1
|
Martín-Vírgala J, Miranda-Prieto D, Fernández-Villabrille S, Martín-Carro B, González-García N, Bande-Fernández J, Díaz-Corte C, Fernández-Martín JL, Alonso-Montes C, Suárez A, Panizo S, Naves-Díaz M, Rodríguez-Carrio J, Carrillo-López N. Novel T-cell subsets as non-invasive biomarkers of vascular damage along the predialysis stages of chronic kidney disease. Front Med (Lausanne) 2024; 11:1460021. [PMID: 39717170 PMCID: PMC11663642 DOI: 10.3389/fmed.2024.1460021] [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: 07/05/2024] [Accepted: 11/26/2024] [Indexed: 12/25/2024] Open
Abstract
Introduction Cardiovascular disease is the major cause of premature death in chronic kidney disease (CKD) and vascular damage is often detected belatedly, usually evaluated by expensive and invasive techniques. CKD involves specific risk factors that lead to vascular calcification and atherosclerosis, where inflammation plays a critical role. However, there are few inflammation-related markers to predict vascular damage in CKD. This study aimed to investigate immune populations in pre-dialysis patients to (i) identify subset alterations, (ii) assess longitudinal changes, and (iii) evaluate their applicability as biomarkers of subclinical vascular indices. Methods 43 pre-dialysis CKD patients in stages CKD-2 to CKD-5 and 38 controls were recruited at baseline and after 18-month follow-up. Aortic stiffness was determined by carotid-femoral pulse wave velocity (PWV) and abdominal aortic calcification was quantified by the Kauppila index on X-rays. Carotid intima-media thickness, the number of carotid plaques and adventitial neovascularization were evaluated by Superb Microvascular Imaging. Peripheral blood mononuclear cells were isolated and immune cell populations were assessed by flow cytometry: senescent T cells (CD4+CD28null), Tang (CD3+CD31+CD184+) and derived subsets, and monocyte subsets (classical, intermediate and non-classical; and ACE expression). Results Senescent T cells were increased in CKD. Despite Tang levels were unchanged compared to controls, this subset exhibited enhanced immunosenescence traits (CD28null and inverted CD4+CD8+ ratio) in CKD. Furthermore, Tang were negatively correlated with CKD progression. Slight alterations within monocyte subsets were observed. These findings were validated at the 18-month follow-up. Tang were correlated with several subclinical indices, and further analyses revealed an independent effect on PWV and their potential value as biomarkers. Intermediate monocytes were positively correlated with PWV. Conclusion Pre-dialysis CKD stages are hallmarked by alterations in immune cell populations related to vascular homeostasis, including early T-cell immunosenescence traits and a stage-dependent Tang depletion, which was independently related to vascular stiffness. All these features were replicated upon follow-up, thus providing validation toward our results. Our findings pave the ground for future studies addressing the functional contribution of these cellular mediators at the local level, assessing their potential predictive value in the long-term and implementing preventive strategies in the clinical setting.
Collapse
Affiliation(s)
- Julia Martín-Vírgala
- Metabolismo Óseo, Vascular y Enfermedades Inflamatorias Crónicas, Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), Oviedo, Spain
- Bone and Mineral Research Unit, Hospital Universitario Central de Asturias, Oviedo, Spain
- Resultados en Salud 2040 (RICORS2040, Kidney Disease), Oviedo, Spain
| | - Daniel Miranda-Prieto
- Area of Immunology, Department of Functional Biology, Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), University of Oviedo, Oviedo, Spain
- Investigación básica y traslacional en enfermedades inflamatorias crónicas, Instituto de Investigación del Principado de Asturias (ISPA), Oviedo, Spain
| | - Sara Fernández-Villabrille
- Metabolismo Óseo, Vascular y Enfermedades Inflamatorias Crónicas, Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), Oviedo, Spain
- Bone and Mineral Research Unit, Hospital Universitario Central de Asturias, Oviedo, Spain
- Resultados en Salud 2040 (RICORS2040, Kidney Disease), Oviedo, Spain
| | - Beatriz Martín-Carro
- Metabolismo Óseo, Vascular y Enfermedades Inflamatorias Crónicas, Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), Oviedo, Spain
- Bone and Mineral Research Unit, Hospital Universitario Central de Asturias, Oviedo, Spain
- Resultados en Salud 2040 (RICORS2040, Kidney Disease), Oviedo, Spain
| | - Nerea González-García
- Metabolismo Óseo, Vascular y Enfermedades Inflamatorias Crónicas, Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), Oviedo, Spain
- Bone and Mineral Research Unit, Hospital Universitario Central de Asturias, Oviedo, Spain
- Resultados en Salud 2040 (RICORS2040, Kidney Disease), Oviedo, Spain
| | | | - Carmen Díaz-Corte
- Resultados en Salud 2040 (RICORS2040, Kidney Disease), Oviedo, Spain
- Nephrology Unit, Hospital Universitario Central de Asturias, Oviedo, Spain
- Departamento de Medicina, Universidad de Oviedo, Oviedo, Spain
| | - José Luis Fernández-Martín
- Metabolismo Óseo, Vascular y Enfermedades Inflamatorias Crónicas, Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), Oviedo, Spain
- Bone and Mineral Research Unit, Hospital Universitario Central de Asturias, Oviedo, Spain
- Resultados en Salud 2040 (RICORS2040, Kidney Disease), Oviedo, Spain
- Departamento de Medicina, Universidad de Oviedo, Oviedo, Spain
| | - Cristina Alonso-Montes
- Metabolismo Óseo, Vascular y Enfermedades Inflamatorias Crónicas, Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), Oviedo, Spain
- Bone and Mineral Research Unit, Hospital Universitario Central de Asturias, Oviedo, Spain
- Resultados en Salud 2040 (RICORS2040, Kidney Disease), Oviedo, Spain
- Departamento de Medicina, Universidad de Oviedo, Oviedo, Spain
| | - Ana Suárez
- Investigación básica y traslacional en enfermedades inflamatorias crónicas, Instituto de Investigación del Principado de Asturias (ISPA), Oviedo, Spain
| | - Sara Panizo
- Metabolismo Óseo, Vascular y Enfermedades Inflamatorias Crónicas, Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), Oviedo, Spain
- Bone and Mineral Research Unit, Hospital Universitario Central de Asturias, Oviedo, Spain
- Resultados en Salud 2040 (RICORS2040, Kidney Disease), Oviedo, Spain
- Departamento de Medicina, Universidad de Oviedo, Oviedo, Spain
| | - Manuel Naves-Díaz
- Metabolismo Óseo, Vascular y Enfermedades Inflamatorias Crónicas, Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), Oviedo, Spain
- Bone and Mineral Research Unit, Hospital Universitario Central de Asturias, Oviedo, Spain
- Resultados en Salud 2040 (RICORS2040, Kidney Disease), Oviedo, Spain
- Departamento de Medicina, Universidad de Oviedo, Oviedo, Spain
| | - Javier Rodríguez-Carrio
- Area of Immunology, Department of Functional Biology, Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), University of Oviedo, Oviedo, Spain
- Investigación básica y traslacional en enfermedades inflamatorias crónicas, Instituto de Investigación del Principado de Asturias (ISPA), Oviedo, Spain
| | - Natalia Carrillo-López
- Metabolismo Óseo, Vascular y Enfermedades Inflamatorias Crónicas, Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), Oviedo, Spain
- Bone and Mineral Research Unit, Hospital Universitario Central de Asturias, Oviedo, Spain
- Resultados en Salud 2040 (RICORS2040, Kidney Disease), Oviedo, Spain
- Departamento de Medicina, Universidad de Oviedo, Oviedo, Spain
| |
Collapse
|
2
|
Richardson KC, Jung K, Matsubara JA, Choy JC, Granville DJ. Granzyme B in aging and age-related pathologies. Trends Mol Med 2024; 30:1165-1179. [PMID: 39181801 DOI: 10.1016/j.molmed.2024.07.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2024] [Revised: 07/19/2024] [Accepted: 07/24/2024] [Indexed: 08/27/2024]
Abstract
Aging is a major risk factor for pathologies that manifest later in life. Much attention is devoted towards elucidating how prolonged environmental exposures and inflammation promote biological (accelerated) tissue aging. Granzymes, a family of serine proteases, are increasingly recognized for their emerging roles in biological aging and disease. Widely recognized as intracellular mediators of cell death, granzymes, particularly granzyme B (GzmB), also accumulate in the extracellular milieu of tissues with age, contributing to chronic tissue injury, inflammation, and impaired healing. Consequently, this has prompted the field to reconsider how GzmB regulation, accumulation, and proteolysis impact health and disease with age. While GzmB is observed in numerous age-related conditions, the current review focuses on mechanistic studies where proof-of-concept has been forwarded.
Collapse
Affiliation(s)
- Katlyn C Richardson
- International Collaboration On Repair Discoveries (ICORD) Centre, Department of Pathology and Laboratory Medicine, British Columbia Professional Firefighters' Burn and Wound Healing Laboratory, Vancouver Coastal Health Research Institute, University of British Columbia, Vancouver, BC, Canada
| | - Karen Jung
- International Collaboration On Repair Discoveries (ICORD) Centre, Department of Pathology and Laboratory Medicine, British Columbia Professional Firefighters' Burn and Wound Healing Laboratory, Vancouver Coastal Health Research Institute, University of British Columbia, Vancouver, BC, Canada
| | - Joanne A Matsubara
- Department of Ophthalmology and Visual Sciences, University of British Columbia, Vancouver, BC, Canada
| | - Jonathan C Choy
- Department of Molecular Biology and Biochemistry, Simon Fraser University, Burnaby, BC, Canada
| | - David J Granville
- International Collaboration On Repair Discoveries (ICORD) Centre, Department of Pathology and Laboratory Medicine, British Columbia Professional Firefighters' Burn and Wound Healing Laboratory, Vancouver Coastal Health Research Institute, University of British Columbia, Vancouver, BC, Canada; Centre for Heart Lung Innovation, Providence Research, University of British Columbia, Vancouver, BC, Canada.
| |
Collapse
|
3
|
Ntuli Y, Shawcross DL. Infection, inflammation and hepatic encephalopathy from a clinical perspective. Metab Brain Dis 2024; 39:1689-1703. [PMID: 39212845 PMCID: PMC11535002 DOI: 10.1007/s11011-024-01402-y] [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: 04/11/2024] [Accepted: 07/23/2024] [Indexed: 09/04/2024]
Abstract
Hepatic encephalopathy (HE) is a syndrome that is associated with both acute and chronic liver injury. It manifests as a wide spectrum of neuropsychological abnormalities, ranging from subtle impairments in executive higher functions observed in cirrhosis, through to coma in acute liver failure. In acute liver failure, the central role of ammonia in the development of brain oedema has remained undisputed for 130 years. It latterly became apparent that infection and inflammation were profound determinants for the development of severe hepatic encephalopathy, associated with the development of cerebral oedema and intracranial hypertension. The relationship of the development of hepatic encephalopathy with blood ammonia levels in cirrhosis is less clear cut and the synergistic interplay of inflammation and infection with ammonia has been identified as being fundamental in the development and progression of hepatic encephalopathy. A perturbed gut microbiome and the presence of an impaired gut epithelial barrier that facilitates translocation of bacteria and bacterial degradation products into the systemic circulation, inducing systemic inflammation and innate and adaptive immune dysfunction, has now become the focus of therapies that treat hepatic encephalopathy in cirrhosis, and may explain why the prebiotic lactulose and rifaximin are efficacious. This review summarises the current clinical perspective on the roles of inflammation and infection in hepatic encephalopathy and presents the evidence base for existing therapies and those in development in the setting of acute and chronic liver failure.
Collapse
Affiliation(s)
- Yevedzo Ntuli
- School of Immunology and Microbial Sciences, Faculty of Life Sciences and Medicine, King's College London, King's College Hospital, 125 Coldharbour Lane, London, SE5 9NU, UK
- Institute of Liver Studies, King's College Hospital, Denmark Hill, London, SE5 9RS, UK
| | - Debbie L Shawcross
- School of Immunology and Microbial Sciences, Faculty of Life Sciences and Medicine, King's College London, King's College Hospital, 125 Coldharbour Lane, London, SE5 9NU, UK.
- Institute of Liver Studies, King's College Hospital, Denmark Hill, London, SE5 9RS, UK.
| |
Collapse
|
4
|
Ahmed B, Rahman AA, Lee S, Malhotra R. The Implications of Aging on Vascular Health. Int J Mol Sci 2024; 25:11188. [PMID: 39456971 PMCID: PMC11508873 DOI: 10.3390/ijms252011188] [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: 09/18/2024] [Revised: 10/07/2024] [Accepted: 10/11/2024] [Indexed: 10/28/2024] Open
Abstract
Vascular aging encompasses structural and functional changes in the vasculature, significantly contributing to cardiovascular diseases, which are the leading cause of death globally. The incidence and prevalence of these diseases increase with age, with most morbidity and mortality attributed to myocardial infarction and stroke. Diagnosing and intervening in vascular aging while understanding the mechanisms behind age-induced vascular phenotypic and pathophysiological alterations offers the potential for delaying and preventing cardiovascular mortality in an aging population. This review delves into various aspects of vascular aging by examining age-related changes in arterial health at the cellular level, including endothelial dysfunction, cellular senescence, and vascular smooth muscle cell transdifferentiation, as well as at the structural level, including arterial stiffness and changes in wall thickness and diameter. We also explore aging-related changes in perivascular adipose tissue deposition, arterial collateralization, and calcification, providing insights into the physiological and pathological implications. Overall, aging induces phenotypic changes that augment the vascular system's susceptibility to disease, even in the absence of traditional risk factors, such as hypertension, diabetes, obesity, and smoking. Overall, age-related modifications in cellular phenotype and molecular homeostasis increase the vulnerability of the arterial vasculature to structural and functional alterations, thereby accelerating cardiovascular risk. Increasing our understanding of these modifications is crucial for success in delaying or preventing cardiovascular diseases. Non-invasive techniques, such as measuring carotid intima-media thickness, pulse wave velocity, and flow-mediated dilation, as well as detecting vascular calcifications, can be used for the early detection of vascular aging. Targeting specific pathological mechanisms, such as cellular senescence and enhancing angiogenesis, holds promise for innovative therapeutic approaches.
Collapse
Affiliation(s)
- Bulbul Ahmed
- Evans Department of Medicine and Whitaker Cardiovascular Institute, Boston University School of Medicine, Boston, MA 02118, USA;
| | - Ahmed A. Rahman
- Department of Pediatric Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
| | - Sujin Lee
- Division of Vascular Surgery, Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA;
| | - Rajeev Malhotra
- Division of Cardiology, Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
| |
Collapse
|
5
|
Konishi T, Kawakami R, Vozenilek AE, Ghosh SKB, Xu W, Grogan A, Shah P, Tanaka T, Sekimoto T, Shiraki T, Kawai K, Sato Y, Mori M, Sakamoto A, Hisadome H, Ashida K, Bellissard A, Williams D, Dryanovski D, Kutys R, Cheng Q, Romero M, Chahal D, Virmani R, Finn AV. Mechanisms of Medial Wall Thinning in Chronic Total Occlusion. JACC Cardiovasc Interv 2024; 17:1719-1728. [PMID: 38970581 DOI: 10.1016/j.jcin.2024.05.013] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/10/2024] [Revised: 04/25/2024] [Accepted: 05/07/2024] [Indexed: 07/08/2024]
Abstract
BACKGROUND The success rate of percutaneous coronary intervention (PCI) for chronic total occlusion (CTO) is lower and the risk for complications higher compared with other non-CTO PCI. Although interventionalists focus on intimal plaque characteristics, the coronary media is an important (especially for techniques involving antegrade dissection and re-entry) but poorly understood structure in CTO PCI. OBJECTIVES The aim of the present study was to investigate coronary medial wall thinning in CTO lesions and determine how this thinning might affect CTO PCI. METHODS A total of 2,586 sections were investigated, from arteries with evidence of CTO from 54 subjects (1,383 sections) and arteries without evidence of CTO from 54 subjects with non-coronary-related deaths (1,203 sections) after matching for age, gender, body weight, and body height. RESULTS The medial thickness in subjects with CTO was lower than that in those with non-coronary-related death (P < 0.001). In subjects with CTO, CTO lesions had thinner medial walls compared with those with lower luminal narrowing (P < 0.001). At the CTO distal segments, the 6- to 12-mm distal segment from the distal end of the CTO had significantly less luminal narrowing (P < 0.001), and similar medial thickness, compared with the distal end of the CTO. Immunohistochemical analysis revealed that short-duration CTO had more cleaved caspase-3-positive cells in media and had significantly more CD3+, CD4+, CD8+, and CD4+CD28null T cells compared with long-duration CTO. CONCLUSIONS CTO lesions demonstrated coronary medial thinning compared with non-CTO lesions. Further investigation of the cause-and-effect relationship among inflammation, apoptosis, and coronary medial wall thinning is warranted in future mechanistic studies.
