1
|
Tobar CGR, Urmendiz YDMM, Vallejo MA, Manquillo DF, Castaño VEN, Caicedo AIO, Tobar LLM, Vargas JAG, Cuellar RAD. Immunomodulatory effect of Tityus sp. in mononuclear cells extracted from the blood of rheumatoid arthritis patients. J Venom Anim Toxins Incl Trop Dis 2024; 30:e20230064. [PMID: 39445068 PMCID: PMC11498904 DOI: 10.1590/1678-9199-jvatitd-2023-0064] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2023] [Accepted: 06/10/2024] [Indexed: 10/25/2024] Open
Abstract
Background Pathophysiological mechanisms of rheumatoid arthritis arise because of a proinflammatory environment, generated by the interaction of autoreactive lymphocytes and proinflammatory mediators. Current strategies to mitigate the progression of the disease produce adverse effects, so there is a need for new therapeutic strategies and molecular targets to treat this disease. In this context, evidence suggests that scorpion venoms could modulate the immune response and some important cellular mechanisms of pharmacological interest. To evaluate the immunomodulatory effect of the venom of Tityus sp. (a possible new species close to Tityus metuendus) peripheral blood mononuclear cells of women diagnosed with RA were compared to cells of a control group. Methods A case-control study was conducted with a sample of 10 women with a confirmed diagnosis of RA and controls matched by sex and age. The cytotoxicity of the venom was evaluated to find sublethal concentrations of the venom, and subsequently, their immunomodulatory capacity in terms of percentage of proliferation, cell activation, and cytokines production. Results the venom of Tityus sp. produced a decrease in the percentage of proliferation in the CD3+, CD3+CD4+, and CD3+CD8+ cell subpopulations of RA patients and healthy controls, at concentrations of 252 and 126 µg/mL. However, the venom did not induce significant differences in the percentage of cell activation markers. The venom caused a decrease in IL-10 at a concentration of 252 µg/mL compared to untreated cells from patients and controls. The remaining cytokines did not show significant differences. Conclusion the venom of Tityus sp. is a potential source of molecules with immunomodulatory ability in CD4 and CD8 T lymphocytes. This result directs venom characterization studies to identify pharmacological targets with immunomodulatory capacity in T lymphocytes to enhance research in the treatment of autoimmune disorders such as RA.
Collapse
Affiliation(s)
- Cindy Gabriela Rivera Tobar
- Grupo de Investigación en Inmunología y Enfermedades Infecciosas,
Programa de Medicina, Departamento de Patología, Facultad de Ciencias de la Salud,
Universidad del Cauca, Popayán, Colombia
| | - Yisel del Mar Morales Urmendiz
- Grupo de Investigación en Inmunología y Enfermedades Infecciosas,
Programa de Medicina, Departamento de Patología, Facultad de Ciencias de la Salud,
Universidad del Cauca, Popayán, Colombia
| | - Marcela Alejandra Vallejo
- Grupo de Investigación en Inmunología y Enfermedades Infecciosas,
Programa de Medicina, Departamento de Patología, Facultad de Ciencias de la Salud,
Universidad del Cauca, Popayán, Colombia
| | - Diego Felipe Manquillo
- Grupo de Investigación en Inmunología y Enfermedades Infecciosas,
Programa de Medicina, Departamento de Patología, Facultad de Ciencias de la Salud,
Universidad del Cauca, Popayán, Colombia
| | - Victoria Eugenia Niño Castaño
- Grupo de Investigación en Inmunología y Enfermedades Infecciosas,
Programa de Medicina, Departamento de Patología, Facultad de Ciencias de la Salud,
Universidad del Cauca, Popayán, Colombia
| | - Ana Isabel Ospina Caicedo
- Grupo de Investigación en Salud, Programa de Medicina, Departamento
de Medicina Interna, Facultad de Ciencias de la Salud, Universidad del Cauca,
Popayán, Colombia
| | - Leydy Lorena Mendoza Tobar
- Grupo de Investigaciones Herpetológicas y Toxinológicas (GIHT),
Departamento de Biología, Facultad de Ciencias Naturales, Exactas y de la Educación,
Universidad del Cauca, Popayán, Colombia
| | - Jimmy Alexander Guerrero Vargas
- Grupo de Investigaciones Herpetológicas y Toxinológicas (GIHT),
Departamento de Biología, Facultad de Ciencias Naturales, Exactas y de la Educación,
Universidad del Cauca, Popayán, Colombia
- Grupo de Investigaciones Herpetológicas y Toxinológicas (GIHT),
Centro de Investigaciones Biomédicas - Bioterio (CIBUC-Bioterio), Museo de Historia
Natural, Departamento de Biología, Facultad de Ciencias Naturales, Exactas y de la
Educación, Universidad del Cauca, Popayán, Colombia
| | - Rosa Amalia Dueñas Cuellar
- Grupo de Investigación en Inmunología y Enfermedades Infecciosas,
Programa de Medicina, Departamento de Patología, Facultad de Ciencias de la Salud,
Universidad del Cauca, Popayán, Colombia
| |
Collapse
|
2
|
Yasumizu Y, Takeuchi D, Morimoto R, Takeshima Y, Okuno T, Kinoshita M, Morita T, Kato Y, Wang M, Motooka D, Okuzaki D, Nakamura Y, Mikami N, Arai M, Zhang X, Kumanogoh A, Mochizuki H, Ohkura N, Sakaguchi S. Single-cell transcriptome landscape of circulating CD4 + T cell populations in autoimmune diseases. CELL GENOMICS 2024; 4:100473. [PMID: 38359792 PMCID: PMC10879034 DOI: 10.1016/j.xgen.2023.100473] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/09/2023] [Revised: 09/07/2023] [Accepted: 12/05/2023] [Indexed: 02/17/2024]
Abstract
CD4+ T cells are key mediators of various autoimmune diseases; however, their role in disease progression remains unclear due to cellular heterogeneity. Here, we evaluated CD4+ T cell subpopulations using decomposition-based transcriptome characterization and canonical clustering strategies. This approach identified 12 independent gene programs governing whole CD4+ T cell heterogeneity, which can explain the ambiguity of canonical clustering. In addition, we performed a meta-analysis using public single-cell datasets of over 1.8 million peripheral CD4+ T cells from 953 individuals by projecting cells onto the reference and cataloging cell frequency and qualitative alterations of the populations in 20 diseases. The analyses revealed that the 12 transcriptional programs were useful in characterizing each autoimmune disease and predicting its clinical status. Moreover, genetic variants associated with autoimmune diseases showed disease-specific enrichment within the 12 gene programs. The results collectively provide a landscape of single-cell transcriptomes of CD4+ T cell subpopulations involved in autoimmune disease.
Collapse
Affiliation(s)
- Yoshiaki Yasumizu
- Department of Experimental Immunology, Immunology Frontier Research Center, Osaka University, Osaka, Japan; Department of Neurology, Graduate School of Medicine, Osaka University, Osaka, Japan; Integrated Frontier Research for Medical Science Division, Institute for Open and Transdisciplinary Research Initiatives (OTRI), Osaka University, Osaka, Japan
| | - Daiki Takeuchi
- Department of Experimental Immunology, Immunology Frontier Research Center, Osaka University, Osaka, Japan; Faculty of Medicine, Osaka University, Osaka, Japan
| | - Reo Morimoto
- Department of Experimental Immunology, Immunology Frontier Research Center, Osaka University, Osaka, Japan
| | - Yusuke Takeshima
- Department of Experimental Immunology, Immunology Frontier Research Center, Osaka University, Osaka, Japan
| | - Tatsusada Okuno
- Department of Neurology, Graduate School of Medicine, Osaka University, Osaka, Japan
| | - Makoto Kinoshita
- Department of Neurology, Graduate School of Medicine, Osaka University, Osaka, Japan
| | - Takayoshi Morita
- Department of Respiratory Medicine and Clinical Immunology, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Yasuhiro Kato
- Department of Respiratory Medicine and Clinical Immunology, Osaka University Graduate School of Medicine, Osaka, Japan; Department of Immunopathology, Immunology Frontier Research Center, Osaka University, Osaka, Japan
| | - Min Wang
- Clinical Immunology Center, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China; Department of Rheumatology, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing, China
| | - Daisuke Motooka
- Integrated Frontier Research for Medical Science Division, Institute for Open and Transdisciplinary Research Initiatives (OTRI), Osaka University, Osaka, Japan; Genome Information Research Center, Research Institute for Microbial Diseases, Osaka University, Osaka, Japan
| | - Daisuke Okuzaki
- Integrated Frontier Research for Medical Science Division, Institute for Open and Transdisciplinary Research Initiatives (OTRI), Osaka University, Osaka, Japan; Genome Information Research Center, Research Institute for Microbial Diseases, Osaka University, Osaka, Japan
| | - Yamami Nakamura
- Department of Experimental Immunology, Immunology Frontier Research Center, Osaka University, Osaka, Japan
| | - Norihisa Mikami
- Department of Experimental Immunology, Immunology Frontier Research Center, Osaka University, Osaka, Japan
| | - Masaya Arai
- Department of Experimental Immunology, Immunology Frontier Research Center, Osaka University, Osaka, Japan
| | - Xuan Zhang
- Department of Rheumatology, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing, China
| | - Atsushi Kumanogoh
- Integrated Frontier Research for Medical Science Division, Institute for Open and Transdisciplinary Research Initiatives (OTRI), Osaka University, Osaka, Japan; Department of Respiratory Medicine and Clinical Immunology, Osaka University Graduate School of Medicine, Osaka, Japan; Department of Immunopathology, Immunology Frontier Research Center, Osaka University, Osaka, Japan; Center for Infectious Diseases for Education and Research, Osaka University, Osaka, Japan
| | - Hideki Mochizuki
- Department of Neurology, Graduate School of Medicine, Osaka University, Osaka, Japan; Integrated Frontier Research for Medical Science Division, Institute for Open and Transdisciplinary Research Initiatives (OTRI), Osaka University, Osaka, Japan
| | - Naganari Ohkura
- Department of Experimental Immunology, Immunology Frontier Research Center, Osaka University, Osaka, Japan; Department of Frontier Research in Tumor Immunology, Graduate School of Medicine, Osaka University, Osaka, Japan.
| | - Shimon Sakaguchi
- Department of Experimental Immunology, Immunology Frontier Research Center, Osaka University, Osaka, Japan; Department of Experimental Immunology, Institute for Life and Medical Sciences, Kyoto University, Kyoto, Japan.
| |
Collapse
|
3
|
Identification of proteinase 3 autoreactive CD4 +T cells and their T-cell receptor repertoires in antineutrophil cytoplasmic antibody-associated vasculitis. Kidney Int 2023; 103:973-985. [PMID: 36804380 DOI: 10.1016/j.kint.2023.01.023] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2022] [Revised: 01/10/2023] [Accepted: 01/19/2023] [Indexed: 02/17/2023]
Abstract
Antineutrophil cytoplasmic antibody (ANCA)-associated vasculitis (AAV) is an autoimmune disease involving autoreactivity to proteinase 3 (PR3) as demonstrated by presence of ANCAs. While autoantibodies are screened for diagnosis, autoreactive T cells and their features are less well-studied. Here, we investigated PR3-specific CD4+T cell responses and features of autoreactive T cells in patients with PR3-AAV, using a cohort of 72 patients with either active or inactive disease. Autoreactive PR3-specific CD4+T cells producing interferon γ in response to protein stimulation were found to express the G-protein coupled receptor 56 (GPR56), a cell surface marker that distinguishes T cells with cytotoxic capacity. GPR56+CD4+T cells were significantly more prominent in the blood of patients with inactive as compared to active disease, suggesting that these cells were affected by immunosuppression and/or that they migrated from the circulation to sites of organ involvement. Indeed, GPR56+CD4+T cells were identified in T-cell infiltrates of affected kidneys and an association with immunosuppressive therapy was found. Moreover, distinct TCR gene segment usage and shared (public) T cell clones were found for the PR3-reactive TCRs. Shared T cell clones were found in different patients with AAV carrying the disease-associated HLA-DP allele, demonstrating convergence of the autoreactive T cell repertoire. Thus, we identified a CD4+T cell signature in blood and in affected kidneys that display PR3 autoreactivity and associates with T cell cytotoxicity. Our data provide a basis for novel rationales for both immune monitoring and future therapeutic intervention in PR3-AAV.
Collapse
|
4
|
Single cell sequencing identifies clonally expanded synovial CD4 + T PH cells expressing GPR56 in rheumatoid arthritis. Nat Commun 2022; 13:4046. [PMID: 35831277 PMCID: PMC9279430 DOI: 10.1038/s41467-022-31519-6] [Citation(s) in RCA: 68] [Impact Index Per Article: 22.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2021] [Accepted: 06/01/2022] [Indexed: 11/23/2022] Open
Abstract
Rheumatoid arthritis (RA) is an autoimmune disease affecting synovial joints where different CD4+ T cell subsets may contribute to pathology. Here, we perform single cell sequencing on synovial CD4+ T cells from anti-citrullinated protein antibodies (ACPA)+ and ACPA- RA patients and identify two peripheral helper T cell (TPH) states and a cytotoxic CD4+ T cell subset. We show that the adhesion G-protein coupled receptor 56 (GPR56) delineates synovial CXCL13high TPH CD4+ T cells expressing LAG-3 and the tissue-resident memory receptors CXCR6 and CD69. In ACPA- SF, TPH cells display lower levels of GPR56 and LAG-3. Further, most expanded T cell clones in the joint are within CXCL13high TPH CD4+ T cells. Finally, RNA-velocity analyses suggest a common differentiation pathway between the two TPH clusters and effector CD4+ T cells. Our study provides comprehensive immunoprofiling of the synovial CD4+ T cell subsets in ACPA+ and ACPA- RA. Antibodies against citrullinated proteins (ACPA) can divide rheumatoid arthritis patients but the difference in immune phenotype is not clear. Here the authors characterise T peripheral helper cells in arthritic joints comparing ACPA+ and ACPA- patients showing changes in immune cell phenotype and surface molecules.
