1
|
Hu H, Zhang G, Chen T, Liu Y, Meng L, Holmdahl R, Dai L, Zhao Y. Immunosenescence in autoimmune diseases. Autoimmun Rev 2025; 24:103805. [PMID: 40132774 DOI: 10.1016/j.autrev.2025.103805] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2025] [Revised: 03/21/2025] [Accepted: 03/21/2025] [Indexed: 03/27/2025]
Abstract
Autoimmune diseases (AIDs) are a group of disorders in which the immune system mistakenly attacks the body's own tissues, characterized by the loss of tolerance to self-antigens and destruction of tissues. Aging is a natural process of physiological decline that also alters the immune system, a condition known as immunosenescence. During immunosenescence, the immune system undergoes various changes, including modifications and antigenicity of self-antigens, abnormalities in the quantity, phenotype, and function of lymphocytes and antibodies, as well as a narrowing of the B and T cell receptor repertoire, changes that may increase susceptibility to AIDs. Additionally, senescent immune cells and the senescence-associated secretory phenotype (SASP) contribute to target organ involvement in AIDs, exacerbating chronic inflammation and tissue damage. Mitochondrial dysfunction and metabolic imbalances in AIDs lead to the accumulation of senescent cells, which act as upstream drivers of immunosenescence. In this review, we summarize the bidirectional relationship between AIDs and immunosenescence, as well as its potential mechanisms. Therapeutic approaches targeting immunosenescence in AIDs remain at an early stage. Strategies aimed at resetting or reversing the aging immune system are expected to become a novel direction in the future.
Collapse
Affiliation(s)
- Huifang Hu
- Department of Rheumatology and Immunology, West China Hospital, Sichuan University, 610041 Chengdu, Sichuan, China; Clinical Institute of Inflammation and Immunology, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, 610041 Chengdu, Sichuan, China
| | - Guangyue Zhang
- Department of Rheumatology and Immunology, West China Hospital, Sichuan University, 610041 Chengdu, Sichuan, China; Clinical Institute of Inflammation and Immunology, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, 610041 Chengdu, Sichuan, China
| | - Tao Chen
- Department of Rheumatology and Immunology, West China Hospital, Sichuan University, 610041 Chengdu, Sichuan, China; Clinical Institute of Inflammation and Immunology, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, 610041 Chengdu, Sichuan, China
| | - Yi Liu
- Department of Rheumatology and Immunology, West China Hospital, Sichuan University, 610041 Chengdu, Sichuan, China; Clinical Institute of Inflammation and Immunology, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, 610041 Chengdu, Sichuan, China
| | - Liesu Meng
- Department of Rheumatology, and National Joint Engineering Research Center of Biodiagnostics and Biotherapy, Second Affiliated Hospital, Xi'an Jiaotong University, Xi'an, Shaanxi 710004, PR China
| | - Rikard Holmdahl
- Medical Inflammation Research, Division of Immunology, Department of Medical Biochemistry and Biophysics, Karolinska Institute, Stockholm, Sweden
| | - Lunzhi Dai
- National Clinical Research Center for Geriatrics, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, 610041 Chengdu, Sichuan, China.
| | - Yi Zhao
- Department of Rheumatology and Immunology, West China Hospital, Sichuan University, 610041 Chengdu, Sichuan, China; Clinical Institute of Inflammation and Immunology, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, 610041 Chengdu, Sichuan, China.
| |
Collapse
|
2
|
Han F, Riaz F, Pu J, Gao R, Yang L, Wang Y, Song J, Liang Y, Wu Z, Li C, Tang J, Xu X, Wang X. Connecting the Dots: Telomere Shortening and Rheumatic Diseases. Biomolecules 2024; 14:1261. [PMID: 39456194 PMCID: PMC11506250 DOI: 10.3390/biom14101261] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2024] [Revised: 09/24/2024] [Accepted: 10/04/2024] [Indexed: 10/28/2024] Open
Abstract
Telomeres, repetitive sequences located at the extremities of chromosomes, play a pivotal role in sustaining chromosomal stability. Telomerase is a complex enzyme that can elongate telomeres by appending telomeric repeats to chromosome ends and acts as a critical factor in telomere dynamics. The gradual shortening of telomeres over time is a hallmark of cellular senescence and cellular death. Notably, telomere shortening appears to result from the complex interplay of two primary mechanisms: telomere shelterin complexes and telomerase activity. The intricate interplay of genetic, environmental, and lifestyle influences can perturb telomere replication, incite oxidative stress damage, and modulate telomerase activity, collectively resulting in shifts in telomere length. This age-related process of telomere shortening plays a considerable role in various chronic inflammatory and oxidative stress conditions, including cancer, cardiovascular disease, and rheumatic disease. Existing evidence has shown that abnormal telomere shortening or telomerase activity abnormalities are present in the pathophysiological processes of most rheumatic diseases, including different disease stages and cell types. The impact of telomere shortening on rheumatic diseases is multifaceted. This review summarizes the current understanding of the link between telomere length and rheumatic diseases in clinical patients and examines probable telomere shortening in peripheral blood mononuclear cells and histiocytes. Therefore, understanding the intricate interaction between telomere shortening and various rheumatic diseases will help in designing personalized treatment and control measures for rheumatic disease.
Collapse
Affiliation(s)
- Fang Han
- Department of Rheumatology and Immunology, Tongji Hospital, School of Medicine, Tongji University, No. 389 Xincun Road, Shanghai 200065, China; (F.H.); (J.P.); (R.G.); (L.Y.); (Y.W.); (J.S.); (Y.L.); (Z.W.); (C.L.); (J.T.)
| | - Farooq Riaz
- Faculty of Pharmaceutical Sciences, Shenzhen University of Advanced Technology, Shenzhen 518000, China;
- Center for Cancer Immunology, Shenzhen Institute of Advanced Technology (SIAT), Chinese Academy of Sciences (CAS), 1068 Xueyuan Avenue, Shenzhen 518055, China
| | - Jincheng Pu
- Department of Rheumatology and Immunology, Tongji Hospital, School of Medicine, Tongji University, No. 389 Xincun Road, Shanghai 200065, China; (F.H.); (J.P.); (R.G.); (L.Y.); (Y.W.); (J.S.); (Y.L.); (Z.W.); (C.L.); (J.T.)
| | - Ronglin Gao
- Department of Rheumatology and Immunology, Tongji Hospital, School of Medicine, Tongji University, No. 389 Xincun Road, Shanghai 200065, China; (F.H.); (J.P.); (R.G.); (L.Y.); (Y.W.); (J.S.); (Y.L.); (Z.W.); (C.L.); (J.T.)
| | - Lufei Yang
- Department of Rheumatology and Immunology, Tongji Hospital, School of Medicine, Tongji University, No. 389 Xincun Road, Shanghai 200065, China; (F.H.); (J.P.); (R.G.); (L.Y.); (Y.W.); (J.S.); (Y.L.); (Z.W.); (C.L.); (J.T.)
| | - Yanqing Wang
- Department of Rheumatology and Immunology, Tongji Hospital, School of Medicine, Tongji University, No. 389 Xincun Road, Shanghai 200065, China; (F.H.); (J.P.); (R.G.); (L.Y.); (Y.W.); (J.S.); (Y.L.); (Z.W.); (C.L.); (J.T.)
| | - Jiamin Song
- Department of Rheumatology and Immunology, Tongji Hospital, School of Medicine, Tongji University, No. 389 Xincun Road, Shanghai 200065, China; (F.H.); (J.P.); (R.G.); (L.Y.); (Y.W.); (J.S.); (Y.L.); (Z.W.); (C.L.); (J.T.)
| | - Yuanyuan Liang
- Department of Rheumatology and Immunology, Tongji Hospital, School of Medicine, Tongji University, No. 389 Xincun Road, Shanghai 200065, China; (F.H.); (J.P.); (R.G.); (L.Y.); (Y.W.); (J.S.); (Y.L.); (Z.W.); (C.L.); (J.T.)
| | - Zhenzhen Wu
- Department of Rheumatology and Immunology, Tongji Hospital, School of Medicine, Tongji University, No. 389 Xincun Road, Shanghai 200065, China; (F.H.); (J.P.); (R.G.); (L.Y.); (Y.W.); (J.S.); (Y.L.); (Z.W.); (C.L.); (J.T.)
| | - Chunrui Li
- Department of Rheumatology and Immunology, Tongji Hospital, School of Medicine, Tongji University, No. 389 Xincun Road, Shanghai 200065, China; (F.H.); (J.P.); (R.G.); (L.Y.); (Y.W.); (J.S.); (Y.L.); (Z.W.); (C.L.); (J.T.)
| | - Jianping Tang
- Department of Rheumatology and Immunology, Tongji Hospital, School of Medicine, Tongji University, No. 389 Xincun Road, Shanghai 200065, China; (F.H.); (J.P.); (R.G.); (L.Y.); (Y.W.); (J.S.); (Y.L.); (Z.W.); (C.L.); (J.T.)
| | - Xianghuai Xu
- Department of Pulmonary and Critical Care Medicine, Tongji Hospital, School of Medicine, Tongji University, No. 389 Xincun Road, Shanghai 200065, China;
| | - Xuan Wang
- Department of Rheumatology and Immunology, Tongji Hospital, School of Medicine, Tongji University, No. 389 Xincun Road, Shanghai 200065, China; (F.H.); (J.P.); (R.G.); (L.Y.); (Y.W.); (J.S.); (Y.L.); (Z.W.); (C.L.); (J.T.)
| |
Collapse
|
3
|
Shi J, Zhang M, Zhang L, Yu X, Sun L, Liu J, Zhao Y, Zheng W. Shelterin dysfunction promotes CD4+ T cell senescence in Behçet's disease. Rheumatology (Oxford) 2024; 63:2819-2827. [PMID: 38145496 DOI: 10.1093/rheumatology/kead703] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2023] [Revised: 11/26/2023] [Accepted: 12/16/2023] [Indexed: 12/27/2023] Open
Abstract
OBJECTIVES To investigate the potential role of shelterin dysfunction in naïve CD4+ T cells in the pathogenesis of Behçet's disease (BD). METHODS Naïve CD4+ T cells were isolated from 40 BD patients and 40 sex- and age-matched healthy controls (HC). Senescent profiles, shelterin subunits expression, telomere length, telomerase activity and critical DNA damage response (DDR) were evaluated. Telomere repeat factor-2 (TRF2) silencing was conducted for further validation. RESULTS Compared with HC, BD patients had significantly decreased naïve CD4+ T cells, increased cell apoptosis, senescence, and productions of TNF-α and IFN-γ upon activation. Notably, BD naïve CD4+ T cells had shortened telomere, impaired telomerase activity, and expressed lower levels of shelterin subunits TRF2, TRF1- and TRF2-Interacting Nuclear Protein 2 (TIN2) and Repressor/Activator Protein 1 (RAP1). Furthermore, BD naïve CD4+ T cells exhibited significantly increased DDR, evidenced by elevated phosphorylated ataxia telangiectasia (AT) mutated (pATM), phosphorylated p53 (pp53) and p21. Finally, TRF2 silencing markedly upregulated DDR, apoptosis and proinflammatory cytokines production in HC naïve CD4+ T cells. CONCLUSION Our study demonstrated that TRF2 deficiency in BD naïve CD4+ T cells promoted cell apoptosis and senescence, leading to proinflammatory cytokines overproduction. Therefore, restoring TRF2 might be a promising therapeutic strategy for BD.