Collapse
Affiliation(s)
- Takao Konishi
- Department of Cardiovascular Pathology, CVPath Institute, Gaithersburg, Maryland, USA; Department of Cardiovascular Medicine, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Sapporo, Japan
| | - Rika Kawakami
- Department of Cardiovascular Pathology, CVPath Institute, Gaithersburg, Maryland, USA
| | - Aimee E Vozenilek
- Department of Cardiovascular Pathology, CVPath Institute, Gaithersburg, Maryland, USA
| | - Saikat Kumar B Ghosh
- Department of Cardiovascular Pathology, CVPath Institute, Gaithersburg, Maryland, USA
| | - Weili Xu
- Department of Cardiovascular Pathology, CVPath Institute, Gaithersburg, Maryland, USA
| | - Alyssa Grogan
- Department of Cardiovascular Pathology, CVPath Institute, Gaithersburg, Maryland, USA
| | - Palak Shah
- Department of Cardiovascular Pathology, CVPath Institute, Gaithersburg, Maryland, USA
| | - Takamasa Tanaka
- Department of Cardiovascular Pathology, CVPath Institute, Gaithersburg, Maryland, USA
| | - Teruo Sekimoto
- Department of Cardiovascular Pathology, CVPath Institute, Gaithersburg, Maryland, USA
| | - Tatsuya Shiraki
- Department of Cardiovascular Pathology, CVPath Institute, Gaithersburg, Maryland, USA
| | - Kenji Kawai
- Department of Cardiovascular Pathology, CVPath Institute, Gaithersburg, Maryland, USA
| | - Yu Sato
- Department of Cardiovascular Pathology, CVPath Institute, Gaithersburg, Maryland, USA
| | - Masayuki Mori
- Department of Cardiovascular Pathology, CVPath Institute, Gaithersburg, Maryland, USA
| | - Atsushi Sakamoto
- Department of Cardiovascular Pathology, CVPath Institute, Gaithersburg, Maryland, USA
| | | | - Kazuhiro Ashida
- Cardiovascular Center, Seirei Yokohama Hospital, Yokohama, Japan
| | - Arielle Bellissard
- Department of Cardiovascular Pathology, CVPath Institute, Gaithersburg, Maryland, USA
| | - Desiree Williams
- Department of Cardiovascular Pathology, CVPath Institute, Gaithersburg, Maryland, USA
| | - Dilyan Dryanovski
- Department of Cardiovascular Pathology, CVPath Institute, Gaithersburg, Maryland, USA
| | - Robert Kutys
- Department of Cardiovascular Pathology, CVPath Institute, Gaithersburg, Maryland, USA
| | - Qi Cheng
- Department of Cardiovascular Pathology, CVPath Institute, Gaithersburg, Maryland, USA
| | - Maria Romero
- Department of Cardiovascular Pathology, CVPath Institute, Gaithersburg, Maryland, USA
| | - Diljon Chahal
- School of Medicine, University of Maryland, Baltimore, Maryland, USA
| | - Renu Virmani
- Department of Cardiovascular Pathology, CVPath Institute, Gaithersburg, Maryland, USA
| | - Aloke V Finn
- Department of Cardiovascular Pathology, CVPath Institute, Gaithersburg, Maryland, USA; School of Medicine, University of Maryland, Baltimore, Maryland, USA.
| |
Collapse
|
6
|
Lee ES, Nguyen N, Young BE, Wee H, Wazny V, Lee KL, Tay KY, Goh LL, Chioh FW, Law MC, Lee IR, Ang LT, Loh KM, Chan MY, Fan BE, Dalan R, Lye DC, Renia L, Cheung C. Inflammatory risk contributes to post-COVID endothelial dysfunction through anti-ACKR1 autoantibody. Life Sci Alliance 2024; 7:e202402598. [PMID: 38740432 PMCID: PMC11091471 DOI: 10.26508/lsa.202402598] [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: 01/16/2024] [Revised: 04/26/2024] [Accepted: 04/29/2024] [Indexed: 05/16/2024] Open
Abstract
Subclinical vascular impairment can be exacerbated in individuals who experience sustained inflammation after COVID-19 infection. Our study explores the prevalence and impact of autoantibodies on vascular dysfunction in healthy COVID-19 survivors, an area that remains inadequately investigated. Focusing on autoantibodies against the atypical chemokine receptor 1 (ACKR1), COVID-19 survivors demonstrated significantly elevated anti-ACKR1 autoantibodies, correlating with systemic cytokines, circulating damaged endothelial cells, and endothelial dysfunction. An independent cohort linked these autoantibodies to increased vascular disease outcomes during a median 6.7-yr follow-up. We analyzed a single-cell transcriptome atlas of endothelial cells from diverse mouse tissues, identifying enriched Ackr1 expressions in venous regions of the brain and soleus muscle vasculatures, which holds intriguing implications for tissue-specific venous thromboembolism manifestations reported in COVID-19. Functionally, purified immunoglobulin G (IgG) extracted from patient plasma did not trigger cell apoptosis or increase barrier permeability in human vein endothelial cells. Instead, plasma IgG enhanced antibody-dependent cellular cytotoxicity mediated by patient PBMCs, a phenomenon alleviated by blocking peptide or liposome ACKR1 recombinant protein. The blocking peptide uncovered that purified IgG from COVID-19 survivors possessed potential epitopes in the N-terminal extracellular domain of ACKR1, which effectively averted antibody-dependent cellular cytotoxicity. Our findings offer insights into therapeutic development to mitigate autoantibody reactivity in blood vessels in chronic inflammation.
Collapse
Affiliation(s)
- Ee-Soo Lee
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, Singapore
| | - Nhi Nguyen
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, Singapore
| | - Barnaby E Young
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, Singapore
- Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- Department of Infectious Diseases, Tan Tock Seng Hospital, Singapore, Singapore
- National Centre for Infectious Diseases, Singapore, Singapore
| | - Hannah Wee
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, Singapore
| | - Vanessa Wazny
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, Singapore
| | - Khang Leng Lee
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, Singapore
| | - Kai Yi Tay
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, Singapore
| | - Liuh Ling Goh
- Department of Endocrinology, Tan Tock Seng Hospital, Singapore, Singapore
| | - Florence Wj Chioh
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, Singapore
| | - Michelle Cy Law
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, Singapore
| | - I Russel Lee
- National Centre for Infectious Diseases, Singapore, Singapore
| | - Lay Teng Ang
- Stanford Institute for Stem Cell Biology and Regenerative Medicine, Stanford University, Stanford, CA, USA
| | - Kyle M Loh
- Stanford Institute for Stem Cell Biology and Regenerative Medicine, Stanford University, Stanford, CA, USA
- Department of Developmental Biology, Stanford Cardiovascular Institute, Stanford University, Stanford, CA, USA
| | - Mark Y Chan
- Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- National University Heart Centre, National University Health System, Singapore, Singapore
| | - Bingwen E Fan
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, Singapore
- Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- Department of Haematology, Tan Tock Seng Hospital, Singapore, Singapore
- Department of Laboratory Medicine, Khoo Teck Puat Hospital, Singapore, Singapore
| | - Rinkoo Dalan
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, Singapore
- Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- Department of Endocrinology, Tan Tock Seng Hospital, Singapore, Singapore
| | - David C Lye
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, Singapore
- Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- Department of Infectious Diseases, Tan Tock Seng Hospital, Singapore, Singapore
- National Centre for Infectious Diseases, Singapore, Singapore
| | - Laurent Renia
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, Singapore
- A*STAR Infectious Diseases Labs, Agency for Science, Technology and Research, Singapore, Singapore
| | - Christine Cheung
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, Singapore
- Institute of Molecular and Cell Biology, Agency for Science, Technology and Research, Singapore, Singapore
| |
Collapse
|
7
|
Guan Y, Cao M, Wu X, Yan J, Hao Y, Zhang C. CD28 null T cells in aging and diseases: From biology to assessment and intervention. Int Immunopharmacol 2024; 131:111807. [PMID: 38471362 DOI: 10.1016/j.intimp.2024.111807] [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: 01/02/2024] [Revised: 02/25/2024] [Accepted: 03/04/2024] [Indexed: 03/14/2024]
Abstract
CD28null T cells, an atypical subset characterized by the loss of CD28 costimulatory molecule expression, exhibit functional variants and progressively expand with age. Moreover, T cells with these phenotypes are found in both typical and atypical humoral immune responses. Consequently, they accumulate during infectious diseases, autoimmune disorders, cardiovascular conditions, and neurodegenerative ailments. To provide an in-depth review of the current knowledge regarding CD28null T cells, we specifically focus on their phenotypic and functional characteristics as well as their physiological roles in aging and diseases. While uncertainties regarding the clinical utility remains, we will review the following two crucial research perspectives to explore clinical translational applications of the research on this specific T cell subset: 1) addressing the potential utility of CD28null T cells as immunological markers for prognosis and adverse outcomes in both aging and disease, and 2) speculating on the potential of targeting CD28null T cells as an interventional strategy for preventing or delaying immune aging processes and disease progression.
Collapse
Affiliation(s)
- Yuqi Guan
- Department of Geriatrics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Avenue, Wuhan 430030, China; Key Laboratory of Vascular Aging, Ministry of Education, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Avenue, Wuhan 430030, China
| | - Ming Cao
- Department of Geriatrics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Avenue, Wuhan 430030, China; Key Laboratory of Vascular Aging, Ministry of Education, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Avenue, Wuhan 430030, China
| | - Xiaofen Wu
- Department of Geriatrics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Avenue, Wuhan 430030, China; Key Laboratory of Vascular Aging, Ministry of Education, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Avenue, Wuhan 430030, China
| | - Jinhua Yan
- Department of Geriatrics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Avenue, Wuhan 430030, China; Key Laboratory of Vascular Aging, Ministry of Education, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Avenue, Wuhan 430030, China
| | - Yi Hao
- Department of Geriatrics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Avenue, Wuhan 430030, China; Key Laboratory of Vascular Aging, Ministry of Education, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Avenue, Wuhan 430030, China; Department of Pathogen Biology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China.
| | - Cuntai Zhang
- Department of Geriatrics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Avenue, Wuhan 430030, China; Key Laboratory of Vascular Aging, Ministry of Education, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Avenue, Wuhan 430030, China.
| |
Collapse
|
8
|
Snijckers RPM, Foks AC. Adaptive immunity and atherosclerosis: aging at its crossroads. Front Immunol 2024; 15:1350471. [PMID: 38686373 PMCID: PMC11056569 DOI: 10.3389/fimmu.2024.1350471] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2023] [Accepted: 03/28/2024] [Indexed: 05/02/2024] Open
Abstract
Adaptive immunity plays a profound role in atherosclerosis pathogenesis by regulating antigen-specific responses, inflammatory signaling and antibody production. However, as we age, our immune system undergoes a gradual functional decline, a phenomenon termed "immunosenescence". This decline is characterized by a reduction in proliferative naïve B- and T cells, decreased B- and T cell receptor repertoire and a pro-inflammatory senescence associated secretory profile. Furthermore, aging affects germinal center responses and deteriorates secondary lymphoid organ function and structure, leading to impaired T-B cell dynamics and increased autoantibody production. In this review, we will dissect the impact of aging on adaptive immunity and the role played by age-associated B- and T cells in atherosclerosis pathogenesis, emphasizing the need for interventions that target age-related immune dysfunction to reduce cardiovascular disease risk.
Collapse
Affiliation(s)
| | - Amanda C. Foks
- Division of BioTherapeutics, Leiden Academic Centre for Drug Research, Leiden University, Leiden, Netherlands
| |
Collapse
|
9
|
Hoeks C, Puijfelik FV, Koetzier SC, Rip J, Corsten CEA, Wierenga-Wolf AF, Melief MJ, Stinissen P, Smolders J, Hellings N, Broux B, van Luijn MM. Differential Runx3, Eomes, and T-bet expression subdivides MS-associated CD4 + T cells with brain-homing capacity. Eur J Immunol 2024; 54:e2350544. [PMID: 38009648 DOI: 10.1002/eji.202350544] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2023] [Revised: 11/22/2023] [Accepted: 11/24/2023] [Indexed: 11/29/2023]
Abstract
Multiple sclerosis (MS) is a common and devastating chronic inflammatory disease of the CNS. CD4+ T cells are assumed to be the first to cross the blood-central nervous system (CNS) barrier and trigger local inflammation. Here, we explored how pathogenicity-associated effector programs define CD4+ T cell subsets with brain-homing ability in MS. Runx3- and Eomes-, but not T-bet-expressing CD4+ memory cells were diminished in the blood of MS patients. This decline reversed following natalizumab treatment and was supported by a Runx3+ Eomes+ T-bet- enrichment in cerebrospinal fluid samples of treatment-naïve MS patients. This transcription factor profile was associated with high granzyme K (GZMK) and CCR5 levels and was most prominent in Th17.1 cells (CCR6+ CXCR3+ CCR4-/dim ). Previously published CD28- CD4 T cells were characterized by a Runx3+ Eomes- T-bet+ phenotype that coincided with intermediate CCR5 and a higher granzyme B (GZMB) and perforin expression, indicating the presence of two separate subsets. Under steady-state conditions, granzyme Khigh Th17.1 cells spontaneously passed the blood-brain barrier in vitro. This was only found for other subsets including CD28- cells when using inflamed barriers. Altogether, CD4+ T cells contain small fractions with separate pathogenic features, of which Th17.1 seems to breach the blood-brain barrier as a possible early event in MS.
Collapse
Affiliation(s)
- Cindy Hoeks
- Department of Immunology and Infection, Biomedical Research Institute, Hasselt University, Hasselt, Belgium
- University MS Center (UMSC), Hasselt, Belgium
| | - Fabiënne van Puijfelik
- Department of Immunology, MS Center ErasMS, Erasmus MC, University Medical Center Rotterdam, Rotterdam, the Netherlands
| | - Steven C Koetzier
- Department of Immunology, MS Center ErasMS, Erasmus MC, University Medical Center Rotterdam, Rotterdam, the Netherlands
| | - Jasper Rip
- Department of Immunology, MS Center ErasMS, Erasmus MC, University Medical Center Rotterdam, Rotterdam, the Netherlands
| | - Cato E A Corsten
- Department of Neurology, MS Center ErasMS, Erasmus MC, University Medical Center Rotterdam, Rotterdam, the Netherlands
| | - Annet F Wierenga-Wolf
- Department of Immunology, MS Center ErasMS, Erasmus MC, University Medical Center Rotterdam, Rotterdam, the Netherlands
| | - Marie-José Melief
- Department of Immunology, MS Center ErasMS, Erasmus MC, University Medical Center Rotterdam, Rotterdam, the Netherlands
| | - Piet Stinissen
- Department of Immunology and Infection, Biomedical Research Institute, Hasselt University, Hasselt, Belgium
- University MS Center (UMSC), Hasselt, Belgium
| | - Joost Smolders
- Department of Immunology, MS Center ErasMS, Erasmus MC, University Medical Center Rotterdam, Rotterdam, the Netherlands
- Department of Neurology, MS Center ErasMS, Erasmus MC, University Medical Center Rotterdam, Rotterdam, the Netherlands
- Neuroimmunology Research Group, Netherlands Institute for Neuroscience, Amsterdam, the Netherlands
| | - Niels Hellings
- Department of Immunology and Infection, Biomedical Research Institute, Hasselt University, Hasselt, Belgium
- University MS Center (UMSC), Hasselt, Belgium
| | - Bieke Broux
- Department of Immunology and Infection, Biomedical Research Institute, Hasselt University, Hasselt, Belgium
- University MS Center (UMSC), Hasselt, Belgium
| | - Marvin M van Luijn
- Department of Immunology, MS Center ErasMS, Erasmus MC, University Medical Center Rotterdam, Rotterdam, the Netherlands
| |
Collapse
|
10
|
Hopkin SJ, Pezhman L, Begum J, Kavanagh D, McGettrick HM, Iqbal AJ, Chimen M. Aging modulates homeostatic leukocyte trafficking to the peritoneal cavity in a sex-specific manner. J Leukoc Biol 2023; 114:301-314. [PMID: 37309034 PMCID: PMC10533226 DOI: 10.1093/jleuko/qiad053] [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: 12/01/2022] [Revised: 03/28/2023] [Accepted: 04/28/2023] [Indexed: 06/14/2023] Open
Abstract
Aging is associated with exacerbated systemic inflammation (inflammaging) and the progressive loss of immune system function (immunosenescence). Leukocyte migration is necessary for effective immunity; however, dysregulated trafficking of leukocytes into tissue contributes to inflammaging and the development of age-related inflammatory diseases. Aging modulates leukocyte trafficking under inflammatory conditions; however, whether aging modulates leukocyte trafficking under homeostatic conditions remains to be elucidated. Although immune responses are evidently sexually dimorphic, limited studies have investigated the effect of sex on age-related changes to leukocyte trafficking processes. Here, we investigated age-related and sex-specific changes to the leukocyte populations within the peritoneal cavity of young (3-mo), middle-aged (18-mo) and old (21-mo) male and female wild-type mice in the steady state. We found an age-related increase in the number of leukocytes within the peritoneal cavity of female mice, predominantly B cells, which may reflect increased trafficking through this tissue with age. This was accompanied by an increased inflammatory environment within the aged cavity, including increased levels of chemoattractants, including B cell chemoattractants CXCL13 and CCL21, soluble adhesion molecules, and proinflammatory cytokines, which was more pronounced in aged female mice. Intravital microscopy techniques revealed altered vascular structure and increased vascular permeability within the peritoneal membrane of aged female mice, which may support increased leukocyte trafficking to the cavity with age. Together, these data indicate that aging affects homeostatic leukocyte trafficking processes in a sex-specific fashion.
Collapse
Affiliation(s)
- Sophie J Hopkin
- Institute of Cardiovascular Sciences, University of Birmingham, Birmingham, B15 2TT, United Kingdom
| | - Laleh Pezhman
- Institute of Inflammation and Ageing, University of Birmingham, Birmingham, B15 2TT, United Kingdom
| | - Jenefa Begum
- Institute of Cardiovascular Sciences, University of Birmingham, Birmingham, B15 2TT, United Kingdom
| | - Dean Kavanagh
- Institute of Cardiovascular Sciences, University of Birmingham, Birmingham, B15 2TT, United Kingdom
| | - Helen M McGettrick
- Institute of Inflammation and Ageing, University of Birmingham, Birmingham, B15 2TT, United Kingdom
| | - Asif J Iqbal
- Institute of Cardiovascular Sciences, University of Birmingham, Birmingham, B15 2TT, United Kingdom
| | - Myriam Chimen
- Institute of Inflammation and Ageing, University of Birmingham, Birmingham, B15 2TT, United Kingdom
| |
Collapse
|
11
|
Sato Y, Jain A, Ohtsuki S, Okuyama H, Sturmlechner I, Takashima Y, Le KPC, Bois MC, Berry GJ, Warrington KJ, Goronzy JJ, Weyand CM. Stem-like CD4 + T cells in perivascular tertiary lymphoid structures sustain autoimmune vasculitis. Sci Transl Med 2023; 15:eadh0380. [PMID: 37672564 PMCID: PMC11131576 DOI: 10.1126/scitranslmed.adh0380] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2023] [Accepted: 08/09/2023] [Indexed: 09/08/2023]
Abstract
Autoimmune vasculitis of the medium and large elastic arteries can cause blindness, stroke, aortic arch syndrome, and aortic aneurysm. The disease is often refractory to immunosuppressive therapy and progresses over decades as smoldering aortitis. How the granulomatous infiltrates in the vessel wall are maintained and how tissue-infiltrating T cells and macrophages are replenished are unknown. Single-cell and whole-tissue transcriptomic studies of immune cell populations in vasculitic arteries identified a CD4+ T cell population with stem cell-like features. CD4+ T cells supplying the tissue-infiltrating and tissue-damaging effector T cells survived in tertiary lymphoid structures around adventitial vasa vasora, expressed the transcription factor T cell factor 1 (TCF1), had high proliferative potential, and gave rise to two effector populations, Eomesodermin (EOMES)+ cytotoxic T cells and B cell lymphoma 6 (BCL6)+ T follicular helper-like cells. TCF1hiCD4+ T cells expressing the interleukin 7 receptor (IL-7R) sustained vasculitis in serial transplantation experiments. Thus, TCF1hiCD4+ T cells function as disease stem cells and promote chronicity and autonomy of autoimmune tissue inflammation. Remission-inducing therapies will require targeting stem-like CD4+ T cells instead of only effector T cells.
