1
|
Admasu TD, Yu JS. Harnessing Immune Rejuvenation: Advances in Overcoming T Cell Senescence and Exhaustion in Cancer Immunotherapy. Aging Cell 2025; 24:e70055. [PMID: 40178455 PMCID: PMC12073907 DOI: 10.1111/acel.70055] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2024] [Revised: 02/15/2025] [Accepted: 03/14/2025] [Indexed: 04/05/2025] Open
Abstract
Immunotherapy has transformed the landscape of cancer treatment, with T cell-based strategies at the forefront of this revolution. However, the durability of these responses is frequently undermined by two intertwined phenomena: T cell exhaustion and senescence. While exhaustion is driven by chronic antigen exposure in the immunosuppressive tumor microenvironment, leading to a reversible state of diminished functionality, senescence reflects a more permanent, age- or stress-induced arrest in cellular proliferation and effector capacity. Together, these processes represent formidable barriers to sustained anti-tumor immunity. In this review, we dissect the molecular underpinnings of T cell exhaustion and senescence, revealing how these dysfunctions synergistically contribute to immune evasion and resistance across a range of solid tumors. We explore cutting-edge therapeutic approaches aimed at rewiring the exhausted and senescent T cell phenotypes. These include advances in immune checkpoint blockade, the engineering of "armored" CAR-T cells, senolytic therapies that selectively eliminate senescent cells, and novel interventions that reinvigorate the immune system's capacity for tumor eradication. By spotlighting emerging strategies that target both exhaustion and senescence, we provide a forward-looking perspective on the potential to harness immune rejuvenation. This comprehensive review outlines the next frontier in cancer immunotherapy: unlocking durable responses by overcoming the immune system's intrinsic aging and exhaustion, ultimately paving the way for transformative therapeutic breakthroughs.
Collapse
Affiliation(s)
| | - John S. Yu
- Department of NeurosurgeryCedars‐Sinai Medical CenterLos AngelesCaliforniaUSA
- Kairos PharmaLos AngelesCaliforniaUSA
| |
Collapse
|
2
|
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
|
3
|
Xu G, Wang Y. CNPY2 modulates senescence-associated secretory phenotype in tendon stem/progenitor cells. Tissue Cell 2025; 93:102706. [PMID: 39755057 DOI: 10.1016/j.tice.2024.102706] [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: 06/24/2024] [Revised: 12/22/2024] [Accepted: 12/23/2024] [Indexed: 01/06/2025]
Abstract
Age-related diseases are often linked to chronic inflammation. Senescent cells secrete inflammatory cytokines, chemokines and matrix metalloproteinases, collectively referred to as the senescence-associated secretory phenotype (SASP). The current study discovered that aging leads to the accumulation of senescent tendon stem/progenitor cells (TSPCs) in tendon tissue, resulting in the development of a SASP. Conditioned medium from aged TSPCs induced cellular inflammation in young TSPCs. In addition, we found that Canopy homolog 2 (CNPY2) expression is reduced during tendon aging. CNPY2 deficiency causes TSPCs senescence and SASP. Our findings showed that the NF-κB signaling pathway is activated in CNPY2 knockdown TSPCs, pharmacological inhibition of NF-κB signaling pathway with BMS-345541 attenuated SASP of senescent TSPCs, which indicated that CNPY2 regulates TSPCs SASP might through NF-κB signaling pathway. Our findings suggested that CNPY2 plays an important role in TSPCs senescence and SASP, CNPY2 could be a promising target for age-related tendon disorders.
Collapse
Affiliation(s)
- Gang Xu
- Department of Orthopaedics, Affiliated Hospital of Nantong University, Nantong, Jiangsu, China; Department of Orthopaedics, The Eighth People's Hospital of Tongzhou, Nantong, Jiangsu, China
| | - Youhua Wang
- Department of Orthopaedics, Affiliated Hospital of Nantong University, Nantong, Jiangsu, China.
| |
Collapse
|
4
|
Bracken OV, De Maeyer RPH, Akbar AN. Enhancing immunity during ageing by targeting interactions within the tissue environment. Nat Rev Drug Discov 2025; 24:300-315. [PMID: 39875569 DOI: 10.1038/s41573-024-01126-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/16/2024] [Indexed: 01/30/2025]
Abstract
Immunity declines with age. This results in a higher risk of age-related diseases, diminished ability to respond to new infections and reduced response to vaccines. The causes of this immune dysfunction are cellular senescence, which occurs in both lymphoid and non-lymphoid tissue, and chronic, low-grade inflammation known as 'inflammageing'. In this Review article, we highlight how the processes of inflammation and senescence drive each other, leading to loss of immune function. To break this cycle, therapies are needed that target the interactions between the altered tissue environment and the immune system instead of targeting each component alone. We discuss the relative merits and drawbacks of therapies that are directed at eliminating senescent cells (senolytics) and those that inhibit inflammation (senomorphics) in the context of tissue niches. Furthermore, we discuss therapeutic strategies designed to directly boost immune cell function and improve immune surveillance in tissues.
Collapse
Affiliation(s)
| | - Roel P H De Maeyer
- Division of Medicine, University College London, London, UK
- Botnar Research Centre, Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, University of Oxford, Oxford, UK
| | - Arne N Akbar
- Division of Medicine, University College London, London, UK.
| |
Collapse
|
5
|
Fukumoto T, Shimosawa T, Yakabe M, Yoshida S, Yoshida Y. Recent advances in biomarkers for senescence: Bridging basic research to clinic. Geriatr Gerontol Int 2025; 25:139-147. [PMID: 39754295 DOI: 10.1111/ggi.15054] [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: 03/11/2024] [Revised: 10/31/2024] [Accepted: 12/14/2024] [Indexed: 01/06/2025]
Abstract
In this review, we review the current status of biomarkers for aging and possible perspectives on anti-aging or rejuvenation from the standpoint of biomarkers. Aging is observed in all cells and organs, and we focused on research into senescence in the skin, musculoskeletal system, immune system, and cardiovascular system. Commonly used biomarkers include SA-βgal, cell-cycle markers, senescence-associated secretory phenotype (SASP) factors, damage-associated molecular patterns (DAMPs), and DNA-damage-related markers. In addition, each organ or cell has its specific markers. Generally speaking, a combination of biomarkers is required to define age-related changes. When considering the translation of basic research, biomarkers that are highly sensitive, highly specific, with validation and reliability as well as being non-invasive are optimal; however, currently reported markers do not fulfill the prerequisite for biomarkers. In addition, rodent models of aging do not necessarily represent human aging, and markers in rodent or cell models are not applicable in clinical settings. The prerequisite of clinically applicable biomarkers is that they provide useful information for clinical decision-making, such as predicting disease risk, diagnosing disease, monitoring disease progression, or guiding treatment decisions. Therefore, the development of non-invasive robust, reliable, and useful biomarkers in humans is necessary to develop anti-aging therapy for humans. Geriatr Gerontol Int 2025; 25: 139-147.
Collapse
Affiliation(s)
- Takeshi Fukumoto
- Division of Dermatology, Department of Internal Related, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Tatsuo Shimosawa
- Department of Clinical Laboratory, Graduate School of Medicine, International University of Health and Welfare, Hyogo, Japan
| | - Mitsutaka Yakabe
- Department of Geriatric Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Shota Yoshida
- Department of Geriatric and General Medicine, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Yohko Yoshida
- Department of Advanced Senotherapeutics and Department of Cardiovascular Biology and Medicine, Juntendo University Graduate School of Medicine, Tokyo, Japan
| |
Collapse
|
6
|
Yildiz O, Hunt GP, Schroth J, Dhillon G, Spargo TP, Al-Chalabi A, Koks S, Turner MR, Shaw PJ, Henson SM, Iacoangeli A, Malaspina A. Lipid-mediated resolution of inflammation and survival in amyotrophic lateral sclerosis. Brain Commun 2025; 7:fcae402. [PMID: 39816195 PMCID: PMC11733686 DOI: 10.1093/braincomms/fcae402] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2024] [Revised: 07/26/2024] [Accepted: 01/10/2025] [Indexed: 01/18/2025] Open
Abstract
Neuroinflammation impacts on the progression of amyotrophic lateral sclerosis (ALS), a fatal neurodegenerative disorder. Specialized pro-resolving mediators trigger the resolution of inflammation. We investigate the specialized pro-resolving mediator blood profile and their receptors' expression in peripheral blood mononuclear cells in relation to survival in ALS. People living with ALS (pwALS) were stratified based on bulbar versus limb onset and on key progression metrics using a latent class model, to separate faster progressing from slower progressing ALS. Specialized pro-resolving mediator blood concentrations were measured at baseline and in one additional visit in 20 pwALS and 10 non-neurological controls (Cohort 1). Flow cytometry was used to study the GPR32 and GPR18 resolvin receptors' expression in peripheral blood mononuclear cells from 40 pwALS and 20 non-neurological controls (Cohort 2) at baseline and in two additional visits in 17 pwALS. Survival analysis was performed using Cox proportional hazards models, including known clinical predictors and GPR32 and GPR18 mononuclear cell expression. Differential expression and linear discriminant analyses showed that plasma resolvins were able to distinguish phenotypic variants of ALS from non-neurological controls. RvE3 was elevated in blood from pwALS, whilst RvD1, RvE3, RvT4 and RvD1n-3 DPA were upregulated in A-S and RvD2 in A-F. Compared to non-neurological controls, GPR32 was upregulated in monocytes expressing the active inflammation-suppressing CD11b+ integrin from fast-progressing pwALS, including those with bulbar onset disease (P < 0.0024), whilst GPR32 and GPR18 were downregulated in most B and T cell subtypes. Only GPR18 was upregulated in naïve double positive Tregs, memory cytotoxic Tregs, senescent late memory B cells and late senescent CD8+ T cells from pwALS compared to non-neurological controls (P < 0.0431). Higher GPR32 and GPR18 median expression in blood mononuclear cells was associated with longer survival, with GPR32 expression in classical monocytes (hazard ratio: 0.11, P = 0.003) and unswitched memory B cells (hazard ratio: 0.44, P = 0.008) showing the most significant association, along with known clinical predictors. Low levels of resolvins and downregulation of their membrane receptors in blood mononuclear cells are linked to a faster progression of ALS. Higher mononuclear cell expression of resolvin receptors is a predictor of longer survival. These findings suggest a lipid-mediated neuroprotective response that could be harnessed to develop novel therapeutic strategies and biomarkers for ALS.
Collapse
Affiliation(s)
- Ozlem Yildiz
- Neuromuscular Department, Motor Neuron Disease Centre, Queen Square Institute of Neurology, University College London, London WC1N 3BG, UK
- Neuroscience and Trauma, The Blizard Institute, Queen Mary University of London, London E1 2AT, UK
| | - Guy P Hunt
- Department of Biostatistics and Health Informatics, Institute of Psychiatry, Psychology and Neuroscience, King’s College London, London SE5 8AF, UK
- Department of Basic and Clinical Neuroscience, Maurice Wohl Clinical Neuroscience Institute, Institute of Psychiatry, Psychology and Neuroscience, King’s College London, London SE5 9RT, UK
- Perron Institute for Neurological and Translational Science, Research Institute in Nedlands, WA 6009, Australia
- Centre for Molecular Medicine and Innovative Therapeutics, Murdoch University, Perth 6150, Australia
| | - Johannes Schroth
- Translational Medicine and Therapeutics, William Harvey Research Institute, Queen Mary University of London, London EC1M 6BQ, UK
| | - Gurleen Dhillon
- Neuroscience and Trauma, The Blizard Institute, Queen Mary University of London, London E1 2AT, UK
| | - Thomas P Spargo
- Department of Basic and Clinical Neuroscience, Maurice Wohl Clinical Neuroscience Institute, Institute of Psychiatry, Psychology and Neuroscience, King’s College London, London SE5 9RT, UK
| | - Ammar Al-Chalabi
- Department of Basic and Clinical Neuroscience, Maurice Wohl Clinical Neuroscience Institute, Institute of Psychiatry, Psychology and Neuroscience, King’s College London, London SE5 9RT, UK
- Maurice Wohl Clinical Neuroscience Institute, King’s College Hospital, London SE5 9RS, UK
| | - Sulev Koks
- Perron Institute for Neurological and Translational Science, Research Institute in Nedlands, WA 6009, Australia
- Centre for Molecular Medicine and Innovative Therapeutics, Murdoch University, Perth 6150, Australia
| | - Martin R Turner
- Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford OX3 7JX, UK
| | - Pamela J Shaw
- Sheffield Institute for Translational Neuroscience, University of Sheffield, Sheffield S10 2HQ, UK
| | - Sian M Henson
- Translational Medicine and Therapeutics, William Harvey Research Institute, Queen Mary University of London, London EC1M 6BQ, UK
| | - Alfredo Iacoangeli
- Department of Biostatistics and Health Informatics, Institute of Psychiatry, Psychology and Neuroscience, King’s College London, London SE5 8AF, UK
- Department of Basic and Clinical Neuroscience, Maurice Wohl Clinical Neuroscience Institute, Institute of Psychiatry, Psychology and Neuroscience, King’s College London, London SE5 9RT, UK
- National Institute for Health Research Biomedical Research Centre and Dementia Unit at South London and Maudsley NHS Foundation Trust and King’s College London, London SE5 8AF, UK
| | - Andrea Malaspina
- Neuromuscular Department, Motor Neuron Disease Centre, Queen Square Institute of Neurology, University College London, London WC1N 3BG, UK
- Neuroscience and Trauma, The Blizard Institute, Queen Mary University of London, London E1 2AT, UK
| |
Collapse
|
7
|
Suryadevara V, Hudgins AD, Rajesh A, Pappalardo A, Karpova A, Dey AK, Hertzel A, Agudelo A, Rocha A, Soygur B, Schilling B, Carver CM, Aguayo-Mazzucato C, Baker DJ, Bernlohr DA, Jurk D, Mangarova DB, Quardokus EM, Enninga EAL, Schmidt EL, Chen F, Duncan FE, Cambuli F, Kaur G, Kuchel GA, Lee G, Daldrup-Link HE, Martini H, Phatnani H, Al-Naggar IM, Rahman I, Nie J, Passos JF, Silverstein JC, Campisi J, Wang J, Iwasaki K, Barbosa K, Metis K, Nernekli K, Niedernhofer LJ, Ding L, Wang L, Adams LC, Ruiyang L, Doolittle ML, Teneche MG, Schafer MJ, Xu M, Hajipour M, Boroumand M, Basisty N, Sloan N, Slavov N, Kuksenko O, Robson P, Gomez PT, Vasilikos P, Adams PD, Carapeto P, Zhu Q, Ramasamy R, Perez-Lorenzo R, Fan R, Dong R, Montgomery RR, Shaikh S, Vickovic S, Yin S, Kang S, Suvakov S, Khosla S, Garovic VD, Menon V, Xu Y, Song Y, Suh Y, Dou Z, Neretti N. SenNet recommendations for detecting senescent cells in different tissues. Nat Rev Mol Cell Biol 2024; 25:1001-1023. [PMID: 38831121 PMCID: PMC11578798 DOI: 10.1038/s41580-024-00738-8] [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] [Accepted: 04/15/2024] [Indexed: 06/05/2024]
Abstract
Once considered a tissue culture-specific phenomenon, cellular senescence has now been linked to various biological processes with both beneficial and detrimental roles in humans, rodents and other species. Much of our understanding of senescent cell biology still originates from tissue culture studies, where each cell in the culture is driven to an irreversible cell cycle arrest. By contrast, in tissues, these cells are relatively rare and difficult to characterize, and it is now established that fully differentiated, postmitotic cells can also acquire a senescence phenotype. The SenNet Biomarkers Working Group was formed to provide recommendations for the use of cellular senescence markers to identify and characterize senescent cells in tissues. Here, we provide recommendations for detecting senescent cells in different tissues based on a comprehensive analysis of existing literature reporting senescence markers in 14 tissues in mice and humans. We discuss some of the recent advances in detecting and characterizing cellular senescence, including molecular senescence signatures and morphological features, and the use of circulating markers. We aim for this work to be a valuable resource for both seasoned investigators in senescence-related studies and newcomers to the field.