Collapse
Affiliation(s)
- Jing Shi
- Department of Rheumatology and Clinical Immunology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College; National Clinical Research Center for Dermatologic and Immunologic Diseases, Ministry of Science & Technology, Beijing, China
- Department of Allergy, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Menghao Zhang
- Department of Rheumatology and Clinical Immunology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College; National Clinical Research Center for Dermatologic and Immunologic Diseases, Ministry of Science & Technology, Beijing, China
| | - Lili Zhang
- Department of Rheumatology, Linyi People's Hospital, Linyi, Shandong, China
| | - Xin Yu
- Department of Rheumatology and Clinical Immunology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College; National Clinical Research Center for Dermatologic and Immunologic Diseases, Ministry of Science & Technology, Beijing, China
| | - Luxi Sun
- Department of Respiratory and Critical Care Medicine, Peking Union Medical College Hospital, Beijing, China
| | - Jinjing Liu
- Department of Rheumatology and Clinical Immunology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College; National Clinical Research Center for Dermatologic and Immunologic Diseases, Ministry of Science & Technology, Beijing, China
| | - Yan Zhao
- Department of Rheumatology and Clinical Immunology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College; National Clinical Research Center for Dermatologic and Immunologic Diseases, Ministry of Science & Technology, Beijing, China
| | - Wenjie Zheng
- Department of Rheumatology and Clinical Immunology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College; National Clinical Research Center for Dermatologic and Immunologic Diseases, Ministry of Science & Technology, Beijing, China
| |
Collapse
|
4
|
Yu PJ, Zhou M, Liu Y, Du J. Senescent T Cells in Age-Related Diseases. Aging Dis 2024; 16:AD.2024.0219. [PMID: 38502582 PMCID: PMC11745454 DOI: 10.14336/ad.2024.0219] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2023] [Accepted: 02/18/2024] [Indexed: 03/21/2024] Open
Abstract
Age-induced alterations in human immunity are often considered deleterious and are referred to as immunosenescence. The immune system monitors the number of senescent cells in the body, while immunosenescence may represent the initiation of systemic aging. Immune cells, particularly T cells, are the most impacted and involved in age-related immune function deterioration, making older individuals more prone to different age-related diseases. T-cell senescence can impact the effectiveness of immunotherapies that rely on the immune system's function, including vaccines and adoptive T-cell therapies. The research and practice of using senescent T cells as therapeutic targets to intervene in age-related diseases are in their nascent stages. Therefore, in this review, we summarize recent related literature to investigate the characteristics of senescent T cells as well as their formation mechanisms, relationship with various aging-related diseases, and means of intervention. The primary objective of this article is to explore the prospects and possibilities of therapeutically targeting senescent T cells, serving as a valuable resource for the development of immunotherapy and treatment of age-related diseases.
Collapse
Affiliation(s)
- Pei-Jie Yu
- Beijing Anzhen Hospital, Capital Medical University
- The Key Laboratory of Remodeling-Related Cardiovascular Diseases, Ministry of Education
- Beijing Collaborative Innovative Research Center for Cardiovascular Diseases
- Beijing Institute of Heart Lung and Blood Vessel Diseases, Beijing 100029, China
| | - Mei Zhou
- Beijing Anzhen Hospital, Capital Medical University
- The Key Laboratory of Remodeling-Related Cardiovascular Diseases, Ministry of Education
- Beijing Collaborative Innovative Research Center for Cardiovascular Diseases
- Beijing Institute of Heart Lung and Blood Vessel Diseases, Beijing 100029, China
| | - Yan Liu
- Correspondence should be addressed to: Dr. Jie Du () and Dr. Yan Liu (), Beijing Anzhen Hospital, Capital Medical University, Beijing 100029, China
| | - Jie Du
- Correspondence should be addressed to: Dr. Jie Du () and Dr. Yan Liu (), Beijing Anzhen Hospital, Capital Medical University, Beijing 100029, China
| |
Collapse
|
5
|
Li D, Cao R, Dong W, Cheng M, Pan X, Hu Z, Hao J. Identification of potential biomarkers for ankylosing spondylitis based on bioinformatics analysis. BMC Musculoskelet Disord 2023; 24:413. [PMID: 37226132 DOI: 10.1186/s12891-023-06550-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/25/2023] [Accepted: 05/19/2023] [Indexed: 05/26/2023] Open
Abstract
OBJECTIVE The aim of this study was to search for key genes in ankylosing spondylitis (AS) through comprehensive bioinformatics analysis, thus providing some theoretical support for future diagnosis and treatment of AS and further research. METHODS Gene expression profiles were collected from Gene Expression Omnibus (GEO, http://www.ncbi.nlm.nih.gov/geo/ ) by searching for the term "ankylosing spondylitis". Ultimately, two microarray datasets (GSE73754 and GSE11886) were downloaded from the GEO database. A bioinformatic approach was used to screen differentially expressed genes and perform functional enrichment analysis to obtain biological functions and signalling pathways associated with the disease. Weighted correlation network analysis (WGCNA) was used to further obtain key genes. Immune infiltration analysis was performed using the CIBERSORT algorithm to conduct a correlation analysis of key genes with immune cells. The GWAS data of AS were analysed to identify the pathogenic regions of key genes in AS. Finally, potential therapeutic agents for AS were predicted using these key genes. RESULTS A total of 7 potential biomarkers were identified: DYSF, BASP1, PYGL, SPI1, C5AR1, ANPEP and SORL1. ROC curves showed good prediction for each gene. T cell, CD4 naïve cell, and neutrophil levels were significantly higher in the disease group than in the paired normal group, and key gene expression was strongly correlated with immune cells. CMap results showed that the expression profiles of ibuprofen, forskolin, bongkrek-acid, and cimaterol showed the most significant negative correlation with the expression profiles of disease perturbations, suggesting that these drugs may play a role in AS treatment. CONCLUSION The potential biomarkers of AS screened in this study are closely related to the level of immune cell infiltration and play an important role in the immune microenvironment. This may provide help in the clinical diagnosis and treatment of AS and provide new ideas for further research.
Collapse
Affiliation(s)
- Dongxu Li
- Department of Orthopedics, The First Affiliated Hospital of Chongqing Medical University, Yuzhong, Chongqing, China
- Orthopedic Laboratory of Chongqing Medical University, Yuzhong, Chongqing, China
| | - Ruichao Cao
- Department of Orthopedics, The First Affiliated Hospital of Chongqing Medical University, Yuzhong, Chongqing, China
- Orthopedic Laboratory of Chongqing Medical University, Yuzhong, Chongqing, China
| | - Wei Dong
- Department of Orthopedics, The First Affiliated Hospital of Chongqing Medical University, Yuzhong, Chongqing, China
- Orthopedic Laboratory of Chongqing Medical University, Yuzhong, Chongqing, China
| | - Minghuang Cheng
- Department of Orthopedics, The First Affiliated Hospital of Chongqing Medical University, Yuzhong, Chongqing, China
- Orthopedic Laboratory of Chongqing Medical University, Yuzhong, Chongqing, China
| | - Xiaohan Pan
- Department of Orthopedics, The First Affiliated Hospital of Chongqing Medical University, Yuzhong, Chongqing, China
- Orthopedic Laboratory of Chongqing Medical University, Yuzhong, Chongqing, China
| | - Zhenming Hu
- Department of Orthopedics, The First Affiliated Hospital of Chongqing Medical University, Yuzhong, Chongqing, China
- Orthopedic Laboratory of Chongqing Medical University, Yuzhong, Chongqing, China
| | - Jie Hao
- Department of Orthopedics, The First Affiliated Hospital of Chongqing Medical University, Yuzhong, Chongqing, China.
- Orthopedic Laboratory of Chongqing Medical University, Yuzhong, Chongqing, China.
| |
Collapse
|
6
|
Gudmundsdottir JA, Thorgeirsdottir S, Lundbäck V, Göngrich C, Lingman Framme J, Kindgren E, Rydenman K, Ludviksson BR, Bjarnadottir H, Runarsdottir S, Nilsson S, Zetterström RH, Ekwall O, Lindgren S. Normal neonatal TREC and KREC levels in early onset juvenile idiopathic arthritis. Clin Immunol 2023; 249:109277. [PMID: 36878420 DOI: 10.1016/j.clim.2023.109277] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2023] [Revised: 01/18/2023] [Accepted: 03/01/2023] [Indexed: 03/07/2023]
Abstract
OBJECTIVE Dysregulated central tolerance predisposes to autoimmune diseases. Reduced thymic output as well as compromised central B cell tolerance checkpoints have been proposed in the pathogenesis of juvenile idiopathic arthritis (JIA). The aim of this study was to investigate neonatal levels of T-cell receptor excision circles (TRECs) and kappa-deleting element excision circles (KRECs), as markers of T- and B-cell output at birth, in patients with early onset JIA. METHODS TRECs and KRECs were quantitated by multiplex qPCR from dried blood spots (DBS), collected 2-5 days after birth, in 156 children with early onset JIA and in 312 matched controls. RESULTS When analysed from neonatal dried blood spots, the median TREC level was 78 (IQR 55-113) in JIA cases and 88 (IQR 57-117) copies/well in controls. The median KREC level was 51 (IQR 35-69) and 53 (IQR 35-74) copies/well, in JIA cases and controls, respectively. Stratification by sex and age at disease onset did not reveal any difference in the levels of TRECs and KRECs. CONCLUSION T- and B-cell output at birth, as measured by TREC and KREC levels in neonatal dried blood spots, does not differ in children with early onset JIA compared to controls.
Collapse
Affiliation(s)
- Judith A Gudmundsdottir
- Children's Medical Center, Landspitali, The National University Hospital, Reykjavik, Iceland
| | - Sigridur Thorgeirsdottir
- Department of Pediatrics, Institute of Clinical Sciences, The Sahlgrenska Academy at University of Gothenburg, Gothenburg, Sweden
| | - Veroniqa Lundbäck
- Centre for Inherited Metabolic Diseases, Karolinska University Hospital, Stockholm, Sweden; Department of Molecular Medicine and Surgery, Karolinska Institute, Stockholm, Sweden
| | - Christina Göngrich
- Centre for Inherited Metabolic Diseases, Karolinska University Hospital, Stockholm, Sweden; Department of Molecular Medicine and Surgery, Karolinska Institute, Stockholm, Sweden
| | - Jenny Lingman Framme
- Department of Pediatrics, Institute of Clinical Sciences, The Sahlgrenska Academy at University of Gothenburg, Gothenburg, Sweden; The Department of Pediatrics, Halland Hospital Halmstad, Halmstad, Region Halland, Sweden
| | - Erik Kindgren
- Division of Pediatrics, Department of Biomedical and Clinical Sciences, Linköping University, Linköping, Sweden; Department of Pediatrics, Skaraborgs Hospital Skövde, Region Västra Götaland, Sweden
| | - Karin Rydenman
- Department of Pediatrics, Institute of Clinical Sciences, The Sahlgrenska Academy at University of Gothenburg, Gothenburg, Sweden
| | - Bjorn Runar Ludviksson
- Department of Immunology, Landspitali, The National University Hospital of Iceland, Reykjavik, Iceland
| | - Helga Bjarnadottir
- Department of Immunology, Landspitali, The National University Hospital of Iceland, Reykjavik, Iceland
| | - Saga Runarsdottir
- Department of Genetics and Molecular Medicine, Landspitali, The National University Hospital of Iceland, Reykjavik, Iceland
| | - Staffan Nilsson
- Department of Mathematical Sciences, Chalmers University of Technology, Gothenburg, Sweden; Department of Laboratory Medicine, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Rolf H Zetterström
- Centre for Inherited Metabolic Diseases, Karolinska University Hospital, Stockholm, Sweden; Department of Molecular Medicine and Surgery, Karolinska Institute, Stockholm, Sweden
| | - Olov Ekwall
- Department of Pediatrics, Institute of Clinical Sciences, The Sahlgrenska Academy at University of Gothenburg, Gothenburg, Sweden; Department of Rheumatology and Inflammation Research, The Sahlgrenska Academy at University of Gothenburg, Gothenburg, Sweden
| | - Susanne Lindgren
- Department of Pediatrics, Institute of Clinical Sciences, The Sahlgrenska Academy at University of Gothenburg, Gothenburg, Sweden; Department of Rheumatology and Inflammation Research, The Sahlgrenska Academy at University of Gothenburg, Gothenburg, Sweden.
