1
|
Hu M, Zhou Y, Yao Z, Tang Y, Zhang Y, Liao J, Cai X, Liu L. T cell dysregulation in rheumatoid arthritis: Recent advances and natural product interventions. Int Immunopharmacol 2025; 153:114499. [PMID: 40120382 DOI: 10.1016/j.intimp.2025.114499] [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: 10/25/2024] [Revised: 02/28/2025] [Accepted: 03/13/2025] [Indexed: 03/25/2025]
Abstract
Autoimmune diseases result from chronic and dysregulated activation of the immune system, culminating in pathological self-tissue damage. These disorders are primarily driven by adaptive immune responses, particularly those mediated by T and B lymphocytes, which mistakenly target self-antigens expressed in host tissues. In rheumatoid arthritis (RA), the pathogenesis is closely associated with the emergence of tissue-invasive effector T cells and the functional impairment of regulatory T cells (Tregs), both of which play pivotal roles in disease progression. Therapeutic interventions targeting these dysregulated T cell populations have emerged as a promising strategy for RA management. Although synthetic immunosuppressants remain the mainstay of RA treatment, their long-term application is often hampered by adverse effects, diminished therapeutic efficacy, and poor patient adherence. These limitations highlight the critical need for the development of novel therapeutic approaches. Natural compounds derived from medicinal plants have been widely utilized in the clinical management of RA, with growing evidence supporting their immunomodulatory potential, particularly in restoring T cell-mediated immune tolerance. This review aims to provide a comprehensive overview of recent advances in understanding T cell dysregulation in RA and to elucidate the mechanisms through which natural compounds regulate immune responses. By integrating current findings, this work seeks to offer a theoretical foundation for the optimized use of natural compounds in the treatment of RA, while exploring their potential in advancing precision medicine and personalized therapeutic strategies.
Collapse
Affiliation(s)
- Mingyue Hu
- Department of Rheumatology of First Hospital and School of Chinese Medical Sciences, Hunan University of Chinese Medicine, Changsha, Hunan 410208, China
| | - Yujun Zhou
- The General Surgery Department of Xiangya Hospital Affiliated to Central South University, Changsha, Hunan 410028, China
| | - Zhongliu Yao
- Department of Rheumatology of First Hospital and School of Chinese Medical Sciences, Hunan University of Chinese Medicine, Changsha, Hunan 410208, China
| | - Yuanyuan Tang
- Department of Rheumatology of First Hospital and School of Chinese Medical Sciences, Hunan University of Chinese Medicine, Changsha, Hunan 410208, China
| | - Ye Zhang
- Department of Rheumatology of First Hospital and School of Chinese Medical Sciences, Hunan University of Chinese Medicine, Changsha, Hunan 410208, China
| | - Jing Liao
- Department of Rheumatology of First Hospital and School of Chinese Medical Sciences, Hunan University of Chinese Medicine, Changsha, Hunan 410208, China.
| | - Xiong Cai
- Department of Rheumatology of First Hospital and School of Chinese Medical Sciences, Hunan University of Chinese Medicine, Changsha, Hunan 410208, China.
| | - Liang Liu
- State Key Laboratory of Traditional Chinese Medicine Syndrome, Guangdong Provincial Hospital of Chinese Medicine, Guangdong Provincial Academy of Chinese Medical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong 510006, China.
| |
Collapse
|
2
|
Li C, Lu W, Zhang H. BTB domain and CNC homolog 2: A master regulator that controls immune response and cancer progression. Biochim Biophys Acta Rev Cancer 2025; 1880:189325. [PMID: 40252853 DOI: 10.1016/j.bbcan.2025.189325] [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: 12/08/2024] [Revised: 04/10/2025] [Accepted: 04/13/2025] [Indexed: 04/21/2025]
Abstract
BTB domain and CNC homolog 2 (BACH2) is a transcription repressor from the basic region leucine zipper (bZIP) family. Although BACH2 is predominantly expressed in lymphoid cells, it plays pivotal roles throughout hematological development and differentiation, ranging from the regulation of hematopoietic stem and progenitor cell (HSPC) lineage commitment to the development of both innate and adaptive immune cells. Given its extensive regulation of immunity, it is not surprising that BACH2 has been implicated in cancer, particularly in hematological malignancies. While multiple findings indicate that BACH2 acts primarily as a tumor suppressor, other findings suggest that BACH2, whether within tumor cells or their surrounding microenvironment, may contribute to tumorigenesis and progression, highlighting the complexity of its roles and the diverse networks involved in different contexts. In this review, we provide a comprehensive overview of the evolving roles of BACH2 across various stages of hematopoiesis, with a particular focus on its associations with cancer and its therapeutic potential in a wide range of cancer types.
Collapse
Affiliation(s)
- Chenyang Li
- Institute of Medical Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Kunming, Yunnan 650118, China
| | - Wei Lu
- Institute of Medical Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Kunming, Yunnan 650118, China; Kunming Medical University, Kunming, Yunnan 650500, China
| | - Han Zhang
- Institute of Medical Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Kunming, Yunnan 650118, China.
| |
Collapse
|
3
|
Zheng Q, Lin R, Li Z, Zheng Q, Xu W. Taurine is a potential therapy for rheumatoid arthritis via targeting FOXO3 through cellular senescence and autophagy. PLoS One 2025; 20:e0318311. [PMID: 40238799 PMCID: PMC12002484 DOI: 10.1371/journal.pone.0318311] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2024] [Accepted: 01/13/2025] [Indexed: 04/18/2025] Open
Abstract
BACKGROUND Rheumatoid arthritis (RA) is a chronic inflammatory autoimmune disease closely related to aging with unclear pathogenic mechanisms. This study aims to identify the biomarkers in RA, aging and autophagy using bioinformatics and machine learning and explore the binding stability of taurine to target utilizing computer-aided drug design (CADD). METHODS We identified differentially expressed genes (DEGs) for RA, then crossed with gene libraries for aging and autophagy to identify common genes (Co-genes). We performed Gene Ontology (GO), Kyoto Encyclopedia of the Genome (KEGG), and ClueGO analysis for Co-genes. The Co-genes were subjected to support vector machine-recursive feature elimination (SVM-RFE), Degree, and Betweenness algorithms to get hub genes, then verified by an artificial neural network (ANN). After continuing to perform least absolute shrinkage and selection operator (LASSO) and weighted gene co-expression network analysis (WGCNA) on Co-genes, the results were crossed with hub genes to obtain genes, which were imported into various validation sets for receiver operating characteristics (ROC) to identify key genes. We analyzed the microRNA/TF network, enriched pathways, and immune cell infiltration for key genes. The binding stability of taurine with the target protein was verified by CADD. Finally, we used Western blot for in vitro experimental verification. RESULTS We obtained 74 Co-genes enriched in RA, cellular senescence, and regulation of programmed cell death. The model prediction of hub genes works well in ANN. The key genes (MMP9, CXCL10, IL15, FOXO3) were tested in ROC with excellent efficacy. In RA, FOXO3 expression was down-regulated while MMP9, CXCL10, and IL15 expression were upregulated, and FOXO3 was negatively correlated with MMP9, CXCL10, and IL15. Two miRNAs (hsa-mir-21-5p, hsa-mir-129-2-3p) and four TFs (CTCF, KLF, FOXC1, TP53) were associated with key genes. The immune cells positively correlated with MMP9, CXCL10, and IL15 expression and negatively correlated with FOXO3 expression were Plasma cells, CD8 T cells, memory-activated CD4 T cells, and follicular helper T cells, aggregating in RA. The binding stability of taurine with FOXO3 was verified by molecular docking and molecular dynamics simulation. In vitro experiments have indicated that taurine can upregulate the expression of FOXO3 and treat RA through the FOXO3-Parkin signaling pathway. CONCLUSIONS MMP9, CXCL10, IL15, and FOXO3 are biomarkers of RA, cellular senescence, and autophagy. Taurine might be a promising drug against RA via targeting cellular senescence and autophagy through FOXO3.
Collapse
Affiliation(s)
- Qingcong Zheng
- Department of Spinal Surgery, The First Affiliated Hospital of Fujian Medical University, Fuzhou, China
| | - Rongjie Lin
- Department of Orthopedic Surgery, Fujian Medical University Union Hospital, Fuzhou, China
| | - Zhechen Li
- Department of Spinal Surgery, The First Affiliated Hospital of Fujian Medical University, Fuzhou, China
| | - Qingzhu Zheng
- Department of Laboratory Medicine, Fujian Medical University Union Hospital, Fuzhou, China
| | - Weihong Xu
- Department of Spinal Surgery, The First Affiliated Hospital of Fujian Medical University, Fuzhou, China
| |
Collapse
|
4
|
Yu W, Jie S, Su G, Zhuangwei N, Yiqing Z, Suhong C, Guiyuan L. The ultrafine powder of atractylodis macrocephalae rhizoma improves immune function in naturally aging rats by regulating the PI3K/Akt/NF-κB signaling pathway. Front Pharmacol 2025; 16:1550357. [PMID: 40255567 PMCID: PMC12006087 DOI: 10.3389/fphar.2025.1550357] [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: 12/23/2024] [Accepted: 03/21/2025] [Indexed: 04/22/2025] Open
Abstract
Background The phenomenon of population aging presents a significant global challenge, with the aging population in China steadily increasing. As individuals progress in age, there is a gradual deterioration of human organs and systems, as well as a decline in the immune system, referred to as immunosenescence. Atractylodis macrocephalae rhizoma (BZ) has been historically used in China for its medicinal properties, including gastrointestinal improvement, immunomodulation, anti-aging, antioxidant effects, and anti-tumor effects. Nevertheless, there remains a gap in understanding the pharmacological and molecular mechanisms underlying its anti-immunosenescence effects. Methods This study employed UPLC-ESI-MS and network pharmacology to create a network map of BZ ultrafine powder (BZU) and its aging targets. Enrichment analysis was then used to identify the primary mechanistic pathways underlying BZU's anti-immunosenescence effects. The primary components of BZU were quantitatively analyzed using high-performance liquid chromatography (HPLC). Naturally aging rats were used to examine the effects of different oral doses (0.25, 0.5, and 1 g/kg) of BZU over 5 weeks on aging performance, peripheral blood immunophenotyping and cell count, and splenic lymphocyte proliferation rate. To validate the findings of network pharmacology, quantitative RT-PCR, Western blotting, and immunofluorescence analyses were conducted. Results Our analyses demonstrated that BZU improved various indicators of aging in naturally aging rats, such as increasing the number of voluntary activities, enhance grip strength and fatigue resistance, increasing the microcirculatory blood flow and improving hematological levels. The BZU administration enhanced T and B lymphocyte proliferation and significantly improved the lymphocyte-to-T cell subpopulation ratio. It can elevate serum IL-2 and IL-4 levels while reducing IL-6, IFN-γ and TNF-α levels in naturally aging rats. Finally, it increased CD3 protein expression in the spleen while decreasing protein levels of PI3K, p-AKT, IKKα/β, and NF-κB. It also decreased the mRNA expression of Pik3cg, Akt1, Pdk1 and Nfκb1. Conclusion These findings suggest that BZU may enhance lymphocyte proliferation by inhibiting the PI3K/Akt/NF-κB signaling pathway, correcting immune cell imbalances, reducing inflammatory responses, and ultimately enhancing immune function and potentially delaying aging.
Collapse
Affiliation(s)
| | | | | | | | | | - Chen Suhong
- College of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, China
| | - Lv Guiyuan
- College of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, China
| |
Collapse
|
5
|
Zhou M, Shu Y, Gao J. Thymus Degeneration in Women and the Influence of Female Sexual Hormones on Thymic Epithelial Cells. Int J Mol Sci 2025; 26:3014. [PMID: 40243626 PMCID: PMC11988661 DOI: 10.3390/ijms26073014] [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: 02/20/2025] [Revised: 03/19/2025] [Accepted: 03/24/2025] [Indexed: 04/18/2025] Open
Abstract
The thymus is a central immune organ for T cell development and plays an extremely important role in immune and aging. The unique physiological processes that occur in women, such as the menstrual cycle, pregnancy, and menopause, contribute to sexual dimorphism in thymic immunity. Thymic epithelial cells (TECs) are key stromal cells that affect thymus development and degeneration. Interestingly, TECs in women have stronger proliferation potentiality and ability for output of T cells than those in men. In comparison to men, women exhibit higher susceptibility to autoimmune disease, which can be attributed to lower AIRE expression in the female thymus, which is influenced by fluctuating hormone levels. In this review, we summarize the principles of female thymus regulation by hormones, particularly the influence of female sex hormones in the development and function of TECs, as well as the underlying mechanisms, with the aim of providing new ideas and strategies to inhibit or slow down female thymus degeneration.
Collapse
Affiliation(s)
| | | | - Jianli Gao
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou 310053, China; (M.Z.); (Y.S.)
| |
Collapse
|
6
|
Weyand CM, Goronzy JJ. Immune Aging in Rheumatoid Arthritis. Arthritis Rheumatol 2025. [PMID: 39800938 DOI: 10.1002/art.43105] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2024] [Revised: 11/11/2024] [Accepted: 12/18/2024] [Indexed: 02/12/2025]
Abstract
Rheumatoid arthritis (RA) is a life-long autoimmune disease caused by the confluence of genetic and environmental variables that lead to loss of self-tolerance and persistent joint inflammation. RA occurs at the highest incidence in individuals >65 years old, implicating the aging process in disease susceptibility. Transformative approaches in molecular immunology and in functional genomics have paved the way for pathway paradigms underlying the replacement of immune homeostasis with autodestructive immunity in affected patients, including the process of immune aging. Patients with RA have a signature of premature immune aging, best understood for CD4+ T cells, which function as pathogenic effectors in this HLA class II-associated disease. Premature immune aging is present in healthy HLA-DRB1*04+ individuals, placing accelerated immune aging before joint inflammation. Aging-related molecular abnormalities directly implicated in turning RA CD4+ T cells into proinflammatory effector cells are linked to malfunction of subcellular organelles, such as mitochondria, lysosomes, lipid droplets, and the endoplasmic reticulum. Resulting changes in T cell behavior include cellular hypermobility, tissue invasiveness, unopposed mammalian target of rapamycin complex (mTORC)1 activation, excessive release of tumor necrosis factor, lysosomal failure, clonal expansion, and immunogenic cell death. Aged and metabolically reprogrammed T cells in patients with RA are accompanied by age-associated B cells, which specialize in autoantibody production. Clonal hematopoiesis drives myeloid cell aging by producing aged monocytes and hypermetabolic macrophages, which sustain the process of inflammaging. Here, we synthesize insights into the relationship of RA risk and immune aging and discuss mechanisms through which immune aging can cause autoimmunity.
Collapse
Affiliation(s)
- Cornelia M Weyand
- Mayo Clinic Alix School of Medicine, Mayo Clinic College of Medicine and Science, Rochester, Minnesota, and Stanford University School of Medicine, Stanford, California
| | - Jörg J Goronzy
- Mayo Clinic Alix School of Medicine, Mayo Clinic College of Medicine and Science, Rochester, Minnesota, and Stanford University School of Medicine, Stanford, California
| |
Collapse
|
7
|
Fang S, Jiang M, Jiao J, Zhao H, Liu D, Gao D, Wang T, Yang Z, Yuan H. Unraveling the ROS-Inflammation-Immune Balance: A New Perspective on Aging and Disease. Aging Dis 2025:AD.2024.1253. [PMID: 39812539 DOI: 10.14336/ad.2024.1253] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2024] [Accepted: 01/03/2025] [Indexed: 01/16/2025] Open
Abstract
Increased entropy is a common cause of disease and aging. Lifespan entropy is the overall increase in disorder caused by a person over their lifetime. Aging leads to the excessive production of reactive oxygen species (ROS), which damage the antioxidant system and disrupt redox balance. Organ aging causes chronic inflammation, disrupting the balance of proinflammatory and anti-inflammatory factors. Inflammaging, which is a chronic low-grade inflammatory state, is activated by oxidative stress and can lead to immune system senescence. During this process, entropy increases significantly as the body transitions from a state of low order to high disorder. However, the connection among inflammation, aging, and immune system activity is still not fully understood. This review introduces the idea of the ROS-inflammation-immune balance for the first time and suggests that this balance may be connected to aging and the development of age-related diseases. We also explored how the balance of these three factors controls and affects age-related diseases. Moreover, imbalance in the relationship described above disrupts the regular structures of cells and alters their functions, leading to cellular damage and the emergence of a disorganized state marked by increased entropy. Maintaining a low entropy state is crucial for preventing and reversing aging processes. Consequently, we examined the current preclinical evidence for antiaging medications that target this balance. Ultimately, comprehending the intricate relationships between these three factors and the risk of age-related diseases in organisms will aid in the development of clinical interventions that promote long-term health.
