1
|
Xu Y, Wang Z, Li S, Su J, Gao L, Ou J, Lin Z, Luo OJ, Xiao C, Chen G. An in-depth understanding of the role and mechanisms of T cells in immune organ aging and age-related diseases. SCIENCE CHINA. LIFE SCIENCES 2025; 68:328-353. [PMID: 39231902 DOI: 10.1007/s11427-024-2695-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/24/2024] [Accepted: 07/28/2024] [Indexed: 09/06/2024]
Abstract
T cells play a critical and irreplaceable role in maintaining overall health. However, their functions undergo alterations as individuals age. It is of utmost importance to comprehend the specific characteristics of T-cell aging, as this knowledge is crucial for gaining deeper insights into the pathogenesis of aging-related diseases and developing effective therapeutic strategies. In this review, we have thoroughly examined the existing studies on the characteristics of immune organ aging. Furthermore, we elucidated the changes and potential mechanisms that occur in T cells during the aging process. Additionally, we have discussed the latest research advancements pertaining to T-cell aging-related diseases. These findings provide a fresh perspective for the study of T cells in the context of aging.
Collapse
Affiliation(s)
- Yudai Xu
- Department of Microbiology and Immunology, School of Medicine; Institute of Geriatric Immunology, School of Medicine, Jinan University, Guangzhou, 510632, China
- Key Laboratory of Viral Pathogenesis & Infection Prevention and Control (Jinan University), Ministry of Education, Guangzhou, 510632, China
- Guangdong-Hong Kong-Macau Great Bay Area Geroscience Joint Laboratory, School of Medicine, Jinan University, Guangzhou, 510632, China
| | - Zijian Wang
- Department of Microbiology and Immunology, School of Medicine; Institute of Geriatric Immunology, School of Medicine, Jinan University, Guangzhou, 510632, China
- Key Laboratory of Viral Pathogenesis & Infection Prevention and Control (Jinan University), Ministry of Education, Guangzhou, 510632, China
- Guangdong-Hong Kong-Macau Great Bay Area Geroscience Joint Laboratory, School of Medicine, Jinan University, Guangzhou, 510632, China
| | - Shumin Li
- Department of Microbiology and Immunology, School of Medicine; Institute of Geriatric Immunology, School of Medicine, Jinan University, Guangzhou, 510632, China
- Key Laboratory of Viral Pathogenesis & Infection Prevention and Control (Jinan University), Ministry of Education, Guangzhou, 510632, China
- Guangdong-Hong Kong-Macau Great Bay Area Geroscience Joint Laboratory, School of Medicine, Jinan University, Guangzhou, 510632, China
| | - Jun Su
- First Affiliated Hospital, Jinan University, Guangzhou, 510630, China
| | - Lijuan Gao
- Department of Microbiology and Immunology, School of Medicine; Institute of Geriatric Immunology, School of Medicine, Jinan University, Guangzhou, 510632, China
- Key Laboratory of Viral Pathogenesis & Infection Prevention and Control (Jinan University), Ministry of Education, Guangzhou, 510632, China
- Guangdong-Hong Kong-Macau Great Bay Area Geroscience Joint Laboratory, School of Medicine, Jinan University, Guangzhou, 510632, China
| | - Junwen Ou
- Anti Aging Medical Center, Clifford Hospital, Guangzhou, 511495, China
| | - Zhanyi Lin
- Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, 510080, China
| | - Oscar Junhong Luo
- Department of Systems Biomedical Sciences, School of Medicine, Jinan University, Guangzhou, 510632, China
| | - Chanchan Xiao
- Department of Microbiology and Immunology, School of Medicine; Institute of Geriatric Immunology, School of Medicine, Jinan University, Guangzhou, 510632, China.
- Key Laboratory of Viral Pathogenesis & Infection Prevention and Control (Jinan University), Ministry of Education, Guangzhou, 510632, China.
- Guangdong-Hong Kong-Macau Great Bay Area Geroscience Joint Laboratory, School of Medicine, Jinan University, Guangzhou, 510632, China.
- The Sixth Affiliated Hospital of Jinan University (Dongguan Eastern Central Hospital), Jinan University, Dongguan, 523000, China.
- Zhuhai Institute of Jinan University, Jinan University, Zhuhai, 519070, China.
| | - Guobing Chen
- Department of Microbiology and Immunology, School of Medicine; Institute of Geriatric Immunology, School of Medicine, Jinan University, Guangzhou, 510632, China.
- Key Laboratory of Viral Pathogenesis & Infection Prevention and Control (Jinan University), Ministry of Education, Guangzhou, 510632, China.
- Guangdong-Hong Kong-Macau Great Bay Area Geroscience Joint Laboratory, School of Medicine, Jinan University, Guangzhou, 510632, China.
- The Sixth Affiliated Hospital of Jinan University (Dongguan Eastern Central Hospital), Jinan University, Dongguan, 523000, China.
- Zhuhai Institute of Jinan University, Jinan University, Zhuhai, 519070, China.
| |
Collapse
|
2
|
Wang Y, Zhang Y, Gong G, Liu Q, Li L, Zhang M, Shen S, Wang R, Wu J, Xu W. Single-cell analysis of human peripheral blood reveals high immune response activity in successful ageing individuals. Mech Ageing Dev 2025; 223:112011. [PMID: 39622417 DOI: 10.1016/j.mad.2024.112011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2024] [Revised: 11/21/2024] [Accepted: 11/25/2024] [Indexed: 12/07/2024]
Abstract
Beneficial remodeling of the immune system in successful ageing individuals (centenarians and supercentenarians) is critical for healthy ageing. However, mechanisms for dynamic regulation of immunity during ageing remain unclear. We use single-cell RNA sequencing (scRNA-seq) as an analytical strategy to study the dynamic regulation of immunity during aging and its molecular mechanisms at the single-cell level. We performed an integrative analysis of 87,215 peripheral blood mononuclear cells, from seven supercentenarians, three centenarians, and four elderly controls, generated by single-cell transcriptomics complemented with fluorescence-activated cell sorting. Animals experiments were also conducted to validate the makers of healthy aging found by our bioinformatic analysis and further explore the dynamic of immune changes during aging process. We found that CD8+ effector memory T cells and terminally differentiated B cells were enriched in the longevity group (centenarians and supercentenarians), whereas naïve T cells and Tregs were enriched in elderly controls. CD56dim NK cells in the longevity group activated Fc-γ receptor signaling. The higher antigen-presenting ability of CD14+ monocytes in the longevity group and the CellChat analysis indicated that CD14+ monocytes might assist active T and B cells. Here, we revealed the adaptive immune remodeling geromarkers of immunosenescence in centenarians and supercentenarians, which could be considered as biomarkers of healthy aging, and might help sustain immune responses and achieve exceptional longevity.
