1
|
Ait-Oufella H, Libby P. Inflammation and Atherosclerosis: Prospects for Clinical Trials. Arterioscler Thromb Vasc Biol 2024; 44:1899-1905. [PMID: 39167675 PMCID: PMC11343092 DOI: 10.1161/atvbaha.124.320155] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/23/2024]
Affiliation(s)
- Hafid Ait-Oufella
- Université Paris Cité, INSERM U970, Paris Cardiovascular Research Center, Sorbonne Université, Paris, France
- Medical Intensive Care Unit, Hôpital Saint-Antoine, AP-HP, Sorbonne Université, Paris, France
| | - Peter Libby
- Division of Cardiovascular Medicine, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA
| |
Collapse
|
2
|
Bashore AC, Xue C, Kim E, Yan H, Zhu LY, Pan H, Kissner M, Ross LS, Zhang H, Li M, Reilly MP. Monocyte Single-Cell Multimodal Profiling in Cardiovascular Disease Risk States. Circ Res 2024; 135:685-700. [PMID: 39105287 PMCID: PMC11430373 DOI: 10.1161/circresaha.124.324457] [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: 02/17/2024] [Revised: 07/11/2024] [Accepted: 07/28/2024] [Indexed: 08/07/2024]
Abstract
BACKGROUND Monocytes are a critical innate immune system cell type that serves homeostatic and immunoregulatory functions. They have been identified historically by the cell surface expression of CD14 and CD16. However, recent single-cell studies have revealed that they are much more heterogeneous than previously realized. METHODS We utilized cellular indexing of transcriptomes and epitopes by sequencing (CITE-seq) and single-cell RNA sequencing to describe the comprehensive transcriptional and phenotypic landscape of 437 126 monocytes. RESULTS This high-dimensional multimodal approach identified vast phenotypic diversity and functionally distinct subsets, including IFN-responsive, MHCIIhi (major histocompatibility complex class II), monocyte-platelet aggregates, as well as nonclassical, and several subpopulations of classical monocytes. Using flow cytometry, we validated the existence of MHCII+CD275+ MHCIIhi, CD42b+ monocyte-platelet aggregates, CD16+CD99- nonclassical monocytes, and CD99+ classical monocytes. Each subpopulation exhibited unique characteristics, developmental trajectories, transcriptional regulation, and tissue distribution. In addition, alterations associated with cardiovascular disease risk factors, including race, smoking, and hyperlipidemia were identified. Moreover, the effect of hyperlipidemia was recapitulated in mouse models of elevated cholesterol. CONCLUSIONS This integrative and cross-species comparative analysis provides a new perspective on the comparison of alterations in monocytes in pathological conditions and offers insights into monocyte-driven mechanisms in cardiovascular disease and the potential for monocyte subpopulation targeted therapies.
Collapse
Affiliation(s)
- Alexander C Bashore
- Division of Cardiology, Department of Medicine, Vagelos College of Physicians and Surgeons, Columbia University, New York (A.C.B., C.X., E.K., L.Y.Z., L.S.R., H.Z., M.P.R.)
- Cardiometabolic Genomics Program, Division of Cardiology, Department of Medicine (A.C.B., C.X., E.K., L.Y.Z., L.S.R., H.Z., M.P.R.), Columbia University Irving Medical Center, New York
| | - Chenyi Xue
- Division of Cardiology, Department of Medicine, Vagelos College of Physicians and Surgeons, Columbia University, New York (A.C.B., C.X., E.K., L.Y.Z., L.S.R., H.Z., M.P.R.)
- Cardiometabolic Genomics Program, Division of Cardiology, Department of Medicine (A.C.B., C.X., E.K., L.Y.Z., L.S.R., H.Z., M.P.R.), Columbia University Irving Medical Center, New York
| | - Eunyoung Kim
- Division of Cardiology, Department of Medicine, Vagelos College of Physicians and Surgeons, Columbia University, New York (A.C.B., C.X., E.K., L.Y.Z., L.S.R., H.Z., M.P.R.)
- Cardiometabolic Genomics Program, Division of Cardiology, Department of Medicine (A.C.B., C.X., E.K., L.Y.Z., L.S.R., H.Z., M.P.R.), Columbia University Irving Medical Center, New York
| | - Hanying Yan
- Department of Biostatistics, Epidemiology and Informatics, University of Pennsylvania Perelman School of Medicine, Philadelphia (H.Y., M.L.)
| | - Lucie Y Zhu
- Division of Cardiology, Department of Medicine, Vagelos College of Physicians and Surgeons, Columbia University, New York (A.C.B., C.X., E.K., L.Y.Z., L.S.R., H.Z., M.P.R.)
- Cardiometabolic Genomics Program, Division of Cardiology, Department of Medicine (A.C.B., C.X., E.K., L.Y.Z., L.S.R., H.Z., M.P.R.), Columbia University Irving Medical Center, New York
| | - Huize Pan
- Division of Cardiovascular Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN (H.P.)
| | - Michael Kissner
- Columbia Stem Cell Initiative, Department of Genetics and Development (M.K.), Columbia University Irving Medical Center, New York
| | - Leila S Ross
- Division of Cardiology, Department of Medicine, Vagelos College of Physicians and Surgeons, Columbia University, New York (A.C.B., C.X., E.K., L.Y.Z., L.S.R., H.Z., M.P.R.)
- Cardiometabolic Genomics Program, Division of Cardiology, Department of Medicine (A.C.B., C.X., E.K., L.Y.Z., L.S.R., H.Z., M.P.R.), Columbia University Irving Medical Center, New York
| | - Hanrui Zhang
- Division of Cardiology, Department of Medicine, Vagelos College of Physicians and Surgeons, Columbia University, New York (A.C.B., C.X., E.K., L.Y.Z., L.S.R., H.Z., M.P.R.)
- Cardiometabolic Genomics Program, Division of Cardiology, Department of Medicine (A.C.B., C.X., E.K., L.Y.Z., L.S.R., H.Z., M.P.R.), Columbia University Irving Medical Center, New York
| | - Mingyao Li
- Department of Biostatistics, Epidemiology and Informatics, University of Pennsylvania Perelman School of Medicine, Philadelphia (H.Y., M.L.)
| | - Muredach P Reilly
- Division of Cardiology, Department of Medicine, Vagelos College of Physicians and Surgeons, Columbia University, New York (A.C.B., C.X., E.K., L.Y.Z., L.S.R., H.Z., M.P.R.)