Collapse
|
5
|
Bediaga NG, Garnham AL, Naselli G, Bandala-Sanchez E, Stone NL, Cobb J, Harbison JE, Wentworth JM, Ziegler AG, Couper JJ, Smyth GK, Harrison LC. Cytotoxicity-Related Gene Expression and Chromatin Accessibility Define a Subset of CD4+ T Cells That Mark Progression to Type 1 Diabetes. Diabetes 2022; 71:566-577. [PMID: 35007320 PMCID: PMC8893947 DOI: 10.2337/db21-0612] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/08/2021] [Accepted: 12/12/2021] [Indexed: 11/13/2022]
Abstract
Type 1 diabetes in children is heralded by a preclinical phase defined by circulating autoantibodies to pancreatic islet antigens. How islet autoimmunity is initiated and then progresses to clinical diabetes remains poorly understood. Only one study has reported gene expression in specific immune cells of children at risk associated with progression to islet autoimmunity. We analyzed gene expression with RNA sequencing in CD4+ and CD8+ T cells, natural killer (NK) cells, and B cells, and chromatin accessibility by assay for transposase-accessible chromatin sequencing (ATAC-seq) in CD4+ T cells, in five genetically at risk children with islet autoantibodies who progressed to diabetes over a median of 3 years ("progressors") compared with five children matched for sex, age, and HLA-DR who had not progressed ("nonprogressors"). In progressors, differentially expressed genes (DEGs) were largely confined to CD4+ T cells and enriched for cytotoxicity-related genes/pathways. Several top-ranked DEGs were validated in a semi-independent cohort of 13 progressors and 11 nonprogressors. Flow cytometry confirmed that progression was associated with expansion of CD4+ cells with a cytotoxic phenotype. By ATAC-seq, progression was associated with reconfiguration of regulatory chromatin regions in CD4+ cells, some linked to differentially expressed cytotoxicity-related genes. Our findings suggest that cytotoxic CD4+ T cells play a role in promoting progression to type 1 diabetes.
Collapse
Affiliation(s)
- Naiara G. Bediaga
- The Walter and Eliza Hall Institute of Medical Research, Parkville, Australia
- Department of Medical Biology, The University of Melbourne, Parkville, Australia
| | - Alexandra L. Garnham
- The Walter and Eliza Hall Institute of Medical Research, Parkville, Australia
- Department of Medical Biology, The University of Melbourne, Parkville, Australia
| | - Gaetano Naselli
- The Walter and Eliza Hall Institute of Medical Research, Parkville, Australia
| | - Esther Bandala-Sanchez
- The Walter and Eliza Hall Institute of Medical Research, Parkville, Australia
- Department of Medical Biology, The University of Melbourne, Parkville, Australia
| | - Natalie L. Stone
- The Walter and Eliza Hall Institute of Medical Research, Parkville, Australia
| | - Joanna Cobb
- Murdoch Children’s Research Institute, Parkville, Australia
| | - Jessica E. Harbison
- Department of Endocrinology and Diabetes, Women’s and Children’s Hospital, North Adelaide, Australia
- Robinson Research Institute, The University of Adelaide, Adelaide, Australia
| | - John M. Wentworth
- The Walter and Eliza Hall Institute of Medical Research, Parkville, Australia
- Department of Medical Biology, The University of Melbourne, Parkville, Australia
- Department of Diabetes and Endocrinology, Royal Melbourne Hospital, Parkville, Australia
| | - Annette-G. Ziegler
- Institute of Diabetes Research, Helmholtz Zentrum München, Neuherberg, Germany
| | - Jennifer J. Couper
- Department of Endocrinology and Diabetes, Women’s and Children’s Hospital, North Adelaide, Australia
- Robinson Research Institute, The University of Adelaide, Adelaide, Australia
| | - Gordon K. Smyth
- The Walter and Eliza Hall Institute of Medical Research, Parkville, Australia
- School of Mathematics and Statistics, The University of Melbourne, Parkville, Australia
| | - Leonard C. Harrison
- The Walter and Eliza Hall Institute of Medical Research, Parkville, Australia
- Department of Medical Biology, The University of Melbourne, Parkville, Australia
- Corresponding author: Leonard C. Harrison,
| |
Collapse
|
6
|
Explanation of Structure and Function of kv1.3 Potent Blocker From Mesobuthus eupeus Venom Gland: A New Promise in Drug Development. Jundishapur J Nat Pharm Prod 2022. [DOI: 10.5812/jjnpp.120271] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
Background: Scorpions and other venomous animals are sought with great concern because venom is a source of novel peptides with exciting features. Some toxins of scorpion venom are effectors of potassium channels. Previous studies strongly support the importance of potassium channel toxins for use as pharmacological tools or potential drugs. Objectives: Here, a three-dimensional (3-D) structure and function of a potent acidic blocker of the human voltage-gated potassium ion channel, Kv1.3, previously identified in the scorpion Mesobuthus eupeus venom gland, were interpreted. Methods: The 3-D structure of meuK2-2 was generated using homology modeling. The interaction of meuK2-2 with the Kv1.3 channel was evaluated using a computational protocol employing peptide-protein docking experiments, pose clustering, and 100 ns molecular dynamic simulations to make the 3-D models of the meuK2-2/Kv1.3 complex trustworthy. Results: A CSα/β (cysteine-stabilized α-helical and β-sheet) fold was found for the 3-D structure of meuK2-2. In a different mechanism from what was identified so far, meuK2-2 binds to both turret and pore loop of Kv1.3 through two key residues (Ala28 and Ser11) and H-bonds. The binding of meuK2-2 induces some conformational changes to Kv1.3. Eventually, the side chain of a positively charged amino acid (His9) occupies the channel's pore. All together blocks the ion permeation pathway. Conclusions: MeuK2-2 could block Kv1.3 by a new mechanism. So, it could be a unique target for further investigations to develop a pharmacological tool and potential drug.
Collapse
|
7
|
Kosmaczewska A, Ciszak L, Stosio M, Szteblich A, Madej M, Frydecka I, Wiland P, Szmyrka M. CD4 +CD28 null T cells are expanded in moderately active systemic lupus erythematosus and secrete pro-inflammatory interferon gamma, depending on the Disease Activity Index. Lupus 2020; 29:705-714. [PMID: 32279585 DOI: 10.1177/0961203320917749] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
BACKGROUND Pathogenic CD4+CD28null cells are characterized by inflammatory cytokine synthesis and tropism to the inflamed tissues. Recent studies showed the involvement of CD28null T cells in a severe clinical outcome of lupus. However, their role in moderately active disease is still unresolved. METHODS We examined the levels of circulating CD4+CD28null cells and CD8+CD28null suppressor T cells. We also compared the CD4+CD28null and CD4+CD28+ T-cell functional properties, including the expression of interferon gamma (IFN-γ) and Ki67 among systemic lupus erythematosus (SLE) patients (n = 20) and healthy controls (n = 20). All the patients were under immunosuppressive treatment and exhibited moderate SLE activity (median SLE Disease Activity Index (SLEDAI) = 6). RESULTS In patients, we found elevated CD4+CD28null and unchanged levels of suppressor CD8+CD28null T cells. There was no difference between patients and controls in IFN-γ and Ki67-expressing CD4+, CD4+CD28+, and CD4+CD28null T cells, except for higher IFN-γ levels in CD4+CD28+ T cells in SLE. In each studied group, we observed a higher preponderance of IFN-γ- and Ki67-expressing cells among CD4+CD28null T cells and lower levels of IFN-γ in CD4+CD28null T cells compared to the CD28+ subset. Similarly, Ki67 intensity was decreased in healthy CD4+CD28null cells, whereas in patients, comparably high expression was observed in both subsets. IFN-γ intensity in CD4+CD28null T cells correlated with SLEDAI. CONCLUSION SLE with a moderately active clinical course is characterized by peripheral blood expansion of CD4+CD28null T cells and a normal abundance of suppressor CD8+CD28null T cells. The demonstration that these pathogenic CD4+ T cells, despite the lack of CD28, maintain the ability to produce pro-inflammatory IFN-γ positively correlated with disease activity as well as relatively high proliferative capacity may suggest their potentially predictive role in SLE flares.
Collapse
Affiliation(s)
- Agata Kosmaczewska
- Department of Experimental Therapy, Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, Wroclaw, Poland
| | - Lidia Ciszak
- Department of Experimental Therapy, Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, Wroclaw, Poland
| | - Malgorzata Stosio
- Faculty of Chemistry, Wroclaw University of Science and Technology, Wroclaw, Poland
| | - Aleksandra Szteblich
- Department of Experimental Therapy, Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, Wroclaw, Poland
| | - Marta Madej
- Department of Rheumatology and Internal Diseases, Wroclaw Medical University, Wroclaw, Poland
| | - Irena Frydecka
- Department of Experimental Therapy, Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, Wroclaw, Poland
| | - Piotr Wiland
- Department of Rheumatology and Internal Diseases, Wroclaw Medical University, Wroclaw, Poland
| | - Magdalena Szmyrka
- Department of Rheumatology and Internal Diseases, Wroclaw Medical University, Wroclaw, Poland
| |
Collapse
|
8
|
Update on the Pathomechanism, Diagnosis, and Treatment Options for Rheumatoid Arthritis. Cells 2020; 9:cells9040880. [PMID: 32260219 PMCID: PMC7226834 DOI: 10.3390/cells9040880] [Citation(s) in RCA: 476] [Impact Index Per Article: 95.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2020] [Revised: 03/28/2020] [Accepted: 03/30/2020] [Indexed: 12/18/2022] Open
Abstract
Rheumatoid arthritis (RA) is an autoimmune disease that involves multiple joints bilaterally. It is characterized by an inflammation of the tendon (tenosynovitis) resulting in both cartilage destruction and bone erosion. While until the 1990s RA frequently resulted in disability, inability to work, and increased mortality, newer treatment options have made RA a manageable disease. Here, great progress has been made in the development of disease-modifying anti-rheumatic drugs (DMARDs) which target inflammation and thereby prevent further joint damage. The available DMARDs are subdivided into (1) conventional synthetic DMARDs (methotrexate, hydrochloroquine, and sulfadiazine), (2) targeted synthetic DMARDs (pan-JAK- and JAK1/2-inhibitors), and (3) biologic DMARDs (tumor necrosis factor (TNF)-α inhibitors, TNF-receptor (R) inhibitors, IL-6 inhibitors, IL-6R inhibitors, B cell depleting antibodies, and inhibitors of co-stimulatory molecules). While DMARDs have repeatedly demonstrated the potential to greatly improve disease symptoms and prevent disease progression in RA patients, they are associated with considerable side-effects and high financial costs. This review summarizes our current understanding of the underlying pathomechanism, diagnosis of RA, as well as the mode of action, clinical benefits, and side-effects of the currently available DMARDs.
Collapse
|
9
|
Bano A, Pera A, Almoukayed A, Clarke THS, Kirmani S, Davies KA, Kern F. CD28 null CD4 T-cell expansions in autoimmune disease suggest a link with cytomegalovirus infection. F1000Res 2019; 8. [PMID: 30984377 PMCID: PMC6436193 DOI: 10.12688/f1000research.17119.1] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 03/20/2019] [Indexed: 01/03/2023] Open
Abstract
Immunosenescence is thought to contribute to the increase of autoimmune diseases in older people. Immunosenescence is often associated with the presence of an expanded population of CD4 T cells lacking expression of CD28 (CD28
null). These highly cytotoxic CD4 T cells were isolated from disease-affected tissues in patients with rheumatoid arthritis, systemic lupus erythematosus, multiple sclerosis, or other chronic inflammatory diseases and their numbers appeared to be linked to disease severity. However, we recently demonstrated that the common herpes virus, cytomegalovirus (CMV), not ageing, is the major driver of this subset of cytotoxic T cells. In this review, we discuss how CMV might potentiate and exacerbate autoimmune disease through the expansion of CD28
null CD4 T cells.
Collapse
Affiliation(s)
- Aalia Bano
- Department of Clinical and Experimental medicine, Brighton and Sussex Medical School, Brighton, Sussex, BN1 9PX, UK
| | - Alejandra Pera
- Department of Immunology, Maimonides Institute for Biomedical Research (IMIBIC), Reina Sofia Hospital, University of Cordoba, Av. Menendez Pidal, 14004, Cordoba, Spain
| | - Ahmad Almoukayed
- Department of Clinical and Experimental medicine, Brighton and Sussex Medical School, Brighton, Sussex, BN1 9PX, UK
| | - Thomas H S Clarke
- Department of Clinical and Experimental medicine, Brighton and Sussex Medical School, Brighton, Sussex, BN1 9PX, UK
| | - Sukaina Kirmani
- Department of Clinical and Experimental medicine, Brighton and Sussex Medical School, Brighton, Sussex, BN1 9PX, UK
| | - Kevin A Davies
- Department of Clinical and Experimental medicine, Brighton and Sussex Medical School, Brighton, Sussex, BN1 9PX, UK
| | - Florian Kern
- Department of Clinical and Experimental medicine, Brighton and Sussex Medical School, Brighton, Sussex, BN1 9PX, UK
| |
Collapse
|
10
|
Chemin K, Gerstner C, Malmström V. Effector Functions of CD4+ T Cells at the Site of Local Autoimmune Inflammation-Lessons From Rheumatoid Arthritis. Front Immunol 2019; 10:353. [PMID: 30915067 PMCID: PMC6422991 DOI: 10.3389/fimmu.2019.00353] [Citation(s) in RCA: 161] [Impact Index Per Article: 26.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2018] [Accepted: 02/11/2019] [Indexed: 12/17/2022] Open
Abstract
Infiltration of memory CD4+ T cells in synovial joints of Rheumatoid Arthritis (RA) patients has been reported since decades. Moreover, several genome wide association studies (GWAS) pinpointing a key genetic association between the HLA-DR locus and RA have led to the generally agreed hypothesis that CD4+ T cells are directly implicated in the disease. Still, RA is a heterogeneous disease and much effort has been made to understand its different facets. T cell differentiation is driven by mechanisms including antigen stimulation, co-stimulatory signals and cytokine milieu, all of which are abundant in the rheumatic joint, implying that any T cells migrating into the joint may be further affected locally. In parallel to the characterization and classification of T-cell subsets, the contribution of different effector T cells to RA has been investigated in numerous studies though sometimes with contradictory results. In particular, the frequency of Th1 and Th17 cells has been assessed in the synovial joints with various results that could, at least partly, be explained by the stage of the disease. For regulatory T cells, it is largely accepted that they accumulate in RA synovial fluid and that the equilibrium between regulatory T cells and effector cells is a key factor in controlling inflammation processes involved in RA. Recent phenotypic studies describe the possible implication of a novel subset of peripheral T helper cells (Tph) important for T-B cell cross talk and plasma cell differentiation in the RA joint of ACPA+ (autoantibodies against citrullinated proteins) RA patients. Finally, cytotoxic CD4+ T cells, historically described as increased in the peripheral blood of RA patients have attracted new attention in the last years. In view of the recently identified peripheral T-cell subsets, we will integrate immunological data as well as information on genetic variants and therapeutic strategy outcomes into our current understanding of the width of effector T cells. We will also integrate tissue-resident memory T cell aspects, and discuss similarities and differences with inflammatory conditions in skin (psoriasis) and mucosal organs (Crohn's disease).
