocytic Jurkat cells), we have optimized a rapid and homogeneous methodology for monitoring endogenous, receptor-mediated signaling via STAT 1, STAT 3, or STAT 5 phosphorylation, in response to several agonists. These assays, which can be tailored for both standard research applications or high-throughput drug screening applications, afford quantitative data for receptor-mediated signaling mechanisms in an endogenous, cellular environment.
Collapse
|
19
|
Slattery ML, Fitzpatrick FA. Convergence of hormones, inflammation, and energy-related factors: a novel pathway of cancer etiology. Cancer Prev Res (Phila) 2010; 2:922-30. [PMID: 19892662 DOI: 10.1158/1940-6207.capr-08-0191] [Citation(s) in RCA: 62] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Colorectal cancer (CRC) is a multifactorial disease with several hypothesized etiologic factors including inflammatory processes; hormones such as estrogen, androgen, and insulin; and energy-related factors. We present evidence that integrates these elements in a pathway we call the convergence of hormones, inflammation, and energy-related factors (CHIEF). First, given the physiology of the gut, substantial epidemiologic and molecular data support the hypothesis that activation of innate immunity in the normal gut mucosa by various environmental agents (commensal bacteria, dietary antigens, mucosal irritants, pathogens) and endogenous factors such as estrogen, androgens, and insulin levels provokes basal inflammation as an underlying factor of the association of insulin, estrogen, and energy-related factors with CRC. Second, critical genes involved in this pathway, e.g., phosphatase tensin homologue on chromosome 10 (PTEN) and serine threonine kinase 11 (STK11)/LKB1, are tumor suppressor genes often mutated in intestinal cancer or CRC. Third, laboratory experiments show that cellular PTEN and STK11/LKB1 tumor suppressor enzymes are vulnerable to inactivation by redox-active species, especially chemically reactive lipid mediators of inflammation and redox stress. Epidemiologic data further support the underlying proposal that CHIEF comprises important elements of CRC risk. Although this discussion of the CHIEF pathway focuses on CRC, we believe that this pathway may play an important role in the etiology of other cancers as well.
Collapse
Affiliation(s)
- Martha L Slattery
- Department of Medicine, University of Utah, Salt Lake City, Utah 84132, USA.
| | | |
Collapse
|
20
|
Frisullo G, Nociti V, Iorio R, Patanella AK, Plantone D, Bianco A, Marti A, Cammarota G, Tonali PA, Batocchi AP. T-bet and pSTAT-1 expression in PBMC from coeliac disease patients: new markers of disease activity. Clin Exp Immunol 2009; 158:106-14. [PMID: 19737237 DOI: 10.1111/j.1365-2249.2009.03999.x] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
Coeliac disease (CD) is considered a T cell-mediated autoimmune disease, and up-regulation of T-bet and phosphorylated signal transducers and activators of transcription (pSTAT)1, key transcription factors for the development of T helper type 1 (Th1) cells, has been described in the mucosa of patients with untreated CD. Using transcription factor analysis, we investigated whether T-bet and pSTAT1 expressions are up-regulated in the peripheral blood of CD patients and correlate with disease activity. Using flow cytometry, we analysed T-bet, pSTAT1 and pSTAT3 expression in CD4(+), CD8(+) T cells, CD19(+) B cells and monocytes from peripheral blood of 15 untreated and 15 treated CD patients and 30 controls, and longitudinally in five coeliac patients before and after dietary treatment. We evaluated using enzyme-linked immunosorbent assay (ELISA), interferon (FN)-gamma, interleukin (IL)-17 and IL-10 production by peripheral blood mononuclear cell (PBMC) cultures. T-bet expression in CD4(+), CD8(+) T cells, CD19(+) B cells and monocytes and IFN-gamma production by PBMC was higher in untreated than in treated CD patients and controls. pSTAT1 expression was higher in CD4(+)T cells, B cells and monocytes from untreated than from treated CD patients and controls. pSTAT3 was increased only in monocytes from untreated patients compared with CD-treated patients and controls. The data obtained from the longitudinal evaluation of transcription factors confirmed these results. Flow cytometric analysis of pSTAT1 and T-bet protein expression in peripheral blood mononuclear cells could be useful and sensible markers in the follow-up of CD patients to evaluate disease activity and response to dietary treatment.