Collapse
Affiliation(s)
- Yu Wang
- Key Laboratory of Geriatrics of Jiangsu Province, Department of Geriatrics, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China; Department of Geriatrics, The Fourth Affiliated Hospital of Nanjing Medical University, Nanjing 210031, China
| | - Yuxing Zhang
- Key Laboratory of Geriatrics of Jiangsu Province, Department of Geriatrics, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China
| | - Ge Gong
- Key Laboratory of Geriatrics of Jiangsu Province, Department of Geriatrics, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China
| | - Quanzhong Liu
- Department of Bioinformatics, Nanjing Medical University, Nanjing 211166, China
| | - Liangyu Li
- Department of Bioinformatics, Nanjing Medical University, Nanjing 211166, China; BGI-Shenzhen, Shenzhen, Guangdong 518083, China
| | - Mingjiong Zhang
- Key Laboratory of Geriatrics of Jiangsu Province, Department of Geriatrics, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China
| | - Shuping Shen
- Key Laboratory of Geriatrics of Jiangsu Province, Department of Geriatrics, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China
| | - Ran Wang
- Key Laboratory of Geriatrics of Jiangsu Province, Department of Geriatrics, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China
| | - Jianqing Wu
- Key Laboratory of Geriatrics of Jiangsu Province, Department of Geriatrics, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China.
| | - Wei Xu
- Key Laboratory of Geriatrics of Jiangsu Province, Department of Geriatrics, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China.
| |
Collapse
|
3
|
Martini E, Cremonesi M, Felicetta A, Serio S, Puccio S, Pelamatti E, van Beek JJP, Papadopoulou V, Catalano C, Fanuele F, Giuliano D, Basso G, Bonfiglio CA, Panico C, Vacchiano M, Carullo P, Papa L, D'Andrea C, Tuzger N, Marchini S, Magistroni P, Deaglio S, Amoroso A, Lugli E, Condorelli G, Kallikourdis M. Autoimmune-Like Mechanism in Heart Failure Enables Preventive Vaccine Therapy. Circ Res 2025; 136:4-25. [PMID: 39629560 DOI: 10.1161/circresaha.124.324999] [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: 07/17/2024] [Revised: 10/31/2024] [Accepted: 11/20/2024] [Indexed: 01/04/2025]
Abstract
BACKGROUND Heart failure (HF) is strongly associated with inflammation. In pressure overload (PO)-induced HF, cardiac stress triggers adaptive immunity, ablation or inhibition of which blocks disease progression. We hypothesized that PO-HF might fulfill the often-used criteria of autoimmunity: if so, the associated adaptive immune response would be not only necessary but also sufficient to induce HF; it should also be possible to identify self-antigens driving the autoimmune response. Finally, we hypothesized that such an antigen-specific response can be manipulated to preventively reduce the severity of PO-HF in a tolerizing vaccine. METHODS We used the transfer of lymphocytes or serum from PO-HF mice into healthy recipients to assess whether the adaptive response is sufficient to induce disease. We devised a novel pipeline to identify self-antigens driving the response. We immunized healthy mice with novel antigens to assess whether they induce disease. To determine whether these antigens could be present in human patients, we sought to detect existing responses against these antigens in patients with HF. Finally, we used the antigens in an oral tolerance protocol to preventively protect mice from subsequently induced PO-HF, analyzing the results with next-generation sequencing. RESULTS We found that PO-HF fulfills the criteria of an autoimmune disease, albeit partially, and identified novel cardiac self-antigens, capable of inducing cardiac dysfunction. The novel antigens in a tolerizing vaccine formulation preemptively reduced the severity of disease triggered by subsequent application of PO, via induction of effector regulatory T cells, enabling a potent reduction of PO-driven loss of systolic function, cardiac inflammation, and proinflammatory CD4+ T-cell clonal expansion. CONCLUSIONS We demonstrate that PO-HF is triggered by hemodynamic stress and then sets off an autoimmune-like response against cardiac self-antigens. The antigens can be used to reduce the severity of future-onset disease, via oral tolerization, effectively acting as a protective vaccine.
Collapse
Affiliation(s)
- Elisa Martini
- Adaptive Immunity Laboratory, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Humanitas Research Hospital, Rozzano, Italy (E.M., M.C., E.P., V.P., C.C., F.F., C.A.B., N.T., M.K.)
| | - Marco Cremonesi
- Adaptive Immunity Laboratory, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Humanitas Research Hospital, Rozzano, Italy (E.M., M.C., E.P., V.P., C.C., F.F., C.A.B., N.T., M.K.)
| | - Arianna Felicetta
- Cardio Center, IRCCS Humanitas Research Hospital, Rozzano, Italy (A.F., S.S., C.P., M.V., P.C., L.P., C.D., G.C.)
| | - Simone Serio
- Cardio Center, IRCCS Humanitas Research Hospital, Rozzano, Italy (A.F., S.S., C.P., M.V., P.C., L.P., C.D., G.C.)
- Institute of Genetics and Biomedical Research, Milan Unit, Consiglio Nazionale delle Ricerche, Italy (S.S., S.P.)
| | - Simone Puccio
- Laboratory of Translational Immunology, IRCCS Humanitas Research Hospital, Rozzano, Italy (S.P., J.J.P.v.B., E.L.)
