1
|
Weinstein KN, Domeier PP, Ziegler SF. A splice of life: the discovery, function, and clinical implications of FOXP3 isoforms in autoimmune disease. Int Immunol 2024; 37:83-90. [PMID: 39136284 DOI: 10.1093/intimm/dxae049] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2024] [Accepted: 08/12/2024] [Indexed: 12/28/2024] Open
Abstract
Regulatory T cells (Tregs) are a specialized subset of CD4+ T cells essential for the maintenance of immune homeostasis and prevention of autoimmunity. Treg lineage and functions are programmed by the X-chromosome encoded transcription factor forkhead box P3 (FOXP3). In humans, multiple FOXP3 isoforms are generated through alternative splicing. A full-length isoform containing all coding exons (FOXP3-FL) and a version lacking the second exon (FOXP3-ΔE2) are the predominant FOXP3 isoforms. Additionally, there are two minor isoforms lacking either exon 7 (FOXP3-ΔE7) and both exons 2 and 7 (FOXP3-ΔE2ΔE7). Although healthy humans express approximately equal levels of the FOXP3-FL and FOXP3-ΔE2 isoforms, sole expression of FOXP3-ΔE2 results in the development of a systemic autoimmune disease that resembles immune dysregulation, polyendocrinopathy, enteropathy, X-linked (IPEX) syndrome. These clinical observations strongly suggest functional defects in suppression by Tregs programmed by the FOXP3-ΔE2 isoform. Work from the past two decades has provided phenotypic and functional evidence of differences between Tregs programmed by the FOXP3-FL, FOXP3-ΔE2, and FOXP3-ΔE7 isoforms. In this review, we discuss the discovery of the FOXP3 isoforms, differences in the phenotype and function of Tregs programmed by different FOXP3 isoforms, and the role that these isoforms are known to play in autoimmunity.
Collapse
Affiliation(s)
- Kristin N Weinstein
- Center for Fundamental Immunology, Benaroya Research Institute, Seattle, WA, 98101, USA
- Department of Immunology, University of Washington, Seattle, WA, USA
| | - Phillip P Domeier
- Center for Fundamental Immunology, Benaroya Research Institute, Seattle, WA, 98101, USA
| | - Steven F Ziegler
- Center for Fundamental Immunology, Benaroya Research Institute, Seattle, WA, 98101, USA
| |
Collapse
|
2
|
van Haaren MJH, Steller LB, Vastert SJ, Calis JJA, van Loosdregt J. Get Spliced: Uniting Alternative Splicing and Arthritis. Int J Mol Sci 2024; 25:8123. [PMID: 39125692 PMCID: PMC11311815 DOI: 10.3390/ijms25158123] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2024] [Revised: 07/21/2024] [Accepted: 07/22/2024] [Indexed: 08/12/2024] Open
Abstract
Immune responses demand the rapid and precise regulation of gene protein expression. Splicing is a crucial step in this process; ~95% of protein-coding gene transcripts are spliced during mRNA maturation. Alternative splicing allows for distinct functional regulation, as it can affect transcript degradation and can lead to alternative functional protein isoforms. There is increasing evidence that splicing can directly regulate immune responses. For several genes, immune cells display dramatic changes in isoform-level transcript expression patterns upon activation. Recent advances in long-read RNA sequencing assays have enabled an unbiased and complete description of transcript isoform expression patterns. With an increasing amount of cell types and conditions that have been analyzed with such assays, thousands of novel transcript isoforms have been identified. Alternative splicing has been associated with autoimmune diseases, including arthritis. Here, GWASs revealed that SNPs associated with arthritis are enriched in splice sites. In this review, we will discuss how alternative splicing is involved in immune responses and how the dysregulation of alternative splicing can contribute to arthritis pathogenesis. In addition, we will discuss the therapeutic potential of modulating alternative splicing, which includes examples of spliceform-based biomarkers for disease severity or disease subtype, splicing manipulation using antisense oligonucleotides, and the targeting of specific immune-related spliceforms using antibodies.
Collapse
Affiliation(s)
- Maurice J. H. van Haaren
- Center for Translational Immunology, University Medical Center Utrecht, 3584 CX Utrecht, The Netherlands
| | - Levina Bertina Steller
- Center for Translational Immunology, University Medical Center Utrecht, 3584 CX Utrecht, The Netherlands
| | - Sebastiaan J. Vastert
- Center for Translational Immunology, University Medical Center Utrecht, 3584 CX Utrecht, The Netherlands
- Division of Pediatric Rheumatology and Immunology, Wilhelmina Children’s Hospital, 3584 CX Utrecht, The Netherlands
| | - Jorg J. A. Calis
- Center for Translational Immunology, University Medical Center Utrecht, 3584 CX Utrecht, The Netherlands
| | - Jorg van Loosdregt
- Center for Translational Immunology, University Medical Center Utrecht, 3584 CX Utrecht, The Netherlands
| |
Collapse
|
3
|
Saleh QW, Mohammadnejad A, Tepel M. FOXP3 full length splice variant is associated with kidney allograft tolerance. Front Immunol 2024; 15:1389105. [PMID: 38660296 PMCID: PMC11040551 DOI: 10.3389/fimmu.2024.1389105] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2024] [Accepted: 03/29/2024] [Indexed: 04/26/2024] Open
Abstract
Background Progressive decline of allograft function leads to premature graft loss. Forkhead box P3 (FOXP3), a characteristic gene of T-regulatory cells, is known to be essential for auto-antigen tolerance. We assessed the hypothesis that low FOXP3 mRNA splice variant levels in peripheral blood cells early after transplantation are associated with progressive allograft injury. Methods Blood samples were prospectively collected from 333 incident kidney transplant recipients on the first and 29th postoperative day. We used quantitative polymerase chain reaction to determine transcripts of 3 isotypes of FOXP3 splice variants, including pre-mature FOXP3 and full length FOXP3 (FOXP3fl). We investigated the association between FOXP3 splice variant levels and the declines in estimated glomerular filtration rate (eGFR) of more than 5ml/min/1.73m2 within the first-year post-transplant using logistic regression. Results We observed lower FOXP3fl levels in recipients with declining eGFR (N = 132) than in recipients with stable eGFR (N = 201), (logarithmic value -4.13 [IQR -4.50 to -3.84] vs -4.00 [4.32 to -3.74], p=0.02). In ad hoc analysis pre-transplant FOXP3fl levels were similar in both groups. The association between FOXP3fl and declining eGFR was confirmed by multivariable analysis adjusted for potential confounding factors (Odds Ratio 0.51, 95% confidence interval 0.28 to 0.91: p=0.02). When stratifying FOXP3fl levels into quartiles, recipients with lower day1 FOXP3fl had the highest rate of declining eGFR (p=0.04). Conclusion Low FOXP3fl splice variant levels at the first postoperative day in kidney transplant recipients were associated with severe decline of eGFR, a well-known surrogate for hard endpoints.