| |
Collapse
|
7
|
Hodl I, Sallegger C, Forstner P, Sareban N, Moritz M, Dreo B, Schulz E, Lackner A, Kleinhappl B, Hatzl S, Moazedi-Fürst F, Seifert-Held T, Heschl B, Khalil M, Enzinger C, Greinix H, Stradner MH, Steinmetz I, Schlenke P, Fessler J. Altered cellular immune response to vaccination against SARS-CoV-2 in patients suffering from autoimmunity with B-cell depleting therapy. Microbes Infect 2023; 25:105103. [PMID: 36681177 PMCID: PMC9850844 DOI: 10.1016/j.micinf.2023.105103] [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: 09/26/2022] [Revised: 01/11/2023] [Accepted: 01/12/2023] [Indexed: 01/20/2023]
Abstract
B-cell depleting therapies result in diminished humoral immunity following vaccination against COVID-19, but our understanding on the impact on cellular immune responses is limited. Here, we performed a detailed analysis of cellular immunity following mRNA vaccination in patients receiving B-cell depleting therapy using ELISpot assay and flow cytometry. Anti-SARS-CoV-2 spike receptor-binding domain antibody assays were performed to elucidate B-cell responses. To complement our cellular analysis, we performed immunophenotyping for T- and B-cell subsets. We show that SARS-CoV-2 vaccination using mRNA vaccines elicits cellular T-cell responses in patients under B-cell depleting therapy. Some facets of this immune response including TNFα production of CD4+ T-cells and granzyme B production of CD8+ T-cells, however, are distinctly diminished in these patients. Consequently, it appears that the finely coordinated process of T-cell activation with a uniform involvement of CD4+ and CD8+ T-cells as seen in HCs is disturbed in autoimmune patients. In addition, we observed that immune cell composition does impact cellular immunity as well as sustainability of anti-spike antibody titers. Our data suggest disturbed cellular immunity following mRNA vaccination in patients treated with B-cell depleting therapy. Immune cell composition may be an important determinant for vaccination efficacy.
Collapse
Affiliation(s)
- Isabel Hodl
- Division of Rheumatology and Immunology, Department of Internal Medicine, Medical University of Graz, Graz, Austria
| | - Clarissa Sallegger
- Division of Rheumatology and Immunology, Department of Internal Medicine, Medical University of Graz, Graz, Austria
| | - Patrick Forstner
- Institute of Hygiene, Microbiology and Environmental Medicine, Medical University of Graz, Graz, Austria
| | - Nazanin Sareban
- Department of Blood Group Serology and Transfusion Medicine, Medical University of Graz, Graz, Austria
| | - Martina Moritz
- Department of Blood Group Serology and Transfusion Medicine, Medical University of Graz, Graz, Austria
| | - Barbara Dreo
- Division of Rheumatology and Immunology, Department of Internal Medicine, Medical University of Graz, Graz, Austria
| | - Eduard Schulz
- Division of Hematology, Department of Internal Medicine, Medical University of Graz, Graz, Austria,Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, United States
| | - Angelika Lackner
- Division of Rheumatology and Immunology, Department of Internal Medicine, Medical University of Graz, Graz, Austria
| | - Barbara Kleinhappl
- Institute of Hygiene, Microbiology and Environmental Medicine, Medical University of Graz, Graz, Austria
| | - Stefan Hatzl
- Division of Hematology, Department of Internal Medicine, Medical University of Graz, Graz, Austria
| | - Florentine Moazedi-Fürst
- Division of Rheumatology and Immunology, Department of Internal Medicine, Medical University of Graz, Graz, Austria
| | | | - Bettina Heschl
- Department of Neurology, Medical University of Graz, Graz, Austria
| | - Michael Khalil
- Department of Neurology, Medical University of Graz, Graz, Austria
| | | | - Hildegard Greinix
- Division of Hematology, Department of Internal Medicine, Medical University of Graz, Graz, Austria
| | - Martin H. Stradner
- Division of Rheumatology and Immunology, Department of Internal Medicine, Medical University of Graz, Graz, Austria,Corresponding author. Division of Rheumatology and Immunology Department of Internal Medicine Medical University of Graz Auenbruggerplatz 15, 8036 Graz, Austria. Tel.: +43/316/385-81794; fax: +43/316/385-17813
| | - Ivo Steinmetz
- Institute of Hygiene, Microbiology and Environmental Medicine, Medical University of Graz, Graz, Austria
| | - Peter Schlenke
- Department of Blood Group Serology and Transfusion Medicine, Medical University of Graz, Graz, Austria
| | - Johannes Fessler
- Division of Immunology, Medical University of Graz, Graz, Austria
| |
Collapse
|
8
|
Wei D, Jiang Y, Cheng J, Wang H, Sha K, Zhao J. Assessing the association of leukocyte telomere length with ankylosing spondylitis and rheumatoid arthritis: A bidirectional Mendelian randomization study. Front Immunol 2023; 14:1023991. [PMID: 37033949 PMCID: PMC10080099 DOI: 10.3389/fimmu.2023.1023991] [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: 08/20/2022] [Accepted: 03/08/2023] [Indexed: 04/11/2023] Open
Abstract
Background Telomere length shortening can cause senescence and apoptosis in various immune cells, resulting in immune destabilization and ageing of the organism. In this study, we aimed to systematically assess the causal relationship of leukocyte telomere length (LTL) with ankylosing spondylitis (AS) and rheumatoid arthritis (RA) using a Mendelian randomization study. Methods LTL (n=472174) was obtained from the UK Biobank genome-wide association study pooled data. AS (n=229640), RA (n=212472) were obtained from FinnGen database. MR-Egger, inverse variance weighting, and weighted median methods were used to estimate the effects of causes. Cochran's Q test, MR Egger intercept test, MR-PRESSO, leave-one-out analysis, and funnel plots were used to look at sensitivity, heterogeneity, and multiple effects. Forward MR analysis considered LTL as the exposure and AS, RA as the outcome. Reverse MR analysis considered AS, RA as the exposure and LTL as the outcome. Results In the forward MR analysis, inverse variance-weighted and weighted median analysis results indicated that longer LTL might be associated with increased risk of AS (IVW: OR = 1.55, 95% CI: 1.14-2.11, p = 0.006). MR Egger regression analysis showed no pleiotropy between instrumental variables (IVs) (Egger intercept= 0.008, p = 0.294). The leave-one-out analysis showed that each single nucleotide polymorphism (SNP) of AS was robust to each outcome. No significant causal effects were found between AS, RA and LTL in the reverse MR analysis. Conclusion Longer LTL may be related with an increased risk of developing AS, and these findings provide a foundation for future clinical research on the causal association between LTL and AS.
Collapse
Affiliation(s)
- Donglei Wei
- Department of Orthopedic Trauma and Hand Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China
- Guangxi Key Laboratory of Regenerative Medicine, Orthopaedic Department, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China
| | - Yage Jiang
- Department of Anesthesiology, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China
| | - Jianwen Cheng
- Department of Orthopedic Trauma and Hand Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China
| | - Hui Wang
- Department of Orthopedic Trauma and Hand Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China
| | - Ke Sha
- Department of Orthopedic Trauma and Hand Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China
| | - Jinmin Zhao
- Department of Orthopedic Trauma and Hand Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China
- Guangxi Key Laboratory of Regenerative Medicine, Orthopaedic Department, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China
- *Correspondence: Jinmin Zhao,
| |
Collapse
|
9
|
Liu M, Luo P, Liu L, Wei X, Bai X, Li J, Wu L, Luo M. Immune-mediated inflammatory diseases and leukocyte telomere length: A Mendelian randomization study. Front Genet 2023; 14:1129247. [PMID: 37139230 PMCID: PMC10150136 DOI: 10.3389/fgene.2023.1129247] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2022] [Accepted: 03/20/2023] [Indexed: 05/05/2023] Open
Abstract
Objective: To elucidate the potential causality of leukocyte telomere length (LTL) with immune-mediated inflammatory diseases (IMIDs), we conducted a Mendelian randomization (MR) study. Methods: The genetically predicted causation between LTL and IMIDs was evaluated using a two-sample MR method. We analyzed 16 major IMIDs, which included systemic lupus erythematosus (SLE), inflammatory bowel disease (IBD), ulcerative colitis (UC), Crohn's disease (CD), ankylosing spondylitis (AS), sicca syndrome (SS), rheumatoid arthritis (RA), type 1 diabetes (T1D), primary sclerosing cholangitis (PSC), idiopathic pulmonary fibrosis (IPF), atopic dermatitis (AD), sarcoidosis, hypothyroidism, hyperthyroidism, psoriasis, and childhood asthma. The random-effects inverse-variance weighted (IVW) method was performed as the main analytical approach in MR. Various sensitivity analyses, including MR-Egger, MR robust adjusted profile score (MR-RAPS), weighted median, MR pleiotropy residual sum and outlier (MR-PRESSO) methods, weighted mode, radial plot, and radial regression, were used to guarantee the robustness of the results and detect horizontal pleiotropy. Cochran's Q value was calculated to check for heterogeneity, and the MR Steiger approach was used to test the causal direction. Results: The MR results indicated significant inverse associations of LTL with risks of psoriasis (OR: 0.77, 95% CI: 0.66-0.89, and p = 3.66 × 10-4), SS (OR: 0.75, CI: 0.58-0.98, and p = 0.03), RA (OR: 0.77, 95% CI: 0.68-0.88, and p = 9.85 × 10-5), hypothyroidism (OR: 0.84, 95% CI: 0.78-0.91, and p = 7,08 × 10-6), hyperthyroidism (OR: 0.60, 95% CI: 0.44-0.83, and p = 1.90 × 10-3), sarcoidosis (OR: 0.67, 95% CI: 0.54-0.83, and p = 2.60 × 10-4), and IPF (OR: 0.41, 95% CI: 0.29-0.58, and p = 4.11 × 10-7) in the FinnGen study. We observed that longer LTL was associated with an increased risk of AS susceptibility (OR: 1.51, 95% CI: 1.18-1.94, and p = 9.66 × 10-4). The results of the IVW method showed no causal relationship between TL and SLE (OR: 0.92, 95% CI: 0.62-1.38, and p = 0.69) in the FinnGen study; however, a significantly positive correlation was shown between LTL and SLE in another larger GWAS (OR: 1.87, 95% CI: 1.37-2.54, and p = 8.01 × 10-5). Conclusion: Our findings reveal that abnormal LTL has the potential to increase the risk of IMIDs. Therefore, it could be treated as a predictor and may provide new potential treatment targets for IMIDs. However, the change of LTL may not be the direct cause of IMIDs. Further studies should aim at the pathogenic mechanism or potential protective effects of LTL in IMIDs.