- Cardiometabolic Genomics Program, Division of Cardiology, Department of Medicine (A.C.B., C.X., E.K., L.Y.Z., L.S.R., H.Z., M.P.R.), Columbia University Irving Medical Center, New York
- Irving Institute for Clinical and Translational Research, Columbia University Irving Medical Center, New York (M.P.R.)
| |
Collapse
|
3
|
Ali I, Zhang H, Zaidi SAA, Zhou G. Understanding the intricacies of cellular senescence in atherosclerosis: Mechanisms and therapeutic implications. Ageing Res Rev 2024; 96:102273. [PMID: 38492810 DOI: 10.1016/j.arr.2024.102273] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2023] [Revised: 01/16/2024] [Accepted: 03/13/2024] [Indexed: 03/18/2024]
Abstract
Cardiovascular disease is currently the largest cause of mortality and disability globally, surpassing communicable diseases, and atherosclerosis is the main contributor to this epidemic. Aging is intimately linked to atherosclerosis development and progression, however, the mechanism of aging in atherosclerosis is not well known. To emphasize the significant research on the involvement of senescent cells in atherosclerosis, we begin by outlining compelling evidence that indicates various types of senescent cells and SASP factors linked to atherosclerotic phenotypes. We subsequently provide a comprehensive summary of the existing knowledge, shedding light on the intricate mechanisms through which cellular senescence contributes to the pathogenesis of atherosclerosis. Further, we cover that senescence can be identified by both structural changes and several senescence-associated biomarkers. Finally, we discuss that preventing accelerated cellular senescence represents an important therapeutic potential, as permanent changes may occur in advanced atherosclerosis. Together, the review summarizes the relationship between cellular senescence and atherosclerosis, and inspects the molecular knowledge, and potential clinical significance of senescent cells in developing senescent-based therapy, thus providing crucial insights into their biology and potential therapeutic exploration.
Collapse
Affiliation(s)
- Ilyas Ali
- Department of Medical Cell Biology and Genetics, Guangdong Key Laboratory of Genomic Stability and Disease Prevention, Shenzhen Key Laboratory of Anti-Aging and Regenerative Medicine, and Shenzhen Engineering Laboratory of Regenerative Technologies for Orthopaedic Diseases, Health Sciences Center, Shenzhen University, Shenzhen 518060, PR China; Guangdong Key Laboratory for Biomedical Measurements and Ultrasound Imaging, National-Regional Key Technology Engineering Laboratory for Medical Ultrasound, School of Biomedical Engineering, Shenzhen University Medical School, Shenzhen 518060, PR China
| | - Hongliang Zhang
- Shenzhen University General Hospital, Shenzhen University, Shenzhen 518060, PR China
| | - Syed Aqib Ali Zaidi
- Department of Medical Cell Biology and Genetics, Guangdong Key Laboratory of Genomic Stability and Disease Prevention, Shenzhen Key Laboratory of Anti-Aging and Regenerative Medicine, and Shenzhen Engineering Laboratory of Regenerative Technologies for Orthopaedic Diseases, Health Sciences Center, Shenzhen University, Shenzhen 518060, PR China
| | - Guangqian Zhou
- Department of Medical Cell Biology and Genetics, Guangdong Key Laboratory of Genomic Stability and Disease Prevention, Shenzhen Key Laboratory of Anti-Aging and Regenerative Medicine, and Shenzhen Engineering Laboratory of Regenerative Technologies for Orthopaedic Diseases, Health Sciences Center, Shenzhen University, Shenzhen 518060, PR China; Guangdong Key Laboratory for Biomedical Measurements and Ultrasound Imaging, National-Regional Key Technology Engineering Laboratory for Medical Ultrasound, School of Biomedical Engineering, Shenzhen University Medical School, Shenzhen 518060, PR China.
| |
Collapse
|
4
|
Tang L, Huang ZP, Mei H, Hu Y. Insights gained from single-cell analysis of chimeric antigen receptor T-cell immunotherapy in cancer. Mil Med Res 2023; 10:52. [PMID: 37941075 PMCID: PMC10631149 DOI: 10.1186/s40779-023-00486-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/05/2023] [Accepted: 10/10/2023] [Indexed: 11/10/2023] Open
Abstract
Advances in chimeric antigen receptor (CAR)-T cell therapy have significantly improved clinical outcomes of patients with relapsed or refractory hematologic malignancies. However, progress is still hindered as clinical benefit is only available for a fraction of patients. A lack of understanding of CAR-T cell behaviors in vivo at the single-cell level impedes their more extensive application in clinical practice. Mounting evidence suggests that single-cell sequencing techniques can help perfect the receptor design, guide gene-based T cell modification, and optimize the CAR-T manufacturing conditions, and all of them are essential for long-term immunosurveillance and more favorable clinical outcomes. The information generated by employing these methods also potentially informs our understanding of the numerous complex factors that dictate therapeutic efficacy and toxicities. In this review, we discuss the reasons why CAR-T immunotherapy fails in clinical practice and what this field has learned since the milestone of single-cell sequencing technologies. We further outline recent advances in the application of single-cell analyses in CAR-T immunotherapy. Specifically, we provide an overview of single-cell studies focusing on target antigens, CAR-transgene integration, and preclinical research and clinical applications, and then discuss how it will affect the future of CAR-T cell therapy.
Collapse
Affiliation(s)
- Lu Tang
- Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Hubei Clinical Medical Center of Cell Therapy for Neoplastic Disease, Wuhan, 430022, China
- Key Laboratory of Biological Targeted Therapy, The Ministry of Education, Wuhan, 430022, China
| | - Zhong-Pei Huang
- Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Hubei Clinical Medical Center of Cell Therapy for Neoplastic Disease, Wuhan, 430022, China
- Key Laboratory of Biological Targeted Therapy, The Ministry of Education, Wuhan, 430022, China
| | - Heng Mei
- Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.
- Hubei Clinical Medical Center of Cell Therapy for Neoplastic Disease, Wuhan, 430022, China.
- Key Laboratory of Biological Targeted Therapy, The Ministry of Education, Wuhan, 430022, China.
