1
|
Bahr J, Poschmann G, Jungmann A, Busch M, Ding Z, Vogt J, Zalfen R, Steinhausen J, Euan Martínez AA, Wachtmeister T, Rickert D, Lautwein T, Alter C, Amrute JM, Lavine KJ, Köhrer K, Levkau B, Most P, Stühler K, Hesse J, Schrader J. A secretome atlas of cardiac fibroblasts from healthy and infarcted mouse hearts. Commun Biol 2025; 8:675. [PMID: 40301568 PMCID: PMC12041564 DOI: 10.1038/s42003-025-08083-y] [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: 07/31/2024] [Accepted: 04/12/2025] [Indexed: 05/01/2025] Open
Abstract
Cardiac fibroblasts (CF) are key players after myocardial infarction (MI), but their signaling is only incompletely understood. Here we report a first secretome atlas of CF in control (cCF) and post-MI mouse hearts (miCF), combining a rapid cell isolation technique with SILAC and click chemistry. In CF, numerous paracrine factors involved in immune homeostasis are identified. Comparing secretome, transcriptome (SLAMseq), and cellular proteome disclose protein turnover. In miCF at day 5 post-MI, significantly upregulated proteins include SLIT2, FN1, and CRLF1 in mouse and human samples. Comparing the miCF secretome at days 3 and 5 post-MI reveals the dynamic nature of protein secretion. Specific in-vivo labeling of miCF proteins via biotin ligase TurboID using the POSTN promotor mirrors the in-vitro data. In summary, we identify numerous paracrine factors specifically secreted from CF in mice and humans. This secretome atlas may lead to new biomarkers and/or therapeutic targets for the activated CF.
Collapse
Affiliation(s)
- Jasmin Bahr
- Department of Molecular Cardiology, Medical Faculty and University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Gereon Poschmann
- Institute for Molecular Medicine, Proteome Research, Medical Faculty and University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Andreas Jungmann
- Division of Molecular and Translational Cardiology, Department of Internal Medicine III, Heidelberg University Hospital, Heidelberg, Germany
| | - Martin Busch
- Division of Molecular and Translational Cardiology, Department of Internal Medicine III, Heidelberg University Hospital, Heidelberg, Germany
| | - Zhaoping Ding
- Department of Molecular Cardiology, Medical Faculty and University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Jens Vogt
- Institute of Molecular Medicine III, Medical Faculty and University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Ria Zalfen
- Department of Molecular Cardiology, Medical Faculty and University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Julia Steinhausen
- Department of Molecular Cardiology, Medical Faculty and University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Arlen Aurora Euan Martínez
- Department of Molecular Cardiology, Medical Faculty and University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Thorsten Wachtmeister
- Genomics & Transcriptomics Laboratory, Biological and Medical Research Centre (BMFZ), Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Daniel Rickert
- Genomics & Transcriptomics Laboratory, Biological and Medical Research Centre (BMFZ), Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Tobias Lautwein
- Genomics & Transcriptomics Laboratory, Biological and Medical Research Centre (BMFZ), Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Christina Alter
- Department of Molecular Cardiology, Medical Faculty and University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Junedh M Amrute
- Center for Cardiovascular Research, Department of Medicine, Cardiovascular Division, Washington University School of Medicine, St. Louis, MO, USA
| | - Kory J Lavine
- Center for Cardiovascular Research, Department of Medicine, Cardiovascular Division, Washington University School of Medicine, St. Louis, MO, USA
| | - Karl Köhrer
- Genomics & Transcriptomics Laboratory, Biological and Medical Research Centre (BMFZ), Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Bodo Levkau
- Institute of Molecular Medicine III, Medical Faculty and University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
- CARID, Cardiovascular Research Institute Düsseldorf, Medical Faculty and University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Patrick Most
- Division of Molecular and Translational Cardiology, Department of Internal Medicine III, Heidelberg University Hospital, Heidelberg, Germany
| | - Kai Stühler
- Institute for Molecular Medicine, Proteome Research, Medical Faculty and University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
- Molecular Proteomics Laboratory, Biological and Medical Research Centre (BMFZ), Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Julia Hesse
- Department of Molecular Cardiology, Medical Faculty and University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, Düsseldorf, Germany.
- CARID, Cardiovascular Research Institute Düsseldorf, Medical Faculty and University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, Düsseldorf, Germany.
| | - Jürgen Schrader
- Department of Molecular Cardiology, Medical Faculty and University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, Düsseldorf, Germany.