Collapse
Affiliation(s)
- Qais W. Saleh
- Department of Nephrology, Odense University Hospital, Odense, Denmark
- Cardiovascular and Renal Research, Department of Molecular Medicine, University of Southern Denmark, Odense, Denmark
| | - Afsaneh Mohammadnejad
- Epidemiology, Biostatistics and Biodemography, Department of Public Health, University of Southern Denmark, Odense, Denmark
| | - Martin Tepel
- Department of Nephrology, Odense University Hospital, Odense, Denmark
- Cardiovascular and Renal Research, Department of Molecular Medicine, University of Southern Denmark, Odense, Denmark
| |
Collapse
|
4
|
Wang Y, Fang S, Zhou H. Pathogenic role of Th17 cells in autoimmune thyroid disease and their underlying mechanisms. Best Pract Res Clin Endocrinol Metab 2023; 37:101743. [PMID: 36841747 DOI: 10.1016/j.beem.2023.101743] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/11/2023]
Abstract
Autoimmune thyroid disease, encompassing Graves' disease and Hashimoto's thyroiditis, has a very complex etiology. Pathogenesis of the disease involves both genetic susceptibility and environmental triggers. Traditionally, imbalance of T helper cell 1 and 2 was thought to result in the immune disorders in Graves' disease and Hashimoto's thyroiditis. However, increasing evidence recently revealed the important role of T helper 17 cell and its relative cellular and secretory components in the pathogenesis and progression of autoimmune thyroid disease. This review is aimed to summarize the published studies on the involvement of T helper 17 cell in autoimmune thyroid disease and discuss the underlying regulatory mechanisms, which could possibly serve as the foundation of discovering new therapeutic targets.
Collapse
Affiliation(s)
- Yi Wang
- Department of Ophthalmology, Shanghai Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, Shanghai 200011, China; Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai 200011, China; Chinese Consortium for Thyroid Eye Disease (CCTED), China; Department of Immunology and Microbiology, Shanghai Institute of Immunology, Shanghai JiaoTong University School of Medicine, Shanghai 200025, China.
| | - Sijie Fang
- Department of Ophthalmology, Shanghai Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, Shanghai 200011, China; Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai 200011, China; Chinese Consortium for Thyroid Eye Disease (CCTED), China; Department of Immunology and Microbiology, Shanghai Institute of Immunology, Shanghai JiaoTong University School of Medicine, Shanghai 200025, China.
| | - Huifang Zhou
- Department of Ophthalmology, Shanghai Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, Shanghai 200011, China; Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai 200011, China; Chinese Consortium for Thyroid Eye Disease (CCTED), China.
| |
Collapse
|
5
|
Du J, Wang Q, Yang S, Chen S, Fu Y, Spath S, Domeier P, Hagin D, Anover-Sombke S, Haouili M, Liu S, Wan J, Han L, Liu J, Yang L, Sangani N, Li Y, Lu X, Janga SC, Kaplan MH, Torgerson TR, Ziegler SF, Zhou B. FOXP3 exon 2 controls T reg stability and autoimmunity. Sci Immunol 2022; 7:eabo5407. [PMID: 35749515 PMCID: PMC9333337 DOI: 10.1126/sciimmunol.abo5407] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Differing from the mouse Foxp3 gene that encodes only one protein product, human FOXP3 encodes two major isoforms through alternative splicing-a longer isoform (FOXP3 FL) containing all the coding exons and a shorter isoform lacking the amino acids encoded by exon 2 (FOXP3 ΔE2). The two isoforms are naturally expressed in humans, yet their differences in controlling regulatory T cell phenotype and functionality remain unclear. In this study, we show that patients expressing only the shorter isoform fail to maintain self-tolerance and develop immunodeficiency, polyendocrinopathy, and enteropathy X-linked (IPEX) syndrome. Mice with Foxp3 exon 2 deletion have excessive follicular helper T (TFH) and germinal center B (GC B) cell responses, and develop systemic autoimmune disease with anti-dsDNA and antinuclear autoantibody production, as well as immune complex glomerulonephritis. Despite having normal suppressive function in in vitro assays, regulatory T cells expressing FOXP3 ΔE2 are unstable and sufficient to induce autoimmunity when transferred into Tcrb-deficient mice. Mechanistically, the FOXP3 ΔE2 isoform allows increased expression of selected cytokines, but decreased expression of a set of positive regulators of Foxp3 without altered binding to these gene loci. These findings uncover indispensable functions of the FOXP3 exon 2 region, highlighting a role in regulating a transcriptional program that maintains Treg stability and immune homeostasis.
Collapse
Affiliation(s)
- Jianguang Du
- Department of Pediatrics, HB Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Qun Wang
- Department of Pediatrics, HB Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Shuangshuang Yang
- Department of Pediatrics, HB Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN 46202, USA
- Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Si Chen
- Department of Pediatrics, HB Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN 46202, USA
- Department of Immunology, Shenzhen University School of Medicine, Shenzhen 518060, China
| | - Yongyao Fu
- Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Sabine Spath
- Center for Fundamental Immunology, Benaroya Research Institute, Seattle, WA 98101, USA
| | - Phillip Domeier
- Center for Fundamental Immunology, Benaroya Research Institute, Seattle, WA 98101, USA
| | - David Hagin
- Allen Institute for Immunology, Seattle, WA and secondary affiliation as University of Washington, Seattle, WA 98109; Department of Pediatrics, University of Washington; Center for Immunity and Immunotherapies, Seattle Children’s Hospital Research Institute, Seattle, WA 98101, USA
| | - Stephanie Anover-Sombke
- Allen Institute for Immunology, Seattle, WA and secondary affiliation as University of Washington, Seattle, WA 98109; Department of Pediatrics, University of Washington; Center for Immunity and Immunotherapies, Seattle Children’s Hospital Research Institute, Seattle, WA 98101, USA
| | - Maya Haouili
- Department of Pediatrics, HB Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Sheng Liu
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Jun Wan
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Lei Han
- Department of Pediatrics, HB Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Juli Liu
- Department of Pediatrics, HB Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Lei Yang
- Department of Pediatrics, HB Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Neel Sangani
- Department of BioHealth Informatics, School of Informatics and Computing, Indiana University–Purdue University Indianapolis; Center for Computational Biology and Bioinformatics, Indiana University School of Medicine, Indianapolis, Indiana, 46202, USA
| | - Yujing Li
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Xiongbin Lu
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Sarath Chandra Janga
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN 46202, USA
- Department of BioHealth Informatics, School of Informatics and Computing, Indiana University–Purdue University Indianapolis; Center for Computational Biology and Bioinformatics, Indiana University School of Medicine, Indianapolis, Indiana, 46202, USA
| | - Mark H. Kaplan
- Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Troy R. Torgerson
- Allen Institute for Immunology, Seattle, WA and secondary affiliation as University of Washington, Seattle, WA 98109; Department of Pediatrics, University of Washington; Center for Immunity and Immunotherapies, Seattle Children’s Hospital Research Institute, Seattle, WA 98101, USA
| | - Steven F. Ziegler
- Center for Fundamental Immunology, Benaroya Research Institute, Seattle, WA 98101, USA
| | - Baohua Zhou
- Department of Pediatrics, HB Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN 46202, USA
- Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| |
Collapse
|
6
|
Sato Y, Liu J, Lee E, Perriman R, Roncarolo MG, Bacchetta R. Co-Expression of FOXP3FL and FOXP3Δ2 Isoforms Is Required for Optimal Treg-Like Cell Phenotypes and Suppressive Function. Front Immunol 2021; 12:752394. [PMID: 34737751 PMCID: PMC8560788 DOI: 10.3389/fimmu.2021.752394] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2021] [Accepted: 09/21/2021] [Indexed: 11/13/2022] Open
Abstract
FOXP3 is the master transcription factor in both murine and human FOXP3+ regulatory T cells (Tregs), a T-cell subset with a central role in controlling immune responses. Loss of the functional Foxp3 protein in scurfy mice leads to acute early-onset lethal lymphoproliferation. Similarly, pathogenic FOXP3 mutations in humans lead to immunodysregulation, polyendocrinopathy, enteropathy, and X-linked (IPEX) syndrome, which are characterized by systemic autoimmunity that typically begins in the first year of life. However, although pathogenic FOXP3 mutations lead to overlapping phenotypic consequences in both systems, FOXP3 in human Tregs, but not mouse, is expressed as two predominant isoforms, the full length (FOXP3FL) and the alternatively spliced isoform, delta 2 (FOXP3Δ2). Here, using CRISPR/Cas9 to generate FOXP3 knockout CD4+ T cells (FOXP3KOGFP CD4+ T cells), we restore the expression of each isoform by lentiviral gene transfer to delineate their functional roles in human Tregs. When compared to FOXP3FL or FOXP3Δ2 alone, or double transduction of the same isoform, co-expression of FOXP3FL and FOXP3Δ2 induced the highest overall FOXP3 protein expression in FOXP3KOGFP CD4+ T cells. This condition, in turn, led to optimal acquisition of Treg-like cell phenotypes including downregulation of cytokines, such as IL-17, and increased suppressive function. Our data confirm that co-expression of FOXP3FL and FOXP3Δ2 leads to optimal Treg-like cell function and supports the need to maintain the expression of both when engineering therapeutics designed to restore FOXP3 function in otherwise deficient cells.