- Institute of Genetics and Biomedical Research, Milan Unit, Consiglio Nazionale delle Ricerche, Italy (S.S., S.P.)
| | - Erica Pelamatti
- Adaptive Immunity Laboratory, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Humanitas Research Hospital, Rozzano, Italy (E.M., M.C., E.P., V.P., C.C., F.F., C.A.B., N.T., M.K.)
| | - Jasper J P van Beek
- Laboratory of Translational Immunology, IRCCS Humanitas Research Hospital, Rozzano, Italy (S.P., J.J.P.v.B., E.L.)
| | - Vasiliki Papadopoulou
- Adaptive Immunity Laboratory, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Humanitas Research Hospital, Rozzano, Italy (E.M., M.C., E.P., V.P., C.C., F.F., C.A.B., N.T., M.K.)
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, Italy (V.P., C.C., F.F., C.P., C.D., N.T., G.C., M.K.)
| | - Chiara Catalano
- Adaptive Immunity Laboratory, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Humanitas Research Hospital, Rozzano, Italy (E.M., M.C., E.P., V.P., C.C., F.F., C.A.B., N.T., M.K.)
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, Italy (V.P., C.C., F.F., C.P., C.D., N.T., G.C., M.K.)
| | - Francesca Fanuele
- Adaptive Immunity Laboratory, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Humanitas Research Hospital, Rozzano, Italy (E.M., M.C., E.P., V.P., C.C., F.F., C.A.B., N.T., M.K.)
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, Italy (V.P., C.C., F.F., C.P., C.D., N.T., G.C., M.K.)
| | - Desirée Giuliano
- Humanitas Genomics Facility, IRCCS Humanitas Research Hospital, Rozzano, Italy (D.G., G.B., S.M.)
| | - Gianluca Basso
- Humanitas Genomics Facility, IRCCS Humanitas Research Hospital, Rozzano, Italy (D.G., G.B., S.M.)
| | - Cecilia Assunta Bonfiglio
- Adaptive Immunity Laboratory, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Humanitas Research Hospital, Rozzano, Italy (E.M., M.C., E.P., V.P., C.C., F.F., C.A.B., N.T., M.K.)
- Institute for Cardiovascular Prevention, Ludwig-Maximilians-Universität München, Germany (C.A.B.)
| | - Cristina Panico
- Cardio Center, IRCCS Humanitas Research Hospital, Rozzano, Italy (A.F., S.S., C.P., M.V., P.C., L.P., C.D., G.C.)
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, Italy (V.P., C.C., F.F., C.P., C.D., N.T., G.C., M.K.)
| | - Marco Vacchiano
- Cardio Center, IRCCS Humanitas Research Hospital, Rozzano, Italy (A.F., S.S., C.P., M.V., P.C., L.P., C.D., G.C.)
| | - Pierluigi Carullo
- Cardio Center, IRCCS Humanitas Research Hospital, Rozzano, Italy (A.F., S.S., C.P., M.V., P.C., L.P., C.D., G.C.)
| | - Laura Papa
- Cardio Center, IRCCS Humanitas Research Hospital, Rozzano, Italy (A.F., S.S., C.P., M.V., P.C., L.P., C.D., G.C.)
| | - Carla D'Andrea
- Cardio Center, IRCCS Humanitas Research Hospital, Rozzano, Italy (A.F., S.S., C.P., M.V., P.C., L.P., C.D., G.C.)
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, Italy (V.P., C.C., F.F., C.P., C.D., N.T., G.C., M.K.)
| | - Naz Tuzger
- Adaptive Immunity Laboratory, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Humanitas Research Hospital, Rozzano, Italy (E.M., M.C., E.P., V.P., C.C., F.F., C.A.B., N.T., M.K.)
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, Italy (V.P., C.C., F.F., C.P., C.D., N.T., G.C., M.K.)
| | - Sergio Marchini
- Humanitas Genomics Facility, IRCCS Humanitas Research Hospital, Rozzano, Italy (D.G., G.B., S.M.)
| | - Paola Magistroni
- Immunogenetics and Transplant Biology Service, Città della Salute e della Scienza Hospital, Turin, Italy (P.M., S.D., A.A.)
| | - Silvia Deaglio
- Immunogenetics and Transplant Biology Service, Città della Salute e della Scienza Hospital, Turin, Italy (P.M., S.D., A.A.)
- Department of Medical Sciences, University of Turin, Italy (S.D., A.A.)
| | - Antonio Amoroso
- Immunogenetics and Transplant Biology Service, Città della Salute e della Scienza Hospital, Turin, Italy (P.M., S.D., A.A.)
- Department of Medical Sciences, University of Turin, Italy (S.D., A.A.)
| | - Enrico Lugli
- Laboratory of Translational Immunology, IRCCS Humanitas Research Hospital, Rozzano, Italy (S.P., J.J.P.v.B., E.L.)
| | - Gianluigi Condorelli
- Cardio Center, IRCCS Humanitas Research Hospital, Rozzano, Italy (A.F., S.S., C.P., M.V., P.C., L.P., C.D., G.C.)
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, Italy (V.P., C.C., F.F., C.P., C.D., N.T., G.C., M.K.)
| | - Marinos Kallikourdis
- Adaptive Immunity Laboratory, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Humanitas Research Hospital, Rozzano, Italy (E.M., M.C., E.P., V.P., C.C., F.F., C.A.B., N.T., M.K.)
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, Italy (V.P., C.C., F.F., C.P., C.D., N.T., G.C., M.K.)
| |
Collapse
|
4
|
Jung M, Kim H, Choi E, Shin MK, Shin SJ. Enhancing vaccine effectiveness in the elderly to counter antibiotic resistance: The potential of adjuvants via pattern recognition receptors. Hum Vaccin Immunother 2024; 20:2317439. [PMID: 39693178 DOI: 10.1080/21645515.2024.2317439] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2023] [Revised: 01/24/2024] [Accepted: 02/07/2024] [Indexed: 12/20/2024] Open
Abstract
Vaccines are an effective way to prevent the emergence and spread of antibiotic resistance by preventing diseases and establishing herd immunity. However, the reduced effectiveness of vaccines in the elderly due to immunosenescence is one of the significant contributors to the increasing antibiotic resistance. To counteract this decline and enhance vaccine effectiveness in the elderly, adjuvants play a pivotal role. Adjuvants are designed to augment the effectiveness of vaccines by activating the innate immune system, particularly through pattern recognition receptors on antigen-presenting cells. To improve vaccine effectiveness in the elderly using adjuvants, it is imperative to select the appropriate adjuvants based on an understanding of immunosenescence and the mechanisms of adjuvant functions. This review demonstrates the phenomenon of immunosenescence and explores various types of adjuvants, including their mechanisms and their potential in improving vaccine effectiveness for the elderly, thereby contributing to developing more effective vaccines for this vulnerable demographic.