Collapse
Affiliation(s)
- Yuki Sato
- Department of Medicine, Mayo Clinic College of Medicine and
Science, Rochester, MN 55905, USA
- Department of Cardiovascular Disease, Mayo Clinic College
of Medicine and Science, Rochester, MN 55905, USA
- Department of Immunology, Mayo Clinic College of Medicine
and Science, Rochester, MN 55905, USA
| | - Abhinav Jain
- Department of Immunology, Mayo Clinic College of Medicine
and Science, Rochester, MN 55905, USA
| | - Shozo Ohtsuki
- Department of Medicine, Mayo Clinic College of Medicine and
Science, Rochester, MN 55905, USA
- Department of Cardiovascular Disease, Mayo Clinic College
of Medicine and Science, Rochester, MN 55905, USA
- Department of Immunology, Mayo Clinic College of Medicine
and Science, Rochester, MN 55905, USA
| | - Hirohisa Okuyama
- Department of Immunology, Mayo Clinic College of Medicine
and Science, Rochester, MN 55905, USA
| | - Ines Sturmlechner
- Department of Immunology, Mayo Clinic College of Medicine
and Science, Rochester, MN 55905, USA
| | - Yoshinori Takashima
- Department of Medicine, Mayo Clinic College of Medicine and
Science, Rochester, MN 55905, USA
- Department of Cardiovascular Disease, Mayo Clinic College
of Medicine and Science, Rochester, MN 55905, USA
- Department of Immunology, Mayo Clinic College of Medicine
and Science, Rochester, MN 55905, USA
| | - Kevin-Phu C Le
- Department of Medicine, Mayo Clinic College of Medicine and
Science, Rochester, MN 55905, USA
- Department of Cardiovascular Disease, Mayo Clinic College
of Medicine and Science, Rochester, MN 55905, USA
- Department of Immunology, Mayo Clinic College of Medicine
and Science, Rochester, MN 55905, USA
| | - Melanie C. Bois
- Department of Laboratory Medicine and Pathology, Mayo
Clinic College of Medicine and Science, Rochester, MN 55905, USA
| | - Gerald J. Berry
- Department of Pathology, School of Medicine, Stanford
University, Stanford, CA 94305, USA
| | - Kenneth J. Warrington
- Department of Medicine, Mayo Clinic College of Medicine and
Science, Rochester, MN 55905, USA
| | - Jorg J. Goronzy
- Department of Medicine, Mayo Clinic College of Medicine and
Science, Rochester, MN 55905, USA
- Department of Immunology, Mayo Clinic College of Medicine
and Science, Rochester, MN 55905, USA
- Department of Medicine, School of Medicine, Stanford
University, Stanford, CA 94305, USA
| | - Cornelia M. Weyand
- Department of Medicine, Mayo Clinic College of Medicine and
Science, Rochester, MN 55905, USA
- Department of Cardiovascular Disease, Mayo Clinic College
of Medicine and Science, Rochester, MN 55905, USA
- Department of Immunology, Mayo Clinic College of Medicine
and Science, Rochester, MN 55905, USA
- Department of Medicine, School of Medicine, Stanford
University, Stanford, CA 94305, USA
| |
Collapse
|
12
|
Liu Q, Zheng Y, Goronzy JJ, Weyand CM. T cell aging as a risk factor for autoimmunity. J Autoimmun 2023; 137:102947. [PMID: 36357240 PMCID: PMC10164202 DOI: 10.1016/j.jaut.2022.102947] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2022] [Accepted: 10/23/2022] [Indexed: 11/09/2022]
Abstract
Immune aging is a complex process rendering the host susceptible to cancer, infection, and insufficient tissue repair. Many autoimmune diseases preferentially occur during the second half of life, counterintuitive to the concept of excess adaptive immunity driving immune-mediated tissue damage. T cells are particularly susceptible to aging-imposed changes, as they are under extreme proliferative pressure to fulfill the demands of clonal expansion and of homeostatic T cell repopulation. T cells in older adults have a footprint of genetic and epigenetic changes, lack mitochondrial fitness, and fail to maintain proteostasis, diverging them from host protection to host injury. Here, we review recent progress in understanding how the human T-cell system ages and the evidence detailing how T cell aging contributes to autoimmune conditions. T cell aging is now recognized as a risk determinant in two prototypic autoimmune syndromes; rheumatoid arthritis and giant cell arteritis. The emerging concept adds susceptibility to autoimmune and autoinflammatory disease to the spectrum of aging-imposed adaptations and opens new opportunities for immunomodulatory therapy by restoring the functional intactness of aging T cells.
Collapse
Affiliation(s)
- Qingxiang Liu
- Department of Medicine, Mayo Clinic Alix School of Medicine, Rochester, MN 55905, USA; Department of Immunology, Mayo Clinic College of Medicine and Science, Rochester, MN, USA
| | - Yanyan Zheng
- Department of Medicine, Mayo Clinic Alix School of Medicine, Rochester, MN 55905, USA; Department of Immunology, Mayo Clinic College of Medicine and Science, Rochester, MN, USA; Department of Cardiovascular Medicine, Mayo Alix School of Medicine, Rochester, MN, USA
| | - Jorg J Goronzy
- Department of Medicine, Mayo Clinic Alix School of Medicine, Rochester, MN 55905, USA; Department of Immunology, Mayo Clinic College of Medicine and Science, Rochester, MN, USA; Department of Medicine, Stanford University School of Medicine, Stanford, CA 94306, USA
| | - Cornelia M Weyand
- Department of Medicine, Mayo Clinic Alix School of Medicine, Rochester, MN 55905, USA; Department of Immunology, Mayo Clinic College of Medicine and Science, Rochester, MN, USA; Department of Cardiovascular Medicine, Mayo Alix School of Medicine, Rochester, MN, USA; Department of Medicine, Stanford University School of Medicine, Stanford, CA 94306, USA.
| |
Collapse
|
13
|
Moulana M. Androgen-Induced Cardiovascular Risk in Polycystic Ovary Syndrome: The Role of T Lymphocytes. Life (Basel) 2023; 13:life13041010. [PMID: 37109539 PMCID: PMC10145997 DOI: 10.3390/life13041010] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2023] [Revised: 04/10/2023] [Accepted: 04/11/2023] [Indexed: 04/29/2023] Open
Abstract
An estimated 15-20% of reproductive-age women are affected by polycystic ovary syndrome (PCOS). PCOS is associated with substantial metabolic and cardiovascular long-term consequences. In young women with PCOS, several cardiovascular risk factors may be found, including chronic inflammation, high blood pressure, and elevated leukocytes. These women are at an increased risk of cardiovascular diseases (CVD), not only during the reproductive years, but also with aging and menopause; therefore, the early prevention and treatment of future cardiovascular adverse effects are necessary. The fundamental characteristic of PCOS is hyperandrogenemia, which is associated with increased pro-inflammatory cytokines and T lymphocytes. Whether these factors play a role in the pathophysiology of hypertension, a risk factor of CVD, due to PCOS is not well established. This review will briefly discuss how a modest increase in androgens in females is linked to the development of hypertension through pro-inflammatory cytokines and T lymphocyte subsets and the promotion of renal injury. Moreover, it reveals a few existing research gaps in this area, including the lack of specific therapy directed at androgen-induced inflammation and immune activation, thus emphasizing the necessity to explore the systemic inflammation in women with PCOS to halt the inevitable inflammatory process targeting the underlying abnormalities of CVD.
Collapse
Affiliation(s)
- Mohadetheh Moulana
- Department of Psychiatry and Human Behavior, Women's Health Research Center, University of Mississippi Medical Center, 2500 North State Street, Jackson, MS 39216, USA
| |
Collapse
|
14
|
Booth LK, Redgrave RE, Tual-Chalot S, Spyridopoulos I, Phillips HM, Richardson GD. Heart Disease and Ageing: The Roles of Senescence, Mitochondria, and Telomerase in Cardiovascular Disease. Subcell Biochem 2023; 103:45-78. [PMID: 37120464 DOI: 10.1007/978-3-031-26576-1_4] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/01/2023]
Abstract
During ageing molecular damage leads to the accumulation of several hallmarks of ageing including mitochondrial dysfunction, cellular senescence, genetic instability and chronic inflammation, which contribute to the development and progression of ageing-associated diseases including cardiovascular disease. Consequently, understanding how these hallmarks of biological ageing interact with the cardiovascular system and each other is fundamental to the pursuit of improving cardiovascular health globally. This review provides an overview of our current understanding of how candidate hallmarks contribute to cardiovascular diseases such as atherosclerosis, coronary artery disease and subsequent myocardial infarction, and age-related heart failure. Further, we consider the evidence that, even in the absence of chronological age, acute cellular stress leading to accelerated biological ageing expedites cardiovascular dysfunction and impacts on cardiovascular health. Finally, we consider the opportunities that modulating hallmarks of ageing offer for the development of novel cardiovascular therapeutics.
Collapse
Affiliation(s)
- Laura K Booth
- Translational and Clinical Research Institute, Vascular Biology and Medicine Theme, Newcastle University, Newcastle upon Tyne, UK
| | - Rachael E Redgrave
- Biosciences Institute, Vascular Biology and Medicine Theme, Newcastle University, Newcastle upon Tyne, UK
| | - Simon Tual-Chalot
- Biosciences Institute, Vascular Biology and Medicine Theme, Newcastle University, Newcastle upon Tyne, UK
| | - Ioakim Spyridopoulos
- Translational and Clinical Research Institute, Vascular Biology and Medicine Theme, Newcastle University, Newcastle upon Tyne, UK
| | - Helen M Phillips
- Biosciences Institute, Vascular Biology and Medicine Theme, Newcastle University, Newcastle upon Tyne, UK
| | - Gavin D Richardson
- Biosciences Institute, Vascular Biology and Medicine Theme, Newcastle University, Newcastle upon Tyne, UK.
| |
Collapse
|
15
|
Chi M, Tian Z, Ma K, Li Y, Wang L, Nasser MI, Liu C. The diseased kidney: aging and senescent immunology. IMMUNITY & AGEING 2022; 19:58. [PMCID: PMC9666969 DOI: 10.1186/s12979-022-00313-9] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/11/2022] [Accepted: 10/23/2022] [Indexed: 11/17/2022]
Abstract
AbstractImmunosenescence is the deterioration of the innate and adaptive immune systems associated with aging and is primarily characterized by a reduction in T cell production and accumulation of atypical subsets. Age-related immunological dysfunction leads to impaired immune protection and persistent low-grade chronic inflammation, resulting in a decreased vaccination response and increased vulnerability to infection, cancer, cardiovascular disease, and autoimmune disease in the elderly. As the elderly constitute a growing proportion of the population with renal disease, immunosenescence is a normal aging process that is prevalent among older people. In addition, immunosenescence seems to be more pronounced in patients with kidney diseases than in healthy controls, as shown by severe chronic inflammation, accumulation of immune cells with the senescent phenotype (CD28− T cells, CD14+CD16+ monocytes), and proinflammatory cytokine production. Immunosenescence inhibits immunological clearance and renal tissue regeneration, thereby increasing the risk of permanent renal damage, infection, and cardiovascular events in patients with kidney disease, lowering the prognosis, and even influencing the efficacy of renal replacement treatment. Biological drugs (senomorphics and senolytics) target the aging immune system and exert renoprotective effects. This review aims to emphasize the features of immunosenescence and its influence on kidney diseases and immunotherapy, highlighting the future directions of kidney disease treatment using senescence-focused techniques.
Collapse
|
16
|
Tian W, Zhang T, Wang X, Zhang J, Ju J, Xu H. Global research trends in atherosclerosis: A bibliometric and visualized study. Front Cardiovasc Med 2022; 9:956482. [PMID: 36082127 PMCID: PMC9445883 DOI: 10.3389/fcvm.2022.956482] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2022] [Accepted: 08/03/2022] [Indexed: 11/13/2022] Open
Abstract
BackgroundIncreasing evidence has spurred a considerable evolution of concepts related to atherosclerosis, prompting the need to provide a comprehensive view of the growing literature. By retrieving publications in the Web of Science Core Collection (WoSCC) of Clarivate Analytics, we conducted a bibliometric analysis of the scientific literature on atherosclerosis to describe the research landscape.MethodsA search was conducted of the WoSCC for articles and reviews serving exclusively as a source of information on atherosclerosis published between 2012 and 2022. Microsoft Excel 2019 was used to chart the annual productivity of research relevant to atherosclerosis. Through CiteSpace and VOSviewer, the most prolific countries or regions, authors, journals, and resource-, intellectual-, and knowledge-sharing in atherosclerosis research, as well as co-citation analysis of references and keywords, were analyzed.ResultsA total of 20,014 publications were retrieved. In terms of publications, the United States remains the most productive country (6,390, 31,93%). The most publications have been contributed by Johns Hopkins Univ (730, 3.65%). ALVARO ALONSO produced the most published works (171, 0.85%). With a betweenness centrality of 0.17, ERIN D MICHOS was the most influential author. The most prolific journal was identified as Atherosclerosis (893, 4.46%). Circulation received the most co-citations (14,939, 2.79%). Keywords with the ongoing strong citation bursts were “nucleotide-binding oligomerization (NOD), Leucine-rich repeat (LRR)-containing protein (NLRP3) inflammasome,” “short-chain fatty acids (SCFAs),” “exosome,” and “homeostasis,” etc.ConclusionThe research on atherosclerosis is driven mostly by North America and Europe. Intensive research has focused on the link between inflammation and atherosclerosis, as well as its complications. Specifically, the NLRP3 inflammasome, interleukin-1β, gut microbiota and SCFAs, exosome, long non-coding RNAs, autophagy, and cellular senescence were described to be hot issues in the field.
Collapse
Affiliation(s)
- Wende Tian
- Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
- Graduate School, China Academy of Chinese Medical Sciences, Beijing, China
- National Clinical Research Center for Chinese Medicine Cardiology, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Tai Zhang
- Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
- Graduate School, China Academy of Chinese Medical Sciences, Beijing, China
- Department of Gastroenterology, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Xinyi Wang
- Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
- Graduate School, China Academy of Chinese Medical Sciences, Beijing, China
- National Clinical Research Center for Chinese Medicine Cardiology, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Jie Zhang
- National Clinical Research Center for Chinese Medicine Cardiology, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
- Graduate School, Beijing University of Chinese Medicine, Beijing, China
| | - Jianqing Ju
- Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
- National Clinical Research Center for Chinese Medicine Cardiology, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
- *Correspondence: Jianqing Ju,
| | - Hao Xu
- Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
- National Clinical Research Center for Chinese Medicine Cardiology, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
- Hao Xu,
| |
Collapse
|
17
|
Ghamar Talepoor A, Doroudchi M. Immunosenescence in atherosclerosis: A role for chronic viral infections. Front Immunol 2022; 13:945016. [PMID: 36059478 PMCID: PMC9428721 DOI: 10.3389/fimmu.2022.945016] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2022] [Accepted: 07/26/2022] [Indexed: 01/10/2023] Open
Abstract
Immune system is a versatile and dynamic body organ which offers survival and endurance of human beings in their hostile living environment. However, similar to other cells, immune cells are hijacked by senescence. The ageing immune cells lose their beneficial functions but continue to produce inflammatory mediators which draw other immune and non-immune cells to the senescence loop. Immunosenescence has been shown to be associated with different pathological conditions and diseases, among which atherosclerosis has recently come to light. There are common drivers of both immunosenescence and atherosclerosis; e.g. inflammation, reactive oxygen species (ROS), chronic viral infections, genomic damage, oxidized-LDL, hypertension, cigarette smoke, hyperglycaemia, and mitochondrial failure. Chronic viral infections induce inflammaging, sustained cytokine signaling, ROS generation and DNA damage which are associated with atherogenesis. Accumulating evidence shows that several DNA and RNA viruses are stimulators of immunosenescence and atherosclerosis in an interrelated network. DNA viruses such as CMV, EBV and HBV upregulate p16, p21 and p53 senescence-associated molecules; induce inflammaging, metabolic reprogramming of infected cells, replicative senescence and telomere shortening. RNA viruses such as HCV and HIV induce ROS generation, DNA damage, induction of senescence-associated secretory phenotype (SASP), metabolic reprogramming of infected cells, G1 cell cycle arrest, telomere shortening, as well as epigenetic modifications of DNA and histones. The newly emerged SARS-CoV-2 virus is also a potent inducer of cytokine storm and SASP. The spike protein of SARS-CoV-2 promotes senescence phenotype in endothelial cells by augmenting p16, p21, senescence-associated β-galactosidase (SA-β-Gal) and adhesion molecules expression. The impact of SARS-CoV-2 mega-inflammation on atherogenesis, however, remains to be investigated. In this review we focus on the common processes in immunosenescence and atherogenesis caused by chronic viral infections and discuss the current knowledge on this topic.