Collapse
Affiliation(s)
- Sihang Fang
- The Key Laboratory of Geriatrics, Beijing Institute of Geriatrics, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing Hospital/National Center of Gerontology of National Health Commission, Beijing, China
| | - Mingjun Jiang
- Respiratory Department, Beijing Children's Hospital, Capital Medical University, China National Clinical Research Center of Respiratory Diseases, National Center for Children's Health, Beijing, China
| | - Juan Jiao
- Department of Clinical Laboratory, the Seventh Medical Center of PLA General Hospital, Beijing, China
| | - Hongye Zhao
- Department of Biochemistry and Molecular Biology, The Key Laboratory of Neural and Vascular Biology, Ministry of Education of China, Hebei Medical University, Shijiazhuang, Hebei, China
| | - Dizhi Liu
- The Key Laboratory of Geriatrics, Beijing Institute of Geriatrics, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing Hospital/National Center of Gerontology of National Health Commission, Beijing, China
| | - Danni Gao
- The Key Laboratory of Geriatrics, Beijing Institute of Geriatrics, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing Hospital/National Center of Gerontology of National Health Commission, Beijing, China
| | - Tenger Wang
- The Key Laboratory of Geriatrics, Beijing Institute of Geriatrics, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing Hospital/National Center of Gerontology of National Health Commission, Beijing, China
| | - Ze Yang
- The Key Laboratory of Geriatrics, Beijing Institute of Geriatrics, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing Hospital/National Center of Gerontology of National Health Commission, Beijing, China
| | - Huiping Yuan
- The Key Laboratory of Geriatrics, Beijing Institute of Geriatrics, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing Hospital/National Center of Gerontology of National Health Commission, Beijing, China
| |
Collapse
|
8
|
Priora M, Becciolini A, Celletti E, Di Penta M, Lo Gullo A, Paroli M, Bravi E, Andracco R, Nucera V, Ometto F, Lumetti F, Farina A, Del Medico P, Colina M, Ravagnani V, Scolieri P, Larosa M, Visalli E, Addimanda O, Vitetta R, Volpe A, Bezzi A, Girelli F, Molica Colella AB, Caccavale R, Di Donato E, Adorni G, Santilli D, Lucchini G, Arrigoni E, Sabatini E, Platè I, Mansueto N, Ianniello A, Fusaro E, Ditto MC, Bruzzese V, Camellino D, Bianchi G, Serale F, Foti R, Amato G, De Lucia F, Bosco YD, Foti R, Reta M, Fiorenza A, Rovera G, Marchetta A, Focherini MC, Mascella F, Bernardi S, Sandri G, Giuggioli D, Salvarani C, Franchina V, Molica Colella F, Ferrero G, Ariani A, Parisi S. Effectiveness and Predictors of Long-Term Treatment Response to Tofacitinib in Rheumatoid Arthritis Cohort: General Analysis and Focus on High-Cardiovascular-Risk Subgroup-A Multicenter Study. MEDICINA (KAUNAS, LITHUANIA) 2024; 60:1982. [PMID: 39768862 PMCID: PMC11677110 DOI: 10.3390/medicina60121982] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/28/2024] [Revised: 11/17/2024] [Accepted: 11/25/2024] [Indexed: 01/11/2025]
Abstract
Background and Objectives: The treatment landscape for Rheumatoid Arthritis (RA) has evolved significantly with the introduction of Janus kinase inhibitors (JAKi), such as Tofacitinib (TOFA), which offer a new therapeutic option for patients who have failed or are intolerant to conventional synthetic disease-modifying antirheumatic drugs (csDMARDs). Safety concerns, particularly related to cardiovascular and cancer risks, prompted a need for additional investigation in real-world clinical settings. This study aimed to evaluate the long-term effectiveness and predictors of response to TOFA in two subpopulations of RA patients, categorized by differing cardiovascular risk profiles. Materials and Methods: This was a retrospective, multicenter observational study conducted as part of the BIRRA project, involving 23 Italian rheumatological referral centers. A total of 213 patients diagnosed with RA and treated with TOFA were included, with data collected on baseline demographics, clinical history, disease activity, and comorbidities. Patients were divided into high-risk and low-risk cardiovascular groups based on age (≥65 years) and the presence of at least one cardiovascular risk factor. Disease activity was assessed at baseline, 6 months, and 12 months using DAS28-ESR and DAS28-CRP. Treatment response was evaluated using intention-to-treat (ITT) and per-protocol (PP) approaches. Predictors of low disease activity (LDA) and remission were assessed through logistic regression, and clustering analyses were used to identify subgroups of patients with different therapeutic responses. Results: The study included 213 patients, with 129 classified as high-risk. For the overall cohort, patients achieving LDA and remission at 6 months were 20% and 12%, respectively, for the ITT analysis, and 29% and 14% for the PP analysis. At 12 months, 26% of patients reached LDA, and 17% achieved remission according to ITT, while for the PP analysis, these rates were 30% and 19%, respectively. No significant differences in remission or LDA rates were observed between the high-risk and low-risk groups. In the high-risk subgroup, 17% of patients reached LDA and 9% achieved remission at 6 months (ITT analysis), while these rates increased to 22% and 13%, respectively, in the PP analysis. At 12 months, 22% achieved LDA and 13% achieved remission in the ITT analysis, while 28% and 17% did so in the PP analysis. The reduction in DAS28-ESR and DAS28-CRP scores was significant (p < 0.001) across all time points for both high-risk and low-risk patients. Logistic regression analyses revealed that none of the baseline characteristics-including age, sex, comorbidities, rheumatoid factor, anti-citrullinated protein antibody (ACPA) positivity, initial disease severity, or treatment history-were significant predictors of remission or LDA at 6 or 12 months. The clustering analysis suggested that older patients, particularly those with worse baseline DAS28 scores, tended to show a less favorable response to treatment, potentially indicating impacts of age-related factors such as immunosenescence on therapeutic outcomes. Conclusions: Tofacitinib demonstrated similar effectiveness in both high- and low-risk cardiovascular subgroups of RA patients, with significant reductions in disease activity observed at both 6 and 12 months. Despite safety concerns related to cardiovascular risk, TOFA remained an effective treatment option across patient subgroups, with no significant differences in remission or LDA rates based on cardiovascular risk profiles. Age appeared to negatively impact treatment response, highlighting the role of immunosenescence in RA management. These findings support the use of TOFA as a personalized therapeutic option for RA, emphasizing the need for careful evaluation of cardiovascular and age-related risks in clinical decision-making.
Collapse
Affiliation(s)
- Marta Priora
- Rheumatology Day Hospital and Outpatient Clinic, ASL CN1, 12100 Cuneo, Italy; (M.P.); (F.S.)
| | - Andrea Becciolini
- Internal Medicine and Rheumatology Unit, University Hospital of Parma, 43126 Parma, Italy; (A.B.); (E.D.D.); (G.A.); (D.S.); (G.L.); (A.A.)
| | - Eleonora Celletti
- Rheumatology Unit, Clinica Medica Institute, SS Annunziata Hospital, 66100 Chieti, Italy; (M.D.P.); (E.S.)
| | - Myriam Di Penta
- Rheumatology Unit, Clinica Medica Institute, SS Annunziata Hospital, 66100 Chieti, Italy; (M.D.P.); (E.S.)
| | | | - Marino Paroli
- Department of Clinical, Anesthesiological and Cardiovascular Sciences, Sapienza University of Rome, 00185 Rome, Italy; (M.P.); (R.C.)
| | - Elena Bravi
- Reumathology Unit, Guglielmo da Saliceto Hospital, 29121 Piacenza, Italy; (E.B.); (E.A.); (I.P.)
| | - Romina Andracco
- Rheumatology Unit, Imperia Hospital, 18100 Imperia, Italy; (R.A.); (N.M.)
| | - Valeria Nucera
- Rheumatology Outpatient Unit, ASL Novara, 28100 Novara, Italy; (V.N.); (A.I.)
| | - Francesca Ometto
- Rheumatology Outpatient Clinic, Azienda ULSS 6 Euganea, 35131 Padova, Italy;
| | - Federica Lumetti
- Rheumatology Unit, Azienda USL of Modena and AOU Policlinico of Modena, 41100 Modena, Italy;
| | - Antonella Farina
- Internal Medicine Unit, Rheumatology Outpatient Clinic, Augusto Murri Hospital, 63900 Fermo, Italy;
| | - Patrizia Del Medico
- Rheumatology Outpatient Clinic, Internal Medicine Unit, Civitanova Marche Hospital, 62012 Civitanova Marche, Italy;
| | - Matteo Colina
- Rheumatology Service, Internal Medicine Section, Department of Medicine and Oncology, Santa Maria della Scaletta Hospital, 40026 Imola, Italy;
- Alma Mater Studiorum, Department of Biomedical and Neuromotor Sciences, University of Bologna, 40126 Bologna, Italy
| | - Viviana Ravagnani
- Rheumatology Unit, Santa Chiara Hospital APSS—Trento, 38122 Trento, Italy;
| | - Palma Scolieri
- Rheumatology Unit, Department of Medical Specialties, Nuovo Regina Margherita Hospital, 00154 Roma, Italy; (P.S.); (V.B.)
| | - Maddalena Larosa
- Division of Rheumatology, Department of Medical Specialties, La Colletta Hospital, ASL 3 Genova, 16132 Genova, Italy; (M.L.); (D.C.); (G.B.)
| | - Elisa Visalli
- Rheumatology Unit, Policlinico San Marco Hospital, 95121 Catania, Italy; (E.V.); (R.F.); (G.A.); (F.D.L.); (Y.D.B.); (R.F.)
| | - Olga Addimanda
- Rheumatology Unit, AUSL of Bologna—Policlinico Sant’Orsola—AOU—IRCCS of Bologna, 40138 Bologna, Italy; (O.A.); (M.R.)
| | - Rosetta Vitetta
- Rheumatology Unit, ASL VC Sant’Andrea Hospital, 13100 Vercelli, Italy; (R.V.); (A.F.); (G.R.)
| | - Alessandro Volpe
- Rheumatology Unit, IRCCS Sacro Cuore Don Calabria Hospital, 37024 Negrar di Valpolicella, Italy; (A.V.); (A.M.)
| | - Alessandra Bezzi
- Internal Medicine and Rheumatology Unit, ASL Romagna—Rimini, 47924 Rimini, Italy (M.C.F.); (F.M.)
| | - Francesco Girelli
- Rheumatology Unit, G.B. Morgagni—L. Pierantoni Hospital, 47121 Forli, Italy; (F.G.); (S.B.)
| | | | - Rosalba Caccavale
- Department of Clinical, Anesthesiological and Cardiovascular Sciences, Sapienza University of Rome, 00185 Rome, Italy; (M.P.); (R.C.)
| | - Eleonora Di Donato
- Internal Medicine and Rheumatology Unit, University Hospital of Parma, 43126 Parma, Italy; (A.B.); (E.D.D.); (G.A.); (D.S.); (G.L.); (A.A.)
| | - Giuditta Adorni
- Internal Medicine and Rheumatology Unit, University Hospital of Parma, 43126 Parma, Italy; (A.B.); (E.D.D.); (G.A.); (D.S.); (G.L.); (A.A.)
| | - Daniele Santilli
- Internal Medicine and Rheumatology Unit, University Hospital of Parma, 43126 Parma, Italy; (A.B.); (E.D.D.); (G.A.); (D.S.); (G.L.); (A.A.)
| | - Gianluca Lucchini
- Internal Medicine and Rheumatology Unit, University Hospital of Parma, 43126 Parma, Italy; (A.B.); (E.D.D.); (G.A.); (D.S.); (G.L.); (A.A.)
| | - Eugenio Arrigoni
- Reumathology Unit, Guglielmo da Saliceto Hospital, 29121 Piacenza, Italy; (E.B.); (E.A.); (I.P.)
| | - Emanuela Sabatini
- Rheumatology Unit, Clinica Medica Institute, SS Annunziata Hospital, 66100 Chieti, Italy; (M.D.P.); (E.S.)
| | - Ilaria Platè
- Reumathology Unit, Guglielmo da Saliceto Hospital, 29121 Piacenza, Italy; (E.B.); (E.A.); (I.P.)
| | - Natalia Mansueto
- Rheumatology Unit, Imperia Hospital, 18100 Imperia, Italy; (R.A.); (N.M.)
| | - Aurora Ianniello
- Rheumatology Outpatient Unit, ASL Novara, 28100 Novara, Italy; (V.N.); (A.I.)
| | - Enrico Fusaro
- Rheumatology Department, Azienda Ospedaliero-Universitaria Città della Salute e della Scienza di Torino, 10126 Torino, Italy; (E.F.); (M.C.D.); (S.P.)
| | - Maria Chiara Ditto
- Rheumatology Department, Azienda Ospedaliero-Universitaria Città della Salute e della Scienza di Torino, 10126 Torino, Italy; (E.F.); (M.C.D.); (S.P.)
| | - Vincenzo Bruzzese
- Rheumatology Unit, Department of Medical Specialties, Nuovo Regina Margherita Hospital, 00154 Roma, Italy; (P.S.); (V.B.)
| | - Dario Camellino
- Division of Rheumatology, Department of Medical Specialties, La Colletta Hospital, ASL 3 Genova, 16132 Genova, Italy; (M.L.); (D.C.); (G.B.)
| | - Gerolamo Bianchi
- Division of Rheumatology, Department of Medical Specialties, La Colletta Hospital, ASL 3 Genova, 16132 Genova, Italy; (M.L.); (D.C.); (G.B.)
| | - Francesca Serale
- Rheumatology Day Hospital and Outpatient Clinic, ASL CN1, 12100 Cuneo, Italy; (M.P.); (F.S.)
| | - Rosario Foti
- Rheumatology Unit, Policlinico San Marco Hospital, 95121 Catania, Italy; (E.V.); (R.F.); (G.A.); (F.D.L.); (Y.D.B.); (R.F.)
| | - Giorgio Amato
- Rheumatology Unit, Policlinico San Marco Hospital, 95121 Catania, Italy; (E.V.); (R.F.); (G.A.); (F.D.L.); (Y.D.B.); (R.F.)
| | - Francesco De Lucia
- Rheumatology Unit, Policlinico San Marco Hospital, 95121 Catania, Italy; (E.V.); (R.F.); (G.A.); (F.D.L.); (Y.D.B.); (R.F.)
| | - Ylenia Dal Bosco
- Rheumatology Unit, Policlinico San Marco Hospital, 95121 Catania, Italy; (E.V.); (R.F.); (G.A.); (F.D.L.); (Y.D.B.); (R.F.)
| | - Roberta Foti
- Rheumatology Unit, Policlinico San Marco Hospital, 95121 Catania, Italy; (E.V.); (R.F.); (G.A.); (F.D.L.); (Y.D.B.); (R.F.)
| | - Massimo Reta
- Rheumatology Unit, AUSL of Bologna—Policlinico Sant’Orsola—AOU—IRCCS of Bologna, 40138 Bologna, Italy; (O.A.); (M.R.)
| | - Alessia Fiorenza
- Rheumatology Unit, ASL VC Sant’Andrea Hospital, 13100 Vercelli, Italy; (R.V.); (A.F.); (G.R.)
| | - Guido Rovera
- Rheumatology Unit, ASL VC Sant’Andrea Hospital, 13100 Vercelli, Italy; (R.V.); (A.F.); (G.R.)
| | - Antonio Marchetta
- Rheumatology Unit, IRCCS Sacro Cuore Don Calabria Hospital, 37024 Negrar di Valpolicella, Italy; (A.V.); (A.M.)
| | - Maria Cristina Focherini
- Internal Medicine and Rheumatology Unit, ASL Romagna—Rimini, 47924 Rimini, Italy (M.C.F.); (F.M.)
| | - Fabio Mascella
- Internal Medicine and Rheumatology Unit, ASL Romagna—Rimini, 47924 Rimini, Italy (M.C.F.); (F.M.)
| | - Simone Bernardi
- Rheumatology Unit, G.B. Morgagni—L. Pierantoni Hospital, 47121 Forli, Italy; (F.G.); (S.B.)
| | - Gilda Sandri
- Rheumatology Unit, University of Modena and Reggio Emilia, 41125 Modena, Italy; (G.S.); (D.G.); (C.S.)
| | - Dilia Giuggioli
- Rheumatology Unit, University of Modena and Reggio Emilia, 41125 Modena, Italy; (G.S.); (D.G.); (C.S.)
| | - Carlo Salvarani
- Rheumatology Unit, University of Modena and Reggio Emilia, 41125 Modena, Italy; (G.S.); (D.G.); (C.S.)
| | | | | | - Giulio Ferrero
- Unit of Diagnostic and Interventional Radiology, Santa Corona Hospital, 17027 Pietra Ligure, Italy;
| | - Alarico Ariani
- Internal Medicine and Rheumatology Unit, University Hospital of Parma, 43126 Parma, Italy; (A.B.); (E.D.D.); (G.A.); (D.S.); (G.L.); (A.A.)
| | - Simone Parisi
- Rheumatology Department, Azienda Ospedaliero-Universitaria Città della Salute e della Scienza di Torino, 10126 Torino, Italy; (E.F.); (M.C.D.); (S.P.)
| |
Collapse
|
9
|
Xu F, Li Z, Liu T, Pang X, Fan C, Jiang H. The role of cellular senescence in the pathogenesis of Rheumatoid Arthritis: Focus on IL-6 as a target gene. Cytokine 2024; 184:156762. [PMID: 39326197 DOI: 10.1016/j.cyto.2024.156762] [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: 05/25/2024] [Revised: 08/30/2024] [Accepted: 09/13/2024] [Indexed: 09/28/2024]
Abstract
BACKGROUND Rheumatoid arthritis is a chronic autoimmune disease. However, the specific role of senescence in rheumatoid arthritis (RA) is unknown. This study aimed to identify potential aging-related genes that have diagnostic and therapeutic value for RA. METHODS The GSE89408 dataset was downloaded from the Gene Expression Omnibus (GEO). Aging-related genes were downloaded from the HAGR database. Differentially expressed genes (DEGs) were subsequently identified with the "edgeR" tool. Next, hub genes were identified with a PPI network and CytoHubba analysis. Receiver operating characteristic (ROC) curves were used to evaluate the diagnostic value of these hub genes. Immune infiltration analysis was performed with the CIBERSORT algorithm. Additionally, molecular docking was performed with CB-Dock2. Finally, correlation experiments were performed to validate the bioinformatics and molecular docking results. RESULTS A total of 22 ADEGs were identified. Combined PPI network and CytoHubba analyses identified a total of 7 hub genes, including IL-6, IL7R, IL2RG, CDK1, PTGS2, and LEP, which are associated mainly with inflammation and immune responses. ROC analysis revealed that the hub genes were highly predictive of RA. Analysis of immune infiltration revealed that the 6 hub genes were positively associated with M1 macrophages. Validation experiments revealed that the inhibition of IL-6 significantly decreased the degree of synovial fibroblast (FLS) senescence. Furthermore, molecular docking and validation experiments revealed that IL-6 is a potential target for drug therapy. CONCLUSION This study demonstrated that RA-FLS senescence may promote the development of RA via inflammatory and immune mechanisms. Seven hub genes were identified, of which IL-6 is a reliable biomarker for the diagnosis and treatment of RA.