Collapse
Affiliation(s)
- Vidyani Suryadevara
- Department of Radiology, Molecular Imaging Program at Stanford (MIPS), Stanford University, School of Medicine, Stanford, CA, USA
| | - Adam D Hudgins
- Department of Obstetrics and Gynecology, Columbia University, New York, NY, USA
| | - Adarsh Rajesh
- Sanford Burnham Prebys Medical Discovery Institute, Cancer Genome and Epigenetics Program, La Jolla, CA, USA
| | | | - Alla Karpova
- Department of Medicine, Washington University School of Medicine, St. Louis, MO, USA
| | - Amit K Dey
- National Institute on Aging, NIH, Baltimore, MD, USA
| | - Ann Hertzel
- Department of Biochemistry, Molecular Biology and Biophysics, University of Minnesota, Minneapolis, MN, USA
- Institute on the Biology of Aging and Metabolism, University of Minnesota, Minneapolis, MN, USA
| | - Anthony Agudelo
- Department of Molecular Biology, Cell Biology, and Biochemistry, Brown University, Providence, RI, USA
- Center on the Biology of Aging, Brown University, Providence, RI, USA
| | - Azucena Rocha
- Department of Molecular Biology, Cell Biology, and Biochemistry, Brown University, Providence, RI, USA
- Center on the Biology of Aging, Brown University, Providence, RI, USA
| | - Bikem Soygur
- The Buck Institute for Research on Aging, Novato, CA, USA
| | | | - Chase M Carver
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN, USA
- Robert and Arlene Kogod Center on Aging, Rochester, MN, USA
| | - Cristina Aguayo-Mazzucato
- Islet Cell Biology and Regenerative Medicine, Joslin Diabetes Center, Harvard Medical School, Boston, USA
| | - Darren J Baker
- Robert and Arlene Kogod Center on Aging, Rochester, MN, USA
- Department of Biochemistry and Molecular Biology, Department of Pediatric and Adolescent Medicine, Mayo Clinic, Rochester, MN, USA
| | - David A Bernlohr
- Department of Biochemistry, Molecular Biology and Biophysics, University of Minnesota, Minneapolis, MN, USA
- Institute on the Biology of Aging and Metabolism, University of Minnesota, Minneapolis, MN, USA
| | - Diana Jurk
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN, USA
- Robert and Arlene Kogod Center on Aging, Rochester, MN, USA
- Department of Neurology, Mayo Clinic, Rochester, MN, USA
| | - Dilyana B Mangarova
- Department of Radiology, Molecular Imaging Program at Stanford (MIPS), Stanford University, School of Medicine, Stanford, CA, USA
| | - Ellen M Quardokus
- Department of Intelligent Systems Engineering, Indiana University, Bloomington, IN, USA
| | | | - Elizabeth L Schmidt
- Department of Biochemistry, Molecular Biology and Biophysics, University of Minnesota, Minneapolis, MN, USA
- Institute on the Biology of Aging and Metabolism, University of Minnesota, Minneapolis, MN, USA
| | - Feng Chen
- Department of Medicine, Washington University School of Medicine, St. Louis, MO, USA
| | - Francesca E Duncan
- The Buck Institute for Research on Aging, Novato, CA, USA
- Department of Obstetrics and Gynecology, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | | | - Gagandeep Kaur
- Department of Environmental Medicine, University of Rochester Medical Center, Rochester, NY, USA
| | - George A Kuchel
- UConn Center on Aging, University of Connecticut Health Center, Farmington, CT, USA
- Department of Genetics and Genome Sciences, University of Connecticut Health Center, Farmington, CT, USA
| | - Gung Lee
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN, USA
- Robert and Arlene Kogod Center on Aging, Rochester, MN, USA
| | - Heike E Daldrup-Link
- Department of Radiology, Molecular Imaging Program at Stanford (MIPS), Stanford University, School of Medicine, Stanford, CA, USA
| | - Helene Martini
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN, USA
- Robert and Arlene Kogod Center on Aging, Rochester, MN, USA
| | - Hemali Phatnani
- New York Genome Center, New York, NY, USA
- Department of Neurology, Columbia University Irving Medical Center, New York, NY, USA
| | - Iman M Al-Naggar
- UConn Center on Aging, University of Connecticut Health Center, Farmington, CT, USA
| | - Irfan Rahman
- Department of Environmental Medicine, University of Rochester Medical Center, Rochester, NY, USA
| | - Jia Nie
- Barshop Institute for Longevity and Aging Studies, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
| | - João F Passos
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN, USA
- Robert and Arlene Kogod Center on Aging, Rochester, MN, USA
| | - Jonathan C Silverstein
- Department of Biomedical Informatics, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Judith Campisi
- The Buck Institute for Research on Aging, Novato, CA, USA
| | - Julia Wang
- Department of Medicine, Washington University School of Medicine, St. Louis, MO, USA
| | - Kanako Iwasaki
- Islet Cell Biology and Regenerative Medicine, Joslin Diabetes Center, Harvard Medical School, Boston, USA
| | - Karina Barbosa
- Sanford Burnham Prebys Medical Discovery Institute, Cancer Genome and Epigenetics Program, La Jolla, CA, USA
| | - Kay Metis
- Department of Biomedical Informatics, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Kerem Nernekli
- Department of Radiology, Molecular Imaging Program at Stanford (MIPS), Stanford University, School of Medicine, Stanford, CA, USA
| | - Laura J Niedernhofer
- Department of Biochemistry, Molecular Biology and Biophysics, University of Minnesota, Minneapolis, MN, USA
- Institute on the Biology of Aging and Metabolism, University of Minnesota, Minneapolis, MN, USA
| | - Li Ding
- Department of Medicine, Washington University School of Medicine, St. Louis, MO, USA
| | - Lichao Wang
- UConn Center on Aging, University of Connecticut Health Center, Farmington, CT, USA
- Department of Genetics and Genome Sciences, University of Connecticut Health Center, Farmington, CT, USA
| | - Lisa C Adams
- Department of Radiology, Molecular Imaging Program at Stanford (MIPS), Stanford University, School of Medicine, Stanford, CA, USA
| | - Liu Ruiyang
- Department of Medicine, Washington University School of Medicine, St. Louis, MO, USA
| | - Madison L Doolittle
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN, USA
- Robert and Arlene Kogod Center on Aging, Rochester, MN, USA
- Division of Endocrinology, Diabetes and Metabolism, Mayo Clinic, Rochester, MN, USA
| | - Marcos G Teneche
- Sanford Burnham Prebys Medical Discovery Institute, Cancer Genome and Epigenetics Program, La Jolla, CA, USA
| | - Marissa J Schafer
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN, USA
- Robert and Arlene Kogod Center on Aging, Rochester, MN, USA
- Department of Neurology, Mayo Clinic, Rochester, MN, USA
| | - Ming Xu
- UConn Center on Aging, University of Connecticut Health Center, Farmington, CT, USA
- Department of Genetics and Genome Sciences, University of Connecticut Health Center, Farmington, CT, USA
| | - Mohammadjavad Hajipour
- Department of Radiology, Molecular Imaging Program at Stanford (MIPS), Stanford University, School of Medicine, Stanford, CA, USA
| | | | | | - Nicholas Sloan
- Department of Neurology, Columbia University Medical Center, New York, NY, USA
| | - Nikolai Slavov
- Center on the Biology of Aging, Brown University, Providence, RI, USA
- Department of Bioengineering, Northeastern University, Boston, MA, USA
- Department of Biology, Northeastern University, Boston, MA, USA
- Barnett Institute for Chemical and Biological Analysis, Northeastern University, Boston, MA, USA
| | - Olena Kuksenko
- Department of Neurology, Columbia University Medical Center, New York, NY, USA
| | - Paul Robson
- The Jackson Laboratory for Genomic Medicine, Farmington, CT, USA
- Institute for Systems Genomics, University of Connecticut, Farmington, CT, USA
| | - Paul T Gomez
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN, USA
- Robert and Arlene Kogod Center on Aging, Rochester, MN, USA
| | - Periklis Vasilikos
- Department of Genetics and Development, Columbia University, New York, NY, USA
| | - Peter D Adams
- Sanford Burnham Prebys Medical Discovery Institute, Cancer Genome and Epigenetics Program, La Jolla, CA, USA
| | - Priscila Carapeto
- Islet Cell Biology and Regenerative Medicine, Joslin Diabetes Center, Harvard Medical School, Boston, USA
| | - Quan Zhu
- Center for Epigenomics, University of California, San Diego, CA, USA
| | | | | | - Rong Fan
- Yale-Center for Research on Aging, Yale School of Medicine, New Haven, CT, USA
| | - Runze Dong
- Department of Biochemistry, University of Washington, Seattle, WA, USA
- Graduate Program in Biological Physics, Structure and Design, University of Washington, Seattle, WA, USA
| | - Ruth R Montgomery
- Yale-Center for Research on Aging, Yale School of Medicine, New Haven, CT, USA
| | - Sadiya Shaikh
- Department of Environmental Medicine, University of Rochester Medical Center, Rochester, NY, USA
| | - Sanja Vickovic
- New York Genome Center, New York, NY, USA
- Herbert Irving Institute for Cancer Dynamics, Columbia University, New York, NY, USA
- Department of Biomedical Engineering, Columbia University, New York, NY, USA
- Science for Life Laboratory, Department of Immunology, Genetics and Pathology, Beijer Laboratory for Gene and Neuro Research, Uppsala University, Uppsala, Sweden
| | - Shanshan Yin
- Sanford Burnham Prebys Medical Discovery Institute, Cancer Genome and Epigenetics Program, La Jolla, CA, USA
| | - Shoukai Kang
- Department of Biochemistry, University of Washington, Seattle, WA, USA
| | - Sonja Suvakov
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN, USA
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, MN, USA
| | - Sundeep Khosla
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN, USA
- Robert and Arlene Kogod Center on Aging, Rochester, MN, USA
- Division of Endocrinology, Diabetes and Metabolism, Mayo Clinic, Rochester, MN, USA
| | - Vesna D Garovic
- Department of Obstetrics and Gynecology, Mayo Clinic, Rochester, MN, USA
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, MN, USA
| | - Vilas Menon
- Department of Neurology, Columbia University Irving Medical Center, New York, NY, USA
- Center for Translational and Computational Neuroimmunology, Columbia University Irving Medical Center, New York, NY, USA
| | - Yanxin Xu
- Center for Regenerative Medicine, Massachusetts General Hospital, Boston, MA, USA
- Harvard Stem Cell Institute, Harvard University, Cambridge, MA, USA
- Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Yizhe Song
- Department of Medicine, Washington University School of Medicine, St. Louis, MO, USA
| | - Yousin Suh
- Department of Obstetrics and Gynecology, Columbia University, New York, NY, USA
- Department of Genetics and Development, Columbia University, New York, NY, USA
| | - Zhixun Dou
- Center for Regenerative Medicine, Massachusetts General Hospital, Boston, MA, USA
- Harvard Stem Cell Institute, Harvard University, Cambridge, MA, USA
- Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Nicola Neretti
- Department of Molecular Biology, Cell Biology, and Biochemistry, Brown University, Providence, RI, USA.
- Center on the Biology of Aging, Brown University, Providence, RI, USA.
| |
Collapse
|
8
|
Beckers D, Jainarayanan AK, Dustin ML, Capera J. T Cell Resistance: On the Mechanisms of T Cell Non-activation. Immune Netw 2024; 24:e42. [PMID: 39801736 PMCID: PMC11711127 DOI: 10.4110/in.2024.24.e42] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2024] [Revised: 10/28/2024] [Accepted: 11/12/2024] [Indexed: 01/16/2025] Open
Abstract
Immunological tolerance is a fundamental arm of any functioning immune system. Not only does tolerance mitigate collateral damage from host immune responses, but in doing so permits a robust response sufficient to clear infection as necessary. Yet, despite occupying such a cornerstone, research aiming to unravel the intricacies of tolerance induction is mired by interchangeable and often misused terminologies, with markers and mechanistic pathways that beg the question of redundancy. In this review we aim to define these boarders by providing new perspectives to long-standing theories of tolerance. Given the central role of T cells in enforcing immune cascades, in this review we choose to explore immunological tolerance through the perspective of T cell 'resistance to activation,' to delineate the contexts in which one tolerance mechanism has evolved over the other. By clarifying the important biological markers and cellular players underpinning T cell resistance to activation, we aim to encourage more purposeful and directed research into tolerance and, more-over, potential therapeutic strategies in autoimmune diseases and cancer. The tolerance field is in much need of reclassification and consideration, and in this review, we hope to open that conversation.
Collapse
Affiliation(s)
- Daniel Beckers
- The Kennedy Institute of Rheumatology, Nuffield Department of Orthopaedics Rheumatology & Musculoskeletal Sciences, University of Oxford, Oxford OX3 7FY, United Kingdom
| | - Ashwin K. Jainarayanan
- The Kennedy Institute of Rheumatology, Nuffield Department of Orthopaedics Rheumatology & Musculoskeletal Sciences, University of Oxford, Oxford OX3 7FY, United Kingdom
| | - Michael L. Dustin
- The Kennedy Institute of Rheumatology, Nuffield Department of Orthopaedics Rheumatology & Musculoskeletal Sciences, University of Oxford, Oxford OX3 7FY, United Kingdom
| | - Jesusa Capera
- The Kennedy Institute of Rheumatology, Nuffield Department of Orthopaedics Rheumatology & Musculoskeletal Sciences, University of Oxford, Oxford OX3 7FY, United Kingdom
| |
Collapse
|
9
|
Li W, Qian R, Zhou Z, Wen L, Yin Q, Zhou X, Li X, Cheng J, Zhang X, Zeng X, Wang Z, Huang Y, Wang S, Liao Y, Li Y, Shan S, Zhou M, Wei W, Abdollahi A, August A, Magazine N, Veggiani G, Huang W, Guan D, Zhou C. T cell senescence may contribute to immunothrombosis via Th17 immune transition in COVID-19. Sci Bull (Beijing) 2024; 69:3501-3506. [PMID: 38755088 DOI: 10.1016/j.scib.2024.04.068] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2024] [Revised: 03/07/2024] [Accepted: 04/28/2024] [Indexed: 05/18/2024]
Affiliation(s)
- Wenxing Li
- Department of Radiation Oncology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China; Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Rui Qian
- Department of General Surgery, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Zhaoming Zhou
- Department of Radiation Oncology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China; Department of Radiation Medicine, School of Public Health, Southern Medical University, Guangzhou 510515, China
| | - Lei Wen
- Department of Radiation Oncology, Zhujiang Hospital, Southern Medical University, Guangzhou 510280, China
| | - Quan Yin
- State Key Laboratory of Silkworm Genome Biology, Southwest University, Chongqing 400715, China
| | - Xiang Zhou
- Department of Anesthesiology, General Hospital of Central Theater Command of PLA, Wuhan 430070, China
| | - Xiaowei Li
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Jie Cheng
- Center for Reproductive Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200135, China
| | - Xinlu Zhang
- Department of Cardiology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Xin Zeng
- Department of Radiation Medicine, School of Public Health, Southern Medical University, Guangzhou 510515, China
| | - Zhuoya Wang
- School of Life Science and Biopharmaceutics, Guangdong Pharmaceutical University, Guangzhou 510006, China
| | - Yingying Huang
- CAS Key Laboratory of Computational Biology, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Shengqiang Wang
- Department of Rehabilitation Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Yixing Liao
- Department of Critical Care Medicine, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China
| | - Youjiang Li
- Department of Clinical Laboratory, The Fourth Affiliated Hospital, Zhejiang University School of Medicine, Yiwu 322000, China
| | - Shenbing Shan
- CAS Key Laboratory of Computational Biology, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Meijuan Zhou
- Department of Radiation Medicine, School of Public Health, Southern Medical University, Guangzhou 510515, China
| | - Wu Wei
- CAS Key Laboratory of Computational Biology, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Amir Abdollahi
- Translational Radiation Oncology, German Cancer Research Center (DKFZ) and University Heidelberg School of Medicine, Heidelberg 69120, Germany
| | - Avery August
- Department of Microbiology and Immunology, College of Veterinary Medicine, Cornell University, Ithaca NY 14853, USA
| | - Nicholas Magazine
- Department of Pathobiological Sciences, School of Veterinary Medicine, Louisiana State University, Baton Rouge LA 70803, USA
| | - Gianluca Veggiani
- Department of Pathobiological Sciences, School of Veterinary Medicine, Louisiana State University, Baton Rouge LA 70803, USA
| | - Weishan Huang
- Department of Microbiology and Immunology, College of Veterinary Medicine, Cornell University, Ithaca NY 14853, USA; Department of Pathobiological Sciences, School of Veterinary Medicine, Louisiana State University, Baton Rouge LA 70803, USA.
| | - Daogang Guan
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China.
| | - Cheng Zhou
- Department of Radiation Oncology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China; Translational Radiation Oncology, German Cancer Research Center (DKFZ) and University Heidelberg School of Medicine, Heidelberg 69120, Germany.
| |
Collapse
|
10
|
Chatterjee P, Stevens HY, Kippner LE, Bowles-Welch AC, Drissi H, Mautner K, Yeago C, Gibson G, Roy K. Single-cell transcriptome and crosstalk analysis reveals immune alterations and key pathways in the bone marrow of knee OA patients. iScience 2024; 27:110827. [PMID: 39310769 PMCID: PMC11416684 DOI: 10.1016/j.isci.2024.110827] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2023] [Revised: 06/10/2024] [Accepted: 08/22/2024] [Indexed: 09/25/2024] Open
Abstract
Knee osteoarthritis (OA) is a significant medical and economic burden. To understand systemic immune effects, we performed deep exploration of bone marrow aspirate concentrates (BMACs) from knee-OA patients via single-cell RNA sequencing and proteomic analyses from a randomized clinical trial (MILES: NCT03818737). We found significant cellular and immune alterations in the bone marrow, specifically in MSCs, T cells and NK cells, along with changes in intra-tissue cellular crosstalk during OA progression. Unlike previous studies focusing on injury sites or peripheral blood, our probe into the bone marrow-an inflammation and immune regulation hub-highlights remote organ impact of OA, identifying cell types and pathways for potential therapeutic targeting. Our findings highlight increased cellular senescence and inflammatory pathways, revealing key upstream genes, transcription factors, and ligands. Additionally, we identified significant enrichment in key biological pathways like PI3-AKT-mTOR signaling and IFN responses, showing their potentially crucial role in OA onset and progression.
Collapse
Affiliation(s)
- Paramita Chatterjee
- Marcus Center for Therapeutic Cell Characterization and Manufacturing, The Parker H. Petit Institute for Bioengineering and Biosciences, Georgia Institute of Technology, Atlanta, GA, USA
- The Parker H. Petit Institute for Bioengineering and Biosciences Georgia Institute of Technology, Atlanta, GA, USA
| | - Hazel Y. Stevens
- Marcus Center for Therapeutic Cell Characterization and Manufacturing, The Parker H. Petit Institute for Bioengineering and Biosciences, Georgia Institute of Technology, Atlanta, GA, USA
- The Parker H. Petit Institute for Bioengineering and Biosciences Georgia Institute of Technology, Atlanta, GA, USA
| | - Linda E. Kippner
- Marcus Center for Therapeutic Cell Characterization and Manufacturing, The Parker H. Petit Institute for Bioengineering and Biosciences, Georgia Institute of Technology, Atlanta, GA, USA
- The Parker H. Petit Institute for Bioengineering and Biosciences Georgia Institute of Technology, Atlanta, GA, USA
| | - Annie C. Bowles-Welch
- Marcus Center for Therapeutic Cell Characterization and Manufacturing, The Parker H. Petit Institute for Bioengineering and Biosciences, Georgia Institute of Technology, Atlanta, GA, USA
- The Parker H. Petit Institute for Bioengineering and Biosciences Georgia Institute of Technology, Atlanta, GA, USA
| | - Hicham Drissi
- Department of Orthopaedics, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Kenneth Mautner
- Department of Orthopaedics, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Carolyn Yeago
- The Parker H. Petit Institute for Bioengineering and Biosciences Georgia Institute of Technology, Atlanta, GA, USA
| | - Greg Gibson
- School of Biological Sciences, Georgia Institute of Technology, Atlanta, GA 30332, USA
| | - Krishnendu Roy
- Marcus Center for Therapeutic Cell Characterization and Manufacturing, The Parker H. Petit Institute for Bioengineering and Biosciences, Georgia Institute of Technology, Atlanta, GA, USA
- Department of Biomedical Engineering, School of Engineering, Vanderbilt University, Nashville, TN, USA
- Department of Pathology, Microbiology and Immunology, School of Medicine, Vanderbilt University, Nashville, TN, USA
- Department of Chemical and Biomolecular Engineering, School of Engineering, Vanderbilt University, Nashville, TN, USA
| |
Collapse
|
11
|
Koga HK, Grodstein F, Williams DR, Demeo DL, Kubzansky LD. Relations of optimism and purpose in life to immune markers in aging. J Psychosom Res 2024; 184:111851. [PMID: 38964200 DOI: 10.1016/j.jpsychores.2024.111851] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/11/2024] [Revised: 06/13/2024] [Accepted: 06/21/2024] [Indexed: 07/06/2024]
Abstract
OBJECTIVE Optimism and purpose in life are associated with improved health outcomes. More information is needed on biological mechanisms, including immunosenescence. We investigated if psychological well-being is associated with healthier immunosenescence-related measures including naïve and terminally differentiated CD4+ and CD8+ T cell percentages, CD4+:CD8+, and cytomegalovirus (CMV) IgG response. METHODS Participants were adults over age 50 from the Health and Retirement Study. Optimism was measured using the Life Orientation Test Revised. Purpose in life was assessed using the subscale from the Ryff psychological well-being measure. We examined the cross-sectional associations of optimism and purpose in life with measures of T cell subsets using linear regression and with CMV IgG using ordered logit regression, controlling for potential confounding factors. RESULTS The final analytic sample ranged from 7250 to 7870. After adjusting for sociodemographic factors, a 1-SD increment in optimism was associated with the percentage of naïve CD4+ T cells increasing by 0.6 (95%CI 0.2%, 1.0%). A 1-SD increment in purpose in life was associated with the percentage of naïve CD4+ T cells increasing by 0.9 (95%CI 0.5%, 1.3%) after adjusting for sociodemographic factors and the association was maintained after further adjustments for health conditions, depression, and health behaviors. For naïve CD8+ T cell percentages, CD4:CD8 ratios, and CMV IgG antibodies, associations were seen only in models that adjusted for age. No significant associations were seen in any models for the terminally differentiated CD4+ and CD8+ T cells. CONCLUSIONS We found associations of optimism and purpose in life with naïve CD4+ T cell percentages.
Collapse
Affiliation(s)
- Hayami K Koga
- Department of Social and Behavioral Sciences, Harvard T.H. Chan School of Public Health, Boston, MA, United States of America; Harvard Center for Population and Development Studies, Cambridge, MA, United States of America.
| | - Francine Grodstein
- Rush Alzheimer's Disease Center, Rush University Medical Center, Chicago, IL, United States of America
| | - David R Williams
- Department of Social and Behavioral Sciences, Harvard T.H. Chan School of Public Health, Boston, MA, United States of America; Department of African and African American Studies, Department of Sociology, Harvard University, Cambridge, MA, United States of America
| | - Dawn L Demeo
- Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, United States of America; Channing Division of Network Medicine, Brigham and Women's Hospital, Boston, MA, United States of America
| | - Laura D Kubzansky
- Department of Social and Behavioral Sciences, Harvard T.H. Chan School of Public Health, Boston, MA, United States of America
| |
Collapse
|
12
|
Jiang J, Yang M, Zhu H, Long D, He Z, Liu J, He L, Tan Y, Akbar AN, Reddy V, Zhao M, Long H, Wu H, Lu Q. CD4 +CD57 + senescent T cells as promoters of systemic lupus erythematosus pathogenesis and the therapeutic potential of senolytic BCL-2 inhibitor. Eur J Immunol 2024; 54:e2350603. [PMID: 38752316 DOI: 10.1002/eji.202350603] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2023] [Revised: 03/23/2024] [Accepted: 03/28/2024] [Indexed: 07/07/2024]
Abstract
Systemic lupus erythematosus (SLE) is a complex autoimmune disease characterized by persistent activation of immune cells and overproduction of autoantibodies. The accumulation of senescent T and B cells has been observed in SLE and other immune-mediated diseases. However, the exact mechanistic pathways contributing to this process in SLE remain incompletely understood. In this study, we found that in SLE patients: (1) the frequency of CD4+CD57+ senescent T cells was significantly elevated and positively correlated with disease activity; (2) the expression levels of B-lymphoma-2 (BCL-2) family and interferon-induced genes (ISGs) were significantly upregulated; and (3) in vitro, the cytokine IL-15 stimulation increased the frequency of senescent CD4+ T cells and upregulated the expression of BCL-2 family and ISGs. Further, treatment with ABT-263 (a senolytic BCL-2 inhibitor) in MRL/lpr mice resulted in decreased: (1) frequency of CD4+CD44hiCD62L-PD-1+CD153+ senescent CD4+ T cells; (2) frequency of CD19+CD11c+T-bet+ age-related B cells; (3) level of serum antinuclear antibody; (4) proteinuria; (5) frequency of Tfh cells; and (6) renal histopathological abnormalities. Collectively, these results indicated a dominant role for CD4+CD57+ senescent CD4+ T cells in the pathogenesis of SLE and senolytic BCL-2 inhibitor ABT-263 may be the potential treatment in ameliorating lupus phenotypes.