Collapse
Affiliation(s)
- Yohei Sato
- Department of Pediatrics, Division of Hematology, Oncology, Stem Cell Transplantation and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA, United States
| | - Jessica Liu
- Department of Pediatrics, Division of Hematology, Oncology, Stem Cell Transplantation and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA, United States
| | - Esmond Lee
- Department of Pediatrics, Division of Hematology, Oncology, Stem Cell Transplantation and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA, United States.,Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA, United States
| | - Rhonda Perriman
- Department of Pediatrics, Division of Hematology, Oncology, Stem Cell Transplantation and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA, United States
| | - Maria Grazia Roncarolo
- Department of Pediatrics, Division of Hematology, Oncology, Stem Cell Transplantation and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA, United States.,Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA, United States.,Center for Definitive and Curative Medicine (CDCM), Stanford University School of Medicine, Stanford, CA, United States
| | - Rosa Bacchetta
- Department of Pediatrics, Division of Hematology, Oncology, Stem Cell Transplantation and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA, United States.,Center for Definitive and Curative Medicine (CDCM), Stanford University School of Medicine, Stanford, CA, United States
| |
Collapse
|
7
|
Ibáñez-Costa A, Perez-Sanchez C, Patiño-Trives AM, Luque-Tevar M, Font P, Arias de la Rosa I, Roman-Rodriguez C, Abalos-Aguilera MC, Conde C, Gonzalez A, Pedraza-Arevalo S, Del Rio-Moreno M, Blazquez-Encinas R, Segui P, Calvo J, Ortega Castro R, Escudero-Contreras A, Barbarroja N, Aguirre MA, Castaño JP, Luque RM, Collantes-Estevez E, Lopez-Pedrera C. Splicing machinery is impaired in rheumatoid arthritis, associated with disease activity and modulated by anti-TNF therapy. Ann Rheum Dis 2021; 81:56-67. [PMID: 34625402 PMCID: PMC8762032 DOI: 10.1136/annrheumdis-2021-220308] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2021] [Accepted: 08/18/2021] [Indexed: 12/29/2022]
Abstract
OBJECTIVES To characterise splicing machinery (SM) alterations in leucocytes of patients with rheumatoid arthritis (RA), and to assess its influence on their clinical profile and therapeutic response. METHODS Leucocyte subtypes from 129 patients with RA and 29 healthy donors (HD) were purified, and 45 selected SM elements (SME) were evaluated by quantitative PCR-array based on microfluidic technology (Fluidigm). Modulation by anti-tumour necrosis factor (TNF) therapy and underlying regulatory mechanisms were assessed. RESULTS An altered expression of several SME was found in RA leucocytes. Eight elements (SNRNP70, SNRNP200, U2AF2, RNU4ATAC, RBM3, RBM17, KHDRBS1 and SRSF10) were equally altered in all leucocytes subtypes. Logistic regressions revealed that this signature might: discriminate RA and HD, and anti-citrullinated protein antibodies (ACPAs) positivity; classify high-disease activity (disease activity score-28 (DAS28) >5.1); recognise radiological involvement; and identify patients showing atheroma plaques. Furthermore, this signature was altered in RA synovial fluid and ankle joints of K/BxN-arthritic mice. An available RNA-seq data set enabled to validate data and identified distinctive splicing events and splicing variants among patients with RA expressing high and low SME levels. 3 and 6 months anti-TNF therapy reversed their expression in parallel to the reduction of the inflammatory profile. In vitro, ACPAs modulated SME, at least partially, by Fc Receptor (FcR)-dependent mechanisms. Key inflammatory cytokines further altered SME. Lastly, induced SNRNP70-overexpression and KHDRBS1-overexpression reversed inflammation in lymphocytes, NETosis in neutrophils and adhesion in RA monocytes and influenced activity of RA synovial fibroblasts. CONCLUSIONS Overall, we have characterised for the first time a signature comprising eight dysregulated SME in RA leucocytes from both peripheral blood and synovial fluid, linked to disease pathophysiology, modulated by ACPAs and reversed by anti-TNF therapy.