Collapse
Affiliation(s)
- Karine Chemin
- Division of Rheumatology, Department of Medicine, Karolinska Institute, Karolinska University Hospital Solna, Stockholm, Sweden
| | - Christina Gerstner
- Division of Rheumatology, Department of Medicine, Karolinska Institute, Karolinska University Hospital Solna, Stockholm, Sweden
| | - Vivianne Malmström
- Division of Rheumatology, Department of Medicine, Karolinska Institute, Karolinska University Hospital Solna, Stockholm, Sweden
| |
Collapse
|
11
|
Lynch KD, Keshav S, Chapman RW. The Use of Biologics in Patients with Inflammatory Bowel Disease and Primary Sclerosing Cholangitis. CURRENT HEPATOLOGY REPORTS 2019; 18:115-126. [PMID: 31008013 PMCID: PMC6445403 DOI: 10.1007/s11901-019-00456-2] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
PURPOSE OF REVIEW Biologics are well established in the treatment of many immuno-inflammatory diseases including inflammatory bowel disease (IBD). However, although primary sclerosing cholangitis (PSC) is closely associated with IBD, the role of biologics in PSC remains uncertain. Many new biologics are becoming available to treat IBD, and this review aims to use the experience of biologics in PSC so far to guide more effective evaluation of emerging therapies in the future. RECENT FINDINGS Antibodies to TNF-α were the first biologics used in IBD, and retrospective analysis suggests that they may have some benefit in PSC, even though an early randomised controlled trial (RCT) showed no effect. Mechanistic studies suggest that TNF-α may have a pathogenic role in PSC. An antibody to integrin α4β7 is effective in IBD, and there are emerging data on its effects in PSC, although no RCT data are available. Mechanistic studies suggest that interrupting the migration of lymphocytes is relevant in PSC. Two biologics, targeting vascular adhesion protein-1 (VAP-1), and lysyl oxidase-like 2 (LOXL2) have been tested in RCTs. The trial of anti-VAP1 is ongoing, whilst the anti-LOXL2 trial was negative. SUMMARY Anti-TNF antibodies may benefit PSC when used to treat concomitant IBD, and this may be a direct effect on the liver in a subgroup of patients, or may be an indirect effect of treating IBD. Similarly, anti-integrin therapy may benefit a subset of patients with IBD and PSC. RCTs could decide the role of emerging biologics in PSC, although future trials should be guided by biomarkers that could predict response to the pathway being targeted.
Collapse
Affiliation(s)
- Kate D. Lynch
- Translational Gastroenterology Unit, Nuffield Department of Medicine, Level 5, John Radcliffe Hospital, University of Oxford, Headley Way, Headington, Oxford, OX3 9DU UK
| | - Satish Keshav
- Translational Gastroenterology Unit, Nuffield Department of Medicine, Level 5, John Radcliffe Hospital, University of Oxford, Headley Way, Headington, Oxford, OX3 9DU UK
| | - Roger W. Chapman
- Translational Gastroenterology Unit, Nuffield Department of Medicine, Level 5, John Radcliffe Hospital, University of Oxford, Headley Way, Headington, Oxford, OX3 9DU UK
| |
Collapse
|
12
|
Tanner MR, Pennington MW, Chauhan SS, Laragione T, Gulko PS, Beeton C. KCa1.1 and Kv1.3 channels regulate the interactions between fibroblast-like synoviocytes and T lymphocytes during rheumatoid arthritis. Arthritis Res Ther 2019; 21:6. [PMID: 30612588 PMCID: PMC6322314 DOI: 10.1186/s13075-018-1783-9] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2018] [Accepted: 11/29/2018] [Indexed: 12/22/2022] Open
Abstract
BACKGROUND Fibroblast-like synoviocytes (FLS) and CCR7- effector memory T (TEM) cells are two of the major cell types implicated in the progression of rheumatoid arthritis (RA). In particular, FLS become highly invasive, whereas TEM cells proliferate and secrete proinflammatory cytokines, during RA. FLS and T cells may also interact and influence each other's phenotypes. Inhibition of the pathogenic phenotypes of both FLS and TEM cells can be accomplished by selectively blocking the predominant potassium channels that they upregulate during RA: KCa1.1 (BK, Slo1, MaxiK, KCNMA1) upregulated by FLS and Kv1.3 (KCNA3) upregulated by activated TEM cells. In this study, we investigated the roles of KCa1.1 and Kv1.3 in regulating the interactions between FLS and TEM cells and determined if combination therapies of KCa1.1- and Kv1.3-selective blockers are more efficacious than monotherapies in ameliorating disease in rat models of RA. METHODS We used in vitro functional assays to assess the effects of selective KCa1.1 and Kv1.3 channel inhibitors on the interactions of FLS isolated from rats with collagen-induced arthritis (CIA) with syngeneic TEM cells. We also used flow cytometric analyses to determine the effects of KCa1.1 blockers on the expression of proteins used for antigen presentation on CIA-FLS. Finally, we used the CIA and pristane-induced arthritis models to determine the efficacy of combinatorial therapies of KCa1.1 and Kv1.3 blockers in reducing disease severity compared with monotherapies. RESULTS We show that the interactions of FLS from rats with CIA and of rat TEM cells are regulated by KCa1.1 and Kv1.3. Inhibiting KCa1.1 on FLS reduces the ability of FLS to stimulate TEM cell proliferation and migration, and inhibiting Kv1.3 on TEM cells reduces TEM cells' ability to enhance FLS expression of KCa1.1 and major histocompatibility complex class II protein, as well as stimulates their invasion. Furthermore, we show that combination therapies of selective KCa1.1 and Kv1.3 blockers are more efficacious than monotherapies at reducing signs of disease in two rat models of RA. CONCLUSIONS Our results demonstrate the importance of KCa1.1 and Kv1.3 in regulating FLS and TEM cells during RA, as well as the value of combined therapies targeting both of these cell types to treat RA.
Collapse
Affiliation(s)
- Mark R. Tanner
- Department of Molecular Physiology and Biophysics, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030 USA
- Interdepartmental Graduate Program in Translational Biology and Molecular Medicine, Baylor College of Medicine, Houston, TX USA
| | - Michael W. Pennington
- Peptides International, Inc., Louisville, KY USA
- Present address: Ambiopharm, Inc., North Augusta, SC USA
| | | | - Teresina Laragione
- Division of Rheumatology, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY USA
| | - Pércio S. Gulko
- Division of Rheumatology, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY USA
| | - Christine Beeton
- Department of Molecular Physiology and Biophysics, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030 USA
- Biology of Inflammation Center, Center for Drug Discovery, Cardiovascular Research Institute, and Dan L Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX USA
| |
Collapse
|
13
|
Pieper J, Dubnovitsky A, Gerstner C, James EA, Rieck M, Kozhukh G, Tandre K, Pellegrino S, Gebe JA, Rönnblom L, Sandalova T, Kwok WW, Klareskog L, Buckner JH, Achour A, Malmström V. Memory T cells specific to citrullinated α-enolase are enriched in the rheumatic joint. J Autoimmun 2018; 92:47-56. [PMID: 29853344 DOI: 10.1016/j.jaut.2018.04.004] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2018] [Revised: 04/26/2018] [Accepted: 04/28/2018] [Indexed: 12/29/2022]
Abstract
ACPA-positive rheumatoid arthritis (RA) is associated with distinct HLA-DR alleles and immune responses to many citrullinated self-antigens. Herein we investigated the T cell epitope confined within α-enolase326-340 in the context of HLA-DRB1*04:01 and assessed the corresponding CD4+ T cells in both the circulation and in the rheumatic joint. Comparative crystallographic analyses were performed for the native and citrullinated α-enolase326-340 peptides in complex with HLA-DRB1*04:01. HLA-tetramers assembled with either the native or citrullinated peptide were used for ex vivo and in vitro assessment of α-enolase-specific T cells in peripheral blood, synovial fluid and synovial tissue by flow cytometry. The native and modified peptides take a completely conserved structural conformation within the peptide-binding cleft of HLA-DRB1*04:01. The citrulline residue-327 was located N-terminally, protruding towards TCRs. The frequencies of T cells recognizing native eno326-340 were similar in synovial fluid and peripheral blood, while in contrast, the frequency of T cells recognizing cit-eno326-340 was significantly elevated in synovial fluid compared to peripheral blood (3.6-fold, p = 0.0150). Additionally, citrulline-specific T cells with a memory phenotype were also significantly increased (1.6-fold, p = 0.0052) in synovial fluid compared to peripheral blood. The native T cell epitope confined within α-enolase326-340 does not appear to lead to complete negative selection of cognate CD4+ T cells. In RA patient samples, only T cells recognizing the citrullinated version of α-enolase326-340 were found at elevated frequencies implicating that neo-antigen formation is critical for breach of tolerance.
Collapse
Affiliation(s)
- Jennifer Pieper
- Rheumatology Unit, Department of Medicine Solna, Center for Molecular Medicine, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| | - Anatoly Dubnovitsky
- Rheumatology Unit, Department of Medicine Solna, Center for Molecular Medicine, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden; Science for Life Laboratory, Department of Medicine Solna, Karolinska Institutet, Sweden; Department of Infectious Diseases, Karolinska University Hospital, Stockholm, Sweden
| | - Christina Gerstner
- Rheumatology Unit, Department of Medicine Solna, Center for Molecular Medicine, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| | - Eddie A James
- Tetramer Core, BRI at Virginia Mason, Seattle, WA, USA
| | - Mary Rieck
- Translational Research Program, BRI at Virginia Mason, Seattle, WA, USA
| | - Genadiy Kozhukh
- Rheumatology Unit, Department of Medicine Solna, Center for Molecular Medicine, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden; Science for Life Laboratory, Department of Medicine Solna, Karolinska Institutet, Sweden; Department of Infectious Diseases, Karolinska University Hospital, Stockholm, Sweden
| | - Karolina Tandre
- Department of Medical Sciences, Rheumatology, Science for Life Laboratory, Uppsala University, Uppsala, Sweden
| | - Sara Pellegrino
- DISFARM, Dipartimento di Scienze Farmaceutiche, Sezione Chimica Generale e Organica, Università degli Studi, Milano, Italy
| | - John A Gebe
- Translational Research Program, BRI at Virginia Mason, Seattle, WA, USA
| | - Lars Rönnblom
- Department of Medical Sciences, Rheumatology, Science for Life Laboratory, Uppsala University, Uppsala, Sweden
| | - Tatyana Sandalova
- Science for Life Laboratory, Department of Medicine Solna, Karolinska Institutet, Sweden; Department of Infectious Diseases, Karolinska University Hospital, Stockholm, Sweden
| | - William W Kwok
- Translational Research Program, BRI at Virginia Mason, Seattle, WA, USA
| | - Lars Klareskog
- Rheumatology Unit, Department of Medicine Solna, Center for Molecular Medicine, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| | - Jane H Buckner
- Translational Research Program, BRI at Virginia Mason, Seattle, WA, USA
| | - Adnane Achour
- Science for Life Laboratory, Department of Medicine Solna, Karolinska Institutet, Sweden; Department of Infectious Diseases, Karolinska University Hospital, Stockholm, Sweden
| | - Vivianne Malmström
- Rheumatology Unit, Department of Medicine Solna, Center for Molecular Medicine, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden.
| |
Collapse
|
14
|
CD28neg. T lymphocytes of a melanoma patient harbor tumor immunity and a high frequency of germline-encoded and public TCRs. Immunol Res 2017; 66:79-86. [DOI: 10.1007/s12026-017-8976-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
|
15
|
Pandya JM, Venalis P, Al-Khalili L, Shahadat Hossain M, Stache V, Lundberg IE, Malmström V, Fasth AER. CD4+ and CD8+ CD28(null) T Cells Are Cytotoxic to Autologous Muscle Cells in Patients With Polymyositis. Arthritis Rheumatol 2017; 68:2016-26. [PMID: 26895511 DOI: 10.1002/art.39650] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2015] [Accepted: 02/16/2016] [Indexed: 12/12/2022]
Abstract
OBJECTIVE Inflammatory T cell infiltrates in the skeletal muscle tissue of patients with polymyositis are dominated by CD28-negative effector (CD28(null) ) T cells of both the CD4 and CD8 lineage. These cells are potentially cytotoxic, and the aim of the present study was to develop a fully autologous cell culture system in which to investigate the functional contribution of such CD28(null) T cells to myotoxicity. METHODS In vitro cocultures of autologous skeletal muscle cells and T cell subsets obtained from 5 polymyositis patients were performed. Myotoxicity of T cells was quantified by calcein release and flow cytometric analyses. T cell degranulation was blocked with concanamycin A. Specific blocking of perforin, cytokines, and HLA was performed using antibodies. RESULTS Both CD4+CD28(null) and CD8+CD28(null) T cells induced more muscle cell death than did their CD28+ counterparts. Differentiated muscle cells (myotubes) were more sensitive to T cell-mediated cell death than were their precursors (myoblasts). Both CD8+ and CD4+ CD28(null) T cells displayed perforin polarization toward muscle cells and secreted higher levels of granzyme B and interferon-γ (IFNγ) in coculture than did CD28+ T cells. The myotoxic effects of CD28(null) T cells were reduced upon the blocking of perforin, cytokines, and HLA. Addition of IFNγ or tumor necrosis factor did not induce skeletal muscle cell death in the absence of T cells; however, it did up-regulate HLA expression on muscle cells. CONCLUSION Myotoxicity of CD4+ and CD8+ CD28(null) T cells is mediated by directed perforin-dependent killing and can be further influenced by IFNγ-induced HLA expression on muscle cells. The data suggest that CD28(null) T cells are key effector cells that contribute to the muscle cell damage in polymyositis.
Collapse
Affiliation(s)
- Jayesh M Pandya
- Karolinska University Hospital Solna, Sweden, and Karolinska Institutet, Stockholm, Sweden
| | - Paulius Venalis
- Karolinska University Hospital Solna, Sweden, and Karolinska Institutet, Stockholm, Sweden
| | | | | | - Vanessa Stache
- Karolinska University Hospital Solna, Sweden, and Karolinska Institutet, Stockholm, Sweden
| | - Ingrid E Lundberg
- Karolinska University Hospital Solna, Sweden, and Karolinska Institutet, Stockholm, Sweden
| | - Vivianne Malmström
- Karolinska University Hospital Solna, Sweden, and Karolinska Institutet, Stockholm, Sweden
| | - Andreas E R Fasth
- Andreas E. R. Fasth, PhD: Karolinska lnstitutet, Schering-Plough, MSD, and Novartis, Stockholm, Sweden
| |
Collapse
|
16
|
Apoil PA, Puissant-Lubrano B, Congy-Jolivet N, Peres M, Tkaczuk J, Roubinet F, Blancher A. Influence of age, sex and HCMV-serostatus on blood lymphocyte subpopulations in healthy adults. Cell Immunol 2017; 314:42-53. [PMID: 28219652 DOI: 10.1016/j.cellimm.2017.02.001] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2016] [Revised: 02/03/2017] [Accepted: 02/04/2017] [Indexed: 12/22/2022]
Abstract
Using a standardized immunophenotyping procedure we studied thirty-eight distinct subpopulations of T, B and NK lymphocytes in 253 healthy blood donors aged from 19 to 67. We analysed the influence of age, sex and HCMV seropositivity on each lymphocyte subpopulations and established reference ranges. We observed that aging influences the largest number of lymphocyte subpopulations with a slow increase of CD8+ EMRA T lymphocytes and of the numbers of circulating Tregs. The proportion of HLA-DR+ cells among Tregs increased with age and was correlated to the proportion of HLA-DR+ cells among effector T CD4+ lymphocytes. Sex had a major impact on absolute counts of CD4+ T cells which were higher in females. HCMV-seropositivity was associated with higher frequencies of CD8+ EMRA memory T lymphocytes while a high frequency of terminally differentiated EMRA CD4+ T cells was observed in 80% of HCMV-positive individuals and in none of the HCMV seronegative individuals.