Collapse
Affiliation(s)
- G Frisullo
- Department of Neurosciences, Catholic University, Largo Agostino Gemelli 8, Rome, Italy
| | | | | | | | | | | | | | | | | | | |
Collapse
|
21
|
Celiac disease: from oral tolerance to intestinal inflammation, autoimmunity and lymphomagenesis. Mucosal Immunol 2009; 2:8-23. [PMID: 19079330 DOI: 10.1038/mi.2008.75] [Citation(s) in RCA: 93] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Celiac disease is a multifactorial disorder and provides a privileged model to decipher how the interplay between environmental and genetic factors can alter mucosal tolerance to a food antigen, lead to chronic intestinal inflammation, and ultimately promote T-cell lymphomagenesis. Here we summarize how HLA-DQ2/8 molecules, the main genetic risk factor for this disease can orchestrate a CD4(+) T-cell adaptive immune response against gluten, and discuss recent data which shed light on the innate and adaptive immune stimuli that collaborate to induce a proinflammatory TH1 response, a massive expansion of intraepithelial lymphocytes, and a cytolytic attack of the epithelium. The intestinal immune response driven in genetically predisposed patients by chronic exposure to gluten emerges as the pathological counterpart of normal acute intestinal responses to intracellular pathogens.
Collapse
|
22
|
Garrote JA, Gómez E, León AJ, Bernardo D, Calvo C, Fernández-Salazar L, Blanco-Quirós A, Arranz E. Cytokine, Chemokine and Immune Activation Pathway Profiles in Celiac Disease: An Immune System Activity Screening by Expression Macroarrays. Drug Target Insights 2008. [DOI: 10.4137/dti.s399] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Affiliation(s)
- José A. Garrote
- Group of Mucosal Immunology. Pediatrics and Immunology Areas- Instituto de Biologia y Genética Molecular (IBGM). University of Valladolid. (Spain)
- Research Unit. (Spain)
| | - Emma Gómez
- Group of Mucosal Immunology. Pediatrics and Immunology Areas- Instituto de Biologia y Genética Molecular (IBGM). University of Valladolid. (Spain)
| | - Alberto J. León
- Group of Mucosal Immunology. Pediatrics and Immunology Areas- Instituto de Biologia y Genética Molecular (IBGM). University of Valladolid. (Spain)
| | - David Bernardo
- Group of Mucosal Immunology. Pediatrics and Immunology Areas- Instituto de Biologia y Genética Molecular (IBGM). University of Valladolid. (Spain)
| | | | - Luis Fernández-Salazar
- Adults Digestive Diseases Services. Hospital Clinico Universitario of Valladolid. (Spain)
| | - Alfredo Blanco-Quirós
- Group of Mucosal Immunology. Pediatrics and Immunology Areas- Instituto de Biologia y Genética Molecular (IBGM). University of Valladolid. (Spain)
| | - Eduardo Arranz
- Group of Mucosal Immunology. Pediatrics and Immunology Areas- Instituto de Biologia y Genética Molecular (IBGM). University of Valladolid. (Spain)
| |
Collapse
|
23
|
Martinez-Lostao L, Ordi-Ros J, Balada E, Segarra-Medrano A, Majó-Masferrer J, Labrador-Horrillo M, Vilardell-Tarrés M. Activation of the signal transducer and activator of transcription-1 in diffuse proliferative lupus nephritis. Lupus 2007; 16:483-8. [PMID: 17670846 DOI: 10.1177/0961203307079618] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Difuse proliferative lupus nephritis (DPLN) is the most common and severe form of lupus nephritis. A predominance of IFN-gamma-producing T cells in both peripheral and renal tissues of patients with DPLN has been identified which suggests an important role for cell-mediated immunity in the pathogenesis of this complication in SLE. The biological effects of IFN-gamma rely mainly on the activity of the transcription factor called signal transducer and activator of transcription (STAT)-1. To assess the IFN-gamma/STAT-1 pathway in DPLN, we examined the expression of STAT-1 in renal biopsies from 15 DPLN patients by