- Hubei Key Laboratory of Biological Targeted Therapy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.
| | - Yu Hu
- Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.
- Hubei Clinical Medical Center of Cell Therapy for Neoplastic Disease, Wuhan, 430022, China.
- Key Laboratory of Biological Targeted Therapy, The Ministry of Education, Wuhan, 430022, China.
- Hubei Key Laboratory of Biological Targeted Therapy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.
| |
Collapse
|
5
|
Liu F, Wang Y, Yu J. Role of inflammation and immune response in atherosclerosis: Mechanisms, modulations, and therapeutic targets. Hum Immunol 2023; 84:439-449. [PMID: 37353446 DOI: 10.1016/j.humimm.2023.06.002] [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: 04/21/2023] [Revised: 05/19/2023] [Accepted: 06/08/2023] [Indexed: 06/25/2023]
Abstract
Cardiovascular diseases (CVDs) have emerged as the leading cause of mortality globally, with atherosclerosis being a prominent focus of investigation among medical researchers worldwide. Atherosclerosis is characterized as a disease of the large and medium-sized arteries that is multifocal, accumulative, and immunoinflammatory in nature, resulting from the deposition of lipids. Accumulating evidence suggests that inflammatory responses and immunoregulation play a vital role in the occurrence and development of atherosclerosis. While existing treatments for atherosclerosis can assist in symptom management and slowing disease progression, a complete cure remains elusive. Consequently, there is significant interest in research and development of potential new drugs for this condition. Therefore, this review aims to consolidate the current understanding of the pathogenesis of atherosclerosis with an emphasis on inflammation, immune response and infection. Besides, it examines the effects and mechanisms of immunological modulations in atherosclerosis, and the potential therapeutic targets and drugs for intervening in the inflammatory responses and immunoregulation associated with atherosclerosis. Additionally, novel drug options for treating atherosclerosis are explored within the context of this review.
Collapse
Affiliation(s)
- Fang Liu
- Department of Vascular Surgery, Affiliated Hospital of Jiangsu University, Zhenjiang 212001, China; International Genome Center, Jiangsu University, Zhenjiang 212013, China.
| | - Yijun Wang
- International Genome Center, Jiangsu University, Zhenjiang 212013, China
| | - Jiayin Yu
- International Genome Center, Jiangsu University, Zhenjiang 212013, China
| |
Collapse
|
6
|
Su X, Wang L, Ma N, Yang X, Liu C, Yang F, Li J, Yi X, Xing Y. Immune heterogeneity in cardiovascular diseases from a single-cell perspective. Front Cardiovasc Med 2023; 10:1057870. [PMID: 37180791 PMCID: PMC10167030 DOI: 10.3389/fcvm.2023.1057870] [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: 10/03/2022] [Accepted: 04/10/2023] [Indexed: 05/16/2023] Open
Abstract
A variety of immune cell subsets occupy different niches in the cardiovascular system, causing changes in the structure and function of the heart and vascular system, and driving the progress of cardiovascular diseases (CVDs). The immune cells infiltrating the injury site are highly diverse and integrate into a broad dynamic immune network that controls the dynamic changes of CVDs. Due to technical limitations, the effects and molecular mechanisms of these dynamic immune networks on CVDs have not been fully revealed. With recent advances in single-cell technologies such as single-cell RNA sequencing, systematic interrogation of the immune cell subsets is feasible and will provide insights into the way we understand the integrative behavior of immune populations. We no longer lightly ignore the role of individual cells, especially certain highly heterogeneous or rare subpopulations. We summarize the phenotypic diversity of immune cell subsets and their significance in three CVDs of atherosclerosis, myocardial ischemia and heart failure. We believe that such a review could enhance our understanding of how immune heterogeneity drives the progression of CVDs, help to elucidate the regulatory roles of immune cell subsets in disease, and thus guide the development of new immunotherapies.
Collapse
Affiliation(s)
- Xin Su
- China Academy of Chinese Medical Sciences, Guang’anmen Hospital, Beijing, China
| | - Li Wang
- Department of Breast Surgery, Xingtai People’s Hospital, Xingtai, China
| | - Ning Ma
- Department of Breast Surgery, Dezhou Second People’s Hospital, Dezhou, China
| | - Xinyu Yang
- Fangshan Hospital Beijing University of Chinese Medicine, Beijing, China
| | - Can Liu
- China Academy of Chinese Medical Sciences, Guang’anmen Hospital, Beijing, China
| | - Fan Yang
- China Academy of Chinese Medical Sciences, Guang’anmen Hospital, Beijing, China
| | - Jun Li
- China Academy of Chinese Medical Sciences, Guang’anmen Hospital, Beijing, China
| | - Xin Yi
- Department of Cardiology, Beijing Huimin Hospital, Beijing, China
| | - Yanwei Xing
- China Academy of Chinese Medical Sciences, Guang’anmen Hospital, Beijing, China
| |
Collapse
|
7
|
Chiorescu RM, Mocan M, Inceu AI, Buda AP, Blendea D, Vlaicu SI. Vulnerable Atherosclerotic Plaque: Is There a Molecular Signature? Int J Mol Sci 2022; 23:13638. [PMID: 36362423 PMCID: PMC9656166 DOI: 10.3390/ijms232113638] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2022] [Revised: 10/30/2022] [Accepted: 10/31/2022] [Indexed: 08/18/2023] Open
Abstract
Atherosclerosis and its clinical manifestations, coronary and cerebral artery diseases, are the most common cause of death worldwide. The main pathophysiological mechanism for these complications is the rupture of vulnerable atherosclerotic plaques and subsequent thrombosis. Pathological studies of the vulnerable lesions showed that more frequently, plaques rich in lipids and with a high level of inflammation, responsible for mild or moderate stenosis, are more prone to rupture, leading to acute events. Identifying the vulnerable plaques helps to stratify patients at risk of developing acute vascular events. Traditional imaging methods based on plaque appearance and size are not reliable in prediction the risk of rupture. Intravascular imaging is a novel technique able to identify vulnerable lesions, but it is invasive and an operator-dependent technique. This review aims to summarize the current data from literature regarding the main biomarkers involved in the attempt to diagnose vulnerable atherosclerotic lesions. These biomarkers could be the base for risk stratification and development of the new therapeutic drugs in the treatment of patients with vulnerable atherosclerotic plaques.