Collapse
Affiliation(s)
- P A Apoil
- Laboratoire d'Immunogénétique Moléculaire, EA 3034, Université Paul Sabatier, Toulouse 3, France; Laboratoire d'Immunologie, CHU de Toulouse, France
| | - B Puissant-Lubrano
- Laboratoire d'Immunogénétique Moléculaire, EA 3034, Université Paul Sabatier, Toulouse 3, France; Laboratoire d'Immunologie, CHU de Toulouse, France
| | - N Congy-Jolivet
- Laboratoire d'Immunogénétique Moléculaire, EA 3034, Université Paul Sabatier, Toulouse 3, France; Laboratoire d'Immunologie, CHU de Toulouse, France
| | - M Peres
- Laboratoire d'Immunologie, CHU de Toulouse, France
| | - J Tkaczuk
- Laboratoire d'Immunologie, CHU de Toulouse, France
| | - F Roubinet
- EFS Pyrénées-Méditerranée, Toulouse, France
| | - A Blancher
- Laboratoire d'Immunogénétique Moléculaire, EA 3034, Université Paul Sabatier, Toulouse 3, France; Laboratoire d'Immunologie, CHU de Toulouse, France.
| |
Collapse
|
17
|
Rational design of a Kv1.3 channel-blocking antibody as a selective immunosuppressant. Proc Natl Acad Sci U S A 2016; 113:11501-11506. [PMID: 27663736 DOI: 10.1073/pnas.1612803113] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
A variable region fusion strategy was used to generate an immunosuppressive antibody based on a novel "stalk-knob" structural motif in the ultralong complementary-determining region (CDR) of a bovine antibody. The potent Kv1.3 channel inhibitory peptides Moka1-toxin and Vm24-toxin were grafted into different CDRs of the humanized antibodies BVK and Synagis (Syn) using both β-sheet and coiled-coil linkers. Structure-activity relationship efforts led to generation of the fusion protein Syn-Vm24-CDR3L, which demonstrated excellent selectivity and potency against effector human memory T cells (subnanomolar to picomolar EC50 values). This fusion antibody also had significantly improved plasma half-life and serum stability in rodents compared with the parent Vm24 peptide. Finally, this fusion protein showed potent in vivo efficacy in the delayed type hypersensitivity in rats. These results illustrate the utility of antibody CDR fusions as a general and effective strategy to generate long-acting functional antibodies, and may lead to a selective immunosuppressive antibody for the treatment of autoimmune diseases.
Collapse
|
18
|
Chalan P, van den Berg A, Kroesen BJ, Brouwer L, Boots A. Rheumatoid Arthritis, Immunosenescence and the Hallmarks of Aging. Curr Aging Sci 2016. [PMID: 26212057 PMCID: PMC5388800 DOI: 10.2174/1874609808666150727110744] [Citation(s) in RCA: 80] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Age is the most important risk factor for the development of infectious diseases, cancer and chronic inflammatory diseases including rheumatoid arthritis (RA). The very act of living causes damage to cells. A network of molecular, cellular and physiological maintenance and repair systems creates a buffering capacity against these damages. Aging leads to progressive shrinkage of the buffering capacity and increases vulnerability. In order to better understand the complex mammalian aging processes, nine hallmarks of aging and their interrelatedness were recently put forward. RA is a chronic autoimmune disease affecting the joints. Although RA may develop at a young age, the incidence of RA increases with age. It has been suggested that RA may develop as a consequence of premature aging (immunosenescence) of the immune system. Alternatively, premature aging may be the consequence of the inflammatory state in RA. In an effort to answer this chicken and egg conundrum, we here outline and discuss the nine hallmarks of aging, their contribution to the pre-aged phenotype and the effects of treatment on the reversibility of immunosenescence in RA.
Collapse
Affiliation(s)
| | | | | | | | - Annemieke Boots
- Department of Rheumatology and Clinical Immunology, University of Groningen, University Medical Center Groningen, P.O Box 30.001, 9700 RB, Groningen, The Netherlands.
| |
Collapse
|
19
|
Winchester R, Giles JT, Nativ S, Downer K, Zhang HZ, Bag-Ozbek A, Zartoshti A, Bokhari S, Bathon JM. Association of Elevations of Specific T Cell and Monocyte Subpopulations in Rheumatoid Arthritis With Subclinical Coronary Artery Atherosclerosis. Arthritis Rheumatol 2016; 68:92-102. [PMID: 26360530 DOI: 10.1002/art.39419] [Citation(s) in RCA: 49] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2015] [Accepted: 08/27/2015] [Indexed: 01/20/2023]
Abstract
OBJECTIVE Coronary artery disease (CAD) is the leading cause of excess deaths in rheumatoid arthritis (RA). However, identification of features denoting patients with a risk of developing CAD is lacking. The composition of circulating peripheral blood mononuclear cell (PBMC) subsets in RA patients differs markedly from that in healthy controls with regard to the extent of T cell activation, with clonal expansion and differentiation to effector memory status, and presence of inflammatory monocytes. In this study, we sought to evaluate whether elevations in these PBMC subpopulations in RA patients could denote those with an increased risk of subclinical CAD, as determined by the presence of coronary artery calcification (CAC). METHODS The study cohort comprised 72 patients with RA who underwent cardiac computed tomography to assess CAC. PBMC subsets were determined by multiparameter flow cytometry. Multivariable logistic regression was used to determine the associations between PBMC subpopulations and the presence of CAC. RESULTS Among the 72 patients with RA, 33% had CAC and exhibited significant increases in the levels of circulating CD4 T cell subsets denoting activation and differentiation to the effector memory phenotypes. Analogous increases in the levels of CD8 T cell subsets, as well as in the CD14(high)CD16+ intermediate monocyte subset, were also present in these patients, as compared to those without CAC. The increases in the CD4 and CD8 T cell subsets were highly intercorrelated, whereas the increases in CD14(high)CD16+ monocytes were independent of elevations in the CD4 T cell subsets. After adjustments for relevant confounders, the levels of CD4+CD56+CD57+ T cells and CD14(high)CD16+ monocytes remained associated with the presence of CAC. CONCLUSION These findings indicate that PBMC subsets are markers for the presence of CAC and suggest that mechanisms of atherogenesis in RA may operate in part through the elevations in these subsets, raising further questions about the mechanisms underlying the presence of such alterations in cell composition in patients with RA and the potential for shared etiologic pathways between RA and cardiovascular disease.
Collapse
Affiliation(s)
- Robert Winchester
- Columbia University College of Physicians and Surgeons, New York, New York
| | - Jon T Giles
- Columbia University College of Physicians and Surgeons, New York, New York
| | - Simona Nativ
- Columbia University College of Physicians and Surgeons, New York, New York
| | - Kendall Downer
- Columbia University College of Physicians and Surgeons, New York, New York
| | - Hui-Zhu Zhang
- Columbia University College of Physicians and Surgeons, New York, New York
| | - Ayse Bag-Ozbek
- Columbia University College of Physicians and Surgeons, New York, New York
| | - Afshin Zartoshti
- Columbia University College of Physicians and Surgeons, New York, New York
| | - Sabahat Bokhari
- Columbia University College of Physicians and Surgeons, New York, New York
| | - Joan M Bathon
- Columbia University College of Physicians and Surgeons, New York, New York
| |
Collapse
|
20
|
Pandya JM, Loell I, Hossain MS, Zong M, Alexanderson H, Raghavan S, Lundberg IE, Malmström V. Effects of conventional immunosuppressive treatment on CD244+ (CD28null) and FOXP3+ T cells in the inflamed muscle of patients with polymyositis and dermatomyositis. Arthritis Res Ther 2016; 18:80. [PMID: 27039301 PMCID: PMC4818535 DOI: 10.1186/s13075-016-0974-5] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2015] [Accepted: 03/14/2016] [Indexed: 12/12/2022] Open
Abstract
Background T-cell infiltrates may persist in muscle tissue of polymyositis (PM) and dermatomyositis (DM) patients despite aggressive immunosuppressive treatment. Here, we investigated to what extent persistent T cells in affected muscle were FOXP3+, a marker for regulatory T cells (Tregs), or CD244+, a marker for CD28null T cells, and whether their presence correlated to clinical outcome. The sensitivity of CD28null T cells towards glucocorticoid and Treg-mediated immunosuppression was also investigated. Methods Muscle biopsies from 16 newly diagnosed or untreated patients with PM/DM were investigated by immunohistochemistry for expression of CD3, FOXP3 and CD244 before and after treatment with glucocorticoids and immunosuppressive agents. For clinical evaluation, serum levels of creatine kinase, muscle performance (FI and MMT8), disease activity (MITAX) and disability (HAQ) were measured. In vitro suppressive effects of glucocorticoids and Tregs on T-cell activation were measured by CD69 upregulation. Results Before treatment, CD244+ cells were present at higher proportions compared to FOXP3+ cells in the inflamed muscle. Following treatment, FOXP3+ cell numbers decreased while CD244+ cells persisted. Patients with impaired muscle function (<75 % FI) post-treatment had higher levels of CD244+ cells in the follow-up biopsy compared to those with FI >75 %. MITAX and HAQ correlated with the number of CD244+ cells post-treatment. CD4+CD28null T cells displayed lower sensitivity towards both glucocorticoid and Treg-mediated immunosuppression in vitro compared to their CD28+ counterparts. Conclusions Poor outcome in patients with myositis following immunosuppressive therapy was linked to persistence of CD244+ (CD28null) T cells in muscle tissue, suggesting their resistance against immunosuppression. A relative loss of regulatory T cells could also contribute to poor clinical outcome given their recently ascribed role in muscle tissue regeneration.
Collapse
Affiliation(s)
- Jayesh M Pandya
- Rheumatology Unit, Department of Medicine, Center for Molecular Medicine, Karolinska University Hospital, Solna, Karolinska Institutet, Stockholm, Sweden
| | - Ingela Loell
- Rheumatology Unit, Department of Medicine, Center for Molecular Medicine, Karolinska University Hospital, Solna, Karolinska Institutet, Stockholm, Sweden
| | - Mohammad Shahadat Hossain
- Rheumatology Unit, Department of Medicine, Center for Molecular Medicine, Karolinska University Hospital, Solna, Karolinska Institutet, Stockholm, Sweden
| | - Mei Zong
- Rheumatology Unit, Department of Medicine, Center for Molecular Medicine, Karolinska University Hospital, Solna, Karolinska Institutet, Stockholm, Sweden
| | - Helene Alexanderson
- Department of NVS, Division of Physiotherapy, Solna, Karolinska Institutet, Huddinge and Physiotherapy Clinic, Karolinska University Hospital, Stockholm, Sweden
| | - Sukanya Raghavan
- Rheumatology Unit, Department of Medicine, Center for Molecular Medicine, Karolinska University Hospital, Solna, Karolinska Institutet, Stockholm, Sweden.,Present address: Department of Microbiology and Immunology, Institute for Biomedicine, Gothenburg University, Gothenburg, Sweden
| | - Ingrid E Lundberg
- Rheumatology Unit, Department of Medicine, Center for Molecular Medicine, Karolinska University Hospital, Solna, Karolinska Institutet, Stockholm, Sweden
| | - Vivianne Malmström
- Rheumatology Unit, Department of Medicine, Center for Molecular Medicine, Karolinska University Hospital, Solna, Karolinska Institutet, Stockholm, Sweden.
| |
Collapse
|
21
|
Abstract
A role of genetics in primary sclerosing cholangitis (PSC) development is now firmly established. A total of 16 risk genes have been reported at highly robust ("genome-wide") significance levels, and ongoing efforts suggest that the list will ultimately be considerably longer. Importantly, this genetic risk pool so far accounts for less than 10 % of an estimated overall PSC susceptibility. The relative importance of genetic versus environmental factors (including gene-gene and gene-environment interactions) in remaining aspects of PSC pathogenesis is unknown, and other study designs than genome-wide association studies are needed to explore these aspects. For some of the loci, e.g. HLA and FUT2, distinct interacting environmental factors may exist, and working from the genetic associations may prove one valid path for determining the specific nature of environmental triggers. So far the biological implications for PSC risk genes are typically merely hypothesized based on previously published literature, and there is therefore a strong need for dedicated translational studies to determine their roles within the specific disease context of PSC. Apparently, most risk loci seem to involve in a subset of biological pathways for which genetic associations exist in a multitude of immune-mediated diseases, accounting for both inflammatory bowel disease as well as prototypical autoimmunity. In the present article, we will survey the current knowledge on PSC genetics with a particular emphasis on the pathophysiological insight potentially gained from genetic risk loci involving in this profound immunogenetic pleiotropy.
Collapse
|
22
|
Lovastatin blocks Kv1.3 channel in human T cells: a new mechanism to explain its immunomodulatory properties. Sci Rep 2015; 5:17381. [PMID: 26616555 PMCID: PMC4663632 DOI: 10.1038/srep17381] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2015] [Accepted: 10/29/2015] [Indexed: 12/13/2022] Open
Abstract
Lovastatin is a member of Statins, which are beneficial in a lot of immunologic cardiovascular diseases and T cell-mediated autoimmune diseases. Kv1.3 channel plays important roles in the activation and proliferation of T cells, and have become attractive target for immune-related disorders. The present study was designed to examine the block effect of Lovastatin on Kv1.3 channel in human T cells, and to clarify its new immunomodulatory mechanism. We found that Lovastatin inhibited Kv1.3 currents in a concentration- and voltage-dependent manner, and the IC50 for peak, end of the pulse was 39.81 ± 5.11, 6.92 ± 0.95 μM, respectively. Lovastatin also accelerated the decay rate of current inactivation and negatively shifted the steady-state inactivation curves concentration-dependently, without affecting the activation curve. However, 30 μM Lovastatin had no apparent effect on KCa current in human T cells. Furthermore, Lovastatin inhibited Ca2+ influx, T cell proliferation as well as IL-2 production. The activities of NFAT1 and NF-κB p65/50 were down-regulated by Lovastatin, too. At last, Mevalonate application only partially reversed the inhibition of Lovastatin on IL-2 secretion, and the siRNA against Kv1.3 also partially reduced this inhibitory effect of Lovastatin. In conclusion, Lovastatin can exert immunodulatory properties through the new mechanism of blocking Kv1.3 channel.