Collapse
|
10
|
Chen C, Wang P, Zhang RD, Fang Y, Jiang LQ, Fang X, Zhao Y, Wang DG, Ni J, Pan HF. Mendelian randomization as a tool to gain insights into the mosaic causes of autoimmune diseases. Clin Exp Rheumatol 2022; 21:103210. [PMID: 36273526 DOI: 10.1016/j.autrev.2022.103210] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2022] [Revised: 10/06/2022] [Accepted: 10/11/2022] [Indexed: 12/14/2022]
Abstract
Autoimmune diseases (ADs) are a broad range of disorders which are characterized by long-term inflammation and tissue damage arising from an immune response against one's own tissues. It is now widely accepted that the causes of ADs include environmental factors, genetic susceptibility and immune dysregulation. However, the exact etiology of ADs has not been fully elucidated to date. Because observational studies are plagued by confounding factors and reverse causality, no firm conclusions can be drawn about the etiology of ADs. Over the years, Mendelian randomization (MR) analysis has come into focus, offering unique perspectives and insights into the etiology of ADs and promising the discovery of potential therapeutic interventions. In MR analysis, genetic variation (alleles are randomly dispensed during meiosis, usually irrespective of environmental or lifestyle factors) is used instead of modifiable exposure to explore the link between exposure factors and disease or other outcomes. Therefore, MR analysis can provide a valuable method for exploring the causal relationship between different risk factors and ADs when its inherent assumptions and limitations are fully considered. This review summarized the recent findings of MR in major ADs, including systemic lupus erythematosus (SLE), rheumatoid arthritis (RA), multiple sclerosis (MS), and type 1 diabetes mellitus (T1DM), focused on the effects of different risk factors on ADs risks. In addition, we also discussed the opportunities and challenges of MR methods in ADs research.
Collapse
Affiliation(s)
- Cong Chen
- Department of Epidemiology and Biostatistics, School of Public Health, Anhui Medical University, 81 Meishan Road, Hefei 230032, Anhui, China; Institute of Kidney Disease, Inflammation & Immunity Mediated Diseases, The Second Hospital of Anhui Medical University, China
| | - Peng Wang
- Institute of Kidney Disease, Inflammation & Immunity Mediated Diseases, The Second Hospital of Anhui Medical University, China; Teaching Center for Preventive Medicine, School of Public Health, Anhui Medical University, 81 Meishan Road, Hefei 230032, Anhui, China
| | - Ruo-Di Zhang
- Department of Epidemiology and Biostatistics, School of Public Health, Anhui Medical University, 81 Meishan Road, Hefei 230032, Anhui, China; Institute of Kidney Disease, Inflammation & Immunity Mediated Diseases, The Second Hospital of Anhui Medical University, China
| | - Yang Fang
- Department of Epidemiology and Biostatistics, School of Public Health, Anhui Medical University, 81 Meishan Road, Hefei 230032, Anhui, China; Institute of Kidney Disease, Inflammation & Immunity Mediated Diseases, The Second Hospital of Anhui Medical University, China
| | - Ling-Qiong Jiang
- Department of Epidemiology and Biostatistics, School of Public Health, Anhui Medical University, 81 Meishan Road, Hefei 230032, Anhui, China; Institute of Kidney Disease, Inflammation & Immunity Mediated Diseases, The Second Hospital of Anhui Medical University, China
| | - Xi Fang
- Department of Epidemiology and Biostatistics, School of Public Health, Anhui Medical University, 81 Meishan Road, Hefei 230032, Anhui, China; Institute of Kidney Disease, Inflammation & Immunity Mediated Diseases, The Second Hospital of Anhui Medical University, China
| | - Yan Zhao
- Department of Epidemiology and Biostatistics, School of Public Health, Anhui Medical University, 81 Meishan Road, Hefei 230032, Anhui, China; Institute of Kidney Disease, Inflammation & Immunity Mediated Diseases, The Second Hospital of Anhui Medical University, China
| | - De-Guang Wang
- Institute of Kidney Disease, Inflammation & Immunity Mediated Diseases, The Second Hospital of Anhui Medical University, China; Department of Nephrology, The Second Hospital of Anhui Medical University, Hefei, China.
| | - Jing Ni
- Department of Epidemiology and Biostatistics, School of Public Health, Anhui Medical University, 81 Meishan Road, Hefei 230032, Anhui, China; Institute of Kidney Disease, Inflammation & Immunity Mediated Diseases, The Second Hospital of Anhui Medical University, China.
| | - Hai-Feng Pan
- Department of Epidemiology and Biostatistics, School of Public Health, Anhui Medical University, 81 Meishan Road, Hefei 230032, Anhui, China; Institute of Kidney Disease, Inflammation & Immunity Mediated Diseases, The Second Hospital of Anhui Medical University, China.
| |
Collapse
|
11
|
Gao Y, Cai W, Zhou Y, Li Y, Cheng J, Wei F. Immunosenescence of T cells: a key player in rheumatoid arthritis. Inflamm Res 2022; 71:1449-1462. [DOI: 10.1007/s00011-022-01649-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2022] [Revised: 09/12/2022] [Accepted: 09/15/2022] [Indexed: 11/05/2022] Open
|
12
|
Ao YQ, Jiang JH, Gao J, Wang HK, Ding JY. Recent thymic emigrants as the bridge between thymoma and autoimmune diseases. Biochim Biophys Acta Rev Cancer 2022; 1877:188730. [DOI: 10.1016/j.bbcan.2022.188730] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2022] [Revised: 04/14/2022] [Accepted: 04/15/2022] [Indexed: 11/27/2022]
|
13
|
Guo G, Watterson S, Zhang SD, Bjourson A, McGilligan V, Peace A, Rai TS. The role of senescence in the pathogenesis of atrial fibrillation: A target process for health improvement and drug development. Ageing Res Rev 2021; 69:101363. [PMID: 34023420 DOI: 10.1016/j.arr.2021.101363] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2020] [Revised: 01/24/2021] [Accepted: 05/12/2021] [Indexed: 12/11/2022]
Abstract
Cellular senescence is a state of growth arrest that occurs after cells encounter various stresses. Senescence contributes to tumour suppression, embryonic development, and wound healing. It impacts on the pathology of various diseases by secreting inflammatory chemokines, immune modulators and other bioactive factors. These secretory biosignatures ultimately cause inflammation, tissue fibrosis, immunosenescence and many ageing-related diseases such as atrial fibrillation (AF). Because the molecular mechanisms underpinning AF development remain unclear, current treatments are suboptimal and have serious side effects. In this review, we summarize recent results describing the role of senescence in AF. We propose that senescence factors induce AF and have a causative role. Hence, targeting senescence and its secretory phenotype may attenuate AF.
Collapse
|
14
|
Fessler J, Angiari S. The Role of T Cell Senescence in Neurological Diseases and Its Regulation by Cellular Metabolism. Front Immunol 2021; 12:706434. [PMID: 34335619 PMCID: PMC8317490 DOI: 10.3389/fimmu.2021.706434] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2021] [Accepted: 06/28/2021] [Indexed: 12/28/2022] Open
Abstract
Immunosenescence is a state of dysregulated leukocyte function characterised by arrested cell cycle, telomere shortening, expression of markers of cellular stress, and secretion of pro-inflammatory mediators. Immunosenescence principally develops during aging, but it may also be induced in other pathological settings, such as chronic viral infections and autoimmune diseases. Appearance of senescent immune cells has been shown to potentially cause chronic inflammation and tissue damage, suggesting an important role for this process in organismal homeostasis. In particular, the presence of senescent T lymphocytes has been reported in neurological diseases, with some works pointing towards a direct connection between T cell senescence, inflammation and neuronal damage. In this minireview, we provide an overview on the role of T cell senescence in neurological disorders, in particular in multiple sclerosis and Alzheimer disease. We also discuss recent literature investigating how metabolic remodelling controls the development of a senescence phenotype in T cells. Targeting metabolic pathways involved in the induction of senescent T cells may indeed represent a novel approach to limit their inflammatory activity and prevent neuroinflammation and neurodegeneration.
Collapse
Affiliation(s)
- Johannes Fessler
- Division of Immunology and Pathophysiology, Otto Loewi Research Center, Medical University of Graz, Graz, Austria
| | - Stefano Angiari
- Division of Immunology and Pathophysiology, Otto Loewi Research Center, Medical University of Graz, Graz, Austria
| |
Collapse
|
15
|
Telomeres: New players in immune-mediated inflammatory diseases? J Autoimmun 2021; 123:102699. [PMID: 34265700 DOI: 10.1016/j.jaut.2021.102699] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2021] [Revised: 07/02/2021] [Accepted: 07/05/2021] [Indexed: 12/21/2022]
Abstract
Telomeres are repetitive DNA sequences located at the ends of linear chromosomes that preserve the integrity and stability of the genome. Telomere dysfunctions due to short telomeres or altered telomere structures can ultimately lead to replicative cellular senescence and chromosomal instability, both mechanisms being hallmarks of ageing. Chronic inflammation, oxidative stress and finally telomere length (TL) dynamics have been shown to be involved in various age-related non-communicable diseases (NCDs). Immune-mediated inflammatory diseases (IMIDs), including affections such as inflammatory bowel disease, psoriasis, rheumatoid arthritis, spondyloarthritis and uveitis belong to this group of age-related NCDs. Although in recent years, we have witnessed the emergence of studies in the literature linking these IMIDs to TL dynamics, the causality between these diseases and telomere attrition is still unclear and controversial. In this review, we provide an overview of available studies on telomere dynamics and discuss the utility of TL measurements in immune-mediated inflammatory diseases.
Collapse
|
16
|
Fessler J, Fasching P, Raicht A, Hammerl S, Weber J, Lackner A, Hermann J, Dejaco C, Graninger WB, Schwinger W, Stradner MH. Lymphopenia in primary Sjögren's syndrome is associated with premature aging of naïve CD4+ T cells. Rheumatology (Oxford) 2021; 60:588-597. [PMID: 32227243 DOI: 10.1093/rheumatology/keaa105] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2019] [Revised: 02/13/2020] [Indexed: 11/12/2022] Open
Abstract
OBJECTIVE To investigate peripheral lymphopenia, a frequent finding in primary Sjögren's syndrome (pSS) associated with higher disease activity and increased mortality. METHODS Prospective, cross-sectional study of consecutive patients with pSS (n = 66) and healthy controls (n = 181). Lymphocyte subsets were analysed by flow cytometry, naïve (CD45RA+) and memory (CD45RO+) CD4+ T cells were purified by MACS technology. In vitro proliferation and senescence-associated β-galactosidase (SABG) were assessed by flow cytometry. Telomere length and TCR excision circles (TREC) were measured by real-time PCR. Telomerase activity was analysed according to the telomeric repeat amplification protocols (TRAP). RESULTS In pSS, lymphopenia mainly affected naïve CD4+ T cells. We noted a lower frequency of proliferating naïve CD4+ T cells ex vivo and decreased homeostatic proliferation in response to IL-7 stimulation in vitro. Furthermore, naïve CD4+ T cells exhibited signs of immune cell aging including shortened telomeres, a reduction in IL-7R expression and accumulation of SABG. The senescent phenotype could be explained by telomerase insufficiency and drastically reduced levels of T-cell receptor excision circles (TRECs), indicating a history of extensive post-thymic cell division. TRECs correlated with the number of naïve CD4+ T cells linking the extend of earlier proliferation to the inability to sustain normal cell numbers. CONCLUSION In pSS, evidence for increased proliferation of naïve CD4+ T cells earlier in life is associated with a senescent phenotype unable to sustain homeostasis. The lack of naïve CD4+ T cells forms the basis of lymphopenia frequently observed in pSS.
