1
views
0
recommends
+1 Recommend
0 collections
    0
    shares
      • Record: found
      • Abstract: found
      • Article: found
      Is Open Access

      Role of vascular smooth muscle cell clonality in atherosclerosis

      review-article

      Read this article at

      Bookmark
          There is no author summary for this article yet. Authors can add summaries to their articles on ScienceOpen to make them more accessible to a non-specialist audience.

          Abstract

          Atherosclerotic cardiovascular disease remains the leading cause of death worldwide. While many cell types contribute to the growing atherosclerotic plaque, the vascular smooth muscle cell (SMC) is a major contributor due in part to its remarkable plasticity and ability to undergo phenotype switching in response to injury. SMCs can migrate into the fibrous cap, presumably stabilizing the plaque, or accumulate within the lesional core, possibly accelerating vascular inflammation. How SMCs expand and react to disease stimuli has been a controversial topic for many decades. While early studies relying on X-chromosome inactivation were inconclusive due to low resolution and sensitivity, recent advances in multi-color lineage tracing models have revitalized the concept that SMCs likely expand in an oligoclonal fashion during atherogenesis. Current efforts are focused on determining whether all SMCs have equal capacity for clonal expansion or if a “stem-like” progenitor cell may exist, and to understand how constituents of the clone decide which phenotype they will ultimately adopt as the disease progresses. Mechanistic studies are also beginning to dissect the processes which confer cells with their overall survival advantage, test whether these properties are attributable to intrinsic features of the expanding clone, and define the role of cross-talk between proliferating SMCs and other plaque constituents such as neighboring macrophages. In this review, we aim to summarize the historical perspectives on SMC clonality, highlight unanswered questions, and identify translational issues which may need to be considered as therapeutics directed against SMC clonality are developed as a novel approach to targeting atherosclerosis.

          Related collections

          Most cited references88

          • Record: found
          • Abstract: found
          • Article: not found

          The clonal evolution of tumor cell populations.

          P C Nowell (1976)
          It is proposed that most neoplasms arise from a single cell of origin, and tumor progression results from acquired genetic variability within the original clone allowing sequential selection of more aggressive sublines. Tumor cell populations are apparently more genetically unstable than normal cells, perhaps from activation of specific gene loci in the neoplasm, continued presence of carcinogen, or even nutritional deficiencies within the tumor. The acquired genetic insta0ility and associated selection process, most readily recognized cytogenetically, results in advanced human malignancies being highly individual karyotypically and biologically. Hence, each patient's cancer may require individual specific therapy, and even this may be thwarted by emergence of a genetically variant subline resistant to the treatment. More research should be directed toward understanding and controlling the evolutionary process in tumors before it reaches the late stage usually seen in clinical cancer.
            Bookmark
            • Record: found
            • Abstract: found
            • Article: found

            Clonal Heterogeneity and Tumor Evolution: Past, Present, and the Future

            Intratumor heterogeneity, which fosters tumor evolution, is a key challenge in cancer medicine. Here, we review data and technologies that have revealed intra-tumor heterogeneity across cancer types and the dynamics, constraints, and contingencies inherent to tumor evolution. We emphasize the importance of macro-evolutionary leaps, often involving large-scale chromosomal alterations, in driving tumor evolution and metastasis and consider the role of the tumor microenvironment in engendering heterogeneity and drug resistance. We suggest that bold approaches to drug development, harnessing the adaptive properties of the immune-microenvironment while limiting those of the tumor, combined with advances in clinical trial-design, will improve patient outcome.
              Bookmark
              • Record: found
              • Abstract: found
              • Article: not found