Collapse
|
18
|
Krüger K, Tirekoglou P, Weyh C. Immunological mechanisms of exercise therapy in dyslipidemia. Front Physiol 2022; 13:903713. [PMID: 36003652 PMCID: PMC9393246 DOI: 10.3389/fphys.2022.903713] [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: 03/24/2022] [Accepted: 07/07/2022] [Indexed: 12/05/2022] Open
Abstract
Numerous studies demonstrated the strong link between dyslipidemia and the cardiovascular risk. Physical activity and exercise represent effective prevention and therapy strategies for dyslipidemia and at the same time counteract numerous comorbidities that often accompany the disease. The physiological mechanisms are manifold, and primary mechanisms might be an increased energy consumption and associated adaptations of the substrate metabolism. Recent studies showed that there are bidirectional interactions between dyslipidemia and the immune system. Thus, abnormal blood lipids may favor pro-inflammatory processes, and at the same time inflammatory processes may also promote dyslipidemia. Physical activity has been shown to affect numerous immunological processes and has primarily anti-inflammatory effects. These are manifested by altered leukocyte subtypes, cytokine patterns, stress protein expression, and by reducing hallmarks of immunosenescence. The aim of this review is to describe the effects of exercise on the treatment dyslipidemia and to discuss possible immunological mechanisms against the background of the current literature.
Collapse
Affiliation(s)
- Karsten Krüger
- Department of Exercise Physiology and Sports Therapy, Institute of Sport Science, Justus-Liebig-University Giessen, Giessen, Germany
| | | | | |
Collapse
|
19
|
Hoeks C, Duran G, Hellings N, Broux B. When Helpers Go Above and Beyond: Development and Characterization of Cytotoxic CD4+ T Cells. Front Immunol 2022; 13:951900. [PMID: 35903098 PMCID: PMC9320319 DOI: 10.3389/fimmu.2022.951900] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2022] [Accepted: 06/21/2022] [Indexed: 11/26/2022] Open
Abstract
Once regarded as an experimental artefact, cytotoxic CD4+ T cells (CD4 CTL) are presently recognized as a biologically relevant T cell subset with important functions in anti-viral, anti-tumor, and autoimmune responses. Despite the potentially large impact on their micro-environment, the absolute cell counts of CD4 CTL within the peripheral circulation are relatively low. With the rise of single cell analysis techniques, detection of these cells is greatly facilitated. This led to a renewed appraisal of CD4 CTL and an increased insight into their heterogeneous nature and ontogeny. In this review, we summarize the developmental path from naïve CD4+ T cells to terminally differentiated CD4 CTL, and present markers that can be used to detect or isolate CD4 CTL and their precursors. Subsets of CD4 CTL and their divergent functionalities are discussed. Finally, the importance of local cues as triggers for CD4 CTL differentiation is debated, posing the question whether CD4 CTL develop in the periphery and migrate to site of inflammation when called for, or that circulating CD4 CTL reflect cells that returned to the circulation following differentiation at the local inflammatory site they previously migrated to. Even though much remains to be learned about this intriguing T cell subset, it is clear that CD4 CTL represent interesting therapeutic targets for several pathologies.
Collapse
Affiliation(s)
- Cindy Hoeks
- Neuro Immune Connections & Repair Lab, Department of Immunology and Infection, Biomedical Research Institute, Hasselt University, Hasselt, Belgium
- University MS Center (UMSC), Hasselt, Belgium
| | - Gayel Duran
- Neuro Immune Connections & Repair Lab, Department of Immunology and Infection, Biomedical Research Institute, Hasselt University, Hasselt, Belgium
- University MS Center (UMSC), Hasselt, Belgium
| | - Niels Hellings
- Neuro Immune Connections & Repair Lab, Department of Immunology and Infection, Biomedical Research Institute, Hasselt University, Hasselt, Belgium
- University MS Center (UMSC), Hasselt, Belgium
| | - Bieke Broux
- Neuro Immune Connections & Repair Lab, Department of Immunology and Infection, Biomedical Research Institute, Hasselt University, Hasselt, Belgium
- University MS Center (UMSC), Hasselt, Belgium
- *Correspondence: Bieke Broux,
| |
Collapse
|
20
|
Bazioti V, La Rose AM, Maassen S, Bianchi F, de Boer R, Halmos B, Dabral D, Guilbaud E, Flohr-Svendsen A, Groenen AG, Marmolejo-Garza A, Koster MH, Kloosterhuis NJ, Havinga R, Pranger AT, Langelaar-Makkinje M, de Bruin A, van de Sluis B, Kohan AB, Yvan-Charvet L, van den Bogaart G, Westerterp M. T cell cholesterol efflux suppresses apoptosis and senescence and increases atherosclerosis in middle aged mice. Nat Commun 2022; 13:3799. [PMID: 35778407 PMCID: PMC9249754 DOI: 10.1038/s41467-022-31135-4] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2021] [Accepted: 05/27/2022] [Indexed: 02/04/2023] Open
Abstract
Atherosclerosis is a chronic inflammatory disease driven by hypercholesterolemia. During aging, T cells accumulate cholesterol, potentially affecting inflammation. However, the effect of cholesterol efflux pathways mediated by ATP-binding cassette A1 and G1 (ABCA1/ABCG1) on T cell-dependent age-related inflammation and atherosclerosis remains poorly understood. In this study, we generate mice with T cell-specific Abca1/Abcg1-deficiency on the low-density-lipoprotein-receptor deficient (Ldlr-/-) background. T cell Abca1/Abcg1-deficiency decreases blood, lymph node, and splenic T cells, and increases T cell activation and apoptosis. T cell Abca1/Abcg1-deficiency induces a premature T cell aging phenotype in middle-aged (12-13 months) Ldlr-/- mice, reflected by upregulation of senescence markers. Despite T cell senescence and enhanced T cell activation, T cell Abca1/Abcg1-deficiency decreases atherosclerosis and aortic inflammation in middle-aged Ldlr-/- mice, accompanied by decreased T cells in atherosclerotic plaques. We attribute these effects to T cell apoptosis downstream of T cell activation, compromising T cell functionality. Collectively, we show that T cell cholesterol efflux pathways suppress T cell apoptosis and senescence, and induce atherosclerosis in middle-aged Ldlr-/- mice.
Collapse
Affiliation(s)
- Venetia Bazioti
- grid.4494.d0000 0000 9558 4598Department of Pediatrics, University Medical Center Groningen, University of Groningen, 9713 AV Groningen, the Netherlands ,grid.5252.00000 0004 1936 973XInstitute for Cardiovascular Prevention (IPEK), Ludwig-Maximilians-Universität, 80336 Munich, Germany
| | - Anouk M. La Rose
- grid.4494.d0000 0000 9558 4598Department of Pediatrics, University Medical Center Groningen, University of Groningen, 9713 AV Groningen, the Netherlands
| | - Sjors Maassen
- grid.4830.f0000 0004 0407 1981Department of Molecular Immunology and Microbiology, Groningen Biomolecular Sciences and Biotechnology Institute, University of Groningen, 9747 AG Groningen, the Netherlands
| | - Frans Bianchi
- grid.4830.f0000 0004 0407 1981Department of Molecular Immunology and Microbiology, Groningen Biomolecular Sciences and Biotechnology Institute, University of Groningen, 9747 AG Groningen, the Netherlands
| | - Rinse de Boer
- grid.4830.f0000 0004 0407 1981Department of Molecular Immunology and Microbiology, Groningen Biomolecular Sciences and Biotechnology Institute, University of Groningen, 9747 AG Groningen, the Netherlands
| | - Benedek Halmos
- grid.4494.d0000 0000 9558 4598Department of Pediatrics, University Medical Center Groningen, University of Groningen, 9713 AV Groningen, the Netherlands
| | - Deepti Dabral
- grid.4830.f0000 0004 0407 1981Department of Molecular Immunology and Microbiology, Groningen Biomolecular Sciences and Biotechnology Institute, University of Groningen, 9747 AG Groningen, the Netherlands
| | - Emma Guilbaud
- grid.462370.40000 0004 0620 5402Institut National de la Santé et de la Recherche Médicale (INSERM) U1065, Université Côte d’Azur, Centre Méditerranéen de Médecine Moléculaire (C3M), Atip-Avenir, Fédération Hospitalo-Universitaire (FHU) Oncoage, 06204 Nice, France
| | - Arthur Flohr-Svendsen
- grid.4494.d0000 0000 9558 4598European Research Institute for the Biology of Ageing, University Medical Center Groningen, University of Groningen, 9713 AV Groningen, the Netherlands
| | - Anouk G. Groenen
- grid.4494.d0000 0000 9558 4598Department of Pediatrics, University Medical Center Groningen, University of Groningen, 9713 AV Groningen, the Netherlands
| | - Alejandro Marmolejo-Garza
- grid.4494.d0000 0000 9558 4598Department of Pediatrics, University Medical Center Groningen, University of Groningen, 9713 AV Groningen, the Netherlands
| | - Mirjam H. Koster
- grid.4494.d0000 0000 9558 4598Department of Pediatrics, University Medical Center Groningen, University of Groningen, 9713 AV Groningen, the Netherlands
| | - Niels J. Kloosterhuis
- grid.4494.d0000 0000 9558 4598Department of Pediatrics, University Medical Center Groningen, University of Groningen, 9713 AV Groningen, the Netherlands
| | - Rick Havinga
- grid.4494.d0000 0000 9558 4598Department of Pediatrics, University Medical Center Groningen, University of Groningen, 9713 AV Groningen, the Netherlands
| | - Alle T. Pranger
- grid.4494.d0000 0000 9558 4598Laboratory of Medicine, University Medical Center Groningen, University of Groningen, 9713 AV Groningen, the Netherlands
| | - Miriam Langelaar-Makkinje
- grid.4494.d0000 0000 9558 4598Department of Pediatrics, University Medical Center Groningen, University of Groningen, 9713 AV Groningen, the Netherlands
| | - Alain de Bruin
- grid.4494.d0000 0000 9558 4598Department of Pediatrics, University Medical Center Groningen, University of Groningen, 9713 AV Groningen, the Netherlands ,grid.5477.10000000120346234Department of Biomolecular Health Sciences, Dutch Molecular Pathology Center, Faculty of Veterinary Medicine, Utrecht University, 3584 CL Utrecht, the Netherlands
| | - Bart van de Sluis
- grid.4494.d0000 0000 9558 4598Department of Pediatrics, University Medical Center Groningen, University of Groningen, 9713 AV Groningen, the Netherlands
| | - Alison B. Kohan
- grid.21925.3d0000 0004 1936 9000Division of Endocrinology and Metabolism, Department of Medicine, University of Pittsburgh, Pittsburgh, PA 15260 USA
| | - Laurent Yvan-Charvet
- grid.462370.40000 0004 0620 5402Institut National de la Santé et de la Recherche Médicale (INSERM) U1065, Université Côte d’Azur, Centre Méditerranéen de Médecine Moléculaire (C3M), Atip-Avenir, Fédération Hospitalo-Universitaire (FHU) Oncoage, 06204 Nice, France
| | - Geert van den Bogaart
- grid.4830.f0000 0004 0407 1981Department of Molecular Immunology and Microbiology, Groningen Biomolecular Sciences and Biotechnology Institute, University of Groningen, 9747 AG Groningen, the Netherlands
| | - Marit Westerterp
- grid.4494.d0000 0000 9558 4598Department of Pediatrics, University Medical Center Groningen, University of Groningen, 9713 AV Groningen, the Netherlands
| |
Collapse
|
21
|
Ng C, Lee KL, Muthiah MD, Wu KX, Chioh FWJ, Tan K, Soon GST, Shabbir A, Loo WM, Low ZS, Chen Q, Tan NS, Ng HH, Dan YY, Cheung C. Endothelial‐immune crosstalk contributes to vasculopathy in nonalcoholic fatty liver disease. EMBO Rep 2022; 23:e54271. [PMID: 35403791 PMCID: PMC9171677 DOI: 10.15252/embr.202154271] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2021] [Revised: 03/19/2022] [Accepted: 03/24/2022] [Indexed: 11/17/2022] Open
Abstract
The top cause of mortality in patients with nonalcoholic fatty liver disease (NAFLD) is cardiovascular complications. However, mechanisms of NAFLD‐associated vasculopathy remain understudied. Here, we show that blood outgrowth endothelial cells (BOECs) from NAFLD subjects exhibit global transcriptional upregulation of chemokines and human leukocyte antigens. In mouse models of diet‐induced NAFLD, we confirm heightened endothelial expressions of CXCL12 in the aortas and the liver vasculatures, and increased retention of infiltrated leukocytes within the vessel walls. To elucidate endothelial‐immune crosstalk, we performed immunoprofiling by single‐cell analysis, uncovering T cell intensification in NAFLD patients. Functionally, treatment with a CXCL12‐neutralizing antibody is effective at moderating the enhanced chemotactic effect of NAFLD BOECs in recruiting CD8+ T lymphocytes. Interference with the CXCL12‐CXCR4 axis using a CXCR4 antagonist also averts the impact of immune cell transendothelial migration and restores endothelial barrier integrity. Clinically, we detect threefold more circulating damaged endothelial cells in NAFLD patients than in healthy controls. Our work provides insight into the modulation of interactions with effector immune cells to mitigate endothelial injury in NAFLD.
Collapse
Affiliation(s)
- Chun‐Yi Ng
- Lee Kong Chian School of Medicine Nanyang Technological University Singapore Singapore
| | - Khang Leng Lee
- Lee Kong Chian School of Medicine Nanyang Technological University Singapore Singapore
| | - Mark Dhinesh Muthiah
- Yong Loo Lin School of Medicine National University of Singapore Singapore Singapore
- Department of Medicine National University Health System Singapore Singapore
| | - Kan Xing Wu
- Lee Kong Chian School of Medicine Nanyang Technological University Singapore Singapore
| | | | - Konstanze Tan
- Lee Kong Chian School of Medicine Nanyang Technological University Singapore Singapore
| | | | - Asim Shabbir
- Yong Loo Lin School of Medicine National University of Singapore Singapore Singapore
- Department of Surgery University Surgical Cluster National University Health System Singapore Singapore
| | - Wai Mun Loo
- Department of Medicine National University Health System Singapore Singapore
| | - Zun Siong Low
- Lee Kong Chian School of Medicine Nanyang Technological University Singapore Singapore
| | - Qingfeng Chen
- Institute of Molecular and Cell Biology Agency for Science Technology and Research (A*STAR) Singapore Singapore
| | - Nguan Soon Tan
- Lee Kong Chian School of Medicine Nanyang Technological University Singapore Singapore
- School of Biological Sciences Nanyang Technological University Singapore Singapore
| | - Huck Hui Ng
- Lee Kong Chian School of Medicine Nanyang Technological University Singapore Singapore
- Yong Loo Lin School of Medicine National University of Singapore Singapore Singapore
- Institute of Molecular and Cell Biology Agency for Science Technology and Research (A*STAR) Singapore Singapore
- School of Biological Sciences Nanyang Technological University Singapore Singapore
- Genome Institute of Singapore Agency for Science Technology and Research (A*STAR) Singapore Singapore
| | - Yock Young Dan
- Yong Loo Lin School of Medicine National University of Singapore Singapore Singapore
- Department of Medicine National University Health System Singapore Singapore
| | - Christine Cheung
- Lee Kong Chian School of Medicine Nanyang Technological University Singapore Singapore
- Institute of Molecular and Cell Biology Agency for Science Technology and Research (A*STAR) Singapore Singapore
| |
Collapse
|
22
|
Zhao TV, Sato Y, Goronzy JJ, Weyand CM. T-Cell Aging-Associated Phenotypes in Autoimmune Disease. FRONTIERS IN AGING 2022; 3:867950. [PMID: 35821833 PMCID: PMC9261367 DOI: 10.3389/fragi.2022.867950] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/01/2022] [Accepted: 03/28/2022] [Indexed: 01/10/2023]
Abstract
The aging process causes profound restructuring of the host immune system, typically associated with declining host protection against cancer and infection. In the case of T cells, aging leads to the accumulation of a diverse set of T-cell aging-associated phenotypes (TASP), some of which have been implicated in driving tissue inflammation in autoimmune diseases. T cell aging as a risk determinant for autoimmunity is exemplified in two classical autoimmune conditions: rheumatoid arthritis (RA), a disease predominantly affecting postmenopausal women, and giant cell arteritis (GCA), an inflammatory vasculopathy exclusively occurring during the 6th-9th decade of life. Pathogenic T cells in RA emerge as a consequence of premature immune aging. They have shortening and fragility of telomeric DNA ends and instability of mitochondrial DNA. As a result, they produce a distinct profile of metabolites, disproportionally expand their endoplasmic reticulum (ER) membranes and release excess amounts of pro-inflammatory effector cytokines. Characteristically, they are tissue invasive, activate the inflammasome and die a pyroptotic death. Patients with GCA expand pathogenic CD4+ T cells due to aberrant expression of the co-stimulatory receptor NOTCH1 and the failure of the PD-1/PD-L1 immune checkpoint. In addition, GCA patients lose anti-inflammatory Treg cells, promoting tissue-destructive granulomatous vasculitis. In summary, emerging data identify T cell aging as a risk factor for autoimmune disease and directly link TASPs to the breakdown of T cell tolerance and T-cell-induced tissue inflammation.
Collapse
Affiliation(s)
- Tuantuan V. Zhao
- Mayo Clinic Alix School of Medicine, College of Medicine and Science, Rochester, MN, United States
| | - Yuki Sato
- Mayo Clinic Alix School of Medicine, College of Medicine and Science, Rochester, MN, United States
| | - Jorg J. Goronzy
- Mayo Clinic Alix School of Medicine, College of Medicine and Science, Rochester, MN, United States
- School of Medicine, Stanford University, Stanford, CA, United States
| | - Cornelia M. Weyand
- Mayo Clinic Alix School of Medicine, College of Medicine and Science, Rochester, MN, United States
- School of Medicine, Stanford University, Stanford, CA, United States
| |
Collapse
|
23
|
Liu D, Liu J, Zhang D, Yang W. Advances in relationship between cell senescence and atherosclerosis. Zhejiang Da Xue Xue Bao Yi Xue Ban 2022; 51:95-101. [PMID: 35576118 PMCID: PMC9109755 DOI: 10.3724/zdxbyxb-2021-0270] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2021] [Accepted: 11/03/2021] [Indexed: 06/15/2023]
Abstract
Cellular senescence is a biological process associated with the degeneration of cell structure and function, which contribute to age-related diseases. Atherosclerosis is a chronic inflammatory disease that can cause a variety of cardiovascular disorders. In this article, we review the effects of cellular senescence on the development of atherosclerosis through diverse physiopathological changes, focusing on the alterations in senescent organelles and the increased senescence-associated secretory phenotype (SASP), and exploring the relevant therapeutic strategies for atherosclerosis by clearing senescent cells and reducing SASP, to provide new insights for the treatment of atherosclerosis.