Collapse
Affiliation(s)
- Johannes Fessler
- Department of Rheumatology and Immunology, Medical University of Graz, Graz, Austria.,Department of Neurology, Harvard Medical School, Brigham and Women's Hospital, Harvard, MA, USA
| | - Patrizia Fasching
- Department of Rheumatology and Immunology, Medical University of Graz, Graz, Austria
| | - Andrea Raicht
- Department of Pediatric Hemato-Oncology, Medical University of Graz, Graz, Austria
| | - Sabrina Hammerl
- Department of Rheumatology and Immunology, Medical University of Graz, Graz, Austria
| | - Jennifer Weber
- Department of Rheumatology and Immunology, Medical University of Graz, Graz, Austria
| | - Angelika Lackner
- Department of Rheumatology and Immunology, Medical University of Graz, Graz, Austria
| | - Josef Hermann
- Department of Rheumatology and Immunology, Medical University of Graz, Graz, Austria
| | - Christian Dejaco
- Department of Rheumatology and Immunology, Medical University of Graz, Graz, Austria.,Servizio di Reumatologia, Azienda Sanitaria dell'Alto Adige, Ospedale di Brunico, Brunico, Italy
| | - Winfried B Graninger
- Department of Rheumatology and Immunology, Medical University of Graz, Graz, Austria
| | - Wolfgang Schwinger
- Department of Pediatric Hemato-Oncology, Medical University of Graz, Graz, Austria
| | - Martin H Stradner
- Department of Rheumatology and Immunology, Medical University of Graz, Graz, Austria
| |
Collapse
|
17
|
Hoffmann-Vold AM, Maher TM, Philpot EE, Ashrafzadeh A, Distler O. Assessment of recent evidence for the management of patients with systemic sclerosis-associated interstitial lung disease: a systematic review. ERJ Open Res 2021; 7:00235-2020. [PMID: 33644224 PMCID: PMC7897846 DOI: 10.1183/23120541.00235-2020] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2020] [Accepted: 12/09/2020] [Indexed: 12/12/2022] Open
Abstract
This systematic review summarises current evidence to help guide treatment decisions for patients with systemic sclerosis (SSc)-associated interstitial lung disease (ILD). A systematic search of the literature (January 2012 to April 2018), including grey literature (searched between 1992 and 2011), was conducted using multiple electronic databases. Guidelines, meta-analyses, randomised controlled trials and observational studies reporting on risk stratification, screening, diagnosis, treatment and management outcomes for patients with SSc-ILD were included. A quality assessment of the included evidence was undertaken. In total, 2464 publications were identified and 280 included. Multiple independent risk factors for ILD in patients with SSc were identified, including older age, male sex and baseline pulmonary function. High-resolution computed tomography (HRCT) has been used for characterising ILD in patients with SSc, and pulmonary function tests are a key adjunctive component in the diagnostic and monitoring pathway. The clinical value of biomarkers relating to SSc-ILD diagnosis or assessment for disease progression is unknown at present. Immunosuppressive therapy (monotherapy or combined therapy) is the current standard of care for SSc-ILD; long-term evidence for effective and safe treatment of SSc-ILD is limited. Identification of patients at risk for SSc-ILD remains challenging. HRCT and pulmonary function tests are key to diagnosing and monitoring for disease progression. Although immunosuppressive therapy is considered current first-line treatment, it is partly associated with adverse effects and long-term follow-up evidence is limited. Novel therapies and biomarkers should be further explored in well-controlled clinical studies.
Collapse
Affiliation(s)
- Anna-Maria Hoffmann-Vold
- Dept of Rheumatology, Oslo University Hospital-Rikshospitalet, Oslo, Norway
- These authors contributed equally
| | - Toby M. Maher
- National Institute of Health Research Respiratory Clinical Research Facility, Royal Brompton Hospital, London, UK
- Fibrosis Research Group, National Heart and Lung Institute, Imperial College London, London, UK
- These authors contributed equally
| | | | - Ali Ashrafzadeh
- Rheumatology Center of Excellence, IQVIA, San Diego, CA, USA
| | - Oliver Distler
- Dept of Rheumatology, University Hospital Zurich, Zurich, Switzerland
| |
Collapse
|
18
|
Souto-Carneiro MM, Klika KD, Abreu MT, Meyer AP, Saffrich R, Sandhoff R, Jennemann R, Kraus FV, Tykocinski L, Eckstein V, Carvalho L, Kriegsmann M, Giese T, Lorenz HM, Carvalho RA. Effect of Increased Lactate Dehydrogenase A Activity and Aerobic Glycolysis on the Proinflammatory Profile of Autoimmune CD8+ T Cells in Rheumatoid Arthritis. Arthritis Rheumatol 2020; 72:2050-2064. [PMID: 32602217 DOI: 10.1002/art.41420] [Citation(s) in RCA: 70] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2019] [Accepted: 06/23/2020] [Indexed: 12/11/2022]
Abstract
OBJECTIVE CD8+ T cells contribute to rheumatoid arthritis (RA) by releasing proinflammatory and cytolytic mediators, even in a challenging hypoxic and nutrient-poor microenvironment such as the synovial membrane. This study was undertaken to explore the mechanisms through which CD8+ T cells meet their metabolic demands in the blood and synovial membrane of patients with RA. METHODS Purified blood CD8+ T cells from patients with RA, patients with psoriatic arthritis (PsA), and patients with spondyloarthritis (SpA), as well as healthy control subjects, and CD8+ T cells from RA synovial membrane were stimulated in medium containing 13 C-labeled metabolic substrates in the presence or absence of metabolic inhibitors, under conditions of normoxia or hypoxia. The production of metabolic intermediates was quantified by 1 H-nuclear magnetic resonance. The expression of metabolic enzymes, transcription factors, and immune effector molecules was assessed at both the messenger RNA (mRNA) and protein levels. CD8+ T cell functional studies were performed. RESULTS RA blood CD8+ T cells met their metabolic demands through aerobic glycolysis, production of uniformly 13 C-enriched lactate in the RA blood (2.6 to 3.7-fold higher than in patients with SpA, patients with PsA, and healthy controls; P < 0.01), and induction of glutaminolysis. Overexpression of Warburg effect-linked enzymes in all RA CD8+ T cell subsets maintained this metabolic profile, conferring to the cells the capacity to proliferate under hypoxia and low-glucose conditions. In all RA CD8+ T cell subsets, lactate dehydrogenase A (LDHA) was overexpressed at the mRNA level (P < 0.03 versus controls; n = 6 per group) and protein level (P < 0.05 versus controls; n = 17 RA patients, n = 9 controls). In RA blood, inhibition of LDHA with FX11 led to reductions in lipogenesis, migration and proliferation of CD8+ T cells, and CD8+ T cell effector functions, while production of reactive oxygen species was increased by 1.5-fold (P < 0.03 versus controls). Following inhibition of LDHA with FX11, RA CD8+ T cells lost their capacity to induce healthy B cells to develop a proinflammatory phenotype. Similar metabolic alterations were observed in RA CD8+ T cells from the synovial membrane. CONCLUSION Remodeling glucose and glutamine metabolism in RA CD8+ T cells by targeting LDHA activity can reduce the deleterious inflammatory and cytolytic contributions of these cells to the development of autoimmunity.
Collapse
Affiliation(s)
| | | | - Mónica T Abreu
- Faculty of Medicine, University of Coimbra, Coimbra, Portugal
| | - André P Meyer
- University Hospital Heidelberg, Heidelberg, Germany, and Zentrum für Kinder- und Jugendmedizin, University Hospital Freiburg, Freiburg, Germany
| | - Rainer Saffrich
- Medical Faculty Mannheim, University Hospital Heidelberg, Heidelberg, Germany, and Deutsches Rotes Kreuz Baden-Württemberg-Hessen, Mannheim, Germany
| | | | | | | | | | | | - Lina Carvalho
- Faculty of Medicine, University of Coimbra, Coimbra, Portugal
| | | | | | | | - Rui A Carvalho
- University Hospital Heidelberg, Heidelberg, Germany, and Faculty of Science and Technology, University of Coimbra, Coimbra, Portugal
| |
Collapse
|
19
|
Seo MR, Kim JW, Park EJ, Jung SM, Sung YK, Kim H, Kim G, Kim HS, Lee MS, Lee J, Hur JA, Chin BS, Eom JS, Baek HJ. Recommendations for the management of patients with systemic rheumatic diseases during the coronavirus disease pandemic. Korean J Intern Med 2020; 35:1317-1332. [PMID: 32972125 PMCID: PMC7652644 DOI: 10.3904/kjim.2020.417] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/12/2020] [Revised: 09/06/2020] [Accepted: 09/24/2020] [Indexed: 02/06/2023] Open
Abstract
Patients with systemic rheumatic diseases (SRD) are vulnerable for coronavirus disease (COVID-19). The Korean College of Rheumatology recognized the urgent need to develop recommendations for rheumatologists and other physicians to manage patients with SRD during the COVID-19 pandemic. The working group was organized and was responsible for selecting key health questions, searching and reviewing the available literature, and formulating statements. The appropriateness of the statements was evaluated by voting panels using the modified Delphi method. Four general principles and thirteen individual recommendations were finalized through expert consensus based on the available evidence. The recommendations included preventive measures against COVID-19, medicinal treatment for stable or active SRD patients without COVID-19, medicinal treatment for SRD patients with COVID-19, and patient evaluation and monitoring. Medicinal treatments were categorized according to the status with respect to both COVID-19 and SRD. These recommendations should serve as a reference for individualized treatment for patients with SRD. As new evidence is emerging, an immediate update will be required.