Collapse
Affiliation(s)
- Myunghwan Jung
- Department of Microbiology, Institute of Medical Science, Department of Convergence Medical Science, BK21 Center for Human Resource Development in the Bio-Health Industry, Gyeongsang National University College of Medicine, Jinju, South Korea
| | - Hongmin Kim
- Department of Microbiology, Institute for Immunology and Immunological Diseases, Graduate School of Medical Science, Brain Korea 21 Project, Yonsei University College of Medicine, Seoul, South Korea
| | - Eunsol Choi
- Department of Microbiology, Institute for Immunology and Immunological Diseases, Graduate School of Medical Science, Brain Korea 21 Project, Yonsei University College of Medicine, Seoul, South Korea
| | - Min-Kyoung Shin
- Department of Microbiology, Institute of Medical Science, Department of Convergence Medical Science, BK21 Center for Human Resource Development in the Bio-Health Industry, Gyeongsang National University College of Medicine, Jinju, South Korea
| | - Sung Jae Shin
- Department of Microbiology, Institute for Immunology and Immunological Diseases, Graduate School of Medical Science, Brain Korea 21 Project, Yonsei University College of Medicine, Seoul, South Korea
| |
Collapse
|
5
|
Silva RCMC. The dichotomic role of cytokines in aging. Biogerontology 2024; 26:17. [PMID: 39621124 DOI: 10.1007/s10522-024-10152-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2024] [Accepted: 10/30/2024] [Indexed: 12/11/2024]
Abstract
The chronic inflammation present in aged individuals is generally depicted as a detrimental player for longevity. Here, it is discussed several beneficial effects associated with the cytokines that are chronically elevated in inflammaging. These cytokines, such as IL-1β, type I interferons, IL-6 and TNF positively regulate macroautophagy, mitochondrial function, anti-tumor immune responses and skeletal muscle biogenesis, possibly contributing to longevity. On the other side, the detrimental and antagonistic role of these cytokines including the induction of sarcopenia, tissue damage and promotion of tumorigenesis are also discussed, underscoring the dichotomy associated with inflammaging and its players. In addition, it is discussed the role of the anti-inflammatory cytokine IL-10 and other cytokines that affect aging in a more linear way, such as IL-11, which promotes senescence, and IL-4 and IL-15, which promotes longevity. It is also discussed more specific regulators of aging that are downstream cytokines-mediated signaling.
Collapse
|
6
|
Wang B, Han J, Elisseeff JH, Demaria M. The senescence-associated secretory phenotype and its physiological and pathological implications. Nat Rev Mol Cell Biol 2024; 25:958-978. [PMID: 38654098 DOI: 10.1038/s41580-024-00727-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/21/2024] [Indexed: 04/25/2024]
Abstract
Cellular senescence is a state of terminal growth arrest associated with the upregulation of different cell cycle inhibitors, mainly p16 and p21, structural and metabolic alterations, chronic DNA damage responses, and a hypersecretory state known as the senescence-associated secretory phenotype (SASP). The SASP is the major mediator of the paracrine effects of senescent cells in their tissue microenvironment and of various local and systemic biological functions. In this Review, we discuss the composition, dynamics and heterogeneity of the SASP as well as the mechanisms underlying its induction and regulation. We describe the various biological properties of the SASP, its beneficial and detrimental effects in different physiological and pathological settings, and its impact on overall health span. Finally, we discuss the use of the SASP as a biomarker and of SASP inhibitors as senomorphic interventions to treat cancer and other age-related conditions.
Collapse
Affiliation(s)
- Boshi Wang
- European Research Institute for the Biology of Ageing (ERIBA), University Medical Center Groningen (UMCG), University of Groningen (RUG), Groningen, Netherlands
| | - Jin Han
- Translational Tissue Engineering Center, Wilmer Eye Institute, and Department of Biomedical Engineering, John Hopkins University School of Medicine, Baltimore MD, MD, USA
| | - Jennifer H Elisseeff
- Translational Tissue Engineering Center, Wilmer Eye Institute, and Department of Biomedical Engineering, John Hopkins University School of Medicine, Baltimore MD, MD, USA
| | - Marco Demaria
- European Research Institute for the Biology of Ageing (ERIBA), University Medical Center Groningen (UMCG), University of Groningen (RUG), Groningen, Netherlands.
| |
Collapse
|
7
|
Kalinoski H, Daoud A, Rusinkevich V, Jurčová I, Talor MV, Welsh RA, Hughes D, Zemanová K, Stříž I, Hooper JE, Kautzner J, Peichl P, Melenovský V, Won T, Čiháková D. Injury-induced myosin-specific tissue-resident memory T cells drive immune checkpoint inhibitor myocarditis. Proc Natl Acad Sci U S A 2024; 121:e2323052121. [PMID: 39378095 PMCID: PMC11494310 DOI: 10.1073/pnas.2323052121] [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: 01/21/2024] [Accepted: 09/10/2024] [Indexed: 10/10/2024] Open
Abstract
Cardiac myosin-specific (MyHC) T cells drive the disease pathogenesis of immune checkpoint inhibitor-associated myocarditis (ICI-myocarditis). To determine whether MyHC T cells are tissue-resident memory T (TRM) cells, we characterized cardiac TRM cells in naive mice and established that they have a distinct phenotypic and transcriptional profile that can be defined by their upregulation of CD69, PD-1, and CXCR6. We then investigated the effects of cardiac injury through a modified experimental autoimmune myocarditis mouse model and an ischemia-reperfusion injury mouse model and determined that cardiac inflammation induces the recruitment of autoreactive MyHC TRM cells, which coexpress PD-1 and CD69. To investigate whether the recruited MyHC TRM cells could increase susceptibility to ICI-myocarditis, we developed a two-hit ICI-myocarditis mouse model where cardiac injury was induced, mice were allowed to recover, and then were treated with anti-PD-1 antibodies. We determined that mice who recover from cardiac injury are more susceptible to ICI-myocarditis development. We found that murine and human TRM cells share a similar location in the heart and aggregate along the perimyocardium. We phenotyped cells obtained from pericardial fluid from patients diagnosed with dilated cardiomyopathy and ischemic cardiomyopathy and established that pericardial T cells are predominantly CD69+ TRM cells that up-regulate PD-1. Finally, we determined that human pericardial macrophages produce IL-15, which supports and maintains pericardial TRM cells.