Collapse
Affiliation(s)
- Dekun Liu
- 1. Faculty of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan 250300, China
| | - Jiali Liu
- 1. Faculty of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan 250300, China
| | - Dan Zhang
- 3. Innovation Institute of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan 250300, China
| | - Wenqing Yang
- 2. Experimental Center, Shandong University of Traditional Chinese Medicine, Jinan 250300, China
| |
Collapse
|
24
|
Abstract
Two vasculitides, giant cell arteritis (GCA) and Takayasu arteritis (TAK), are recognized as autoimmune and autoinflammatory diseases that manifest exclusively within the aorta and its large branches. In both entities, the age of the affected host is a critical risk factor. TAK manifests during the 2nd-4th decade of life, occurring while the immune system is at its height of performance. GCA is a disease of older individuals, with infrequent cases during the 6th decade and peak incidence during the 8th decade of life. In both vasculitides, macrophages and T cells infiltrate into the adventitia and media of affected vessels, induce granulomatous inflammation, cause vessel wall destruction, and reprogram vascular cells to drive adventitial and neointimal expansion. In GCA, abnormal immunity originates in an aged immune system and evolves within the aged vascular microenvironment. One hallmark of the aging immune system is the preferential loss of CD8+ T cell function. Accordingly, in GCA but not in TAK, CD8+ effector T cells play a negligible role and anti-inflammatory CD8+ T regulatory cells are selectively impaired. Here, we review current evidence of how the process of immunosenescence impacts the risk for GCA and how fundamental differences in the age of the immune system translate into differences in the granulomatous immunopathology of TAK versus GCA.
Collapse
|
25
|
Herman AB, Occean JR, Sen P. Epigenetic dysregulation in cardiovascular aging and disease. THE JOURNAL OF CARDIOVASCULAR AGING 2021; 1. [PMID: 34790973 PMCID: PMC8594871 DOI: 10.20517/jca.2021.16] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Cardiovascular disease (CVD) is the leading cause of mortality and morbidity for all sexes, racial and ethnic groups. Age, and its associated physiological and pathological consequences, exacerbate CVD incidence and progression, while modulation of biological age with interventions track with cardiovascular health. Despite the strong link between aging and CVD, surprisingly few studies have directly investigated heart failure and vascular dysfunction in aged models and subjects. Nevertheless, strong correlations have been found between heart disease, atherosclerosis, hypertension, fibrosis, and regeneration efficiency with senescent cell burden and its proinflammatory sequelae. In agreement, senotherapeutics have had success in reducing the detrimental effects in experimental models of cardiovascular aging and disease. Aside from senotherapeutics, cellular reprogramming strategies targeting epigenetic enzymes remain an unexplored yet viable option for reversing or delaying CVD. Epigenetic alterations comprising local and global changes in DNA and histone modifications, transcription factor binding, disorganization of the nuclear lamina, and misfolding of the genome are hallmarks of aging. Limited studies in the aging cardiovascular system of murine models or human patient samples have identified strong correlations between the epigenome, age, and senescence. Here, we compile the findings in published studies linking epigenetic changes to CVD and identify clear themes of epigenetic deregulation during aging. Pending direct investigation of these general mechanisms in aged tissues, this review predicts that future work will establish epigenetic rejuvenation as a potent method to delay CVD.
Collapse
Affiliation(s)
- Allison B Herman
- Laboratory of Genetics and Genomics, National Institute on Aging, NIH, Baltimore, MD 21224, USA
| | - James R Occean
- Laboratory of Genetics and Genomics, National Institute on Aging, NIH, Baltimore, MD 21224, USA
| | - Payel Sen
- Laboratory of Genetics and Genomics, National Institute on Aging, NIH, Baltimore, MD 21224, USA
| |
Collapse
|
26
|
Rao M, Wang X, Guo G, Wang L, Chen S, Yin P, Chen K, Chen L, Zhang Z, Chen X, Hu X, Hu S, Song J. Resolving the intertwining of inflammation and fibrosis in human heart failure at single-cell level. Basic Res Cardiol 2021; 116:55. [PMID: 34601654 DOI: 10.1007/s00395-021-00897-1] [Citation(s) in RCA: 104] [Impact Index Per Article: 26.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/05/2021] [Revised: 09/13/2021] [Accepted: 09/21/2021] [Indexed: 12/13/2022]
Abstract
Inflammation and fibrosis are intertwined mechanisms fundamentally involved in heart failure. Detailed deciphering gene expression perturbations and cell-cell interactions of leukocytes and non-myocytes is required to understand cell-type-specific pathology in the failing human myocardium. To this end, we performed single-cell RNA sequencing and single T cell receptor sequencing of 200,615 cells in both human dilated cardiomyopathy (DCM) and ischemic cardiomyopathy (ICM) hearts. We sampled both lesion and mild-lesion tissues from each heart to sequentially capture cellular and molecular alterations to different extents of cardiac fibrosis. By which, left (lesion) and right ventricle (mild-lesion) for DCM hearts were harvest while infarcted (lesion) and non-infarcted area (mild-lesion) were dissected from ICM hearts. A novel transcription factor AEBP1 was identified as a crucial cardiac fibrosis regulator in ACTA2+ myofibroblasts. Within fibrotic myocardium, an infiltration of a considerable number of leukocytes was witnessed, especially cytotoxic and exhausted CD8+ T cells and pro-inflammatory CD4+ T cells. Furthermore, a subset of tissue-resident macrophage, CXCL8hiCCR2+HLA-DRhi macrophage was particularly identified in severely fibrotic area, which interacted with activated endothelial cell via DARC, that potentially facilitate leukocyte recruitment and infiltration in human heart failure.
Collapse
Affiliation(s)
- Man Rao
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, Chinese Academy of Medical Science, PUMC, 167 Beilishi Road, Xicheng District, Beijing, 100037, China.,National Clinical Research Center for Orthopedics, Sports Medicine and Rehabilitation, General Hospital of Chinese PLA, Beijing, 100039, China
| | - Xiliang Wang
- BIOPIC and School of Life Sciences, Peking University, No. 5 Yiheyuan Road, Haidian District, Beijing, 100871, China.,Analytical Biosciences Limited, Beijing, 100084, China
| | - Guangran Guo
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, Chinese Academy of Medical Science, PUMC, 167 Beilishi Road, Xicheng District, Beijing, 100037, China.,Department of Thoracic Surgery, Sun Yat-Sen University Cancer Center, Guangzhou, 510060, China
| | - Li Wang
- BIOPIC and School of Life Sciences, Peking University, No. 5 Yiheyuan Road, Haidian District, Beijing, 100871, China
| | - Shi Chen
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, Chinese Academy of Medical Science, PUMC, 167 Beilishi Road, Xicheng District, Beijing, 100037, China
| | - Pengbin Yin
- National Clinical Research Center for Orthopedics, Sports Medicine and Rehabilitation, General Hospital of Chinese PLA, Beijing, 100039, China
| | - Kai Chen
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, Chinese Academy of Medical Science, PUMC, 167 Beilishi Road, Xicheng District, Beijing, 100037, China
| | - Liang Chen
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, Chinese Academy of Medical Science, PUMC, 167 Beilishi Road, Xicheng District, Beijing, 100037, China
| | - Zemin Zhang
- BIOPIC and School of Life Sciences, Peking University, No. 5 Yiheyuan Road, Haidian District, Beijing, 100871, China.,Beijing Advanced Innovation Center for Genomics, Peking University, Beijing, 100871, China.,Peking-Tsinghua Center for Life Sciences, Peking University, Beijing, 100871, China
| | - Xiao Chen
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, Chinese Academy of Medical Science, PUMC, 167 Beilishi Road, Xicheng District, Beijing, 100037, China
| | - Xueda Hu
- BIOPIC and School of Life Sciences, Peking University, No. 5 Yiheyuan Road, Haidian District, Beijing, 100871, China. .,Analytical Biosciences Limited, Beijing, 100084, China.
| | - Shengshou Hu
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, Chinese Academy of Medical Science, PUMC, 167 Beilishi Road, Xicheng District, Beijing, 100037, China.
| | - Jiangping Song
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, Chinese Academy of Medical Science, PUMC, 167 Beilishi Road, Xicheng District, Beijing, 100037, China.
| |
Collapse
|
27
|
The impact of CD4 +CD28 null T lymphocytes on atrial fibrillation: a potential pathophysiological pathway. Inflamm Res 2021; 70:1011-1014. [PMID: 34536081 PMCID: PMC8572810 DOI: 10.1007/s00011-021-01502-w] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2021] [Revised: 09/04/2021] [Accepted: 09/07/2021] [Indexed: 11/12/2022] Open
Abstract
Introduction Atrial fibrillation (AF) represents the most common cardiac arrhythmia in daily clinical practice and substantially impacts affected patients by elevation of both morbidity and mortality. Previous investigations proved that inflammatory processes are closely linked to this multifactorial pathogenesis—especially autoreactive CD4+CD28null T cells received in-depth attention. Purpose Consequently, a potential pathophysiological pathway of the impact of CD4+CD28null T lymphocytes on the development and progression AF can be outlined. Conclusion Considering the available data in the literature, it needs to be assumed that CD4+CD28null T lymphocytes are mainly involved in the development of AF and disease progression. Of utmost importance, it can be considered as the result of a T-cell-mediated auto-immune reaction among myocardial tissue. However, mechanisms which recruit CD4+CD28null cells in cardiac tissue remain unclear and need further investigation.
Collapse
|
28
|
Rodríguez-Carrio J, Carrillo-López N, Ulloa C, Martín-Carro B, Rodríguez-Suárez C, Naves-Díaz M, Sánchez-Álvarez E, Rodríguez-García M, Arcidiacono MV, Fernández-Mariño B, Cannata-Andía JB, Suárez A, Dusso AS. Novel Immune Cell Subsets Exhibit Different Associations With Vascular Outcomes in Chronic Kidney Disease Patients-Identifying Potential Biomarkers. Front Med (Lausanne) 2021; 8:618286. [PMID: 34113627 PMCID: PMC8185045 DOI: 10.3389/fmed.2021.618286] [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: 10/16/2020] [Accepted: 04/07/2021] [Indexed: 11/17/2022] Open
Abstract
Background and Aims: Alterations in novel immune cell subsets, such as angiogenic T cells (Tang), senescent T cells (CD4+CD28null), and monocyte subsets are associated with impaired vascular homeostasis in several inflammatory conditions. However, mediators underlying vascular deterioration in chronic kidney disease (CKD) are poorly characterized. This study assessed their role in the vascular deterioration of CKD using a broad spectrum of surrogate markers ranging from altered functionality to overt calcification. Methods: Tang (CD3+CD31+CXCR4+), CD4+CD28null cells, and monocytes [CD14/CD16 subsets and angiotensin-converting enzyme (ACE) expression] were measured in peripheral blood by flow cytometry in 33 CKD stage 5 patients undergoing peritoneal dialysis (CKD5-PD) and 15 healthy controls (HCs). Analyses were replicated in a hemodialysis cohort. Vascular surrogate markers (including adventitial vasa vasorum, pulse wave velocity, intima-media thickness, and vascular calcification) were assessed by appropriate imaging methods. Results: In CKD5-PD, decreased Tang levels (p < 0.001) were unrelated to clinical features or traditional cardiovascular (CV) risk factors but correlated negatively with troponin T levels (r = −0.550, p = 0.003). Instead, CD4+CD28null frequency was increased (p < 0.001), especially in those with vascular calcifications. Quantitative and qualitative differences were also observed within the monocyte pool, a shift toward CD16+ subsets and ACE expression being found in CKD. Equivalent results were observed in the replication cohort. Each subset associated distinctly with adverse vascular outcomes in univariate and multivariate analyses: while Tang depletion was linked to poor vascular function and subclinical atherosclerosis, increases in CD4+CD28null were associated with overt vascular thickening and calcification. Monocytes were not independently associated with vascular outcomes in CKD patients. Conclusions: Novel T cell and monocyte subsets are altered in CKD. Altered T-cell subpopulations, but not monocytes, exhibited distinct associations with different vascular outcomes in CKD. Tang are emerging biomarkers of subclinical vascular deterioration in CKD.
Collapse
Affiliation(s)
- Javier Rodríguez-Carrio
- Area of Immunology, Department of Functional Biology, University of Oviedo, Oviedo, Spain.,Bone and Mineral Research Unit, Hospital Universitario Central de Asturias, Oviedo, Spain.,Instituto de Investigación Sanitaria del Principado de Asturias, Oviedo, Spain
| | - Natalia Carrillo-López
- Bone and Mineral Research Unit, Hospital Universitario Central de Asturias, Oviedo, Spain.,Instituto de Investigación Sanitaria del Principado de Asturias, Oviedo, Spain.,REDinREN-ISCIII, Oviedo, Spain
| | - Catalina Ulloa
- Bone and Mineral Research Unit, Hospital Universitario Central de Asturias, Oviedo, Spain.,Department of Nephrology, Hospital Universitario de Cabueñes, Gijón, Spain
| | - Beatriz Martín-Carro
- Bone and Mineral Research Unit, Hospital Universitario Central de Asturias, Oviedo, Spain.,Instituto de Investigación Sanitaria del Principado de Asturias, Oviedo, Spain.,REDinREN-ISCIII, Oviedo, Spain
| | | | - Manuel Naves-Díaz
- Bone and Mineral Research Unit, Hospital Universitario Central de Asturias, Oviedo, Spain.,Instituto de Investigación Sanitaria del Principado de Asturias, Oviedo, Spain.,REDinREN-ISCIII, Oviedo, Spain
| | - Emilio Sánchez-Álvarez
- REDinREN-ISCIII, Oviedo, Spain.,Department of Nephrology, Hospital Universitario de Cabueñes, Gijón, Spain
| | - Minerva Rodríguez-García
- REDinREN-ISCIII, Oviedo, Spain.,Division of Nephrology, Hospital Universitario Central de Asturias, Oviedo, Spain
| | | | | | - Jorge B Cannata-Andía
- Bone and Mineral Research Unit, Hospital Universitario Central de Asturias, Oviedo, Spain.,Instituto de Investigación Sanitaria del Principado de Asturias, Oviedo, Spain.,REDinREN-ISCIII, Oviedo, Spain.,Department of Medicine, University of Oviedo, Oviedo, Spain
| | - Ana Suárez
- Area of Immunology, Department of Functional Biology, University of Oviedo, Oviedo, Spain.,Department of Medicine, University of Oviedo, Oviedo, Spain
| | - Adriana S Dusso
- Bone and Mineral Research Unit, Hospital Universitario Central de Asturias, Oviedo, Spain.,Instituto de Investigación Sanitaria del Principado de Asturias, Oviedo, Spain.,REDinREN-ISCIII, Oviedo, Spain.,Department of Medicine, University of Oviedo, Oviedo, Spain
| |
Collapse
|
29
|
Formentini M, Navas A, Hassouneh F, Lopez-Sejas N, Alonso C, Tarazona R, Solana R, Pera A. Impact of CMV and age on T cell subsets defined by CD161, CD300a, and/or CD57 expression in healthy Andalusians. J Gerontol A Biol Sci Med Sci 2021; 76:1946-1953. [PMID: 33993242 DOI: 10.1093/gerona/glab140] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2020] [Indexed: 11/14/2022] Open
Abstract
Immunosenescence affects innate and adaptive immunity impairing the response to pathogens and vaccines. Chronic infection with cytomegalovirus (CMV) has been shown to drive 'early immunosenescence' and can considerably impact both the function and phenotype of immune cells, especially T cells. We have previously shown that the expression of CD57, CD300a, and CD161 was differentially affected by age and chronic CMV infection, indicating that these markers are a hallmark of CMV infection and T cell ageing. The aim of this present study was to clarify whether these three markers define distinct T cell subpopulations with a specific functional and molecular signature. Specifically, we analyzed the effect of age and chronic CMV infection on the functionality of T cells according to CD161, CD300a, and CD57 expression. We found that these markers defined different T cell subsets, both at the phenotypic and functional levels. CD57 was the best biomarker for CD4+ T cell cytotoxicity and was a hallmark of CMV infection. CD300a+ T cells were heterogeneous and included different cell subsets. The population of CD161+ T cells dramatically decreased with age, independently of CMV infection, and represented a sign of age-associated immune system alterations. The latter could contribute to an increased risk of autoimmune disease and infection in older adults. Our results underline the importance of better understanding the factors involved in the immunosenescence process to be able to uncover new biomarkers and open new avenues for the investigation and development of novel age-related disease therapies.