Collapse
Affiliation(s)
- Fengxia Xu
- Clinical Research Experiment Center, The First Affiliated Hospital of Anhui University of Chinese Medicine, Hefei 230012, Anhui, China; College of Pharmacy, Anhui University of Chinese Medicine, Hefei 230012, Anhui, China
| | - Zhen Li
- Clinical Research Experiment Center, The First Affiliated Hospital of Anhui University of Chinese Medicine, Hefei 230012, Anhui, China; College of Pharmacy, Anhui University of Chinese Medicine, Hefei 230012, Anhui, China
| | - Tao Liu
- Clinical Research Experiment Center, The First Affiliated Hospital of Anhui University of Chinese Medicine, Hefei 230012, Anhui, China; College of Pharmacy, Anhui University of Chinese Medicine, Hefei 230012, Anhui, China
| | - Xue Pang
- Clinical Research Experiment Center, The First Affiliated Hospital of Anhui University of Chinese Medicine, Hefei 230012, Anhui, China; College of Pharmacy, Anhui University of Chinese Medicine, Hefei 230012, Anhui, China
| | - Chang Fan
- Clinical Research Experiment Center, The First Affiliated Hospital of Anhui University of Chinese Medicine, Hefei 230012, Anhui, China
| | - Hui Jiang
- Clinical Research Experiment Center, The First Affiliated Hospital of Anhui University of Chinese Medicine, Hefei 230012, Anhui, China; College of Pharmacy, Anhui University of Chinese Medicine, Hefei 230012, Anhui, China.
| |
Collapse
|
10
|
Li H, Lin S, Wang Y, Shi Y, Fang X, Wang J, Cui H, Bian Y, Qi X. Immunosenescence: A new direction in anti-aging research. Int Immunopharmacol 2024; 141:112900. [PMID: 39137628 DOI: 10.1016/j.intimp.2024.112900] [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: 02/16/2024] [Revised: 07/22/2024] [Accepted: 08/05/2024] [Indexed: 08/15/2024]
Abstract
The immune system is a major regulatory system of the body, that is composed of immune cells, immune organs, and related signaling factors. As an organism ages, observable age-related changes in the function of the immune system accumulate in a process described as 'immune aging. Research has shown that the impact of aging on immunity is detrimental, with various dysregulated responses that affect the function of immune cells at the cellular level. For example, increased aging has been shown to result in the abnormal chemotaxis of neutrophils and decreased phagocytosis of macrophages. Age-related diminished functionality of immune cell types has direct effects on host fitness, leading to poorer responses to vaccination, more inflammation and tissue damage, as well as autoimmune disorders and the inability to control infections. Similarly, age impacts the function of the immune system at the organ level, resulting in decreased hematopoietic function in the bone marrow, a gradual deficiency of catalase in the thymus, and thymic atrophy, resulting in reduced production of related immune cells such as B cells and T cells, further increasing the risk of autoimmune disorders in the elderly. As the immune function of the body weakens, aging cells and inflammatory factors cannot be cleared, resulting in a cycle of increased inflammation that accumulates over time. Cumulatively, the consequences of immune aging increase the likelihood of developing age-related diseases, such as Alzheimer's disease, atherosclerosis, and osteoporosis, among others. Therefore, targeting the age-related changes that occur within cells of the immune system might be an effective anti-aging strategy. In this article, we summarize the relevant literature on immune aging research, focusing on its impact on aging, in hopes of providing new directions for anti-aging research.
Collapse
Affiliation(s)
- Hanzhou Li
- Tianjin University of Traditional Chinese Medicine, Tianjin, China; Tianjin Union Medical Center, Tianjin, China
| | - Shan Lin
- Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Yuming Wang
- Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Yuexuan Shi
- Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Xixing Fang
- College of Traditional Chinese Medicine, Changchun University of Traditional Chinese Medicine, Changchun, China
| | - Jida Wang
- Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Huantian Cui
- Yunnan University of Chinese Medicine, Yunnan, China.
| | - Yuhong Bian
- Tianjin University of Traditional Chinese Medicine, Tianjin, China.
| | - Xin Qi
- Tianjin University of Traditional Chinese Medicine, Tianjin, China; Tianjin Union Medical Center, Tianjin, China.
| |
Collapse
|
11
|
Bolton C. Review of evidence linking exposure to environmental stressors and associated alterations in the dynamics of immunosenescence (ISC) with the global increase in multiple sclerosis (MS). Immun Ageing 2024; 21:73. [PMID: 39438909 PMCID: PMC11494837 DOI: 10.1186/s12979-024-00473-w] [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: 08/13/2024] [Accepted: 10/07/2024] [Indexed: 10/25/2024]
Abstract
Historical survey confirms that, over the latter part of the 20th century, autoimmune-based diseases, including multiple sclerosis (MS), have shown a worldwide increase in incidence and prevalence. Analytical population studies have established that the exponential rise in MS is not solely due to improvements in diagnosis and healthcare but relates to an increase in autoimmune risk factors. Harmful environmental exposures, including non-communicable social determinants of health, anthropogens and indigenous or transmissible microbes, constitute a group of causal determinants that have been closely linked with the global rise in MS cases. Exposure to environmental stressors has profound effects on the adaptive arm of the immune system and, in particular, the associated intrinsic process of immune ageing or immunosenescence (ISC). Stressor-related disturbances to the dynamics of ISC include immune cell-linked untimely or premature (p) alterations and an accelerated replicative (ar) change. A recognised immune-associated feature of MS is pISC and current evidence supports the presence of an arISC during the disease. Moreover, collated data illustrates the immune-associated alterations that characterise pISC and arISC are inducible by environmental stressors strongly implicated in causing duplicate changes in adaptive immune cells during MS. The close relationship between exposure to environmental risk factors and the induction of pISC and arISC during MS offers a valid mechanism through which pro-immunosenescent stressors may act and contribute to the recorded increase in the global rate and number of new cases of the disease. Confirmation of alterations to the dynamics of ISC during MS provides a rational and valuable therapeutic target for the use of senolytic drugs to either prevent accumulation and enhance ablation of less efficient untimely senescent adaptive immune cells or decelerate the dysregulated process of replicative proliferation. A range of senotherapeutics are available including kinase and transcriptase inhibitors, rapalogs, flavanols and genetically-engineered T cells and the use of selective treatments to control emerging and unspecified aspects of pISC and arISC are discussed.
Collapse
|
12
|
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
|
13
|
Parab A, Bhatt LK. T-cell metabolism in rheumatoid arthritis: focus on mitochondrial and lysosomal dysfunction. Immunopharmacol Immunotoxicol 2024; 46:378-384. [PMID: 38478010 DOI: 10.1080/08923973.2024.2330645] [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: 06/07/2023] [Accepted: 03/08/2024] [Indexed: 03/20/2024]
Abstract
INTRODUCTION Rheumatoid arthritis (RA) is a chronic inflammatory disease characterized by immune cell dysregulation, synovial hyperplasia, and progressive cartilage destruction. The loss of immunological self-tolerance against autoantigens is the crucial insult responsible for the pathogenesis of RA. These immune abnormalities are experienced many years before the onset of clinical arthritis. OBJECTIVE This review aims to discuss the metabolic status of T-cells in RA and focuses mainly on mitochondrial and lysosomal dysfunctions involved in altering the T-cell metabolism. DISCUSSION T-cells are identified as the primary initiators of immunological abnormalities in RA. These RA T-cells show a distinct metabolic pattern compared to the healthy individuals. Dampened glycolytic flux, poor ATP production, and shifting of glucose to the pentose phosphate pathway resulting in increased NADPH and decreased ROS levels are the common metabolic patterns observed in RA T-cells. Defective mtDNA due to lack of MRE11A gene, a key molecular actor for resection, and inefficient lysosomal function due to misplacement of AMPK on the lysosomal surface were found to be responsible for mitochondrial and lysosome dysfunction in RA. Targeting this mechanism in RA can alleviate aggressive T-cell phenotype and may control the severity of RA.
Collapse
Affiliation(s)
- Asmita Parab
- Department of Pharmacology, SVKM's Dr. Bhanuben Nanavati College of Pharmacy, Mumbai, India
| | - Lokesh Kumar Bhatt
- Department of Pharmacology, SVKM's Dr. Bhanuben Nanavati College of Pharmacy, Mumbai, India
| |
Collapse
|
14
|
Sagy I, Schwarzfuchs O, Zeller L, Ling E, Babiev AS, Abu-Shakra M. Short- and Long-Term Mortality of Hospitalized Patients With Autoimmune Rheumatic Diseases and Serious Infections: A National Cohort Study. J Rheumatol 2024; 51:517-522. [PMID: 38302166 DOI: 10.3899/jrheum.2023-1063] [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] [Accepted: 01/22/2024] [Indexed: 02/03/2024]
Abstract
OBJECTIVE Infectious conditions are a significant cause of mortality in autoimmune rheumatic diseases (ARD). Among patients hospitalized with an infection, we compared in-hospital and long-term (3-year) mortality between those with and without ARD. METHODS This retrospective analysis included members of the largest health maintenance organization in Israel, aged > 18 years at the first episode of infection, who required hospitalization during 2003-2019. We compared in-hospital mortality and the results of a 3-year landmark analysis of those who survived the index hospitalization between patients with ARD, according to disease subgroups, and patients without ARD. Additionally, we compared mortality outcomes among patients with ARD, according to subgroup diagnosis, matched in a 1:3 ratio by age, sex, and ethnicity to patients without ARD. RESULTS Included were 365,247 patients who were admitted for the first time with the diagnosis of a serious infection. Of these, we identified 9755 with rheumatoid arthritis (RA), 1351 with systemic lupus erythematosus, 2120 with spondyloarthritis (SpA), 584 with systemic sclerosis, and 3214 with vasculitis. In a matched multivariate analysis, the risk for in-hospital mortality was lower among patients with RA (odds ratio [OR] 0.89, 95% CI 0.81-0.97) and SpA (OR 0.77, 95% CI 0.63-0.94). In a similar analysis, the risk of 3-year mortality was lower among patients with RA (hazard ratio [HR] 0.82, 95% CI 0.78-0.86) and vasculitis (HR 0.86, 95% CI 0.80-0.93). CONCLUSION Among patients hospitalized for an infection, the risk of in-hospital and 3-year mortality was not increased among those with ARD compared to those without ARD.
Collapse
Affiliation(s)
- Iftach Sagy
- I. Sagy, MD, PhD, Rheumatology Disease Unit, and Clinical Research Center, Soroka University Medical Center, and Faculty of Health Sciences, Ben Gurion University of the Negev;
| | - Omer Schwarzfuchs
- O. Schwarzfuchs, BSc, Faculty of Health Sciences, Ben Gurion University of the Negev
| | - Lior Zeller
- L. Zeller, MD, E. Ling, MD, PhD, M. Abu-Shakra, MD, Rheumatology Disease Unit, Soroka University Medical Center, and Faculty of Health Sciences, Ben Gurion University of the Negev
| | - Eduard Ling
- L. Zeller, MD, E. Ling, MD, PhD, M. Abu-Shakra, MD, Rheumatology Disease Unit, Soroka University Medical Center, and Faculty of Health Sciences, Ben Gurion University of the Negev
| | - Amit Shira Babiev
- A.S. Babiev, BSc, Clinical Research Center, Soroka University Medical Center, Beer Sheva, Israel
| | - Mahmoud Abu-Shakra
- L. Zeller, MD, E. Ling, MD, PhD, M. Abu-Shakra, MD, Rheumatology Disease Unit, Soroka University Medical Center, and Faculty of Health Sciences, Ben Gurion University of the Negev
| |
Collapse
|
15
|
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
|
16
|
Bi J, Zhang C, Lu C, Mo C, Zeng J, Yao M, Jia B, Liu Z, Yuan P, Xu S. Age-related bone diseases: Role of inflammaging. J Autoimmun 2024; 143:103169. [PMID: 38340675 DOI: 10.1016/j.jaut.2024.103169] [Citation(s) in RCA: 19] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2023] [Revised: 01/03/2024] [Accepted: 01/19/2024] [Indexed: 02/12/2024]
Abstract
Bone aging is characterized by an imbalance in the physiological and pathological processes of osteogenesis, osteoclastogenesis, adipogenesis, and chondrogenesis, resulting in exacerbated bone loss and the development of age-related bone diseases, including osteoporosis, osteoarthritis, rheumatoid arthritis, and periodontitis. Inflammaging, a novel concept in the field of aging research, pertains to the persistent and gradual escalation of pro-inflammatory reactions during the aging process. This phenomenon is distinguished by its low intensity, systemic nature, absence of symptoms, and potential for management. The mechanisms by which inflammaging contribute to age-related chronic diseases, particularly in the context of age-related bone diseases, remain unclear. The precise manner in which systemic inflammation induces bone aging and consequently contributes to the development of age-related bone diseases has yet to be fully elucidated. This article primarily examines the mechanisms underlying inflammaging and its association with age-related bone diseases, to elucidate the potential mechanisms of inflammaging in age-related bone diseases and offer insights for developing preventive and therapeutic strategies for such conditions.