Collapse
Affiliation(s)
- Mi Ryoung Seo
- Division of Rheumatology, Department of Internal Medicine, Gil Medical Center, Gachon University College of Medicine, Incheon, Korea
| | - Ji-Won Kim
- Division of Rheumatology, Department of Internal Medicine, Daegu Catholic University School of Medicine, Daegu, Korea
| | - Eun-Jung Park
- Division of Rheumatology, Department of Internal Medicine, National Medical Center, Seoul, Korea
| | - Seung Min Jung
- Division of Rheumatology, Department of Internal Medicine, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Yoon-Kyoung Sung
- Department of Rheumatology, Hanyang University Hospital for Rheumatic Diseases, Seoul, Korea
| | - Hyungjin Kim
- Department of Medical Humanities, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Gunwoo Kim
- Division of Rheumatology, Department of Internal Medicine, Daegu Fatima Hospital, Daegu, Korea
| | - Hyun-Sook Kim
- Division of Rheumatology, Department of Internal Medicine, Soonchunhyang University Seoul Hospital, Seoul, Korea
| | - Myeung-Su Lee
- Division of Rheumatology, Department of Internal Medicine, Wonkwang University Hospital, Iksan, Korea
| | - Jisoo Lee
- Division of Rheumatology, Department of Internal Medicine, Ewha Womans University College of Medicine, Seoul, Korea
| | - Ji An Hur
- Division of Infectious Diseases, Department of Internal Medicine, Yeungnam University Medical Center, Daegu, Korea
| | - Bum Sik Chin
- Division of Infectious Diseases, Department of Internal Medicine, National Medical Center, Seoul, Korea
| | - Joong Sik Eom
- Division of Infectious Diseases, Department of Internal Medicine, Gil Medical Center, Gachon University College of Medicine, Incheon, Korea
| | - Han Joo Baek
- Division of Rheumatology, Department of Internal Medicine, Gil Medical Center, Gachon University College of Medicine, Incheon, Korea
| | - The Korean College of Rheumatology working group
- Division of Rheumatology, Department of Internal Medicine, Gil Medical Center, Gachon University College of Medicine, Incheon, Korea
- Division of Rheumatology, Department of Internal Medicine, Daegu Catholic University School of Medicine, Daegu, Korea
- Division of Rheumatology, Department of Internal Medicine, National Medical Center, Seoul, Korea
- Division of Rheumatology, Department of Internal Medicine, College of Medicine, The Catholic University of Korea, Seoul, Korea
- Department of Rheumatology, Hanyang University Hospital for Rheumatic Diseases, Seoul, Korea
- Department of Medical Humanities, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
- Division of Rheumatology, Department of Internal Medicine, Daegu Fatima Hospital, Daegu, Korea
- Division of Rheumatology, Department of Internal Medicine, Soonchunhyang University Seoul Hospital, Seoul, Korea
- Division of Rheumatology, Department of Internal Medicine, Wonkwang University Hospital, Iksan, Korea
- Division of Rheumatology, Department of Internal Medicine, Ewha Womans University College of Medicine, Seoul, Korea
- Division of Infectious Diseases, Department of Internal Medicine, Yeungnam University Medical Center, Daegu, Korea
- Division of Infectious Diseases, Department of Internal Medicine, National Medical Center, Seoul, Korea
- Division of Infectious Diseases, Department of Internal Medicine, Gil Medical Center, Gachon University College of Medicine, Incheon, Korea
| |
Collapse
|
20
|
Seo MR, Kim JW, Park EJ, Jung SM, Sung YK, Kim H, Kim G, Kim HS, Lee MS, Lee J, Hur J, Chin BS, Eom JS, Baek HJ. Recommendations for the Management of Patients With Systemic Rheumatic Diseases During the Coronavirus Disease Pandemic. JOURNAL OF RHEUMATIC DISEASES 2020. [DOI: 10.4078/jrd.2020.27.4.218] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Affiliation(s)
- Mi Ryoung Seo
- Division of Rheumatology, Department of Internal Medicine, Gachon University College of Medicine, Gil Medical Center, Incheon, Korea
| | - Ji-Won Kim
- Division of Rheumatology, Department of Internal Medicine, Daegu Catholic University School of Medicine, Daegu, Korea
| | - Eun-Jung Park
- Division of Rheumatology, Department of Internal Medicine, National Medical Center, Seoul, Korea
| | - Seung Min Jung
- Division of Rheumatology, Department of Internal Medicine, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Yoon-Kyoung Sung
- Department of Rheumatology, Hanyang University Hospital for Rheumatic Diseases, Seoul, Korea
| | - Hyungjin Kim
- Department of Medical Humanities, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Gunwoo Kim
- Division of Rheumatology, Department of Internal Medicine, Daegu Fatima Hospital, Daegu, Korea
| | - Hyun-Sook Kim
- Division of Rheumatology, Department of Internal Medicine, Soonchunhyang University Seoul Hospital, Seoul, Korea
| | - Myeung-Su Lee
- Division of Rheumatology, Department of Internal Medicine, Wonkwang University Hospital, Iksan, Korea
| | - Jisoo Lee
- Division of Rheumatology, Department of Internal Medicine, Ewha Womans University College of Medicine, Seoul, Korea
| | - Jian Hur
- Division of Infectious Diseases, Department of Internal Medicine, Yeungnam University Medical Center, Daegu, Korea
| | - Bum Sik Chin
- Division of Infectious Diseases, Department of Internal Medicine, National Medical Center, Seoul, Korea
| | - Joong Sik Eom
- Division of Infectious Diseases, Department of Internal Medicine, Gachon University College of Medicine, Gil Medical Center, Incheon, Korea
| | - Han Joo Baek
- Division of Rheumatology, Department of Internal Medicine, Gachon University College of Medicine, Gil Medical Center, Incheon, Korea
| | | |
Collapse
|
21
|
Zeng Z, Zhang W, Qian Y, Huang H, Wu DJH, He Z, Ye D, Mao Y, Wen C. Association of telomere length with risk of rheumatoid arthritis: a meta-analysis and Mendelian randomization. Rheumatology (Oxford) 2020; 59:940-947. [PMID: 31697380 DOI: 10.1093/rheumatology/kez524] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2019] [Revised: 09/30/2019] [Indexed: 12/30/2022] Open
Abstract
OBJECTIVE To evaluate the telomere length (TL) in patients with RA relative to that in controls and to test whether TL is causally associated with risk of RA. METHODS Systematic review and meta-analysis of relevant literature was conducted to evaluate the association between TL and RA. Standardized mean differences with 95% CIs of TL in RA patients relative to controls were pooled using fixed or random-effects models. TL-related single-nucleotide polymorphisms were selected from a genome-wide association study of 37 684 individuals, and summary statistics of RA were obtained from a genome-wide association study meta-analysis including 14 361 RA patients and 43 923 controls. Mendelian randomization was performed using the inverse-variance weighted, weighted-median and likelihood-based methods. Sensitivity analyses were performed to test the robustness of the association. RESULTS In the meta-analysis of 911 RA patients and 2498 controls, we found that patients with RA had a significantly shorter TL compared with controls (standardized mean differences = -0.50; 95% CI -0.88, -0.11; P = 0.012). In the Mendelian randomization analysis, we found that genetically predicted longer TL was associated with a reduced risk of RA [odds ratio = 0.68; 95% CI 0.54, 0.86; P = 0.002 using the inverse-variance weighted method]. Sensitivity analyses using alternative Mendelian randomization approaches yielded similar findings, suggesting the robustness of the causal association. CONCLUSION Our study provides evidence for a negative causal association of TL with risk of RA. Further studies are warranted to elucidate the underlying mechanism for the role of telomeres in the development of RA.
Collapse
Affiliation(s)
- Zhen Zeng
- School of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
| | - Wanting Zhang
- School of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
| | - Yu Qian
- School of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
| | - Huijun Huang
- School of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
| | - David J H Wu
- School of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China.,University of Minnesota Medical School, University of Minnesota, Minneapolis, MN, USA
| | - Zhixing He
- School of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
| | - Ding Ye
- School of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
| | - Yingying Mao
- School of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
| | - Chengping Wen
- School of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
| |
Collapse
|
22
|
Effects of Probiotic Strains on Disease Activity and Enteric Permeability in Psoriatic Arthritis-A Pilot Open-Label Study. Nutrients 2020; 12:nu12082337. [PMID: 32764250 PMCID: PMC7468965 DOI: 10.3390/nu12082337] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Revised: 07/28/2020] [Accepted: 08/02/2020] [Indexed: 12/16/2022] Open
Abstract
(1) Background: Psoriatic Arthritis (PsA) is a painful disease of the joints and spine. Recent reports observed distinct enteric dysbiosis in PsA; intake of probiotic strains is considered to ameliorate enteric dysbiosis. If probiotics are effective in PsA is elusive. (2) Methods: In this pilot open-label study we enrolled 10 PsA patients with low to medium disease activity who received probiotics for 12 weeks. Analysis of faecal zonulin, α1-antitrypsin and calprotectin, as well as peripheral immune phenotyping was performed at baseline, after 12 weeks and 12 weeks after termination of probiotic intake. (3) Results: All patients showed increased levels of the enteric permeability marker zonulin which correlated with the frequency of peripheral Th17 cells. Calprotectin, a marker for intestinal inflammation was elevated in 6 out of 10 patients. Probiotic intake resulted in a reduction of disease activity and gut permeability. These effects, however, were not sustained beyond termination of probiotic intake. (4) Conclusions: PsA patients suffer from enhanced enteric permeability and inflammation. Probiotics may ameliorate disease activity in PsA by targeting these alterations.
Collapse
|
23
|
Sun Q, Liu J, Cheng G, Dai M, Liu J, Qi Z, Zhao J, Li W, Kong F, Liu G, Björkholm M, Xu D. The telomerase gene polymorphisms, but not telomere length, increase susceptibility to primary glomerulonephritis/end stage renal diseases in females. J Transl Med 2020; 18:184. [PMID: 32366311 PMCID: PMC7199377 DOI: 10.1186/s12967-020-02347-3] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2019] [Accepted: 04/17/2020] [Indexed: 12/19/2022] Open
Abstract
Background Primary glomerulonephritis (GN) is the leading cause of chronic kidney disease (CKD) and frequently progresses into end stage renal diseases (ESRDs). Shorter leukocyte telomere length (LTL) has been implicated in the CKD susceptibility and diminished kidney function, however, it is unclear whether the variants in telomerase genes contribute to risk to GN/CKD/ESRD. Here we address this issue by determining their association with the genetic variants of rs12696304 at the telomerase RNA component (TERC) and rs2736100 at the telomerase reverse transcriptase (TERT) loci. Methods The study includes 769 patients (243 primary GN-derived CKD and 526 ESRD cases) and sex-/age-matched healthy controls. Genomic DNA was extracted from peripheral blood of both controls and patients. Genotyping of rs12696304 and rs2736100 variants was carried out using PCR-based assays. Leukocyte telomere length (LTL) was determined using quantitative PCR (qPCR). Results A significantly higher frequency of TERC rs12696304 G allele was observed in patients and associated with increased disease risk (C vs G: OR = 1.334, 95% CI 1.112–1.586, P = 0.001; CC + GC vs GG: OR = 1.334, 95% CI 1.122–1.586, P = 0.001). Further analyses showed that such significant differences were only present between female controls and patients (C vs G: OR = 1.483, 95% CI 1.140–1.929, P = 0.003; CC + GC vs CC: OR = 1.692, 95% CI 1.202–2.383, P = 0.003), but not males. There were no differences in rs2736100 variants between controls and patients, but female ESRD patients carried significantly higher C allele frequencies than did female controls (A vs C: OR = 1.306, 95% CI 1.005–1.698, P = 0.046; AA vs CC: OR = 1.781, 95% CI 1.033–3.070, P = 0.037). There was no difference in LTL between controls and patients. Conclusions Our results reveal that the TERC rs12696304 and TERT rs2736100 polymorphisms, but not LTL per se, contribute to GN/CDK/ESRD risk.
Collapse
Affiliation(s)
- Qing Sun
- Central Research Laboratory, Shandong University Second Hospital, Jinan, 250035, People's Republic of China
| | - Junli Liu
- Laboratory for Molecular Diagnostics, Shandong University Second Hospital, Jinan, 250035, People's Republic of China
| | - Guanghui Cheng
- Central Research Laboratory, Shandong University Second Hospital, Jinan, 250035, People's Republic of China
| | - Mingkai Dai
- Central Research Laboratory, Shandong University Second Hospital, Jinan, 250035, People's Republic of China
| | - Jiaxi Liu
- Central Research Laboratory, Shandong University Second Hospital, Jinan, 250035, People's Republic of China
| | - Zhenqiang Qi
- Department of Nephrology, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, 250013, People's Republic of China
| | - Jingjie Zhao
- Laboratory for Molecular Diagnostics, Shandong University Second Hospital, Jinan, 250035, People's Republic of China
| | - Wei Li
- Department of Nephrology, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, 250013, People's Republic of China.
| | - Feng Kong
- Central Research Laboratory, Shandong University Second Hospital, Jinan, 250035, People's Republic of China.
| | - Gang Liu
- Nephrology Research Institute of Shandong University, Shandong University Second Hospital, Jinan, 250035, People's Republic of China.
| | - Magnus Björkholm
- Department of Medicine, Center for Molecular Medicine and Bioclinicum, Karolinska Institutet, Karolinska University Hospital Solna, SE-171 76, Stockholm, Sweden
| | - Dawei Xu
- Department of Medicine, Center for Molecular Medicine and Bioclinicum, Karolinska Institutet, Karolinska University Hospital Solna, SE-171 76, Stockholm, Sweden.
| |
Collapse
|
24
|
Lu Y, Jiang H, Li B, Cao L, Shen Q, Yi W, Ju Z, Chen L, Han F, Appelgren D, Segelmark M, de Buhr N, von Köckritz-Blickwede M, Chen J. Telomere dysfunction promotes small vessel vasculitis via the LL37-NETs-dependent mechanism. ANNALS OF TRANSLATIONAL MEDICINE 2020; 8:357. [PMID: 32355801 PMCID: PMC7186648 DOI: 10.21037/atm.2020.02.130] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Background Small vessel vasculitis (SVV) is a group of systemic autoimmune diseases that are mediated by neutrophil extracellular traps (NETs) in response to cathelicidin LL37, an aging molecular marker, which could be induced by telomere dysfunction. Therefore, in this study, we evaluated the hypothesis that telomere dysfunction in neutrophils may promote SVV via an LL37-NETs-dependent mechanism. Methods We contrasted the release of neutrophil NETs from mice with telomere dysfunction, mice with DNA damage and wide-type mice. Neutrophil telomere length, the expression of LL37, and the formation of NETs were measured in SVV patients and healthy controls (HCs). The co-expression of γH2AX, LL37, and NETs were detected in SVV patients to evaluate the association of the immune aging of neutrophils and pro-inflammatory conditions. LL37 inhibitor was used to verify its key role in NETs release in SVV patients and DNA damage mice. Results We found that NETs were over-induced by telomere dysfunction and DNA damage in mice, which may be associated with a marked increase in LL37. For patients with SVV, telomeres in neutrophils were significantly shortened, which was also associated with higher levels of LL37 and NETs. Inhibition of LL37 reduced the NETs released from neutrophils. Conclusions Taken together, the results of these studies suggest that dysfunction of telomeres may promote SVV through the mechanism of LL37-dependent NETs. Thus, targeting the LL37-NETs may be a novel therapy for SVV.