Collapse
Affiliation(s)
- Martina Formentini
- Maimonides Institute for Biomedical Research of Cordoba (IMIBIC), Córdoba, Spain
| | - Ana Navas
- Maimonides Institute for Biomedical Research of Cordoba (IMIBIC), Córdoba, Spain.,Immunology and Allergy service, Reina Sofia University Hospital, Córdoba, Spain
| | - Fakhri Hassouneh
- Maimonides Institute for Biomedical Research of Cordoba (IMIBIC), Córdoba, Spain
| | - Nelson Lopez-Sejas
- Maimonides Institute for Biomedical Research of Cordoba (IMIBIC), Córdoba, Spain
| | - Corona Alonso
- Maimonides Institute for Biomedical Research of Cordoba (IMIBIC), Córdoba, Spain.,Immunology and Allergy service, Reina Sofia University Hospital, Córdoba, Spain
| | - Raquel Tarazona
- Immunology Unit, Department of Physiology, University of Extremadura, Cáceres, Spain
| | - Rafael Solana
- Maimonides Institute for Biomedical Research of Cordoba (IMIBIC), Córdoba, Spain.,Immunology and Allergy service, Reina Sofia University Hospital, Córdoba, Spain.,Department of Cell Biology, Physiology and Immunology, University of Córdoba, Córdoba, Spain
| | - Alejandra Pera
- Maimonides Institute for Biomedical Research of Cordoba (IMIBIC), Córdoba, Spain.,Department of Cell Biology, Physiology and Immunology, University of Córdoba, Córdoba, Spain
| |
Collapse
|
30
|
Stojanović SD, Fiedler J, Bauersachs J, Thum T, Sedding DG. Senescence-induced inflammation: an important player and key therapeutic target in atherosclerosis. Eur Heart J 2021; 41:2983-2996. [PMID: 31898722 PMCID: PMC7453834 DOI: 10.1093/eurheartj/ehz919] [Citation(s) in RCA: 119] [Impact Index Per Article: 29.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/22/2019] [Revised: 09/13/2019] [Accepted: 12/12/2019] [Indexed: 12/21/2022] Open
Abstract
Inflammation is a hallmark and potent driver of pathological vascular remodelling in atherosclerosis. However, current anti-inflammatory therapeutic strategies have shown mixed results. As an alternative perspective on the conundrum of chronic inflammation emerging evidence points towards a small subset of senescent cells as a critical player and central node driving atherosclerosis. Senescent cells belonging to various cell types are a dominant and chronic source of a large array of pro-inflammatory cytokines and various additional plaque destabilizing factors, being involved with various aspects of atherosclerosis pathogenesis. Antagonizing these key agitators of local chronic inflammation and plaque instability may provide a causative and multi-purpose therapeutic strategy to treat atherosclerosis. Anti-senescence treatment options with translational potential are currently in development. However, several questions and challenges remain to be addressed before these novel treatment approaches may enter the clinical setting.
Collapse
Affiliation(s)
- Stevan D Stojanović
- Institute of Molecular and Translational Therapeutic Strategies (IMTTS), Hannover Medical School, Carl-Neuberg-Strasse 1, 30625 Hannover, Germany.,Department of Cardiology and Angiology, Hannover Medical School, Carl-Neuberg-Strasse 1, 30625 Hannover, Germany
| | - Jan Fiedler
- Institute of Molecular and Translational Therapeutic Strategies (IMTTS), Hannover Medical School, Carl-Neuberg-Strasse 1, 30625 Hannover, Germany
| | - Johann Bauersachs
- Department of Cardiology and Angiology, Hannover Medical School, Carl-Neuberg-Strasse 1, 30625 Hannover, Germany
| | - Thomas Thum
- Institute of Molecular and Translational Therapeutic Strategies (IMTTS), Hannover Medical School, Carl-Neuberg-Strasse 1, 30625 Hannover, Germany
| | - Daniel G Sedding
- Department of Internal Medicine III, Cardiology, Angiology and Intensive Care Medicine, Martin-Luther-University Halle (Saale), Ernst-Grube-Strasse 40, 06120 Halle (Saale), Germany
| |
Collapse
|
31
|
Boßlau TK, Wasserfurth P, Krüger B, Reichel T, Palmowski J, Nebl J, Weyh C, Schenk A, Joisten N, Stahl F, Thoms S, Gebhardt K, Hahn A, Krüger K. Abdominal Obesity-Related Disturbance of Insulin Sensitivity Is Associated with CD8 + EMRA Cells in the Elderly. Cells 2021; 10:cells10050998. [PMID: 33922813 PMCID: PMC8146929 DOI: 10.3390/cells10050998] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2021] [Revised: 04/21/2021] [Accepted: 04/22/2021] [Indexed: 12/24/2022] Open
Abstract
Aging and overweight increase the risk of developing type 2 diabetes mellitus. In this cross-sectional study, we aimed to investigate the potential mediating role of T-EMRA cells and inflammatory markers in the development of a decreased insulin sensitivity. A total of 134 healthy older volunteers were recruited (age 59.2 (SD 5.6) years). T cell subpopulations were analyzed by flow cytometry. Furthermore, body composition, HOMA-IR, plasma tryptophan (Trp) metabolites, as well as cytokines and adipokines were determined. Using subgroup and covariance analyses, the influence of BMI on the parameters was evaluated. Moreover, correlation, multiple regression, and mediation analyses were performed. In the subgroup of participants with obesity, an increased proportion of CD8+EMRA cells and elevated concentrations of plasma kynurenine (KYN) were found compared to the lower-weight subgroups. Linear regression analysis revealed that an elevated HOMA-IR could be predicted by a higher proportion of CD8+EMRA cells and KYN levels. A mediation analysis showed a robust indirect effect of the Waist-to-hip ratio on HOMA-IR mediated by CD8+EMRA cells. Thus, the deleterious effects of abdominal obesity on glucose metabolism might be mediated by CD8+EMRA cells in the elderly. Longitudinal studies should validate this assumption and analyze the suitability of CD8+EMRA cells as early predictors of incipient prediabetes.
Collapse
Affiliation(s)
- Tim K. Boßlau
- Department of Exercise Physiology and Sports Therapy, Institute of Sports Science, Justus-Liebig-University Giessen, Kugelberg 62, 35394 Giessen, Germany; (T.K.B.); (T.R.); (J.P.); (C.W.); (K.G.)
| | - Paulina Wasserfurth
- Institute of Food Science and Human Nutrition, Leibniz University Hannover, Am Kleinen Felde 30, 30159 Hannover, Germany; (P.W.); (J.N.); (A.H.)
| | - Britta Krüger
- Nemolab, Institute of Sports Science, Justus-Liebig-University Giessen, Kugelberg 62, 35394 Giessen, Germany;
| | - Thomas Reichel
- Department of Exercise Physiology and Sports Therapy, Institute of Sports Science, Justus-Liebig-University Giessen, Kugelberg 62, 35394 Giessen, Germany; (T.K.B.); (T.R.); (J.P.); (C.W.); (K.G.)
| | - Jana Palmowski
- Department of Exercise Physiology and Sports Therapy, Institute of Sports Science, Justus-Liebig-University Giessen, Kugelberg 62, 35394 Giessen, Germany; (T.K.B.); (T.R.); (J.P.); (C.W.); (K.G.)
| | - Josefine Nebl
- Institute of Food Science and Human Nutrition, Leibniz University Hannover, Am Kleinen Felde 30, 30159 Hannover, Germany; (P.W.); (J.N.); (A.H.)
| | - Christopher Weyh
- Department of Exercise Physiology and Sports Therapy, Institute of Sports Science, Justus-Liebig-University Giessen, Kugelberg 62, 35394 Giessen, Germany; (T.K.B.); (T.R.); (J.P.); (C.W.); (K.G.)
| | - Alexander Schenk
- Department of Performance and Health, Institute of Sports and Sport Science, Technical University Dortmund, Otto-Hahn-Straße 3, 44227 Dortmund, Germany; (A.S.); (N.J.)
| | - Niklas Joisten
- Department of Performance and Health, Institute of Sports and Sport Science, Technical University Dortmund, Otto-Hahn-Straße 3, 44227 Dortmund, Germany; (A.S.); (N.J.)
| | - Frank Stahl
- Institute of Technical Chemistry, Leibniz University Hannover, Callinstrasse 5, 30167 Hannover, Germany; (F.S.); (S.T.)
| | - Stefanie Thoms
- Institute of Technical Chemistry, Leibniz University Hannover, Callinstrasse 5, 30167 Hannover, Germany; (F.S.); (S.T.)
| | - Kristina Gebhardt
- Department of Exercise Physiology and Sports Therapy, Institute of Sports Science, Justus-Liebig-University Giessen, Kugelberg 62, 35394 Giessen, Germany; (T.K.B.); (T.R.); (J.P.); (C.W.); (K.G.)
| | - Andreas Hahn
- Institute of Food Science and Human Nutrition, Leibniz University Hannover, Am Kleinen Felde 30, 30159 Hannover, Germany; (P.W.); (J.N.); (A.H.)
| | - Karsten Krüger
- Department of Exercise Physiology and Sports Therapy, Institute of Sports Science, Justus-Liebig-University Giessen, Kugelberg 62, 35394 Giessen, Germany; (T.K.B.); (T.R.); (J.P.); (C.W.); (K.G.)
- Correspondence: ; Tel.: +49-641-992-5211
| |
Collapse
|
32
|
Chioh FW, Fong SW, Young BE, Wu KX, Siau A, Krishnan S, Chan YH, Carissimo G, Teo LL, Gao F, Tan RS, Zhong L, Koh AS, Tan SY, Tambyah PA, Renia L, Ng LF, Lye DC, Cheung C. Convalescent COVID-19 patients are susceptible to endothelial dysfunction due to persistent immune activation. eLife 2021; 10:64909. [PMID: 33752798 PMCID: PMC7987341 DOI: 10.7554/elife.64909] [Citation(s) in RCA: 101] [Impact Index Per Article: 25.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2020] [Accepted: 02/15/2021] [Indexed: 12/18/2022] Open
Abstract
Numerous reports of vascular events after an initial recovery from COVID-19 form our impetus to investigate the impact of COVID-19 on vascular health of recovered patients. We found elevated levels of circulating endothelial cells (CECs), a biomarker of vascular injury, in COVID-19 convalescents compared to healthy controls. In particular, those with pre-existing conditions (e.g., hypertension, diabetes) had more pronounced endothelial activation hallmarks than non-COVID-19 patients with matched cardiovascular risk. Several proinflammatory and activated T lymphocyte-associated cytokines sustained from acute infection to recovery phase, which correlated positively with CEC measures, implicating cytokine-driven endothelial dysfunction. Notably, we found higher frequency of effector T cells in our COVID-19 convalescents compared to healthy controls. The activation markers detected on CECs mapped to counter receptors found primarily on cytotoxic CD8+ T cells, raising the possibility of cytotoxic effector cells targeting activated endothelial cells. Clinical trials in preventive therapy for post-COVID-19 vascular complications may be needed.
Collapse
Affiliation(s)
- Florence Wj Chioh
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, Singapore
| | - Siew-Wai Fong
- A*STAR ID Labs, Agency for Science, Technology and Research, Singapore, Singapore.,Singapore Immunology Network, Agency for Science, Technology and Research, Singapore, Singapore.,Department of Biological Sciences, National University of Singapore, Singapore, Singapore
| | - Barnaby E Young
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, Singapore.,National Centre for Infectious Diseases, Singapore, Singapore.,Department of Infectious Diseases, Tan Tock Seng Hospital, Singapore, Singapore
| | - Kan-Xing Wu
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, Singapore
| | - Anthony Siau
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, Singapore
| | - Shuba Krishnan
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, Singapore.,Division of Clinical Microbiology, Department of Laboratory Medicine, Karolinska Institute, ANA Futura, Campus Flemingsberg, Stockholm, Sweden
| | - Yi-Hao Chan
- A*STAR ID Labs, Agency for Science, Technology and Research, Singapore, Singapore.,Singapore Immunology Network, Agency for Science, Technology and Research, Singapore, Singapore
| | - Guillaume Carissimo
- A*STAR ID Labs, Agency for Science, Technology and Research, Singapore, Singapore.,Singapore Immunology Network, Agency for Science, Technology and Research, Singapore, Singapore
| | - Louis Ly Teo
- National Heart Centre Singapore, Singapore, Singapore.,Duke-NUS Medical School, Singapore, Singapore
| | - Fei Gao
- National Heart Centre Singapore, Singapore, Singapore.,Duke-NUS Medical School, Singapore, Singapore
| | - Ru San Tan
- National Heart Centre Singapore, Singapore, Singapore.,Duke-NUS Medical School, Singapore, Singapore
| | - Liang Zhong
- National Heart Centre Singapore, Singapore, Singapore.,Duke-NUS Medical School, Singapore, Singapore
| | - Angela S Koh
- National Heart Centre Singapore, Singapore, Singapore.,Duke-NUS Medical School, Singapore, Singapore
| | - Seow-Yen Tan
- Department of Infectious Diseases, Changi General Hospital, Singapore, Singapore
| | - Paul A Tambyah
- Department of Medicine, National University Hospital, Singapore, Singapore
| | - Laurent Renia
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, Singapore.,A*STAR ID Labs, Agency for Science, Technology and Research, Singapore, Singapore.,Singapore Immunology Network, Agency for Science, Technology and Research, Singapore, Singapore
| | - Lisa Fp Ng
- A*STAR ID Labs, Agency for Science, Technology and Research, Singapore, Singapore.,Singapore Immunology Network, Agency for Science, Technology and Research, Singapore, Singapore
| | - David C Lye
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, Singapore.,National Centre for Infectious Diseases, Singapore, Singapore.,Department of Infectious Diseases, Tan Tock Seng Hospital, Singapore, Singapore.,Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Christine Cheung
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, Singapore.,Institute of Molecular and Cell Biology, Agency for Science, Technology and Research, Singapore, Singapore
| |
Collapse
|
33
|
Marchini T, Hansen S, Wolf D. ApoB-Specific CD4 + T Cells in Mouse and Human Atherosclerosis. Cells 2021; 10:446. [PMID: 33669769 PMCID: PMC7922692 DOI: 10.3390/cells10020446] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2021] [Revised: 02/15/2021] [Accepted: 02/17/2021] [Indexed: 12/11/2022] Open
Abstract
Atherosclerosis is a chronic inflammatory condition of the arterial wall that leads to the formation of vessel-occluding plaques within the subintimal space of middle-sized and larger arteries. While traditionally understood as a myeloid-driven lipid-storage disease, growing evidence suggests that the accumulation of low-density lipoprotein cholesterol (LDL-C) ignites an autoimmune response with CD4+ T-helper (TH) cells that recognize self-peptides from Apolipoprotein B (ApoB), the core protein of LDL-C. These autoreactive CD4+ T cells home to the atherosclerotic plaque, clonally expand, instruct other cells in the plaque, and induce clinical plaque instability. Recent developments in detecting antigen-specific cells at the single cell level have demonstrated that ApoB-reactive CD4+ T cells exist in humans and mice. Their phenotypes and functions deviate from classical immunological concepts of distinct and terminally differentiated TH immunity. Instead, ApoB-specific CD4+ T cells have a highly plastic phenotype, can acquire several, partially opposing and mixed transcriptional programs simultaneously, and transit from one TH subset into another over time. In this review, we highlight adaptive immune mechanisms in atherosclerosis with a focus on CD4+ T cells, introduce novel technologies to detect ApoB-specific CD4+ T cells at the single cell level, and discuss the potential impact of ApoB-driven autoimmunity in atherosclerosis.
Collapse
Affiliation(s)
- Timoteo Marchini
- Department of Cardiology and Angiology I, University Heart Center Freiburg, Hugstetterstraße 55, 79106 Freiburg, Germany; (T.M.); (S.H.)
- Faculty of Medicine, University of Freiburg, Breisacherstraße 153, 79110 Freiburg, Germany
- Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires, CONICET, Instituto de Bioquímica y Medicina Molecular (IBIMOL), Junín 954, C1113 AAD Buenos Aires, Argentina
| | - Sophie Hansen
- Department of Cardiology and Angiology I, University Heart Center Freiburg, Hugstetterstraße 55, 79106 Freiburg, Germany; (T.M.); (S.H.)
- Faculty of Medicine, University of Freiburg, Breisacherstraße 153, 79110 Freiburg, Germany
| | - Dennis Wolf
- Department of Cardiology and Angiology I, University Heart Center Freiburg, Hugstetterstraße 55, 79106 Freiburg, Germany; (T.M.); (S.H.)
- Faculty of Medicine, University of Freiburg, Breisacherstraße 153, 79110 Freiburg, Germany
| |
Collapse
|
34
|
Zhao L, Wang Q, Zhou B, Zhang L, Zhu H. The Role of Immune Cells in the Pathogenesis of Idiopathic Inflammatory Myopathies. Aging Dis 2021; 12:247-260. [PMID: 33532139 PMCID: PMC7801271 DOI: 10.14336/ad.2020.0410] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2020] [Accepted: 04/10/2020] [Indexed: 12/15/2022] Open
Abstract
Idiopathic inflammatory myopathies (IIMs) are chronic autoimmune disorders involving multiple organs, such as the muscle, skin, lungs and joints. Although the detailed pathogenesis of IIMs remains unclear, immune mechanisms have long been recognised as of key importance. Immune cells contribute to many inflammatory processes via intercellular interactions and secretion of inflammatory factors, and many studies have demonstrated the participation of a variety of immune cells, such as T cells and B cells, in the development of IIMs. Here, we summarise the current knowledge regarding immune cells in IIM patients and discuss their potential roles in IIM pathogenesis.