Collapse
Affiliation(s)
- Jiaming Bi
- Department of Endodontics, Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou, Guangdong, China
| | - Caimei Zhang
- Department of Endodontics, Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou, Guangdong, China
| | - Caihong Lu
- Department of Endodontics, Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou, Guangdong, China
| | - Chuzi Mo
- Department of Endodontics, Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou, Guangdong, China
| | - Jiawei Zeng
- Department of Endodontics, Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou, Guangdong, China
| | - Mingyan Yao
- Department of Endocrinology, The Third Hospital of Hebei Medical University, Shijiazhuang, China; Department of Endocrinology, Baoding No.1 Central Hospital, Baoding, China
| | - Bo Jia
- Department of Oral and Maxillofacial Surgery, Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou, Guangdong, China
| | - Zhongjun Liu
- Department of Endodontics, Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou, Guangdong, China
| | - Peiyan Yuan
- Department of Endodontics, Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou, Guangdong, China.
| | - Shuaimei Xu
- Department of Endodontics, Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou, Guangdong, China.
| |
Collapse
|
17
|
Gautam S, Kumar R, Kumar U, Kumar S, Luthra K, Dada R. Yoga maintains Th17/Treg cell homeostasis and reduces the rate of T cell aging in rheumatoid arthritis: a randomized controlled trial. Sci Rep 2023; 13:14924. [PMID: 37696876 PMCID: PMC10495372 DOI: 10.1038/s41598-023-42231-w] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2023] [Accepted: 09/07/2023] [Indexed: 09/13/2023] Open
Abstract
The pathogenesis of rheumatoid arthritis (RA) is characterized by a Th17/Treg cell imbalance. A pro-inflammatory cytokine milieu that promotes the continued proliferation of Th17 cells is related to the development of autoinflammation. In RA, T cells have several hallmarks of cellular aging, and they accumulate DNA damage, predisposing to the occurrence of mutations and epigenetic alterations. Since the onset, progression, and treatment response are influenced by a variety of external stressors and environmental factors, this study aimed to evaluate the impact of 8-week yoga practice on disease severity, T cell subsets, markers of T cell ageing and inflammation, epigenetic alterations and gene expression patterns in active RA patients on standard disease-modifying anti-rheumatic drugs (DMARDs). A total of 64 participants with active RA were randomized into 2 groups, yoga group (n = 32) or non-yoga group (n = 32); that were assessed for disease severity, at baseline and after 8 week duration, for Disease Activity Score (DAS28-ESR), T cell subsets [Th17 (CD3+ CD4+ IL17+ RORγt+) cells and Treg (CD3+ CD4+ CD25+ CD127-Foxp3+) cells], markers of T cell aging [aged Th17 cells (CD3+ CD4+ IL17+ RORγt+ CD28-) and aged Treg cells (CD3+ CD4+ CD25+ CD127-Foxp3+ CD28-)], pro-inflammatory markers [IL-6, and IL-17], anti-inflammatory markers [TGF-β, and IL-10], epigenetic alterations [5-methyl cytosine, 5-hydroxymethyl cytosine, and HDAC1] and gene expression patterns [RORγt, FoxP3, IL-17, IL-6, TGF-β, CXCL2, CXCR2, and JUN]. In yoga group, there was a significant improvement in DAS28-ESR scores at the end of 8-weeks of yoga program. The Th17 cells and aged T cell subsets showed a significant decline whereas Treg cell population showed a significant elevation in yoga group. There were significant improvements observed in epigenetic markers as well as inflammatory markers post 8-weeks of yoga practice. The yoga group showed downregulation of RORγt, IL-17, IL-6, CXCL2, CXCR2, and upregulation of FoxP3 and TGF-β transcripts. Yoga enables the maintenance of immune-homeostasis as evident by increased Treg cell population and reduced Th17 cell population. Yoga reduces the rate of immunological aging in T cells, as seen by the reduction in population of aged Th17 cells and aged Treg cells. Yoga positively modifies transcriptome and epigenome by normalization of various inflammatory markers, gene expression patterns and epigenetic alterations. Taken together, yoga reduces RA severity, and aids in immune-modulation and hence can be beneficial as an adjunct therapy.
Collapse
Affiliation(s)
- Surabhi Gautam
- Department of Anatomy, Molecular Reproduction and Genetics Facility, All India Institute of Medical Sciences (AIIMS), New Delhi, India
- Department of Orthopaedics, Emory Musculoskeletal Institute, Emory University School of Medicine, Atlanta, USA
| | - Romsha Kumar
- Department of Biochemistry, All India Institute of Medical Sciences (AIIMS), New Delhi, India
| | - Uma Kumar
- Department of Rheumatology, All India Institute of Medical Sciences (AIIMS), New Delhi, India
| | - Sanjeev Kumar
- Department of Biochemistry, All India Institute of Medical Sciences (AIIMS), New Delhi, India
| | - Kalpana Luthra
- Department of Biochemistry, All India Institute of Medical Sciences (AIIMS), New Delhi, India
| | - Rima Dada
- Department of Anatomy, Molecular Reproduction and Genetics Facility, All India Institute of Medical Sciences (AIIMS), New Delhi, India.
| |
Collapse
|
18
|
Garley M, Omeljaniuk WJ, Motkowski R, Ratajczak-Wrona W, JabŁOŃSka E, Filipkowski D, Charkiewicz AE. Immunoaging - the effect of age on serum levels of NET biomarkers in men: a pilot study. Int J Occup Med Environ Health 2023; 36:333-348. [PMID: 37681423 PMCID: PMC10664002 DOI: 10.13075/ijomeh.1896.02125] [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: 12/13/2022] [Accepted: 05/25/2023] [Indexed: 09/09/2023] Open
Abstract
OBJECTIVES The study aimed to evaluate the impact of aging on the formation of neutrophil extracellular traps (NETs). The impaired formation of NETs is the cause of an abnormal innate immune response. MATERIAL AND METHODS The study included a total of 45 healthy male subjects of different age groups. Whole blood was collected from the subjects, and the concentration of myeloperoxidase (MPO), the main biocidal protein in NETs, was determined in serum using ELISA. The serum levels of circulating free DNA (cfDNA), which are the structural basis of NETs, were also measured by fluorescence. In addition, the white blood cell count was determined, whole blood smear was evaluated, and the neutrophillymphocyte ratio was calculated. The variations in the levels of NET biomarkers were analyzed in different age groups. RESULTS The low levels of MPO (243.70 ng/ml) and cfDNA (6.24 ng/100 μl) in boys indicated neutrophil insufficiency for NETosis in children. A progressive increase in the levels of MPO and cfDNA with age was observed among adolescents (420.91, p = 0.04; 13.55, p = 0.03, respectively), with the highest level noted in the healthy adult group (466.58, p = 0.01; 14.07, p = 0.01, respectively). The levels of the studied parameters were comparable in adolescents and young adults, which proved that the NETosis process was appropriate and suggested the attainment of neutrophil maturity for the release of NETs in adolescence. The levels of MPO and cfDNA were low in older men (225.46, p < 0.01; 5.19, p < 0.01, respectively) indicating impaired NET formation. CONCLUSIONS Data on the generation of NETs in different age groups obtained in this study can allow a better understanding of the ontogenesis of the immune system in terms of the course of NETosis, and also indicate the need to support nonspecific responses in children and adults. Further research should be performed to determine the possibility of regulating the NETosis process. Int J Occup Med Environ Health. 2023;36(3):333-48.
Collapse
Affiliation(s)
- Marzena Garley
- Medical University of Bialystok, Department of Immunology, Białystok, Poland
| | | | - RadosŁAw Motkowski
- Medical University of Bialystok, University Children's Clinical Hospital, Department of Pediatrics, Rheumatology, Immunology and Metabolic Bone Diseases, Białystok, Poland
| | | | - Ewa JabŁOŃSka
- Medical University of Bialystok, Department of Immunology, Białystok, Poland
| | - Daniel Filipkowski
- Medical University of Bialystok, Students' Scientific Society, Department of Immunology, Białystok, Poland
| | | |
Collapse
|
19
|
Amezcua-Guerra LM, Espinosa-Bautista F, Hopf-Estandía K, Valdivieso-Ruiz M, Coronel D, Robledo S, Ramos-Rosillo V, Del Rocío Martínez-Alvarado M, Patlán M, Páez A, Silveira LH, Tavera-Alonso C, Massó F, Soto-Fajardo C, Pineda C. Senescent CD4 +CD28 null cells are increased in chronic hyperuricemia, show aberrant effector phenotypes, and are reversed after allopurinol therapy: a proof-of-concept pilot study. Clin Rheumatol 2023:10.1007/s10067-023-06595-8. [PMID: 37072512 DOI: 10.1007/s10067-023-06595-8] [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: 02/20/2023] [Revised: 04/04/2023] [Accepted: 04/05/2023] [Indexed: 04/20/2023]
Abstract
To characterize CD4+CD28null cells in chronic hyperuricemia and investigate whether allopurinol could restore CD28 expression and the balance of T helper phenotypes. Asymptomatic individuals with chronic hyperuricemia and ultrasonographic findings evocative of urate deposition in the joints. Age- and gender-matched normouricemic individuals were also studied. Oral allopurinol at 150 mg/day for 4 weeks, followed by 300 mg/day through week 12. Color-flow cytometry on peripheral blood mononuclear cells (PBMC) with antibodies against CD4, CD28, T-bet (Th1), GATA-3 (Th2), and RORγt (Th17). Six patients (five men, median age of 53 years) and seven controls were studied. At baseline, hyperuricemic patients had more CD4+CD28null/CD4+ cells than normouricemic subjects (36.8% vs. 6.1%; p = 0.001), with a predominance of T-bet+ cells (98.5% vs. 6.6%; p = 0.001) and few RORγt+ cells (0.7% vs. 89.4%; p = 0.014). In hyperuricemic patients, the number of CD4+ cells/10,000 PBMC was similar before and after allopurinol (3378 vs. 3954; p = 0.843). Conversely, CD4+CD28null cells decreased from 36.8% (23.0-43.7) to 15.8% (4.7-28.1; p = 0.031). CD4+CD28nullT-bet+ cells decreased from 98.5% (95.0-99.4) to 88.3% (75.2-98.9; p = 0.062), CD4+CD28nullGATA-3+ cells increased from 0% (0-4.0) to 2.8% (0.1-15.6; p = 0.156), and CD4+CD28nullRORγt+ cells increased from 0.7% (0.4-7.0) to 4.5% (1.3-28.1; p = 0.031). The CD4+CD28null cell subset is abnormally expanded in chronic hyperuricemia, despite the absence of overt urate-related disease. Allopurinol may partially restore CD28 expression on CD4+ cells while enhancing the homeostatic balance of T helper phenotypes. ClinicalTrials.gov, number NCT04012294.
Collapse
Affiliation(s)
- Luis M Amezcua-Guerra
- Immunology Department, Instituto Nacional de Cardiología Ignacio Chávez, Juan Badiano 1, Sección XVI, Tlalpan, 14080, Mexico City, Mexico.
- Health Care Department, Universidad Autónoma Metropolitana Xochimilco, Mexico City, Mexico.
| | - Fernanda Espinosa-Bautista
- Immunology Department, Instituto Nacional de Cardiología Ignacio Chávez, Juan Badiano 1, Sección XVI, Tlalpan, 14080, Mexico City, Mexico
- Master in Chemobiological Sciences, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, Mexico City, Mexico
- Red MEDICI, Facultad de Estudios Superiores Iztacala, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | - Karen Hopf-Estandía
- Immunology Department, Instituto Nacional de Cardiología Ignacio Chávez, Juan Badiano 1, Sección XVI, Tlalpan, 14080, Mexico City, Mexico
| | - Melisa Valdivieso-Ruiz
- Rheumatology Division, Instituto Nacional de Rehabilitación Luis Guillermo Ibarra Ibarra, Mexico City, Mexico
| | - Dania Coronel
- Immunology Department, Instituto Nacional de Cardiología Ignacio Chávez, Juan Badiano 1, Sección XVI, Tlalpan, 14080, Mexico City, Mexico
| | - Sandra Robledo
- Immunology Department, Instituto Nacional de Cardiología Ignacio Chávez, Juan Badiano 1, Sección XVI, Tlalpan, 14080, Mexico City, Mexico
- Master in Chemobiological Sciences, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, Mexico City, Mexico
- Red MEDICI, Facultad de Estudios Superiores Iztacala, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | - Varna Ramos-Rosillo
- Immunology Department, Instituto Nacional de Cardiología Ignacio Chávez, Juan Badiano 1, Sección XVI, Tlalpan, 14080, Mexico City, Mexico
| | | | - Mariana Patlán
- UNAM/INC Translational Research Unit, Instituto Nacional de Cardiología Ignacio Chávez, Mexico City, Mexico
| | - Araceli Páez
- UNAM/INC Translational Research Unit, Instituto Nacional de Cardiología Ignacio Chávez, Mexico City, Mexico
| | - Luis H Silveira
- Rheumatology Department, Instituto Nacional de Cardiología Ignacio Chávez, Mexico City, Mexico
| | | | - Felipe Massó
- UNAM/INC Translational Research Unit, Instituto Nacional de Cardiología Ignacio Chávez, Mexico City, Mexico
| | - Carina Soto-Fajardo
- Rheumatology Division, Instituto Nacional de Rehabilitación Luis Guillermo Ibarra Ibarra, Mexico City, Mexico
| | - Carlos Pineda
- Rheumatology Division, Instituto Nacional de Rehabilitación Luis Guillermo Ibarra Ibarra, Mexico City, Mexico
| |
Collapse
|
20
|
Weyand CM, Wu B, Huang T, Hu Z, Goronzy JJ. Mitochondria as disease-relevant organelles in rheumatoid arthritis. Clin Exp Immunol 2023; 211:208-223. [PMID: 36420636 PMCID: PMC10038327 DOI: 10.1093/cei/uxac107] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Revised: 10/18/2022] [Accepted: 11/18/2022] [Indexed: 11/25/2022] Open
Abstract
Mitochondria are the controllers of cell metabolism and are recognized as decision makers in cell death pathways, organizers of cytoplasmic signaling networks, managers of cellular stress responses, and regulators of nuclear gene expression. Cells of the immune system are particularly dependent on mitochondrial resources, as they must swiftly respond to danger signals with activation, trafficking, migration, and generation of daughter cells. Analogously, faulty immune responses that lead to autoimmunity and tissue inflammation rely on mitochondria to supply energy, cell building blocks and metabolic intermediates. Emerging data endorse the concept that mitochondrial fitness, and the lack of it, is of particular relevance in the autoimmune disease rheumatoid arthritis (RA) where deviations of bioenergetic and biosynthetic flux affect T cells during early and late stages of disease. During early stages of RA, mitochondrial deficiency allows naïve RA T cells to lose self-tolerance, biasing fundamental choices of the immune system toward immune-mediated tissue damage and away from host protection. During late stages of RA, mitochondrial abnormalities shape the response patterns of RA effector T cells engaged in the inflammatory lesions, enabling chronicity of tissue damage and tissue remodeling. In the inflamed joint, autoreactive T cells partner with metabolically reprogrammed tissue macrophages that specialize in antigen-presentation and survive by adapting to the glucose-deplete tissue microenvironment. Here, we summarize recent data on dysfunctional mitochondria and mitochondria-derived signals relevant in the RA disease process that offer novel opportunities to deter autoimmune tissue inflammation by metabolic interference.
Collapse
Affiliation(s)
- Cornelia M Weyand
- Department of Medicine, Mayo Clinic Alix School of Medicine, Rochester, MN 55905, USA
- Department of Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Bowen Wu
- Department of Medicine, Mayo Clinic Alix School of Medicine, Rochester, MN 55905, USA
| | - Tao Huang
- Department of Medicine, Mayo Clinic Alix School of Medicine, Rochester, MN 55905, USA
| | - Zhaolan Hu
- Department of Medicine, Mayo Clinic Alix School of Medicine, Rochester, MN 55905, USA
| | - Jörg J Goronzy
- Department of Medicine, Mayo Clinic Alix School of Medicine, Rochester, MN 55905, USA
- Department of Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA
| |
Collapse
|
21
|
Lu Y, Ruan Y, Hong P, Rui K, Liu Q, Wang S, Cui D. T-cell senescence: A crucial player in autoimmune diseases. Clin Immunol 2023; 248:109202. [PMID: 36470338 DOI: 10.1016/j.clim.2022.109202] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2022] [Revised: 10/24/2022] [Accepted: 11/23/2022] [Indexed: 12/12/2022]
Abstract
Senescent T cells are proliferative disabled lymphocytes that lack antigen-specific responses. The development of T-cell senescence in autoimmune diseases contributes to immunological disorders and disease progression. Senescent T cells lack costimulatory markers with the reduction of T cell receptor repertoire and the uptake of natural killer cell receptors. Senescent T cells exert cytotoxic effects through the expression of perforin, granzymes, tumor necrosis factor, and other molecules without the antigen-presenting process. DNA damage accumulation, telomere damage, and limited DNA repair capacity are important features of senescent T cells. Impaired mitochondrial function and accumulation of reactive oxygen species contribute to T cell senescence. Alleviation of T-cell senescence could provide potential targets for the treatment of autoimmune diseases.