Collapse
Affiliation(s)
- Hannah Kalinoski
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD21205
| | - Abdel Daoud
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD21205
| | - Vitali Rusinkevich
- Department of Pathology, School of Medicine, Johns Hopkins University, Baltimore, MD21205
| | - Ivana Jurčová
- Department of Cardiology, Institute for Clinical and Experimental Medicine, Prague140 21, Czech Republic
| | - Monica V. Talor
- Department of Pathology, School of Medicine, Johns Hopkins University, Baltimore, MD21205
| | - Robin A. Welsh
- Department of Pathology, School of Medicine, Johns Hopkins University, Baltimore, MD21205
| | - David Hughes
- Department of Pathology, School of Medicine, Johns Hopkins University, Baltimore, MD21205
- Department of Chemical and Biomolecular Engineering, Whiting School of Engineering, Johns Hopkins University, Baltimore, MD21205
| | - Kateřina Zemanová
- Department of Clinical and Transplant Immunology, Institute for Clinical and Experimental Medicine, Prague140 21, Czech Republic
| | - Ilja Stříž
- Department of Clinical and Transplant Immunology, Institute for Clinical and Experimental Medicine, Prague140 21, Czech Republic
| | - Jody E. Hooper
- Department of Pathology, School of Medicine, Johns Hopkins University, Baltimore, MD21205
| | - Josef Kautzner
- Department of Cardiology, Institute for Clinical and Experimental Medicine, Prague140 21, Czech Republic
| | - Petr Peichl
- Department of Cardiology, Institute for Clinical and Experimental Medicine, Prague140 21, Czech Republic
| | - Vojtěch Melenovský
- Department of Cardiology, Institute for Clinical and Experimental Medicine, Prague140 21, Czech Republic
| | - Taejoon Won
- Department of Pathology, School of Medicine, Johns Hopkins University, Baltimore, MD21205
| | - Daniela Čiháková
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD21205
- Department of Pathology, School of Medicine, Johns Hopkins University, Baltimore, MD21205
| |
Collapse
|
8
|
Vyas V, Sandhar B, Keane JM, Wood EG, Blythe H, Jones A, Shahaj E, Fanti S, Williams J, Metic N, Efremova M, Ng HL, Nageswaran G, Byrne S, Feldhahn N, Marelli-Berg F, Chain B, Tinker A, Finlay MC, Longhi MP. Tissue-resident memory T cells in epicardial adipose tissue comprise transcriptionally distinct subsets that are modulated in atrial fibrillation. NATURE CARDIOVASCULAR RESEARCH 2024; 3:1067-1082. [PMID: 39271815 PMCID: PMC11399095 DOI: 10.1038/s44161-024-00532-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/20/2023] [Accepted: 07/29/2024] [Indexed: 09/15/2024]
Abstract
Atrial fibrillation (AF) is the most common sustained arrhythmia and carries an increased risk of stroke and heart failure. Here we investigated how the immune infiltrate of human epicardial adipose tissue (EAT), which directly overlies the myocardium, contributes to AF. Flow cytometry analysis revealed an enrichment of tissue-resident memory T (TRM) cells in patients with AF. Cellular indexing of transcriptomes and epitopes by sequencing (CITE-seq) and single-cell T cell receptor (TCR) sequencing identified two transcriptionally distinct CD8+ TRM cells that are modulated in AF. Spatial transcriptomic analysis of EAT and atrial tissue identified the border region between the tissues to be a region of intense inflammatory and fibrotic activity, and the addition of TRM populations to atrial cardiomyocytes demonstrated their ability to differentially alter calcium flux as well as activate inflammatory and apoptotic signaling pathways. This study identified EAT as a reservoir of TRM cells that can directly modulate vulnerability to cardiac arrhythmia.
Collapse
Affiliation(s)
- Vishal Vyas
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, UK
- Department of Cardiology, Barts Heart Centre, St. Bartholomew's Hospital, London, UK
| | - Balraj Sandhar
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Jack M Keane
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Elizabeth G Wood
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Hazel Blythe
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Aled Jones
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Eriomina Shahaj
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Silvia Fanti
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Jack Williams
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Nasrine Metic
- Cancer Research UK, Barts Centre, Queen Mary University of London, London, UK
| | - Mirjana Efremova
- Cancer Research UK, Barts Centre, Queen Mary University of London, London, UK
| | - Han Leng Ng
- Department of Immunology and Inflammation, Centre for Haematology, Faculty of Medicine, Imperial College London, London, UK
| | - Gayathri Nageswaran
- UCL Division of Infection and Immunity, University College London, London, UK
| | - Suzanne Byrne
- UCL Division of Infection and Immunity, University College London, London, UK
| | - Niklas Feldhahn
- Department of Immunology and Inflammation, Centre for Haematology, Faculty of Medicine, Imperial College London, London, UK
| | - Federica Marelli-Berg
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Benny Chain
- UCL Division of Infection and Immunity, University College London, London, UK
| | - Andrew Tinker
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Malcolm C Finlay
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, UK
- Department of Cardiology, Barts Heart Centre, St. Bartholomew's Hospital, London, UK
| | - M Paula Longhi
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, UK.