- CARID, Cardiovascular Research Institute Düsseldorf, Medical Faculty and University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, Düsseldorf, Germany.
| |
Collapse
|
2
|
Li G, Ni C, Wang J, Zhang F, Fu Z, Wang L, Wang B, Liu Y, Zhao J, Li M, Lin H, Liao F, Ye S, Zhang Y, Cai J, Shi S, Zhong Z, Shi Y, He J, Xiong X, Xu Y, Chen J, Zhu W, Wang Y, Wang J, Hu X. Dynamic molecular atlas of cardiac fibrosis at single-cell resolution shows CD248 in cardiac fibroblasts orchestrates interactions with immune cells. NATURE CARDIOVASCULAR RESEARCH 2025; 4:380-396. [PMID: 40148545 DOI: 10.1038/s44161-025-00617-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/04/2024] [Accepted: 01/30/2025] [Indexed: 03/29/2025]
Abstract
Post-injury remodeling is a complex process involving temporal specific cellular interactions in the injured tissue where the resident fibroblasts play multiple roles. Here, we performed single-cell and spatial transcriptome analysis in human and mouse infarcted hearts to dissect the molecular basis of these interactions. We identified a unique fibroblast subset with high CD248 expression, strongly associated with extracellular matrix remodeling. Genetic Cd248 deletion in fibroblasts mitigated cardiac fibrosis and dysfunction following ischemia/reperfusion. Mechanistically, CD248 stabilizes type I transforming growth factor beta receptor and thus upregulates fibroblast ACKR3 expression, leading to enhanced T cell retention. This CD248-mediated fibroblast-T cell interaction is required to sustain fibroblast activation and scar expansion. Disrupting this interaction using monoclonal antibody or chimeric antigen receptor T cell reduces T cell infiltration and consequently ameliorates cardiac fibrosis and dysfunction. Our findings reveal a CD248+ fibroblast subpopulation as a key regulator of immune-fibroblast cross-talk and a potential therapy to treat tissue fibrosis.
Collapse
Affiliation(s)
- Guohua Li
- Department of Cardiology, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
- State Key Laboratory of Transvascular Implantation Devices, Hangzhou, China
- Heart Regeneration and Repair Key Laboratory of Zhejiang Province, Hangzhou, China
| | - Cheng Ni
- Department of Cardiology, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
- State Key Laboratory of Transvascular Implantation Devices, Hangzhou, China
- Heart Regeneration and Repair Key Laboratory of Zhejiang Province, Hangzhou, China
| | - Jiacheng Wang
- Department of Cardiology, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
- State Key Laboratory of Transvascular Implantation Devices, Hangzhou, China
- Heart Regeneration and Repair Key Laboratory of Zhejiang Province, Hangzhou, China
| | - Feimu Zhang
- Department of Cardiology, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
- State Key Laboratory of Transvascular Implantation Devices, Hangzhou, China
- Heart Regeneration and Repair Key Laboratory of Zhejiang Province, Hangzhou, China
| | - Zaiyang Fu
- Department of Cardiology, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
- State Key Laboratory of Transvascular Implantation Devices, Hangzhou, China
- Heart Regeneration and Repair Key Laboratory of Zhejiang Province, Hangzhou, China
| | - Lingjun Wang
- Department of Cardiology, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
- State Key Laboratory of Transvascular Implantation Devices, Hangzhou, China
- Heart Regeneration and Repair Key Laboratory of Zhejiang Province, Hangzhou, China
| | - Biqing Wang
- Department of Cardiology, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
- State Key Laboratory of Transvascular Implantation Devices, Hangzhou, China
- Heart Regeneration and Repair Key Laboratory of Zhejiang Province, Hangzhou, China
| | - Ye Liu
- Department of Cardiology, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
- State Key Laboratory of Transvascular Implantation Devices, Hangzhou, China
- Heart Regeneration and Repair Key Laboratory of Zhejiang Province, Hangzhou, China
| | - Jing Zhao
- Department of Cardiology, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
- State Key Laboratory of Transvascular Implantation Devices, Hangzhou, China