Collapse
Affiliation(s)
- Yinyun Lu
- Department of Infectious Diseases, Shaoxing People's Hospital, Shaoxing, China
| | - Yongchun Ruan
- Department of Infectious Diseases, Shaoxing People's Hospital, Shaoxing, China
| | - Pan Hong
- Department of Hematology, Shaoxing People's Hospital, Shaoxing, China
| | - Ke Rui
- Department of Transfusion, Shaoxing People's Hospital, Shaoxing, China
| | - Qi Liu
- Department of Laboratory Medicine, Affiliated Hospital of Jiangsu University, Zhenjiang, China.
| | - Shengjun Wang
- Department of Immunology, Jiangsu Key Laboratory of Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang, China; Department of Laboratory Medicine, The Affiliated People's Hospital, Jiangsu University, Zhenjiang, China.
| | - Dawei Cui
- The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.
| |
Collapse
|
22
|
Vitamin D as a Shield against Aging. Int J Mol Sci 2023; 24:ijms24054546. [PMID: 36901976 PMCID: PMC10002864 DOI: 10.3390/ijms24054546] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Revised: 02/17/2023] [Accepted: 02/22/2023] [Indexed: 03/02/2023] Open
Abstract
Aging can be seen as a physiological progression of biomolecular damage and the accumulation of defective cellular components, which trigger and amplify the process, toward whole-body function weakening. Senescence initiates at the cellular level and consists in an inability to maintain homeostasis, characterized by the overexpression/aberrant expression of inflammatory/immune/stress responses. Aging is associated with significant modifications in immune system cells, toward a decline in immunosurveillance, which, in turn, leads to chronic elevation of inflammation/oxidative stress, increasing the risk of (co)morbidities. Albeit aging is a natural and unavoidable process, it can be regulated by some factors, like lifestyle and diet. Nutrition, indeed, tackles the mechanisms underlying molecular/cellular aging. Many micronutrients, i.e., vitamins and elements, can impact cell function. This review focuses on the role exerted by vitamin D in geroprotection, based on its ability to shape cellular/intracellular processes and drive the immune response toward immune protection against infections and age-related diseases. To this aim, the main biomolecular paths underlying immunosenescence and inflammaging are identified as biotargets of vitamin D. Topics such as heart and skeletal muscle cell function/dysfunction, depending on vitamin D status, are addressed, with comments on hypovitaminosis D correction by food and supplementation. Albeit research has progressed, still limitations exist in translating knowledge into clinical practice, making it necessary to focus attention on the role of vitamin D in aging, especially considering the growing number of older individuals.
Collapse
|
23
|
Zhang L, Jiao W, Deng H, Hu C, Xu J, Yu J, Liu L, Zhang M, Liu J, Chen G. High-throughput Treg cell receptor sequencing reveals differential immune repertoires in rheumatoid arthritis with kidney deficiency. PeerJ 2023; 11:e14837. [PMID: 36751634 PMCID: PMC9899432 DOI: 10.7717/peerj.14837] [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: 08/08/2022] [Accepted: 01/10/2023] [Indexed: 02/05/2023] Open
Abstract
Background Regulatory T (Treg) cells are important immune cells that are regulated by adaptive immunity in the composition of Treg-cell subsets and T-cell receptors (TCRs). Treg cells are related to most autoimmune diseases, such as rheumatoid arthritis (RA). In traditional Chinese medicine (TCM), RA is typically attributed to kidney deficiency (KD) associated with the immunosenescence that causes immune dysfunction and the impaired function of Treg cells. So far, however, no mechanism related to KD and immune repertoires has been identified in RA. Methods Flow cytometry and high-throughput Treg-cell receptor sequencing were used to investigate the amount of different Treg-cell subsets and the diversity of TCRs between RA patients and healthy subjects, as well as between KD RA and non-KD RA patients. RT-qPCR was used to validate the high-throughput sequencing results. Results The data showed that the amount of naïve Treg cells in KD patients was less than in non-KD RA patients (P = 0.004) with no significant differences observed between other subsets. In the TCR of Treg cells, the length of complementarity determining region 3 (CDR3) was low and clonotypes increased in the KD group compared with the non-KD group. The diversity and abundance of Treg TCRs were low, as determined by the Hill number. In addition, several V(D)J combinations, such as T-cell receptor beta variable 7-2 (TRBV7-2), TRBV11-1, TRBV13, TRBV15, and TRBJ2-3, varied significantly between the two groups, indicating that KD causes Treg dysfunction. RT-qPCR shows that FOXP3 expression in peripheral blood Treg is lower in KD than in non-KD. Conclusion The results demonstrate the close correlation between KD and immune repertoires in RA and provide a new evaluation method for RA in TCM.
Collapse
Affiliation(s)
- Lu Zhang
- First Clinical Medical School, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Wei Jiao
- First Clinical Medical School, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Hui Deng
- First Clinical Medical School, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Congqi Hu
- Department of Rheumatology, First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Jia Xu
- First Clinical Medical School, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Jiahui Yu
- First Clinical Medical School, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Lijuan Liu
- Department of Rheumatology, First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Mingying Zhang
- Department of Rheumatology, First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Jiduo Liu
- Department of Rheumatology, First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Guangxing Chen
- Department of Rheumatology, First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China,Baiyun Hospital of the First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| |
Collapse
|
24
|
Role of T Cells in the Pathogenesis of Rheumatoid Arthritis: Focus on Immunometabolism Dysfunctions. Inflammation 2023; 46:88-102. [PMID: 36215002 DOI: 10.1007/s10753-022-01751-9] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2022] [Revised: 09/20/2022] [Accepted: 09/30/2022] [Indexed: 11/05/2022]
Abstract
Evidence demonstrated that metabolic-associated T cell abnormalities could be detected in the early stage of RA development. In this context, molecular evaluations have revealed changes in metabolic pathways, leading to the aggressive phenotype of RA T cells. A growing list of genes is downregulated or upregulated in RA T cells, and most of these genes with abnormal expression fall into the category of metabolic pathways. It has been shown that RA T cells shunt glucose towards the pentose phosphate pathway (PPP), which is associated with a high level of nicotinamide adenine dinucleotide phosphate (NADPH) and intermediate molecules. An increased level of NADPH inhibits ATM activation and thereby increases the proliferation capabilities of the RA T cells. Defects in the DNA repair nuclease MRE11A cause failures in repairing mitochondrial DNA, resulting in inhibiting the fatty acid oxidation pathway and further elevated cytoplasmic lipid droplets. Accumulated lipid droplets employ to generate lipid membranes for the cell building program and are also used to form the front-end membrane ruffles that are accomplices with invasive phenotypes of RA T cells. Metabolic pathway involvement in RA pathogenesis expands the pathogenic concept of the disease beyond the common view of autoimmunity triggered by autoantigen recognition. Increased knowledge about metabolic pathways' implications in RA pathogenesis paves the way to understand better the environment/gene interactions and host/microbiota interactions and introduce potential therapeutic approaches. This review summarized emerging data about the roles of T cells in RA pathogenesis with a focus on immunometabolism dysfunctions and how these metabolic alterations can affect the disease process.
Collapse
|
25
|
Lin W, Wang M, Xu L, Tortorella M, Li G. Cartilage organoids for cartilage development and cartilage-associated disease modeling. Front Cell Dev Biol 2023; 11:1125405. [PMID: 36824369 PMCID: PMC9941961 DOI: 10.3389/fcell.2023.1125405] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2022] [Accepted: 01/09/2023] [Indexed: 01/31/2023] Open
Abstract
Cartilage organoids have emerged as powerful modelling technology for recapitulation of joint embryonic events, and cartilage regeneration, as well as pathophysiology of cartilage-associated diseases. Recent breakthroughs have uncovered "mini-joint" models comprising of multicellular components and extracellular matrices of joint cartilage for development of novel disease-modifying strategies for personalized therapeutics of cartilage-associated diseases. Here, we hypothesized that LGR5-expressing embryonic joint chondroprogenitor cells are ideal stem cells for the generation of cartilage organoids as "mini-joints" ex vivo "in a dish" for embryonic joint development, cartilage repair, and cartilage-associated disease modelling as essential research models of drug screening for further personalized regenerative therapy. The pilot research data suggested that LGR5-GFP-expressing embryonic joint progenitor cells are promising for generation of cartilage organoids through gel embedding method, which may exert various preclinical and clinical applications for realization of personalized regenerative therapy in the future.
Collapse
Affiliation(s)
- Weiping Lin
- Centre for Regenerative Medicine and Health, Hong Kong Institute of Science & Innovation, Chinese Academy of Sciences, Hong Kong, Hong Kong SAR, China,The Fifth Affiliated Hospital of Guangzhou Medical University, Guangzhou, China,*Correspondence: Weiping Lin, ; Liangliang Xu, ; Micky Tortorella, ; Gang Li,
| | - Min Wang
- Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, China
| | - Liangliang Xu
- The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou, China,*Correspondence: Weiping Lin, ; Liangliang Xu, ; Micky Tortorella, ; Gang Li,
| | - Micky Tortorella
- Centre for Regenerative Medicine and Health, Hong Kong Institute of Science & Innovation, Chinese Academy of Sciences, Hong Kong, Hong Kong SAR, China,Drug Discovery Pipeline at the Guangzhou Institutes for Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China,*Correspondence: Weiping Lin, ; Liangliang Xu, ; Micky Tortorella, ; Gang Li,
| | - Gang Li
- Musculoskeletal Research Laboratory, Department of Orthopaedics & Traumatology, Faculty of Medicine, The Chinese University of Hong Kong, Prince of Wales Hospital, Hong Kong SAR, China,Stem Cells and Regenerative Medicine Laboratory, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Prince of Wales Hospital, Hong Kong SAR, China,Shenzhen Research Institute, The Chinese University of Hong Kong, Shenzhen, China,*Correspondence: Weiping Lin, ; Liangliang Xu, ; Micky Tortorella, ; Gang Li,
| |
Collapse
|
26
|
Chintapula U, Chikate T, Sahoo D, Kieu A, Guerrero Rodriguez ID, Nguyen KT, Trott D. Immunomodulation in age-related disorders and nanotechnology interventions. WILEY INTERDISCIPLINARY REVIEWS. NANOMEDICINE AND NANOBIOTECHNOLOGY 2023; 15:e1840. [PMID: 35950266 PMCID: PMC9840662 DOI: 10.1002/wnan.1840] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/24/2021] [Revised: 05/19/2022] [Accepted: 06/01/2022] [Indexed: 01/31/2023]
Abstract
Recently, the aging population has increased exponentially around the globe bringing more challenges to improve quality of life in those populations while reducing the economic burden on healthcare systems. Aging is associated with changes in the immune system culminating in detrimental effects such as immune dysfunction, immunosenescence, and chronic inflammation. Age-related decline of immune functions is associated with various pathologies including cardiovascular, autoimmune, neurodegenerative, and infectious diseases to name a few. Conventional treatment addresses the onset of age-related diseases by early detection of risk factors, administration of vaccines as preventive care, immunomodulatory treatment, and other dietary supplements. However, these approaches often come with systemic side-effects, low bioavailability of therapeutic agents, and poor outcomes seen in the elderly. Recent innovations in nanotechnology have led to the development of novel biomaterials/nanomaterials, which explore targeted drug delivery and immunomodulatory interactions in vivo. Current nanotechnology-based immunomodulatory approaches that have the potential to be used as therapeutic interventions for some prominent age-related diseases are discussed here. Finally, we explore challenges and future aspects of nanotechnology in the treatments of age-related disorders to improve quality of life in the elderly. This article is categorized under: Therapeutic Approaches and Drug Discovery > Nanomedicine for Cardiovascular Disease Therapeutic Approaches and Drug Discovery > Nanomedicine for Neurological Disease Therapeutic Approaches and Drug Discovery > Emerging Technologies.
Collapse
Affiliation(s)
- Uday Chintapula
- Department of Bioengineering, University of Texas at Arlington, Arlington, Texas, USA
- Joint Bioengineering Program, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Tanmayee Chikate
- Department of Bioengineering, University of Texas at Arlington, Arlington, Texas, USA
| | - Deepsundar Sahoo
- Department of Bioengineering, University of Texas at Arlington, Arlington, Texas, USA
| | - Amie Kieu
- Department of Bioengineering, University of Texas at Arlington, Arlington, Texas, USA
| | | | - Kytai T. Nguyen
- Department of Bioengineering, University of Texas at Arlington, Arlington, Texas, USA
- Joint Bioengineering Program, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Daniel Trott
- Department of Kinesiology, University of Texas at Arlington, Arlington, Texas, USA
| |
Collapse
|
27
|
Ravaei A, Zimmer-Bensch G, Govoni M, Rubini M. lncRNA-mediated synovitis in rheumatoid arthritis: A perspective for biomarker development. PROGRESS IN BIOPHYSICS AND MOLECULAR BIOLOGY 2022; 175:103-119. [PMID: 36126801 DOI: 10.1016/j.pbiomolbio.2022.09.007] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/17/2022] [Revised: 07/28/2022] [Accepted: 09/13/2022] [Indexed: 06/15/2023]
Abstract
Long noncoding RNAs (lncRNAs) are a regulatory class of noncoding RNAs with a wide range of activities such as transcriptional and post-transcriptional regulations. Emerging evidence has demonstrated that various lncRNAs contribute to the initiation and progression of Rheumatoid Arthritis (RA) through distinctive mechanisms. The present study reviews the recent findings on lncRNA role in RA development. It focuses on the involvement of different lncRNAs in the main steps of RA pathogenesis including T cell activation, cytokine dysregulation, fibroblast-like synoviocyte (FLS) activation and joint destruction. Besides, it discusses the current findings on RA diagnosis and the potential of lncRNAs as diagnostic, prognostic and predictive biomarkers in Rheumatology clinic.
Collapse
Affiliation(s)
- Amin Ravaei
- Department of Neurosciences and Rehabilitation, Section of Medical Biochemistry, Molecular Biology and Genetics, University of Ferrara, Ferrara, Italy.
| | - Geraldine Zimmer-Bensch
- Division of Neuroepigenetics, Institute of Zoology (Biology II), RWTH Aachen University, Aachen, Germany.
| | - Marcello Govoni
- Department of Medical Science, Section of Rheumatology, University of Ferrara, Ferrara, Italy.
| | - Michele Rubini
- Department of Neurosciences and Rehabilitation, Section of Medical Biochemistry, Molecular Biology and Genetics, University of Ferrara, Ferrara, Italy.
| |
Collapse
|
28
|
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
|
29
|
Natalini JG, England BR, Baker JF, Chen Q, Singh N, Mahajan TD, Roul P, Thiele GM, Sauer BC, Mikuls TR, Johnson FB, Kawut SM. Associations between shortened telomeres and rheumatoid arthritis-associated interstitial lung disease among U.S. Veterans. Respir Med 2022; 201:106943. [PMID: 35947933 PMCID: PMC10120870 DOI: 10.1016/j.rmed.2022.106943] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/13/2022] [Revised: 07/20/2022] [Accepted: 07/22/2022] [Indexed: 11/29/2022]
Abstract
BACKGROUND Shortened telomeres are associated with several different subtypes of interstitial lung disease (ILD), although studies of telomere length and ILD in rheumatoid arthritis (RA) are lacking. METHODS Within the Veterans Affairs Rheumatoid Arthritis (VARA) registry, we performed cross-sectional and case-control studies of prevalent and incident ILD, respectively. We randomly selected a subset of RA patients with ILD and individually matched them to RA patients without ILD according to age, sex, and VARA enrollment date. Telomere length was measured on peripheral blood leukocytes collected at registry enrollment using quantitative PCR (T/S ratio). Short telomeres were defined as a T/S ratio in the lowest 10th percentile of the cohort. RESULTS Our cross-sectional study cohort was comprised of 54 RA-ILD patients and 92 RA-non-ILD patients. T/S ratios significantly differed between patients with and without prevalent ILD (1.56 [IQR 1.30, 1.78] vs. 1.96 [IQR 1.65, 2.27], p < 0.001). Similarly, prevalence of ILD was significantly higher in patients with short vs. normal-length telomeres (73.3% vs. 32.8%, p = 0.002). Short telomeres were independently associated with an increased odds of prevalent ILD compared to normal-length telomeres (adjusted OR 6.60, 95% CI 1.78-24.51, p = 0.005). In our case-control analysis, comprised of 22 incident RA-ILD cases and 36 RA-non-ILD controls, short telomeres were not associated with incident RA-ILD (adjusted OR 0.90, 95% CI 0.06-13.4, p = 0.94). CONCLUSION Short telomeres were strongly associated with prevalent but not incident ILD among patients with RA. Additional studies are needed to better understand telomere length dynamics among RA patients with and without ILD.