Collapse
Affiliation(s)
- Alejandro Ibáñez-Costa
- Rheumatology Service, Maimonides Institute of Biomedical Research of Cordoba (IMIBIC),Reina Sofia University Hospital, University of Córdoba, Cordoba, Spain
| | - Carlos Perez-Sanchez
- Rheumatology Service, Maimonides Institute of Biomedical Research of Cordoba (IMIBIC),Reina Sofia University Hospital, University of Córdoba, Cordoba, Spain
| | - Alejandra María Patiño-Trives
- Rheumatology Service, Maimonides Institute of Biomedical Research of Cordoba (IMIBIC),Reina Sofia University Hospital, University of Córdoba, Cordoba, Spain
| | - Maria Luque-Tevar
- Rheumatology Service, Maimonides Institute of Biomedical Research of Cordoba (IMIBIC),Reina Sofia University Hospital, University of Córdoba, Cordoba, Spain
| | - Pilar Font
- Rheumatology Service, Maimonides Institute of Biomedical Research of Cordoba (IMIBIC),Reina Sofia University Hospital, University of Córdoba, Cordoba, Spain
| | - Ivan Arias de la Rosa
- Rheumatology Service, Maimonides Institute of Biomedical Research of Cordoba (IMIBIC),Reina Sofia University Hospital, University of Córdoba, Cordoba, Spain
| | - Cristobal Roman-Rodriguez
- Rheumatology Service, Maimonides Institute of Biomedical Research of Cordoba (IMIBIC),Reina Sofia University Hospital, University of Córdoba, Cordoba, Spain
| | - Mª Carmen Abalos-Aguilera
- Rheumatology Service, Maimonides Institute of Biomedical Research of Cordoba (IMIBIC),Reina Sofia University Hospital, University of Córdoba, Cordoba, Spain
| | - Carmen Conde
- Laboratorio de Investigación 8, Instituto de Investigación Sanitaria (IDIS), Hospital Clinico de Santiago (CHUS), Santiago de Compostela, Spain
| | - Antonio Gonzalez
- Experimental and Observational Rheumatology, Hospital Clinico Universitario de Santiago, Santiago de Compostela, Spain
| | - Sergio Pedraza-Arevalo
- Department of Cell Biology, Physiology and Immunology, Maimonides Institute of Biomedical Research of Cordoba (IMIBIC),Reina Sofia University Hospital, University of Córdoba and CIBER Fisiopatologia de la Obesidad y Nutricion (CIBERobn), Cordoba, Spain
| | - Mercedes Del Rio-Moreno
- Department of Cell Biology, Physiology and Immunology, Maimonides Institute of Biomedical Research of Cordoba (IMIBIC),Reina Sofia University Hospital, University of Córdoba and CIBER Fisiopatologia de la Obesidad y Nutricion (CIBERobn), Cordoba, Spain
| | - Ricardo Blazquez-Encinas
- Department of Cell Biology, Physiology and Immunology, Maimonides Institute of Biomedical Research of Cordoba (IMIBIC),Reina Sofia University Hospital, University of Córdoba and CIBER Fisiopatologia de la Obesidad y Nutricion (CIBERobn), Cordoba, Spain
| | - Pedro Segui
- Radiology Service, Maimonides Institute of Biomedical Research of Cordoba (IMIBIC),Reina Sofia University Hospital, University of Córdoba, Cordoba, Spain
| | - Jerusalem Calvo
- Rheumatology Service, Maimonides Institute of Biomedical Research of Cordoba (IMIBIC),Reina Sofia University Hospital, University of Córdoba, Cordoba, Spain
| | - Rafaela Ortega Castro
- Rheumatology Service, Maimonides Institute of Biomedical Research of Cordoba (IMIBIC),Reina Sofia University Hospital, University of Córdoba, Cordoba, Spain
| | - Alejandro Escudero-Contreras
- Rheumatology Service, Maimonides Institute of Biomedical Research of Cordoba (IMIBIC),Reina Sofia University Hospital, University of Córdoba, Cordoba, Spain
| | - Nuria Barbarroja
- Rheumatology Service, Maimonides Institute of Biomedical Research of Cordoba (IMIBIC),Reina Sofia University Hospital, University of Córdoba, Cordoba, Spain
| | - Mª Angeles Aguirre
- Rheumatology Service, Maimonides Institute of Biomedical Research of Cordoba (IMIBIC),Reina Sofia University Hospital, University of Córdoba, Cordoba, Spain
| | - Justo P Castaño
- Department of Cell Biology, Physiology and Immunology, Maimonides Institute of Biomedical Research of Cordoba (IMIBIC),Reina Sofia University Hospital, University of Córdoba and CIBER Fisiopatologia de la Obesidad y Nutricion (CIBERobn), Cordoba, Spain
| | - Raul M Luque
- Department of Cell Biology, Physiology and Immunology, Maimonides Institute of Biomedical Research of Cordoba (IMIBIC),Reina Sofia University Hospital, University of Córdoba and CIBER Fisiopatologia de la Obesidad y Nutricion (CIBERobn), Cordoba, Spain
| | - Eduardo Collantes-Estevez
- Rheumatology Service, Maimonides Institute of Biomedical Research of Cordoba (IMIBIC),Reina Sofia University Hospital, University of Córdoba, Cordoba, Spain
| | - Chary Lopez-Pedrera
- Rheumatology Service, Maimonides Institute of Biomedical Research of Cordoba (IMIBIC),Reina Sofia University Hospital, University of Córdoba, Cordoba, Spain
| |
Collapse
|
8
|
Ren P, Lu L, Cai S, Chen J, Lin W, Han F. Alternative Splicing: A New Cause and Potential Therapeutic Target in Autoimmune Disease. Front Immunol 2021; 12:713540. [PMID: 34484216 PMCID: PMC8416054 DOI: 10.3389/fimmu.2021.713540] [Citation(s) in RCA: 51] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2021] [Accepted: 07/29/2021] [Indexed: 11/13/2022] Open
Abstract
Alternative splicing (AS) is a complex coordinated transcriptional regulatory mechanism. It affects nearly 95% of all protein-coding genes and occurs in nearly all human organs. Aberrant alternative splicing can lead to various neurological diseases and cancers and is responsible for aging, infection, inflammation, immune and metabolic disorders, and so on. Though aberrant alternative splicing events and their regulatory mechanisms are widely recognized, the association between autoimmune disease and alternative splicing has not been extensively examined. Autoimmune diseases are characterized by the loss of tolerance of the immune system towards self-antigens and organ-specific or systemic inflammation and subsequent tissue damage. In the present review, we summarized the most recent reports on splicing events that occur in the immunopathogenesis of systemic lupus erythematosus (SLE) and rheumatoid arthritis (RA) and attempted to clarify the role that splicing events play in regulating autoimmune disease progression. We also identified the changes that occur in splicing factor expression. The foregoing information might improve our understanding of autoimmune diseases and help develop new diagnostic and therapeutic tools for them.
Collapse
Affiliation(s)
- Pingping Ren
- Kidney Disease Center, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Key Laboratory of Nephropathy, Zhejiang Province, Hangzhou, China
- Institute of Nephropathy, Zhejiang University, Hangzhou, China
| | - Luying Lu
- Kidney Disease Center, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Key Laboratory of Nephropathy, Zhejiang Province, Hangzhou, China
- Institute of Nephropathy, Zhejiang University, Hangzhou, China
| | - Shasha Cai
- Kidney Disease Center, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Department of Nephrology, The First People’s Hospital of Wenling, Taizhou, China
| | - Jianghua Chen
- Kidney Disease Center, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Key Laboratory of Nephropathy, Zhejiang Province, Hangzhou, China
- Institute of Nephropathy, Zhejiang University, Hangzhou, China
| | - Weiqiang Lin
- Kidney Disease Center, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Key Laboratory of Nephropathy, Zhejiang Province, Hangzhou, China
- Institute of Nephropathy, Zhejiang University, Hangzhou, China
- Institute of Translational Medicine, Zhejiang University of Medicine, Hangzhou, China
| | - Fei Han
- Kidney Disease Center, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Key Laboratory of Nephropathy, Zhejiang Province, Hangzhou, China
- Institute of Nephropathy, Zhejiang University, Hangzhou, China
| |
Collapse
|
9
|
Aravilli RK, Vikram SL, Kohila V. The Functional Impact of Alternative Splicing and Single Nucleotide Polymorphisms in Rheumatoid Arthritis. Curr Pharm Biotechnol 2021; 22:1014-1029. [PMID: 33001009 DOI: 10.2174/1389201021666201001142416] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2020] [Revised: 09/05/2020] [Accepted: 09/08/2020] [Indexed: 11/22/2022]
Abstract
Advances in genomics and proteomics aid the identification of genes associated with various diseases. Genome-Wide Association Studies (GWAS) have identified multiple loci as risk alleles for susceptibility to Rheumatoid Arthritis (RA). A bisection of RA risk can be attributed to genetic factors. Over 100 associated genetic loci that encompass immune regulatory factors have been found to be linked with RA. Aberrant Single Nucleotide Polymorphisms (SNPs) and alternative splicing mechanisms in such loci induce RA. These aberrations are viewed as potential therapeutic targets due to their association with a multitude of diseases. This review presents a few imperious genes whose alterations can cause severe bone deformities culminating in RA.