Collapse
Affiliation(s)
- Lijuan Zhao
- Department of Rheumatology, Xiangya Hospital, Central South University, Changsha, Hunan, China.
| | - Qi Wang
- Department of Radiology, Hunan Provincial People's Hospital and The First Affiliated Hospital of Hunan Normal University, Changsha, Hunan, China.
| | - Bin Zhou
- Department of Nephrology, The Affiliated Hospital of Qingdao University, Qingdao, China.
| | - Lihua Zhang
- Department of Rheumatology, Hunan Provincial People's Hospital and The First Affiliated Hospital of Hunan Normal University, Changsha, Hunan, China.
| | - Honglin Zhu
- Department of Rheumatology, Xiangya Hospital, Central South University, Changsha, Hunan, China.
| |
Collapse
|
35
|
Heinbokel T, Quante M, Iske J, Nian Y, Maenosono R, Minami K, Liu Y, Azuma H, Elkhal A, Tullius SG. CTLA4-Ig prolongs graft survival specifically in young but not old mice. Am J Transplant 2021; 21:488-502. [PMID: 32717114 PMCID: PMC7855762 DOI: 10.1111/ajt.16218] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2020] [Revised: 06/22/2020] [Accepted: 07/09/2020] [Indexed: 01/25/2023]
Abstract
Elderly organ transplant recipients have remained underrepresented in clinical trials, despite representing a rapidly growing population. Here, we assessed age-specific effects of CTLA4-Ig (cytotoxic T-lymphocyte antigen 4-Ig), a fusion protein blocking costimulatory signaling between antigen-presenting cells and T cells through CD28. Cardiac allografts in young mice (2-3 months) treated with CTLA4-Ig survived indefinitely, whereas 80% of old recipients (18 months) had lost their graft after 100 days. CTLA4-Ig was also significantly less effective in older recipients of skin transplants. CTLA4-Ig reduced CD4+ central memory and effector memory T cells and diminished systemic interferon-gamma levels only in young recipients. These differences corresponded to a reduced expression of CD28 on antigen-experienced CD4+ T cells in old mice. In support, adoptive transfer of old CD4+ T cells that were transfected with a lentiviral vector inducing constant expression of CD28 accelerated the rejection of allogeneic skin grafts in young RAG2-/- recipient mice. Regulatory T cells (Tregs), in contrast, demonstrated an increased expression of CD28 with aging and CTLA4-Ig treatment in old recipients resulted in reduced frequencies, compromised proliferation, and diminished suppressive capacity of Tregs. These findings may prove to have unique clinical consequences for immunosuppression in the growing population of elderly transplant recipients.
Collapse
Affiliation(s)
- Timm Heinbokel
- Transplant Surgery Research Laboratory and Division of Transplant Surgery, Brigham and Women’s Hospital, Harvard Medical School, Boston, USA,Department of Nephrology, Charité – Universitätsmedizin Berlin, Berlin, Germany
| | - Markus Quante
- Transplant Surgery Research Laboratory and Division of Transplant Surgery, Brigham and Women’s Hospital, Harvard Medical School, Boston, USA,Department of General, Visceral and Transplant Surgery, Tuebingen University Hospital, Tuebingen, Germany
| | - Jasper Iske
- Transplant Surgery Research Laboratory and Division of Transplant Surgery, Brigham and Women’s Hospital, Harvard Medical School, Boston, USA,Institute of Transplant Immunology, Hannover Medical School, Hannover, Germany
| | - Yeqi Nian
- Transplant Surgery Research Laboratory and Division of Transplant Surgery, Brigham and Women’s Hospital, Harvard Medical School, Boston, USA
| | - Ryoichi Maenosono
- Transplant Surgery Research Laboratory and Division of Transplant Surgery, Brigham and Women’s Hospital, Harvard Medical School, Boston, USA,Department of Urology, Osaka Medical College, Osaka, Japan
| | - Koichiro Minami
- Transplant Surgery Research Laboratory and Division of Transplant Surgery, Brigham and Women’s Hospital, Harvard Medical School, Boston, USA,Department of Urology, Osaka Medical College, Osaka, Japan
| | - Yang Liu
- Transplant Surgery Research Laboratory and Division of Transplant Surgery, Brigham and Women’s Hospital, Harvard Medical School, Boston, USA
| | - Haruhito Azuma
- Department of Urology, Osaka Medical College, Osaka, Japan
| | - Abdallah Elkhal
- Transplant Surgery Research Laboratory and Division of Transplant Surgery, Brigham and Women’s Hospital, Harvard Medical School, Boston, USA
| | - Stefan G. Tullius
- Transplant Surgery Research Laboratory and Division of Transplant Surgery, Brigham and Women’s Hospital, Harvard Medical School, Boston, USA
| |
Collapse
|
36
|
Cao M, Ruan L, Huang Y, Wang J, Yan J, Sang Y, Li S, Wang G, Wu X. Premature CD4 + T Cells Senescence Induced by Chronic Infection in Patients with Acute Coronary Syndrome. Aging Dis 2020; 11:1471-1480. [PMID: 33269101 PMCID: PMC7673853 DOI: 10.14336/ad.2020.0203] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2019] [Accepted: 02/03/2020] [Indexed: 01/10/2023] Open
Abstract
Acquired immune responses mediated by CD4+ T cells contribute to the initiation and progression of acute coronary syndrome (ACS). ACS patients show acquired immune system abnormalities that resemble the characteristics of autoimmune dysfunction described in the elderly. This study aimed to investigate the role of premature CD4+ T cells senescence in ACS and the underlying mechanism. We compared the immunological status of 25 ACS patients, 15 young healthy individuals (C1), and 20 elderly individuals with absence of ACS (C2). The percentages of CD4+ T lymphocyte subsets (including naïve, regulatory, memory and effector T cells) in peripheral blood were analyzed. In ACS patients, a significant expansion of CD4+CD28null effector T cells and a decline of CD4+CD25+CD62L+Treg cells were observed. In addition, patients with ACS showed an accelerated loss of CD4+CD45RA+CD62L+ naïve T cells and a compensatory increase in the number of CD4+CD45RO+ memory T cells. ACS patients demonstrated no significant difference in frequency of T cell receptor excision circles (TRECs) compared to age-matched healthy volunteers. The expression of p16Ink4a was increased while CD62L was decreased in CD4+CD28null T cells of ACS patients. Compared to healthy donors, ACS patients demonstrated the lowest telomerase activity in both CD4+CD28+and CD4+CD28null T cells. The serum levels of C-reactive protein, Cytomegalovirus IgG, Helicobactor pylori IgG and Chlamydia pneumonia IgG were significantly higher in ACS patients. The results suggested that the percentage of CD4+ T cell subpopulations correlated with chronic infection, which contributes to immunosenescence. In conclusion, chronic infection induced senescence of premature CD4+T cells, which may be responsible for the development of ACS.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Xiaofen Wu
- Department of Gerontology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| |
Collapse
|
37
|
Paccosi S, Pala L, Cresci B, Silvano A, Cecchi M, Caporale R, Maria Rotella C, Parenti A. Insulin resistance and obesity affect monocyte-derived dendritic cell phenotype and function. Diabetes Res Clin Pract 2020; 170:108528. [PMID: 33157116 DOI: 10.1016/j.diabres.2020.108528] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/06/2020] [Revised: 09/10/2020] [Accepted: 10/26/2020] [Indexed: 12/12/2022]
Abstract
AIM Cardiovascular disease (CVD) is prevalent in women after menopause, which may be associated with obesity, insulin resistance and metaflammation. Despite the recognized role of immunological mechanisms in vascular remodeling, the role of dendritic cells (DCs) is still unclear. The aim was to characterize monocyte-derived DCs (Mo-DC) in post-menopausal patients with type 2 diabetes (T2DM) and obese woman, without clinical manifestations of atherosclerosis. METHODS Obese post-menopausal women with or without T2DM were enrolled and were compared to age-matched healthy women. DCs obtained from patients were phenotypically and functionally characterized by flow cytometry and mixed lymphocyte reaction. MRNA integrins expression was assessed by real time RT-PCR; circulating fetuin-A and adiponectin levels were measured by ELISA. RESULTS Phenotypic dysregulation of Mo-DC reported was related to a defective allogenic lymphocyte stimulation and to an increased mRNA of CD11c, CD18 and DC-SIGN/CD209 which regulate their adhesion to vascular wall cells. Fetuin-A and adiponectin levels were significantly altered and negatively correlated. Hyperglycaemia significantly impaired CD14+ transdifferentiation into Mo-DC. CONCLUSIONS These data show a dysfunction of Mo-DCs obtained from precursors isolated from T2DM obese post-menopausal woman without any documented clinical CV event. Association of obesity to diabetes seems to worsen DC's phenotype and function and increase vascular inflammation.
Collapse
Affiliation(s)
- Sara Paccosi
- Department of Health Sciences, Clinical Pharmacology and Oncology Section, University of Florence, Florence, Italy
| | - Laura Pala
- Diabetology, Careggi University Hospital, Florence, Italy
| | - Barbara Cresci
- Diabetology, Careggi University Hospital, Florence, Italy
| | - Angela Silvano
- Department of Health Sciences, Clinical Pharmacology and Oncology Section, University of Florence, Florence, Italy
| | - Marta Cecchi
- Department of Health Sciences, Clinical Pharmacology and Oncology Section, University of Florence, Florence, Italy
| | - Roberto Caporale
- Cytofluorimetry and Immunotherapy Diagnostic Center, Careggi University Hospital, Florence, Italy
| | - Carlo Maria Rotella
- Department of Biomedical Clinical and Experimental Sciences, Endocrine Unit, University of Florence, Florence, Italy
| | - Astrid Parenti
- Department of Health Sciences, Clinical Pharmacology and Oncology Section, University of Florence, Florence, Italy.
| |
Collapse
|
38
|
Jin Y, Dong Y, Zhang J, Sun J, Liu Y, Chen Y. The toxicity of cell therapy: Mechanism, manifestations, and challenges. J Appl Toxicol 2020; 41:659-667. [DOI: 10.1002/jat.4100] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2020] [Revised: 10/09/2020] [Accepted: 10/13/2020] [Indexed: 12/20/2022]
Affiliation(s)
- Yongjia Jin
- Shanghai Electric Power Hospital Shanghai China
| | - Yan Dong
- Department of Musculoskeletal Oncology Fudan University Shanghai Cancer Center Shanghai China
- Department of Oncology Shanghai Medical College, Fudan University Shanghai China
| | | | | | | | - Yong Chen
- Department of Musculoskeletal Oncology Fudan University Shanghai Cancer Center Shanghai China
- Department of Oncology Shanghai Medical College, Fudan University Shanghai China
| |
Collapse
|
39
|
Machado-Oliveira G, Ramos C, Marques ARA, Vieira OV. Cell Senescence, Multiple Organelle Dysfunction and Atherosclerosis. Cells 2020; 9:E2146. [PMID: 32977446 PMCID: PMC7598292 DOI: 10.3390/cells9102146] [Citation(s) in RCA: 45] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2020] [Revised: 09/19/2020] [Accepted: 09/20/2020] [Indexed: 01/10/2023] Open
Abstract
Atherosclerosis is an age-related disorder associated with long-term exposure to cardiovascular risk factors. The asymptomatic progression of atherosclerotic plaques leads to major cardiovascular diseases (CVD), including acute myocardial infarctions or cerebral ischemic strokes in some cases. Senescence, a biological process associated with progressive structural and functional deterioration of cells, tissues and organs, is intricately linked to age-related diseases. Cell senescence involves coordinated modifications in cellular compartments and has been demonstrated to contribute to different stages of atheroma development. Senescence-based therapeutic strategies are currently being pursued to treat and prevent CVD in humans in the near-future. In addition, distinct experimental settings allowed researchers to unravel potential approaches to regulate anti-apoptotic pathways, facilitate excessive senescent cell clearance and eventually reverse atherogenesis to improve cardiovascular function. However, a deeper knowledge is required to fully understand cellular senescence, to clarify senescence and atherogenesis intertwining, allowing researchers to establish more effective treatments and to reduce the cardiovascular disorders' burden. Here, we present an objective review of the key senescence-related alterations of the major intracellular organelles and analyze the role of relevant cell types for senescence and atherogenesis. In this context, we provide an updated analysis of therapeutic approaches, including clinically relevant experiments using senolytic drugs to counteract atherosclerosis.
Collapse
Affiliation(s)
- Gisela Machado-Oliveira
- CEDOC, NOVA Medical School, Faculdade de Ciências Médicas, Universidade NOVA de Lisboa, 1169-056 Lisboa, Portugal; (C.R.); (A.R.A.M.)
| | | | | | - Otília V. Vieira
- CEDOC, NOVA Medical School, Faculdade de Ciências Médicas, Universidade NOVA de Lisboa, 1169-056 Lisboa, Portugal; (C.R.); (A.R.A.M.)
| |
Collapse
|
40
|
Kosmaczewska A, Ciszak L, Stosio M, Szteblich A, Madej M, Frydecka I, Wiland P, Szmyrka M. CD4 +CD28 null T cells are expanded in moderately active systemic lupus erythematosus and secrete pro-inflammatory interferon gamma, depending on the Disease Activity Index. Lupus 2020; 29:705-714. [PMID: 32279585 DOI: 10.1177/0961203320917749] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
BACKGROUND Pathogenic CD4+CD28null cells are characterized by inflammatory cytokine synthesis and tropism to the inflamed tissues. Recent studies showed the involvement of CD28null T cells in a severe clinical outcome of lupus. However, their role in moderately active disease is still unresolved. METHODS We examined the levels of circulating CD4+CD28null cells and CD8+CD28null suppressor T cells. We also compared the CD4+CD28null and CD4+CD28+ T-cell functional properties, including the expression of interferon gamma (IFN-γ) and Ki67 among systemic lupus erythematosus (SLE) patients (n = 20) and healthy controls (n = 20). All the patients were under immunosuppressive treatment and exhibited moderate SLE activity (median SLE Disease Activity Index (SLEDAI) = 6). RESULTS In patients, we found elevated CD4+CD28null and unchanged levels of suppressor CD8+CD28null T cells. There was no difference between patients and controls in IFN-γ and Ki67-expressing CD4+, CD4+CD28+, and CD4+CD28null T cells, except for higher IFN-γ levels in CD4+CD28+ T cells in SLE. In each studied group, we observed a higher preponderance of IFN-γ- and Ki67-expressing cells among CD4+CD28null T cells and lower levels of IFN-γ in CD4+CD28null T cells compared to the CD28+ subset. Similarly, Ki67 intensity was decreased in healthy CD4+CD28null cells, whereas in patients, comparably high expression was observed in both subsets. IFN-γ intensity in CD4+CD28null T cells correlated with SLEDAI. CONCLUSION SLE with a moderately active clinical course is characterized by peripheral blood expansion of CD4+CD28null T cells and a normal abundance of suppressor CD8+CD28null T cells. The demonstration that these pathogenic CD4+ T cells, despite the lack of CD28, maintain the ability to produce pro-inflammatory IFN-γ positively correlated with disease activity as well as relatively high proliferative capacity may suggest their potentially predictive role in SLE flares.
Collapse
Affiliation(s)
- Agata Kosmaczewska
- Department of Experimental Therapy, Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, Wroclaw, Poland
| | - Lidia Ciszak
- Department of Experimental Therapy, Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, Wroclaw, Poland
| | - Malgorzata Stosio
- Faculty of Chemistry, Wroclaw University of Science and Technology, Wroclaw, Poland
| | - Aleksandra Szteblich
- Department of Experimental Therapy, Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, Wroclaw, Poland
| | - Marta Madej
- Department of Rheumatology and Internal Diseases, Wroclaw Medical University, Wroclaw, Poland
| | - Irena Frydecka
- Department of Experimental Therapy, Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, Wroclaw, Poland
| | - Piotr Wiland
- Department of Rheumatology and Internal Diseases, Wroclaw Medical University, Wroclaw, Poland
| | - Magdalena Szmyrka
- Department of Rheumatology and Internal Diseases, Wroclaw Medical University, Wroclaw, Poland
| |
Collapse
|
41
|
Orlik C, Deibel D, Küblbeck J, Balta E, Ganskih S, Habicht J, Niesler B, Schröder-Braunstein J, Schäkel K, Wabnitz G, Samstag Y. Keratinocytes costimulate naive human T cells via CD2: a potential target to prevent the development of proinflammatory Th1 cells in the skin. Cell Mol Immunol 2020; 17:380-394. [PMID: 31324882 PMCID: PMC7109061 DOI: 10.1038/s41423-019-0261-x] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2019] [Accepted: 06/27/2019] [Indexed: 12/01/2022] Open
Abstract
The interplay between keratinocytes and immune cells, especially T cells, plays an important role in the pathogenesis of chronic inflammatory skin diseases. During psoriasis, keratinocytes attract T cells by releasing chemokines, while skin-infiltrating self-reactive T cells secrete proinflammatory cytokines, e.g., IFNγ and IL-17A, that cause epidermal hyperplasia. Similarly, in chronic graft-versus-host disease, allogenic IFNγ-producing Th1/Tc1 and IL-17-producing Th17/Tc17 cells are recruited by keratinocyte-derived chemokines and accumulate in the skin. However, whether keratinocytes act as nonprofessional antigen-presenting cells to directly activate naive human T cells in the epidermis remains unknown. Here, we demonstrate that under proinflammatory conditions, primary human keratinocytes indeed activate naive human T cells. This activation required cell contact and costimulatory signaling via CD58/CD2 and CD54/LFA-1. Naive T cells costimulated by keratinocytes selectively differentiated into Th1 and Th17 cells. In particular, keratinocyte-initiated Th1 differentiation was dependent on costimulation through CD58/CD2. The latter molecule initiated STAT1 signaling and IFNγ production in T cells. Costimulation of T cells by keratinocytes resulting in Th1 and Th17 differentiation represents a new explanation for the local enrichment of Th1 and Th17 cells in the skin of patients with a chronic inflammatory skin disease. Consequently, local interference with T cell-keratinocyte interactions may represent a novel strategy for the treatment of Th1 and Th17 cell-driven skin diseases.
