24
views
0
recommends
+1 Recommend
0 collections
    0
    shares
      • Record: found
      • Abstract: found
      • Article: found
      Is Open Access

      Single-cell expression profiling reveals dynamic flux of cardiac stromal, vascular and immune cells in health and injury

      research-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

          Besides cardiomyocytes (CM), the heart contains numerous interstitial cell types which play key roles in heart repair, regeneration and disease, including fibroblast, vascular and immune cells. However, a comprehensive understanding of this interactive cell community is lacking. We performed single-cell RNA-sequencing of the total non-CM fraction and enriched ( Pdgfra-GFP +) fibroblast lineage cells from murine hearts at days 3 and 7 post-sham or myocardial infarction (MI) surgery. Clustering of >30,000 single cells identified >30 populations representing nine cell lineages, including a previously undescribed fibroblast lineage trajectory present in both sham and MI hearts leading to a uniquely activated cell state defined in part by a strong anti-WNT transcriptome signature. We also uncovered novel myofibroblast subtypes expressing either pro-fibrotic or anti-fibrotic signatures. Our data highlight non-linear dynamics in myeloid and fibroblast lineages after cardiac injury, and provide an entry point for deeper analysis of cardiac homeostasis, inflammation, fibrosis, repair and regeneration.

          eLife digest

          In our bodies, heart attacks lead to cell death and inflammation. This is then followed by a healing phase where the organ repairs itself. There are many types of heart cells, from muscle and pacemaker cells that help to create the beating motion, to so-called fibroblasts that act as a supporting network. Yet, it is still unclear how individual cells participate in the heart's response to injury.

          All cells possess the same genetic information, but they turn on or off different genes depending on the specific tasks that they need to perform. Spotting which genes are activated in individual cells can therefore provide clues about their exact roles in the body. Until recently, technological limitations meant that this information was difficult to access, because it was only possible to capture the global response of a group of cells in a sample.

          A new method called single-cell RNA sequencing is now allowing researchers to study the activities of many genes in thousands of individual cells at the same time. Here, Farbehi, Patrick et al. performed single-cell RNA sequencing on over 30,000 individual cells from healthy and injured mouse hearts. Computational approaches were then used to cluster cells into groups according to the activities of their genes.

          The experiments identified over 30 distinct sub-types of cell, including several that were previously unknown. For example, a group of fibroblasts that express a gene called Wif1 was discovered. Previous genetic studies have shown that Wif1 is essential for the heart's response to injury. Further experiments by Farbehi, Patrick et al. indicated that this new sub-type of cells may control the timing of the different aspects of heart repair after damage.

          Tens of millions of people around the world suffer from heart attacks and other heart diseases. Knowing how different types of heart cells participate in repair mechanisms may help to find new targets for drugs and other treatments.

          Related collections

          Most cited references50

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

          Cardiac Fibrosis: The Fibroblast Awakens.

          Myocardial fibrosis is a significant global health problem associated with nearly all forms of heart disease. Cardiac fibroblasts comprise an essential cell type in the heart that is responsible for the homeostasis of the extracellular matrix; however, upon injury, these cells transform to a myofibroblast phenotype and contribute to cardiac fibrosis. This remodeling involves pathological changes that include chamber dilation, cardiomyocyte hypertrophy and apoptosis, and ultimately leads to the progression to heart failure. Despite the critical importance of fibrosis in cardiovascular disease, our limited understanding of the cardiac fibroblast impedes the development of potential therapies that effectively target this cell type and its pathological contribution to disease progression. This review summarizes current knowledge regarding the origins and roles of fibroblasts, mediators and signaling pathways known to influence fibroblast function after myocardial injury, as well as novel therapeutic strategies under investigation to attenuate cardiac fibrosis.
            Bookmark
            • Record: found
            • Abstract: found
            • Article: not found

            Macrophages are required for neonatal heart regeneration.

            Myocardial infarction (MI) leads to cardiomyocyte death, which triggers an immune response that clears debris and restores tissue integrity. In the adult heart, the immune system facilitates scar formation, which repairs the damaged myocardium but compromises cardiac function. In neonatal mice, the heart can regenerate fully without scarring following MI; however, this regenerative capacity is lost by P7. The signals that govern neonatal heart regeneration are unknown. By comparing the immune response to MI in mice at P1 and P14, we identified differences in the magnitude and kinetics of monocyte and macrophage responses to injury. Using a cell-depletion model, we determined that heart regeneration and neoangiogenesis following MI depends on neonatal macrophages. Neonates depleted of macrophages were unable to regenerate myocardia and formed fibrotic scars, resulting in reduced cardiac function and angiogenesis. Immunophenotyping and gene expression profiling of cardiac macrophages from regenerating and nonregenerating hearts indicated that regenerative macrophages have a unique polarization phenotype and secrete numerous soluble factors that may facilitate the formation of new myocardium. Our findings suggest that macrophages provide necessary signals to drive angiogenesis and regeneration of the neonatal mouse heart. Modulating inflammation may provide a key therapeutic strategy to support heart regeneration.
              Bookmark
              • Record: found
              • Abstract: found
              • Article: not found

              Self-renewing resident cardiac macrophages limit adverse remodeling following myocardial infarction