Collapse
Affiliation(s)
- Jake G Natalini
- Division of Pulmonary, Critical Care, and Sleep Medicine, Department of Medicine, New York University Grossman School of Medicine, New York, NY, United States.
| | - Bryant R England
- Division of Rheumatology and Immunology, Department of Internal Medicine, University of Nebraska Medical Center, Omaha, NE, United States; VA Nebraska-Western Iowa Health Care System, Omaha, NE, United States
| | - Joshua F Baker
- Division of Rheumatology, Department of Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, United States; Corporal Michael J. Crescenz VA Medical Center, Philadelphia, PA, United States
| | - Qijun Chen
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, United States
| | - Namrata Singh
- Division of Rheumatology, Department of Medicine, University of Washington School of Medicine, Seattle, WA, United States
| | - Tina D Mahajan
- Division of Rheumatology and Immunology, Department of Internal Medicine, University of Nebraska Medical Center, Omaha, NE, United States
| | - Punyasha Roul
- Division of Rheumatology and Immunology, Department of Internal Medicine, University of Nebraska Medical Center, Omaha, NE, United States
| | - Geoffrey M Thiele
- Division of Rheumatology and Immunology, Department of Internal Medicine, University of Nebraska Medical Center, Omaha, NE, United States
| | - Brian C Sauer
- Division of Epidemiology, University of Utah School of Medicine, Salt Lake City, UT, United States
| | - Ted R Mikuls
- Division of Rheumatology and Immunology, Department of Internal Medicine, University of Nebraska Medical Center, Omaha, NE, United States; VA Nebraska-Western Iowa Health Care System, Omaha, NE, United States
| | - F Bradley Johnson
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, United States
| | - Steven M Kawut
- Division of Pulmonary, Allergy, and Critical Care, Department of Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, United States
| |
Collapse
|
30
|
Callender LA, Carroll EC, Garrod-Ketchley C, Schroth J, Bystrom J, Berryman V, Pattrick M, Campbell-Richards D, Hood GA, Hitman GA, Finer S, Henson SM. Altered Nutrient Uptake Causes Mitochondrial Dysfunction in Senescent CD8 + EMRA T Cells During Type 2 Diabetes. FRONTIERS IN AGING 2022; 2:681428. [PMID: 35821991 PMCID: PMC9261431 DOI: 10.3389/fragi.2021.681428] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/16/2021] [Accepted: 07/22/2021] [Indexed: 01/04/2023]
Abstract
Mitochondrial health and cellular metabolism can heavily influence the onset of senescence in T cells. CD8+ EMRA T cells exhibit mitochondrial dysfunction and alterations to oxidative phosphorylation, however, the metabolic properties of senescent CD8+ T cells from people living with type 2 diabetes (T2D) are not known. We show here that mitochondria from T2D CD8+ T cells had a higher oxidative capacity together with increased levels of mitochondrial reactive oxgen species (mtROS), compared to age-matched control cells. While fatty acid uptake was increased, fatty acid oxidation was impaired in T2D CD8+ EMRA T cells, which also showed an accumulation of lipid droplets and decreased AMPK activity. Increasing glucose and fatty acids in healthy CD8+ T cells resulted in increased p-p53 expression and a fragmented mitochondrial morphology, similar to that observed in T2D CD8+ EMRA T cells. The resulting mitochondrial changes are likely to have a profound effect on T cell function. Consequently, a better understanding of these metabolic abnormalities is crucial as metabolic manipulation of these cells may restore correct T cell function and help reduce the impact of T cell dysfunction in T2D.
Collapse
Affiliation(s)
- Lauren A Callender
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom
| | - Elizabeth C Carroll
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom
| | - Conor Garrod-Ketchley
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom
| | - Johannes Schroth
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom
| | - Jonas Bystrom
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom
| | | | | | | | - Gillian A Hood
- Institute of Population Health Sciences, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom
| | - Graham A Hitman
- Blizard Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom
| | - Sarah Finer
- Barts Health NHS Trust, London, United Kingdom.,Institute of Population Health Sciences, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom
| | - Sian M Henson
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom
| |
Collapse
|
31
|
Greisen SR, Aspari M, Deleuran B. Co-Inhibitory Molecules – Their Role in Health and Autoimmunity; Highlighted by Immune Related Adverse Events. Front Immunol 2022; 13:883733. [PMID: 35784333 PMCID: PMC9243421 DOI: 10.3389/fimmu.2022.883733] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2022] [Accepted: 05/10/2022] [Indexed: 12/18/2022] Open
Abstract
Immune checkpoint receptors are key players in regulating the immune response. They are responsible for both generating an immune response sufficient to kill invading pathogens, balancing the same response, and protecting against tissue destruction or the development of autoimmune events. The central role of the co-inhibitory receptors also referred to as inhibitory immune checkpoints, including PD-1 and CTLA-4 has become especially evident with the cancer treatments targeting these receptors. Blocking these pathways enhances the immune activity, resulting in both an increased chance of cancer clearance, at the same time induction of immune-related adverse events (irAE). Some of these irAE progress into actual autoimmune diseases with autoantibodies and symptoms, undistinguished from the naturally occurring diseases. This review will take advantage of the lessons learned from immune checkpoint blockade and relate this knowledge to our understanding of the same pathways in naturally occurring autoimmune diseases, mainly focusing on rheumatic diseases.
Collapse
Affiliation(s)
- Stinne R. Greisen
- Department of Biomedicine, Aarhus University, Aarhus, Denmark
- Department of Rheumatology, Aarhus University Hospital, Aarhus, Denmark
- *Correspondence: Stinne R. Greisen,
| | - Maithri Aspari
- Department of Biomedicine, Aarhus University, Aarhus, Denmark
| | - Bent Deleuran
- Department of Biomedicine, Aarhus University, Aarhus, Denmark
- Department of Rheumatology, Aarhus University Hospital, Aarhus, Denmark
| |
Collapse
|
32
|
Deng R, Wang Y, Bu Y, Wu H. BNIP3 mediates the different adaptive responses of fibroblast-like synovial cells to hypoxia in patients with osteoarthritis and rheumatoid arthritis. Mol Med 2022; 28:64. [PMID: 35690741 PMCID: PMC9188199 DOI: 10.1186/s10020-022-00490-9] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2021] [Accepted: 05/25/2022] [Indexed: 01/01/2023] Open
Abstract
Background Hypoxia is one of the important characteristics of synovial microenvironment in rheumatoid arthritis (RA), and plays an important role in synovial hyperplasia. In terms of cell survival, fibroblast-like synovial cells (FLSs) are relatively affected by hypoxia. In contrast, fibroblast-like synovial cells from patients with RA (RA-FLSs) are particularly resistant to hypoxia-induced cell death. The purpose of this study was to evaluate whether fibroblast-like synovial cells in patients with osteoarthritis (OA-FLSs) and RA-FLSs have the same adaptation to hypoxia. Methods CCK-8, flow cytometry and BrdU were used to detect the proliferation of OA-FLSs and RA-FLSs under different oxygen concentrations. Apoptosis was detected by AV/PI, TUNEL and Western blot, mitophagy was observed by electron microscope, laser confocal microscope and Western blot, the state of mitochondria was detected by ROS and mitochondrial membrane potential by flow cytometry, BNIP3 and HIF-1α were detected by Western blot and RT-qPCR. The silencing of BNIP3 was achieved by stealth RNA system technology. Results After hypoxia, the survival rate of OA-FLSs decreased, while the proliferation activity of RA-FLSs further increased. Hypoxia induced an increase in apoptosis and inhibition of mitophagy in OA-FLSs, but not in RA-FLSs. Hypoxia led to a more lasting adaptive response. RA-FLSs displayed a more significant increase in the expression of genes transcriptionally regulated by HIF-1α. Interestingly, they showed higher BNIP3 expression than OA-FLSs, and showed stronger mitophagy and proliferation activities. BNIP3 siRNA experiment confirmed the potential role of BNIP3 in the survival of RA-FLSs. Inhibition of BNIP3 resulted in the decrease of cell proliferation, mitophagy and the increase of apoptosis. Conclusion In summary, RA-FLSs maintained intracellular redox balance through mitophagy to promote cell survival under hypoxia. The mitophagy of OA-FLSs was too little to maintain the redox balance of mitochondria, resulting in apoptosis. The difference of mitophagy between OA-FLSs and RA-FLSs under hypoxia is mediated by the level of BNIP3 expression.
Collapse
Affiliation(s)
- Ran Deng
- Key Laboratory of Xin'an Medicine, Ministry of Education, Hefei, 230012, China.,Anhui University of Chinese Medicine, Qian Jiang Road 1, Hefei, 230012, China.,Anhui Province Key Laboratory of Research &, Development of Chinese Medicine, Hefei, 230012, China
| | - Yan Wang
- Key Laboratory of Xin'an Medicine, Ministry of Education, Hefei, 230012, China.,Anhui University of Chinese Medicine, Qian Jiang Road 1, Hefei, 230012, China.,Anhui Province Key Laboratory of Research &, Development of Chinese Medicine, Hefei, 230012, China
| | - Yanhong Bu
- Key Laboratory of Xin'an Medicine, Ministry of Education, Hefei, 230012, China.,Anhui University of Chinese Medicine, Qian Jiang Road 1, Hefei, 230012, China.,Anhui Province Key Laboratory of Research &, Development of Chinese Medicine, Hefei, 230012, China
| | - Hong Wu
- Key Laboratory of Xin'an Medicine, Ministry of Education, Hefei, 230012, China. .,Anhui University of Chinese Medicine, Qian Jiang Road 1, Hefei, 230012, China. .,Anhui Province Key Laboratory of Research &, Development of Chinese Medicine, Hefei, 230012, China. .,College of Pharmacy, Anhui University of Chinese Medicine, Hefei, 230012, China.
| |
Collapse
|
33
|
Fearon U, Hanlon MM, Floudas A, Veale DJ. Cellular metabolic adaptations in rheumatoid arthritis and their therapeutic implications. Nat Rev Rheumatol 2022; 18:398-414. [PMID: 35440762 DOI: 10.1038/s41584-022-00771-x] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/04/2022] [Indexed: 12/16/2022]
Abstract
Activation of endothelium and immune cells is fundamental to the initiation of autoimmune diseases such as rheumatoid arthritis (RA), and it results in trans-endothelial cell migration and synovial fibroblast proliferation, leading to joint destruction. In RA, the synovial microvasculature is highly dysregulated, resulting in inefficient oxygen perfusion to the synovium, which, along with the high metabolic demands of activated immune and stromal cells, leads to a profoundly hypoxic microenvironment. In inflamed joints, infiltrating immune cells and synovial resident cells have great requirements for energy and nutrients, and they adapt their metabolic profiles to generate sufficient energy to support their highly activated inflammatory states. This shift in metabolic capacity of synovial cells enables them to produce the essential building blocks to support their proliferation, activation and invasiveness. Furthermore, it results in the accumulation of metabolic intermediates and alteration of redox-sensitive pathways, affecting signalling pathways that further potentiate the inflammatory response. Importantly, the inflamed synovium is a multicellular tissue, with cells differing in their metabolic requirements depending on complex cell-cell interactions, nutrient supply, metabolic intermediates and transcriptional regulation. Therefore, understanding the complex interplay between metabolic and inflammatory pathways in synovial cells in RA will provide insight into the underlying mechanisms of disease pathogenesis.
Collapse
Affiliation(s)
- Ursula Fearon
- Molecular Rheumatology, Trinity Biomedical Sciences Institute, TCD, Dublin, Ireland. .,EULAR Centre of Excellence, Centre for Arthritis and Rheumatic Diseases, St Vincent's University Hospital, Dublin, Ireland.
| | - Megan M Hanlon
- Molecular Rheumatology, Trinity Biomedical Sciences Institute, TCD, Dublin, Ireland.,EULAR Centre of Excellence, Centre for Arthritis and Rheumatic Diseases, St Vincent's University Hospital, Dublin, Ireland
| | - Achilleas Floudas
- Molecular Rheumatology, Trinity Biomedical Sciences Institute, TCD, Dublin, Ireland.,EULAR Centre of Excellence, Centre for Arthritis and Rheumatic Diseases, St Vincent's University Hospital, Dublin, Ireland
| | - Douglas J Veale
- EULAR Centre of Excellence, Centre for Arthritis and Rheumatic Diseases, St Vincent's University Hospital, Dublin, Ireland
| |
Collapse
|
34
|
Taucher E, Mykoliuk I, Lindenmann J, Smolle-Juettner FM. Implications of the Immune Landscape in COPD and Lung Cancer: Smoking Versus Other Causes. Front Immunol 2022; 13:846605. [PMID: 35386685 PMCID: PMC8978964 DOI: 10.3389/fimmu.2022.846605] [Citation(s) in RCA: 35] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2021] [Accepted: 02/28/2022] [Indexed: 12/30/2022] Open
Abstract
Cigarette smoking is reported in about one third of adults worldwide. A strong relationship between cigarette smoke exposure and chronic obstructive pulmonary disease (COPD) as well as lung cancer has been proven. However, about 15% of lung cancer cases, and between one fourth and one third of COPD cases, occur in never-smokers. The effects of cigarette smoke on the innate as well as the adaptive immune system have been widely investigated. It is assumed that certain immunologic features contribute to lung cancer and COPD development in the absence of smoking as the major risk factor. In this article, we review different immunological aspects of lung cancer and COPD with a special focus on non-smoking related risk factors.
Collapse
Affiliation(s)
- Elisabeth Taucher
- Division of Pulmonology, Department of Internal Medicine, Medical University Graz, Graz, Austria
| | - Iurii Mykoliuk
- Division of Thoracic Surgery, Department of Surgery, Medical University Graz, Graz, Austria
| | - Joerg Lindenmann
- Division of Thoracic Surgery, Department of Surgery, Medical University Graz, Graz, Austria
| | | |
Collapse
|
35
|
Amoriello R, Mariottini A, Ballerini C. Immunosenescence and Autoimmunity: Exploiting the T-Cell Receptor Repertoire to Investigate the Impact of Aging on Multiple Sclerosis. Front Immunol 2021; 12:799380. [PMID: 34925384 PMCID: PMC8673061 DOI: 10.3389/fimmu.2021.799380] [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: 10/21/2021] [Accepted: 11/16/2021] [Indexed: 01/08/2023] Open
Abstract
T-cell receptor (TCR) repertoire diversity is a determining factor for the immune system capability in fighting infections and preventing autoimmunity. During life, the TCR repertoire diversity progressively declines as a physiological aging progress. The investigation of TCR repertoire dynamics over life represents a powerful tool unraveling the impact of immunosenescence in health and disease. Multiple Sclerosis (MS) is a demyelinating, inflammatory, T-cell mediated autoimmune disease of the Central Nervous System in which age is crucial: it is the most widespread neurological disease among young adults and, furthermore, patients age may impact on MS progression and treatments outcome. Crossing knowledge on the TCR repertoire dynamics over MS patients' life is fundamental to investigate disease mechanisms, and the advent of high- throughput sequencing (HTS) has significantly increased our knowledge on the topic. Here we report an overview of current literature about the impact of immunosenescence and age-related TCR dynamics variation in autoimmunity, including MS.
Collapse
Affiliation(s)
- Roberta Amoriello
- Dipartimento di Medicina Sperimentale e Clinica (DMSC), Laboratory of Neuroimmunology, University of Florence, Florence, Italy
| | - Alice Mariottini
- Dipartimento di Neuroscienze, Psicologia, Area del Farmaco e Salute del Bambino (NEUROFARBA), University of Florence, Florence, Italy
| | - Clara Ballerini
- Dipartimento di Medicina Sperimentale e Clinica (DMSC), Laboratory of Neuroimmunology, University of Florence, Florence, Italy
| |
Collapse
|
36
|
Salminen A. Immunosuppressive network promotes immunosenescence associated with aging and chronic inflammatory conditions. J Mol Med (Berl) 2021; 99:1553-1569. [PMID: 34432073 PMCID: PMC8384586 DOI: 10.1007/s00109-021-02123-w] [Citation(s) in RCA: 64] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2021] [Revised: 07/21/2021] [Accepted: 07/30/2021] [Indexed: 01/10/2023]
Abstract
The functional competence of the immune system gradually declines with aging, a process called immunosenescence. The age-related remodelling of the immune system affects both adaptive and innate immunity. In particular, a chronic low-grade inflammation, termed inflammaging, is associated with the aging process. Immunosenescence not only is present in inflammaging state, but it also occurs in several pathological conditions in conjunction with chronic inflammation. It is known that persistent inflammation stimulates a counteracting compensatory immunosuppression intended to protect host tissues. Inflammatory mediators enhance myelopoiesis and induce the generation of immature myeloid-derived suppressor cells (MDSC) which in mutual cooperation stimulates the immunosuppressive network. Immunosuppressive cells, especially MDSCs, regulatory T cells (Treg), and M2 macrophages produce immunosuppressive factors, e.g., TGF-β, IL-10, ROS, arginase-1 (ARG1), and indoleamine 2,3-dioxygenase (IDO), which suppress the functions of CD4/CD8T and B cells as well as macrophages, natural killer (NK) cells, and dendritic cells. The immunosuppressive armament (i) inhibits the development and proliferation of immune cells, (ii) decreases the cytotoxic activity of CD8T and NK cells, (iii) prevents antigen presentation and antibody production, and (iv) suppresses responsiveness to inflammatory mediators. These phenotypes are the hallmarks of immunosenescence. Immunosuppressive factors are able to control the chromatin landscape, and thus, it seems that the immunosenescence state is epigenetically regulated.