Collapse
Affiliation(s)
- R Kowshik Aravilli
- Department of Biotechnology, National Institute of Technology Warangal, Warangal, India
| | - S Laveen Vikram
- Department of Computer Science and Engineering, Alagappa University, Karaikudi, India
| | - V Kohila
- Department of Biotechnology, National Institute of Technology Warangal, Warangal, India
| |
Collapse
|
10
|
Liu Y, Jarjour W, Olsen N, Zheng SG. Traitor or warrior-Treg cells sneaking into the lesions of psoriatic arthritis. Clin Immunol 2020; 215:108425. [PMID: 32305454 DOI: 10.1016/j.clim.2020.108425] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2020] [Revised: 04/14/2020] [Accepted: 04/14/2020] [Indexed: 01/01/2023]
Abstract
Regulatory T (Treg) cells have been recognized to maintain immune tolerance, which contributes to prevention of autoimmune diseases. However, recent evidence has demonstrated different characteristics of these cells between those that are in circulation compared to those in various local tissues. In addition, the ability of Treg cells to have plasticity in certain disease settings and in inflammatory lesions has been increasingly recognized. Herein we summarize updated knowledge of Treg biology and discuss the current understanding of tissue-resident Treg cells in psoriatic arthritis (PsA), attempting to provide new insights into precise role of Treg cells in the immune response and as a possible therapeutic intervention in patients with PsA.
Collapse
Affiliation(s)
- Yan Liu
- Institute of Clinical Immunology, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou 510620, China
| | - Wael Jarjour
- Department of Internal Medicine, The Ohio State University College of Medicine, Wexner Medical Center, Columbus 43210, USA
| | - Nancy Olsen
- Department of Medicine, The Penn State Hershey College of Medicine, Hershey 17031, USA
| | - Song Guo Zheng
- Department of Internal Medicine, The Ohio State University College of Medicine, Wexner Medical Center, Columbus 43210, USA.
| |
Collapse
|
11
|
Dekkema GJ, Bijma T, Jellema PG, Van Den Berg A, Kroesen BJ, Stegeman CA, Heeringa P, Abdulahad WH, Sanders JS. Increased miR-142-3p Expression Might Explain Reduced Regulatory T Cell Function in Granulomatosis With Polyangiitis. Front Immunol 2019; 10:2170. [PMID: 31572380 PMCID: PMC6751284 DOI: 10.3389/fimmu.2019.02170] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2019] [Accepted: 08/28/2019] [Indexed: 12/16/2022] Open
Abstract
Objectives: Regulatory T cells (Tregs) are frequently functionally impaired in patients with granulomatosis with polyangiitis (GPA). However, the mechanism underlying their impaired function is unknown. Here, we hypothesized that Treg dysfunction in GPA is due to altered microRNA (miRNA) expression. Methods: RNA isolated from FACS-sorted memory (M) Tregs (CD4+CD45RO+CD25+CD127-) of 8 healthy controls (HCs) and 8 GPA patients without treatment was subjected to miRNA microarray analysis. Five differentially expressed miRNAs were validated in a larger cohort by reverse transcriptase quantitative polymerase chain reaction (RT-qPCR). An miRNA target gene database search revealed targets that were tested with RT-qPCR in MTregs from patients and HCs. cAMP levels were measured using flow cytometry. Results: Microarray analysis revealed 19 differentially expressed miRNAs, of which miR-142-3p was confirmed to be significantly upregulated in MTregs from GPA patients compared to those from HCs (1.9-fold, p = 0.03). In vitro overexpression of miR-142-3p lowered the suppressive capacity of MTregs (2.1-fold, p = 0.03), and miR-142-3p expression correlated negatively with the suppressive capacity (rho = -0.446, p = 0.04). Overexpression of miR-142-3p significantly decreased cAMP levels (p = 0.02) and tended to decrease the mRNA levels of a predicted target gene, adenylate cyclase 9 (ADCY9; p = 0.06). In comparison to those from HCs, MTregs from GPA patients had lower ADCY9 mRNA levels (2-fold, p = 0.008) and produced significantly less cAMP after stimulation. Importantly, induction of cAMP production in miR-142-3p overexpressed MTregs by forskolin restored their suppressive function in vitro. Conclusion: Overexpression of miR-142-3p in MTregs from GPA patients might cause functional impairment by targeting ADCY9, which leads to the suppression of cAMP production.
Collapse
Affiliation(s)
- Gerjan J Dekkema
- Department of Pathology and Medical Biology, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| | - Theo Bijma
- Department of Pathology and Medical Biology, University Medical Center Groningen, University of Groningen, Groningen, Netherlands.,Department of Internal Medicine, Division of Nephrology, University Medical Center Groningen, Groningen, Netherlands
| | - Pytrick G Jellema
- Department of Pathology and Medical Biology, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| | - Anke Van Den Berg
- Department of Pathology and Medical Biology, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| | - Bart-Jan Kroesen
- Medical Immunology Laboratory, Department of Laboratory Medicine, University Medical Center Groningen, Groningen, Netherlands
| | - Coen A Stegeman
- Department of Internal Medicine, Division of Nephrology, University Medical Center Groningen, Groningen, Netherlands
| | - Peter Heeringa
- Department of Pathology and Medical Biology, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| | - Wayel H Abdulahad
- Department of Pathology and Medical Biology, University Medical Center Groningen, University of Groningen, Groningen, Netherlands.,Department of Rheumatology and Clinical Immunology, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| | - Jan-Stephan Sanders
- Department of Internal Medicine, Division of Nephrology, University Medical Center Groningen, Groningen, Netherlands
| |
Collapse
|
12
|
Preventing psoriatic arthritis: focusing on patients with psoriasis at increased risk of transition. Nat Rev Rheumatol 2019; 15:153-166. [DOI: 10.1038/s41584-019-0175-0] [Citation(s) in RCA: 131] [Impact Index Per Article: 21.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
|
13
|
Abstract
FOXP3 is the lineage-defining transcription factor of CD4+ CD25+ regulatory T cells. While many aspects of its regulation, interaction, and function are conserved among species, alternatively spliced FOXP3 isoforms are expressed only in human cells. This review summarizes current knowledge about alternative splicing of FOXP3 and the specific functions of FOXP3 isoforms in health and disease. Future perspectives in research and the therapeutic potential of manipulating alternative splicing of FOXP3 are discussed.