Collapse
Affiliation(s)
- Yingying Lu
- Kidney Disease Center, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou 310003, China.,Kidney Disease Immunology Laboratory, The Third Grade Laboratory, State Administration of Traditional Chinese Medicine of PR China, Hangzhou 310003, China.,Key Laboratory of Multiple Organ Transplantation, Ministry of Health, Hangzhou 310003, China.,Key Laboratory of Nephropathy, Hangzhou 310003, China
| | - Hong Jiang
- Kidney Disease Center, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou 310003, China.,Kidney Disease Immunology Laboratory, The Third Grade Laboratory, State Administration of Traditional Chinese Medicine of PR China, Hangzhou 310003, China.,Key Laboratory of Multiple Organ Transplantation, Ministry of Health, Hangzhou 310003, China.,Key Laboratory of Nephropathy, Hangzhou 310003, China
| | - Bingjue Li
- Kidney Disease Center, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou 310003, China.,Kidney Disease Immunology Laboratory, The Third Grade Laboratory, State Administration of Traditional Chinese Medicine of PR China, Hangzhou 310003, China.,Key Laboratory of Multiple Organ Transplantation, Ministry of Health, Hangzhou 310003, China.,Key Laboratory of Nephropathy, Hangzhou 310003, China
| | - Luxi Cao
- Kidney Disease Center, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou 310003, China.,Kidney Disease Immunology Laboratory, The Third Grade Laboratory, State Administration of Traditional Chinese Medicine of PR China, Hangzhou 310003, China.,Key Laboratory of Multiple Organ Transplantation, Ministry of Health, Hangzhou 310003, China.,Key Laboratory of Nephropathy, Hangzhou 310003, China
| | - Qixia Shen
- Kidney Disease Center, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou 310003, China.,Kidney Disease Immunology Laboratory, The Third Grade Laboratory, State Administration of Traditional Chinese Medicine of PR China, Hangzhou 310003, China.,Key Laboratory of Multiple Organ Transplantation, Ministry of Health, Hangzhou 310003, China.,Key Laboratory of Nephropathy, Hangzhou 310003, China
| | - Weiwei Yi
- Institute of Aging Research and Max-Planck-Research Group on Stem Cell Aging, Hangzhou Normal University, Hangzhou 311121, China
| | - Zhenyu Ju
- Institute of Aging Research and Max-Planck-Research Group on Stem Cell Aging, Hangzhou Normal University, Hangzhou 311121, China
| | - Liangliang Chen
- Kidney Disease Center, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou 310003, China.,Kidney Disease Immunology Laboratory, The Third Grade Laboratory, State Administration of Traditional Chinese Medicine of PR China, Hangzhou 310003, China.,Key Laboratory of Multiple Organ Transplantation, Ministry of Health, Hangzhou 310003, China.,Key Laboratory of Nephropathy, Hangzhou 310003, China
| | - Fei Han
- Kidney Disease Center, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou 310003, China.,Kidney Disease Immunology Laboratory, The Third Grade Laboratory, State Administration of Traditional Chinese Medicine of PR China, Hangzhou 310003, China.,Key Laboratory of Multiple Organ Transplantation, Ministry of Health, Hangzhou 310003, China.,Key Laboratory of Nephropathy, Hangzhou 310003, China
| | - Daniel Appelgren
- Department of Medical and Health Sciences (IMH), Linkoping University, Linkoping, Sweden
| | - Mårten Segelmark
- Department of Medical and Health Sciences (IMH), Linkoping University, Linkoping, Sweden
| | - Nicole de Buhr
- Department of Physiological Chemistry, University of Veterinary Medicine Hannover, Hannover, Germany.,Department of Physiological Chemistry and Research Center for Emerging Infections and Zoonoses (RIZ), University of Veterinary Medicine Hannover, 30559 Hannover, Germany
| | - Maren von Köckritz-Blickwede
- Department of Physiological Chemistry, University of Veterinary Medicine Hannover, Hannover, Germany.,Department of Physiological Chemistry and Research Center for Emerging Infections and Zoonoses (RIZ), University of Veterinary Medicine Hannover, 30559 Hannover, Germany
| | - Jianghua Chen
- Kidney Disease Center, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou 310003, China.,Kidney Disease Immunology Laboratory, The Third Grade Laboratory, State Administration of Traditional Chinese Medicine of PR China, Hangzhou 310003, China.,Key Laboratory of Multiple Organ Transplantation, Ministry of Health, Hangzhou 310003, China.,Key Laboratory of Nephropathy, Hangzhou 310003, China
| |
Collapse
|
25
|
Gracey E, Yao Y, Qaiyum Z, Lim M, Tang M, Inman RD. Altered Cytotoxicity Profile of CD8+ T Cells in Ankylosing Spondylitis. Arthritis Rheumatol 2020; 72:428-434. [PMID: 31599089 DOI: 10.1002/art.41129] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2019] [Accepted: 10/01/2019] [Indexed: 02/03/2023]
Abstract
OBJECTIVE Ankylosing spondylitis (AS) is an inflammatory arthritis in which men have a higher risk of developing progressive axial disease than women. Transcriptomic studies have shown reduced expression of cytotoxic cell genes in the blood of AS patients. HLA-B27 contributes the greatest risk for AS, suggesting a role for CD8+ T cells. This study was undertaken to profile AS patient cytotoxic cells with the hypothesis that an alteration in CD8+ T cells might explain the aberrant cytotoxic profile observed in patients. METHODS Whole blood was examined for GZM and PRF1 gene expression by quantitative polymerase chain reaction. Serum and synovial fluid (SF) were examined for granzyme and perforin 1 expression by bead array, and blood and SF mononuclear cells were examined for granzyme and perforin 1 expression by fluorescence-activated cell sorting (FACS). RESULTS GZM and PRF1 gene expression were both reduced in AS patients compared to healthy controls, especially in men. Perforin 1, but not granzyme, protein levels were reduced in AS patient serum. Granzymes were elevated in AS SF, but not in rheumatoid arthritis or osteoarthritis SF. FACS revealed a reduction in granzyme-positive and perforin 1-positive lymphocytes, but not an intrinsic defect in CD8+ T cell granzyme or perforin 1 production. CD8+ T cell frequency was reduced in the blood and increased in the SF of AS patients. CONCLUSION Our findings indicate that AS patients have an altered cytotoxic T cell profile. These data suggest that CD8+ T cells with a cytotoxic phenotype are recruited to the joints, where they exhibit an activated phenotype. Thus, a central role for CD8+ T cells in AS may have been overlooked and deserves further study.
Collapse
Affiliation(s)
- Eric Gracey
- University of Toronto, Krembil Research Institute, Toronto Western Hospital, and University Health Network, Toronto, Ontario, Canada
| | - Yuchen Yao
- University of Toronto, Krembil Research Institute, and University Health Network, Toronto, Ontario, Canada
| | - Zoya Qaiyum
- Krembil Research Institute, Toronto Western Hospital, and University Health Network, Toronto, Ontario, Canada
| | - Melissa Lim
- Krembil Research Institute, Toronto Western Hospital, and University Health Network, Toronto, Ontario, Canada
| | - Michael Tang
- Krembil Research Institute, Toronto Western Hospital, and University Health Network, Toronto, Ontario, Canada
| | - Robert D Inman
- University of Toronto, Krembil Research Institute, Toronto Western Hospital, and University Health Network, Toronto, Ontario, Canada
| |
Collapse
|
26
|
Tachikart Y, Malaise O, Mumme M, Jorgensen C, Brondello JM. Seno-suppressive molecules as new therapeutic perspectives in rheumatic diseases. Biochem Pharmacol 2019; 165:126-133. [PMID: 30878551 DOI: 10.1016/j.bcp.2019.03.017] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2019] [Accepted: 03/12/2019] [Indexed: 01/10/2023]
Abstract
Over the past years, through in vitro studies and unique animal models, biologists and clinicians have demonstrated that cellular senescence is at the root of numerous age-related chronic diseases including osteoarthritis and osteoporosis. This non-proliferative cellular syndrome can modify other surrounding tissue-resident cells through the establishment of a deleterious catabolic and inflammatory microenvironment. Targeting these deleterious cells through local or systemic seno-therapeutic agent delivery in pre-clinical models improves dramatically clinical signs and extends health span. In this review, we will summarize the current knowledge on cellular senescence, list the different strategies for identifying seno-suppressive therapeutic agents and their translations to rheumatic diseases.
Collapse
Affiliation(s)
- Yassin Tachikart
- IRMB (Institut of Regenerative Medicine and Biotherapies), Inserm U1183, Univ Montpellier, Montpellier, France
| | - Olivier Malaise
- IRMB (Institut of Regenerative Medicine and Biotherapies), Inserm U1183, Univ Montpellier, Montpellier, France; GIGA Research (Groupe Interdisciplinaire de Genoproteomique Appliquée), CHU de Liège & Université de Liège, Liège, Belgium
| | - Marcus Mumme
- IRMB (Institut of Regenerative Medicine and Biotherapies), Inserm U1183, Univ Montpellier, Montpellier, France; Clinic for Orthopedics and Traumatology, University Hospital of Basel, Basel, Switzerland
| | - Christian Jorgensen
- IRMB (Institut of Regenerative Medicine and Biotherapies), Inserm U1183, Univ Montpellier, Montpellier, France; Service de Rhumatologie, CHU La Peyronie, Montpellier, France
| | - Jean-Marc Brondello
- IRMB (Institut of Regenerative Medicine and Biotherapies), Inserm U1183, Univ Montpellier, Montpellier, France.
| |
Collapse
|
27
|
Fessler J, Husic R, Schwetz V, Lerchbaum E, Aberer F, Fasching P, Ficjan A, Obermayer-Pietsch B, Duftner C, Graninger W, Stradner MH, Dejaco C. Senescent T-Cells Promote Bone Loss in Rheumatoid Arthritis. Front Immunol 2018; 9:95. [PMID: 29472917 PMCID: PMC5810289 DOI: 10.3389/fimmu.2018.00095] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2017] [Accepted: 01/12/2018] [Indexed: 12/18/2022] Open
Abstract
Objective T-cells are critical players in the pathogenesis of osteoporosis in patients with rheumatoid arthritis (RA). Premature senescence of lymphocytes including the accumulation of senescent CD4+ T-cells is a hallmark feature of RA. Whether T-cell senescence is associated with bone loss in RA patients is elusive so far. Methods This includes a prospective study of consecutive patients with RA (n = 107), patients with primary osteopenia/-porosis (n = 75), and healthy individuals (n = 38). Bone mineral density (BMD) was determined by dual-energy X-ray absorptiometry scan. Flow cytometry, magnetic-associated cell sorting, and cell culture experiments were performed to analyze the pro-osteoclastic phenotype and the function of senescent CD4+CD28− T-cells. Results Patients with osteopenia/-porosis yielded a higher prevalence of senescent CD4+CD28− T-cells than individuals with normal BMD, in the RA, as well as in the non-RA cohort. Receptor activator of nuclear factor kappa-B ligand (RANKL) was expressed at higher levels on CD4+CD28− T-cells as compared to CD28+ T-cells. Stimulation with interleukin-15 led to an up-regulation of RANKL expression, particularly on CD28− T-cells. CD4+CD28− T-cells induced osteoclastogenesis more efficiently than CD28+ T-cells. Conclusion Our data indicate that senescent T-cells promote osteoclastogenesis more efficiently than conventional CD28+ T-cells, which might contribute to the pathogenesis of systemic bone loss in RA and primary osteoporosis.