Collapse
Affiliation(s)
- Jiao Jiang
- Hospital for Skin Diseases, Institute of Dermatology, Chinese Academy of Medical Sciences & Peking Union Medical College, Nanjing, Jiangsu, China
- Department of Dermatology, Hunan Key Laboratory of Medical Epigenomics, The Second Xiangya Hospital of Central South University, Changsha, Hunan, China
- Key Laboratory of Basic and Translational Research on Immune-Mediated Skin Diseases, Chinese Academy of Medical Sciences, Nanjing, Jiang Su, China
- Jiangsu Key Laboratory of Molecular Biology for Skin Diseases and STIs, Nanjing, Jiang Su, China
| | - Ming Yang
- Department of Dermatology, Hunan Key Laboratory of Medical Epigenomics, The Second Xiangya Hospital of Central South University, Changsha, Hunan, China
| | - Huan Zhu
- Department of Dermatology, Hunan Key Laboratory of Medical Epigenomics, The Second Xiangya Hospital of Central South University, Changsha, Hunan, China
| | - Di Long
- Department of Dermatology, Hunan Key Laboratory of Medical Epigenomics, The Second Xiangya Hospital of Central South University, Changsha, Hunan, China
| | - Zhenghao He
- Department of Dermatology, Hunan Key Laboratory of Medical Epigenomics, The Second Xiangya Hospital of Central South University, Changsha, Hunan, China
| | - Juan Liu
- Department of Dermatology, Hunan Key Laboratory of Medical Epigenomics, The Second Xiangya Hospital of Central South University, Changsha, Hunan, China
| | - Liting He
- Department of Dermatology, Hunan Key Laboratory of Medical Epigenomics, The Second Xiangya Hospital of Central South University, Changsha, Hunan, China
| | - Yixin Tan
- Department of Dermatology, Hunan Key Laboratory of Medical Epigenomics, The Second Xiangya Hospital of Central South University, Changsha, Hunan, China
| | - Arne N Akbar
- Division of Medicine, University College London, London, United Kingdom
| | - Venkat Reddy
- Division of Medicine, University College London, London, United Kingdom
| | - Ming Zhao
- Hospital for Skin Diseases, Institute of Dermatology, Chinese Academy of Medical Sciences & Peking Union Medical College, Nanjing, Jiangsu, China
- Department of Dermatology, Hunan Key Laboratory of Medical Epigenomics, The Second Xiangya Hospital of Central South University, Changsha, Hunan, China
- Key Laboratory of Basic and Translational Research on Immune-Mediated Skin Diseases, Chinese Academy of Medical Sciences, Nanjing, Jiang Su, China
- Jiangsu Key Laboratory of Molecular Biology for Skin Diseases and STIs, Nanjing, Jiang Su, China
| | - Hai Long
- Department of Dermatology, Hunan Key Laboratory of Medical Epigenomics, The Second Xiangya Hospital of Central South University, Changsha, Hunan, China
| | - Haijing Wu
- Department of Dermatology, Hunan Key Laboratory of Medical Epigenomics, The Second Xiangya Hospital of Central South University, Changsha, Hunan, China
| | - Qianjin Lu
- Hospital for Skin Diseases, Institute of Dermatology, Chinese Academy of Medical Sciences & Peking Union Medical College, Nanjing, Jiangsu, China
- Department of Dermatology, Hunan Key Laboratory of Medical Epigenomics, The Second Xiangya Hospital of Central South University, Changsha, Hunan, China
- Key Laboratory of Basic and Translational Research on Immune-Mediated Skin Diseases, Chinese Academy of Medical Sciences, Nanjing, Jiang Su, China
- Jiangsu Key Laboratory of Molecular Biology for Skin Diseases and STIs, Nanjing, Jiang Su, China
| |
Collapse
|
13
|
Andreu-Sánchez S, Ripoll-Cladellas A, Culinscaia A, Bulut O, Bourgonje AR, Netea MG, Lansdorp P, Aubert G, Bonder MJ, Franke L, Vogl T, van der Wijst MG, Melé M, Van Baarle D, Fu J, Zhernakova A. Antibody signatures against viruses and microbiome reflect past and chronic exposures and associate with aging and inflammation. iScience 2024; 27:109981. [PMID: 38868191 PMCID: PMC11167443 DOI: 10.1016/j.isci.2024.109981] [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: 02/16/2024] [Revised: 04/16/2024] [Accepted: 05/13/2024] [Indexed: 06/14/2024] Open
Abstract
Encounters with pathogens and other molecules can imprint long-lasting effects on our immune system, influencing future physiological outcomes. Given the wide range of microbes to which humans are exposed, their collective impact on health is not fully understood. To explore relations between exposures and biological aging and inflammation, we profiled an antibody-binding repertoire against 2,815 microbial, viral, and environmental peptides in a population cohort of 1,443 participants. Utilizing antibody-binding as a proxy for past exposures, we investigated their impact on biological aging, cell composition, and inflammation. Immune response against cytomegalovirus (CMV), rhinovirus, and gut bacteria relates with telomere length. Single-cell expression measurements identified an effect of CMV infection on the transcriptional landscape of subpopulations of CD8 and CD4 T-cells. This examination of the relationship between microbial exposures and biological aging and inflammation highlights a role for chronic infections (CMV and Epstein-Barr virus) and common pathogens (rhinoviruses and adenovirus C).
Collapse
Affiliation(s)
- Sergio Andreu-Sánchez
- Department of Genetics, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
- Department of Pediatrics, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - Aida Ripoll-Cladellas
- Life Sciences Department, Barcelona Supercomputing Center, 08034 Barcelona, Catalonia, Spain
| | - Anna Culinscaia
- Department of Genetics, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - Ozlem Bulut
- Department of Internal Medicine and Radboud Center for Infectious Diseases (RCI), Radboudumc, Nijmegen, the Netherlands
| | - Arno R. Bourgonje
- Department of Gastroenterology and Hepatology, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
- The Henry D. Janowitz Division of Gastroenterology, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Mihai G. Netea
- Department of Internal Medicine and Radboud Center for Infectious Diseases (RCI), Radboudumc, Nijmegen, the Netherlands
- Department for Immunology & Metabolism, Life and Medical Sciences Institute (LIMES), University of Bonn, Bonn, Germany
| | - Peter Lansdorp
- Terry Fox Laboratory, British Columbia Cancer Research Center, Vancouver, BC, Canada
- Departments of Hematology and Medical Genetics, University of British Columbia, Vancouver, BC, Canada
| | - Geraldine Aubert
- Terry Fox Laboratory, British Columbia Cancer Research Center, Vancouver, BC, Canada
- Repeat Diagnostics Inc, Vancouver, BC, Canada
| | - Marc Jan Bonder
- Department of Genetics, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - Lude Franke
- Department of Genetics, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - Thomas Vogl
- Center for Cancer Research, Medical University of Vienna, Wien, Austria
| | - Monique G.P. van der Wijst
- Department of Genetics, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - Marta Melé
- Life Sciences Department, Barcelona Supercomputing Center, 08034 Barcelona, Catalonia, Spain
| | - Debbie Van Baarle
- Department of Medical Microbiology and Infection Prevention, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Jingyuan Fu
- Department of Genetics, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
- Department of Pediatrics, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - Alexandra Zhernakova
- Department of Genetics, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| |
Collapse
|
14
|
Mosebarger A, Vidal MS, Bento GFC, Lintao RCV, Severino MEL, Kumar Kammala A, Menon R. Immune cells at the feto-maternal interface: Comprehensive characterization and insights into term labor. J Reprod Immunol 2024; 163:104239. [PMID: 38493591 DOI: 10.1016/j.jri.2024.104239] [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: 11/05/2023] [Revised: 02/05/2024] [Accepted: 03/07/2024] [Indexed: 03/19/2024]
Abstract
Immune cells at the feto-maternal interface play an important role in pregnancy; starting at implantation, maintenance of pregnancy, and parturition. The role of decidual immune cells in induction of labor still needs to be understood. Published reports on this topic show heterogeneity in methods of cell isolation, assay, analysis and cellular characterization making it difficult to collate available information in order to understand the contribution of immune cells at term leading to parturition. In the present study, available literature was reviewed to study the differences in immune cells between the decidua basalis and decidua parietalis, as well as between immune cells in term and preterm labor. Additionally, immune cells at the decidua parietalis were isolated from term not in labor (TNL) or term in labor (TL) samples and characterized via flow cytometry using a comprehensive, high-dimensional antibody panel. This allowed a full view of immune cell differences without combining multiple studies, which must include variation in isolation and analysis methods, for more conclusive data. The ratio of cells found in decidua parietalis in this study generally matched those reported in the literature, although we report a lower percentage of natural killer (NK) cells at term. We report that CD4 expression on CD8- NK cells decreased in term labor compared to not in labor samples, suggesting that natural killer cells may be migrating to other sites during labor. Also, we report a decrease in CD38 expression on CD8+ CD57+ T cells in labor, indicative of cytotoxic T cell senescence. Our study provides a comprehensive status of immune cells at the decidua-chorion interface at term.
Collapse
Affiliation(s)
- Angela Mosebarger
- Division of Basic and Translational Research, Department of Obstetrics and Gynecology, University of Texas Medical Branch at Galveston, Galveston, TX, USA
| | - Manuel S Vidal
- Division of Basic and Translational Research, Department of Obstetrics and Gynecology, University of Texas Medical Branch at Galveston, Galveston, TX, USA; Department of Biochemistry and Molecular Biology, College of Medicine, University of Philippines Manila, Manila, Philippines
| | | | - Ryan C V Lintao
- Division of Basic and Translational Research, Department of Obstetrics and Gynecology, University of Texas Medical Branch at Galveston, Galveston, TX, USA; Department of Biochemistry and Molecular Biology, College of Medicine, University of Philippines Manila, Manila, Philippines
| | - Mary Elise L Severino
- Division of Basic and Translational Research, Department of Obstetrics and Gynecology, University of Texas Medical Branch at Galveston, Galveston, TX, USA; Department of Biochemistry and Molecular Biology, College of Medicine, University of Philippines Manila, Manila, Philippines
| | - Ananth Kumar Kammala
- Division of Basic and Translational Research, Department of Obstetrics and Gynecology, University of Texas Medical Branch at Galveston, Galveston, TX, USA
| | - Ramkumar Menon
- Division of Basic and Translational Research, Department of Obstetrics and Gynecology, University of Texas Medical Branch at Galveston, Galveston, TX, USA.
| |
Collapse
|
15
|
Zhu L, Zhang L, Qi J, Ye Z, Nie G, Leng S. Machine learning-derived immunosenescence index for predicting outcome and drug sensitivity in patients with skin cutaneous melanoma. Genes Immun 2024; 25:219-231. [PMID: 38811681 DOI: 10.1038/s41435-024-00278-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2024] [Revised: 05/16/2024] [Accepted: 05/21/2024] [Indexed: 05/31/2024]
Abstract
The functions of immunosenescence are closely related to skin cutaneous melanoma (SKCM). The aim of this study is to uncover the characteristics of immunosenescence index (ISI) to identify novel biomarkers and potential targets for treatment. Firstly, integrated bioinformatics analysis was carried out to identify risk prognostic genes, and their expression and prognostic value were evaluated. Then, we used the computational algorithm to estimate ISI. Finally, the distribution characteristics and clinical significance of ISI in SKCM by using multi-omics analysis. Patients with a lower ISI had a favorable survival rate, lower chromosomal instability, lower somatic copy-number alterations, lower somatic mutations, higher immune infiltration, and sensitive to immunotherapy. The ISI exhibited robust, which was validated in multiple datasets. Besides, the ISI is more effective than other published signatures in predicting survival outcomes for patients with SKCM. Single-cell analysis revealed higher ISI was specifically expressed in monocytes, and correlates with the differentiation fate of monocytes in SKCM. Besides, individuals exhibiting elevated ISI levels could potentially receive advantages from chemotherapy, and promising compounds with the potential to target high ISI were recognized. The ISI model is a valuable tool in categorizing SKCM patients based on their prognosis, gene mutation signatures, and response to immunotherapy.
Collapse
Affiliation(s)
- Linyu Zhu
- Department of Dermatovenereology, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, China
| | - Lvya Zhang
- Traditional Chinese Medicine department, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, 518107, Guangdong, China
| | - Junhua Qi
- Research Center, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, China
- Department of Clinical Laboratory, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, China
| | - Zhiyu Ye
- Traditional Chinese Medicine department, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, 518107, Guangdong, China.
| | - Gang Nie
- Department of Dermatovenereology, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, China.
| | - Shaolong Leng
- Department of Dermatovenereology, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, China.
| |
Collapse
|
16
|
Hambright WS, Duke VR, Goff AD, Goff AW, Minas LT, Kloser H, Gao X, Huard C, Guo P, Lu A, Mitchell J, Mullen M, Su C, Tchkonia T, Espindola Netto JM, Robbins PD, Niedernhofer LJ, Kirkland JL, Bahney CS, Philippon M, Huard J. Clinical validation of C 12FDG as a marker associated with senescence and osteoarthritic phenotypes. Aging Cell 2024; 23:e14113. [PMID: 38708778 PMCID: PMC11113632 DOI: 10.1111/acel.14113] [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: 05/01/2023] [Revised: 01/23/2024] [Accepted: 01/29/2024] [Indexed: 05/07/2024] Open
Abstract
Chronic conditions associated with aging have proven difficult to prevent or treat. Senescence is a cell fate defined by loss of proliferative capacity and the development of a pro-inflammatory senescence-associated secretory phenotype comprised of cytokines/chemokines, proteases, and other factors that promotes age-related diseases. Specifically, an increase in senescent peripheral blood mononuclear cells (PBMCs), including T cells, is associated with conditions like frailty, rheumatoid arthritis, and bone loss. However, it is unknown if the percentage of senescent PBMCs associated with age-associated orthopedic decline could be used for potential diagnostic or prognostic use in orthopedics. Here, we report senescent cell detection using the fluorescent compound C12FDG to quantify PBMCs senescence across a large cohort of healthy and osteoarthritic patients. There is an increase in the percent of circulating C12FDG+ PBMCs that is commensurate with increases in age and senescence-related serum biomarkers. Interestingly, C12FDG+ PBMCs and T cells also were found to be elevated in patients with mild to moderate osteoarthritis, a progressive joint disease that is strongly associated with inflammation. The percent of C12FDG+ PBMCs and age-related serum biomarkers were decreased in a small subgroup of study participants taking the senolytic drug fisetin. These results demonstrate quantifiable measurements in a large group of participants that could create a composite score of healthy aging sensitive enough to detect changes following senolytic therapy and may predict age-related orthopedic decline. Detection of peripheral senescence in PBMCs and subsets using C12FDG may be clinically useful for quantifying cellular senescence and determining how and if it plays a pathological role in osteoarthritic progression.
Collapse
Affiliation(s)
- William S. Hambright
- Center for Regenerative Sports MedicineSteadman Philippon Research InstituteVailColoradoUSA
| | - Victoria R. Duke
- Center for Regenerative Sports MedicineSteadman Philippon Research InstituteVailColoradoUSA
| | - Adam D. Goff
- Center for Regenerative Sports MedicineSteadman Philippon Research InstituteVailColoradoUSA
| | - Alex W. Goff
- Center for Regenerative Sports MedicineSteadman Philippon Research InstituteVailColoradoUSA
| | - Lucas T. Minas
- Center for Regenerative Sports MedicineSteadman Philippon Research InstituteVailColoradoUSA
| | - Heidi Kloser
- Center for Regenerative Sports MedicineSteadman Philippon Research InstituteVailColoradoUSA
| | - Xueqin Gao
- Center for Regenerative Sports MedicineSteadman Philippon Research InstituteVailColoradoUSA
| | - Charles Huard
- Center for Regenerative Sports MedicineSteadman Philippon Research InstituteVailColoradoUSA
| | - Ping Guo
- Center for Regenerative Sports MedicineSteadman Philippon Research InstituteVailColoradoUSA
| | - Aiping Lu
- Center for Regenerative Sports MedicineSteadman Philippon Research InstituteVailColoradoUSA
| | - John Mitchell
- Center for Regenerative Sports MedicineSteadman Philippon Research InstituteVailColoradoUSA
| | - Michael Mullen
- Center for Regenerative Sports MedicineSteadman Philippon Research InstituteVailColoradoUSA
| | - Charles Su
- Center for Regenerative Sports MedicineSteadman Philippon Research InstituteVailColoradoUSA
| | - Tamara Tchkonia
- Department of Physiology and Biomedical EngineeringMayo ClinicRochesterMinnesotaUSA
| | | | - Paul D. Robbins
- Department of Biochemistry and Molecular Biology and Biophysics, Institute on the Biology of Aging and MetabolismUniversity of MinnesotaMinneapolisMinnesotaUSA
| | - Laura J. Niedernhofer
- Department of Biochemistry and Molecular Biology and Biophysics, Institute on the Biology of Aging and MetabolismUniversity of MinnesotaMinneapolisMinnesotaUSA
| | - James L. Kirkland
- Department of Physiology and Biomedical EngineeringMayo ClinicRochesterMinnesotaUSA
- Division of General Internal Medicine, Department of MedicineMayo ClinicRochesterMinnesotaUSA
| | - Chelsea S. Bahney
- Center for Regenerative Sports MedicineSteadman Philippon Research InstituteVailColoradoUSA
- Orthopaedic Trauma InstituteUniversity of California San FranciscoSan FranciscoCaliforniaUSA
| | - Marc Philippon
- Center for Regenerative Sports MedicineSteadman Philippon Research InstituteVailColoradoUSA
- The Steadman ClinicVailColoradoUSA
| | - Johnny Huard
- Center for Regenerative Sports MedicineSteadman Philippon Research InstituteVailColoradoUSA
| |
Collapse
|
17
|
Giovenzana A, Bezzecchi E, Bichisecchi A, Cardellini S, Ragogna F, Pedica F, Invernizzi F, Di Filippo L, Tomajer V, Aleotti F, Scotti GM, Socci C, Cesana G, Olmi S, Morelli MJ, Falconi M, Giustina A, Bonini C, Piemonti L, Ruggiero E, Petrelli A. Fat-to-blood recirculation of partially dysfunctional PD-1 +CD4 Tconv cells is associated with dysglycemia in human obesity. iScience 2024; 27:109032. [PMID: 38380252 PMCID: PMC10877684 DOI: 10.1016/j.isci.2024.109032] [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: 08/09/2023] [Revised: 01/03/2024] [Accepted: 01/23/2024] [Indexed: 02/22/2024] Open
Abstract
Obesity is characterized by the accumulation of T cells in insulin-sensitive tissues, including the visceral adipose tissue (VAT), that can interfere with the insulin signaling pathway eventually leading to insulin resistance (IR) and type 2 diabetes. Here, we found that PD-1+CD4 conventional T (Tconv) cells, endowed with a transcriptomic and functional profile of partially dysfunctional cells, are diminished in VAT of obese patients with dysglycemia (OB-Dys), without a concomitant increase in apoptosis. These cells showed enhanced capacity to recirculate into the bloodstream and had a non-restricted TCRβ repertoire divergent from that of normoglycemic obese and lean individuals. PD-1+CD4 Tconv were reduced in the circulation of OB-Dys, exhibited an altered migration potential, and were detected in the liver of patients with non-alcoholic steatohepatitis. The findings suggest a potential role for partially dysfunctional PD-1+CD4 Tconv cells as inter-organ mediators of IR in obese patients with dysglycemic.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | | | | | - Stefano Olmi
- San Marco Hospital GSD, Zingonia, Bergamo, Italy
- Università Vita-Salute San Raffaele, Milan, Italy
| | | | - Massimo Falconi
- IRCCS Ospedale San Raffaele, Milan, Italy
- Università Vita-Salute San Raffaele, Milan, Italy
| | - Andrea Giustina
- IRCCS Ospedale San Raffaele, Milan, Italy
- Università Vita-Salute San Raffaele, Milan, Italy
| | - Chiara Bonini
- IRCCS Ospedale San Raffaele, Milan, Italy
- Università Vita-Salute San Raffaele, Milan, Italy
| | - Lorenzo Piemonti
- IRCCS Ospedale San Raffaele, Milan, Italy
- Università Vita-Salute San Raffaele, Milan, Italy
| | | | | |
Collapse
|
18
|
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
|
19
|
Parisi S, Andrea B, Chiara DM, Lo Gullo A, Maddalena L, Palma S, Olga A, Massimo R, Paroli M, Rosalba C, Elisa V, Rosario F, Giorgio A, Francesco DL, Ylenia DB, Roberta F, Antonella F, Francesco G, Simone B, Dario C, Gerolamo B, Matteo C, Romina A, Natalia M, Giulio F, Patrizia DM, Aldo MC, Veronica F, Francesco MC, Federica L, Gilda S, Carlo S, Marta P, Aurora I, Valeria N, Daniele S, Gianluca L, Adorni G, Eleonora DD, Elena B, Ilaria P, Eugenio A, Alessandra B, Cristina FM, Fabio M, Vincenzo B, Viviana R, Alessia F, Guido R, Rosetta V, Antonio M, Alessandro V, Francesca O, Alarico A, Enrico F. Analysis of survival rate and persistence predictors of baricitinib in real-world data from a large cohort of rheumatoid arthritis patients. CURRENT RESEARCH IN PHARMACOLOGY AND DRUG DISCOVERY 2024; 6:100178. [PMID: 38419949 PMCID: PMC10899020 DOI: 10.1016/j.crphar.2024.100178] [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: 10/31/2023] [Revised: 01/23/2024] [Accepted: 02/12/2024] [Indexed: 03/02/2024] Open
Abstract
Objectives The persistence in therapy of rheumatoid arthritis drugs and particularly bDMARD is a limiting factor for their long-term use. The randomized controlled trials (RCTs) may not reflect real-world contexts due to strict inclusion and exclusion criteria. Baricitinib, which targets both JAK1 and JAK2, has been used in Italy for several years. The aim of this multi-center study is to assess the real world persistence on therapy of baricitinib in RA patients and to identify predictive factors of baricitinib's survival rate. Methods This is a retrospective, multicentric, Italian, longitudinal study. All patients were enrolled according to the following criteria: a) age ≥ 18 years old; b) diagnosed with RA according 2010 ACR/EULAR classification criteria; c) treated with baricitinib. In order to describe baricitinib clinical efficacy, the survival rate was evaluated by The Kaplan-Meier curve. Then, predictive factors of drug retention rate were assessed by performing the Cox analysis, identifying which risk factors influenced treatment persistence. Results Overall, we included 478 patients treated with baricitinib. Among them, 380 (79.5%) were females. Baricitinib's survival rate was 94.6% at 6 months, 87.9% at 12 months, 81.7% at 24 months and 53.4% at 48 months. The Cox analysis regression showed that a higher bDMARDs/tsDMARD line of therapy seems to be a negative prognostic factor for the drug retention rate (HR 1.26 CI 95% 1.07-1.49, p = 0.006. Conclusion Real-life study confirms baricitinib effectiveness up to 4 years, but previous treatment with bDMARDs was a negative prognostic factor for its survival rate.