Collapse
|
23
|
Lima X, Cintra M, Piaza A, Mamoni R, Oliveira R, Magalhães R, Blotta M. Frequency and characteristics of circulating CD4+
CD28null
T cells in patients with psoriasis. Br J Dermatol 2015; 173:998-1005. [DOI: 10.1111/bjd.13993] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/16/2015] [Indexed: 12/24/2022]
Affiliation(s)
- X.T. Lima
- Department of Clinical Pathology; University of Campinas (UNICAMP); Campinas São Paulo Brazil
- Department of Dermatology; University of Campinas (UNICAMP); Campinas São Paulo Brazil
- School of Medicine; University of Fortaleza (UNIFOR); Fortaleza Ceará Brazil
| | - M.L. Cintra
- Department of Clinical Pathology; University of Campinas (UNICAMP); Campinas São Paulo Brazil
| | - A.C. Piaza
- Department of Clinical Pathology; University of Campinas (UNICAMP); Campinas São Paulo Brazil
| | - R.L. Mamoni
- Department of Clinical Pathology; University of Campinas (UNICAMP); Campinas São Paulo Brazil
| | - R.T. Oliveira
- Department of Clinical Pathology; University of Campinas (UNICAMP); Campinas São Paulo Brazil
| | - R.F. Magalhães
- Department of Dermatology; University of Campinas (UNICAMP); Campinas São Paulo Brazil
| | - M.H. Blotta
- Department of Clinical Pathology; University of Campinas (UNICAMP); Campinas São Paulo Brazil
| |
Collapse
|
24
|
Wu SJ, Villarreal DO, Shedlock DJ, Weiner DB. Synthetic DNA approach to cytomegalovirus vaccine/immune therapy. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2015; 848:131-48. [PMID: 25757619 DOI: 10.1007/978-1-4939-2432-5_7] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
There is no licensed vaccine or cure for human cytomegalovirus (CMV), a ubiquitous β-herpes virus that infects 60-95 % of adults worldwide. Infection is a major cause of congenital abnormalities in newborns, contributes to development of childhood cerebral palsy and medulloblastoma, can result in severe disease in immunocompromised patients, and is a major impediment during successful organ transplantation. While CMV has been increasingly associated with numerous inflammatory diseases and cancers, only recently has it been correlated with increased risk of heart disease in adults, the number-one killer in the USA. These data, among others, suggest that subclinical CMV infection, or microinfection, in healthy individuals may play more of a causative role than an epiphenomenon in development of CMV-associated pathologies. Due to the myriad of diseases and complications associated with CMV, an efficacious vaccine would be highly valuable in reducing human morbidity and mortality as well as saving billions of dollars in annual health-care costs and disability adjusted life years (DALY) in the developing world. Therefore, the development of a safe efficacious CMV vaccine or immune therapy is paramount to the public health. This review aims to provide a brief overview on aspects of CMV infection and disease and focuses on current vaccine strategies. The use of new synthetic DNA vaccines might offer one such approach to this difficult problem.
Collapse
Affiliation(s)
- Stephan J Wu
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, 505A Stellar-Chance Laboratories Curie Blvd, Philadelphia, 19104, PA, USA
| | | | | | | |
Collapse
|
25
|
Chang SC, Huq R, Chhabra S, Beeton C, Pennington MW, Smith BJ, Norton RS. N-Terminally extended analogues of the K⁺ channel toxin from Stichodactyla helianthus as potent and selective blockers of the voltage-gated potassium channel Kv1.3. FEBS J 2015; 282:2247-59. [PMID: 25864722 DOI: 10.1111/febs.13294] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2014] [Revised: 03/22/2015] [Accepted: 04/06/2015] [Indexed: 12/20/2022]
Abstract
The voltage-gated potassium channel Kv1.3 is an important target for the treatment of autoimmune diseases and asthma. Blockade of Kv1.3 by the sea anemone peptide K⁺-channel toxin from Stichodactyla helianthus (ShK) inhibits the proliferation of effector memory T lymphocytes and ameliorates autoimmune diseases in animal models. However, the lack of selectivity of ShK for Kv1.3 over the Kv1.1 subtype has driven a search for Kv1.3-selective analogues. In the present study, we describe N-terminally extended analogues of ShK that contain a negatively-charged Glu, designed to mimic the phosphonate adduct in earlier Kv1.3-selective analogues, and consist entirely of common protein amino acids. Molecular dynamics simulations indicated that a Trp residue at position [-3] of the tetrapeptide extension could form stable interactions with Pro377 of Kv1.3 and best discriminates between Kv1.3 and Kv1.1. This led to the development of ShK with an N-terminal Glu-Trp-Ser-Ser extension ([EWSS]ShK), which inhibits Kv1.3 with an IC₅₀ of 34 pm and is 158-fold selective for Kv1.3 over Kv1.1. In addition, [EWSS]ShK is more than 2900-fold more selective for Kv1.3 over Kv1.2 and KCa3.1 channels. As a highly Kv1.3-selective analogue of ShK based entirely on protein amino acids, which can be produced by recombinant expression, this peptide is a valuable addition to the complement of therapeutic candidates for the treatment of autoimmune diseases.
Collapse
Affiliation(s)
- Shih C Chang
- Medicinal Chemistry, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Vic., Australia
| | - Redwan Huq
- Department of Molecular Physiology and Biophysics, Baylor College of Medicine, Houston, TX, USA
| | - Sandeep Chhabra
- Medicinal Chemistry, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Vic., Australia
| | - Christine Beeton
- Department of Molecular Physiology and Biophysics, Baylor College of Medicine, Houston, TX, USA
| | | | - Brian J Smith
- Department of Chemistry and Physics, La Trobe Institute for Molecular Science, La Trobe University, Melbourne, Vic., Australia
| | - Raymond S Norton
- Medicinal Chemistry, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Vic., Australia
| |
Collapse
|
26
|
The story of CD4+ CD28- T cells revisited: solved or still ongoing? J Immunol Res 2015; 2015:348746. [PMID: 25834833 PMCID: PMC4365319 DOI: 10.1155/2015/348746] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2014] [Revised: 02/10/2015] [Accepted: 02/19/2015] [Indexed: 02/07/2023] Open
Abstract
CD4+CD28− T cells are a unique type of proinflammatory T cells characterised by blockade of costimulatory CD28 receptor expression at the transcriptional level, which is still reversible by IL-12. In healthy individuals older than 65 years, these cells may accumulate to up to 50% of total CD4+ T lymphocytes as in many immune-mediated diseases, immunodeficiency, and specific infectious diseases. Here we focus on CD4+CD28− T cells in chronic immune-mediated diseases, summarizing various phenotypic and functional characteristics, which vary depending on the underlying disease, disease activity, and concurrent treatment. CD4+CD28− T cells present as effector/memory cells with increased replicative history and oligoclonality but reduced apoptosis. As an alternative costimulatory signal instead of CD28, not only natural killer cell receptors and Toll-like receptors, but also CD47, CTLA-4, OX40, and 4-1BB have to be considered. The proinflammatory and cytotoxic capacities of these cells indicate an involvement in progression and maintenance of chronic immune-mediated disease. So far it has been shown that treatment with TNF-α blockers, abatacept, statins, and polyclonal antilymphocyte globulins (ATG) mediates reduction of the CD4+CD28− T cell level. The clinical relevance of targeting CD4+CD28− T cells as a therapeutic option has not been examined so far.
Collapse
|
27
|
Yin S, Hu Q, Luo J, Li Y, Lu C, Chen X, Hu H. Loureirin B, an essential component of Sanguis Draxonis, inhibits Kv1.3 channel and suppresses cytokine release from Jurkat T cells. Cell Biosci 2014; 4:78. [PMID: 25937895 PMCID: PMC4417528 DOI: 10.1186/2045-3701-4-78] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2014] [Accepted: 12/04/2014] [Indexed: 12/21/2022] Open
Abstract
Sanguis draxonis (SD), also known as “Dragon’s Blood”, is a traditional herb medicine that has been used to treat a variety of complications with unknown mechanisms. Recent studies show that SD displays immunosuppressive activities and improves symptoms of type I diabetes in animal models. However, the mechanisms underlying SD’s immunosuppressive actions are not completely understood. The voltage-gated Kv1.3 channel plays a critical role in the pathogenesis of autoimmune diseases by regulating the functions of both T cells and B cells. Here we investigated the effect of SD and one of its active components loureirin B (LrB) on Kv1.3. Both SD and LrB inhibited Kv1.3-mediated currents, produced a membrane depolarization, and reduced Ca2+ influx in Jurkat T cells. In addition, application of LrB inhibited phytohemagglutinin (PHA)-induced IL-2 release from activated Jurkat T cells. Furthermore, point mutations in the selective filter region significantly reduced the inhibitory effect of LrB on Kv1.3. The results of these experiments provide evidence that LrB is a channel blocker of Kv1.3 by interacting with amino acid residues in its selective filter region. Direct inhibition of Kv1.3 in T cells by SD and LrB might be the cellular and molecular basis of SD-mediated immunosuppression.
Collapse
Affiliation(s)
- Shijin Yin
- College of pharmacy, South-Central University for Nationalities, Wuhan, 430074 P R China ; Center for the Study of Itch, Department of Anesthesiology, Washington University School of Medicine, St. Louis, MO 63110 USA
| | - Qinglan Hu
- College of pharmacy, South-Central University for Nationalities, Wuhan, 430074 P R China
| | - Jialie Luo
- Center for the Study of Itch, Department of Anesthesiology, Washington University School of Medicine, St. Louis, MO 63110 USA
| | - Yuxin Li
- College of pharmacy, South-Central University for Nationalities, Wuhan, 430074 P R China
| | - Chunlan Lu
- College of pharmacy, South-Central University for Nationalities, Wuhan, 430074 P R China
| | - Xuan Chen
- College of pharmacy, South-Central University for Nationalities, Wuhan, 430074 P R China
| | - Hongzhen Hu
- Center for the Study of Itch, Department of Anesthesiology, Washington University School of Medicine, St. Louis, MO 63110 USA
| |
Collapse
|
28
|
Liaskou E, Jeffery LE, Trivedi PJ, Reynolds GM, Suresh S, Bruns T, Adams DH, Sansom DM, Hirschfield GM. Loss of CD28 expression by liver-infiltrating T cells contributes to pathogenesis of primary sclerosing cholangitis. Gastroenterology 2014; 147:221-232.e7. [PMID: 24726754 PMCID: PMC4961260 DOI: 10.1053/j.gastro.2014.04.003] [Citation(s) in RCA: 76] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/06/2014] [Revised: 04/01/2014] [Accepted: 04/04/2014] [Indexed: 12/21/2022]
Abstract
BACKGROUND & AIMS T-cell-mediated biliary injury is a feature of primary sclerosing cholangitis (PSC). We studied the roles of CD28(-) T cells in PSC and their regulation by vitamin D. METHODS Peripheral and liver-infiltrating mononuclear cells were isolated from blood or fresh liver tissue. We analyzed numbers, phenotypes, functions, and localization patterns of CD28(-) T cells, along with their ability to activate biliary epithelial cells. We measured levels of tumor necrosis factor (TNF)α in liver tissues from patients with PSC and the effects of exposure to active vitamin D (1,25[OH]2D3) on expression of CD28. RESULTS A significantly greater proportion of CD4(+) and CD8(+) T cells that infiltrated liver tissues of patients with PSC were CD28(-), compared with control liver tissue (CD4(+): 30.3% vs 2.5%, P < .0001; and CD8(+): 68.5% vs 31.9%, P < .05). The mean percentage of CD4(+)CD28(-) T cells in liver tissues from patients with PSC was significantly higher than from patients with primary biliary cirrhosis or nonalcoholic steatohepatitis (P < .05). CD28(-) T cells were activated CD69(+)CD45RA(-) C-C chemokine receptor (CCR)7(-) effector memory and perforin(+) granzyme B(+) cytotoxic cells, which express CD11a, CX3CR1, C-X3-C motif receptor 6 (CXCR6), and CCR10-consistent with their infiltration of liver and localization around bile ducts. Compared with CD28(+) T cells, activated CD28(-) T cells produced significantly higher levels of interferon γ and TNFα (P < .05), and induced up-regulation of intercellular cell adhesion molecule-1, HLA-DR, and CD40 by primary epithelial cells (3.6-fold, 1.5-fold, and 1.2-fold, respectively). Liver tissue from patients with PSC contained high levels of TNFα; TNFα down-regulated the expression of CD28 by T cells in vitro (P < .01); this effect was prevented by administration of 1,25(OH)2D3 (P < .05). CONCLUSIONS Inflammatory CD28(-) T cells accumulate in livers of patients with PSC and localize around bile ducts. The TNFα-rich microenvironment of this tissue promotes inflammation; these effects are reversed by vitamin D in vitro.