Collapse
Affiliation(s)
- Christian Orlik
- Institute of Immunology, Section Molecular Immunology, Heidelberg University, Im Neuenheimer Feld 305, 69120, Heidelberg, Germany
| | - Daniel Deibel
- Institute of Immunology, Section Molecular Immunology, Heidelberg University, Im Neuenheimer Feld 305, 69120, Heidelberg, Germany
| | - Johanna Küblbeck
- Institute of Immunology, Section Molecular Immunology, Heidelberg University, Im Neuenheimer Feld 305, 69120, Heidelberg, Germany
| | - Emre Balta
- Institute of Immunology, Section Molecular Immunology, Heidelberg University, Im Neuenheimer Feld 305, 69120, Heidelberg, Germany
| | - Sabina Ganskih
- Institute of Immunology, Section Molecular Immunology, Heidelberg University, Im Neuenheimer Feld 305, 69120, Heidelberg, Germany
| | - Jüri Habicht
- Institute of Immunology, Section Molecular Immunology, Heidelberg University, Im Neuenheimer Feld 305, 69120, Heidelberg, Germany
| | - Beate Niesler
- Institute of Human Genetics, Department of Human Molecular Genetics, and nCounter Core Facility, Heidelberg University, Im Neuenheimer Feld 366, 69120, Heidelberg, Germany
| | - Jutta Schröder-Braunstein
- Institute of Immunology, Section Molecular Immunology, Heidelberg University, Im Neuenheimer Feld 305, 69120, Heidelberg, Germany
| | - Knut Schäkel
- Department of Dermatology, Heidelberg University, Im Neuenheimer Feld 440, 69120, Heidelberg, Germany
| | - Guido Wabnitz
- Institute of Immunology, Section Molecular Immunology, Heidelberg University, Im Neuenheimer Feld 305, 69120, Heidelberg, Germany
| | - Yvonne Samstag
- Institute of Immunology, Section Molecular Immunology, Heidelberg University, Im Neuenheimer Feld 305, 69120, Heidelberg, Germany.
| |
Collapse
|
42
|
DeMizio DJ, Geraldino-Pardilla LB. Autoimmunity and Inflammation Link to Cardiovascular Disease Risk in Rheumatoid Arthritis. Rheumatol Ther 2020; 7:19-33. [PMID: 31853784 PMCID: PMC7021876 DOI: 10.1007/s40744-019-00189-0] [Citation(s) in RCA: 59] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2019] [Indexed: 12/19/2022] Open
Abstract
Rheumatoid arthritis (RA) patients have a 50% increased risk of cardiovascular (CV)-related morbidity and mortality. This excess CV risk is closely linked to RA disease severity and chronic inflammation, hence is largely underestimated by traditional risk calculators such as the Framingham Risk Score. Epidemiological studies have shown that patients with RA are more likely to have silent ischemic heart disease, develop heart failure, and experience sudden death compared with controls. Elevations in pro-inflammatory cytokines, circulating autoantibodies, and specific T cell subsets, are believed to drive these findings by promoting atherosclerotic plaque formation and cardiac remodeling. Current European League Against Rheumatism (EULAR) guidelines state that rheumatologists are responsible for the assessment and coordination of CV disease (CVD) risk management in patients with RA, yet the optimal means to do so remain unclear. While these guidelines focus on disease activity control to mitigate excess CV risk, rather than providing a precise algorithm for choice of therapy, studies suggest a differential impact on CV risk of non-biologic disease-modifying anti-rheumatic drugs (DMARDs), biologic DMARDs, and small molecule-based therapy. In this review, we explore the mechanisms linking the pathophysiologic intrinsic features of RA with the increased CVD risk in this population, and the impact of different RA therapies on CV outcomes.
Collapse
Affiliation(s)
- Daniel J DeMizio
- Division of Rheumatology, Department of Medicine, Vagelos College of Physicians and Surgeons, Columbia University Irving Medical Center, New York, NY, USA
| | - Laura B Geraldino-Pardilla
- Division of Rheumatology, Department of Medicine, Vagelos College of Physicians and Surgeons, Columbia University Irving Medical Center, New York, NY, USA.
| |
Collapse
|
43
|
Zahran AM, Abdallah AM, Saad K, Osman NS, Youssef MAM, Abdel-Raheem YF, Elsayh KI, Abo Elgheet AM, Darwish SF, Alblihed MA, Elhoufey A. Peripheral Blood B and T Cell Profiles in Children with Active Juvenile Idiopathic Arthritis. Arch Immunol Ther Exp (Warsz) 2019; 67:427-432. [PMID: 31535168 DOI: 10.1007/s00005-019-00560-7] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2019] [Accepted: 09/09/2019] [Indexed: 02/07/2023]
Abstract
Juvenile idiopathic arthritis (JIA) is one of the most common autoimmune diseases in children. Our study aimed to evaluate the peripheral blood B and T lymphocyte subpopulations in children with JIA. This case-control study included 20 children with JIA as well as 20 healthy children with matching age and sex as a control group. All patients included in the study were in activity as determined by visual analog scale. In addition to complete clinical evaluation, basic investigations, peripheral blood B and T lymphocyte subpopulations were done to all participants by flow cytometry. JIA patients displayed a significant decrease in IgM memory B lymphocytes, switched memory B lymphocytes, and total memory B lymphocytes when compared to the healthy controls. The percentages of naïve B lymphocytes were significantly increased in JIA patients than in controls. Total T lymphocytes, CD8+CD28null cells, and CD4+CD28null cells were significantly increased in JIA patients as compared to controls. In conclusion; JIA patients have an alteration in both B and T lymphocytes with the predisposition of memory cells which may have a role in sustaining the JIA disease activity.
Collapse
Affiliation(s)
- Asmaa M Zahran
- Department of Clinical Pathology, South Egypt Cancer Institute, Assiut, Egypt
| | - Alameldin M Abdallah
- Department of Pediatrics, Faculty of Medicine, Assiut University, Assiut, 71516, Egypt
| | - Khaled Saad
- Department of Pediatrics, Faculty of Medicine, Assiut University, Assiut, 71516, Egypt.
| | - Naglaa S Osman
- Department of Pediatrics, Faculty of Medicine, Assiut University, Assiut, 71516, Egypt
| | - Mervat A M Youssef
- Department of Pediatrics, Faculty of Medicine, Assiut University, Assiut, 71516, Egypt
| | | | - Khalid I Elsayh
- Department of Pediatrics, Faculty of Medicine, Assiut University, Assiut, 71516, Egypt
| | - Amir M Abo Elgheet
- Department of Pediatrics, Faculty of Medicine, Assiut University, Assiut, 71516, Egypt
| | - Sanaa F Darwish
- Department of Microbiology, Faculty of Pharmacy, Cairo University, Cairo, Egypt
| | - Mohamd A Alblihed
- Department of Medical Biochemistry, School of Medicine Taif University, Taif, Kingdom of Saudi Arabia
| | - Amira Elhoufey
- Department of Community Health Nursing, Faculty of Nursing, Assiut University, Assiut, Egypt
- Department of Community Health Nursing, Sabia University College, Jazan University, Jazan, Kingdom of Saudi Arabia
| |
Collapse
|
44
|
Shahbaz SK, Sadeghi M, Koushki K, Penson PE, Sahebkar A. Regulatory T cells: Possible mediators for the anti-inflammatory action of statins. Pharmacol Res 2019; 149:104469. [PMID: 31577918 DOI: 10.1016/j.phrs.2019.104469] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/24/2019] [Revised: 09/26/2019] [Accepted: 09/27/2019] [Indexed: 02/07/2023]
Abstract
Statins beside their main effect on reducing the progression of cardiovascular disease through pharmacological inhibition of the endogenous cholesterol synthesis, have additional pleiotropic effects including antiinflammatory effects mediated through the induction of suppressor regulatory T cells (Tregs). Statin-induced expansion of Tregs reduces chronic inflammation and may have beneficial effects in autoimmune diseases. However, statins could represent a double-edged sword in immunomodulation. Drugs that act by increasing the concentration of Tregs could enhance the risk of cancers, particularly in the elderly and may have adverse effects in neurodegenerative disorders and infectious diseases. In the present paper, we review the experimental studies that evaluate the effects of statins on Treg cells in autoimmune and inflammatory diseases and we discuss potential therapeutic applications of statins in this setting.
Collapse
Affiliation(s)
- Sanaz Keshavarz Shahbaz
- Department of Immunology, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mahvash Sadeghi
- Department of Immunology, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Khadije Koushki
- Department of Immunology, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Peter E Penson
- School of Pharmacy and Biomolecular Sciences, Liverpool John Moores University, Liverpool, UK
| | - Amirhossein Sahebkar
- Halal Research Center of IRI, FDA, Tehran, Iran; Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran; Neurogenic Inflammation Research Center, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
45
|
Abstract
Immunosenescence involves a series of ageing-induced alterations in the immune system and is characterized by two opposing hallmarks: defective immune responses and increased systemic inflammation. The immune system is modulated by intrinsic and extrinsic factors and undergoes profound changes in response to the ageing process. Immune responses are therefore highly age-dependent. Emerging data show that immunosenescence underlies common mechanisms responsible for several age-related diseases and is a plastic state that can be modified and accelerated by non-heritable environmental factors and pharmacological intervention. In the kidney, resident macrophages and fibroblasts are continuously exposed to components of the external environment, and the effects of cellular reprogramming induced by local immune responses, which accumulate with age, might have a role in the increased susceptibility to kidney disease among elderly individuals. Additionally, because chronic kidney disease, especially end-stage renal disease, is often accompanied by immunosenescence, which affects these patients independently of age, and many kidney diseases are strongly age-associated, treatment approaches that target immunosenescence might be particularly clinically relevant.
Collapse
|
46
|
Lack of association of tumor necrosis factor superfamily member 4 (TNFSF4) gene polymorphisms (rs3850641 and rs17568) with coronary heart disease and stroke: A systematic review and meta-analysis. Anatol J Cardiol 2019; 19:86-93. [PMID: 29424751 PMCID: PMC5864823 DOI: 10.14744/anatoljcardiol.2017.8069] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
Abstract
Objective: To evaluate the association between the tumor necrosis factor superfamily member 4 (TNFSF4) gene polymorphisms and common cardiovascular and cerebrovascular diseases. Methods: A literature-based search was conducted through databases including PubMed, EMBASE, Cochrane Library, CNKI, and WanFang data. Crude odds ratios (ORs) and 95% confidence intervals (CI) were calculated to estimate the strength of the association between TNFSF4 polymorphisms (rs3850641 and rs17568) and the risk of coronary heart disease (CHD) and stroke. Results: Overall, 11 eligible studies were included in this meta-analysis. G allele was showed not to be associated with CHD and stroke, compared with A allele (rs3850641: OR=1.02, 95% CI=0.89–1.17; rs17568: OR=1.09, 95% CI=0.89–1.33). Genotypic analysis demonstrated that there was no significant association between the risk of CHD and stroke and rs3850641 [homozygous comparison (GG vs. AA): OR=1.05, 95% CI=0.74–1.50; heterozygous comparison (GA vs. AA): OR=1.00, 95% CI=0.88–1.13; recessive model (GG vs. GA+AA): OR=1.04, 95% CI=0.76–1.43; dominant model (GG+GA vs. AA): OR=1.01, 95% CI=0.88–1.17]. Similarly, no susceptibility between CHD and stroke and rs17568 polymorphism was uncovered (GG vs. AA: OR=1.04, 95% CI=0.74–1.46; GA vs. AA: OR=1.07, 95% CI=0.62–1.83; GG+GA vs. AA: OR=1.13, 95% CI=0.82–1.56; GG vs. GA+AA: OR=1.01, 95% CI=0.74–1.39). Conclusion: The present study demonstrated that there is no significant relationship between TNFSF4 gene polymorphism and cerebrovascular and cardiovascular diseases.
Collapse
|
47
|
Okba AM, Abd El Raouf Raafat M, Nazmy Farres M, Abd El Nour Melek N, Amin MM, Gendy NN. Expanded peripheral CD4 +CD28 null T cells and its association with atherosclerotic changes in patients with end stage renal disease on hemodialysis. Hum Immunol 2019; 80:748-754. [PMID: 30853362 DOI: 10.1016/j.humimm.2019.03.008] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2018] [Revised: 02/25/2019] [Accepted: 03/06/2019] [Indexed: 10/27/2022]
Abstract
End-stage renal disease (ESRD) patients, including those on hemodialysis, possess a high risk for cardiovascular diseases, as the first leading cause of death among them. Traditional risk factors do not utterly elucidate this. Throughout the last two decades, CD4+CD28null T cells; an unusual subset of T lymphocytes, was detected high with excess cardiovascular (CV) mortality. We aimed to investigate the circulating CD4+CD28null T cells frequency in ESRD patients on hemodialysis and to evaluate their relationship with atherosclerotic changes. High-resolution carotid ultrasonography was done to assess the common carotid artery intima media thickness in a number of ESRD patients, accordingly patients were selected and subdivided into two groups; 30 with atherosclerosis (mean [SD] age, 51.6 [6.3] years) and 30 without (mean [SD] age, 48.9 [5.5] years). Another 30 healthy individuals (mean [SD] age, 48.5 [6.8] years) were enrolled. Analysis of CD4+CD28null T-cells frequency by flow-cytometry was performed in all studied subjects. CD4+CD28null T cell percentage was significantly higher in ESRD patients, (mean [SD], 7.3 [2.7] %) compared to healthy individuals (mean [SD], 3.0 [0.8] %), (p < 0.001). Additionally, the expansion of these unusual T lymphocytes was significantly higher in ESRD patients with atherosclerotic changes (mean [SD], 9.47 [0.75] %) compared to those without atherosclerosis (mean [SD], 5.22 [2.14] %), (p < 0.001). In conclusion circulating CD4+CD28null T lymphocyte population showed expansion in ESRD patients, and of interest in correlation to preclinical atherosclerotic changes.
Collapse
Affiliation(s)
- Ashraf Mahmoud Okba
- Department of Internal Medicine, Clinical Immunology and Allergy, Faculty of Medicine, Ain Shams University, Egypt
| | | | - Mohamed Nazmy Farres
- Department of Internal Medicine, Clinical Immunology and Allergy, Faculty of Medicine, Ain Shams University, Egypt
| | - Nermine Abd El Nour Melek
- Department of Internal Medicine, Clinical Immunology and Allergy, Faculty of Medicine, Ain Shams University, Egypt
| | - Mariam Maged Amin
- Department of Internal Medicine, Clinical Immunology and Allergy, Faculty of Medicine, Ain Shams University, Egypt.
| | - Nelly Nader Gendy
- Department of Internal Medicine and Nephrology, Theodor Bilharz Research Institute, Egypt
| |
Collapse
|
48
|
Loss of Regulatory Immune Function in Coronary Artery Disease Patients from the Indian Population. J Cardiovasc Transl Res 2019; 12:378-388. [DOI: 10.1007/s12265-019-09872-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/22/2018] [Accepted: 02/13/2019] [Indexed: 10/27/2022]
|
49
|
Jiang Y, Liu X, Du Y, Zhou S. rs1234313 and rs45454293 are risk factors of cerebral arterial thrombosis, large artery atherosclerosis, and carotid plaque in the Han Chinese population: a case-control study. BMC Neurol 2019; 19:31. [PMID: 30797237 PMCID: PMC6387510 DOI: 10.1186/s12883-019-1259-9] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2018] [Accepted: 02/15/2019] [Indexed: 02/02/2023] Open
Abstract
Background Ischemic stroke is a leading cause of mortality and morbidity worldwide. Stenosis or blockage of an artery from atherosclerosis can cause insufficient cerebral blood supply, which leads to ischemic stroke. It has been reported that the polymorphisms of TNFSF4 (tumor necrosis factor super family member 4) are associated with multiple autoimmune diseases. However, it is still unclear whether TNFSF4 gene polymorphisms are associated with ischemic stroke in the Han Chinese population. Here we analyzed the association between TNFSF4 single nucleotide polymorphisms (SNPs) and cerebral arterial thrombosis in the Han Chinese population. Method We consecutively recruited 481 patients with cerebral arterial thrombosis and 538 healthy controls. Neck ultrasonography and magnetic resonance imaging (MRI) were used to evaluate large artery atherosclerosis (LAA) and small vessel disease (SVD), as well as the thickness and calcification of carotid artery. DNA was purified from the peripheral blood samples. TNFSF4 SNPs, rs1234313 and rs45454293, were genotyped using PCR. Results rs1234313 SNP had a significant correlation with the LAA and SVD subtypes in allelic (G vs A), dominate (GG/GA vs AA) and genotypic (GA vs AA; GG vs AA) models, as well as with the calcification of carotid plaque in dominant (GG/GA vs AA, p = 0.022) and genotypic (GA vs AA, p = 0.01) models. rs45454293 SNP had a significant correlation with the LAA and SVD subtypes in allelic (G vs A) and genotypic models, as well as with the thick carotid plaque in allelic (G vs A, p = 0.01) model. Conclusion TNFSF4 SNPs, rs1234313 and rs45454293, are associated with the risk of specific subtypes of cerebral arterial thrombosis in the Han Chinese population.
Collapse
Affiliation(s)
- Yan Jiang
- Department of Neurology, Qilu Hospital of Shandong University and Brain Science Research Institute, Shandong University, 107 Wenhuaxi Road, Jinan, Shandong, 250012, People's Republic of China.,Department of Neurology, The First Affiliated Hospital of USTC, Anhui Provincial Hospital, Hefei, 230001, People's Republic of China
| | - Xiaomin Liu
- Department of Neurology, The First Affiliated Hospital of USTC, Anhui Provincial Hospital, Hefei, 230001, People's Republic of China
| | - Yifeng Du
- Department of Neurology, Shandong Provincial Hospital, Shandong University, Jinan, 250013, China
| | - Shengnian Zhou
- Department of Neurology, Qilu Hospital of Shandong University and Brain Science Research Institute, Shandong University, 107 Wenhuaxi Road, Jinan, Shandong, 250012, People's Republic of China.
| |
Collapse
|
50
|
Abstract
The diminished capacity for wound healing in patients with diabetes contributes to morbidity through ulceration and recurrent infections, loss of function and decreased workplace productivity, increased hospitalisation rates, and rising health-care costs. These are due to diabetes' effects on signalling molecules, cellular cascades, different cell populations, and the vasculature. The function of multiple immune system components including cellular response, blood factors, and vascular tone are all negatively impacted by diabetes. The purpose of this paper is to review the current understanding of immune and vascular dysfunction contributing to impaired wound healing mechanisms in the diabetic population. Normal wound healing mechanisms are reviewed followed by diabetic aberrations to immune and inflammatory function and atherogenesis and angiopathy. DECLARATION OF INTEREST The authors have no financial or personal relationships to people or organisations that could potentially and inappropriately influence their work and conclusions.
Collapse
Affiliation(s)
- A S Ahmed
- Baylor College of Medicine, 1 Baylor Plaza, Houston, Texas 77030
| | - E L Antonsen
- Baylor College of Medicine, 1 Baylor Plaza, Houston, Texas 77030.,Department of Medicine - Section of Emergency Medicine, 1 Baylor Plaza, Houston, Texas.,Department of Medicine - Section of Space Medicine, 1 Baylor Plaza, Houston, Texas
| |
Collapse
|