Collapse
Affiliation(s)
- Simone Parisi
- Azienda Ospedaliero Universitaria Città della Salute e della Scienza di Torino, Rheumatology Unit, Turin, Italy
| | - Becciolini Andrea
- Azienda Ospedaliero-Universitaria di Parma, Department of Medicine, Internal Medicine and Rheumatology Unit, Parma, Italy
| | - Ditto Maria Chiara
- Azienda Ospedaliero Universitaria Città della Salute e della Scienza di Torino, Rheumatology Unit, Turin, Italy
| | - Alberto Lo Gullo
- Azienda Ospedaliera di Rilievo Nazionale e di Alta Specializzazione Garibaldi Ospedale Garibaldi-Nesima, Rheumatology Unit, Catania, Italy
| | - Larosa Maddalena
- Azienda Sanitaria Locale 3 Genovese, Division of Rheumatology - Medical Specialties Department, Genoa, Italy
| | - Scolieri Palma
- Ospedale Nuovo Regina Margherita, Internal Medicine and Rheumatology Unit, Rome, Italy
| | - Addimanda Olga
- IRCCS Azienda Ospedaliero-Universitaria di Bologna Policlinico S Orsola, Department of Internal Medicine-Rheumatology, Bologna, Italy
| | - Reta Massimo
- IRCCS Azienda Ospedaliero-Universitaria di Bologna Policlinico S Orsola, Department of Internal Medicine-Rheumatology, Bologna, Italy
| | - Marino Paroli
- University of Rome La Sapienza, Department of Clinical, Anesthesiological and Cardiovascular Sciences, Polo Pontino, Latina, Italy
| | - Caccavale Rosalba
- University of Rome La Sapienza, Department of Clinical, Anesthesiological and Cardiovascular Sciences, Polo Pontino, Latina, Italy
| | - Visalli Elisa
- Azienda Ospedaliero Universitaria Policlinico Vittorio Emanuele Catania, Division of Rheumatology, A.O.U. “Policlinico San Marco”, Catania, Italy
| | - Foti Rosario
- Azienda Ospedaliero Universitaria Policlinico Vittorio Emanuele Catania, Division of Rheumatology, A.O.U. “Policlinico San Marco”, Catania, Italy
| | - Amato Giorgio
- Azienda Ospedaliero Universitaria Policlinico Vittorio Emanuele Catania, Division of Rheumatology, A.O.U. “Policlinico San Marco”, Catania, Italy
| | - De Lucia Francesco
- Azienda Ospedaliero Universitaria Policlinico Vittorio Emanuele Catania, Division of Rheumatology, A.O.U. “Policlinico San Marco”, Catania, Italy
| | - Dal Bosco Ylenia
- Azienda Ospedaliero Universitaria Policlinico Vittorio Emanuele Catania, Division of Rheumatology, A.O.U. “Policlinico San Marco”, Catania, Italy
| | - Foti Roberta
- Azienda Ospedaliero Universitaria Policlinico Vittorio Emanuele Catania, Division of Rheumatology, A.O.U. “Policlinico San Marco”, Catania, Italy
| | - Farina Antonella
- ASUR Area Vasta 4 Fermo, Ospedale A Murri, Internal Medicine Unit, Rheumatology outpatient clinic, Fermo, Italy
| | | | - Bernardi Simone
- Morgagni-Pierantoni Hospital, Rheumatology Unit, Forlì, Italy
| | - Camellino Dario
- Azienda Sanitaria Locale 3 Genovese, Division of Rheumatology - Medical Specialties Department, Genoa, Italy
| | - Bianchi Gerolamo
- Azienda Sanitaria Locale 3 Genovese, Division of Rheumatology - Medical Specialties Department, Genoa, Italy
| | - Colina Matteo
- Azienda USL di Imola, Department of Internal Medicine and Oncology. Service of Rheumatology, Imola, Italy
- University of Bologna, Department of Biomedical and Neuromotor Sciences, Imola, Italy
| | - Andracco Romina
- Hospital Santa Corona Pietra Ligure, Internal Medicine Unit, Rheumatology outpatient clinic, Unit of Diagnostic and Interventional Radiology, Pietra Ligure, Italy
| | - Mansueto Natalia
- Hospital Santa Corona Pietra Ligure, Internal Medicine Unit, Rheumatology outpatient clinic, Unit of Diagnostic and Interventional Radiology, Pietra Ligure, Italy
| | - Ferrero Giulio
- Hospital Santa Corona Pietra Ligure, Internal Medicine Unit, Rheumatology outpatient clinic, Unit of Diagnostic and Interventional Radiology, Pietra Ligure, Italy
| | - Del Medico Patrizia
- Civitanova Marche Hospital, Rheumatology outpatient clinic, Internal Medicine Unit, Civitanova Marche, Italy
| | | | | | | | | | - Sandri Gilda
- University of Modena and Reggio Emilia, Rheumatology Unit, Modena and Reggio Emilia, Italy
| | - Salvarani Carlo
- University of Modena and Reggio Emilia, Rheumatology Unit, Modena and Reggio Emilia, Italy
| | - Priora Marta
- ASL 15 Cuneo, Rheumatology Day Hospital and outpatient clinic, Mondovì, Italy
| | | | - Nucera Valeria
- ASL 13 Novara, Rheumatology Outpatient Unit, Novara, Italy
| | - Santilli Daniele
- Azienda Ospedaliero-Universitaria di Parma, Department of Medicine, Internal Medicine and Rheumatology Unit, Parma, Italy
| | - Lucchini Gianluca
- Azienda Ospedaliero-Universitaria di Parma, Department of Medicine, Internal Medicine and Rheumatology Unit, Parma, Italy
| | - Giuditta Adorni
- Azienda Ospedaliero-Universitaria di Parma, Department of Medicine, Internal Medicine and Rheumatology Unit, Parma, Italy
| | - Di Donato Eleonora
- Azienda Ospedaliero-Universitaria di Parma, Department of Medicine, Internal Medicine and Rheumatology Unit, Parma, Italy
| | - Bravi Elena
- Guglielmo da Saliceto Hospital, Department of Medicine, Internal Medicine and Rheumatology Unit, Piacenza, Italy
| | - Platè Ilaria
- Guglielmo da Saliceto Hospital, Department of Medicine, Internal Medicine and Rheumatology Unit, Piacenza, Italy
| | - Arrigoni Eugenio
- Guglielmo da Saliceto Hospital, Department of Medicine, Internal Medicine and Rheumatology Unit, Piacenza, Italy
| | - Bezzi Alessandra
- ASL 13 Rimini, Internal Medicine and Rheumatology Unit, Rimini, Italy
| | | | - Mascella Fabio
- ASL 13 Rimini, Internal Medicine and Rheumatology Unit, Rimini, Italy
| | - Bruzzese Vincenzo
- Ospedale Nuovo Regina Margherita, Internal Medicine and Rheumatology Unit, Rome, Italy
| | | | | | - Rovera Guido
- PO S Andrea di Vercelli, Unit of Rheumatology, Vercelli, Italy
| | - Vitetta Rosetta
- PO S Andrea di Vercelli, Unit of Rheumatology, Vercelli, Italy
| | - Marchetta Antonio
- IRCCS Ospedale Sacro Cuore Don Calabria, Rheumatology Unit, Negrar, Italy
| | - Volpe Alessandro
- IRCCS Ospedale Sacro Cuore Don Calabria, Rheumatology Unit, Negrar, Italy
| | - Ometto Francesca
- Azienda ULSS 6 Euganea, Rheumatology Outpatient Clinic, Padova, Italy
| | - Ariani Alarico
- Azienda Ospedaliero-Universitaria di Parma, Department of Medicine, Internal Medicine and Rheumatology Unit, Parma, Italy
| | - Fusaro Enrico
- Azienda Ospedaliero Universitaria Città della Salute e della Scienza di Torino, Rheumatology Unit, Turin, Italy
| |
Collapse
|
20
|
Endo T, Kobayashi K, Matsumura T, Emori C, Ozawa M, Kawamoto S, Okuzaki D, Shimada K, Miyata H, Shimada K, Kodani M, Ishikawa-Yamauchi Y, Motooka D, Hara E, Ikawa M. Multiple ageing effects on testicular/epididymal germ cells lead to decreased male fertility in mice. Commun Biol 2024; 7:16. [PMID: 38177279 PMCID: PMC10766604 DOI: 10.1038/s42003-023-05685-2] [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: 03/02/2023] [Accepted: 12/07/2023] [Indexed: 01/06/2024] Open
Abstract
In mammals, females undergo reproductive cessation with age, whereas male fertility gradually declines but persists almost throughout life. However, the detailed effects of ageing on germ cells during and after spermatogenesis, in the testis and epididymis, respectively, remain unclear. Here we comprehensively examined the in vivo male fertility and the overall organization of the testis and epididymis with age, focusing on spermatogenesis, and sperm function and fertility, in mice. We first found that in vivo male fertility decreased with age, which is independent of mating behaviors and testosterone levels. Second, overall sperm production in aged testes was decreased; about 20% of seminiferous tubules showed abnormalities such as germ cell depletion, sperm release failure, and perturbed germ cell associations, and the remaining 80% of tubules contained lower number of germ cells because of decreased proliferation of spermatogonia. Further, the spermatozoa in aged epididymides exhibited decreased total cell numbers, abnormal morphology/structure, decreased motility, and DNA damage, resulting in low fertilizing and developmental rates. We conclude that these multiple ageing effects on germ cells lead to decreased in vivo male fertility. Our present findings are useful to better understand the basic mechanism behind the ageing effect on male fertility in mammals including humans.
Collapse
Affiliation(s)
- Tsutomu Endo
- Immunology Frontier Research Center, Osaka University, Osaka, Japan.
- Research Institute for Microbial Diseases, Osaka University, Osaka, Japan.
- Department of Experimental Animal Model for Human Disease, Center for Experimental Animals, Tokyo Medical and Dental University, Tokyo, Japan.
- Graduate School of Agricultural and Life Sciences, The University of Tokyo, Tokyo, Japan.
| | - Kiyonori Kobayashi
- Research Institute for Microbial Diseases, Osaka University, Osaka, Japan
- Graduate School of Frontier Biosciences, Osaka University, Osaka, Japan
| | - Takafumi Matsumura
- Research Institute for Microbial Diseases, Osaka University, Osaka, Japan
- Graduate School of Pharmaceutical Sciences, Osaka University, Osaka, Japan
| | - Chihiro Emori
- Research Institute for Microbial Diseases, Osaka University, Osaka, Japan
| | - Manabu Ozawa
- The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Shimpei Kawamoto
- Research Institute for Microbial Diseases, Osaka University, Osaka, Japan
| | - Daisuke Okuzaki
- Research Institute for Microbial Diseases, Osaka University, Osaka, Japan
| | - Keisuke Shimada
- Research Institute for Microbial Diseases, Osaka University, Osaka, Japan
| | - Haruhiko Miyata
- Research Institute for Microbial Diseases, Osaka University, Osaka, Japan
| | - Kentaro Shimada
- Research Institute for Microbial Diseases, Osaka University, Osaka, Japan
- Graduate School of Pharmaceutical Sciences, Osaka University, Osaka, Japan
| | - Mayo Kodani
- Research Institute for Microbial Diseases, Osaka University, Osaka, Japan
- Graduate School of Pharmaceutical Sciences, Osaka University, Osaka, Japan
| | - Yu Ishikawa-Yamauchi
- The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
- Department of Regenerative Medicine, Yokohama City University Graduate School of Medicine, Kanagawa, Japan
| | - Daisuke Motooka
- Research Institute for Microbial Diseases, Osaka University, Osaka, Japan
| | - Eiji Hara
- Immunology Frontier Research Center, Osaka University, Osaka, Japan
- Research Institute for Microbial Diseases, Osaka University, Osaka, Japan
- Graduate School of Frontier Biosciences, Osaka University, Osaka, Japan
- Graduate School of Medicine, Osaka University, Osaka, Japan
| | - Masahito Ikawa
- Immunology Frontier Research Center, Osaka University, Osaka, Japan.
- Research Institute for Microbial Diseases, Osaka University, Osaka, Japan.
- Graduate School of Pharmaceutical Sciences, Osaka University, Osaka, Japan.
- The Institute of Medical Science, The University of Tokyo, Tokyo, Japan.
- Graduate School of Medicine, Osaka University, Osaka, Japan.
| |
Collapse
|
21
|
Haro S, Gomez-Lahoz AM, Monserrat J, Atienza-Pérez M, Fraile-Martinez O, Ortega MA, García-Montero C, Díaz D, Lopez-Dolado E, Álvarez-Mon M. Patients with Chronic Spinal Cord Injury Display a Progressive Alteration over the Years of the Activation Stages of the T Lymphocyte Compartment. Int J Mol Sci 2023; 24:17596. [PMID: 38139422 PMCID: PMC10744286 DOI: 10.3390/ijms242417596] [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: 11/13/2023] [Revised: 12/12/2023] [Accepted: 12/16/2023] [Indexed: 12/24/2023] Open
Abstract
Spinal cord injury (SCI) is a serious medical condition associated with severe morbidities and disability. Chronic SCI patients present an enhanced susceptibility to infections and comorbidities with inflammatory pathogenesis. Chronic SCI appears to be associated with a systemic dysfunction of the immune system. We investigated the alteration of the pivotal CD4+ and CD8+ T lymphocytes in patients with chronic SCI at different years of evolution. A clinically homogenous population of 105 patients with chronic SCI (31 with time of evolution less than 5 years (SCI SP); 32 early chronic (SCI ECP) with time of evolution between 5 and 15 years; and 42 late chronic (SCI LCP) with time of evolution more than 15 years) and 38 healthy controls were enrolled. SCI ECP and SCI LCP patients showed significant CD4+ and CD8+ T lymphopenia, ascribed to a reduction in naïve and CM subsets. Furthermore, SCI ECP and SCI LCP patients showed a significant reduction in the expression of CD28 on CD8+ T lymphocytes. The expression of CCR6 by CD4+ T lymphocytes was decreased during the evolution of chronic SCI, but on CD8+ T lymphocytes, it was observed during the first 15 years of evolution. In conclusion, the chronic SCI course with severe damage to T lymphocytes mainly worsens over the years of disease evolution.
Collapse
Affiliation(s)
- Sergio Haro
- Department of Medicine and Medical Specialities, University of Alcala, Crta N-II km 33.6, 28871 Alcala de Henares, Spain; (S.H.); (A.M.G.-L.); (J.M.); (O.F.-M.); (M.A.O.); (C.G.-M.); (D.D.); (E.L.-D.)
- Ramón y Cajal Institute of Sanitary Research (IRYCIS), 28034 Madrid, Spain
| | - Ana M. Gomez-Lahoz
- Department of Medicine and Medical Specialities, University of Alcala, Crta N-II km 33.6, 28871 Alcala de Henares, Spain; (S.H.); (A.M.G.-L.); (J.M.); (O.F.-M.); (M.A.O.); (C.G.-M.); (D.D.); (E.L.-D.)
- Ramón y Cajal Institute of Sanitary Research (IRYCIS), 28034 Madrid, Spain
| | - Jorge Monserrat
- Department of Medicine and Medical Specialities, University of Alcala, Crta N-II km 33.6, 28871 Alcala de Henares, Spain; (S.H.); (A.M.G.-L.); (J.M.); (O.F.-M.); (M.A.O.); (C.G.-M.); (D.D.); (E.L.-D.)
- Ramón y Cajal Institute of Sanitary Research (IRYCIS), 28034 Madrid, Spain
| | - Mar Atienza-Pérez
- Service of Rehabilitation, National Hospital for Paraplegic Patients, Carr. de la Peraleda, S/N, 45004 Toledo, Spain;
| | - Oscar Fraile-Martinez
- Department of Medicine and Medical Specialities, University of Alcala, Crta N-II km 33.6, 28871 Alcala de Henares, Spain; (S.H.); (A.M.G.-L.); (J.M.); (O.F.-M.); (M.A.O.); (C.G.-M.); (D.D.); (E.L.-D.)
- Ramón y Cajal Institute of Sanitary Research (IRYCIS), 28034 Madrid, Spain
| | - Miguel A. Ortega
- Department of Medicine and Medical Specialities, University of Alcala, Crta N-II km 33.6, 28871 Alcala de Henares, Spain; (S.H.); (A.M.G.-L.); (J.M.); (O.F.-M.); (M.A.O.); (C.G.-M.); (D.D.); (E.L.-D.)
- Ramón y Cajal Institute of Sanitary Research (IRYCIS), 28034 Madrid, Spain
| | - Cielo García-Montero
- Department of Medicine and Medical Specialities, University of Alcala, Crta N-II km 33.6, 28871 Alcala de Henares, Spain; (S.H.); (A.M.G.-L.); (J.M.); (O.F.-M.); (M.A.O.); (C.G.-M.); (D.D.); (E.L.-D.)
- Ramón y Cajal Institute of Sanitary Research (IRYCIS), 28034 Madrid, Spain
| | - David Díaz
- Department of Medicine and Medical Specialities, University of Alcala, Crta N-II km 33.6, 28871 Alcala de Henares, Spain; (S.H.); (A.M.G.-L.); (J.M.); (O.F.-M.); (M.A.O.); (C.G.-M.); (D.D.); (E.L.-D.)
- Ramón y Cajal Institute of Sanitary Research (IRYCIS), 28034 Madrid, Spain
| | - Elisa Lopez-Dolado
- Department of Medicine and Medical Specialities, University of Alcala, Crta N-II km 33.6, 28871 Alcala de Henares, Spain; (S.H.); (A.M.G.-L.); (J.M.); (O.F.-M.); (M.A.O.); (C.G.-M.); (D.D.); (E.L.-D.)
- Service of Rehabilitation, National Hospital for Paraplegic Patients, Carr. de la Peraleda, S/N, 45004 Toledo, Spain;
| | - Melchor Álvarez-Mon
- Department of Medicine and Medical Specialities, University of Alcala, Crta N-II km 33.6, 28871 Alcala de Henares, Spain; (S.H.); (A.M.G.-L.); (J.M.); (O.F.-M.); (M.A.O.); (C.G.-M.); (D.D.); (E.L.-D.)