- Heart Regeneration and Repair Key Laboratory of Zhejiang Province, Hangzhou, China
| | - Mo Li
- Department of Cardiology, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
- State Key Laboratory of Transvascular Implantation Devices, Hangzhou, China
- Heart Regeneration and Repair Key Laboratory of Zhejiang Province, Hangzhou, China
| | - Hao Lin
- Department of Cardiology, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
- State Key Laboratory of Transvascular Implantation Devices, Hangzhou, China
- Heart Regeneration and Repair Key Laboratory of Zhejiang Province, Hangzhou, China
| | - Fei Liao
- Department of Cardiology, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
- State Key Laboratory of Transvascular Implantation Devices, Hangzhou, China
- Heart Regeneration and Repair Key Laboratory of Zhejiang Province, Hangzhou, China
| | - Shuchang Ye
- Department of Cardiology, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
- State Key Laboratory of Transvascular Implantation Devices, Hangzhou, China
- Heart Regeneration and Repair Key Laboratory of Zhejiang Province, Hangzhou, China
| | - Yu Zhang
- Department of Cardiology, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
- State Key Laboratory of Transvascular Implantation Devices, Hangzhou, China
- Heart Regeneration and Repair Key Laboratory of Zhejiang Province, Hangzhou, China
| | - Jiayue Cai
- Department of Cardiology, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
- State Key Laboratory of Transvascular Implantation Devices, Hangzhou, China
- Heart Regeneration and Repair Key Laboratory of Zhejiang Province, Hangzhou, China
| | - Shaohui Shi
- Liangzhu Laboratory, Zhejiang University, Hangzhou, China
| | - Zhiwei Zhong
- Department of Cardiology, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
- State Key Laboratory of Transvascular Implantation Devices, Hangzhou, China
- Heart Regeneration and Repair Key Laboratory of Zhejiang Province, Hangzhou, China
| | - Yanna Shi
- Department of Cardiology, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
- State Key Laboratory of Transvascular Implantation Devices, Hangzhou, China
- Heart Regeneration and Repair Key Laboratory of Zhejiang Province, Hangzhou, China
| | - Junhua He
- Department of Cardiology, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
- State Key Laboratory of Transvascular Implantation Devices, Hangzhou, China
- Heart Regeneration and Repair Key Laboratory of Zhejiang Province, Hangzhou, China
| | - Xushen Xiong
- Liangzhu Laboratory, Zhejiang University, Hangzhou, China
| | - Yang Xu
- Department of Cardiology, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
- State Key Laboratory of Transvascular Implantation Devices, Hangzhou, China
- Heart Regeneration and Repair Key Laboratory of Zhejiang Province, Hangzhou, China
| | - Jinghai Chen
- Department of Cardiology, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
- State Key Laboratory of Transvascular Implantation Devices, Hangzhou, China
- Heart Regeneration and Repair Key Laboratory of Zhejiang Province, Hangzhou, China
| | - Wei Zhu
- Department of Cardiology, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
- State Key Laboratory of Transvascular Implantation Devices, Hangzhou, China
- Heart Regeneration and Repair Key Laboratory of Zhejiang Province, Hangzhou, China
| | - Yibin Wang
- Programme in Cardiovascular and Metabolic Diseases, Duke-NUS Medical School, Singapore, Singapore
| | - Jian'an Wang
- Department of Cardiology, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
- State Key Laboratory of Transvascular Implantation Devices, Hangzhou, China
- Heart Regeneration and Repair Key Laboratory of Zhejiang Province, Hangzhou, China
- Research Center for Life Science and Human Health, Binjiang Institute of Zhejiang University, Hangzhou, China
| | - Xinyang Hu
- Department of Cardiology, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China.
- State Key Laboratory of Transvascular Implantation Devices, Hangzhou, China.
- Heart Regeneration and Repair Key Laboratory of Zhejiang Province, Hangzhou, China.
- Research Center for Life Science and Human Health, Binjiang Institute of Zhejiang University, Hangzhou, China.