Collapse
Affiliation(s)
- Evaggelia Liaskou
- Centre for Liver Research and National Institute for Health Research Biomedical Research Unit in Liver Disease
| | - Louisa E. Jeffery
- Medical Research Council Centre for Immune Regulation, School of Immunity and Infection, Institute of Biomedical Research, University of Birmingham, Birmingham, United Kingdom
| | - Palak J. Trivedi
- Centre for Liver Research and National Institute for Health Research Biomedical Research Unit in Liver Disease
| | - Gary M. Reynolds
- Centre for Liver Research and National Institute for Health Research Biomedical Research Unit in Liver Disease
| | - Shankar Suresh
- Centre for Liver Research and National Institute for Health Research Biomedical Research Unit in Liver Disease
| | - Tony Bruns
- Centre for Liver Research and National Institute for Health Research Biomedical Research Unit in Liver Disease,Department of Internal Medicine IV, Gastroenterology, Hepatology and Infectious Disease,Center for Sepsis Control and Care, Jena University Hospital, Friedrich Schiller University of Jena, Jena, Germany
| | - David H. Adams
- Centre for Liver Research and National Institute for Health Research Biomedical Research Unit in Liver Disease
| | - David M. Sansom
- Medical Research Council Centre for Immune Regulation, School of Immunity and Infection, Institute of Biomedical Research, University of Birmingham, Birmingham, United Kingdom,Institute for Immunity and Transplantation, University College London, Royal Free Campus, London, United Kingdom
| | - Gideon M. Hirschfield
- Centre for Liver Research and National Institute for Health Research Biomedical Research Unit in Liver Disease
| |
Collapse
|
29
|
Human cytomegalovirus and autoimmune disease. BIOMED RESEARCH INTERNATIONAL 2014; 2014:472978. [PMID: 24967373 PMCID: PMC4022258 DOI: 10.1155/2014/472978] [Citation(s) in RCA: 91] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 01/27/2014] [Accepted: 03/17/2014] [Indexed: 11/28/2022]
Abstract
Human cytomegalovirus (HCMV) represents a prototypic pathogenic member of the β-subgroup of the herpesvirus family. A range of HCMV features like its lytic replication in multiple tissues, the lifelong persistence through periods of latency and intermitting reactivation, the extraordinary large proteome, and extensive manipulation of adaptive and innate immunity make HCMV a high profile candidate for involvement in autoimmune disorders. We surveyed the available literature for reports on HCMV association with onset or exacerbation of autoimmune disease. A causative linkage between HCMV and systemic lupus erythematosus (SLE), systemic sclerosis (SSc), diabetes mellitus type 1, and rheumatoid arthritis (RA) is suggested by the literature. However, a clear association of HCMV seroprevalence and disease could not be established, leaving the question open whether HCMV could play a coresponsible role for onset of disease. For convincing conclusions population-based prospective studies must be performed in the future. Specific immunopathogenic mechanisms by which HCMV could contribute to the course of autoimmune disease have been suggested, for example, molecular mimicry by UL94 in SSc and UL83/pp65 in SLE patients, as well as aggravation of joint inflammation by induction and expansion of CD4+/CD28− T-cells in RA patients. Further studies are needed to validate these findings and to lay the grounds for targeted therapeutic intervention.
Collapse
|
30
|
Pieper J, Johansson S, Snir O, Linton L, Rieck M, Buckner JH, Winqvist O, van Vollenhoven R, Malmström V. Peripheral and site-specific CD4(+) CD28(null) T cells from rheumatoid arthritis patients show distinct characteristics. Scand J Immunol 2014; 79:149-55. [PMID: 24313359 DOI: 10.1111/sji.12139] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2013] [Accepted: 11/19/2013] [Indexed: 12/28/2022]
Abstract
Proinflammatory CD4(+) CD28(null) T cells are frequently found in the circulation of patients with rheumatoid arthritis (RA), but are less common in the rheumatic joint. In the present study, we sought to identify functional differences between CD4(+) CD28(null) T cells from blood and synovial fluid in comparison with conventional CD28-expressing CD4(+) T cells. Forty-four patients with RA, displaying a distinct CD4(+) CD28(null) T cell population in blood, were recruited for this study; the methylation status of the IFNG locus was examined in isolated T cell subsets, and intracellular cytokine production (IFN-γ, TNF, IL-17) and chemokine receptor expression (CXCR3, CCR6 and CCR7) were assessed by flow cytometry on T cells from the two compartments. Circulating CD4(+) CD28(null) T cells were significantly more hypomethylated in the CNS-1 region of the IFNG locus than conventional CD4(+) CD28(+) T cells and produced higher levels of both IFN-γ and TNF after TCR cross-linking. CD4(+) CD28(null) T cells from the site of inflammation expressed significantly more CXCR3 and CCR6 compared to their counterparts in blood. While IL-17A production could hardly be detected in CD4(+) CD28(null) cells from the blood, a significant production was observed in CD4(+) CD28(null) T cells from synovial fluid. CD4(+) CD28(null) T cells were not only found to differ from conventional CD4(+) CD28(+) T cells in the circulation, but we could also demonstrate that synovial CD4(+) CD28(null) T cells showed additional effector functions (IL-17 coproduction) as compared to the same subset in peripheral blood, suggesting an active role for these cells in the perpetuation of inflammation in the subset of patients having a CD28(null) population.
Collapse
Affiliation(s)
- J Pieper
- Rheumatology Unit, Department of Medicine at Karolinska University Hospital, Karolinska Institute, Solna, Stockholm, Sweden
| | | | | | | | | | | | | | | | | |
Collapse
|
31
|
Sheu TT, Chiang BL, Yen JH, Lin WC. Premature CD4+ T cell aging and its contribution to lymphopenia-induced proliferation of memory cells in autoimmune-prone non-obese diabetic mice. PLoS One 2014; 9:e89379. [PMID: 24586733 PMCID: PMC3935863 DOI: 10.1371/journal.pone.0089379] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2013] [Accepted: 01/21/2014] [Indexed: 01/15/2023] Open
Abstract
Lymphopenia-induced proliferation (LIP), a mechanism to maintain a constant number of T cells in circulation, occurs in both normal aging and autoimmune disease. The incidence of most autoimmune diseases increases with age, and premature CD4(+) T cell aging has been reported in several autoimmune diseases. In this study, we tested the hypothesis that premature CD4(+) T cell aging can cause autoimmune disease by examining whether premature CD4(+) T cell aging exists and causes LIP in our mouse model. Non-obese diabetic (NOD) mice were used because, in addition to Treg defects, the LIP of T cells has been shown to plays a causative role in the development of insulin-dependent diabetes mellitus (IDDM) in these mice. We found that with advancing age, NOD mice exhibited an accelerated decrease in the number of CD4(+) T cells due to the loss of naïve cells. This was accompanied by an increase in the percentage of memory cells, leading to a reduced naïve/memory ratio. In addition, both the percentage of CD28(+) cells in CD4(+) T cells and IL-2 production decreased, while the percentage of FAS(+)CD44(+) increased, suggesting that NOD mice exhibit premature CD4(+) T cell aging. This process preferentially contributed to LIP of memory cells. Therefore, our results suggest that premature CD4(+) T cell aging underlies the development of IDDM in NOD mice. Given that CD28 and IL-2 play important roles in Treg function, the relationships between premature CD4(+) T cell aging and lymphopenia as well as Treg defects in autoimmune-prone NOD mice are proposed.
Collapse
Affiliation(s)
- Ting-Ting Sheu
- Department of Immunology, Tzu Chi University, Hualien, Taiwan, Republic of China
- Institute of Microbiology, Immunology and Biochemistry, Tzu Chi University, Hualien, Taiwan, Republic of China
- * E-mail:
| | - Bor-Luen Chiang
- Graduate Institute of Immunology, National Taiwan University, Taipei, Taiwan, Republic of China
- Department of Medical Research, National Taiwan University Hospital, Taipei, Taiwan, Republic of China
| | - Jui-Hung Yen
- Department of Molecular Biology and Human Genetics, Tzu Chi University, Hualien, Taiwan, Republic of China
| | - Wen-Chi Lin
- Institute of Microbiology, Immunology and Biochemistry, Tzu Chi University, Hualien, Taiwan, Republic of China
| |
Collapse
|
32
|
Péguillet I, Milder M, Louis D, Vincent-Salomon A, Dorval T, Piperno-Neumann S, Scholl SM, Lantz O. High numbers of differentiated effector CD4 T cells are found in patients with cancer and correlate with clinical response after neoadjuvant therapy of breast cancer. Cancer Res 2014; 74:2204-16. [PMID: 24535711 DOI: 10.1158/0008-5472.can-13-2269] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
CD4(+) T cells influence tumor immunity in complex ways that are not fully understood. In this study, we characterized a population of human differentiated effector CD4(+) T cells that is defined by low levels of the interleukin (IL)-2 and IL-7 receptors (CD25(-)CD127(-)). We found that this cell population expands in patients with various types of cancer, including breast cancer, to represent 2% to 20% of total CD4(+) blood T lymphocytes as compared with only 0.2% to 2% in healthy individuals. Notably, these CD25(-)CD127(-)CD4 T cells expressed effector markers such as CD244 and CD11b with low levels of CD27, contrasting with the memory phenotype dominating this population in healthy individuals. These cells did not cycle in patients, nor did they secrete IL-10 or IL-17, but instead displayed cytotoxic features. Moreover, they encompassed oligoclonal expansions paralleling an expansion of effector CD8(+) T cells that included tumor antigen-specific T cells. During neoadjuvant chemotherapy in patients with breast cancer, we found that the increase in CD25(-)CD127(-) CD4(+) T cells correlated with tumor regression. This observation suggested that CD4(+) T cells included tumor antigen-specific cells, which may be generated by or participate in tumor regressions during chemotherapy. In summary, our results lend support to the hypothesis that CD4(+) T cells are involved in human antitumor responses.
Collapse
Affiliation(s)
- Isabelle Péguillet
- Authors' Affiliations: Clinical immunology Laboratory; Center of Clinical Investigations CICBT507 IGR/Curie; Departments of Pathology, and Medical Oncology; and Inserm U932, Institut Curie, Paris, France
| | | | | | | | | | | | | | | |
Collapse
|
33
|
Hautz T, Krapf C, Grahammer J, Zelger B, Hickethier T, Seger C, Eberhart N, Wallner C, Messner F, Kotsch K, Griesmacher A, Brandacher G, Lee WPA, Margreiter R, Pratschke J, Glossmann H, Schneeberger S. Targeting the Kv1.3 potassium channel for immunosuppression in vascularized composite allotransplantation - a pilot study. Transpl Int 2013; 26:552-61. [PMID: 23489391 DOI: 10.1111/tri.12080] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2012] [Revised: 03/19/2012] [Accepted: 02/04/2013] [Indexed: 12/19/2022]
Abstract
Kv1.3-channels are critically involved in activation and function of effector memory T cells. Blocking Kv1.3-channels was investigated for its effect on skin rejection in a rat limb-transplantation-model. Animals received the Kv1.3-blocker correolide C systemically or locally as intra-graft-treatment in combination with tacrolimus. Systemic (intraperitoneal) administration of correolide C resulted in slight, but significant prolongation of allograft survival compared with untreated and placebo treated controls. In 4/6 correolide C treated animals, histology showed an intact epidermis and a mild infiltrate by day 10. High correolide C plasma trough levels correlated with prolonged allograft survival. A decrease in CD4+ and CD8+ effector memory T cells was observed in allograft skin, peripheral blood and the spleen on day 5. When applied subcutaneously in combination with systemic tacrolimus (30 days+/-anti-lymphocyte serum) detectable, but insignificant prolongation of graft survival was achieved. 2/5 animals showed an intact epidermis and a mild infiltrate until day 45. Tapering systemic tacrolimus and weaning on day 50 resulted in rejection by day 55, regardless of local correolide C treatment. Subcutaneous injection did not lead to systemic plasma levels. The Kv1.3-channel is a potential drug target worth exploring in more detail for immunosuppression in vascularized composite allotransplantation.
Collapse
Affiliation(s)
- Theresa Hautz
- Department of Visceral, Transplant and Thoracic Surgery, Center of Operative Medicine, Innsbruck Medical University, Innsbruck, Austria
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
34
|
Zhao N, Dong Q, Du LL, Fu XX, Du YM, Liao YH. Potent suppression of Kv1.3 potassium channel and IL-2 secretion by diphenyl phosphine oxide-1 in human T cells. PLoS One 2013; 8:e64629. [PMID: 23717641 PMCID: PMC3661503 DOI: 10.1371/journal.pone.0064629] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2012] [Accepted: 04/16/2013] [Indexed: 11/18/2022] Open
Abstract
Diphenyl phosphine oxide-1 (DPO-1) is a potent Kv1.5 channel inhibitor that has therapeutic potential for the treatment of atrial fibrillation. Many other Kv1.5 channel blockers also potently inhibit the Kv1.3 channel, but whether DPO-1 blocks Kv1.3 channels has not been investigated. The Kv1.3 channel is highly expressed in activated T cells, which is considered a favorable target for immunomodulation. Accordingly, we hypothesized that DPO-1 may exert immunosuppressive and anti-inflammatory effects by inhibiting Kv1.3 channel activity. In this study, DPO-1 blocked Kv1.3 current in a voltage-dependent and concentration-dependent manner, with IC₅₀ values of 2.58 µM in Jurkat cells and 3.11 µM in human peripheral blood T cells. DPO-1 also accelerated the inactivation rate and negatively shifted steady-state inactivation. Moreover, DPO-1 at 3 µM had no apparent effect on the Ca²⁺ activated potassium channel (K(Ca)) current in both Jurkat cells and human peripheral blood T cells. In Jurkat cells, pre-treatment with DPO-1 for 24 h decreased Kv1.3 current density, and protein expression by 48±6% and 60±9%, at 3 and 10 µM, respectively (both p<0.05). In addition, Ca²⁺ influx to Ca²⁺-depleted cells was blunted and IL-2 production was also reduced in activated Jurkat cells. IL-2 secretion was also inhibited by the Kv1.3 inhibitors margatoxin and charybdotoxin. Our results demonstrate for the first time that that DPO-1, at clinically relevant concentrations, blocks Kv1.3 channels, decreases Kv1.3 channel expression and suppresses IL-2 secretion. Therefore, DPO-1 may be a useful treatment strategy for immunologic disorders.
Collapse
Affiliation(s)
- Ning Zhao
- Research Center of Ion Channelopathy, Institute of Cardiovascular Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, P.R. China
| | - Qian Dong
- Research Center of Ion Channelopathy, Institute of Cardiovascular Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, P.R. China
| | - Li-Li Du
- Research Center of Ion Channelopathy, Institute of Cardiovascular Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, P.R. China
| | - Xiao-Xing Fu
- Research Center of Ion Channelopathy, Institute of Cardiovascular Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, P.R. China
| | - Yi-Mei Du
- Research Center of Ion Channelopathy, Institute of Cardiovascular Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, P.R. China
- * E-mail: (Y-MD); (Y-HL)
| | - Yu-Hua Liao
- Research Center of Ion Channelopathy, Institute of Cardiovascular Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, P.R. China
- * E-mail: (Y-MD); (Y-HL)
| |
Collapse
|
35
|
Feske S, Skolnik EY, Prakriya M. Ion channels and transporters in lymphocyte function and immunity. Nat Rev Immunol 2012; 12:532-47. [PMID: 22699833 DOI: 10.1038/nri3233] [Citation(s) in RCA: 350] [Impact Index Per Article: 26.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Lymphocyte function is regulated by a network of ion channels and transporters in the plasma membrane of B and T cells. These proteins modulate the cytoplasmic concentrations of diverse cations, such as calcium, magnesium and zinc ions, which function as second messengers to regulate crucial lymphocyte effector functions, including cytokine production, differentiation and cytotoxicity. The repertoire of ion-conducting proteins includes calcium release-activated calcium (CRAC) channels, P2X receptors, transient receptor potential (TRP) channels, potassium channels, chloride channels and magnesium and zinc transporters. This Review discusses the roles of ion conduction pathways in lymphocyte function and immunity.