- Ramón y Cajal Institute of Sanitary Research (IRYCIS), 28034 Madrid, Spain
- Immune System Diseases-Rheumatology Service and Internal Medicine, Prince of Asturias University Hospital (CIBEREHD), 28806 Alcala de Henares, Spain
| |
Collapse
|
22
|
Ligotti ME, Accardi G, Aiello A, Aprile S, Calabrò A, Caldarella R, Caruso C, Ciaccio M, Corsale AM, Dieli F, Di Simone M, Giammanco GM, Mascarella C, Akbar AN, Meraviglia S, Candore G. Sicilian semi- and supercentenarians: identification of age-related T-cell immunophenotype to define longevity trait. Clin Exp Immunol 2023; 214:61-78. [PMID: 37395602 PMCID: PMC10711357 DOI: 10.1093/cei/uxad074] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2023] [Revised: 05/12/2023] [Accepted: 06/30/2023] [Indexed: 07/04/2023] Open
Abstract
The immunophenotype of oldest centenarians, i.e. semi- and supercentenarians, could provide important information about their ability to adapt to factors associated with immune changes, including ageing per se and chronic Cytomegalovirus infection. We investigated, by flow cytometry, variations in percentages and absolute numbers of immune cell subsets, focusing on T cells, and pro-inflammatory parameters in a cohort of 28 women and 26 men (age range 19-110 years). We observed variability in hallmarks of immunosenescence related to age and Cytomegalovirus serological status. The eight oldest centenarians showed the lowest percentages of naïve T cells, due to their age, and the highest percentages of T-effector memory cells re-expressing CD45RA (TEMRA), according to their cytomegalovirus status, and high levels of serum pro-inflammatory parameters, although their means were lower than that of remaining 90+ donors. Some of them showed CD8 naïve and TEMRA percentages, and exhaustion/pro-inflammatory markers comparable to the younger ones. Our study supports the suggestion that immune ageing, especially of oldest centenarians, exhibits great variability that is not only attributable to a single contributor but should also be the full result of a combination of several factors. Everyone ages differently because he/she is unique in genetics and experience of life and this applies even more to the immune system; everybody has had a different immunological history. Furthermore, our findings on inflammatory markers, TEMRA and CMV seropositivity in centenarians, discussed in the light of the most recent literature, suggest that these changes might be not unfavourable for centenarians, and in particular for the oldest ones.
Collapse
Affiliation(s)
- Mattia Emanuela Ligotti
- Laboratory of Immunopathology and Immunosenescence, Department of Biomedicine, Neurosciences and Advanced Diagnostics, University of Palermo, Palermo, Italy
| | - Giulia Accardi
- Laboratory of Immunopathology and Immunosenescence, Department of Biomedicine, Neurosciences and Advanced Diagnostics, University of Palermo, Palermo, Italy
| | - Anna Aiello
- Laboratory of Immunopathology and Immunosenescence, Department of Biomedicine, Neurosciences and Advanced Diagnostics, University of Palermo, Palermo, Italy
| | - Stefano Aprile
- Laboratory of Immunopathology and Immunosenescence, Department of Biomedicine, Neurosciences and Advanced Diagnostics, University of Palermo, Palermo, Italy
- Unit of Transfusion Medicine, San Giovanni di Dio Hospital, Agrigento, Italy
| | - Anna Calabrò
- Laboratory of Immunopathology and Immunosenescence, Department of Biomedicine, Neurosciences and Advanced Diagnostics, University of Palermo, Palermo, Italy
| | - Rosalia Caldarella
- Department of Laboratory Medicine, University Hospital “P. Giaccone”, Palermo, Italy
| | - Calogero Caruso
- Laboratory of Immunopathology and Immunosenescence, Department of Biomedicine, Neurosciences and Advanced Diagnostics, University of Palermo, Palermo, Italy
| | - Marcello Ciaccio
- Department of Laboratory Medicine, University Hospital “P. Giaccone”, Palermo, Italy
- Section of Clinical Biochemistry, Clinical Molecular Medicine and Clinical Laboratory Medicine Department of Biomedicine, Neurosciences and Advanced Diagnostics, University of Palermo, Palermo, Italy
| | - Anna Maria Corsale
- Central Laboratory of Advanced Diagnosis and Biomedical Research, University Hospital “P. Giaccone”, Palermo, Italy
- Department of Health Promotion Sciences, Maternal and Infant Care, Internal Medicine and Medical Specialties “G. D’Alessandro”, University of Palermo, Palermo, Italy
| | - Francesco Dieli
- Central Laboratory of Advanced Diagnosis and Biomedical Research, University Hospital “P. Giaccone”, Palermo, Italy
- Department of Biomedicine, Neurosciences and Advanced Diagnostics, University of Palermo, Palermo, Italy
| | - Marta Di Simone
- Central Laboratory of Advanced Diagnosis and Biomedical Research, University Hospital “P. Giaccone”, Palermo, Italy
- Department of Health Promotion Sciences, Maternal and Infant Care, Internal Medicine and Medical Specialties “G. D’Alessandro”, University of Palermo, Palermo, Italy
| | - Giovanni Maurizio Giammanco
- Section of Microbiology, Department of Health Promotion Sciences, Maternal and Infant Care, Internal Medicine and Medical Specialties “G. D’Alessandro”, University of Palermo, Palermo, Italy
| | - Chiara Mascarella
- Section of Microbiology, Department of Health Promotion Sciences, Maternal and Infant Care, Internal Medicine and Medical Specialties “G. D’Alessandro”, University of Palermo, Palermo, Italy
| | - Arne N Akbar
- Division of Medicine, Experimental and Therapeutic Medicine, University College London, London, UK
| | - Serena Meraviglia
- Central Laboratory of Advanced Diagnosis and Biomedical Research, University Hospital “P. Giaccone”, Palermo, Italy
- Department of Biomedicine, Neurosciences and Advanced Diagnostics, University of Palermo, Palermo, Italy
| | - Giuseppina Candore
- Laboratory of Immunopathology and Immunosenescence, Department of Biomedicine, Neurosciences and Advanced Diagnostics, University of Palermo, Palermo, Italy
| |
Collapse
|
23
|
Wang L, Zhang X, Zhang H, Lu K, Li M, Li X, Ou Y, Zhao X, Wu X, Wu X, Liu J, Xing M, Liu H, Zhang Y, Tan Y, Li F, Deng X, Deng J, Zhang X, Li J, Zhao Y, Ding Q, Wang H, Wang X, Luo Y, Zhou B, Zhang H. Excessive apoptosis of Rip1-deficient T cells leads to premature aging. EMBO Rep 2023; 24:e57925. [PMID: 37965894 PMCID: PMC10702839 DOI: 10.15252/embr.202357925] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2023] [Revised: 10/29/2023] [Accepted: 11/02/2023] [Indexed: 11/16/2023] Open
Abstract
In mammals, the most remarkable T cell variations with aging are the shrinking of the naïve T cell pool and the enlargement of the memory T cell pool, which are partially caused by thymic involution. However, the mechanism underlying the relationship between T-cell changes and aging remains unclear. In this study, we find that T-cell-specific Rip1 KO mice show similar age-related T cell changes and exhibit signs of accelerated aging-like phenotypes, including inflammation, multiple age-related diseases, and a shorter lifespan. Mechanistically, Rip1-deficient T cells undergo excessive apoptosis and promote chronic inflammation. Consistent with this, blocking apoptosis by co-deletion of Fadd in Rip1-deficient T cells significantly rescues lymphopenia, the imbalance between naïve and memory T cells, and aging-like phenotypes, and prolongs life span in T-cell-specific Rip1 KO mice. These results suggest that the reduction and hyperactivation of T cells can have a significant impact on organismal health and lifespan, underscoring the importance of maintaining T cell homeostasis for healthy aging and prevention or treatment of age-related diseases.
Collapse
Affiliation(s)
- Lingxia Wang
- CAS Key Laboratory of Nutrition, Metabolism and Food Safety, Shanghai Institute of Nutrition and HealthUniversity of Chinese Academy of Sciences, Chinese Academy of SciencesShanghaiChina
| | - Xixi Zhang
- CAS Key Laboratory of Nutrition, Metabolism and Food Safety, Shanghai Institute of Nutrition and HealthUniversity of Chinese Academy of Sciences, Chinese Academy of SciencesShanghaiChina
| | - Haiwei Zhang
- CAS Key Laboratory of Nutrition, Metabolism and Food Safety, Shanghai Institute of Nutrition and HealthUniversity of Chinese Academy of Sciences, Chinese Academy of SciencesShanghaiChina
| | - Kaili Lu
- Department of NeurologyShanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of MedicineShanghaiChina
| | - Ming Li
- CAS Key Laboratory of Nutrition, Metabolism and Food Safety, Shanghai Institute of Nutrition and HealthUniversity of Chinese Academy of Sciences, Chinese Academy of SciencesShanghaiChina
| | - Xiaoming Li
- CAS Key Laboratory of Nutrition, Metabolism and Food Safety, Shanghai Institute of Nutrition and HealthUniversity of Chinese Academy of Sciences, Chinese Academy of SciencesShanghaiChina
| | - Yangjing Ou
- CAS Key Laboratory of Nutrition, Metabolism and Food Safety, Shanghai Institute of Nutrition and HealthUniversity of Chinese Academy of Sciences, Chinese Academy of SciencesShanghaiChina
| | - Xiaoming Zhao
- CAS Key Laboratory of Nutrition, Metabolism and Food Safety, Shanghai Institute of Nutrition and HealthUniversity of Chinese Academy of Sciences, Chinese Academy of SciencesShanghaiChina
| | - Xiaoxia Wu
- CAS Key Laboratory of Nutrition, Metabolism and Food Safety, Shanghai Institute of Nutrition and HealthUniversity of Chinese Academy of Sciences, Chinese Academy of SciencesShanghaiChina
| | - Xuanhui Wu
- CAS Key Laboratory of Nutrition, Metabolism and Food Safety, Shanghai Institute of Nutrition and HealthUniversity of Chinese Academy of Sciences, Chinese Academy of SciencesShanghaiChina
| | - Jianling Liu
- CAS Key Laboratory of Nutrition, Metabolism and Food Safety, Shanghai Institute of Nutrition and HealthUniversity of Chinese Academy of Sciences, Chinese Academy of SciencesShanghaiChina
| | - Mingyan Xing
- CAS Key Laboratory of Nutrition, Metabolism and Food Safety, Shanghai Institute of Nutrition and HealthUniversity of Chinese Academy of Sciences, Chinese Academy of SciencesShanghaiChina
| | - Han Liu
- CAS Key Laboratory of Nutrition, Metabolism and Food Safety, Shanghai Institute of Nutrition and HealthUniversity of Chinese Academy of Sciences, Chinese Academy of SciencesShanghaiChina
| | - Yue Zhang
- Department of Anesthesiology, Ruijin HospitalShanghai Jiao Tong University School of MedicineShanghaiChina
| | - Yongchang Tan
- Department of Anesthesiology, Ruijin HospitalShanghai Jiao Tong University School of MedicineShanghaiChina
| | - Fang Li
- Department of Anesthesiology, Shanghai First People's HospitalShanghai Jiaotong UniversityShanghaiChina
| | - Xiaoxue Deng
- CAS Key Laboratory of Molecular Virology and ImmunologyUniversity of Chinese Academy of Sciences, Chinese Academy of SciencesShanghaiChina
| | - Jiangshan Deng
- Department of NeurologyShanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of MedicineShanghaiChina
| | - Xiaojie Zhang
- Department of NeurologyShanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of MedicineShanghaiChina
| | - Jinbao Li
- Department of Anesthesiology, Shanghai First People's HospitalShanghai Jiaotong UniversityShanghaiChina
| | - Yuwu Zhao
- Department of NeurologyShanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of MedicineShanghaiChina
| | - Qiurong Ding
- CAS Key Laboratory of Nutrition, Metabolism and Food Safety, Shanghai Institute of Nutrition and HealthUniversity of Chinese Academy of Sciences, Chinese Academy of SciencesShanghaiChina
| | - Haikun Wang
- CAS Key Laboratory of Molecular Virology and ImmunologyUniversity of Chinese Academy of Sciences, Chinese Academy of SciencesShanghaiChina
| | - Xiuzhe Wang
- Department of NeurologyShanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of MedicineShanghaiChina
| | - Yan Luo
- Department of Anesthesiology, Ruijin HospitalShanghai Jiao Tong University School of MedicineShanghaiChina
| | - Ben Zhou
- CAS Key Laboratory of Nutrition, Metabolism and Food Safety, Shanghai Institute of Nutrition and HealthUniversity of Chinese Academy of Sciences, Chinese Academy of SciencesShanghaiChina
| | - Haibing Zhang
- CAS Key Laboratory of Nutrition, Metabolism and Food Safety, Shanghai Institute of Nutrition and HealthUniversity of Chinese Academy of Sciences, Chinese Academy of SciencesShanghaiChina
| |
Collapse
|
24
|
Snyder ME, Anderson MR, Benvenuto LJ, Sutton RM, Bondonese A, Koshy R, Burke R, Clifford S, Craig A, Iasella CJ, Hannan SJ, Popescu I, Zhang Y, Sanchez PG, Alder JK, McDyer JF. Impact of age and telomere length on circulating T cells and rejection risk after lung transplantation for idiopathic pulmonary fibrosis. J Heart Lung Transplant 2023; 42:1666-1677. [PMID: 37544465 PMCID: PMC10839116 DOI: 10.1016/j.healun.2023.08.001] [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: 09/26/2022] [Revised: 07/18/2023] [Accepted: 08/01/2023] [Indexed: 08/08/2023] Open
Abstract
BACKGROUND Most idiopathic pulmonary fibrosis (IPF) lung transplant recipients (IPF-LTRs) have short telomere (ST) length. Inherited mutations in telomere-related genes are associated with the development of T cell immunodeficiency. Despite this, IPF-LTRs with telomere-related rare variants are not protected from acute cellular rejection (ACR). We set out to determine the impact of both age and telomere length on the circulating T cell compartment and ACR burden of IPF-LTRs. METHODS We identified 106 IPF-LTRs who had telomere length testing using flowFISH (57 with short telomeres and 49 with long telomeres) as well as a subset from both cohorts who had cryopreserved PBMC at least 1 time point, 6 months posttransplantation. Circulating T cells from before transplantation and at 6 and 12 months posttransplantation were analyzed using multiparameter flow cytometry to study phenotype and functional capacity, and bulk T cell receptor sequencing was performed to study repertoire diversity. Linear regression was used to study the relationship of age and telomere length on early (within 1 year) and late (between 1 and 2 years) ACR. RESULTS IPF-LTRs with ST were found to have premature "aging" of their circulating T cell compartment, with age-agnostic elevations in posttransplant terminal differentiation of CD8+ T cells, increased granzyme B positivity of both CD8+ and CD4+ T cells, upregulation of the exhaustion marker, CD57, and chemotactic protein CCR5, and enhanced T cell receptor clonal expansion. Additionally, we found a significant decline in early ACR burden with increasing age, but only in the ST cohort. CONCLUSIONS IPF-LTRs with ST have premature "aging" of their circulating T cell compartment posttransplantation and a clear age-related decline in ACR burden.
Collapse
Affiliation(s)
- Mark E Snyder
- Department of Medicine, University of Pittsburgh, Pittsburgh, PA, USA; Department of Immunology, University of Pittsburgh, Pittsburgh, PA, USA; Starzl Transplantation Institute, Pittsburgh, Pennsylvania.
| | - Michaela R Anderson
- Department of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Luke J Benvenuto
- Department of Medicine, Columbia University Irving Medical Center, New York, New York
| | - Rachel M Sutton
- Department of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - Anna Bondonese
- Department of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - Ritchie Koshy
- Department of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - Robin Burke
- Department of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - Sarah Clifford
- Department of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - Andrew Craig
- Department of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - Carlo J Iasella
- Department of Medicine, University of Pittsburgh, Pittsburgh, PA, USA; Department of Pharmacology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Stefanie J Hannan
- Department of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - Iulia Popescu
- Department of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - Yingze Zhang
- Department of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - Pablo G Sanchez
- Department of Cardiothoracic Surgery, University of Pittsburgh, Pittsburgh, PA, USA
| | - Jonathan K Alder
- Department of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - John F McDyer
- Department of Medicine, University of Pittsburgh, Pittsburgh, PA, USA; Starzl Transplantation Institute, Pittsburgh, Pennsylvania.
| |
Collapse
|
25
|
Hultberg J, Blixt E, Göransson R, Adolfsson J, Govender M, Larsson M, Nilsdotter-Augustinsson Å, Ernerudh J, Nyström S. In-depth immune profiling reveals advanced B- and T-cell differentiation to be associated with Th1-driven immune dysregulation in common variable immunodeficiency. Clin Immunol 2023; 257:109816. [PMID: 37918468 DOI: 10.1016/j.clim.2023.109816] [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] [Revised: 10/25/2023] [Accepted: 10/27/2023] [Indexed: 11/04/2023]
Abstract
Common variable immunodeficiency (CVID) is an inborn error of immunity characterized by low levels of antibodies. In addition to infections, many patients also suffer from T-helper 1-driven immune dysregulation, which is associated with increased mortality. The aim of this study was to perform in-depth characterization of the T and the B cell compartments in a well-defined cohort of patients affected by CVID and correlate the findings to the level of clinical immune dysregulation. We used mass cytometry, targeted proteomics, flow cytometry and functional assays to delineate the immunological phenotype of 15 CVID-affected patients with different levels of immune dysregulation. Unbiased clustering of T cell mass cytometry data correlated with CVID-related immune dysregulation and plasma protein profiles. Expanded CXCR3+ T-bet-expressing B cells correlated with effector memory CD4+ T cell clusters, and increased plasma levels of CXCR3-ligands. Our findings indicate an interplay between B cells and T cells in CVID-related immune dysregulation and provide a better understanding of the underlying pathological mechanisms.
Collapse
Affiliation(s)
- Jonas Hultberg
- Division of Molecular Medicine and Virology, Department of Biomedical and Clinical Sciences, Linköping University, Sweden
| | - Emelie Blixt
- Department of Clinical Immunology and Transfusion Medicine, and Department of Biomedical and Clinical Sciences, Linköping University, Sweden
| | - Robin Göransson
- Department of Clinical Immunology and Transfusion Medicine, and Department of Biomedical and Clinical Sciences, Linköping University, Sweden
| | - Jörgen Adolfsson
- Division of Molecular Medicine and Virology, Department of Biomedical and Clinical Sciences, Linköping University, Sweden
| | - Melissa Govender
- Division of Molecular Medicine and Virology, Department of Biomedical and Clinical Sciences, Linköping University, Sweden
| | - Marie Larsson
- Division of Molecular Medicine and Virology, Department of Biomedical and Clinical Sciences, Linköping University, Sweden
| | - Åsa Nilsdotter-Augustinsson
- Division of Inflammation and Infection, Department of Biomedical and Clinical Sciences, Linköping University, Sweden
| | - Jan Ernerudh
- Department of Clinical Immunology and Transfusion Medicine, and Department of Biomedical and Clinical Sciences, Linköping University, Sweden
| | - Sofia Nyström
- Department of Clinical Immunology and Transfusion Medicine, and Department of Biomedical and Clinical Sciences, Linköping University, Sweden.
| |
Collapse
|
26
|
Quinn KM, Vicencio DM, La Gruta NL. The paradox of aging: Aging-related shifts in T cell function and metabolism. Semin Immunol 2023; 70:101834. [PMID: 37659169 DOI: 10.1016/j.smim.2023.101834] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/13/2023] [Revised: 08/17/2023] [Accepted: 08/17/2023] [Indexed: 09/04/2023]
Abstract
T cell survival, differentiation after stimulation, and function are intrinsically linked to distinct cellular metabolic states. The ability of T cells to readily transition between metabolic states enables flexibility to meet the changing energy demands defined by distinct effector states or T cell lineages. Immune aging is characterized, in part, by the loss of naïve T cells, accumulation of senescent T cells, severe dysfunction in memory phenotype T cells in particular, and elevated levels of inflammatory cytokines, or 'inflammaging'. Here, we review our current understanding of the phenotypic and functional changes that occur with aging in T cells, and how they relate to metabolic changes in the steady state and after T cell activation. We discuss the apparent contradictions in the aging T cell phenotype - where enhanced differentiation states and metabolic profiles in the steady state can correspond to a diminished capacity to adapt metabolically and functionally after T cell activation. Finally, we discuss key recent studies that indicate the enormous potential for aged T cell metabolism to induce systemic inflammaging and organism-wide multimorbidity, resulting in premature death.