| |
Collapse
|
9
|
Shi Y, Zhao L, Wang J, Liu X, Bai Y, Cong H, Li X. Empagliflozin protects against heart failure with preserved ejection fraction partly by inhibiting the senescence-associated STAT1-STING axis. Cardiovasc Diabetol 2024; 23:269. [PMID: 39044275 PMCID: PMC11267814 DOI: 10.1186/s12933-024-02366-0] [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: 04/18/2024] [Accepted: 07/16/2024] [Indexed: 07/25/2024] Open
Abstract
Heart failure with preserved ejection fraction (HFpEF) is a mortal clinical syndrome without effective therapies. Empagliflozin (EMPA) improves cardiovascular outcomes in HFpEF patients, but the underlying mechanism remains elusive. Here, mice were fed a high-fat diet (HFD) supplemented with L-NAME for 12 weeks and subsequently intraperitoneally injected with EMPA for another 4 weeks. A 4D-DIA proteomic assay was performed to detect protein changes in the failing hearts. We identified 310 differentially expressed proteins (DEPs) (ctrl vs. HFpEF group) and 173 DEPs (HFpEF vs. EMPA group). The regulation of immune system processes was enriched in all groups and the interferon response genes (STAT1, Ifit1, Ifi35 and Ifi47) were upregulated in HFpEF mice but downregulated after EMPA administration. In addition, EMPA treatment suppressed the increase in the levels of aging markers (p16 and p21) in HFpEF hearts. Further bioinformatics analysis verified STAT1 as the hub transcription factor during pathological changes in HFpEF mice. We next treated H9C2 cells with IFN-γ, a primary agonist of STAT1 phosphorylation, to investigate whether EMPA plays a beneficial role by blocking STAT1 activation. Our results showed that IFN-γ treatment caused cardiomyocyte senescence and STAT1 activation, which were inhibited by EMPA administration. Notably, STAT1 inhibition significantly reduced cellular senescence possibly by regulating STING expression. Our findings revealed that EMPA mitigates cardiac inflammation and aging in HFpEF mice by inhibiting STAT1 activation. The STAT1-STING axis may act as a pivotal mechanism in the pathogenesis of HFpEF, especially under inflammatory and aging conditions.
Collapse
Affiliation(s)
- Ying Shi
- Tianjin Chest Hospital, Tianjin Medical University, Tianjin, 300070, China
- Tianjin Key Laboratory of Cardiovascular Emergency and Critical Care, Tianjin Municipal Science and Technology Bureau, Tianjin, 300222, China
- Tianjin Institute of Cardiovascular Disease, Tianjin Chest Hospital, Tianjin, 300222, China
| | - Lili Zhao
- Tianjin Institute of Cardiovascular Disease, Tianjin Chest Hospital, Tianjin, 300222, China
| | - Jing Wang
- Tianjin Chest Hospital, Tianjin Medical University, Tianjin, 300070, China
| | - Xiankun Liu
- Department of Cardiac Surgery, Tianjin Chest Hospital, Tianjin, 300222, China
| | - Yiming Bai
- Department of Cardiac Surgery, Tianjin Chest Hospital, Tianjin, 300222, China
| | - Hongliang Cong
- Tianjin Chest Hospital, Tianjin Medical University, Tianjin, 300070, China.
- Tianjin Key Laboratory of Cardiovascular Emergency and Critical Care, Tianjin Municipal Science and Technology Bureau, Tianjin, 300222, China.
- Tianjin Institute of Cardiovascular Disease, Tianjin Chest Hospital, Tianjin, 300222, China.
- Department of Cardiology, Tianjin Chest Hospital, Tianjin, 300222, China.
| | - Ximing Li
- Tianjin Key Laboratory of Cardiovascular Emergency and Critical Care, Tianjin Municipal Science and Technology Bureau, Tianjin, 300222, China.
- Chest Hospital, Tianjin University, Tianjin, 300072, China.
- Department of Cardiology, Tianjin Chest Hospital, Tianjin, 300222, China.
| |
Collapse
|
10
|
Won T, Song EJ, Kalinoski HM, Moslehi JJ, Čiháková D. Autoimmune Myocarditis, Old Dogs and New Tricks. Circ Res 2024; 134:1767-1790. [PMID: 38843292 DOI: 10.1161/circresaha.124.323816] [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: 04/04/2024] [Accepted: 05/08/2024] [Indexed: 06/12/2024]
Abstract
Autoimmunity significantly contributes to the pathogenesis of myocarditis, underscored by its increased frequency in autoimmune diseases such as systemic lupus erythematosus and polymyositis. Even in cases of myocarditis caused by viral infections, dysregulated immune responses contribute to pathogenesis. However, whether triggered by existing autoimmune conditions or viral infections, the precise antigens and immunologic pathways driving myocarditis remain incompletely understood. The emergence of myocarditis associated with immune checkpoint inhibitor therapy, commonly used for treating cancer, has afforded an opportunity to understand autoimmune mechanisms in myocarditis, with autoreactive T cells specific for cardiac myosin playing a pivotal role. Despite their self-antigen recognition, cardiac myosin-specific T cells can be present in healthy individuals due to bypassing the thymic selection stage. In recent studies, novel modalities in suppressing the activity of pathogenic T cells including cardiac myosin-specific T cells have proven effective in treating autoimmune myocarditis. This review offers an overview of the current understanding of heart antigens, autoantibodies, and immune cells as the autoimmune mechanisms underlying various forms of myocarditis, along with the latest updates on clinical management and prospects for future research.