              Macrophages promote both injury and repair following myocardial infarction, but discriminating functions within mixed populations remains challenging. Here we used fate mapping and single-cell transcriptomics to demonstrate that at steady state, TIMD4+LYVE1+MHC-IIloCCR2− resident cardiac macrophages self-renew with negligible blood monocyte input. Monocytes partially replaced resident TIMD4−LYVE1−MHC-IIhiCCR2− macrophages and fully replaced TIMD4−LYVE1−MHC-IIhiCCR2+ macrophages, revealing a hierarchy of monocyte contribution to functionally distinct macrophage subsets. Ischemic injury reduced TIMD4+ and TIMD4− resident macrophage abundance within infarcted tissue while recruited, CCR2+ monocyte-derived macrophages adopted multiple cell fates, including those nearly indistinguishable from resident macrophages. Despite this similarity, inducible depletion of resident macrophages using a Cx3cr1-based system led to impaired cardiac function and promoted adverse remodeling primarily within the peri-infarct zone, highlighting a non-redundant, cardioprotective role of resident cardiac macrophages. Lastly, we demonstrate the ability of TIMD4 to be used as a durable lineage marker of a subset of resident cardiac macrophages.
                Bookmark

                Author and article information

                Contributors
                Role: Reviewing Editor
                Role: Senior Editor
                Journal
                eLife
                Elife
                eLife
                eLife
                eLife Sciences Publications, Ltd
                2050-084X
                26 March 2019
                2019
                : 8
                : e43882
                Affiliations
                [1 ]Victor Chang Cardiac Research Institute DarlinghurstAustralia
                [2 ]deptStem Cells Australia, Melbourne Brain Centre University of Melbourne VictoriaAustralia
                [3 ]deptGarvan Weizmann Centre for Cellular Genomics Garvan Institute of Medical Research SydneyAustralia
                [4 ]deptGraduate School of Biomedical Engineering UNSW Sydney KensingtonAustralia
                [5 ]deptSt. Vincent's Clinical School UNSW Sydney KensingtonAustralia
                [6 ]deptSchool of Dentistry, Faculty of Medicine and Health University of Sydney, Westmead Hospital WestmeadAustralia
                [7 ]deptSchool of Biotechnology and Biomolecular Science UNSW Sydney KensingtonAustralia
                University of Pennsylvania United States
                Howard Hughes Medical Institute and Institute of Genetic Medicine, Johns Hopkins University School of Medicine United States
                University of Pennsylvania United States
                Author notes
                [†]

                These authors contributed equally to this work.

                Author information
                http://orcid.org/0000-0003-0956-1026
                http://orcid.org/0000-0002-9950-9792
                Article
                43882
                10.7554/eLife.43882
                6459677
                30912746
                6794ec2b-afc5-465c-b736-a01caf6061ef
                © 2019, Farbehi et al

                This article is distributed under the terms of the Creative Commons Attribution License, which permits unrestricted use and redistribution provided that the original author and source are credited.

                History
                : 26 November 2018
                : 25 March 2019
                Funding
                Funded by: FundRef http://dx.doi.org/10.13039/501100001773, University of New South Wales;
                Award Recipient :
                Funded by: FundRef http://dx.doi.org/10.13039/501100001030, National Heart Foundation of Australia;
                Award ID: 100848
                Award Recipient :
                Funded by: FundRef http://dx.doi.org/10.13039/501100000925, National Health and Medical Research Council;
                Award ID: 1105271
                Award Recipient :
                Funded by: FundRef http://dx.doi.org/10.13039/501100000970, Stem Cells Australia;
                Award ID: SR110001002
                Award Recipient :
                Funded by: FundRef http://dx.doi.org/10.13039/501100000925, National Health and Medical Research Council;
                Award ID: 1074386
                Award Recipient :
                Funded by: FundRef http://dx.doi.org/10.13039/501100001674, Fondation Leducq;
                Award ID: 15CVD03
                Award Recipient :
                Funded by: St. Vincent's Clinic Foundation;
                Award ID: 100711
                Award Recipient :
                Funded by: FundRef http://dx.doi.org/10.13039/501100001674, Fondation Leducq;
                Award ID: 13CVD01
                Award Recipient :
                Funded by: FundRef http://dx.doi.org/10.13039/501100000925, National Health and Medical Research Council;
                Award ID: 573707
                Award Recipient :
                Funded by: FundRef http://dx.doi.org/10.13039/501100000925, National Health and Medical Research Council;
                Award ID: 1118576
                Award Recipient :
                Funded by: New South Wales Cardiovascular Research Network;
                Award Recipient :
                The funders had no role in study design, data collection and interpretation, or the decision to submit the work for publication.
                Categories
                Research Article
                Cell Biology
                Computational and Systems Biology
                Custom metadata
                Comprehensive scRNA-seq analysis of cardiac stromal cells in healthy and injured hearts reveals novel cell types and non-linear cell dynamics, providing new insights into cardiac inflammation, fibrosis and repair.

                Life sciences
                heart,myocardial infarction,scrna-seq,mouse
                Life sciences
                heart, myocardial infarction, scrna-seq, mouse

                Comments

                Comment on this article