Collapse
Affiliation(s)
- Stefan Feske
- Department of Pathology, New York University Langone Medical Center, New York, New York 10016, USA.
| | | | | |
Collapse
|
36
|
Yang XF, Yang Y, Lian YT, Wang ZH, Li XW, Cheng LX, Liu JP, Wang YF, Gao X, Liao YH, Wang M, Zeng QT, Liu K. The antibody targeting the E314 peptide of human Kv1.3 pore region serves as a novel, potent and specific channel blocker. PLoS One 2012; 7:e36379. [PMID: 22558454 PMCID: PMC3338681 DOI: 10.1371/journal.pone.0036379] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2011] [Accepted: 04/04/2012] [Indexed: 01/08/2023] Open
Abstract
Selective blockade of Kv1.3 channels in effector memory T (T(EM)) cells was validated to ameliorate autoimmune or autoimmune-associated diseases. We generated the antibody directed against one peptide of human Kv1.3 (hKv1.3) extracellular loop as a novel and possible Kv1.3 blocker. One peptide of hKv1.3 extracellular loop E3 containing 14 amino acids (E314) was chosen as an antigenic determinant to generate the E314 antibody. The E314 antibody specifically recognized 63.8KD protein stably expressed in hKv1.3-HEK 293 cell lines, whereas it did not recognize or cross-react to human Kv1.1(hKv1.1), Kv1.2(hKv1.2), Kv1.4(hKv1.4), Kv1.5(hKv1.5), KCa3.1(hKCa3.1), HERG, hKCNQ1/hKCNE1, Nav1.5 and Cav1.2 proteins stably expressed in HEK 293 cell lines or in human atrial or ventricular myocytes by Western blotting analysis and immunostaining detection. By the technique of whole-cell patch clamp, the E314 antibody was shown to have a directly inhibitory effect on hKv1.3 currents expressed in HEK 293 or Jurkat T cells and the inhibition showed a concentration-dependence. However, it exerted no significant difference on hKv1.1, hKv1.2, hKv1.4, hKv1.5, hKCa3.1, HERG, hKCNQ1/hKCNE1, L-type Ca(2+) or voltage-gated Na(+) currents. The present study demonstrates that the antibody targeting the E314 peptide of hKv1.3 pore region could be a novel, potent and specific hKv1.3 blocker without affecting a variety of closely related K(v)1 channels, KCa3.1 channels and functional cardiac ion channels underlying central nervous system (CNS) disorders or drug-acquired arrhythmias, which is required as a safe clinic-promising channel blocker.
Collapse
Affiliation(s)
- Xiao-Fang Yang
- Department of Cardiology, Union Hospital, Huazhong University of Science and Technology, Wuhan, China
| | - Yong Yang
- Department of Cardiology, Union Hospital, Huazhong University of Science and Technology, Wuhan, China
| | - Yi-Tian Lian
- Department of Cardiology, Union Hospital, Huazhong University of Science and Technology, Wuhan, China
| | - Zhao-Hui Wang
- Department of Geriatrics, Union Hospital, Huazhong University of Science and Technology, Wuhan, China
| | - Xiao-Wei Li
- Department of Cardiology, Tianjin Chest Hospital, Tianjin, China
| | - Long-Xian Cheng
- Department of Cardiology, Union Hospital, Huazhong University of Science and Technology, Wuhan, China
| | - Jin-Ping Liu
- Department of Cardiovascular Surgery, Union Hospital, Huazhong University of Science and Technology, Wuhan, China
| | - Yan-Fu Wang
- Department of Cardiology, Affiliated Hospital, Jining Medical College, Shandong, China
| | - Xiang Gao
- Department of Geriatrics, Tongji Hospital, Huazhong University of Science and Technology, Wuhan, China
| | - Yu-Hua Liao
- Department of Cardiology, Union Hospital, Huazhong University of Science and Technology, Wuhan, China
| | - Min Wang
- Department of Cardiology, Union Hospital, Huazhong University of Science and Technology, Wuhan, China
| | - Qiu-Tang Zeng
- Department of Cardiology, Union Hospital, Huazhong University of Science and Technology, Wuhan, China
| | - Kun Liu
- Department of Cardiology, Union Hospital, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
37
|
Ponchel F, Vital E, Kingsbury SR, El-Sherbiny YM. CD4+T-cell subsets in rheumatoid arthritis. ACTA ACUST UNITED AC 2012. [DOI: 10.2217/ijr.11.69] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
|
38
|
Pinto-Medel MJ, García-León JA, Oliver-Martos B, López-Gómez C, Luque G, Arnáiz-Urrutia C, Orpez T, Marín-Bañasco C, Fernández O, Leyva L. The CD4+ T-cell subset lacking expression of the CD28 costimulatory molecule is expanded and shows a higher activation state in multiple sclerosis. J Neuroimmunol 2012; 243:1-11. [PMID: 22261542 DOI: 10.1016/j.jneuroim.2011.11.008] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2011] [Revised: 11/15/2011] [Accepted: 11/16/2011] [Indexed: 11/30/2022]
Abstract
Multiple sclerosis (MS) is a chronic debilitating disease, in which T-cells are considered to play a pivotal role. CD28 is the quintessential costimulatory molecule on T-cells and its expression declines progressively with repeated stimulations, leading to the generation of CD28(-) T-cells. Our aim was to examine whether CD4(+)CD28(-) T-cells were enriched in MS patients, and characterize the phenotype of this subset in MS patients and healthy controls (HC). All these changes could provide these CD4(+)CD28(-) T-cell characteristics that might be involved in the pathogenesis of MS, turning this T-cell subset into a potential target for future therapeutic strategies.
Collapse
Affiliation(s)
- María Jesús Pinto-Medel
- Research Laboratory, Hospital Regional Universitario Carlos Haya and Fundación IMABIS, Málaga, Spain
| | | | | | | | | | | | | | | | | | | |
Collapse
|
39
|
Chi V, Pennington MW, Norton RS, Tarcha EJ, Londono LM, Sims-Fahey B, Upadhyay SK, Lakey JT, Iadonato S, Wulff H, Beeton C, Chandy KG. Development of a sea anemone toxin as an immunomodulator for therapy of autoimmune diseases. Toxicon 2011; 59:529-46. [PMID: 21867724 DOI: 10.1016/j.toxicon.2011.07.016] [Citation(s) in RCA: 170] [Impact Index Per Article: 12.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2011] [Revised: 06/16/2011] [Accepted: 07/20/2011] [Indexed: 01/05/2023]
Abstract
Electrophysiological and pharmacological studies coupled with molecular identification have revealed a unique network of ion channels--Kv1.3, KCa3.1, CRAC (Orai1 + Stim1), TRPM7, Cl(swell)--in lymphocytes that initiates and maintains the calcium signaling cascade required for activation. The expression pattern of these channels changes during lymphocyte activation and differentiation, allowing the functional network to adapt during an immune response. The Kv1.3 channel is of interest because it plays a critical role in subsets of T and B lymphocytes implicated in autoimmune disorders. The ShK toxin from the sea anemone Stichodactyla helianthus is a potent blocker of Kv1.3. ShK-186, a synthetic analog of ShK, is being developed as a therapeutic for autoimmune diseases, and is scheduled to begin first-in-man phase-1 trials in 2011. This review describes the journey that has led to the development of ShK-186.
Collapse
Affiliation(s)
- Victor Chi
- Department of Physiology and Biophysics, UC Irvine, Irvine, CA 92697, USA
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
40
|
Pandya JM, Fasth AER, Zong M, Arnardottir S, Dani L, Lindroos E, Malmström V, Lundberg IE. Expanded T cell receptor Vβ-restricted T cells from patients with sporadic inclusion body myositis are proinflammatory and cytotoxic CD28null T cells. ACTA ACUST UNITED AC 2010; 62:3457-66. [PMID: 20662057 DOI: 10.1002/art.27665] [Citation(s) in RCA: 59] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
OBJECTIVE Sporadic inclusion body myositis (IBM) is characterized by T cell infiltrates in muscle tissue, but their functional role is unclear. Systemic signs of inflammation are lacking, and the absence of beneficial effects following immunosuppression has challenged the notion of a role for the immune system. This study was undertaken to investigate the phenotype and functionality of T cells, specifically a subset of proinflammatory, cytotoxic, and apoptosis-resistant T cells defined as CD28(null) T cells, in the pathogenesis of sporadic IBM. METHODS A cohort of 27 patients with sporadic IBM was analyzed for the frequency of circulating and muscle-infiltrating CD28(null) T cells. The T cell receptor (TCR) V(β) usage was determined using flow cytometry and immunohistochemistry. Anti-CD3-stimulated peripheral blood mononuclear cells were analyzed for intracellular interferon-γ and cytotoxic potential by flow cytometry. RESULTS We found striking accumulations of both CD8+CD28(null) and CD4+CD28(null) T cells, which represented the TCR V(β) -expanded T cells in sporadic IBM. Such CD28(null) T cells were abundant both in the inflamed muscle tissue and in the circulation. Although the specific TCR V(β) expansions varied between patients, both CD8+CD28(null) and CD4+CD28(null) T cells consistently displayed a highly proinflammatory and cytotoxic potential. CONCLUSION Our results suggest that CD28null T cell expansions represent the previously described expanded T cell subsets in sporadic IBM, and their proinflammatory capacity and presence in both muscle tissue and the circulation may imply a role of immune activation in sporadic IBM. In addition, CD4+CD28(null) T cells may exert cytotoxic effects directly on muscle fibers due to a cytotoxic potential similar to that in CD8+ T cells.
Collapse
Affiliation(s)
- Jayesh M Pandya
- Rheumatology Unit, Karolinska University Hospital, Stockholm, Sweden.
| | | | | | | | | | | | | | | |
Collapse
|
41
|
Eastwood D, Findlay L, Poole S, Bird C, Wadhwa M, Moore M, Burns C, Thorpe R, Stebbings R. Monoclonal antibody TGN1412 trial failure explained by species differences in CD28 expression on CD4+ effector memory T-cells. Br J Pharmacol 2010; 161:512-26. [PMID: 20880392 PMCID: PMC2990151 DOI: 10.1111/j.1476-5381.2010.00922.x] [Citation(s) in RCA: 206] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2010] [Revised: 03/26/2010] [Accepted: 03/30/2010] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND AND PURPOSE In 2006, a life-threatening 'cytokine storm', not predicted by pre-clinical safety testing, rapidly occurred in all six healthy volunteers during the phase I clinical trial of the CD28 superagonist monoclonal antibody (mAb) TGN1412. To date, no unequivocal explanation for the failure of TGN1412 to stimulate profound cytokine release in vitro or in vivo in species used for pre-clinical safety testing has been established. Here, we have identified a species difference almost certainly responsible for this disparate immunopharmacology. EXPERIMENTAL APPROACH Polychromatic flow cytometry and intracellular cytokine staining were employed to dissect the in vitro immunopharmacology of TGN1412 and other therapeutic mAbs at the cellular level to identify differences between humans and species used for pre-clinical safety testing. KEY RESULTS In vitro IL-2 and IFN-γ release from CD4+ effector memory T-cells were key indicators of a TGN1412-type response. This mechanism of cytokine release differed from that of other therapeutic mAbs, which can cause adverse reactions, because these other mAbs stimulate cytokine release primarily from natural killer cells. In contrast to humans, CD28 is not expressed on the CD4+ effector memory T-cells of all species used for pre-clinical safety testing, so cannot be stimulated by TGN1412. CONCLUSIONS AND IMPLICATIONS It is likely that activation of CD4+ effector memory T-cells by TGN1412 was responsible for the cytokine storm. Lack of CD28 expression on the CD4+ effector memory T-cells of species used for pre-clinical safety testing of TGN1412 offers an explanation for the failure to predict a 'cytokine storm' in humans.
Collapse
Affiliation(s)
- D Eastwood
- Biotherapeutics Group, NIBSC, Potters Bar, Hertfordshire, UK
| | | | | | | | | | | | | | | | | |
Collapse
|
42
|
Fasth AER, Björkström NK, Anthoni M, Malmberg KJ, Malmström V. Activating NK-cell receptors co-stimulate CD4(+)CD28(-) T cells in patients with rheumatoid arthritis. Eur J Immunol 2010; 40:378-87. [PMID: 19904767 DOI: 10.1002/eji.200939399] [Citation(s) in RCA: 55] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Effector T-cell responses can be modulated by competing positive or negative signals transduced by NK-cell receptors (NKR). In the CD4(+) T-cell population, the expression of NKR is primarily found in the CD4(+)CD28(-) T-cell subset, also known as CD28(null) T cells. These T cells are frequently found in rheumatoid arthritis (RA) and other inflammatory disorders, suggesting that signaling through NKR may play a role in the autoimmune reaction. Here we aimed to dissect the phenotype and function of NKR-expressing CD4(+)CD28(-) T cells in patients with RA. By analyzing a broad array of NKR on CD4(+)CD28(-) T cells we found a significant expression of the co-activating receptors 2B4 (CD244), DNAM-1 (CD226), and CRACC. Pair-wise ligations of 2B4 with DNAM-1 and/or NKG2D lead to increased effector functions of primary CD4(+)CD28(-) T cells to suboptimal levels of anti-CD3 stimulation. Using multi-parameter flow cytometry, we demonstrate that such co-ligation led to an increased magnitude in overall responsiveness without changing qualitative aspects of the response. Altogether these results demonstrate a pattern of additive effects in NKR-mediated functional modulation of CD4(+)CD28(-) T cells in RA. This may have consequences for the inflammatory responses imposed by these cells, thus influencing disease manifestations.
Collapse
Affiliation(s)
- Andreas E R Fasth
- Rheumatology Unit, Department of Medicine, Center for Molecular Medicine, Karolinska Institutet, Karolinska University Hospital Solna, Stockholm, Sweden
| | | | | | | | | |
Collapse
|
43
|
Mayerl C, Prelog M. Immunosenescence and juvenile idiopathic arthritis. Autoimmun Rev 2010; 11:297-300. [PMID: 20172056 DOI: 10.1016/j.autrev.2010.02.015] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2010] [Accepted: 02/15/2010] [Indexed: 11/17/2022]
Abstract
Aging of the immune system (immunosenescence) is characterized by diminished thymus function, decreased output of recent thymic emigrants, compensatory peripheral proliferation of mature T cells and oligoclonal expansions of specific CD28(-) T cells. Clinical consequences are poor responses to new antigens or vaccinations, increased infection rates with higher morbidity and mortality, and increasing incidence of autoimmune diseases with advancing age. Premature immunosenescence is suggested to play a role in the pathogenesis of adult rheumatoid arthritis and in children with juvenile idiopathic arthritis (JIA). However, so far, there is not enough evidence for supporting one of the two theories: the first, favoring premature immunosenscence in children developing autoimmune conditions as the primary defect causing break-down of self-tolerance; the second, that premature immunosenescence in children with autoimmune disorders is secondary to chronic stimulation and activation of the immune system by inflammatory processes by the autoimmune disease itself. This contradictory view of etiology and pathogenesis of autoimmune diseases in the very young underlines the need for population-based longitudinal studies on immune-risk factors for autoimmune diseases beginning at infancy.