Collapse
Affiliation(s)
- Taejoon Won
- Department of Pathobiology, College of Veterinary Medicine, University of Illinois Urbana-Champaign (T.W.)
| | - Evelyn J Song
- Section of Cardio-Oncology and Immunology, Division of Cardiology and the Cardiovascular Research Institute, University of California San Francisco (E.J.S., J.J.M.)
| | - Hannah M Kalinoski
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Johns Hopkins University Bloomberg School of Public Health, Baltimore, MD (H.M.K., D.Č)
| | - Javid J Moslehi
- Section of Cardio-Oncology and Immunology, Division of Cardiology and the Cardiovascular Research Institute, University of California San Francisco (E.J.S., J.J.M.)
| | - Daniela Čiháková
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Johns Hopkins University Bloomberg School of Public Health, Baltimore, MD (H.M.K., D.Č)
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD (D.Č)
| |
Collapse
|
11
|
Zhang H, Muhetarijiang M, Chen RJ, Hu X, Han J, Zheng L, Chen T. Mitochondrial Dysfunction: A Roadmap for Understanding and Tackling Cardiovascular Aging. Aging Dis 2024:AD.2024.0058. [PMID: 38739929 DOI: 10.14336/ad.2024.0058] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2024] [Accepted: 05/08/2024] [Indexed: 05/16/2024] Open
Abstract
Cardiovascular aging is a progressive remodeling process constituting a variety of cellular and molecular alterations that are closely linked to mitochondrial dysfunction. Therefore, gaining a deeper understanding of the changes in mitochondrial function during cardiovascular aging is crucial for preventing cardiovascular diseases. Cardiac aging is accompanied by fibrosis, cardiomyocyte hypertrophy, metabolic changes, and infiltration of immune cells, collectively contributing to the overall remodeling of the heart. Similarly, during vascular aging, there is a profound remodeling of blood vessel structure. These remodeling present damage to endothelial cells, increased vascular stiffness, impaired formation of new blood vessels (angiogenesis), the development of arteriosclerosis, and chronic vascular inflammation. This review underscores the role of mitochondrial dysfunction in cardiac aging, exploring its impact on fibrosis and myocardial alterations, metabolic remodeling, immune response remodeling, as well as in vascular aging in the heart. Additionally, we emphasize the significance of mitochondria-targeted therapies in preventing cardiovascular diseases in the elderly.
Collapse
Affiliation(s)
- Han Zhang
- Department of Cardiology, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| | - Mairedan Muhetarijiang
- Department of Cardiology, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| | - Ryan J Chen
- School of Medicine, Zhejiang University, Hangzhou, China
| | - Xiaosheng Hu
- Department of Cardiology, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| | - Jie Han
- Department of Cardiology, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| | - Liangrong Zheng
- Department of Cardiology, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| | - Ting Chen
- Department of Cardiology, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
- Key Laboratory of Precision Medicine for Atherosclerotic Diseases of Zhejiang Province, Affiliated First Hospital of Ningbo University, Ningbo, China
| |
Collapse
|
12
|
Figueiredo JC, Bhowmick NA, Karlstaedt A. Metabolic basis of cardiac dysfunction in cancer patients. Curr Opin Cardiol 2024; 39:138-147. [PMID: 38386340 PMCID: PMC11185275 DOI: 10.1097/hco.0000000000001118] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/23/2024]
Abstract
PURPOSE OF REVIEW The relationship between metabolism and cardiovascular diseases is complex and bidirectional. Cardiac cells must adapt metabolic pathways to meet biosynthetic demands and energy requirements to maintain contractile function. During cancer, this homeostasis is challenged by the increased metabolic demands of proliferating cancer cells. RECENT FINDINGS Tumors have a systemic metabolic impact that extends beyond the tumor microenvironment. Lipid metabolism is critical to cancer cell proliferation, metabolic adaptation, and increased cardiovascular risk. Metabolites serve as signals which provide insights for diagnosis and prognosis in cardio-oncology patients. SUMMARY Metabolic processes demonstrate a complex relationship between cancer cell states and cardiovascular remodeling with potential for therapeutic interventions.
Collapse
Affiliation(s)
- Jane C. Figueiredo
- Department of Medicine, Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, CA
| | - Neil Adri Bhowmick
- Department of Medicine, Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, CA
- Division of Hematology and Oncology, Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Anja Karlstaedt
- Advanced Clinical Biosystems Research Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
- Department of Cardiology, Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, CA
| |
Collapse
|
13
|
Tran DT, Batchu SN, Advani A. Interferons and interferon-related pathways in heart disease. Front Cardiovasc Med 2024; 11:1357343. [PMID: 38665231 PMCID: PMC11043610 DOI: 10.3389/fcvm.2024.1357343] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2023] [Accepted: 04/01/2024] [Indexed: 04/28/2024] Open
Abstract
Interferons (IFNs) and IFN-related pathways play key roles in the defence against microbial infection. However, these processes may also be activated during the pathogenesis of non-infectious diseases, where they may contribute to organ injury, or function in a compensatory manner. In this review, we explore the roles of IFNs and IFN-related pathways in heart disease. We consider the cardiac effects of type I IFNs and IFN-stimulated genes (ISGs); the emerging role of the cyclic GMP-AMP synthase (cGAS)-stimulator of interferon genes (STING) pathway; the seemingly paradoxical effects of the type II IFN, IFN-γ; and the varied actions of the interferon regulatory factor (IRF) family of transcription factors. Recombinant IFNs and small molecule inhibitors of mediators of IFN receptor signaling are already employed in the clinic for the treatment of some autoimmune diseases, infections, and cancers. There has also been renewed interest in IFNs and IFN-related pathways because of their involvement in SARS-CoV-2 infection, and because of the relatively recent emergence of cGAS-STING as a pattern recognition receptor-activated pathway. Whether these advances will ultimately result in improvements in the care of those experiencing heart disease remains to be determined.