Collapse
Affiliation(s)
- Roxana Mihaela Chiorescu
- Internal Medicine Department, Iuliu Hatieganu University of Medicine and Pharmacy, 400012 Cluj-Napoca, Romania
- Department of Internal Medicine, Emergency Clinical County Hospital, 400006 Cluj-Napoca, Romania
| | - Mihaela Mocan
- Internal Medicine Department, Iuliu Hatieganu University of Medicine and Pharmacy, 400012 Cluj-Napoca, Romania
- Department of Internal Medicine, Emergency Clinical County Hospital, 400006 Cluj-Napoca, Romania
| | - Andreea Ioana Inceu
- Department of Pharmacology, Toxicology and Clinical Pharmacology, Iuliu Hatieganu University of Medicine, 400349 Cluj-Napoca, Romania
- Department of Cardiology, Nicolae Stăncioiu Heart Institute, 400001 Cluj-Napoca, Romania
| | - Andreea Paula Buda
- Department of Cardiology, Nicolae Stăncioiu Heart Institute, 400001 Cluj-Napoca, Romania
| | - Dan Blendea
- Department of Cardiology, Nicolae Stăncioiu Heart Institute, 400001 Cluj-Napoca, Romania
- Department of Cardiology, Iuliu Hațieganu University of Medicine and Pharmacy, 400437 Cluj-Napoca, Romania
| | - Sonia Irina Vlaicu
- Internal Medicine Department, Iuliu Hatieganu University of Medicine and Pharmacy, 400012 Cluj-Napoca, Romania
- Department of Internal Medicine, Emergency Clinical County Hospital, 400006 Cluj-Napoca, Romania
| |
Collapse
|
8
|
Guillamat-Prats R, Hering D, Derle A, Rami M, Härdtner C, Santovito D, Rinne P, Bindila L, Hristov M, Pagano S, Vuilleumier N, Schmid S, Janjic A, Enard W, Weber C, Maegdefessel L, Faussner A, Hilgendorf I, Steffens S. GPR55 in B cells limits atherosclerosis development and regulates plasma cell maturation. NATURE CARDIOVASCULAR RESEARCH 2022; 1:1056-1071. [PMID: 36523570 PMCID: PMC7613934 DOI: 10.1038/s44161-022-00155-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 01/05/2022] [Accepted: 09/27/2022] [Indexed: 06/17/2023]
Abstract
Dissecting the pathways regulating the adaptive immune response in atherosclerosis is of particular therapeutic interest. Here we report that the lipid G-protein coupled receptor GPR55 is highly expressed by splenic plasma cells (PC), upregulated in mouse spleens during atherogenesis and human unstable or ruptured compared to stable plaques. Gpr55-deficient mice developed larger atherosclerotic plaques with increased necrotic core size compared to their corresponding controls. Lack of GPR55 hyperactivated B cells, disturbed PC maturation and resulted in immunoglobulin (Ig)G overproduction. B cell-specific Gpr55 depletion or adoptive transfer of Gpr55-deficient B cells was sufficient to promote plaque development and elevated IgG titers. In vitro, the endogenous GPR55 ligand lysophsophatidylinositol (LPI) enhanced PC proliferation, whereas GPR55 antagonism blocked PC maturation and increased their mitochondrial content. Collectively, these discoveries provide previously undefined evidence for GPR55 in B cells as a key modulator of the adaptive immune response in atherosclerosis.
Collapse
Affiliation(s)
- Raquel Guillamat-Prats
- Institute for Cardiovascular Prevention (IPEK), Ludwig-Maximilians-Universität (LMU) Munich, Munich, Germany
| | - Daniel Hering
- Institute for Cardiovascular Prevention (IPEK), Ludwig-Maximilians-Universität (LMU) Munich, Munich, Germany
| | - Abhishek Derle
- Institute for Cardiovascular Prevention (IPEK), Ludwig-Maximilians-Universität (LMU) Munich, Munich, Germany
| | - Martina Rami
- Institute for Cardiovascular Prevention (IPEK), Ludwig-Maximilians-Universität (LMU) Munich, Munich, Germany
| | - Carmen Härdtner
- Department of Cardiology and Angiology I, Heart Center and Faculty of Medicine, University of Freiburg. Freiburg, Germany
| | - Donato Santovito
- Institute for Cardiovascular Prevention (IPEK), Ludwig-Maximilians-Universität (LMU) Munich, Munich, Germany
- German Center for Cardiovascular Research (DZHK), Partner Site Munich Heart Alliance (MHA), Munich, Germany
- Institute for Genetic and Biomedical Research (IRGB), Unit of Milan, National Research Council, Milan, Italy
| | - Petteri Rinne
- Institute of Biomedicine, University of Turku, Turku, Finland
| | - Laura Bindila
- Institute of Physiological Chemistry, University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
| | - Michael Hristov
- Institute for Cardiovascular Prevention (IPEK), Ludwig-Maximilians-Universität (LMU) Munich, Munich, Germany
| | - Sabrina Pagano
- Division of Laboratory Medicine, Diagnostic Department, Geneva University Hospitals and Faculty of Medicine
| | - Nicolas Vuilleumier
- Division of Laboratory Medicine, Diagnostic Department, Geneva University Hospitals and Faculty of Medicine
| | - Sofie Schmid
- Department of Vascular and Endovascular Surgery, Klinikum rechts der Isar - Technical University Munich (TUM), Munich, Germany
| | - Aleksandar Janjic
- Anthropology and Human Genomics, Faculty of Biology, Ludwig-Maximilians University, Martinsried, Germany
| | - Wolfgang Enard
- Anthropology and Human Genomics, Faculty of Biology, Ludwig-Maximilians University, Martinsried, Germany
| | - Christian Weber
- Institute for Cardiovascular Prevention (IPEK), Ludwig-Maximilians-Universität (LMU) Munich, Munich, Germany
- German Center for Cardiovascular Research (DZHK), Partner Site Munich Heart Alliance (MHA), Munich, Germany
- Department of Biochemistry, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University Medical Centre, 6229 ER Maastricht, The Netherlands
- Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
| | - Lars Maegdefessel
- German Center for Cardiovascular Research (DZHK), Partner Site Munich Heart Alliance (MHA), Munich, Germany
- Department of Vascular and Endovascular Surgery, Klinikum rechts der Isar - Technical University Munich (TUM), Munich, Germany
| | - Alexander Faussner
- Institute for Cardiovascular Prevention (IPEK), Ludwig-Maximilians-Universität (LMU) Munich, Munich, Germany
| | - Ingo Hilgendorf
- Department of Cardiology and Angiology I, Heart Center and Faculty of Medicine, University of Freiburg. Freiburg, Germany