Collapse
Affiliation(s)
- Christina Mayerl
- Division of Experimental Pathophysiologie and Immunology, Department Biocenter, Medical University Innsbruck, Austria
| | | |
Collapse
|
44
|
Radovits BJ, Kievit W, Laan RFJM. Tumour necrosis factor-alpha antagonists in the management of rheumatoid arthritis in the elderly: a review of their efficacy and safety. Drugs Aging 2010; 26:647-64. [PMID: 19685931 DOI: 10.2165/11316460-000000000-00000] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Rheumatoid arthritis (RA) is a chronic inflammatory disease that frequently affects people aged >or=65 years, causing significant impairment with pain and functional disability. Elderly RA patients have specific problems, including co-morbid diseases, numerous concomitant medications, greater number of damaged joints as a result of longer disease duration and often a more severe disease presentation in elderly-onset RA. These factors, together with an age-related decline in the immune defence mechanisms, make elderly patients more vulnerable. The new era of biologic medications has made intensive treatment of RA patients possible. Anti-tumour necrosis factor-alpha (anti-TNFalpha) agents can cause a dramatic improvement in disease activity and functional capacity, making complete remission of RA a possible target. TNFalpha has been shown to play an important role in both the healthy aging process and age-related diseases such as RA. Targeting this cytokine in elderly patients is therefore reasonable. However, it is not clear whether treatment effects can be reached to the same extent in both elderly and younger patients and whether anti-TNFalpha treatment specifically increases the risk of certain adverse events in elderly RA patients. This review discusses the currently available evidence relating to the efficacy and safety of anti-TNFalpha medication in RA patients aged >or=65 years treated in clinical trials and observational studies. Despite a slightly less robust effect in elderly patients, anti-TNFalpha treatment has a similar long-term efficacy in patients aged >or=65 years and patients aged <65 years. The majority of the study results showed that anti-TNFalpha treatment does not elevate the risk of infections beyond the risk found in age-matched controls. When a moderate increase in risk was found, this occurred equally in elderly and younger patients. Furthermore, whereas anti-TNFalpha agents were found to be relatively safe in the treatment of elderly RA patients, treatment with corticosteroids significantly elevated the risk of serious infections. Corticosteroids are frequently used in elderly patients, but the evidence suggests that preference should increasingly be given to anti-TNFalpha agents, for which the expected benefits will mostly outweigh the modestly increased risks of associated adverse events.
Collapse
Affiliation(s)
- Beáta J Radovits
- Department of Rheumatology, Radboud University Nijmegen Medical Centre, Nijmegen, the Netherlands
| | | | | |
Collapse
|
45
|
Fasth AER, Dastmalchi M, Rahbar A, Salomonsson S, Pandya JM, Lindroos E, Nennesmo I, Malmberg KJ, Söderberg-Nauclér C, Trollmo C, Lundberg IE, Malmström V. T cell infiltrates in the muscles of patients with dermatomyositis and polymyositis are dominated by CD28null T cells. THE JOURNAL OF IMMUNOLOGY 2009; 183:4792-9. [PMID: 19752224 DOI: 10.4049/jimmunol.0803688] [Citation(s) in RCA: 110] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Dermatomyositis and polymyositis are disabling rheumatic diseases characterized by an appreciable number of T cells infiltrating muscle tissue. The precise phenotype, function and specificity of these cells remain elusive. In this study, we aimed to characterize T cells in muscle tissue and circulation and to investigate their association to clinical phenotype. Twenty-four patients with dermatomyositis and 42 with polymyositis were screened for frequency of CD4+CD28(null) and CD8+CD28(null) T cells in peripheral blood by flow cytometry. Presence of these cells in inflamed muscle tissue from 13 of these patients was analyzed by three-color immunofluorescence microscopy. Effector functions, proliferation and Ag specificity were analyzed by flow cytometry after in vitro stimulation. The clinical relevance of CD28(null) T cells was analyzed by multiple regression analyses including six separate and combined disease variables. We demonstrate that muscle-infiltrating T cells are predominantly CD4+CD28(null) and CD8+CD28(null) T cells in patients with dermatomyositis and polymyositis. Muscle-infiltrating CD28(null) T cells were found already at time of diagnosis. Disease activity correlated with the frequency of CD8+ T cells in the inflamed muscles of polymyositis patients. Circulating CD4+CD28(null) and CD8+CD28(null) T cells were significantly more frequent in human CMV (HCMV) seropositive individuals, responded to HCMV Ag stimulation, and correlated with disease duration. These cells also display a proinflammatory cytokine profile, contain perforin and lack the costimulatory molecule CD28. Our observations imply that CD28(null) T cells represent clinically important effector cells in dermatomyositis and polymyositis, and that HCMV might play a role in propagating disease in a subset of patients.
Collapse
Affiliation(s)
- Andreas E R Fasth
- Rheumatology Unit, Center for Molecular Medicine L8:04, Karolinska University Hospital, Stockholm S-171 76, Sweden.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
46
|
Wang Y, Bai J, Li F, Wang H, Fu X, Zhao T, Xu W, Zhang J, Ni B, Wu Y. Characteristics of Expanded CD4+CD28nullT Cells in Patients with Chronic Hepatitis B. Immunol Invest 2009; 38:434-46. [DOI: 10.1080/08820130902943105] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
|
47
|
Prelog M, Schwarzenbrunner N, Sailer-Höck M, Kern H, Klein-Franke A, Ausserlechner MJ, Koppelstaetter C, Brunner A, Duftner C, Dejaco C, Strasak AM, Müller T, Zimmerhackl LB, Brunner J. Premature aging of the immune system in children with juvenile idiopathic arthritis. ACTA ACUST UNITED AC 2008; 58:2153-62. [PMID: 18576332 DOI: 10.1002/art.23599] [Citation(s) in RCA: 54] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Abstract
OBJECTIVE Juvenile idiopathic arthritis (JIA) is an autoimmune disease of the young. The pathogenesis is not completely understood. Premature aging, associated thymic involution, and compensatory autoproliferation could play important roles in the pathogenesis of autoimmunity. We undertook this study to determine whether patients with JIA demonstrate premature immunosenescence. METHODS To test this hypothesis, we measured 3 indicators of aging: the percentages and total counts of peripheral blood naive T cells, the frequency of T cell receptor excision circles (TRECs) in naive T cells, and telomeric erosion and Ki-67 expression as estimates of the replicative history of homeostatic proliferation. RESULTS JIA patients showed an accelerated loss of CD4+,CD45RA+,CD62L+ naive T cells with advancing age and a compensatory increase in the number of CD4+,CD45RO+ memory T cells. JIA patients demonstrated a significantly decreased frequency of TRECs in CD4+,CD45RA+ naive T cells compared with age-matched healthy donors (P = 0.002). TREC numbers correlated with age only in healthy donors (P = 0.0001). Telomeric erosion in CD4+,CD45RA+ naive T cells was increased in JIA patients (P = 0.01). The percentages of Ki-67-positive CD4+,CD45RA+ naive T cells were increased in JIA patients (P = 0.001) and correlated with disease duration (P = 0.003), which was also an independent factor contributing to telomeric erosion (P = 0.04). CONCLUSION Our findings suggest that age-inappropriate T cell senescence and disturbed T cell homeostasis may contribute to the development of JIA. In patients with JIA, dysfunction in the ability to reconstitute the T cell compartment should be considered when exploring new therapeutic strategies.
Collapse
Affiliation(s)
- Martina Prelog
- Department of Pediatrics, Medical University Innsbruck, Innsbruck, Austria.
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
48
|
Miyazaki Y, Iwabuchi K, Kikuchi S, Fukazawa T, Niino M, Hirotani M, Sasaki H, Onoé K. Expansion of CD4+CD28- T cells producing high levels of interferon-{gamma} in peripheral blood of patients with multiple sclerosis. Mult Scler 2008; 14:1044-55. [PMID: 18573819 DOI: 10.1177/1352458508092809] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
CD4(+) T cells that lack surface expression of the CD28 co-stimulatory molecule (CD4(+)CD28(-) T cells) were expanded in peripheral blood of patients with multiple sclerosis (MS) [5.20 +/- 1.67% vs 13.00 +/- 2.68% (healthy controls (HC) versus patients with MS)]. Both the CD4(+)CD28(+) and CD4(+)CD28(-) T-cell populations of patients with MS produced higher levels of interferon (IFN)-gamma compared with those in HC. In particular, the proportion of IFN-gamma(+) cells among CD4(+)CD28(-) T cells from patients with MS was considerably high. However, expression of co-stimulatory molecules including inducible costimulator (ICOS), activating natural killer receptors, or members of tumor necrosis factor receptor family that replace CD28 in CD4(+)CD28(-) T cells of patients with MS could not be identified. A unique subpopulation bearing the CD45RA(high)CCR7(-) phenotype was identified among the CD4(+)CD28(-) T cells of some patients with MS. Because only MS samples contained this CD45RA(high)CCR7(-) population attributed to terminally differentiated effector memory cells and lacked naive CD45RA(high)CCR7(+) cells, we suggest that CD4(+)CD28(-) T cells of patients with MS represent a cell population which is in more differentiated state than healthy subjects. In patients treated with IFN-beta-1b, IFN-gamma production from CD4(+)CD28(+) T cells was suppressed compared with that in untreated patients. On the contrary, in the CD4(+)CD28(-) population, production of IFN-gamma in IFN-beta-1b-treated patients was not significantly suppressed compared with that in untreated patients with MS. Thus, an additional treatment strategy that specifically targets this cell population may enhance the beneficial effect of IFN-beta on MS.
Collapse
Affiliation(s)
- Y Miyazaki
- Division of Immunobiology, Research Section of Pathophysiology, Institute for Genetic Medicine, Hokkaido University, Sapporo, Japan
| | | | | | | | | | | | | | | |
Collapse
|
49
|
Fasth AER, Snir O, Johansson AAT, Nordmark B, Rahbar A, af Klint E, Björkström NK, Ulfgren AK, van Vollenhoven RF, Malmström V, Trollmo C. Skewed distribution of proinflammatory CD4+CD28null T cells in rheumatoid arthritis. Arthritis Res Ther 2008; 9:R87. [PMID: 17825098 PMCID: PMC2212553 DOI: 10.1186/ar2286] [Citation(s) in RCA: 67] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2007] [Revised: 08/23/2007] [Accepted: 09/07/2007] [Indexed: 12/20/2022] Open
Abstract
Expanded populations of CD4+ T cells lacking the co-stimulatory molecule CD28 (CD4+CD28null T cells) have been reported in several inflammatory disorders. In rheumatoid arthritis, increased frequencies of CD4+CD28null T cells in peripheral blood have previously been associated with extra-articular manifestations and human cytomegalovirus (HCMV) infection, but their presence in and contribution to joint manifestations is not clear. In the present article we investigated the distribution of CD4+CD28null T cells in the synovial membrane, synovial fluid and peripheral blood of RA patients, and analysed the association with erosive disease and anti-citrullinated protein antibodies. CD4+CD28null T cells were infrequent in the synovial membrane and synovial fluid, despite significant frequencies in the circulation. Strikingly, the dominant TCR-Vbeta subsets of CD4+CD28null T cells in peripheral blood were often absent in synovial fluid. CD4+CD28null T cells in blood and synovial fluid showed specificity for HCMV antigens, and their presence was clearly associated with HCMV seropositivity but not with anti-citrullinated protein antibodies in the serum or synovial fluid, nor with erosive disease. Together these data imply a primary role for CD4+CD28null T cells in manifestations elsewhere than in the joints of patients with HCMV-seropositive rheumatoid arthritis.
Collapse
Affiliation(s)
- Andreas ER Fasth
- Rheumatology Unit, Department of Medicine, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| | - Omri Snir
- Rheumatology Unit, Department of Medicine, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| | - Anna AT Johansson
- Rheumatology Unit, Department of Medicine, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| | - Birgitta Nordmark
- Rheumatology Unit, Department of Medicine, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| | - Afsar Rahbar
- Department of Medicine, Center for Molecular Medicine, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| | - Erik af Klint
- Rheumatology Unit, Department of Medicine, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| | - Niklas K Björkström
- Center for Infectious Medicine, Department of Medicine, Karolinska Institutet, Karolinska University Hospital Huddinge, Stockholm, Sweden
| | - Ann-Kristin Ulfgren
- Rheumatology Unit, Department of Medicine, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| | - Ronald F van Vollenhoven
- Rheumatology Unit, Department of Medicine, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| | - Vivianne Malmström
- Rheumatology Unit, Department of Medicine, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| | - Christina Trollmo
- Rheumatology Unit, Department of Medicine, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| |
Collapse
|
50
|
Söderberg-Nauclér C. HCMV microinfections in inflammatory diseases and cancer. J Clin Virol 2007; 41:218-23. [PMID: 18164235 DOI: 10.1016/j.jcv.2007.11.009] [Citation(s) in RCA: 116] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2007] [Revised: 10/22/2007] [Accepted: 11/05/2007] [Indexed: 01/10/2023]
Abstract
Human cytomegalovirus (HCMV) is a wide-spread human virus that was mainly known to cause disease in immunocompromised patients. A new entity of infection can be diagnosed with high sensitive techniques; HCMV microinfections that often exhibit an altered pattern of IE protein expression. We have recently discovered that HCMV microinfections are very common in patients with inflammatory diseases and certain cancers. The discovery of active HCMV infections in tissue specimens from patients with inflammatory diseases raises the question of whether the infection is an epiphenomenon or whether the virus plays a causative role in disease development. After a primary infection, which is generally asymptomatic in immunocompetent individuals, HCMV establishes latency and persists in its host. In infected cells, the virus can produce over 250 proteins, but only about 50-60 are believed to be essential for viral replication. Thus, the vast majority of these viral proteins enable the virus to co-exist with its host. Such proteins act through highly sophisticated mechanisms to control different cellular and immunological functions in order to facilitate viral production and to avoid detection and elimination of the virus by the immune system. These proteins may also contribute to the development of common inflammation-related diseases.
Collapse
Affiliation(s)
- Cecilia Söderberg-Nauclér
- Karolinska Insititutet, Center for Molecular Medicine, L8:03, Department of Medicine, Karolinska Hospital, SE-171 76 Stockholm, Sweden.
| |
Collapse
|