Collapse
Affiliation(s)
- Antero Salminen
- Department of Neurology, Institute of Clinical Medicine, University of Eastern Finland, P.O. Box 1627, 70211, Kuopio, Finland.
| |
Collapse
|
37
|
Andreas N, Müller S, Templin N, Jordan PM, Schuhwerk H, Müller M, Gerstmeier J, Miek L, Andreas S, Werz O, Kamradt T. Incidence and severity of G6PI-induced arthritis are not increased in genetically distinct mouse strains upon aging. Arthritis Res Ther 2021; 23:222. [PMID: 34429153 PMCID: PMC8383389 DOI: 10.1186/s13075-021-02596-7] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2021] [Accepted: 08/02/2021] [Indexed: 12/02/2022] Open
Abstract
Background The incidence of rheumatoid arthritis is correlated with age. In this study, we analyzed the association of the incidence and severity of glucose-6-phosphate isomerase (G6PI)-induced arthritis with age in two different mouse strains. Methods Young and very old mice from two different arthritis-susceptible wild-type mouse strains were analyzed after a single subcutaneous injection of G6PI s.c. The metabolism and the function of synoviocytes were analyzed in vitro, the production of bioactive lipid mediators by myeloid cells and synoviocytes was assessed in vitro and ex vivo by UPLC-MS-MS, and flow cytometry was used to verify age-related changes of immune cell composition and function. Results While the severity of arthritis was independent from age, the onset was delayed in old mice. Old mice showed common signs of immune aging like thymic atrophy associated with decreased CD4+ effector T cell numbers. Despite its decrease, the effector T helper (Th) cell compartment in old mice was reactive and functionally intact, and their Tregs exhibited unaltered suppressive capacities. In homeostasis, macrophages and synoviocytes from old mice produced higher amounts of pro-inflammatory cyclooxygenase (COX)-derived products. However, this functional difference did not remain upon challenge in vitro nor upon arthritis reactions ex vivo. Conclusion While old mice show a higher baseline of inflammatory functions, this does not result in increased reaction towards self-antigens in arthritis-susceptible mouse strains. Together, our data from two different mouse strains show that the susceptibility for G6PI-induced arthritis is not age-dependent. Supplementary Information The online version contains supplementary material available at 10.1186/s13075-021-02596-7.
Collapse
Affiliation(s)
- Nico Andreas
- Institute of Immunology, University Hospital Jena, Leutragraben 3, 07743, Jena, Germany.
| | - Sylvia Müller
- Institute of Immunology, University Hospital Jena, Leutragraben 3, 07743, Jena, Germany
| | - Nicole Templin
- Institute of Immunology, University Hospital Jena, Leutragraben 3, 07743, Jena, Germany
| | - Paul M Jordan
- Department of Pharmaceutical/Medicinal Chemistry, Friedrich Schiller University, Philosophenweg 14, 07743, Jena, Germany
| | - Harald Schuhwerk
- Institute of Immunology, University Hospital Jena, Leutragraben 3, 07743, Jena, Germany
| | - Michael Müller
- Institute of Immunology, University Hospital Jena, Leutragraben 3, 07743, Jena, Germany
| | - Jana Gerstmeier
- Department of Pharmaceutical/Medicinal Chemistry, Friedrich Schiller University, Philosophenweg 14, 07743, Jena, Germany
| | - Laura Miek
- Department of Pharmaceutical/Medicinal Chemistry, Friedrich Schiller University, Philosophenweg 14, 07743, Jena, Germany
| | - Saskia Andreas
- Department of Pharmaceutical/Medicinal Chemistry, Friedrich Schiller University, Philosophenweg 14, 07743, Jena, Germany
| | - Oliver Werz
- Department of Pharmaceutical/Medicinal Chemistry, Friedrich Schiller University, Philosophenweg 14, 07743, Jena, Germany
| | - Thomas Kamradt
- Institute of Immunology, University Hospital Jena, Leutragraben 3, 07743, Jena, Germany.
| |
Collapse
|
38
|
González-Osuna L, Sierra-Cristancho A, Rojas C, Cafferata EA, Melgar-Rodríguez S, Cárdenas AM, Vernal R. Premature Senescence of T-cells Favors Bone Loss During Osteolytic Diseases. A New Concern in the Osteoimmunology Arena. Aging Dis 2021; 12:1150-1161. [PMID: 34341698 PMCID: PMC8279535 DOI: 10.14336/ad.2021.0110] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2020] [Accepted: 01/10/2021] [Indexed: 12/15/2022] Open
Abstract
Cellular senescence is a biological process triggered in response to time-accumulated DNA damage, which prioritizes cell survival over cell function. Particularly, senescent T lymphocytes can be generated prematurely during chronic inflammatory diseases regardless of chronological aging. These senescent T lymphocytes are characterized by the loss of CD28 expression, a co-stimulatory receptor that mediates antigen presentation and effective T-cell activation. An increased number of premature senescent CD4+CD28- T lymphocytes has been frequently observed in osteolytic diseases, including rheumatoid arthritis, juvenile idiopathic arthritis, ankylosing spondylitis, osteopenia, osteoporosis, and osteomyelitis. Indeed, CD4+CD28- T lymphocytes produce higher levels of osteoclastogenic molecular mediators directly related to pathologic bone loss, such as tumor necrosis factor (TNF)-α, interleukin (IL)-17A, and receptor-activator of nuclear factor κB ligand (RANKL), as compared with regular CD4+CD28+ T lymphocytes. In addition, premature senescent CD8+CD28- T lymphocytes have been negatively associated with bone healing and regeneration by inhibiting osteoblast differentiation and mesenchymal stromal cell survival. Therefore, accumulated evidence supports the role of senescent T lymphocytes in osteoimmunology. Moreover, premature senescence of T-cells seems to be associated with the functional imbalance between the osteolytic T-helper type-17 (Th17) and bone protective T regulatory (Treg) lymphocytes, as well as the phenotypic instability of Treg lymphocytes responsible for its trans-differentiation into RANKL-producing exFoxp3Th17 cells, a key cellular phenomenon directly related to bone loss. Herein, we present a framework for the understanding of the pathogenic characteristics of T lymphocytes with a premature senescent phenotype; and particularly, we revise and discuss their role in the osteoimmunology of osteolytic diseases.
Collapse
Affiliation(s)
- Luis González-Osuna
- Periodontal Biology Laboratory, Faculty of Dentistry, Universidad de Chile, Santiago, Chile.
| | - Alfredo Sierra-Cristancho
- Periodontal Biology Laboratory, Faculty of Dentistry, Universidad de Chile, Santiago, Chile.
- Faculty of Dentistry, Universidad Andres Bello, Santiago, Chile.
| | - Carolina Rojas
- Periodontal Biology Laboratory, Faculty of Dentistry, Universidad de Chile, Santiago, Chile.
| | - Emilio A Cafferata
- Periodontal Biology Laboratory, Faculty of Dentistry, Universidad de Chile, Santiago, Chile.
- Department of Periodontology, School of Dentistry, Universidad Científica del Sur, Lima, Perú.
| | - Samanta Melgar-Rodríguez
- Periodontal Biology Laboratory, Faculty of Dentistry, Universidad de Chile, Santiago, Chile.
- Department of Conservative Dentistry, Faculty of Dentistry, Universidad de Chile, Santiago, Chile.
| | - Angélica M Cárdenas
- Periodontal Biology Laboratory, Faculty of Dentistry, Universidad de Chile, Santiago, Chile.
- Health Sciences Division, Faculty of Dentistry, Universidad Santo Tomás, Bucaramanga, Colombia.
| | - Rolando Vernal
- Periodontal Biology Laboratory, Faculty of Dentistry, Universidad de Chile, Santiago, Chile.
- Department of Conservative Dentistry, Faculty of Dentistry, Universidad de Chile, Santiago, Chile.
| |
Collapse
|
39
|
Sultan OM, Alghazali DM, Al-Tameemi H, Abed M, Hawiji DA, Abu Ghniem MN, Al-Obaidi L, Abedtwfeq RH. Pattern and Age Distribution of COVID-19 on Pulmonary Computed Tomography. Curr Med Imaging 2021; 17:775-780. [PMID: 33357198 DOI: 10.2174/1573405616666201223144539] [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: 06/05/2020] [Revised: 10/11/2020] [Accepted: 11/12/2020] [Indexed: 11/22/2022]
Abstract
BACKGROUND COVID-19 has emerged recently and has become a global concern. Computed tomography (CT) plays a vital role in the diagnosis. OBJECTIVES To characterize the pulmonary CT findings and distributions of COVID-19 infection in regard to different age groups. METHODS Chest CT scan of 104 symptomatic patients with COVID-19 infection from 7 Iraqi isolation centers were retrospectively analyzed between March 10th to April 5th, 2020. Patients were sub-classified according to their ages into three groups (young adult:20-39 years, middle age:40-59 years, and old age:60-90 years). RESULTS The most common findings were ground-glass opacities (GGO) (92.3%, followed by consolidation (27.9%), bronchovascular thickening (15.4%), and crazy-paving (12.5%). Less commonly, there were tree-in-bud (6.7%), pulmonary nodules (5.8%), bronchiectasis (3.8%), pleural effusion (1.9%), and cavitation (1%). There were no hallo signs, reversed hallo signs, and mediastinal lymphadenopathy. Pulmonary changes were unilateral in 16.7% and bilateral in 83.3%, central in 14.6%, peripheral in 57.3%, and diffuse (central and peripheral) in 28.1%. Most cases showed multi- lobar changes (70.8%), while the lower lobe was more commonly involved (17.7%) than the middle lobe/lingula (8.3%) and upper lobe (3.1%). In unilateral involvement, changes were more on the right (68.8%) than the left (31.2%) side. Compared with middle and old age groups, young adult patients showed significantly lesser frequency of consolidation (17% vs. 13.3% and 37%), diffuse changes 28.1% (14.2% vs. 35.3% and 40.5%), bilateral disease (71.4% vs. 94.1% and 85.2%), and multi-lobar involvement (51.4% vs. 82.4% and 81.4%) respectively. CONCLUSION Bilateral and peripheral GGO were the most frequent findings with the right and lower lobar predilection. The pattern and the distribution of CT changes seem to be age-specific.
Collapse
Affiliation(s)
- Omar Muayad Sultan
- College of Medicine, Lecturer of radiodiagnosis, Director of Medical Education Unit (MEU), Tikrit University, Tikrit, Iraq
| | - Dhia Mahdey Alghazali
- Department of Degnostic Imaging, Al-Imam Al-Hussein Medical City. Karbala 56001, Iraq
| | | | - Mohammed Abed
- Department of Radiology, Al-Yarmuk Teaching Hospital, Baghdad 10001, Iraq
| | | | | | | | | |
Collapse
|
40
|
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
|
41
|
Qiu J, Wu B, Goodman SB, Berry GJ, Goronzy JJ, Weyand CM. Metabolic Control of Autoimmunity and Tissue Inflammation in Rheumatoid Arthritis. Front Immunol 2021; 12:652771. [PMID: 33868292 PMCID: PMC8050350 DOI: 10.3389/fimmu.2021.652771] [Citation(s) in RCA: 68] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2021] [Accepted: 03/15/2021] [Indexed: 12/19/2022] Open
Abstract
Like other autoimmune diseases, rheumatoid arthritis (RA) develops in distinct stages, with each phase of disease linked to immune cell dysfunction. HLA class II genes confer the strongest genetic risk to develop RA. They encode for molecules essential in the activation and differentiation of T cells, placing T cells upstream in the immunopathology. In Phase 1 of the RA disease process, T cells lose a fundamental function, their ability to be self-tolerant, and provide help for autoantibody-producing B cells. Phase 2 begins many years later, when mis-differentiated T cells gain tissue-invasive effector functions, enter the joint, promote non-resolving inflammation, and give rise to clinically relevant arthritis. In Phase 3 of the RA disease process, abnormal innate immune functions are added to adaptive autoimmunity, converting synovial inflammation into a tissue-destructive process that erodes cartilage and bone. Emerging data have implicated metabolic mis-regulation as a fundamental pathogenic pathway in all phases of RA. Early in their life cycle, RA T cells fail to repair mitochondrial DNA, resulting in a malfunctioning metabolic machinery. Mitochondrial insufficiency is aggravated by the mis-trafficking of the energy sensor AMPK away from the lysosomal surface. The metabolic signature of RA T cells is characterized by the shunting of glucose toward the pentose phosphate pathway and toward biosynthetic activity. During the intermediate and terminal phase of RA-imposed tissue inflammation, tissue-residing macrophages, T cells, B cells and stromal cells are chronically activated and under high metabolic stress, creating a microenvironment poor in oxygen and glucose, but rich in metabolic intermediates, such as lactate. By sensing tissue lactate, synovial T cells lose their mobility and are trapped in the tissue niche. The linkage of defective DNA repair, misbalanced metabolic pathways, autoimmunity, and tissue inflammation in RA encourages metabolic interference as a novel treatment strategy during both the early stages of tolerance breakdown and the late stages of tissue inflammation. Defining and targeting metabolic abnormalities provides a new paradigm to treat, or even prevent, the cellular defects underlying autoimmune disease.
Collapse
Affiliation(s)
- Jingtao Qiu
- Department of Medicine, Stanford University School of Medicine, Stanford, CA, United States
| | - Bowen Wu
- Department of Medicine, Stanford University School of Medicine, Stanford, CA, United States
| | - Stuart B Goodman
- Department of Orthopedic Surgery, Stanford University School of Medicine, Stanford, CA, United States
| | - Gerald J Berry
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, United States
| | - Jorg J Goronzy
- Department of Medicine, Stanford University School of Medicine, Stanford, CA, United States
| | - Cornelia M Weyand
- Department of Medicine, Stanford University School of Medicine, Stanford, CA, United States
| |
Collapse
|
42
|
Expansion of CD4 T Lymphocytes Expressing Interleukin 17 and Tumor Necrosis Factor in Patients with Major Depressive Disorder. J Pers Med 2021; 11:jpm11030220. [PMID: 33808804 PMCID: PMC8003656 DOI: 10.3390/jpm11030220] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2021] [Revised: 02/27/2021] [Accepted: 03/16/2021] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND We have investigated the distribution of the Th1, Th2 and Th17 subsets in circulating CD4+ T lymphocytes and their naïve (TN), effector (TE), central (TCM) and effector memory (TEM) activation/differentiation stages in patients with major depressive disorder (MDD). METHODS Thirty MDD patients and 30 healthy controls were studied. The counts of circulating CD4+ T lymphocytes and their distribution on the TN, TE, TCM and TEM activation/differentiation stages were analyzed by polychromatic flow cytometry. The intracytoplasmic interferon gamma (IFNγ), interleukin (IL)-4, IL-17A and tumor necrosis factor alpha (TNF-alpha) and membrane CD28 expression were also measured. The serum IFNγ, IL-4, Il-17A and TNF-alpha were measured by Luminex, respectively. RESULTS MDD patients had normal counts of CD4+ T lymphocytes and of their TN, TCM and TEM subsets but increased number and percentage of TE CD4+ subset. CD4+ T lymphocytes had significantly enhanced percentage of cells that express IL-17 and TNF-alpha explained by the expansions found in the TN, TCM and, TEM and TCM, TEM and TE activation/differentiation stages, respectively. A selective increase in the percentages of TCM and TEM expressing IFNγ was also observed. We found a significant correlation between the percentages of CD4+ T lymphocytes expressing IFNγ and TNF-alpha in these patients. MDD patients showed increased serum levels of IL-17 and TNF-alpha, but normal IFNγ and IL-4 concentration. LIMITATIONS the cross-sectional nature of the study could be considered a limitation. CONCLUSIONS MDD patients have abnormal circulating CD4+ T lymphocytes with expansion of the IL-17 and TNF-alpha expressing cells as well as increased levels of circulating IL-17 and TNF-alpha.
Collapse
|
43
|
Reitsema RD, Boots AMH, van der Geest KSM, Sandovici M, Heeringa P, Brouwer E. CD8+ T Cells in GCA and GPA: Bystanders or Active Contributors? Front Immunol 2021; 12:654109. [PMID: 33815414 PMCID: PMC8015776 DOI: 10.3389/fimmu.2021.654109] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2021] [Accepted: 03/01/2021] [Indexed: 12/28/2022] Open
Abstract
Vasculitis refers to inflammation of blood vessels and can cause a variety of serious complications depending on which vessels are affected. Two different forms of vasculitis are Giant Cell Arteritis (GCA) and Granulomatosis with Polyangiitis (GPA). GCA is the most common form of vasculitis in adults affecting the large arteries and can lead to visual impairment and development of aneurysms. GPA affects small- and medium-sized blood vessels predominantly in the lungs and kidneys resulting in organ failure. Both diseases can potentially be fatal. Although the pathogenesis of GCA and GPA are incompletely understood, a prominent role for CD4+ T cells has been implicated in both diseases. More recently, the role of CD8+ T cells has gained renewed interest. CD8+ T cells are important players in the adaptive immune response against intracellular microorganisms. After a general introduction on the different forms of vasculitis and their association with infections and CD8+ T cells, we review the current knowledge on CD8+ T-cell involvement in the immunopathogenesis of GCA and GPA focusing on phenotypic and functional features of circulating and lesional CD8+ T cells. Furthermore, we discuss to which extent aging is associated with CD8+ T-cell phenotype and function in GCA and GPA.