- Institute for Experimental Cardiovascular Medicine, Heart Center and Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Sabine Steffens
- Institute for Cardiovascular Prevention (IPEK), Ludwig-Maximilians-Universität (LMU) Munich, Munich, Germany
- German Center for Cardiovascular Research (DZHK), Partner Site Munich Heart Alliance (MHA), Munich, Germany
| |
Collapse
|
9
|
Novel Diagnostic Biomarkers Related to Oxidative Stress and Macrophage Ferroptosis in Atherosclerosis. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:8917947. [PMID: 36035208 PMCID: PMC9410850 DOI: 10.1155/2022/8917947] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/01/2022] [Revised: 07/15/2022] [Accepted: 07/20/2022] [Indexed: 12/25/2022]
Abstract
Atherosclerosis (AS) is a chronic inflammatory disease, which has a complex interplay between altered immune metabolism and oxidative stress. Therefore, we aimed to determine the oxidative stress and immune-related biomarkers in AS. Differential gene expression analyses are based on the GSE100927 dataset in the Gene Expression Omnibus (GEO), and 389 oxidative stress (OS) genes are identified based on gene set enrichment analysis (GSEA). We identified 74 differentially expressed genes related to oxidative stress (DEOSGs). “CIBERSORT” and “WGCNA” R Packages were used to compare the differences in immune infiltration levels between AS and control samples. The DEOSGs (N = 74) were intersected with the key module's genes of WGCNA (N = 972), and 27 differentially expressed immune-related oxidative stress genes (DEIOSGs) were obtained. To identify the pivotal genes, a protein-protein interaction (PPI) network was constructed using the STRING database and the Cytoscape software. MMP9, ALOX5, NCF2, NCF, and NCF4 were identified as diagnostic markers of AS, and we validated them in the GSE57691 dataset. The expression levels of the five diagnostic genes were significantly highly expressed in the AS group. Correlation analysis and single-cell analysis revealed that five diagnostic genes were mainly correlated with macrophages M1. We, respectively, intersected differentially expressed genes (DEGs) with ferroptosis gene set, necroptosis gene set, and pyroptosis gene set. The findings suggested that ALOX5 and NCF2 were differentially expressed genes of ferroptosis. High expression of five hub genes in RAW264.7 macrophages were confirmed by PCR. High ALOX5 and NCF2 expression levels in plaque tissues were confirmed by immunohistochemistry (IHC) and western blotting. Our study identified that MMP9, ALOX5, NCF2, NCF1, and NCF4 were diagnostic genes of AS and associated with oxidative stress. ALOX5 and NCF2 may be involved in the formation of the necrotic core in AS by regulating macrophage ferroptosis.
Collapse
|
10
|
Meneri M, Bonato S, Gagliardi D, Comi GP, Corti S. New Insights into Cerebral Vessel Disease Landscapes at Single-Cell Resolution: Pathogenetic and Therapeutic Perspectives. Biomedicines 2022; 10:1693. [PMID: 35884997 PMCID: PMC9313091 DOI: 10.3390/biomedicines10071693] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2022] [Revised: 07/03/2022] [Accepted: 07/11/2022] [Indexed: 11/19/2022] Open
Abstract
Cerebrovascular diseases are a leading cause of death and disability globally. The development of new therapeutic targets for cerebrovascular diseases (e.g., ischemic, and hemorrhagic stroke, vascular dementia) is limited by a lack of knowledge of the cellular and molecular biology of health and disease conditions and the factors that cause injury to cerebrovascular structures. Here, we describe the role of advances in omics technology, particularly RNA sequencing, in studying high-dimensional, multifaceted profiles of thousands of individual blood and vessel cells at single-cell resolution. This analysis enables the dissection of the heterogeneity of diseased cerebral vessels and their atherosclerotic plaques, including the microenvironment, cell evolutionary trajectory, and immune response pathway. In animal models, RNA sequencing permits the tracking of individual cells (including immunological, endothelial, and vascular smooth muscle cells) that compose atherosclerotic plaques and their alteration under experimental settings such as phenotypic transition. We describe how single-cell RNA transcriptomics in humans allows mapping to the molecular and cellular levels of atherosclerotic plaques in cerebral arteries, tracking individual lymphocytes and macrophages, and how these data can aid in identifying novel immune mechanisms that could be exploited as therapeutic targets for cerebrovascular diseases. Single-cell multi-omics approaches will likely provide the unprecedented resolution and depth of data needed to generate clinically relevant cellular and molecular signatures for the precise treatment of cerebrovascular diseases.
Collapse
Affiliation(s)
- Megi Meneri
- Dino Ferrari Centre, Department of Pathophysiology and Transplantation (DEPT), University of Milan, 20122 Milan, Italy; (M.M.); (D.G.); (G.P.C.)
- Neurology Unit, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, 20122 Milan, Italy
| | - Sara Bonato
- Stroke Unit, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, 20122 Milan, Italy;
| | - Delia Gagliardi
- Dino Ferrari Centre, Department of Pathophysiology and Transplantation (DEPT), University of Milan, 20122 Milan, Italy; (M.M.); (D.G.); (G.P.C.)
- Neurology Unit, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, 20122 Milan, Italy
| | - Giacomo P. Comi
- Dino Ferrari Centre, Department of Pathophysiology and Transplantation (DEPT), University of Milan, 20122 Milan, Italy; (M.M.); (D.G.); (G.P.C.)
- Neuromuscular and Rare Diseases Unit, Department of Neuroscience, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, 20122 Milan, Italy
| | - Stefania Corti
- Dino Ferrari Centre, Department of Pathophysiology and Transplantation (DEPT), University of Milan, 20122 Milan, Italy; (M.M.); (D.G.); (G.P.C.)