Collapse
Affiliation(s)
- Reiner K W Mailer
- Institute of Clinical Chemistry and Laboratory Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany.,Cardiovascular Medicine Unit, Department of Medicine, Karolinska Insititutet, Stockholm, Sweden
| |
Collapse
|
14
|
Bliddal S, Nielsen CH, Feldt-Rasmussen U. Recent advances in understanding autoimmune thyroid disease: the tallest tree in the forest of polyautoimmunity. F1000Res 2017; 6:1776. [PMID: 29043075 PMCID: PMC5621109 DOI: 10.12688/f1000research.11535.1] [Citation(s) in RCA: 63] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 09/29/2017] [Indexed: 12/17/2022] Open
Abstract
Autoimmune thyroid disease (AITD) is often observed together with other autoimmune diseases. The coexistence of two or more autoimmune diseases in the same patient is referred to as polyautoimmunity, and AITD is the autoimmune disease most frequently involved. The occurrence of polyautoimmunity has led to the hypothesis that the affected patients suffer from a generalized dysregulation of their immune system. The present review summarizes recent discoveries unravelling the immunological mechanisms involved in autoimmunity, ranging from natural autoimmunity to disease-specific autoimmunity. Furthermore, the clinical grounds for considering AITD in a setting of polyautoimmunity are explored. A better understanding of these may pave the way for designing new treatment modalities targeting the underlying immune dysregulation when AITD appears in the context of polyautoimmunity.
Collapse
Affiliation(s)
- Sofie Bliddal
- Department of Medical Endocrinology, Section 2132, Copenhagen University Hospital (Rigshospitalet), Blegdamsvej 9, 2100 Copenhagen, Denmark
| | - Claus Henrik Nielsen
- Institute for Inflammation Research, Center for Rheumatology and Spine Diseases, section 7521, Copenhagen University Hospital (Rigshospitalet), Blegdamsvej 9, 2100 Copenhagen, Denmark
| | - Ulla Feldt-Rasmussen
- Department of Medical Endocrinology, Section 2132, Copenhagen University Hospital (Rigshospitalet), Blegdamsvej 9, 2100 Copenhagen, Denmark
| |
Collapse
|
15
|
Abstract
The proper restraint of the destructive potential of the immune system is essential for maintaining health. Regulatory T (Treg) cells ensure immune homeostasis through their defining ability to suppress the activation and function of other leukocytes. The expression of the transcription factor forkhead box protein P3 (FOXP3) is a well-recognized characteristic of Treg cells, and FOXP3 is centrally involved in the establishment and maintenance of the Treg cell phenotype. In this Review, we summarize how the expression and activity of FOXP3 are regulated across multiple layers by diverse factors. The therapeutic implications of these topics for cancer and autoimmunity are also discussed.
Collapse
|
16
|
Szentpetery A, Heffernan E, Gogarty M, Mellerick L, McCormack J, Haroon M, Elmamoun M, Gallagher P, Kelly G, Fabre A, Kirby B, FitzGerald O. Abatacept reduces synovial regulatory T-cell expression in patients with psoriatic arthritis. Arthritis Res Ther 2017; 19:158. [PMID: 28679449 PMCID: PMC5498994 DOI: 10.1186/s13075-017-1364-3] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2016] [Accepted: 06/09/2017] [Indexed: 12/17/2022] Open
Abstract
Background The aim was to study changes in immunohistochemical expression markers of synovial and skin inflammation, clinical outcomes and magnetic resonance imaging (MRI) scores with abatacept treatment in patients with psoriatic arthritis (PsA). Methods Biological-treatment-naïve PsA patients with active disease including synovitis of a knee were enrolled in this single-centre, crossover study. Patients were randomised to receive intravenous abatacept 3 mg/kg of body weight or placebo infusion on day 1, 15 and 29; thereafter abatacept 10 mg/kg of body weight was administered every 28 days for 5 months. Clinical data were collected at each visit. Synovial biopsy of the involved knee was obtained at baseline and 2 and 6 months. MRI of the same knee and skin biopsy was performed prior to arthroscopy. Results Fifteen patients were recruited. Significant improvements in the joint-related measures were observed; 90% were European League Against Rheumatism criteria responders and 30% achieved psoriasis area severity index (PASI)50 at 6 months. Reduction in synovitis (P = 0.016) and vascularity (P = 0.039) macroscopic scores consistent with decrease in total MRI score (P = 0.016) were noticed. Abatacept decreased the immunohistological expression of FOXP3+ cells (P = 0.027), specifically the expression of CD4+FOXP3+ regulatory T cells (Tregs) (P = 0.008) in the synovium over 6 months. There was no significant clinical or immunohistological change in any of the skin measures. Conclusion This is the first study assessing synovial and psoriatic skin immunpathological changes following abatacept treatment in PsA. Reduction in Treg expression in the synovium but not in the psoriatic lesion suggests abnormal Treg function in PsA with differential suppressive capacity in the synovium compared to the lesional skin. The results of this study demonstrate that abatacept 10 mg/kg of body weight might be an effective treatment option for joint disease in patients with PsA. Trial registration Irish Health Products Regulatory Authority. Trial registration number: CT 900/489/1 – Abatacept (case number: 2077284, EudraCT Number: 2009-017525-19, Protocol number: 77777). Registered on 12 March 2010.
Collapse
Affiliation(s)
- Agnes Szentpetery
- Department of Rheumatology, St. Vincent's University Hospital, Dublin, Ireland
| | - Eric Heffernan
- Department of Radiology, St. Vincent's University Hospital, Dublin, Ireland
| | | | - Lisa Mellerick
- Department of Histopathology, St. Vincent's University Hospital, Dublin, Ireland
| | - Janet McCormack
- Research Pathology, Immunohistochemistry Core Facility, UCD Conway Institute, UCD School of Medicine, Dublin, Ireland
| | - Muhammad Haroon
- Department of Rheumatology, St. Vincent's University Hospital, Dublin, Ireland
| | - Musaab Elmamoun
- Department of Rheumatology, St. Vincent's University Hospital, Dublin, Ireland
| | - Phil Gallagher
- Department of Rheumatology, St. Vincent's University Hospital, Dublin, Ireland
| | - Genevieve Kelly
- Department of Dermatology, St. Vincent's University Hospital, Dublin, Ireland
| | - Aurelie Fabre
- Department of Histopathology, St. Vincent's University Hospital, Dublin, Ireland.,Research Pathology, Immunohistochemistry Core Facility, UCD Conway Institute, UCD School of Medicine, Dublin, Ireland
| | - Brian Kirby
- Department of Dermatology, St. Vincent's University Hospital, Dublin, Ireland
| | - Oliver FitzGerald
- Department of Rheumatology, St. Vincent's University Hospital, Dublin, Ireland.
| |
Collapse
|
17
|
Rojas J, Teran-Angel G, Barbosa L, Peterson DL, Berrueta L, Salmen S. Activation-dependent mitochondrial translocation of Foxp3 in human hepatocytes. Exp Cell Res 2016; 343:159-167. [PMID: 27068374 DOI: 10.1016/j.yexcr.2016.04.008] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2015] [Revised: 03/24/2016] [Accepted: 04/07/2016] [Indexed: 12/28/2022]
Abstract
Foxp3 is considered to be the master regulator for the development and function of regulatory T cells (Treg). Recently Foxp3, has been detected in extra lymphoid tissue, and in hepatocytes and has been associated with hepatocellular carcinoma (HCC), although its role has not been defined. Since it is expected that there is a relationship between protein localization, activity and cellular function, the aim of this study was to explore the subcellular localization of Foxp3 in resting and stimulated human hepatocytes. Foxp3 expression was measured by flow cytometry, subcellular fractioning, and immunofluorescence, and this data was used to track the shuttling of Foxp3 in different subcellular compartments in hepatocytes (HepG2 cell line), stimulated by using the PKC activators (PMA), core and preS1/2 antigen from hepatitis B virus (HBV). Our data shows that besides the nuclear location, mitochondrial translocation was detected after stimulation with PMA and at to a lesser extent, with preS1/2. In addition, Foxp3 is localizes at outer mitochondrial membrane. These results suggest a non-canonical role of Foxp3 in the mitochondrial compartment in human hepatocytes, and opens a new field about their role in liver damages during HBV infection.