Collapse
Affiliation(s)
- Kylie M Quinn
- School of Health and Biomedical Sciences, Royal Melbourne Institute of Technology University, Bundoora, Victoria, Australia; Immunity Program and Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, Clayton, Victoria, Australia
| | - Daniela M Vicencio
- Immunity Program and Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, Clayton, Victoria, Australia; Division of Biomedical Sciences, Warwick Medical School, The University of Warwick, Coventry, UK
| | - Nicole L La Gruta
- Immunity Program and Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, Clayton, Victoria, Australia.
| |
Collapse
|
27
|
Wang A, Liu W, Jin Y, Wei B, Fan Y, Guo X, Gou X. Identification of immunological characteristics and cuproptosis-related molecular clusters in Rheumatoid arthritis. Int Immunopharmacol 2023; 123:110804. [PMID: 37595490 DOI: 10.1016/j.intimp.2023.110804] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2023] [Revised: 08/09/2023] [Accepted: 08/11/2023] [Indexed: 08/20/2023]
Abstract
BACKGROUND Rheumatoid arthritis (RA) is a chronic autoimmune disease characterised by progressive articular damage, functional loss, and comorbidities. The relationship between cuproptosis, a form of programmed cell death, and RA remains unknown. Therefore, this study aimed to explore cuproptosis-related molecular clusters in RA. METHODS Gene expression profiles of GSE93272 were downloaded from the Gene Expression Omnibus to identify the expression profiles of cuproptosis regulators and the immune infiltration characteristics of RA. The molecular clusters of cuproptosis-related genes and the related immune cell infiltration were explored. Cluster-specific differentially expressed genes were identified using the weighted gene co-expression network analysis. Further, an external dataset (GSE15573) was used, and an enzyme-linked immunosorbent assay was performed to validate the predictive efficiency. RESULTS Thirteen cuproptosis-related genes and activated immune responses were identified between patients with RA and controls. Immune infiltration revealed significant immunological heterogeneity in the two cuproptosis-related molecular clusters in RA. Functional enrichment indicated that Cluster1 and Cluster2 were predominantly enriched in the toll-like receptor signalling pathway and regulation of autophagy, respectively. Further, the performance of FAM96A and CGRRF1 genes in the external validation dataset was observed to be relatively satisfactory (area under the receiver operating characteristic curve = 0.687 and 0.674, respectively). Based on our serum samples, FAM96A and CGRRF1 both exhibited higher expression levels in patients with RA (p = 0.001; p = 0.000). CONCLUSIONS Our study systematically illustrated the involvement of cuproptosis in the progression of RA, and explored the pathogenic mechanisms and novel therapeutic strategies for RA, targeting FAM96A and CGRRF1.
Collapse
Affiliation(s)
- Aihua Wang
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin 300193, China; National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin 300381, China
| | - Wei Liu
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin 300193, China; National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin 300381, China.
| | - Yue Jin
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin 300193, China; National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin 300381, China
| | - Bowen Wei
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin 300193, China; National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin 300381, China
| | - Yihua Fan
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin 300193, China; National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin 300381, China
| | - Xiaojing Guo
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin 300193, China; National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin 300381, China
| | - Xiaoping Gou
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin 300193, China; National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin 300381, China
| |
Collapse
|
28
|
Vrbic M, Milinkovic A. Two vicious circles associated with the aging of the immune system in the development of severe forms of COVID-19. FRONTIERS IN AGING 2023; 4:1260053. [PMID: 37780864 PMCID: PMC10537960 DOI: 10.3389/fragi.2023.1260053] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/21/2023] [Accepted: 09/04/2023] [Indexed: 10/03/2023]
Abstract
Background: The immune-inflammatory response is the basis of the pathophysiology of SARS-Cov-2 infection. In severe cases of COVID-19 uncontrolled systemic inflammatory response causes multiorgan dysfunction (MODS), as the most common immediate cause of death. Unfavorable outcome of the COVID-19 most often occurs in elderly patients. The aim of the study was to establish parameters with prognostic significance in severe cases of COVID-19 according to life years, laboratory markers of sepsis and MODS, as well as the number of peripheral CD4+ and CD8+T lymphocytes in 20 consecutively selected critically ill patients. Results: Eleven subjects were male, 9 female, mean age 73.45 ± 11.59, among which the oldest patient was 94 and the youngest 43 years. All the patients met the sepsis and MODS criteria. Increased age and low CD4+ and CD8+T cell counts were identified as independent predictors of death. Only the two youngest patients (43 and 50 years old) survived 28 days, and they are the only ones with a CD4 lymphocyte count above 500 cells/mm3. Conclusion: Senescence of the immune system is mostly characterized by reduced regenerative capacity of adaptive immunity with diminished ability to respond to new antigens and a manifested proinflammatory phenotype. Additional reduction of protective capacity by further deterioration of T cell quantity and quality due to sepsis itself and mutual interaction of senescent T cells and vascular endothelial cells in the induction of cytokine storm represent two complementary vicious cycles in the development of sepsis-related multiorgan dysfunction.
Collapse
Affiliation(s)
| | - Ana Milinkovic
- Chelsea and Westminster Foundation Trust and Imperial College London, London, United Kingdom
| |
Collapse
|
29
|
Tourountzis T, Lioulios G, Van Laecke S, Ginikopoulou E, Nikolaidou V, Moysidou E, Stai S, Christodoulou M, Fylaktou A, Glorieux G, Stangou M. Immunosenescence and Immune Exhaustion Are Associated with Levels of Protein-Bound Uremic Toxins in Patients on Hemodialysis. Biomedicines 2023; 11:2504. [PMID: 37760945 PMCID: PMC10525954 DOI: 10.3390/biomedicines11092504] [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: 08/06/2023] [Revised: 09/04/2023] [Accepted: 09/08/2023] [Indexed: 09/29/2023] Open
Abstract
BACKGROUND The accumulation of protein-bound uremic toxins (PBUTs) in chronic kidney disease may affect patients' immune status. The aim of the study was to evaluate their potential impacts on lymphocyte alterations in patients on hemodialysis (HD). METHODS The plasma levels of PBUTs were assessed in 54 patients on HD and 31 healthy individuals, using ultra-performance liquid chromatography. The results correlated with the senescent and exhausted status of lymphocytes, based on certain surface molecules, analyzed by flow cytometry. RESULTS The plasma levels of PBUTs were significantly increased in the patients on HD compared with the healthy controls. The patients with residual kidney function had reduced hippuric acid (HA) levels, total (p = 0.03) and free (p = 0.04), and free IxS levels (p = 0.02). The total and free HA levels correlated negatively with less differentiated subpopulations, CD4+CD45RA+CD31+ (p = 0.037 and p = 0.027), CD8+CD28+CD57- (p = 0.01, p = 0.01), and naïve B cells (CD19+IgD+CD27-) (p = 0.04, p = 0.03). Both the total and the free pCS levels correlated positively with exhausted CD4 cells, p = 0.02 and p = 0.01, respectively. A multivariate analysis showed that IxS and age were the main independent parameters implicated in the reduction intotal CD4 and B lymphocytes and their naïve and early differentiated subsets. CONCLUSIONS Increased PBUTs levels are associated with immune disturbances of patients on HD, HA, and IxS in the immunosenescent and pCS in the immunoexhaustion alterations.
Collapse
Affiliation(s)
| | - Georgios Lioulios
- Department of Nephrology, General Hospital “Hippokratio”, School of Medicine, Aristotle University of Thessaloniki, 54642 Thessaloniki, Greece; (G.L.); (E.M.); (S.S.); (M.C.); (M.S.)
| | - Steven Van Laecke
- Department of Internal Medicine and Pediatrics, Nephrology Unit, Ghent University Hospital, 9000 Gent, Belgium; (S.V.L.); (G.G.)
| | | | - Vasiliki Nikolaidou
- Department of Immunology, National Peripheral Histocompatibility Center, General Hospital “Hippokratio”, 54642 Thessaloniki, Greece; (V.N.); (A.F.)
| | - Eleni Moysidou
- Department of Nephrology, General Hospital “Hippokratio”, School of Medicine, Aristotle University of Thessaloniki, 54642 Thessaloniki, Greece; (G.L.); (E.M.); (S.S.); (M.C.); (M.S.)
| | - Stamatia Stai
- Department of Nephrology, General Hospital “Hippokratio”, School of Medicine, Aristotle University of Thessaloniki, 54642 Thessaloniki, Greece; (G.L.); (E.M.); (S.S.); (M.C.); (M.S.)
| | - Michalis Christodoulou
- Department of Nephrology, General Hospital “Hippokratio”, School of Medicine, Aristotle University of Thessaloniki, 54642 Thessaloniki, Greece; (G.L.); (E.M.); (S.S.); (M.C.); (M.S.)
| | - Asimina Fylaktou
- Department of Immunology, National Peripheral Histocompatibility Center, General Hospital “Hippokratio”, 54642 Thessaloniki, Greece; (V.N.); (A.F.)
| | - Griet Glorieux
- Department of Internal Medicine and Pediatrics, Nephrology Unit, Ghent University Hospital, 9000 Gent, Belgium; (S.V.L.); (G.G.)
| | - Maria Stangou
- Department of Nephrology, General Hospital “Hippokratio”, School of Medicine, Aristotle University of Thessaloniki, 54642 Thessaloniki, Greece; (G.L.); (E.M.); (S.S.); (M.C.); (M.S.)
| |
Collapse
|
30
|
Terrabuio E, Zenaro E, Constantin G. The role of the CD8+ T cell compartment in ageing and neurodegenerative disorders. Front Immunol 2023; 14:1233870. [PMID: 37575227 PMCID: PMC10416633 DOI: 10.3389/fimmu.2023.1233870] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2023] [Accepted: 07/13/2023] [Indexed: 08/15/2023] Open
Abstract
CD8+ lymphocytes are adaptive immunity cells with the particular function to directly kill the target cell following antigen recognition in the context of MHC class I. In addition, CD8+ T cells may release pro-inflammatory cytokines, such as tumor necrosis factor-α (TNF-α) and interferon-γ (IFN-γ), and a plethora of other cytokines and chemoattractants modulating immune and inflammatory responses. A role for CD8+ T cells has been suggested in aging and several diseases of the central nervous system (CNS), including Alzheimer's disease, Parkinson's disease, multiple sclerosis, amyotrophic lateral sclerosis, limbic encephalitis-induced temporal lobe epilepsy and Susac syndrome. Here we discuss the phenotypic and functional alterations of CD8+ T cell compartment during these conditions, highlighting similarities and differences between CNS disorders. Particularly, we describe the pathological changes in CD8+ T cell memory phenotypes emphasizing the role of senescence and exhaustion in promoting neuroinflammation and neurodegeneration. We also discuss the relevance of trafficking molecules such as selectins, mucins and integrins controlling the extravasation of CD8+ T cells into the CNS and promoting disease development. Finally, we discuss how CD8+ T cells may induce CNS tissue damage leading to neurodegeneration and suggest that targeting detrimental CD8+ T cells functions may have therapeutic effect in CNS disorders.
Collapse
Affiliation(s)
- Eleonora Terrabuio
- Department of Medicine, Section of General Pathology, University of Verona, Verona, Italy
| | | | | |
Collapse
|
31
|
Hieber C, Grabbe S, Bros M. Counteracting Immunosenescence-Which Therapeutic Strategies Are Promising? Biomolecules 2023; 13:1085. [PMID: 37509121 PMCID: PMC10377144 DOI: 10.3390/biom13071085] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2023] [Revised: 07/03/2023] [Accepted: 07/05/2023] [Indexed: 07/30/2023] Open
Abstract
Aging attenuates the overall responsiveness of the immune system to eradicate pathogens. The increased production of pro-inflammatory cytokines by innate immune cells under basal conditions, termed inflammaging, contributes to impaired innate immune responsiveness towards pathogen-mediated stimulation and limits antigen-presenting activity. Adaptive immune responses are attenuated as well due to lowered numbers of naïve lymphocytes and their impaired responsiveness towards antigen-specific stimulation. Additionally, the numbers of immunoregulatory cell types, comprising regulatory T cells and myeloid-derived suppressor cells, that inhibit the activity of innate and adaptive immune cells are elevated. This review aims to summarize our knowledge on the cellular and molecular causes of immunosenescence while also taking into account senescence effects that constitute immune evasion mechanisms in the case of chronic viral infections and cancer. For tumor therapy numerous nanoformulated drugs have been developed to overcome poor solubility of compounds and to enable cell-directed delivery in order to restore immune functions, e.g., by addressing dysregulated signaling pathways. Further, nanovaccines which efficiently address antigen-presenting cells to mount sustained anti-tumor immune responses have been clinically evaluated. Further, senolytics that selectively deplete senescent cells are being tested in a number of clinical trials. Here we discuss the potential use of such drugs to improve anti-aging therapy.
Collapse
Affiliation(s)
- Christoph Hieber
- Department of Dermatology, University Medical Center of the Johannes Gutenberg-University Mainz, Langenbeckstraße 1, 55131 Mainz, Germany
- Institute of Molecular Biology (IMB), Ackermannweg 4, 55128 Mainz, Germany
| | - Stephan Grabbe
- Department of Dermatology, University Medical Center of the Johannes Gutenberg-University Mainz, Langenbeckstraße 1, 55131 Mainz, Germany
- Institute of Molecular Biology (IMB), Ackermannweg 4, 55128 Mainz, Germany
| | - Matthias Bros
- Department of Dermatology, University Medical Center of the Johannes Gutenberg-University Mainz, Langenbeckstraße 1, 55131 Mainz, Germany
| |
Collapse
|
32
|
Kell L, Simon AK, Alsaleh G, Cox LS. The central role of DNA damage in immunosenescence. FRONTIERS IN AGING 2023; 4:1202152. [PMID: 37465119 PMCID: PMC10351018 DOI: 10.3389/fragi.2023.1202152] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 04/07/2023] [Accepted: 06/22/2023] [Indexed: 07/20/2023]
Abstract
Ageing is the biggest risk factor for the development of multiple chronic diseases as well as increased infection susceptibility and severity of diseases such as influenza and COVID-19. This increased disease risk is linked to changes in immune function during ageing termed immunosenescence. Age-related loss of immune function, particularly in adaptive responses against pathogens and immunosurveillance against cancer, is accompanied by a paradoxical gain of function of some aspects of immunity such as elevated inflammation and increased incidence of autoimmunity. Of the many factors that contribute to immunosenescence, DNA damage is emerging as a key candidate. In this review, we discuss the evidence supporting the hypothesis that DNA damage may be a central driver of immunosenescence through senescence of both immune cells and cells of non-haematopoietic lineages. We explore why DNA damage accumulates during ageing in a major cell type, T cells, and how this may drive age-related immune dysfunction. We further propose that existing immunosenescence interventions may act, at least in part, by mitigating DNA damage and restoring DNA repair processes (which we term "genoprotection"). As such, we propose additional treatments on the basis of their evidence for genoprotection, and further suggest that this approach may provide a viable therapeutic strategy for improving immunity in older people.
Collapse
Affiliation(s)
- Loren Kell
- Department of Biochemistry, University of Oxford, Oxford, United Kingdom
- Botnar Institute for Musculoskeletal Sciences, Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences (NDORMS), University of Oxford, Oxford, United Kingdom
- The Kennedy Institute of Rheumatology, Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences (NDORMS), University of Oxford, Oxford, United Kingdom
| | - Anna Katharina Simon
- The Kennedy Institute of Rheumatology, Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences (NDORMS), University of Oxford, Oxford, United Kingdom
- Max Delbrück Center for Molecular Medicine, Berlin, Germany
| | - Ghada Alsaleh
- Botnar Institute for Musculoskeletal Sciences, Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences (NDORMS), University of Oxford, Oxford, United Kingdom
- The Kennedy Institute of Rheumatology, Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences (NDORMS), University of Oxford, Oxford, United Kingdom
| | - Lynne S. Cox
- Department of Biochemistry, University of Oxford, Oxford, United Kingdom
| |
Collapse
|
33
|
Laphanuwat P, Gomes DCO, Akbar AN. Senescent T cells: Beneficial and detrimental roles. Immunol Rev 2023; 316:160-175. [PMID: 37098109 PMCID: PMC10952287 DOI: 10.1111/imr.13206] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2023] [Revised: 03/20/2023] [Accepted: 04/01/2023] [Indexed: 04/27/2023]
Abstract
As the thymus involutes during aging, the T-cell pool has to be maintained by the periodic expansion of preexisting T cells during adulthood. A conundrum is that repeated episodes of activation and proliferation drive the differentiation of T cells toward replicative senescence, due to telomere erosion. This review discusses mechanisms that regulate the end-stage differentiation (senescence) of T cells. Although these cells, within both CD4 and CD8 compartments, lose proliferative activity after antigen-specific challenge, they acquire innate-like immune function. While this may confer broad immune protection during aging, these senescent T cells may also cause immunopathology, especially in the context of excessive inflammation in tissue microenvironments.
Collapse
Affiliation(s)
- Phatthamon Laphanuwat
- Division of MedicineUniversity College LondonLondonUK
- Department of PharmacologyFaculty of Medicine, Khon Kaen UniversityKhon KaenThailand
| | - Daniel Claudio Oliveira Gomes
- Division of MedicineUniversity College LondonLondonUK
- Núcleo de Doenças InfecciosasUniversidade Federal do Espírito SantoVitoriaBrazil
- Núcleo de BiotecnologiaUniversidade Federal do Espírito SantoVitoriaBrazil
| | - Arne N. Akbar
- Division of MedicineUniversity College LondonLondonUK
| |
Collapse
|
34
|
Klotz L, Antel J, Kuhlmann T. Inflammation in multiple sclerosis: consequences for remyelination and disease progression. Nat Rev Neurol 2023; 19:305-320. [PMID: 37059811 DOI: 10.1038/s41582-023-00801-6] [Citation(s) in RCA: 45] [Impact Index Per Article: 22.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/15/2023] [Indexed: 04/16/2023]
Abstract
Despite the large number of immunomodulatory or immunosuppressive treatments available to treat relapsing-remitting multiple sclerosis (MS), treatment of the progressive phase of the disease has not yet been achieved. This lack of successful treatment approaches is caused by our poor understanding of the mechanisms driving disease progression. Emerging concepts suggest that a combination of persisting focal and diffuse inflammation within the CNS and a gradual failure of compensatory mechanisms, including remyelination, result in disease progression. Therefore, promotion of remyelination presents a promising intervention approach. However, despite our increasing knowledge regarding the cellular and molecular mechanisms regulating remyelination in animal models, therapeutic increases in remyelination remain an unmet need in MS, which suggests that mechanisms of remyelination and remyelination failure differ fundamentally between humans and demyelinating animal models. New and emerging technologies now allow us to investigate the cellular and molecular mechanisms underlying remyelination failure in human tissue samples in an unprecedented way. The aim of this Review is to summarize our current knowledge regarding mechanisms of remyelination and remyelination failure in MS and in animal models of the disease, identify open questions, challenge existing concepts, and discuss strategies to overcome the translational roadblock in the field of remyelination-promoting therapies.
Collapse
Affiliation(s)
- Luisa Klotz
- Department of Neurology with Institute of Translational Neurology, University Hospital Münster, Münster, Germany
| | - Jack Antel
- Neuroimmunology Unit, Montreal Neurological Institute, McGill University, Québec, Canada
| | - Tanja Kuhlmann
- Neuroimmunology Unit, Montreal Neurological Institute, McGill University, Québec, Canada.
- Institute of Neuropathology, University Hospital Münster, Münster, Germany.
| |
Collapse
|
35
|
Kakuda T, Suzuki J, Matsuoka Y, Kikugawa T, Saika T, Yamashita M. Senescent CD8 + T cells acquire NK cell-like innate functions to promote antitumor immunity. Cancer Sci 2023. [PMID: 37186472 DOI: 10.1111/cas.15824] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2022] [Revised: 04/03/2023] [Accepted: 04/06/2023] [Indexed: 05/17/2023] Open
Abstract
It has been suggested that aging of the immune system (immunosenescence) results in a decline in the acquired immune response, which is associated with an increase in age-related tumorigenesis. T-cell senescence plays a critical role in immunosenescence and is involved in the age-related decline of the immune function, which increases susceptibility to certain cancers. However, it has been shown that CD8+ T cells with the senescent T-cell phenotype acquire an natural killer (NK) cell-like function and are involved in tumor elimination. Therefore, the role of senescent CD8+ T cells in tumor immunity remains to be elucidated. In this study, we investigated the role of senescent CD8+ T cells in tumor immunity. In a murine model of transferred with B16 melanoma, lung metastasis was significantly suppressed in aged mice (age ≥30 weeks) in comparison to young mice (age 6-10 weeks). We evaluated the cytotoxic activity of CD8+ T cells in vitro and found that CD8+ T cells from aged mice activated in vitro exhibited increased cytotoxic activity in comparison to those from young mice. We used Menin-deficient effector T cells as a model for senescent CD8+ T cells and found that cytotoxic activity and the expression of NK receptors were upregulated in Menin-deficient senescent CD8+ T cells. Furthermore, Menin-deficient CD8+ T cells can eliminate tumor cells in an antigen-independent manner. These results suggest that senescent effector CD8+ T cells may contribute to tumor immunity in the elderly by acquiring NK-like innate immune functions, such as antigen-independent cytotoxic activity.