- Neurology Unit, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, 20122 Milan, Italy
| |
Collapse
|
11
|
Wang J, Chen X. Junctional Adhesion Molecules: Potential Proteins in Atherosclerosis. Front Cardiovasc Med 2022; 9:888818. [PMID: 35872908 PMCID: PMC9302484 DOI: 10.3389/fcvm.2022.888818] [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: 03/03/2022] [Accepted: 06/21/2022] [Indexed: 11/13/2022] Open
Abstract
Junctional adhesion molecules (JAMs) are cell-cell adhesion molecules of the immunoglobulin superfamily and are involved in the regulation of diverse atherosclerosis-related processes such as endothelial barrier maintenance, leucocytes transendothelial migration, and angiogenesis. To combine and further broaden related results, this review concluded the recent progress in the roles of JAMs and predicted future studies of JAMs in the development of atherosclerosis.
Collapse
Affiliation(s)
- Junqi Wang
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha, China
- Hunan Key Laboratory of Pharmacogenetics, Institute of Clinical Pharmacology, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Xiaoping Chen
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha, China
- Hunan Key Laboratory of Pharmacogenetics, Institute of Clinical Pharmacology, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
- *Correspondence: Xiaoping Chen,
| |
Collapse
|
12
|
Howe KL, Cybulsky M, Fish JE. The Endothelium as a Hub for Cellular Communication in Atherogenesis: Is There Directionality to the Message? Front Cardiovasc Med 2022; 9:888390. [PMID: 35498030 PMCID: PMC9051343 DOI: 10.3389/fcvm.2022.888390] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2022] [Accepted: 03/21/2022] [Indexed: 12/11/2022] Open
Abstract
Endothelial cells line every blood vessel and thereby serve as an interface between the blood and the vessel wall. They have critical functions for maintaining homeostasis and orchestrating vascular pathogenesis. Atherosclerosis is a chronic disease where cholesterol and inflammatory cells accumulate in the artery wall below the endothelial layer and ultimately form plaques that can either progress to occlude the lumen or rupture with thromboembolic consequences - common outcomes being myocardial infarction and stroke. Cellular communication lies at the core of this process. In this review, we discuss traditional (e.g., cytokines, chemokines, nitric oxide) and novel (e.g., extracellular vesicles) modes of endothelial communication with other endothelial cells as well as circulating and vessel wall cells, including monocytes, macrophages, neutrophils, vascular smooth muscle cells and other immune cells, in the context of atherosclerosis. More recently, the growing appreciation of endothelial cell plasticity during atherogenesis suggests that communication strategies are not static. Here, emerging data on transcriptomics in cells during the development of atherosclerosis are considered in the context of how this might inform altered cell-cell communication. Given the unique position of the endothelium as a boundary layer that is activated in regions overlying vascular inflammation and atherosclerotic plaque, there is a potential to exploit the unique features of this group of cells to deliver therapeutics that target the cellular crosstalk at the core of atherosclerotic disease. Data are discussed supporting this concept, as well as inherent pitfalls. Finally, we briefly review the literature for other regions of the body (e.g., gut epithelium) where cells similarly exist as a boundary layer but provide discrete messages to each compartment to govern homeostasis and disease. In this light, the potential for endothelial cells to communicate in a directional manner is explored, along with the implications of this concept - from fundamental experimental design to biomarker potential and therapeutic targets.
Collapse
Affiliation(s)
- Kathryn L. Howe
- Toronto General Hospital Research Institute, University Health Network, Toronto, ON, Canada
- Institute of Medical Science, University of Toronto, Toronto, ON, Canada
- Peter Munk Cardiac Centre, University Health Network, Toronto, ON, Canada
- Division of Vascular Surgery, Department of Surgery, University of Toronto, Toronto, ON, Canada
| | - Myron Cybulsky
- Toronto General Hospital Research Institute, University Health Network, Toronto, ON, Canada
- Peter Munk Cardiac Centre, University Health Network, Toronto, ON, Canada
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada
| | - Jason E. Fish
- Toronto General Hospital Research Institute, University Health Network, Toronto, ON, Canada
- Institute of Medical Science, University of Toronto, Toronto, ON, Canada
- Peter Munk Cardiac Centre, University Health Network, Toronto, ON, Canada
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
13
|
Roy P, Orecchioni M, Ley K. How the immune system shapes atherosclerosis: roles of innate and adaptive immunity. Nat Rev Immunol 2022; 22:251-265. [PMID: 34389841 PMCID: PMC10111155 DOI: 10.1038/s41577-021-00584-1] [Citation(s) in RCA: 216] [Impact Index Per Article: 108.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/24/2021] [Indexed: 02/07/2023]
Abstract
Atherosclerosis is the root cause of many cardiovascular diseases. Extensive research in preclinical models and emerging evidence in humans have established the crucial roles of the innate and adaptive immune systems in driving atherosclerosis-associated chronic inflammation in arterial blood vessels. New techniques have highlighted the enormous heterogeneity of leukocyte subsets in the arterial wall that have pro-inflammatory or regulatory roles in atherogenesis. Understanding the homing and activation pathways of these immune cells, their disease-associated dynamics and their regulation by microbial and metabolic factors will be crucial for the development of clinical interventions for atherosclerosis, including potentially vaccination-based therapeutic strategies. Here, we review key molecular mechanisms of immune cell activation implicated in modulating atherogenesis and provide an update on the contributions of innate and adaptive immune cell subsets in atherosclerosis.
Collapse
Affiliation(s)
- Payel Roy
- Division of Inflammation Biology, La Jolla Institute for Immunology, La Jolla, CA, USA
| | - Marco Orecchioni
- Division of Inflammation Biology, La Jolla Institute for Immunology, La Jolla, CA, USA
| | - Klaus Ley
- Division of Inflammation Biology, La Jolla Institute for Immunology, La Jolla, CA, USA.