Collapse
Affiliation(s)
- Johannes Fessler
- Department of Rheumatology and Immunology, Medical University of Graz, Graz, Austria
| | - Rusmir Husic
- Department of Rheumatology and Immunology, Medical University of Graz, Graz, Austria
| | - Verena Schwetz
- Department of Endocrinology and Diabetology, Medical University of Graz, Graz, Austria
| | - Elisabeth Lerchbaum
- Department of Endocrinology and Diabetology, Medical University of Graz, Graz, Austria
| | - Felix Aberer
- Department of Endocrinology and Diabetology, Medical University of Graz, Graz, Austria
| | - Patrizia Fasching
- Department of Rheumatology and Immunology, Medical University of Graz, Graz, Austria
| | - Anja Ficjan
- Department of Rheumatology and Immunology, Medical University of Graz, Graz, Austria
| | | | - Christina Duftner
- Department of Internal Medicine VI, Innsbruck Medical University, Innsbruck, Austria
| | - Winfried Graninger
- Department of Rheumatology and Immunology, Medical University of Graz, Graz, Austria
| | | | - Christian Dejaco
- Department of Rheumatology and Immunology, Medical University of Graz, Graz, Austria.,Rheumatology Service, South Tyrolean Health Trust, Hospital Bruneck, Bruneck, Italy
| |
Collapse
|
28
|
The influence and impact of ageing and immunosenescence (ISC) on adaptive immunity during multiple sclerosis (MS) and the animal counterpart experimental autoimmune encephalomyelitis (EAE). Ageing Res Rev 2018; 41:64-81. [PMID: 29101043 DOI: 10.1016/j.arr.2017.10.005] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2017] [Revised: 10/23/2017] [Accepted: 10/25/2017] [Indexed: 12/21/2022]
Abstract
The human ageing process encompasses mechanisms that effect a decline in homeostasis with increased susceptibility to disease and the development of chronic life-threatening illness. Increasing age affects the immune system which undergoes a progressive loss of efficiency, termed immunosenescence (ISC), to impact on quantitative and functional aspects of innate and adaptive immunity. The human demyelinating disease multiple sclerosis (MS) and the corresponding animal model experimental autoimmune encephalomyelitis (EAE) are strongly governed by immunological events that primarily involve the adaptive arm of the immune response. MS and EAE are frequently characterised by a chronic pathology and a protracted disease course which thereby creates the potential for exposure to the inherent, on-going effects and consequences of ISC. Collective evidence is presented to confirm the occurrence of established and unendorsed biological markers of ISC during the development of both diseases. Moreover, results are discussed from studies during the course of MS and EAE that reveal a premature upregulation of ISC-related biomarkers which indicates untimely alterations to the adaptive immune system. The effects of ISC and a prematurely aged immune system on autoimmune-associated neurodegenerative conditions such as MS and EAE are largely unknown but current evaluation of data justifies and encourages further investigation.
Collapse
|
29
|
Vazirpanah N, Kienhorst LBE, Van Lochem E, Wichers C, Rossato M, Shiels PG, Dalbeth N, Stamp LK, Merriman TR, Janssen M, Radstake TRDJ, Broen JC. Patients with gout have short telomeres compared with healthy participants: association of telomere length with flare frequency and cardiovascular disease in gout. Ann Rheum Dis 2017; 76:1313-1319. [PMID: 28347991 DOI: 10.1136/annrheumdis-2016-210538] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2016] [Revised: 01/31/2017] [Accepted: 03/02/2017] [Indexed: 01/12/2023]
Abstract
AIM AND BACKGROUND Chronic inflammation associates with increased senescence, which is a strong predictor for cardiovascular disease. We hypothesised that inflammation accelerates senescence and thereby enhances the risk of cardiovascular disease in gout. METHODS We assessed replicative senescence by quantifying telomere length (TL) in a discovery cohort of 145 Dutch patients with gout and 273 healthy individuals and validated our results in 474 patients with gout and 293 healthy participants from New Zealand. Subsequently, we investigated the effect of cardiovascular disease on TL of all participants. Also, we measured TL of CD4+ and CD8+ T lymphocytes, B lymphocytes, monocytes, natural killer cells and plasmacytoid dendritic cells. Additionally, we assessed the potential temporal difference in TL and telomerase activity. RESULTS TL in PBMCs of healthy donors decreased over time, reflecting normal ageing. Patients with gout demonstrated shorter telomeres (p=0.001, R2=0.01873). In fact, the extent of telomere erosion in patients with gout was higher at any age compared with healthy counterparts at any age (p<0.0001, R2=0.02847). Patients with gout with cardiovascular disease had the shortest telomeres and TL was an independent risk factor for cardiovascular disease in patients with gout (p=0.001). TL was inversely associated with the number of gouty flares (p=0.005). CONCLUSIONS Patients with gout have shorter telomeres than healthy participants, reflecting increased cellular senescence. Telomere shortening was associated with the number of flares and with cardiovascular disease in people with gout.
Collapse
Affiliation(s)
- N Vazirpanah
- Department of Rheumatology and Clinical Immunology, University Medical Centre Utrecht, Utrecht, The Netherlands
- Laboratory of Translational Immunology, University Medical Centre Utrecht, Utrecht, The Netherlands
| | - L B E Kienhorst
- Department of Dermatology, University Medical Centre Utrecht, Utrecht, The Netherlands
| | - E Van Lochem
- Department of Medical Microbiology and Immunology, Rijnstate Hospital, Arnhem, The Netherlands
| | - C Wichers
- Department of Rheumatology and Clinical Immunology, University Medical Centre Utrecht, Utrecht, The Netherlands
- Laboratory of Translational Immunology, University Medical Centre Utrecht, Utrecht, The Netherlands
| | - M Rossato
- Department of Rheumatology and Clinical Immunology, University Medical Centre Utrecht, Utrecht, The Netherlands
- Laboratory of Translational Immunology, University Medical Centre Utrecht, Utrecht, The Netherlands
| | - P G Shiels
- University of Glasgow, Institute of Cancer Sciences, Wolfson-Wohl Translational Cancer Research Centre, Glasgow, UK
| | - N Dalbeth
- University of Auckland, Auckland, New Zealand
| | - L K Stamp
- University of Otago, Christchurch, New Zealand
| | | | - M Janssen
- Department of Medical Microbiology and Immunology, Rijnstate Hospital, Arnhem, The Netherlands
| | - T R D J Radstake
- Department of Rheumatology and Clinical Immunology, University Medical Centre Utrecht, Utrecht, The Netherlands
- Laboratory of Translational Immunology, University Medical Centre Utrecht, Utrecht, The Netherlands
| | - J Ca Broen
- Department of Rheumatology and Clinical Immunology, University Medical Centre Utrecht, Utrecht, The Netherlands
- Laboratory of Translational Immunology, University Medical Centre Utrecht, Utrecht, The Netherlands
| |
Collapse
|
30
|
Fessler J, Raicht A, Husic R, Ficjan A, Schwarz C, Duftner C, Schwinger W, Graninger WB, Stradner MH, Dejaco C. Novel Senescent Regulatory T-Cell Subset with Impaired Suppressive Function in Rheumatoid Arthritis. Front Immunol 2017; 8:300. [PMID: 28373873 PMCID: PMC5357868 DOI: 10.3389/fimmu.2017.00300] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2016] [Accepted: 03/03/2017] [Indexed: 11/15/2022] Open
Abstract
Objective Premature senescence of lymphocytes is a hallmark of inflammatory rheumatic diseases such as rheumatoid arthritis (RA). Early T-cell aging affects conventional T-cells but is presumably not limited to this cell population; rather it might also occur in the regulatory T-cells (Tregs) compartment. In RA, Tregs fail to halt aberrant immune reactions and disease progression. Whether this is associated with early Treg senescence leading to phenotypic and functional changes of this subset is elusive so far. Methods Eighty-four RA patients and 75 healthy controls were prospectively enrolled into the study. Flow cytometry, magnetic-associated cell sorting, and cell culture experiments were performed for phenotypic and functional analyses of Treg subsets. T-cell receptor excision circle (TREC) levels and telomere lengths were determined using RT-PCR. Results In this paper, we describe the novel CD4+FoxP3+CD28− T-cell subset (CD28− Treg-like cells) in RA patients revealing features of both Tregs and senescent T-cells: Treg surface/intracellular markers such as CD25, CTLA-4, and PD-1 as well as FOXP3 were all expressed by CD28− Treg-like cells, and they yielded signs of premature senescence including reduced TREC levels and an accumulation of γH2AX. CD28− Treg-like could be generated in vitro by stimulation of (CD28+) Tregs with TNF-α. CD28− Treg-like cells insufficiently suppressed the proliferation of effector T-cells and yielded a pro-inflammatory cytokine profile. Conclusion In conclusion, we describe a novel T-cell subset with features of Tregs and senescent non-Tregs. These cells may be linked to an aberrant balance between regulatory and effector functions in RA.
Collapse
Affiliation(s)
- Johannes Fessler
- Department of Rheumatology and Immunology, Medical University of Graz , Graz , Austria
| | - Andrea Raicht
- Department of Pediatric Hemato-Oncology, Medical University of Graz , Graz , Austria
| | - Rusmir Husic
- Department of Rheumatology and Immunology, Medical University of Graz , Graz , Austria
| | - Anja Ficjan
- Department of Rheumatology and Immunology, Medical University of Graz , Graz , Austria
| | - Christine Schwarz
- Department of Pediatric Hemato-Oncology, Medical University of Graz , Graz , Austria
| | - Christina Duftner
- Department of Internal Medicine VI, Innsbruck Medical University , Innsbruck , Austria
| | - Wolfgang Schwinger
- Department of Pediatric Hemato-Oncology, Medical University of Graz , Graz , Austria
| | - Winfried B Graninger
- Department of Rheumatology and Immunology, Medical University of Graz , Graz , Austria
| | - Martin H Stradner
- Department of Rheumatology and Immunology, Medical University of Graz , Graz , Austria
| | - Christian Dejaco
- Department of Rheumatology and Immunology, Medical University of Graz, Graz, Austria; Rheumatology Service, South Tyrolian Health Trust, Hospital Bruneck, Bruneck, Italy
| |
Collapse
|
31
|
The Telomere/Telomerase System in Chronic Inflammatory Diseases. Cause or Effect? Genes (Basel) 2016; 7:genes7090060. [PMID: 27598205 PMCID: PMC5042391 DOI: 10.3390/genes7090060] [Citation(s) in RCA: 104] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2016] [Revised: 07/26/2016] [Accepted: 08/29/2016] [Indexed: 12/23/2022] Open
Abstract
Telomeres are specialized nucleoprotein structures located at the end of linear chromosomes and telomerase is the enzyme responsible for telomere elongation. Telomerase activity is a key component of many cancer cells responsible for rapid cell division but it has also been found by many laboratories around the world that telomere/telomerase biology is dysfunctional in many other chronic conditions as well. These conditions are characterized by chronic inflammation, a situation mostly overlooked by physicians regarding patient treatment. Among others, these conditions include diabetes, renal failure, chronic obstructive pulmonary disease, etc. Since researchers have in many cases identified the association between telomerase and inflammation but there are still many missing links regarding this correlation, the latest findings about this phenomenon will be discussed by reviewing the literature. Our focus will be describing telomere/telomerase status in chronic diseases under the prism of inflammation, reporting molecular findings where available and proposing possible future approaches.
Collapse
|
32
|
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
|