immunohistochemical staining with an anti-STAT-1 antibody. The expression of STAT-1 in renal tissues was correlated with several clinical and laboratory findings in these DNPN patients.STAT-1 was activated in the tubular cells in all DPLN patients. Seven of 15 DPLN biopsies (46.7%) showed positive cells in glomeruli. Five of these seven DPLN biopsies (71.4%) with positive glomerular cells showed a serum creatinine >1.5 mg/mL at the time the biopsy was carried out whereas only one of eight DPLN biopsy specimens (12.5%) without positive glomerular cells, showed a serum creatinine >1.5 mg/mL (P = 0.041). Moreover, the percentage of DPLN patients with a worse renal outcome in those who showed expression of STAT-1 in glomerulari were higher in comparison to those without STAT-1 expression (P = 0.041). Our results show that STAT-1 is activated in DPLN suggesting that biological effects of IFN-gamma in renal tissues depend, at least in part, on the activation of STAT-1.
Collapse
Affiliation(s)
- L Martinez-Lostao
- Autoimmune Diseases Research Lab, Vall d'Hebron Research Institute, Barcelona, Spain
| | | | | | | | | | | | | |
Collapse
|
24
|
Terrazzano G, Sica M, Gianfrani C, Mazzarella G, Maurano F, De Giulio B, de Saint-Mezard S, Zanzi D, Maiuri L, Londei M, Jabri B, Troncone R, Auricchio S, Zappacosta S, Carbone E. Gliadin regulates the NK-dendritic cell cross-talk by HLA-E surface stabilization. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2007; 179:372-81. [PMID: 17579058 DOI: 10.4049/jimmunol.179.1.372] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
We analyzed the autologous NK cell interaction with gliadin-presenting dendritic cells. Gliadin is the known Ag priming the celiac disease (CD) pathogenesis. We demonstrate that gliadin prevents immature dendritic cells (iDCs) elimination by NK cells. Furthermore, cooperation between human NK cells-iDCs and T cells increases IFN-gamma production of anti-gliadin immune response. Gliadin fractions were analyzed for their capability to stabilize HLA-E molecules. The alpha and omega fractions conferred the protection from NK cell lysis to iDCs and increased their HLA-E expression. Gliadin pancreatic enzyme digest and a peptide derived from gliadin alpha increased HLA-E levels on murine RMA-S/HLA-E-transfected cells. Analysis of HLA-E expression in the small intestinal mucosa of gluten-containing diet celiac patients and organ culture experiments confirmed the in vitro data.
Collapse
Affiliation(s)
- Giuseppe Terrazzano
- Department of Cellular and Molecular Biology and Pathology, University Federico II, Naples, Italy.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
25
|
Abstract
Celiac disease (CD) is an intestinal disorder caused by an altered immune response against wheat gluten, a common dietary antigen, and related cereal proteins. Both CD4+ and CD8+ T cells have a role in inducing the intestinal damage, although recent studies have also pinpointed the involvement of the innate immune response in CD pathogenesis. So far, the only available treatment for CD is the strict avoidance of gluten in the diet, but the poor compliance and the associated complications demand alternative therapies. During the last decade, the knowledge of genetic, molecular and cellular mechanisms leading to CD pathogenesis made great progress. The improved understanding of gluten peptides activating either adaptive or innate immune response, of HLA restriction molecules, as well as of cytokines that mediate most of the inflammatory reactions, opens several new promising perspectives for therapeutic intervention. This review discusses both molecular and cellular strategies to treat CD, including the use of proteolytic enzymes active on gluten peptides, antibodies neutralising IL-15 and IFN-gamma, drugs targeting HLA, regulatory cytokines and T cells.