Collapse
Affiliation(s)
- Toshio Kakuda
- Department of Urologye, Graduate School of Medicin, Ehime University, Toon, Japan
- Department of Immunology, Graduate School of Medicine, Ehime University, Toon, Japan
| | - Junpei Suzuki
- Department of Immunology, Graduate School of Medicine, Ehime University, Toon, Japan
| | - Yuko Matsuoka
- Translational Research Center, Ehime University Hospital, Ehime University, Toon, Japan
| | - Tadahiko Kikugawa
- Department of Urologye, Graduate School of Medicin, Ehime University, Toon, Japan
| | - Takashi Saika
- Department of Urologye, Graduate School of Medicin, Ehime University, Toon, Japan
| | - Masakatsu Yamashita
- Department of Immunology, Graduate School of Medicine, Ehime University, Toon, Japan
| |
Collapse
|
36
|
Liu Z, Zhang J, Liu H, Shen H, Meng N, Qi X, Ding K, Song J, Fu R, Ding D, Feng G. BSA-AIE Nanoparticles with Boosted ROS Generation for Immunogenic Cell Death Immunotherapy of Multiple Myeloma. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2023; 35:e2208692. [PMID: 36529696 DOI: 10.1002/adma.202208692] [Citation(s) in RCA: 40] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/21/2022] [Revised: 11/15/2022] [Indexed: 06/17/2023]
Abstract
The main obstacle of multiple myeloma (MM) therapy is the compromised immune microenvironment, which leads to MM relapses and extramedullary disease progression. In this study, a novel strategy is reported of enhanced immunogenic cell death (ICD) immunotherapy with aggregation-induced emission (AIE) photosensitizer-loaded bovine serum albumin (BSA) nanoparticles (referred as BSA/TPA-Erdn), which can activate T cells, convert the cold tumor to hot, and reverse T cell senescence to restore the immune microenvironment for MM treatment. Loading AIE photosensitizer into the hydrophobic domain of BSA proteins significantly immobilizes the molecular geometry, which massively increases reactive oxygen species (ROS) generation and elicits a promising ICD immune response. Employing a NOD-SCID IL-2receptor gamma null mice model with MM patients' monocytes, it is shown that BSA/TPA-Erdn can simulate human dentric cell maturation, activate functional T lymphocytes, and increase additional polarization and differentiation signals to deliver a promising immunotherapy performance. Intriguingly, for the first time, it is shown that BSA/TPA-Erdn can greatly reverse T cell senescence, a main challenge in treating MM. Additionally, BSA/TPA-Erdn can effectively recruit more functional T lymphocytes into MM tumor. As a consequence, BSA/TPA-Erdn restores MM immune microenvironment and shows the best MM tumor eradication performance, which shall pave new insights for MM treatment in clinical practices.
Collapse
Affiliation(s)
- Zhaoyun Liu
- Department of Hematology, Tianjin Medical University General Hospital, Tianjin, 300052, China
| | - Jingtian Zhang
- State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Bioactive, Materials, Ministry of Education, and College of Life Sciences, Nankai University, Tianjin, 300071, China
| | - Hui Liu
- Department of Hematology, Tianjin Medical University General Hospital, Tianjin, 300052, China
| | - Hongli Shen
- Department of Hematology, Tianjin Medical University General Hospital, Tianjin, 300052, China
| | - Nanhao Meng
- Department of Hematology, Tianjin Medical University General Hospital, Tianjin, 300052, China
| | - Xinwen Qi
- State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Bioactive, Materials, Ministry of Education, and College of Life Sciences, Nankai University, Tianjin, 300071, China
| | - Kai Ding
- Department of Hematology, Tianjin Medical University General Hospital, Tianjin, 300052, China
| | - Jia Song
- Department of Hematology, Tianjin Medical University General Hospital, Tianjin, 300052, China
| | - Rong Fu
- Department of Hematology, Tianjin Medical University General Hospital, Tianjin, 300052, China
| | - Dan Ding
- State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Bioactive, Materials, Ministry of Education, and College of Life Sciences, Nankai University, Tianjin, 300071, China
| | - Guangxue Feng
- State Key Laboratory of Luminescent Materials and Devices, Guangdong Provincial Key Laboratory of Luminescence from Molecular Aggregates, AIE Institute, School of Materials Science and Engineering, South China University of Technology, Guangzhou, 510640, China
| |
Collapse
|
37
|
Kang YE, Yi HS, Yeo MK, Kim JT, Park D, Jung Y, Kim OS, Lee SE, Kim JM, Joung KH, Lee JH, Ku BJ, Lee M, Kim HJ. Increased Pro-Inflammatory T Cells, Senescent T Cells, and Immune-Check Point Molecules in the Placentas of Patients With Gestational Diabetes Mellitus. J Korean Med Sci 2022; 37:e338. [PMID: 36513052 PMCID: PMC9745681 DOI: 10.3346/jkms.2022.37.e338] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/22/2022] [Accepted: 09/22/2022] [Indexed: 11/18/2022] Open
Abstract
BACKGROUND Gestational diabetes mellitus (GDM) is the most common metabolic complication of pregnancy. To define the altered pathway in GDM placenta, we investigated the transcriptomic profiles from human placenta between GDM and controls. METHODS Clinical parameters and postpartum complications were reviewed in all participants. Differentially expressed canonical pathways were analyzed between the GDM and control groups based on transcriptomic analysis. CD4+ T, CD8+ T, and senescent T cell subsets were determined by flow cytometry based on staining for specific intracellular cytokines. RESULTS Gene ontology analysis revealed that the placenta of GDM revealed upregulation of diverse mitochondria or DNA replication related pathways and downregulation of T-cell immunity related pathways. The maternal placenta of the GDM group had a higher proportion of CD4+ T and CD8+ T cells than the control group. Interestingly, senescent CD4+ T cells tended to increase and CD8+ T cells were significantly increased in GDM compared to controls, along with increased programmed cell death-1 (CD274+) expression. Programmed death-ligand 1 expression in syncytotrophoblasts was also significantly increased in patients with GDM. CONCLUSION This study demonstrated increased proinflammatory T cells, senescent T cells and immune-check point molecules in GDM placentas, suggesting that changes in senescent T cells and immune-escape signaling might be related to the pathophysiology of GDM.
Collapse
Affiliation(s)
- Yea Eun Kang
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Chungnam National University College of Medicine, Daejeon, Korea
| | - Hyon-Seung Yi
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Chungnam National University College of Medicine, Daejeon, Korea
- Laboratory of Endocrinology and Immune System, Chungnam National University College of Medicine, Daejeon, Korea
| | - Min-Kyung Yeo
- Department of Pathology, Chungnam National University College of Medicine, Daejeon, Korea
| | - Jung Tae Kim
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Chungnam National University College of Medicine, Daejeon, Korea
| | - Danbit Park
- Department of Obstetrics and Gynecology, Chungnam National University Hospital, Daejeon, Korea
| | - Yewon Jung
- Department of Obstetrics and Gynecology, Chungnam National University Sejong Hospital, Sejong, Korea
| | - Ok Soon Kim
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Chungnam National University College of Medicine, Daejeon, Korea
| | - Seong Eun Lee
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Chungnam National University College of Medicine, Daejeon, Korea
| | - Ji Min Kim
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Chungnam National University College of Medicine, Daejeon, Korea
| | - Kyong Hye Joung
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Chungnam National University College of Medicine, Daejeon, Korea
| | - Ju Hee Lee
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Chungnam National University College of Medicine, Daejeon, Korea
| | - Bon Jeong Ku
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Chungnam National University College of Medicine, Daejeon, Korea
| | - Mina Lee
- Department of Obstetrics and Gynecology, Chungnam National University College of Medicine, Daejeon, Korea.
| | - Hyun Jin Kim
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Chungnam National University College of Medicine, Daejeon, Korea.
| |
Collapse
|
38
|
Foster MA, Bentley C, Hazeldine J, Acharjee A, Nahman O, Shen-Orr SS, Lord JM, Duggal NA. Investigating the potential of a prematurely aged immune phenotype in severely injured patients as predictor of risk of sepsis. Immun Ageing 2022; 19:60. [PMID: 36471343 PMCID: PMC9720981 DOI: 10.1186/s12979-022-00317-5] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2022] [Accepted: 11/24/2022] [Indexed: 12/07/2022]
Abstract
BACKGROUND Traumatic injury elicits a hyperinflammatory response and remodelling of the immune system leading to immuneparesis. This study aimed to evaluate whether traumatic injury results in a state of prematurely aged immune phenotype to relate this to clinical outcomes and a greater risk of developing additional morbidities post-injury. METHODS AND FINDINGS Blood samples were collected from 57 critically injured patients with a mean Injury Severity Score (ISS) of 26 (range 15-75 years), mean age of 39.67 years (range 20-84 years), and 80.7% males, at days 3, 14, 28 and 60 post-hospital admission. 55 healthy controls (HC), mean age 40.57 years (range 20-85 years), 89.7% males were also recruited. The phenotype and frequency of adaptive immune cells were used to calculate the IMM-AGE score, an indicator of the degree of phenotypic ageing of the immune system. IMM-AGE was elevated in trauma patients at an early timepoint (day 3) in comparison with healthy controls (p < 0.001), driven by an increase in senescent CD8 T cells (p < 0.0001), memory CD8 T cells (p < 0.0001) and regulatory T cells (p < 0.0001) and a reduction in naïve CD8 T cells (p < 0.001) and overall T cell lymphopenia (p < 0 .0001). These changes persisted to day 60. Furthermore, the IMM-AGE scores were significantly higher in trauma patients (mean score 0.72) that developed sepsis (p = 0.05) in comparison with those (mean score 0.61) that did not. CONCLUSIONS The profoundly altered peripheral adaptive immune compartment after critical injury can be used as a potential biomarker to identify individuals at a high risk of developing sepsis and this state of prematurely aged immune phenotype in biologically young individuals persists for up to two months post-hospitalisation, compromising the host immune response to infections. Reversing this aged immune system is likely to have a beneficial impact on short- and longer-term outcomes of trauma survivors.
Collapse
Affiliation(s)
- Mark A Foster
- NIHR-Surgical Reconstruction and Microbiology Research Centre, Queen Elizabeth Hospital Birmingham, University Hospitals Birmingham NHS Foundation Trust, Birmingham, UK
- Royal Centre for Defence Medicine, Queen Elizabeth Hospital Birmingham, Birmingham, UK
| | - Conor Bentley
- NIHR-Surgical Reconstruction and Microbiology Research Centre, Queen Elizabeth Hospital Birmingham, University Hospitals Birmingham NHS Foundation Trust, Birmingham, UK
- Royal Centre for Defence Medicine, Queen Elizabeth Hospital Birmingham, Birmingham, UK
| | - Jon Hazeldine
- MRC-Versus Arthritis Centre for Musculoskeletal Ageing Research, Institute of Inflammation and Ageing, University of Birmingham, Birmingham, UK
| | - Animesh Acharjee
- Institute of Cancer and Genomic Sciences, Centre for Computational Biology, University of Birmingham, Birmingham, B15 2TT, UK
| | - Ornit Nahman
- Faculty of Medicine, Technion - Israel Institute of Technology, Haifa, Israel
| | - Shai S Shen-Orr
- Faculty of Medicine, Technion - Israel Institute of Technology, Haifa, Israel
| | - Janet M Lord
- MRC-Versus Arthritis Centre for Musculoskeletal Ageing Research, Institute of Inflammation and Ageing, University of Birmingham, Birmingham, UK
| | - Niharika A Duggal
- MRC-Versus Arthritis Centre for Musculoskeletal Ageing Research, Institute of Inflammation and Ageing, University of Birmingham, Birmingham, UK.
| |
Collapse
|
39
|
Abstract
PURPOSE OF REVIEW Systemic sclerosis (SSc) is a chronic rheumatic disease that is characterized by immune activation, vasculopathy and fibrosis of the skin and internal organs. It has been proposed that premature onset of ageing pathways and associated senescent changes in cells contribute to the clinical and pathological features of SSc. The aim of this review is to critically review recent insights into the involvement of cellular senescence in SSc. RECENT FINDINGS Cellular senescence plays a critical role in SSc pathogenesis, particularly involving endothelial cells and fibroblasts. Immunosenescence could also contribute to SSc pathogenesis by direct alteration of cellular functions or indirect promotion of defective immune surveillance. Molecular studies have shed some light on how cellular senescence contributes to fibrosis. Recent and planned proof-of-concept trials using senotherapeutics showed promising results in fibrotic diseases, including SSc. SUMMARY There is increasing evidence implicating cellular senescence in SSc. The mechanisms underlying premature cellular senescence in SSc, and its potential role in pathogenesis, merit further investigation. Emerging drugs targeting senescence-related pathways might be potential therapeutic options for SSc.
Collapse
Affiliation(s)
- Pei-Suen Tsou
- Division of Rheumatology, Department of Internal Medicine, University of Michigan, Ann Arbor, MI, USA
- Scleroderma Program, University of Michigan, Ann Arbor, MI, USA
| | - Bo Shi
- Department of Dermatology, Northwestern University, Chicago, IL, USA
| | - John Varga
- Division of Rheumatology, Department of Internal Medicine, University of Michigan, Ann Arbor, MI, USA
- Scleroderma Program, University of Michigan, Ann Arbor, MI, USA
| |
Collapse
|
40
|
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
|
41
|
Luís C, Maduro AT, Pereira P, Mendes JJ, Soares R, Ramalho R. Nutritional senolytics and senomorphics: Implications to immune cells metabolism and aging – from theory to practice. Front Nutr 2022; 9:958563. [PMID: 36159455 PMCID: PMC9493043 DOI: 10.3389/fnut.2022.958563] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Accepted: 08/16/2022] [Indexed: 11/28/2022] Open
Abstract
Aging is a natural physiological process, but one that poses major challenges in an increasingly aging society prone to greater health risks such as diabetes, cardiovascular disease, cancer, frailty, increased susceptibility to infection, and reduced response to vaccine regimens. The loss of capacity for cell regeneration and the surrounding tissue microenvironment itself is conditioned by genetic, metabolic, and even environmental factors, such as nutrition. The senescence of the immune system (immunosenescence) represents a challenge, especially when associated with the presence of age-related chronic inflammation (inflammaging) and affecting the metabolic programming of immune cells (immunometabolism). These aspects are linked to poorer health outcomes and therefore present an opportunity for host-directed interventions aimed at both eliminating senescent cells and curbing the underlying inflammation. Senotherapeutics are a class of drugs and natural products that delay, prevent, or reverse the senescence process – senolytics; or inhibit senescence-associated secretory phenotype – senomorphics. Natural senotherapeutics from food sources – nutritional senotherapeutics – may constitute an interesting way to achieve better age-associated outcomes through personalized nutrition. In this sense, the authors present herein a framework of nutritional senotherapeutics as an intervention targeting immunosenescence and immunometabolism, identifying research gaps in this area, and gathering information on concluded and ongoing clinical trials on this subject. Also, we present future directions and ideation for future clinical possibilities in this field.
Collapse
Affiliation(s)
- Carla Luís
- Department of Biomedicine, Faculdade de Medicina da Universidade do Porto, Porto, Portugal
- i3S – Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal
| | - Ana T. Maduro
- Department of Biomedicine, Faculdade de Medicina da Universidade do Porto, Porto, Portugal
- i3S – Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal
| | - Paula Pereira
- Nutritional Immunology – Clinical and Experimental Lab (NICE Lab), Clinical Research Unit, Centro de Investigação Interdisciplinar Egas Moniz (CiiEM, U4585 FCT), Egas Moniz Higher Education School, Monte de Caparica, Portugal
- Applied Nutrition Study Group (Grupo de Estudos em Nutrição Aplicada – G.E.N.A.-IUEM), Egas Moniz Higher Education School, Monte de Caparica, Portugal
- Instituto Universitário Egas Moniz, Egas Moniz Higher Education School, Monte de Caparica, Portugal
| | - José João Mendes
- Nutritional Immunology – Clinical and Experimental Lab (NICE Lab), Clinical Research Unit, Centro de Investigação Interdisciplinar Egas Moniz (CiiEM, U4585 FCT), Egas Moniz Higher Education School, Monte de Caparica, Portugal
- Instituto Universitário Egas Moniz, Egas Moniz Higher Education School, Monte de Caparica, Portugal
| | - Raquel Soares
- Department of Biomedicine, Faculdade de Medicina da Universidade do Porto, Porto, Portugal
- i3S – Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal
| | - Renata Ramalho
- Nutritional Immunology – Clinical and Experimental Lab (NICE Lab), Clinical Research Unit, Centro de Investigação Interdisciplinar Egas Moniz (CiiEM, U4585 FCT), Egas Moniz Higher Education School, Monte de Caparica, Portugal
- Applied Nutrition Study Group (Grupo de Estudos em Nutrição Aplicada – G.E.N.A.-IUEM), Egas Moniz Higher Education School, Monte de Caparica, Portugal
- Instituto Universitário Egas Moniz, Egas Moniz Higher Education School, Monte de Caparica, Portugal
- *Correspondence: Renata Ramalho,
| |
Collapse
|
42
|
Zhao J, Guo S, Schrodi SJ, He D. Cuproptosis and cuproptosis-related genes in rheumatoid arthritis: Implication, prospects, and perspectives. Front Immunol 2022; 13:930278. [PMID: 35990673 PMCID: PMC9386151 DOI: 10.3389/fimmu.2022.930278] [Citation(s) in RCA: 35] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2022] [Accepted: 07/18/2022] [Indexed: 11/14/2022] Open
Abstract
Rheumatoid arthritis (RA) is an autoimmune disease that severely affects patients' physical and mental health, leading to chronic synovitis and destruction of bone joints. Although various available clinical treatment options exist, patients respond with varying efficacies due to multiple factors, and there is an urgent need to discover new treatment options to improve clinical outcomes. Cuproptosis is a newly characterized form of cell death. Copper causes cuproptosis by binding to lipid-acylated components of the tricarboxylic acid cycle, leading to protein aggregation, loss of iron-sulfur cluster proteins, and eventually proteotoxic stress. Targeting copper cytotoxicity and cuproptosis are considered potential options for treating oncological diseases. The synovial hypoxic environment and the presence of excessive glycolysis in multiple cells appear to act as inhibitors of cuproptosis, which can lead to excessive survival and proliferation of multiple immune cells, such as fibroblast-like synoviocytes, effector T cells, and macrophages, further mediating inflammation and bone destruction in RA. Therefore, in this study, we attempted to elaborate and summarize the linkage of cuproptosis and key genes regulating cuproptosis to the pathological mechanisms of RA and their effects on a variety of immune cells. This study aimed to provide a theoretical basis and support for translating preclinical and experimental results of RA to clinical protocols.
Collapse
Affiliation(s)
- Jianan Zhao
- Department of Rheumatology, Shanghai Guanghua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Guanghua Clinical Medical College, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Institute of Arthritis Research in Integrative Medicine, Shanghai Academy of Traditional Chinese Medicine, Shanghai, China
| | - Shicheng Guo
- Computation and Informatics in Biology and Medicine, University of Wisconsin-Madison, Madison, WI, United States
- Department of Medical Genetics, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, United States
| | - Steven J. Schrodi
- Computation and Informatics in Biology and Medicine, University of Wisconsin-Madison, Madison, WI, United States
- Department of Medical Genetics, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, United States
| | - Dongyi He
- Department of Rheumatology, Shanghai Guanghua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Guanghua Clinical Medical College, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Institute of Arthritis Research in Integrative Medicine, Shanghai Academy of Traditional Chinese Medicine, Shanghai, China
- Arthritis Institute of Integrated Traditional and Western Medicine, Shanghai Chinese Medicine Research Institute, Shanghai, China
| |
Collapse
|
43
|
Chaib S, Tchkonia T, Kirkland JL. Cellular senescence and senolytics: the path to the clinic. Nat Med 2022; 28:1556-1568. [PMID: 35953721 PMCID: PMC9599677 DOI: 10.1038/s41591-022-01923-y] [Citation(s) in RCA: 516] [Impact Index Per Article: 172.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2022] [Accepted: 06/28/2022] [Indexed: 01/10/2023]
Abstract
Interlinked and fundamental aging processes appear to be a root-cause contributor to many disorders and diseases. One such process is cellular senescence, which entails a state of cell cycle arrest in response to damaging stimuli. Senescent cells can arise throughout the lifespan and, if persistent, can have deleterious effects on tissue function due to the many proteins they secrete. In preclinical models, interventions targeting those senescent cells that are persistent and cause tissue damage have been shown to delay, prevent or alleviate multiple disorders. In line with this, the discovery of small-molecule senolytic drugs that selectively clear senescent cells has led to promising strategies for preventing or treating multiple diseases and age-related conditions in humans. In this Review, we outline the rationale for senescent cells as a therapeutic target for disorders across the lifespan and discuss the most promising strategies-including recent and ongoing clinical trials-for translating small-molecule senolytics and other senescence-targeting interventions into clinical use.
Collapse
Affiliation(s)
- Selim Chaib
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN, USA
| | - Tamar Tchkonia
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN, USA
| | - James L Kirkland
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN, USA.