Collapse
Affiliation(s)
| | | | - Andrew Advani
- Keenan Research Centre for Biomedical Science and Li Ka Shing Knowledge Institute, St. Michael’s Hospital, Toronto, ON, Canada
| |
Collapse
|
14
|
Fraccarollo D, Geffers R, Galuppo P, Bauersachs J. Mineralocorticoid receptor promotes cardiac macrophage inflammaging. Basic Res Cardiol 2024; 119:243-260. [PMID: 38329499 PMCID: PMC11008080 DOI: 10.1007/s00395-024-01032-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/06/2023] [Revised: 12/21/2023] [Accepted: 01/03/2024] [Indexed: 02/09/2024]
Abstract
Inflammaging, a pro-inflammatory status that characterizes aging and primarily involving macrophages, is a master driver of age-related diseases. Mineralocorticoid receptor (MR) activation in macrophages critically regulates inflammatory and fibrotic processes. However, macrophage-specific mechanisms and the role of the macrophage MR for the regulation of inflammation and fibrotic remodeling in the aging heart have not yet been elucidated. Transcriptome profiling of cardiac macrophages from male/female young (4 months-old), middle (12 months-old) and old (18 and 24 months-old) mice revealed that myeloid cell-restricted MR deficiency prevents macrophage differentiation toward a pro-inflammatory phenotype. Pathway enrichment analysis showed that several biological processes related to inflammation and cell metabolism were modulated by the MR in aged macrophages. Further, transcriptome analysis of aged cardiac fibroblasts revealed that macrophage MR deficiency reduced the activation of pathways related to inflammation and upregulation of ZBTB16, a transcription factor involved in fibrosis. Phenotypic characterization of macrophages showed a progressive replacement of the TIMD4+MHC-IIneg/low macrophage population by TIMD4+MHC-IIint/high and TIMD4-MHC-IIint/high macrophages in the aging heart. By integrating cell sorting and transwell experiments with TIMD4+/TIMD4-macrophages and fibroblasts from old MRflox/MRLysMCre hearts, we showed that the inflammatory crosstalk between TIMD4- macrophages and fibroblasts may imply the macrophage MR and the release of mitochondrial superoxide anions. Macrophage MR deficiency reduced the expansion of the TIMD4- macrophage population and the emergence of fibrotic niches in the aging heart, thereby protecting against cardiac inflammation, fibrosis, and dysfunction. This study highlights the MR as an important mediator of cardiac macrophage inflammaging and age-related fibrotic remodeling.
Collapse
Affiliation(s)
- Daniela Fraccarollo
- Department of Cardiology and Angiology, Hannover Medical School, Carl-Neuberg-Str.1 30625, Hannover, Germany.
| | - Robert Geffers
- Research Group Genome Analytics, Helmholtz Centre for Infection Research, Brunswick, Germany
| | - Paolo Galuppo
- Department of Cardiology and Angiology, Hannover Medical School, Carl-Neuberg-Str.1 30625, Hannover, Germany
| | - Johann Bauersachs
- Department of Cardiology and Angiology, Hannover Medical School, Carl-Neuberg-Str.1 30625, Hannover, Germany.
| |
Collapse
|
15
|
Harding D, Marelli-Berg F. Interferon-gamma signs off an old heart. Cardiovasc Res 2023; 119:2387-2389. [PMID: 37883721 DOI: 10.1093/cvr/cvad151] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/28/2023] Open
Affiliation(s)
- Daniel Harding
- Faculty of Medicine and Dentistry, William Harvey Research Institute, Queen Mary University of London, Charterhouse Square, London EC1M 6BQ, UK
| | - Federica Marelli-Berg
- Faculty of Medicine and Dentistry, William Harvey Research Institute, Queen Mary University of London, Charterhouse Square, London EC1M 6BQ, UK
| |
Collapse
|
16
|
Molinaro C, Scalise M, Leo I, Salerno L, Sabatino J, Salerno N, De Rosa S, Torella D, Cianflone E, Marino F. Polarizing Macrophage Functional Phenotype to Foster Cardiac Regeneration. Int J Mol Sci 2023; 24:10747. [PMID: 37445929 DOI: 10.3390/ijms241310747] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2023] [Revised: 06/22/2023] [Accepted: 06/26/2023] [Indexed: 07/15/2023] Open
Abstract
There is an increasing interest in understanding the connection between the immune and cardiovascular systems, which are highly integrated and communicate through finely regulated cross-talking mechanisms. Recent evidence has demonstrated that the immune system does indeed have a key role in the response to cardiac injury and in cardiac regeneration. Among the immune cells, macrophages appear to have a prominent role in this context, with different subtypes described so far that each have a specific influence on cardiac remodeling and repair. Similarly, there are significant differences in how the innate and adaptive immune systems affect the response to cardiac damage. Understanding all these mechanisms may have relevant clinical implications. Several studies have already demonstrated that stem cell-based therapies support myocardial repair. However, the exact role that cardiac macrophages and their modulation may have in this setting is still unclear. The current need to decipher the dual role of immunity in boosting both heart injury and repair is due, at least for a significant part, to unresolved questions related to the complexity of cardiac macrophage phenotypes. The aim of this review is to provide an overview on the role of the immune system, and of macrophages in particular, in the response to cardiac injury and to outline, through the modulation of the immune response, potential novel therapeutic strategies for cardiac regeneration.
Collapse
Affiliation(s)
- Claudia Molinaro
- Department of Medical and Surgical Sciences, Magna Graecia University, 88100 Catanzaro, Italy
| | - Mariangela Scalise
- Department of Experimental and Clinical Medicine, Magna Graecia University, 88100 Catanzaro, Italy
| | - Isabella Leo
- Department of Experimental and Clinical Medicine, Magna Graecia University, 88100 Catanzaro, Italy
| | - Luca Salerno
- Department of Experimental and Clinical Medicine, Magna Graecia University, 88100 Catanzaro, Italy
| | - Jolanda Sabatino
- Department of Experimental and Clinical Medicine, Magna Graecia University, 88100 Catanzaro, Italy
| | - Nadia Salerno
- Department of Experimental and Clinical Medicine, Magna Graecia University, 88100 Catanzaro, Italy
| | - Salvatore De Rosa
- Department of Medical and Surgical Sciences, Magna Graecia University, 88100 Catanzaro, Italy
| | - Daniele Torella
- Department of Experimental and Clinical Medicine, Magna Graecia University, 88100 Catanzaro, Italy
| | - Eleonora Cianflone
- Department of Medical and Surgical Sciences, Magna Graecia University, 88100 Catanzaro, Italy
| | - Fabiola Marino
- Department of Experimental and Clinical Medicine, Magna Graecia University, 88100 Catanzaro, Italy
| |
Collapse
|