Collapse
|
26
|
Chen W, Khurana Hershey GK. Signal transducer and activator of transcription signals in allergic disease. J Allergy Clin Immunol 2007; 119:529-41; quiz 542-3. [PMID: 17336608 DOI: 10.1016/j.jaci.2007.01.004] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2006] [Revised: 01/03/2007] [Accepted: 01/05/2007] [Indexed: 01/05/2023]
Abstract
Signal transducer and activator of transcription (STAT) proteins are a group of transcription factors that transmit signals from the extracellular milieu of cells to the nucleus. They are crucial for the signaling of many cytokines that are mediators of allergic inflammation and impact various cell types critical to allergy including epithelial cells, mast cells, lymphocytes, dendritic cells, and eosinophils. Dysregulation of STAT signaling has been implicated in allergic disease, highlighting the importance of these ubiquitous molecules in allergic inflammation and the potential of these pathways as a target for therapeutic intervention. This review will summarize the current understanding of the roles of STAT signaling in allergic disease and the potential of targeting STATs for the treatment of allergic disorders, emphasizing recent observations.
Collapse
Affiliation(s)
- Weiguo Chen
- Division of Allergy and Immunology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA
| | | |
Collapse
|
27
|
Branski D, Fasano A, Troncone R. Latest developments in the pathogenesis and treatment of celiac disease. J Pediatr 2006; 149:295-300. [PMID: 16939736 DOI: 10.1016/j.jpeds.2006.06.003] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/21/2005] [Revised: 05/05/2006] [Accepted: 06/05/2006] [Indexed: 01/01/2023]
Affiliation(s)
- David Branski
- Department of Pediatrics, Hadassah University Hospitals, Hadassah Medical Organization, 91120 Jerusalem, Israel.
| | | | | |
Collapse
|
28
|
Diosdado B, Monsuur AJ, Mearin ML, Mulder C, Wijmenga C. The downstream modulator of interferon-γ, STAT1 is not genetically associated to the Dutch coeliac disease population. Eur J Hum Genet 2006; 14:1120-4. [PMID: 16773129 DOI: 10.1038/sj.ejhg.5201667] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022] Open
Abstract
Coeliac disease (CD) is a complex genetic disorder. Its etiology is owing to multiple genes and environmental factors, such as gluten. The first event in the pathogenesis of CD after the ingestion of gluten is the activation of a Th1 immune response that leads to villous atrophy. Although this immune response seems crucial to the disease's development, only the HLA-DQ2/DQ8 genes have been identified as causative immune genes related to CD. Recently, the activation of the transcription factor STAT1 and changes in its expression levels have confirmed the participation of the Janus kinase-signal transducer and activator of transcription pathway in CD. Furthermore, as the STAT-1 gene is a positional candidate located in the CELIAC3 locus on chromosome 2, we speculate that alterations in this gene could be primarily responsible for the aberrant immune response that characterizes CD. Based on this functional and genetic evidence, we investigated the primary contribution of STAT-1 to CD. We performed a comprehensive genetic association study using five tag SNPs fully covering the STAT-1 gene in a Dutch cohort of 355 independent CD cases and 360 healthy controls. Neither the alleles, nor the genotypes in the case-control genetic association studies, nor the haplotype analysis showed any association to the STAT-1 gene in the Dutch CD population. Our results do not point to a primary involvement of the STAT-1 gene in the Dutch CD population.