- Division of General Internal Medicine, Department of Medicine, Mayo Clinic, Rochester, MN, USA.
| |
Collapse
|
44
|
Walker KA, Basisty N, Wilson DM, Ferrucci L. Connecting aging biology and inflammation in the omics era. J Clin Invest 2022; 132:e158448. [PMID: 35838044 PMCID: PMC9282936 DOI: 10.1172/jci158448] [Citation(s) in RCA: 83] [Impact Index Per Article: 27.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
Aging is characterized by the accumulation of damage to macromolecules and cell architecture that triggers a proinflammatory state in blood and solid tissues, termed inflammaging. Inflammaging has been implicated in the pathogenesis of many age-associated chronic diseases as well as loss of physical and cognitive function. The search for mechanisms that underlie inflammaging focused initially on the hallmarks of aging, but it is rapidly expanding in multiple directions. Here, we discuss the threads connecting cellular senescence and mitochondrial dysfunction to impaired mitophagy and DNA damage, which may act as a hub for inflammaging. We explore the emerging multi-omics efforts that aspire to define the complexity of inflammaging - and identify molecular signatures and novel targets for interventions aimed at counteracting excessive inflammation and its deleterious consequences while preserving the physiological immune response. Finally, we review the emerging evidence that inflammation is involved in brain aging and neurodegenerative diseases. Our goal is to broaden the research agenda for inflammaging with an eye on new therapeutic opportunities.
Collapse
Affiliation(s)
- Keenan A. Walker
- Intramural Research Program of the National Institute on Aging, NIH, Baltimore, Maryland, USA
| | - Nathan Basisty
- Intramural Research Program of the National Institute on Aging, NIH, Baltimore, Maryland, USA
| | - David M. Wilson
- Biomedical Research Institute, Faculty of Medicine and Life Sciences, Hasselt University, Diepenbeek, Belgium
| | - Luigi Ferrucci
- Intramural Research Program of the National Institute on Aging, NIH, Baltimore, Maryland, USA
| |
Collapse
|
45
|
Damian L, Login CC, Solomon C, Belizna C, Encica S, Urian L, Jurcut C, Stancu B, Vulturar R. Inclusion Body Myositis and Neoplasia: A Narrative Review. Int J Mol Sci 2022; 23:ijms23137358. [PMID: 35806366 PMCID: PMC9266341 DOI: 10.3390/ijms23137358] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2022] [Revised: 06/28/2022] [Accepted: 06/28/2022] [Indexed: 02/04/2023] Open
Abstract
Inclusion body myositis (IBM) is an acquired, late-onset inflammatory myopathy, with both inflammatory and degenerative pathogenesis. Although idiopathic inflammatory myopathies may be associated with malignancies, IBM is generally not considered paraneoplastic. Many studies of malignancy in inflammatory myopathies did not include IBM patients. Indeed, IBM is often diagnosed only after around 5 years from onset, while paraneoplastic myositis is generally defined as the co-occurrence of malignancy and myopathy within 1 to 3 years of each other. Nevertheless, a significant association with large granular lymphocyte leukemia has been recently described in IBM, and there are reports of cancer-associated IBM. We review the pathogenic mechanisms supposed to be involved in IBM and outline the common mechanisms in IBM and malignancy, as well as the therapeutic perspectives. The terminally differentiated, CD8+ highly cytotoxic T cells expressing NK features are central in the pathogenesis of IBM and, paradoxically, play a role in some cancers as well. Interferon gamma plays a central role, mostly during the early stages of the disease. The secondary mitochondrial dysfunction, the autophagy and cell cycle dysregulation, and the crosstalk between metabolic and mitogenic pathways could be shared by IBM and cancer. There are intermingled subcellular mechanisms in IBM and neoplasia, and probably their co-existence is underestimated. The link between IBM and cancers deserves further interest, in order to search for efficient therapies in IBM and to improve muscle function, life quality, and survival in both diseases.
Collapse
Affiliation(s)
- Laura Damian
- Centre for Rare Autoimmune and Autoinflammatory Diseases (ERN-ReCONNET), Department of Rheumatology, Emergency Clinical County Hospital Cluj, 400347 Cluj-Napoca, Romania;
- CMI Reumatologie Dr. Damian, 6-8 Petru Maior St., 400002 Cluj-Napoca, Romania
| | - Cristian Cezar Login
- Department of Physiology, “Iuliu Hațieganu” University of Medicine and Pharmacy, 400006 Cluj-Napoca, Romania
- Correspondence:
| | - Carolina Solomon
- Radiology Department, “Iuliu Hațieganu” University of Medicine and Pharmacy, 400006 Cluj-Napoca, Romania;
- Radiology Department, Emergency Clinical County Hospital Cluj, 400006 Cluj-Napoca, Romania
| | - Cristina Belizna
- UMR CNRS 6015—INSERM U1083, University of Angers, 49100 Angers, France;
- Internal Medicine Department Clinique de l’Anjou, Angers and Vascular and Coagulation Department, University Hospital Angers, 49100 Angers, France
| | - Svetlana Encica
- Department of Pathology, “Niculae Stancioiu” Heart Institute Cluj-Napoca, 19-21 Calea Moților St., 400001 Cluj-Napoca, Romania;
| | - Laura Urian
- Department of Hematology, “Iuliu Hațieganu” University of Medicine and Pharmacy, 400004 Cluj-Napoca, Romania;
- Department of Hematology, Ion Chiricuta Clinical Cancer Center, 400014 Cluj-Napoca, Romania
| | - Ciprian Jurcut
- Department of Internal Medicine, “Carol Davila” Central Military Emergency University Hospital, Calea Plevnei No 134, 010825 Bucharest, Romania;
| | - Bogdan Stancu
- 2nd Surgical Department, “Iuliu Hațieganu” University of Medicine and Pharmacy, 400012 Cluj-Napoca, Romania;
| | - Romana Vulturar
- Department of Molecular Sciences, “Iuliu Hațieganu” University of Medicine and Pharmacy, 400349 Cluj-Napoca, Romania;
- Cognitive Neuroscience Laboratory, University “Babes-Bolyai” Cluj-Napoca, 400294 Cluj-Napoca, Romania
| |
Collapse
|
46
|
Wagner KD, Wagner N. The Senescence Markers p16INK4A, p14ARF/p19ARF, and p21 in Organ Development and Homeostasis. Cells 2022; 11:cells11121966. [PMID: 35741095 PMCID: PMC9221567 DOI: 10.3390/cells11121966] [Citation(s) in RCA: 68] [Impact Index Per Article: 22.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2022] [Revised: 06/15/2022] [Accepted: 06/15/2022] [Indexed: 02/07/2023] Open
Abstract
It is widely accepted that senescent cells accumulate with aging. They are characterized by replicative arrest and the release of a myriad of factors commonly called the senescence-associated secretory phenotype. Despite the replicative cell cycle arrest, these cells are metabolically active and functional. The release of SASP factors is mostly thought to cause tissue dysfunction and to induce senescence in surrounding cells. As major markers for aging and senescence, p16INK4, p14ARF/p19ARF, and p21 are established. Importantly, senescence is also implicated in development, cancer, and tissue homeostasis. While many markers of senescence have been identified, none are able to unambiguously identify all senescent cells. However, increased levels of the cyclin-dependent kinase inhibitors p16INK4A and p21 are often used to identify cells with senescence-associated phenotypes. We review here the knowledge of senescence, p16INK4A, p14ARF/p19ARF, and p21 in embryonic and postnatal development and potential functions in pathophysiology and homeostasis. The establishment of senolytic therapies with the ultimate goal to improve healthy aging requires care and detailed knowledge about the involvement of senescence and senescence-associated proteins in developmental processes and homeostatic mechanism. The review contributes to these topics, summarizes open questions, and provides some directions for future research.
Collapse
|
47
|
Hu H, Tang N, Zhang F, Li L, Li L. Bioinformatics and System Biology Approach to Identify the Influences of COVID-19 on Rheumatoid Arthritis. Front Immunol 2022; 13:860676. [PMID: 35464423 PMCID: PMC9021444 DOI: 10.3389/fimmu.2022.860676] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2022] [Accepted: 03/16/2022] [Indexed: 02/05/2023] Open
Abstract
Background Severe coronavirus disease 2019 (COVID -19) has led to a rapid increase in mortality worldwide. Rheumatoid arthritis (RA) was a high-risk factor for severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection, whereas the molecular mechanisms underlying RA and CVOID-19 are not well understood. The objectives of this study were to analyze potential molecular mechanisms and identify potential drugs for the treatment of COVID-19 and RA using bioinformatics and a systems biology approach. Methods Two Differentially expressed genes (DEGs) sets extracted from GSE171110 and GSE1775544 datasets were intersected to generate common DEGs, which were used for functional enrichment, pathway analysis, and candidate drugs analysis. Results A total of 103 common DEGs were identified in the two datasets between RA and COVID-19. A protein-protein interaction (PPI) was constructed using various combinatorial statistical methods and bioinformatics tools. Subsequently, hub genes and essential modules were identified from the PPI network. In addition, we performed functional analysis and pathway analysis under ontological conditions and found that there was common association between RA and progression of COVID-19 infection. Finally, transcription factor-gene interactions, protein-drug interactions, and DEGs-miRNAs coregulatory networks with common DEGs were also identified in the datasets. Conclusion We successfully identified the top 10 hub genes that could serve as novel targeted therapy for COVID-19 and screened out some potential drugs useful for COVID-19 patients with RA.
Collapse
Affiliation(s)
- Huan Hu
- Department of Rheumatology and Immunology, The Affiliated Hospital of Guizhou Medical University, Guiyang, China.,Clinical Medical College, Guizhou Medical University, Guiyang, China
| | - Nana Tang
- Medical Intensive Care Unit, The Affiliated Hospital of Guizhou Medical University, Guiyang, China
| | - Facai Zhang
- Department of Urology/Institute of Urology, West China Hospital, Sichuan University, Chengdu, China
| | - Li Li
- Medical Intensive Care Unit, The Affiliated Hospital of Guizhou Medical University, Guiyang, China
| | - Long Li
- Department of Rheumatology and Immunology, The Affiliated Hospital of Guizhou Medical University, Guiyang, China
| |
Collapse
|
48
|
Hu C, Zhang X, Teng T, Ma ZG, Tang QZ. Cellular Senescence in Cardiovascular Diseases: A Systematic Review. Aging Dis 2022; 13:103-128. [PMID: 35111365 PMCID: PMC8782554 DOI: 10.14336/ad.2021.0927] [Citation(s) in RCA: 87] [Impact Index Per Article: 29.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2021] [Accepted: 09/27/2021] [Indexed: 12/12/2022] Open
Abstract
Aging is a prominent risk factor for cardiovascular diseases, which is the leading cause of death around the world. Recently, cellular senescence has received potential attention as a promising target in preventing cardiovascular diseases, including acute myocardial infarction, atherosclerosis, cardiac aging, pressure overload-induced hypertrophy, heart regeneration, hypertension, and abdominal aortic aneurysm. Here, we discuss the mechanisms underlying cellular senescence and describe the involvement of senescent cardiovascular cells (including cardiomyocytes, endothelial cells, vascular smooth muscle cells, fibroblasts/myofibroblasts and T cells) in age-related cardiovascular diseases. Then, we highlight the targets (SIRT1 and mTOR) that regulating cellular senescence in cardiovascular disorders. Furthermore, we review the evidence that senescent cells can exert both beneficial and detrimental implications in cardiovascular diseases on a context-dependent manner. Finally, we summarize the emerging pro-senescent or anti-senescent interventions and discuss their therapeutic potential in preventing cardiovascular diseases.
Collapse
Affiliation(s)
- Can Hu
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan 430060, China
- Hubei Key Laboratory of Metabolic and Chronic Diseases, Wuhan 430060, China
| | - Xin Zhang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan 430060, China
- Hubei Key Laboratory of Metabolic and Chronic Diseases, Wuhan 430060, China
| | - Teng Teng
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan 430060, China
- Hubei Key Laboratory of Metabolic and Chronic Diseases, Wuhan 430060, China
| | - Zhen-Guo Ma
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan 430060, China
- Hubei Key Laboratory of Metabolic and Chronic Diseases, Wuhan 430060, China
| | - Qi-Zhu Tang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan 430060, China
- Hubei Key Laboratory of Metabolic and Chronic Diseases, Wuhan 430060, China
| |
Collapse
|
49
|
Liisborg C, Skov V, Kjær L, Hasselbalch HC, Sørensen TL. Retinal drusen in patients with chronic myeloproliferative blood cancers are associated with an increased proportion of senescent T cells and signs of an aging immune system. Aging (Albany NY) 2021; 13:25763-25777. [PMID: 34954692 PMCID: PMC8751607 DOI: 10.18632/aging.203803] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2021] [Accepted: 12/13/2021] [Indexed: 12/13/2022]
Abstract
The cause of age-related macular degeneration (AMD) is unknown, but evidence indicates that both innate and adaptive immunity play a role in the pathogenesis. Our recent work has investigated AMD in patients with myeloproliferative neoplasms (MPNs) since they have increased drusen and AMD prevalence. We have previously found increased levels of chronic low-grade inflammation (CLI) in MPN patients with drusen (MPNd) compared to MPN patients with normal retinas (MPNn). CLI and AMD are both associated with aging, and we, therefore, wanted to study immunosenescence markers in MPNd, MPNn, and AMD. The purpose was to identify differences between MPNd and MPNn, which might reveal novel information relevant to drusen pathophysiology and thereby the AMD pathogenesis. Our results suggest that MPNd have a T cell differentiation profile resembling AMD and more effector memory T cells than MPNn. The senescence-associated-secretory-phenotype (SASP) is associated with effector T cells. SASP is thought to play a role in driving CLI seen with advancing age. Senescent cells with SASP may damage healthy tissue, including the eye tissues affected in AMD. The finding of increased effector cells in MPNd could implicate a role for adaptive immunity and senescent T cells together with increased CLI in drusen pathophysiology.
Collapse
Affiliation(s)
- Charlotte Liisborg
- Department of Ophthalmology, Zealand University Hospital, Roskilde 4000, Denmark
- Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen 2200, Denmark
| | - Vibe Skov
- Department of Hematology, Zealand University Hospital, Roskilde 4000, Denmark
| | - Lasse Kjær
- Department of Hematology, Zealand University Hospital, Roskilde 4000, Denmark
| | - Hans Carl Hasselbalch
- Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen 2200, Denmark
- Department of Hematology, Zealand University Hospital, Roskilde 4000, Denmark
| | - Torben Lykke Sørensen
- Department of Ophthalmology, Zealand University Hospital, Roskilde 4000, Denmark
- Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen 2200, Denmark
| |
Collapse
|
50
|
Zhang C, Lei L, Yang X, Ma K, Zheng H, Su Y, Jiao A, Wang X, Liu H, Zou Y, Shi L, Zhou X, Sun C, Hou Y, Xiao Z, Zhang L, Zhang B. Single-cell sequencing reveals antitumor characteristics of intratumoral immune cells in old mice. J Immunother Cancer 2021; 9:jitc-2021-002809. [PMID: 34642245 PMCID: PMC8513495 DOI: 10.1136/jitc-2021-002809] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/26/2021] [Indexed: 12/14/2022] Open
Abstract
Background Aging has long been thought to be a major risk factor for various types of cancers. However, accumulating evidence indicates increased resistance of old animals to tumor growth. An in-depth understanding of how old individuals defend against tumor invasion requires further investigations. Methods We revealed age-associated alterations in tumor-infiltrating immune cells between young and old mice using single-cell RNA and coupled T cell receptor (TCR) sequencing analysis. Multiple bioinformatics methods were adopted to analyze the characteristics of the transcriptome between two groups. To explore the impacts of young and old CD8+ T cells on tumor growth, mice were treated with anti-CD8 antibody every 3 days starting 7 days after tumor inoculation. Flow cytometry was used to validate the differences indicated by sequencing analysis between young and old mice. Results We found a higher proportion of cytotoxic CD8+ T cells, naturally occurring Tregs, conventional dendritic cell (DC), and M1-like macrophages in tumors of old mice compared with a higher percentage of exhausted CD8+ T cells, induced Tregs, plasmacytoid DC, and M2-like macrophages in young mice. Importantly, TCR diversity analysis showed that top 10 TCR clones consisted primarily of exhausted CD8+ T cells in young mice whereas top clones were predominantly cytotoxic CD8+ T cells in old mice. Old mice had more CD8+ T cells with a ‘progenitor’ and less ‘terminally’ exhausted phenotypes than young mice. Consistently, trajectory inference demonstrated that CD8+ T cells preferentially differentiated into cytotoxic cells in old mice in contrast to exhausted cells in young mice. Importantly, elimination of CD8+ T cells in old mice during tumor growth significantly accelerated tumor development. Moreover, senescent features were demonstrated in exhausted but not cytotoxic CD8+ T cells regardless of young and old mice. Conclusions Our data revealed that a significantly higher proportion of effector immune cells in old mice defends against tumor progression, providing insights into understanding the altered kinetics of cancer development and the differential response to immunotherapeutic modulation in elderly patients.
Collapse
Affiliation(s)
- Cangang Zhang
- Department of Pathogenic Microbiology and Immunology, Xi'an Jiaotong University, Xi'an, China.,Department of Pathogenic Microbiology and Immunology, Xi'an Jiaotong University Health Science Center, Xi'an, China
| | - Lei Lei
- Department of Pathogenic Microbiology and Immunology, Xi'an Jiaotong University, Xi'an, China.,Department of Pathogenic Microbiology and Immunology, Xi'an Jiaotong University Health Science Center, Xi'an, China.,Key Laboratory of Environment and Genes Related to Diseases, Ministry of Education, Xi'an, China
| | - Xiaofeng Yang
- Department of Pathogenic Microbiology and Immunology, Xi'an Jiaotong University, Xi'an, China.,Department of Pathogenic Microbiology and Immunology, Xi'an Jiaotong University Health Science Center, Xi'an, China.,Key Laboratory of Environment and Genes Related to Diseases, Ministry of Education, Xi'an, China
| | - Kaili Ma
- Institute of Systems Medicine, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.,Suzhou Institute of Systems Medicine, Suzhou, China
| | - Huiqiang Zheng
- Department of Pathogenic Microbiology and Immunology, Xi'an Jiaotong University, Xi'an, China.,Department of Pathogenic Microbiology and Immunology, Xi'an Jiaotong University Health Science Center, Xi'an, China
| | - Yanhong Su
- Department of Pathogenic Microbiology and Immunology, Xi'an Jiaotong University, Xi'an, China.,Department of Pathogenic Microbiology and Immunology, Xi'an Jiaotong University Health Science Center, Xi'an, China
| | - Anjun Jiao
- Department of Pathogenic Microbiology and Immunology, Xi'an Jiaotong University, Xi'an, China.,Department of Pathogenic Microbiology and Immunology, Xi'an Jiaotong University Health Science Center, Xi'an, China
| | - Xin Wang
- Department of Pathogenic Microbiology and Immunology, Xi'an Jiaotong University, Xi'an, China.,Department of Pathogenic Microbiology and Immunology, Xi'an Jiaotong University Health Science Center, Xi'an, China
| | - Haiyan Liu
- Department of Pathogenic Microbiology and Immunology, Xi'an Jiaotong University, Xi'an, China.,Department of Pathogenic Microbiology and Immunology, Xi'an Jiaotong University Health Science Center, Xi'an, China
| | - Yujing Zou
- Division of Hematologic Malignancies and Cellular Therapy, Duke University Medical Center, Durham, North Carolina, USA
| | - Lin Shi
- Department of Pathogenic Microbiology and Immunology, Xi'an Jiaotong University, Xi'an, China.,Department of Pathogenic Microbiology and Immunology, Xi'an Jiaotong University Health Science Center, Xi'an, China
| | - Xiaobo Zhou
- Department of Pathogenic Microbiology and Immunology, Xi'an Jiaotong University, Xi'an, China.,Department of Pathogenic Microbiology and Immunology, Xi'an Jiaotong University Health Science Center, Xi'an, China
| | - Chenming Sun
- Department of Pathogenic Microbiology and Immunology, Xi'an Jiaotong University, Xi'an, China.,Department of Pathogenic Microbiology and Immunology, Xi'an Jiaotong University Health Science Center, Xi'an, China
| | - Yuzhu Hou
- Department of Pathogenic Microbiology and Immunology, Xi'an Jiaotong University, Xi'an, China.,Department of Pathogenic Microbiology and Immunology, Xi'an Jiaotong University Health Science Center, Xi'an, China
| | - Zhengtao Xiao
- Department of Pathogenic Microbiology and Immunology, Xi'an Jiaotong University, Xi'an, China.,Department of Pathogenic Microbiology and Immunology, Xi'an Jiaotong University Health Science Center, Xi'an, China
| | - Lianjun Zhang
- Institute of Systems Medicine, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China .,Suzhou Institute of Systems Medicine, Suzhou, China.,Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Cancer Institute, Xuzhou Medical University, Xuzhou, China
| | - Baojun Zhang
- Department of Pathogenic Microbiology and Immunology, Xi'an Jiaotong University, Xi'an, China .,Department of Pathogenic Microbiology and Immunology, Xi'an Jiaotong University Health Science Center, Xi'an, China.,Key Laboratory of Environment and Genes Related to Diseases, Ministry of Education, Xi'an, China.,Xi'an Key Laboratory of Immune Related Diseases, Xi'an, China
| |
Collapse
|