- Department of Bioengineering, University of California, San Diego, San Diego, CA, USA.
| |
Collapse
|
14
|
Eberhardt N, Giannarelli C. How Single-Cell Technologies Have Provided New Insights Into Atherosclerosis. Arterioscler Thromb Vasc Biol 2022; 42:243-252. [PMID: 35109673 PMCID: PMC8966900 DOI: 10.1161/atvbaha.121.315849] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
The development of innovative single-cell technologies has allowed the high-dimensional transcriptomic and proteomic profiling of individual blood and tissue cells. Recent single-cell studies revealed a new cellular heterogeneity of atherosclerotic plaque tissue and allowed a better understanding of distinct immune functional states in the context of atherosclerosis. In this brief review, we describe how single-cell technologies have shed a new light on the cellular composition of atherosclerotic plaques, and their response to diet perturbations or genetic manipulation in mouse models of atherosclerosis. We discuss how single-cell RNA sequencing, cellular indexing of transcriptomes and epitopes by sequencing, transposase-accessible chromatin with high-throughput sequencing, and cytometry by time-of-flight platforms have empowered the identification of discrete immune, endothelial, and smooth muscle cell alterations in atherosclerosis progression and regression. Finally, we review how single-cell approaches have allowed mapping the cellular and molecular composition of human atherosclerotic plaques and the discovery of new immune alterations in plaques from patients with stroke.
Collapse
Affiliation(s)
- Natalia Eberhardt
- Department of Medicine, Leon H. Charney Division of Cardiology, New York University Grossman School of Medicine, NYU Langone Health, New York (NY), USA.,NYU Cardiovascular Research Center, New York University Grossman School of Medicine, NYU Langone Health, New York (NY), USA
| | - Chiara Giannarelli
- Department of Medicine, Leon H. Charney Division of Cardiology, New York University Grossman School of Medicine, NYU Langone Health, New York (NY), USA.,NYU Cardiovascular Research Center, New York University Grossman School of Medicine, NYU Langone Health, New York (NY), USA.,Department of Pathology, New York University Grossman School of Medicine, NYU Langone Health, New York (NY), USA.,Correspondence to: Chiara Giannarelli, MD, PhD, 435 East 30th street, Science Building, New York, NY, 10016,
| |
Collapse
|
15
|
Gerhardt T, Haghikia A, Stapmanns P, Leistner DM. Immune Mechanisms of Plaque Instability. Front Cardiovasc Med 2022; 8:797046. [PMID: 35087883 PMCID: PMC8787133 DOI: 10.3389/fcvm.2021.797046] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2021] [Accepted: 12/15/2021] [Indexed: 01/08/2023] Open
Abstract
Inflammation crucially drives atherosclerosis from disease initiation to the emergence of clinical complications. Targeting pivotal inflammatory pathways without compromising the host defense could compliment therapy with lipid-lowering agents, anti-hypertensive treatment, and lifestyle interventions to address the substantial residual cardiovascular risk that remains beyond classical risk factor control. Detailed understanding of the intricate immune mechanisms that propel plaque instability and disruption is indispensable for the development of novel therapeutic concepts. In this review, we provide an overview on the role of key immune cells in plaque inception and progression, and discuss recently identified maladaptive immune phenomena that contribute to plaque destabilization, including epigenetically programmed trained immunity in myeloid cells, pathogenic conversion of autoreactive regulatory T-cells and expansion of altered leukocytes due to clonal hematopoiesis. From a more global perspective, the article discusses how systemic crises such as acute mental stress or infection abruptly raise plaque vulnerability and summarizes recent advances in understanding the increased cardiovascular risk associated with COVID-19 disease. Stepping outside the box, we highlight the role of gut dysbiosis in atherosclerosis progression and plaque vulnerability. The emerging differential role of the immune system in plaque rupture and plaque erosion as well as the limitations of animal models in studying plaque disruption are reviewed.
Collapse
Affiliation(s)
- Teresa Gerhardt
- Charité – Universitätsmedizin Berlin, Department of Cardiology, Berlin, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Berlin, Berlin, Germany
- Berlin Institute of Health at Charité – Universitätsmedizin Berlin, Berlin, Germany
| | - Arash Haghikia
- Charité – Universitätsmedizin Berlin, Department of Cardiology, Berlin, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Berlin, Berlin, Germany
- Berlin Institute of Health at Charité – Universitätsmedizin Berlin, Berlin, Germany
| | - Philip Stapmanns
- Charité – Universitätsmedizin Berlin, Department of Cardiology, Berlin, Germany
| | - David Manuel Leistner
- Charité – Universitätsmedizin Berlin, Department of Cardiology, Berlin, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Berlin, Berlin, Germany
- Berlin Institute of Health at Charité – Universitätsmedizin Berlin, Berlin, Germany
- *Correspondence: David Manuel Leistner
| |
Collapse
|
16
|
Bilirubin ameliorates murine atherosclerosis through inhibiting cholesterol synthesis and reshaping the immune system. J Transl Med 2022; 20:1. [PMID: 34980160 PMCID: PMC8722314 DOI: 10.1186/s12967-021-03207-4] [Citation(s) in RCA: 39] [Impact Index Per Article: 19.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2021] [Accepted: 12/17/2021] [Indexed: 12/15/2022] Open
Abstract
Atherosclerosis is a chronic inflammatory disease caused mainly by lipid accumulation and excessive inflammatory immune response. Although the lipid-lowering and cardioprotective properties of bilirubin, as well as the negative relationship between bilirubin and atherosclerosis, were well documented, it is not yet clear whether bilirubin can attenuate atherosclerosis in vivo. In this study, we investigated the role of bilirubin in improving atherosclerosis. We found that mildly elevated bilirubin significantly reduced the risk factors of atherosclerosis, such as plasma glucose, total cholesterol, and low-density lipoprotein cholesterol, and the formation of atherosclerotic plaques, liver total cholesterol, and cholesterol ester concentration in apolipoprotein E-deficient (ApoE-/-) mice fed a western-type (high fat) diet. It was further found that bilirubin could promote the degradation of 3-Hydroxy-3-Methylglutaryl-CoA Reductase (HMGCR), a rate-limiting enzyme for endogenous cholesterol synthesis. Using mass cytometry-based high dimensional single cell analysis, we observed a decrease of natural killer cells and an increase of dendritic cells and myeloid-derived suppressor cells, which all are closely associated with atherosclerosis risk factors and contribute to the improvement of atherosclerosis, in ApoE-/- mice treated with bilirubin. By in-depth analysis, modulation of multiple spleen or peripheral blood T cell clusters exhibiting either positive or negative correlations with total cholesterol or low-density lipoprotein cholesterol was detected after bilirubin treatment. In this study, we demonstrate that bilirubin serves as a negative regulator of atherosclerosis and reduces atherosclerosis by inhibiting cholesterol synthesis and modulating the immune system.