Collapse
Affiliation(s)
- Begoña Diosdado
- Complex Genetics Section, Department of Biomedical Genetics, University Medical Centre Utrecht, Utrecht, The Netherlands
| | | | | | | | | |
Collapse
|
29
|
Resta-Lenert S, Barrett KE. Probiotics and commensals reverse TNF-alpha- and IFN-gamma-induced dysfunction in human intestinal epithelial cells. Gastroenterology 2006; 130:731-46. [PMID: 16530515 DOI: 10.1053/j.gastro.2005.12.015] [Citation(s) in RCA: 227] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/02/2004] [Accepted: 11/23/2005] [Indexed: 12/12/2022]
Abstract
BACKGROUND & AIMS Commensal bacteria are crucial for the development of the mucosal immune system. Probiotics are commensals with special characteristics and may protect mucosal surfaces against pathogens. Pathogens cause significant phenotypic alterations in infected epithelial cells, and probiotics reverse these deleterious responses. We hypothesized that probiotics and/or commensals may also reverse epithelial damage produced by cytokines. METHODS Human intestinal epithelial cells were exposed basolaterally to interferon (IFN)-gamma (10(3) U/mL) or tumor necrosis factor (TNF)-alpha (10 ng/mL) for up to 48 hours and assessed for ion transport, transepithelial resistance (TER), and epithelial permeability in the presence or absence of probiotics (Streptococcus thermophilus [ST] and Lactobacillus acidophilus [LA]), or the commensal, Bacteroides thetaiotaomicron (BT). RESULTS Agonist-stimulated chloride secretion was inhibited by IFN-gamma, an effect prevented by ST/LA or BT. The ability of ST/LA or BT to restore Cl(-) secretion was blocked by inhibitors of p38 MAPK, ERK1, 2, and PI3K. The cystic fibrosis transmembrane conductance regulator (CFTR) and the NKCC1 cotransporter were down-regulated by IFN-gamma, and ST/LA pretreatment reversed this effect. Both TNF-alpha and IFN-gamma significantly reduced TER and increased epithelial permeability, effects prevented by ST/LA or BT. A Janus kinase (JAK) inhibitor synergistically potentiated effects of ST/LA or BT on TER and permeability, but p38, ERK1, 2, or PI3K inhibition did not. Finally, only probiotic-treated epithelial cells exposed to cytokines showed reduced activation of SOCS3 and STAT1,3. CONCLUSIONS Deleterious effects of TNF-alpha and IFN-gamma on epithelial function are prevented by probiotic, and to a lesser extent, commensal pretreatment. These data extend the spectrum of effects of such bacteria on intestinal epithelial function and may justify their use in inflammatory disorders.
Collapse
Affiliation(s)
- Silvia Resta-Lenert
- Department of Medicine, University of California, San Diego, School of Medicine, San Diego, California, USA
| | | |
Collapse
|
30
|
Gianfrani C, Auricchio S, Troncone R. Adaptive and innate immune responses in celiac disease. Immunol Lett 2005; 99:141-5. [PMID: 15876458 DOI: 10.1016/j.imlet.2005.02.017] [Citation(s) in RCA: 97] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2005] [Revised: 02/16/2005] [Accepted: 02/22/2005] [Indexed: 12/13/2022]
Abstract
Celiac disease (CD) is a complex small intestinal disorder due to a dysregulated immune response to wheat gliadin and related proteins which leads to a small intestinal enteropathy. It is generally accepted that CD is a T-cell mediated disease, in which, gliadin derived peptides, either in native form or deamidated by tissue transglutaminase, activate lamina propria infiltrating T lymphocytes which release proinflammatory cytokines. Recent studies indicate that gliadin contains also peptides able to activate an innate immune response. In particular, they induce a selective expansion of IEL, particularly TCRgamma/delta+ and CD8+TCR alpha/beta+ lymphocytes bearing the CD94 NK receptor, as well as a strong epithelial expression of MICA molecules which interact with NKG2D receptor expressed on TCRgamma/delta+ and NK cells. Most of the events of innate immune activation events are inhibited by antibodies neutralizing IL-15, thus confirming the key role of this cytokine as a mediator of intestinal mucosa damage induced by ingestion of gliadin. It remains to be established to what extent the ability of gliadin peptides to activate innate immunity relates to other biological properties exerted not only on celiac cells and tissues; the specificity of celiac patients is probably related to their genetic make up.