Collapse
Affiliation(s)
- Rosanne D Reitsema
- Department of Rheumatology and Clinical Immunology, University of Groningen, University Medical Center Groningen, Groningen, Netherlands
| | - Annemieke M H Boots
- Department of Rheumatology and Clinical Immunology, University of Groningen, University Medical Center Groningen, Groningen, Netherlands
| | - Kornelis S M van der Geest
- Department of Rheumatology and Clinical Immunology, University of Groningen, University Medical Center Groningen, Groningen, Netherlands
| | - Maria Sandovici
- Department of Rheumatology and Clinical Immunology, University of Groningen, University Medical Center Groningen, Groningen, Netherlands
| | - Peter Heeringa
- Department of Pathology and Medical Biology, University of Groningen, University Medical Center Groningen, Groningen, Netherlands
| | - Elisabeth Brouwer
- Department of Rheumatology and Clinical Immunology, University of Groningen, University Medical Center Groningen, Groningen, Netherlands
| |
Collapse
|
44
|
Wang Y, Fu Z, Li X, Liang Y, Pei S, Hao S, Zhu Q, Yu T, Pei Y, Yuan J, Ye J, Fu J, Xu J, Hong J, Yang R, Hou H, Huang X, Peng C, Zheng M, Xiao Y. Cytoplasmic DNA sensing by KU complex in aged CD4 + T cell potentiates T cell activation and aging-related autoimmune inflammation. Immunity 2021; 54:632-647.e9. [PMID: 33667382 DOI: 10.1016/j.immuni.2021.02.003] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2020] [Revised: 09/19/2020] [Accepted: 02/08/2021] [Indexed: 02/06/2023]
Abstract
Aging is associated with DNA accumulation and increased homeostatic proliferation of circulating T cells. Although these attributes are associated with aging-related autoimmunity, their direct contributions remain unclear. Conventionally, KU complex, the regulatory subunit of DNA-dependent protein kinase (DNA-PK), together with the catalytic subunit of DNA-PK (DNA-PKcs), mediates DNA damage repair in the nucleus. Here, we found KU complex abundantly expressed in the cytoplasm, where it recognized accumulated cytoplasmic DNA in aged human and mouse CD4+ T cells. This process enhanced T cell activation and pathology of experimental autoimmune encephalomyelitis (EAE) in aged mice. Mechanistically, KU-mediated DNA sensing facilitated DNA-PKcs recruitment and phosphorylation of the kinase ZAK. This activated AKT and mTOR pathways, promoting CD4+ T cell proliferation and activation. We developed a specific ZAK inhibitor, which dampened EAE pathology in aged mice. Overall, these findings demonstrate a KU-mediated cytoplasmic DNA-sensing pathway in CD4+ T cells that potentiates aging-related autoimmunity.
Collapse
Affiliation(s)
- Yan Wang
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Zunyun Fu
- Drug Discovery and Design Center, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai 201203, China; School of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Xutong Li
- Drug Discovery and Design Center, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai 201203, China
| | - Yinming Liang
- School of Laboratory Medicine, Xinxiang Medical University, Xinxiang 453003, China
| | - Siyu Pei
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Shumeng Hao
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Qingchen Zhu
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Tao Yu
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Yifei Pei
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Jia Yuan
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Jialin Ye
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Jiemeng Fu
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Jing Xu
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Jin Hong
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China; Comprehensive Breast Health Center, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 200025, China
| | - Ruirui Yang
- Drug Discovery and Design Center, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai 201203, China; Shanghai Institute for Advanced Immunochemical Studies, and School of Life Science and Technology, ShanghaiTech University, Shanghai 200031, China
| | - Hui Hou
- Drug Discovery and Design Center, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai 201203, China
| | - Xinfang Huang
- Department of Rheumatology, Shanghai East Hospital, Tongji University, School of Medicine, Shanghai 200120, China
| | - Chao Peng
- National Facility for Protein Science in Shanghai, Zhangjiang Lab, Shanghai 201210, China
| | - Mingyue Zheng
- Drug Discovery and Design Center, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai 201203, China.
| | - Yichuan Xiao
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China.
| |
Collapse
|
45
|
Nian Y, Iske J, Maenosono R, Minami K, Heinbokel T, Quante M, Liu Y, Azuma H, Yang J, Abdi R, Zhou H, Elkhal A, Tullius SG. Targeting age-specific changes in CD4 + T cell metabolism ameliorates alloimmune responses and prolongs graft survival. Aging Cell 2021; 20:e13299. [PMID: 33497523 PMCID: PMC7884034 DOI: 10.1111/acel.13299] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2020] [Revised: 11/16/2020] [Accepted: 12/08/2020] [Indexed: 12/12/2022] Open
Abstract
Age impacts alloimmunity. Effects of aging on T-cell metabolism and the potential to interfere with immunosuppressants have not been explored yet. Here, we dissected metabolic pathways of CD4+ and CD8+ T cells in aging and offer novel immunosuppressive targets. Upon activation, CD4+ T cells from old mice failed to exhibit adequate metabolic reprogramming resulting into compromised metabolic pathways, including oxidative phosphorylation (OXPHOS) and glycolysis. Comparable results were also observed in elderly human patients. Although glutaminolysis remained the dominant and age-independent source of mitochondria for activated CD4+ T cells, old but not young CD4+ T cells relied heavily on glutaminolysis. Treating young and old murine and human CD4+ T cells with 6-diazo-5-oxo-l-norleucine (DON), a glutaminolysis inhibitor resulted in significantly reduced IFN-γ production and compromised proliferative capacities specifically of old CD4+ T cells. Of translational relevance, old and young mice that had been transplanted with fully mismatched skin grafts and treated with DON demonstrated dampened Th1- and Th17-driven alloimmune responses. Moreover, DON diminished cytokine production and proliferation of old CD4+ T cells in vivo leading to a significantly prolonged allograft survival specifically in old recipients. Graft prolongation in young animals, in contrast, was only achieved when DON was applied in combination with an inhibition of glycolysis (2-deoxy-d-glucose, 2-DG) and OXPHOS (metformin), two alternative metabolic pathways. Notably, metabolic treatment had not been linked to toxicities. Remarkably, immunosuppressive capacities of DON were specific to CD4+ T cells as adoptively transferred young CD4+ T cells prevented immunosuppressive capacities of DON on allograft survival in old recipients. Depletion of CD8+ T cells did not alter transplant outcomes in either young or old recipients. Taken together, our data introduce an age-specific metabolic reprogramming of CD4+ T cells. Targeting those pathways offers novel and age-specific approaches for immunosuppression.
Collapse
Affiliation(s)
- Yeqi Nian
- Division of Transplant Surgery and Transplant Surgery Research Laboratory Brigham and Women's Hospital Harvard Medical School Boston MA USA
- Department of Urology Second Xiangya Hospital Central South University Changsha China
- Department of Kidney Transplantation Tianjin First Central Hospital Nankai University Tianjin China
| | - Jasper Iske
- Division of Transplant Surgery and Transplant Surgery Research Laboratory Brigham and Women's Hospital Harvard Medical School Boston MA USA
- Institute of Transplant Immunology Hannover Medical School Hannover Germany
| | - Ryoichi Maenosono
- Division of Transplant Surgery and Transplant Surgery Research Laboratory Brigham and Women's Hospital Harvard Medical School Boston MA USA
- Department of Urology Osaka Medical College Osaka Japan
| | - Koichiro Minami
- Division of Transplant Surgery and Transplant Surgery Research Laboratory Brigham and Women's Hospital Harvard Medical School Boston MA USA
- Department of Urology Osaka Medical College Osaka Japan
| | - Timm Heinbokel
- Division of Transplant Surgery and Transplant Surgery Research Laboratory Brigham and Women's Hospital Harvard Medical School Boston MA USA
- Department of Pathology Charité – Universitätsmedizin Berlin Berlin Germany
| | - Markus Quante
- Department of General, Visceral‐ and Transplant Surgery University Hospital Tübingen Tubingen Germany
| | - Yang Liu
- Division of Transplant Surgery and Transplant Surgery Research Laboratory Brigham and Women's Hospital Harvard Medical School Boston MA USA
- Institute of Hepatobiliary Diseases Zhongnan Hospital of Wuhan University Wuhan China
| | | | - Jinrui Yang
- Department of Urology Second Xiangya Hospital Central South University Changsha China
| | - Reza Abdi
- Renal Division Transplantation Research Center Brigham and Women's Hospital Harvard Medical School Boston MA USA
| | - Hao Zhou
- Division of Transplant Surgery and Transplant Surgery Research Laboratory Brigham and Women's Hospital Harvard Medical School Boston MA USA
| | - Abdallah Elkhal
- Division of Transplant Surgery and Transplant Surgery Research Laboratory Brigham and Women's Hospital Harvard Medical School Boston MA USA
| | - Stefan G. Tullius
- Division of Transplant Surgery and Transplant Surgery Research Laboratory Brigham and Women's Hospital Harvard Medical School Boston MA USA
| |
Collapse
|
46
|
Wang X, Bootsma H, Terpstra J, Vissink A, van der Vegt B, Spijkervet FKL, Kroese FGM, Pringle S. Progenitor cell niche senescence reflects pathology of the parotid salivary gland in primary Sjögren's syndrome. Rheumatology (Oxford) 2021; 59:3003-3013. [PMID: 32159757 PMCID: PMC7516109 DOI: 10.1093/rheumatology/keaa012] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2019] [Revised: 12/20/2019] [Indexed: 12/13/2022] Open
Abstract
Objective Salivary gland (SG) progenitor cells (SGPCs) maintain SG homeostasis. We have previously shown that in primary Sjögren’s syndrome (pSS), SGPCs are likely to be senescent, and may underpin SG dysfunction. This study assessed the extent of senescence of cells in a SGPC niche in pSS patients’ SGs, and its correlation with functional and clinical parameters. Methods The expression of p16 and p21 as markers of senescence in both total SG epithelium and a SGPC niche (basal striated duct cells, BSD) was examined in SGs of pSS (n = 35), incomplete pSS (n = 13) (patients with some signs of pSS, but not fulfilling all classification criteria) and non-SS sicca control (n = 21) patients. This was correlated with functional and clinical parameters. Results pSS patient SGs contained significantly more p16+ cells both in the epithelium in general (P <0.01) and in the BSD layer (P <0.001), than non-SS SGs. Significant correlations were found in pSS patients between p16+ BSD cells and secretion of unstimulated whole saliva, stimulated whole saliva, stimulated parotid saliva, CD45+ infiltrate, ultrasound total score and ACR-EULAR classification score, but not with EULAR Sjögren’s syndrome disease activity index (ESSDAI) and EULAR Sjögren’s Syndrome Patient Reported Index (ESSPRI) scores. Correlations with total epithelium p16+ cells were weaker. Incomplete pSS patients also had increased numbers of p16+ epithelial and BSD cells. Based on protein and mRNA expression, p21+ appears not to play a significant role in the SG in pSS. Conclusion These findings suggest SGPC senescence may be an early feature of primary Sjögren’s syndrome and may contribute to defective SG function in pSS but not to systemic disease activity.
Collapse
Affiliation(s)
- Xiaoyan Wang
- Department of Rheumatology and Clinical ImmunologyUniversity Medical Center Groningen, Groningen, The Netherlands
| | - Hendrika Bootsma
- Department of Rheumatology and Clinical ImmunologyUniversity Medical Center Groningen, Groningen, The Netherlands
| | - Janneke Terpstra
- Department of Rheumatology and Clinical ImmunologyUniversity Medical Center Groningen, Groningen, The Netherlands
| | - Arjan Vissink
- Department of Oral and Maxillofacial Surgery, University Medical Center Groningen, Groningen, The Netherlands
| | - Bert van der Vegt
- Department of Pathology and Medical Biology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Fred K L Spijkervet
- Department of Oral and Maxillofacial Surgery, University Medical Center Groningen, Groningen, The Netherlands
| | - Frans G M Kroese
- Department of Rheumatology and Clinical ImmunologyUniversity Medical Center Groningen, Groningen, The Netherlands
| | - Sarah Pringle
- Department of Rheumatology and Clinical ImmunologyUniversity Medical Center Groningen, Groningen, The Netherlands
| |
Collapse
|
47
|
Abstract
The immunopathogenesis of rheumatoid arthritis (RA) spans decades, beginning with the production of autoantibodies against post-translationally modified proteins (checkpoint 1). After years of asymptomatic autoimmunity and progressive immune system remodeling, tissue tolerance erodes and joint inflammation ensues as tissue-invasive effector T cells emerge and protective joint-resident macrophages fail (checkpoint 2). The transition of synovial stromal cells into autoaggressive effector cells converts synovitis from acute to chronic destructive (checkpoint 3). The loss of T cell tolerance derives from defective DNA repair, causing abnormal cell cycle dynamics, telomere fragility and instability of mitochondrial DNA. Mitochondrial and lysosomal anomalies culminate in the generation of short-lived tissue-invasive effector T cells. This differentiation defect builds on a metabolic platform that shunts glucose away from energy generation toward the cell building and motility programs. The next frontier in RA is the development of curative interventions, for example, reprogramming T cell defects during the period of asymptomatic autoimmunity.
Collapse
Affiliation(s)
- Cornelia M Weyand
- Department of Medicine, Stanford University School of Medicine, Stanford, CA, USA.
- Department of Medicine, Veterans Affairs Palo Alto Health Care System, Palo Alto, CA, USA.
| | - Jörg J Goronzy
- Department of Medicine, Stanford University School of Medicine, Stanford, CA, USA
- Department of Medicine, Veterans Affairs Palo Alto Health Care System, Palo Alto, CA, USA
| |
Collapse
|
48
|
Gurusamy D, Selvamurugesan V, Kalyanasundaram S, Kalyanaraman S. Is COVID 19, a beginning of new entity called chronic viral systemic inflammatory syndrome? AIMS ALLERGY AND IMMUNOLOGY 2021. [DOI: 10.3934/allergy.2021010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
|
49
|
Gomes JAN, da Silva Dias GA, Fujihara S, Yoshikawa GT, Koyama RVL, Sousa RCM, Quaresma JAS, Fuzii HT. Decrease in naïve T cell production due to HTLV-1-associated myelopathy/tropical spastic paraparesis (HAM/TSP) development. Immunobiology 2020; 226:152050. [PMID: 33338979 DOI: 10.1016/j.imbio.2020.152050] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2020] [Revised: 10/25/2020] [Accepted: 11/29/2020] [Indexed: 10/22/2022]
Abstract
Human T-lymphocytic virus 1 (HTLV-1) is mainly associated with adult T-cell leukemia/lymphoma (ATLL) and HTLV-1-associated myelopathy/tropical spastic paraparesis (HAM/TSP). Patients with HAM/TSP exhibit significant changes in their immune response, and HTLV-1 infection can interfere in cytokine production and perhaps in T cell production. The aims of this study were to evaluate thymic function in HAM/TSP patients and HTLV-1 healthy carriers (HCs) and correlate it to age and interleukin 7 (IL-7) gene expression. Thymic function in 21 HAM/TSP patients and 12 HCs was evaluated by quantifying T cell receptor rearrangement excision circle (TREC) particles and IL-7 gene expression, both measured by quantitative polymerase chain reaction. HAM/TSP patients presented lower TREC particle counts (p = 0.0112) and lower IL-7 expression (p = 0.0102) than HCs. Both TREC particles and IL-7 gene expression were separately analyzed in two age groups: ≤ 59 years and ≥60 years, The ≤59-year-old HAM/TSP patients had a lower TREC count compared with the ≤59-year-old HCs (p = 0.0476). In conclusion, HAM/TSP development could interfere with thymic function because the results showed TREC particle reduction in HAM/TSP patients in relation to HCs, and it could be associated with a concomitant reduction in IL-7 expression.
Collapse
Affiliation(s)
| | | | - Satomi Fujihara
- Instituto de Ciências da Saúde, Universidade Federal do Pará, Belém-Pará, Brazil
| | | | | | | | | | - Hellen Thais Fuzii
- Núcleo de Medicina Tropical, Universidade Federal do Pará, Belém-Pará, Brazil
| |
Collapse
|
50
|
CRELD1 modulates homeostasis of the immune system in mice and humans. Nat Immunol 2020; 21:1517-1527. [PMID: 33169013 DOI: 10.1038/s41590-020-00811-2] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2019] [Accepted: 09/16/2020] [Indexed: 01/01/2023]
Abstract
CRELD1 is a pivotal factor for heart development, the function of which is unknown in adult life. We here provide evidence that CRELD1 is an important gatekeeper of immune system homeostasis. Exploiting expression variance in large human cohorts contrasting individuals with the lowest and highest CRELD1 expression levels revealed strong phenotypic, functional and transcriptional differences, including reduced CD4+ T cell numbers. These findings were validated in T cell-specific Creld1-deficient mice. Loss of Creld1 was associated with simultaneous overactivation and increased apoptosis, resulting in a net loss of T cells with age. Creld1 was transcriptionally and functionally linked to Wnt signaling. Collectively, gene expression variance in large human cohorts combined with murine genetic models, transcriptomics and functional testing defines CRELD1 as an important modulator of immune homeostasis.
Collapse
|