Collapse
Affiliation(s)
- Joselyn Rojas
- Instituto de Inmunología Clínica, Facultad de Medicina, Universidad de Los Andes, Merida, Venezuela
| | - Guillermo Teran-Angel
- Instituto de Inmunología Clínica, Facultad de Medicina, Universidad de Los Andes, Merida, Venezuela
| | - Luisa Barbosa
- Instituto de Inmunología Clínica, Facultad de Medicina, Universidad de Los Andes, Merida, Venezuela
| | - Darrell L Peterson
- Department of Biochemistry and Molecular Biology, Virginia Commonwealth University, Richmond, VA, USA
| | - Lisbeth Berrueta
- Instituto de Inmunología Clínica, Facultad de Medicina, Universidad de Los Andes, Merida, Venezuela; Division of Preventive Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA.
| | - Siham Salmen
- Instituto de Inmunología Clínica, Facultad de Medicina, Universidad de Los Andes, Merida, Venezuela.
| |
Collapse
|
18
|
Karczewski J, Dobrowolska A, Rychlewska-Hańczewska A, Adamski Z. New insights into the role of T cells in pathogenesis of psoriasis and psoriatic arthritis. Autoimmunity 2016; 49:435-450. [DOI: 10.3109/08916934.2016.1166214] [Citation(s) in RCA: 68] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
|
19
|
Fiocco U, Martini V, Accordi B, Caso F, Costa L, Oliviero F, Scanu A, Facco M, Boso D, Gatto M, Felicetti M, Frallonardo P, Ramonda R, Piva L, Zambello R, Agostini C, Scarpa R, Basso G, Semenzato G, Dayer JM, Punzi L, Doria A. Transcriptional network profile on synovial fluid T cells in psoriatic arthritis. Clin Rheumatol 2015; 34:1571-80. [DOI: 10.1007/s10067-015-3002-2] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2015] [Revised: 06/09/2015] [Accepted: 06/28/2015] [Indexed: 11/28/2022]
|
20
|
Julià A, Pinto JA, Gratacós J, Queiró R, Ferrándiz C, Fonseca E, Montilla C, Torre-Alonso JC, Puig L, Pérez Venegas JJ, Fernández Nebro A, Fernández E, Muñoz-Fernández S, Daudén E, González C, Roig D, Sánchez Carazo JL, Zarco P, Erra A, López Estebaranz JL, Rodríguez J, Ramírez DM, de la Cueva P, Vanaclocha F, Herrera E, Castañeda S, Rubio E, Salvador G, Díaz-Torné C, Blanco R, Willisch Domínguez A, Mosquera JA, Vela P, Tornero J, Sánchez-Fernández S, Corominas H, Ramírez J, López-Lasanta M, Tortosa R, Palau N, Alonso A, García-Montero AC, Gelpí JL, Codó L, Day K, Absher D, Myers RM, Cañete JD, Marsal S. A deletion at ADAMTS9-MAGI1 locus is associated with psoriatic arthritis risk. Ann Rheum Dis 2015; 74:1875-81. [PMID: 25990289 DOI: 10.1136/annrheumdis-2014-207190] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2014] [Accepted: 04/23/2015] [Indexed: 12/11/2022]
Abstract
OBJECTIVE Copy number variants (CNVs) have been associated with the risk to develop multiple autoimmune diseases. Our objective was to identify CNVs associated with the risk to develop psoriatic arthritis (PsA) using a genome-wide analysis approach. METHODS A total of 835 patients with PsA and 1498 healthy controls were genotyped for CNVs using the Illumina HumanHap610 BeadChip genotyping platform. Genomic CNVs were characterised using CNstream analysis software and analysed for association using the χ(2) test. The most significant genomic CNV associations with PsA risk were independently tested in a validation sample of 1133 patients with PsA and 1831 healthy controls. In order to test for the specificity of the variants with PsA aetiology, we also analysed the association to a cohort of 822 patients with purely cutaneous psoriasis (PsC). RESULTS A total of 165 common CNVs were identified in the genome-wide analysis. We found a highly significant association of an intergenic deletion between ADAMTS9 and MAGI1 genes on chromosome 3p14.1 (p=0.00014). Using the independent patient and control cohort, we validated the association between ADAMTS9-MAGI1 deletion and PsA risk (p=0.032). Using next-generation sequencing, we characterised the 26 kb associated deletion. Finally, analysing the PsC cohort we found a lower frequency of the deletion compared with the PsA cohort (p=0.0088) and a similar frequency to that of healthy controls (p>0.3). CONCLUSIONS The present genome-wide scan for CNVs associated with PsA risk has identified a new deletion associated with disease risk and which is also differential from PsC risk.