Collapse
|
31
|
Monteleone I, Monteleone G, Del Vecchio Blanco G, Vavassori P, Cucchiara S, MacDonald TT, Pallone F. Regulation of the T helper cell type 1 transcription factor T-bet in coeliac disease mucosa. Gut 2004; 53:1090-5. [PMID: 15247173 PMCID: PMC1774159 DOI: 10.1136/gut.2003.030551] [Citation(s) in RCA: 68] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
BACKGROUND In coeliac disease (CD) mucosa, the histological lesion is associated with marked infiltration of T helper cell type 1 (Th1) cells. However, the molecular mechanisms which regulate Th1 cell differentiation in CD mucosa are unknown. AIMS To analyse expression of transcription factors which control the Th1 cell commitment in CD. PATIENTS Duodenal mucosal samples were taken from untreated CD patients and normal controls. METHODS Interferon gamma (IFN-gamma) and interleukin (IL)-4 RNA expression was examined in T lamina propria lymphocytes by quantitative reverse transcription-polymerase chain reaction. T-bet and STAT-4, two Th1 promoting transcription factors, and STAT-6 and GATA-3, transcription factors which govern T helper cell type 2 (Th2) cell polarisation, were examined in duodenal biopsies by western blotting. The effect of gliadin and IFN-gamma on expression of T-bet was examined in an ex vivo culture of biopsies taken from normal and treated CD patients. RESULTS As expected, IFN-gamma but not IL-4 RNA transcripts were increased in the mucosa of CD patients in comparison with controls. CD mucosal samples consistently exhibited higher levels of T-bet than controls. However, no difference in active STAT-4 expression was seen between CD patients and controls, suggesting that Th1 polarisation was not induced by local IL-12. GATA-3 and STAT-6 were also low in both CD and control mucosa. In normal duodenal biopsies, IFN-gamma stimulated T-bet through a STAT-1 dependent mechanism. Challenge of treated CD but not control biopsies with gliadin enhanced T-bet and this effect was also inhibited by STAT-1 inhibition. CONCLUSIONS This study shows that activation of STAT-1 by IFN-gamma promotes T-bet in CD mucosa.
Collapse
Affiliation(s)
- I Monteleone
- Dipartimento di Medicina Interna e Centro di Eccellenza per lo studio delle malattie complesse e multifattoriali, Università Tor Vergata, Rome, Italy.
| | | | | | | | | | | | | |
Collapse
|
32
|
Cerf-Bensussan N, Cellier C, Heyman M, Brousse N, Schmitz J. Coeliac disease: an update on facts and questions based on the 10th International Symposium on Coeliac Disease. J Pediatr Gastroenterol Nutr 2003; 37:412-21. [PMID: 14508210 DOI: 10.1097/00005176-200310000-00004] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
|
33
|
Mudter J, Neurath MF. The role of signal transducers and activators of transcription in T inflammatory bowel diseases. Inflamm Bowel Dis 2003; 9:332-7. [PMID: 14555918 DOI: 10.1097/00054725-200309000-00008] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Signal transducers and activators of transcription (STAT) proteins are intracellular effector molecules of cytokine-modulated signaling. On the one hand, they play an important role in hematopoiesis and the development of the human immune system. STAT transcription factors are necessary for embryogenesis and the maintenance of the mammalian immune response. In the adult, STAT signaling is responsible for T-cell polarization toward interferon gamma-secreting Th1 T cells or interleukin 4-producing Th2 cells. On the other hand, these proteins are involved in the regulation of T-cell survival. STAT activation is strongly associated with tyrosine phosphorylation by tyrosine kinases, namely Jak1, Jak2, Jak3, and Tyk2. Counterregulatory mechanisms protecting from overwhelming STAT activation are represented by protein inhibitors of activated STATs and the SOCS family proteins. Because STAT proteins are key response elements of cytokine-induced T-cell activation, the characterization of STAT proteins is one step to elucidate disturbed T-cell function in inflammatory bowel disease. In particular, an activation of STAT-4 and STAT-3 in T cells seems to play a key pathogenic role in Crohn's disease.
Collapse
Affiliation(s)
- Jonas Mudter
- 1st Medical Clinic, Johannes Gutenberg University of Mainz, Germany.
| | | |
Collapse
|