Collapse
|
17
|
Sawada H, Daugherty A, Lu HS. From unbiased transcriptomics to understanding the molecular basis of atherosclerosis. Curr Opin Lipidol 2021; 32:328-329. [PMID: 34472541 PMCID: PMC8423091 DOI: 10.1097/mol.0000000000000773] [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: 11/26/2022]
Affiliation(s)
- Hisashi Sawada
- Saha Cardiovascular Research Center, Saha Aortic Center, Department of Physiology, University of Kentucky, Lexington, Kentucky, USA
| | | | | |
Collapse
|
18
|
Vallejo J, Cochain C, Zernecke A, Ley K. Heterogeneity of immune cells in human atherosclerosis revealed by scRNA-Seq. Cardiovasc Res 2021; 117:2537-2543. [PMID: 34343272 DOI: 10.1093/cvr/cvab260] [Citation(s) in RCA: 49] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/04/2021] [Revised: 07/02/2021] [Accepted: 07/30/2021] [Indexed: 12/14/2022] Open
Abstract
Immune cells in atherosclerosis include T, B, natural killer (NK) and NKT cells, macrophages, monocytes, dendritic cells (DCs), neutrophils and mast cells. Advances in single cell RNA sequencing (sRNA-Seq) have refined our understanding of immune cell subsets. Four recent studies have used scRNA-Seq of immune cells in human atherosclerotic lesions and peripheral blood mononuclear cells (PBMCs), some including cell surface phenotypes revealed by oligonucleotide-tagged antibodies, which confirmed known and identified new immune cell subsets and identified genes significantly upregulated in PBMCs from HIV+ subjects with atherosclerosis compared to PBMCs from matched HIV+ subjects without atherosclerosis. The ability of scRNA-Seq to identify cell types is greatly augmented by adding cell surface phenotype using antibody sequencing. In this review we summarize the latest data obtained by scRNA-Seq on plaques and human PBMCs in human subjects with atherosclerosis.
Collapse
Affiliation(s)
- Jenifer Vallejo
- Division of Inflammation Biology, La Jolla Institute for Immunology, California, USA
| | - Clément Cochain
- Institute of Experimental Biomedicine, University Hospital Würzburg, Germany.,Comprehensive Heart Failure Center, University Hospital Würzburg, Germany
| | - Alma Zernecke
- Institute of Experimental Biomedicine, University Hospital Würzburg, Germany
| | - Klaus Ley
- Division of Inflammation Biology, La Jolla Institute for Immunology, California, USA.,Department of Bioengineering, University of California San Diego, California, USA
| |
Collapse
|
19
|
Giró O, Jiménez A, Pané A, Badimon L, Ortega E, Chiva-Blanch G. Extracellular vesicles in atherothrombosis and cardiovascular disease: Friends and foes. Atherosclerosis 2021; 330:61-75. [PMID: 34256307 DOI: 10.1016/j.atherosclerosis.2021.07.002] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/28/2021] [Revised: 06/21/2021] [Accepted: 07/02/2021] [Indexed: 12/16/2022]
Abstract
Extracellular vesicles (EV, exosomes and microvesicles -MV-) are 30-1000 nm particles surrounded by a phospholipid bilayer membrane that are released from almost all cell types through several pathways. EV encapsulate bioactive molecules, and the molecular cargo is determined by the trigger stimulating its release, reflecting its cell origin and biological functions. This review is primarily focused on the latest evidence of the roles of EV, released from cells involved in the different stages of atherothrombosis. The potential translation of this information to the clinical arena is also discussed. EV can have both pro- and anti-atherothrombotic effects depending on several factors, such as the type of vesicle (MV/exosome), its molecular cargo, its cell of origin, and the context in which are generated, i.e., the stimulus triggering its release. In fact, EV actively participate in every step of atherosclerosis onset and progression, and also in thrombus formation leading to a major adverse cardiovascular event. Moreover, EV have a determinant role in fibrous cap stability, thus determining the propensity of the plaque to rupture. On the other hand, and again, conditioned by the context and stimulus instigating its secretion, some EV may have protective biological functions, perhaps as a compensatory mechanism or even with reparative or regenerative potential. Therefore, the study of the implication of EV in atherothrombosis might be of relevance to unveil new therapeutic targets, vectors and biomarkers of cardiovascular disease (CVD).
Collapse
Affiliation(s)
- Oriol Giró
- Department of Endocrinology and Nutrition, August Pi i Sunyer Biomedical Research Institute - IDIBAPS, Hospital Clínic of Barcelona, Barcelona, Spain
| | - Amanda Jiménez
- Department of Endocrinology and Nutrition, August Pi i Sunyer Biomedical Research Institute - IDIBAPS, Hospital Clínic of Barcelona, Barcelona, Spain; Centro de Investigación Biomédica en Red Fisiopatología de la Obesidad y la Nutrición (CIBEROBN), Instituto de Salud Carlos III (ISCIII), Madrid, Spain
| | - Adriana Pané
- Department of Endocrinology and Nutrition, August Pi i Sunyer Biomedical Research Institute - IDIBAPS, Hospital Clínic of Barcelona, Barcelona, Spain
| | - Lina Badimon
- Cardiovascular Program ICCC; Institut de Recerca Hospital Santa Creu i Sant Pau-IIB Sant Pau, Barcelona, Spain; Centro de Investigación Biomédica en Red Cardiovascular (CIBERCV), Instituto de Salud Carlos III (ISCIII), Madrid, Spain
| | - Emilio Ortega
- Department of Endocrinology and Nutrition, August Pi i Sunyer Biomedical Research Institute - IDIBAPS, Hospital Clínic of Barcelona, Barcelona, Spain; Centro de Investigación Biomédica en Red Fisiopatología de la Obesidad y la Nutrición (CIBEROBN), Instituto de Salud Carlos III (ISCIII), Madrid, Spain
| | - Gemma Chiva-Blanch
- Department of Endocrinology and Nutrition, August Pi i Sunyer Biomedical Research Institute - IDIBAPS, Hospital Clínic of Barcelona, Barcelona, Spain; Centro de Investigación Biomédica en Red Fisiopatología de la Obesidad y la Nutrición (CIBEROBN), Instituto de Salud Carlos III (ISCIII), Madrid, Spain.
| |
Collapse
|