              Clonal Hematopoiesis and Risk of Atherosclerotic Cardiovascular Disease

              Background Clonal hematopoiesis of indeterminate potential (CHIP), defined by the presence of an expanded somatic blood cell clone in those without other hematologic abnormalities, is common in older individuals and associates with an increased risk of developing hematologic cancer. We previously found preliminary evidence for an association of CHIP with human atherosclerotic cardiovascular disease, but the nature of this association was unclear. Methods We used whole exome sequencing to detect the presence of CHIP in peripheral blood cells and associated this with coronary heart disease in four case-control studies together comprising 4,794 cases and 3,537 controls. To assess causality, we perturbed the function of Tet2, the second most commonly mutated gene linked to clonal hematopoiesis, in the hematopoietic cells of atherosclerosis-prone mice. Results In nested case-control analyses from two prospective cohorts, carriers of CHIP had a 1.9-fold (95% confidence interval 1.4–2.7) increased risk of coronary heart disease compared to non-carriers. In two retrospective case-control cohorts for early-onset myocardial infarction, those with CHIP had a 4.0-fold greater risk (95% confidence interval 2.4–6.7) of having myocardial infarction. Mutations in DNMT3A, TET2, ASXL1, and JAK2 were each individually associated with coronary heart disease. Those with clonal hematopoiesis also had increased coronary artery calcification, a marker of coronary atherosclerosis burden. Hyperlipidemic mice engrafted with Tet2−/− or Tet2+/− bone marrow developed larger atherosclerotic lesions in the aortic root and aorta than mice receiving control marrow. Analyses of Tet2−/− macrophages demonstrated elevated expression of several chemokine and cytokine genes that contribute to atherosclerosis. Conclusions Clonal hematopoiesis robustly associates with coronary heart disease in humans and causes accelerated atherosclerosis in mice.
                Bookmark

                Author and article information

                Contributors
                Journal
                Front Cardiovasc Med
                Front Cardiovasc Med
                Front. Cardiovasc. Med.
                Frontiers in Cardiovascular Medicine
                Frontiers Media S.A.
                2297-055X
                28 November 2023
                2023
                : 10
                : 1273596
                Affiliations
                [ 1 ]Division of Vascular Surgery, Department of Surgery, Stanford University School of Medicine , Stanford, CA, United States
                [ 2 ]Stanford Cardiovascular Institute , Stanford, CA, United States
                [ 3 ]Division of Cardiovascular Medicine, Department of Medicine, Stanford University School of Medicine , Stanford, CA, United States
                [ 4 ]Department of Pathology and Laboratory Medicine, University of British Columbia , Vancouver, BC, Canada
                Author notes

                Edited by: Daniel Greif, Yale University, United States

                Reviewed by: Julian Albarran Juarez, Aarhus University, Denmark Nhat Tu Le, Houston Methodist Research Institute, United States

                [* ] Correspondence: Nicholas J. Leeper nleeper@ 123456stanford.edu
                Article
                10.3389/fcvm.2023.1273596
                10713728
                38089777
                c2379b9a-364e-44c8-9534-a72778eb82ab
                © 2023 Luo, Fu, Bell, Wang and Leeper.

                This is an open-access article distributed under the terms of the Creative Commons Attribution License (CC BY). The use, distribution or reproduction in other forums is permitted, provided the original author(s) and the copyright owner(s) are credited and that the original publication in this journal is cited, in accordance with accepted academic practice. No use, distribution or reproduction is permitted which does not comply with these terms.

                History
                : 06 August 2023
                : 24 October 2023
                Page count
                Figures: 2, Tables: 1, Equations: 0, References: 88, Pages: 0, Words: 0
                Funding
                Funded by: AHA
                Award ID: 23POST1012196
                Funded by: Damon Runyon Cancer Research Foundation
                Award ID: PST 33-21
                Funded by: Heart and Stroke Foundation of Canada New Investigator Award
                Award ID:  
                Funded by: NIH
                Award ID: R35HL144475
                Funded by: AHA
                Award ID: EIA34770065
                Funded by: Leducq Foundation PlaqOmics consortia
                Award ID: 18CVD02
                The author(s) declare financial support was received for the research, authorship, and/or publication of this article.
                LL is supported by AHA 23POST1012196. CB is supported by the Damon Runyon Cancer Research Foundation PST 33-21. YW is supported by Heart and Stroke Foundation of Canada New Investigator Award. NL is supported by NIH R35HL144475, AHA EIA34770065, and Leducq Foundation PlaqOmics consortia (18CVD02).
                Categories
                Cardiovascular Medicine
                Review
                Custom metadata
                Atherosclerosis and Vascular Medicine

                vascular smooth muscle cell,atherosclerosis,clonality,phenotype plasticity,cardiovascular disease

                Comments

                Comment on this article