Collapse
Affiliation(s)
- Antonio Julià
- Rheumatology Research Group, Vall d'Hebron Research Institute, Barcelona, Spain
| | - José Antonio Pinto
- Rheumatology Department, Complejo Hospitalario Juan Canalejo, A Coruña, Spain
| | - Jordi Gratacós
- Rheumatology Department, Hospital Parc Taulí, Sabadell, Barcelona, Spain
| | - Rubén Queiró
- Rheumatology Department, Hospital Universitario Central de Asturias, Oviedo, Spain
| | - Carlos Ferrándiz
- Dermatology Department, Hospital Universitari Germans Trias i Pujol, Badalona, Barcelona, Spain
| | - Eduardo Fonseca
- Dermatology Department, Complejo Hospitalario Universitario de A Coruña, A Coruña, Spain
| | - Carlos Montilla
- Rheumatology Department, Hospital Virgen de la Vega, Salamanca, Spain
| | | | - Lluís Puig
- Dermatology Department, Hospital de la Santa Creu i Sant Pau, Barcelona, Spain
| | | | - Antonio Fernández Nebro
- UGC Reumatología, Instituto de Investigación Biomédica de Málaga (IBIMA), Hospital Regional Universitario de Málaga, Málaga, Spain
| | - Emilia Fernández
- Department of Dermatology, Hospital Universitario de Salamanca, Salamanca, Spain
| | | | - Esteban Daudén
- Dermatology Department, Hospital Universitario La Princesa, Madrid, Spain
| | - Carlos González
- Rheumatology Department, Hospital Universitario Gregorio Marañón, Madrid, Spain
| | - Daniel Roig
- Rheumatology Service, Hospital Moisès Broggi, Barcelona, Spain
| | | | - Pedro Zarco
- Rheumatology Department, Hospital Universitario Fundación Alcorcón, Madrid, Spain
| | - Alba Erra
- Rheumatology Department, Hospital Sant Rafael, Barcelona, Spain
| | | | - Jesús Rodríguez
- Rheumatology Department, Hospital Universitari de Bellvitge, Barcelona, Spain
| | | | - Pablo de la Cueva
- Department of Dermatology, Hospital Universitario Infanta Leonor, Madrid, Spain
| | | | - Enrique Herrera
- Dermatology Department, Hospital Universitario Virgen de la Victoria, Málaga, Spain
| | - Santos Castañeda
- Rheumatology Department, Hospital Universitario de La Princesa, IIS-Princesa, Madrid, Spain
| | - Esteban Rubio
- Rheumatology Department, Centro de Salud Virgen de los Reyes, Sevilla, Spain
| | - Georgina Salvador
- Rheumatology Department, Hospital Mútua de Terrassa, Terrassa, Spain
| | - César Díaz-Torné
- Rheumatology Unit, Hospital de la Santa Creu i Sant Pau, Barcelona, Spain
| | - Ricardo Blanco
- Rheumatology Department, Hospital Universitario Marqués de Valdecilla, Santander, Spain
| | | | - José Antonio Mosquera
- Rheumatology Department, Complejo Hospitalario Hospital Provincial de Pontevedra, Pontevedra, Spain
| | - Paloma Vela
- Rheumatology Department, Hospital General Universitario de Alicante, Alicante, Spain
| | - Jesús Tornero
- Rheumatology Department, Hospital Universitario Guadalajara, Guadalajara, Spain
| | | | | | - Julio Ramírez
- Rheumatology Department, Hospital Clínic de Barcelona and IDIBAPS, Barcelona, Spain
| | - María López-Lasanta
- Rheumatology Research Group, Vall d'Hebron Research Institute, Barcelona, Spain
| | - Raül Tortosa
- Rheumatology Research Group, Vall d'Hebron Research Institute, Barcelona, Spain
| | - Nuria Palau
- Rheumatology Research Group, Vall d'Hebron Research Institute, Barcelona, Spain
| | - Arnald Alonso
- Rheumatology Research Group, Vall d'Hebron Research Institute, Barcelona, Spain
| | | | - Josep Lluís Gelpí
- Life Sciences, Barcelona Supercomputing Centre, National Institute of Bioinformatics, Barcelona, Spain
| | - Laia Codó
- HudsonAlpha Institute for Biotechnology, Alabama, USA
| | - Kenneth Day
- HudsonAlpha Institute for Biotechnology, Alabama, USA
| | - Devin Absher
- HudsonAlpha Institute for Biotechnology, Alabama, USA
| | | | - Juan D Cañete
- Rheumatology Department, Hospital Clínic de Barcelona and IDIBAPS, Barcelona, Spain
| | - Sara Marsal
- Rheumatology Research Group, Vall d'Hebron Research Institute, Barcelona, Spain
| |
Collapse
|
21
|
Kristensen B, Hegedüs L, Madsen HO, Smith TJ, Nielsen CH. Altered balance between self-reactive T helper (Th)17 cells and Th10 cells and between full-length forkhead box protein 3 (FoxP3) and FoxP3 splice variants in Hashimoto's thyroiditis. Clin Exp Immunol 2015; 180:58-69. [PMID: 25412700 PMCID: PMC4367094 DOI: 10.1111/cei.12557] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/18/2014] [Indexed: 02/06/2023] Open
Abstract
T helper type 17 (Th17) cells play a pathogenic role in autoimmune disease, while interleukin (IL)-10-producing Th10 cells serve a protective role. The balance between the two subsets is regulated by the local cytokine milieu and by the relative expression of intact forkhead box protein 3 (FoxP3) compared to FoxP3Δ2, missing exon 2. Th17 and Th10 cell differentiation has usually been studied using polyclonal stimuli, and little is known about the ability of physiologically relevant self-antigens to induce Th17 or Th10 cell differentiation in autoimmune thyroid disease. We subjected mononuclear cells from healthy donors and patients with Hashimoto's thyroiditis (HT) or Graves' disease (GD) to polyclonal stimulation, or stimulation with human thyroglobulin (TG), human thyroid peroxidase (TPO), or Esherichia coli lipopolysaccharide (LPS). TPO and LPS induced increased differentiation of naive CD4(+) CD45RA(+) CD45R0(-) T cells from HT patients into Th17 cells. Th10 cell proportions were decreased in HT after polyclonal stimulation, but were comparable to those of healthy donors after antigen-specific stimulation. Taken together, our data show that an increased Th17 : Th10 ratio was found in HT patients after stimulation with thyroid-specific self-antigens. We also observed an elevated baseline production of IL-6 and transforming growth factor (TGF)-β1 and of mRNA encoding FoxP3Δ2 rather than intact FoxP3. This may contribute to the skewing towards Th17 cell responses in HT.
Collapse
Affiliation(s)
- B Kristensen
- Institute for Inflammation Research, Department of Infectious Diseases and Rheumatology, Copenhagen University Hospital RigshospitaletCopenhagen, Denmark
- Department of Endocrinology and Metabolism, Odense University HospitalOdense, Denmark
| | - L Hegedüs
- Department of Endocrinology and Metabolism, Odense University HospitalOdense, Denmark
| | - H O Madsen
- The Tissue Typing Laboratory, Department of Clinical Immunology, Copenhagen University Hospital RigshospitaletCopenhagen, Denmark
| | - T J Smith
- Departments of Ophthalmology and Visual Sciences, Kellogg Eye Center, and Internal Medicine, University of Michigan Medical SchoolAnn Arbor, MI, USA
| | - C H Nielsen
- Institute for Inflammation Research, Department of Infectious Diseases and Rheumatology, Copenhagen University Hospital RigshospitaletCopenhagen, Denmark
| |
Collapse
|
22
|
Fan X, Tang L. Aberrant and alternative splicing in skeletal system disease. Gene 2013; 528:21-6. [PMID: 23800666 DOI: 10.1016/j.gene.2013.06.027] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2012] [Revised: 05/24/2013] [Accepted: 06/08/2013] [Indexed: 11/19/2022]
Abstract
The main function of skeletal system is to support the body and help movement. A variety of factors can lead to skeletal system disease, including age, exercise, and of course genetic makeup and expression. Pre-mRNA splicing plays a crucial role in gene expression, by creating multiple protein variants with different biological functions. The recent studies show that several skeletal system diseases are related to pre-mRNA splicing. This review focuses on the relationship between pre-mRNA splicing and skeletal system disease. On the one hand, splice site mutation that leads to aberrant splicing often causes genetic skeletal system disease, like COL1A1, SEDL and LRP5. On the other hand, alternative splicing without genomic mutation may generate some marker protein isoforms, for example, FN, VEGF and CD44. Therefore, understanding the relationship between pre-mRNA splicing and skeletal system disease will aid in uncovering the mechanism of disease and contribute to the future development of gene therapy.
Collapse
Affiliation(s)
- Xin Fan
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing 400044, China
| | | |
Collapse
|