| |
Collapse
|
3
|
Shameem M, Olson SL, Marron Fernandez de Velasco E, Kumar A, Singh BN. Cardiac Fibroblasts: Helping or Hurting. Genes (Basel) 2025; 16:381. [PMID: 40282342 PMCID: PMC12026832 DOI: 10.3390/genes16040381] [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: 02/28/2025] [Revised: 03/22/2025] [Accepted: 03/25/2025] [Indexed: 04/29/2025] Open
Abstract
Cardiac fibroblasts (CFs) are the essential cell type for heart morphogenesis and homeostasis. In addition to maintaining the structural integrity of the heart tissue, muscle fibroblasts are involved in complex signaling cascades that regulate cardiomyocyte proliferation, migration, and maturation. While CFs serve as the primary source of extracellular matrix proteins (ECM), tissue repair, and paracrine signaling, they are also responsible for adverse pathological changes associated with cardiovascular disease. Following activation, fibroblasts produce excessive ECM components that ultimately lead to fibrosis and cardiac dysfunction. Decades of research have led to a much deeper understanding of the role of CFs in cardiogenesis. Recent studies using the single-cell genomic approach have focused on advancing the role of CFs in cellular interactions, and the mechanistic implications involved during cardiovascular development and disease. Arguably, the unique role of fibroblasts in development, tissue repair, and disease progression categorizes them into the friend or foe category. This brief review summarizes the current understanding of cardiac fibroblast biology and discusses the key findings in the context of development and pathophysiological conditions.
Collapse
Affiliation(s)
- Mohammad Shameem
- Department of Rehabilitation Medicine, University of Minnesota, Minneapolis, MN 55455, USA;
| | - Shelby L. Olson
- Department of Biomedical Engineering, University of Minnesota, Minneapolis, MN 55455, USA;
| | | | - Akhilesh Kumar
- Department of Medicine, University of Minnesota, Minneapolis, MN 55455, USA;
- Stem Cell Institute, University of Minnesota, Minneapolis, MN 55455, USA
| | - Bhairab N. Singh
- Department of Rehabilitation Medicine, University of Minnesota, Minneapolis, MN 55455, USA;
- Stem Cell Institute, University of Minnesota, Minneapolis, MN 55455, USA
| |
Collapse
|
4
|
Deng Y, He Y, Xu J, He H, Zhang M, Li G. Cardiac Fibroblasts regulate myocardium and coronary vasculature development via the collagen signaling pathway. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2024.09.11.612512. [PMID: 39314489 PMCID: PMC11418987 DOI: 10.1101/2024.09.11.612512] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/25/2024]
Abstract
The fibroblast (FB), cardiomyocyte (CM), and vascular endothelial cell (Vas_EC) are the three major cell types in the heart, yet their relationships during development are largely unexplored. To address this gap, we employed RNA staining of the FB marker gene Col1a1 together with the CM marker gene Actn2 and the Vas_EC marker gene Cdh5 at different stages. This approach enabled us to discern the anatomical pattern of cardiac FBs and identify approximately one EC and four CMs directly interacting with each FB. Molecularly, through the analysis of single-cell mRNA sequencing (scRNA-seq) data, we unveiled collagen as the top signaling molecule derived from FBs influencing CM and Vas_EC development. Subsequently, we used a Pdgfra-CreER controlled diphtheria toxin A (DTA) system to ablate the FBs at different stages. We found that the ablation of FBs disrupted myocardium and vasculature development and led to embryonic heart defects. Using scRNA-seq, we further profiled the ablated hearts and identified molecular defects in their ventricular CMs and Vas_ECs compared to control hearts. Moreover, we identified a reduction of collagen in the ablated hearts and predicted collagen as the major signaling pathway regulating the differentially expressed genes in the ablated ventricular CMs. Finally, we performed both short-term and long-term fibroblast ablation at the neonatal stage. We found that short-term ablation caused a reduction in collagen and Vas_EC density, while long-term ablation may induce compensatory collagen expression without causing heart function reduction. In summary, our study has identified the function of fibroblasts in regulating myocardium and vasculature development and implicated an important role for the collagen pathway in this process.
Collapse
Affiliation(s)
- Yiting Deng
- Department of Cell Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15201, USA
| | - Yuanhang He
- Tsinghua University, Tsinghua medicine, School of Medicine, Beijing, China
- Department of Cell Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15201, USA
| | - Juan Xu
- Department of Cell Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15201, USA
| | - Haoting He
- Department of Cell Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15201, USA
| | - Manling Zhang
- Vascular Medicine Institute, Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Guang Li
- Department of Cell Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15201, USA
| |
Collapse
|
5
|
Bakhos JJ, Saliba Y, Hajal J, Achkouty G, Oskaridjian H, Albuquerque M, Azevedo C, Semaan A, Suffee N, Balse E, Hatem SN, Fares N. Inhibiting atrial natriuretic peptide clearance reduces myocardial fibrosis and improves cardiac function in diabetic rats. EUROPEAN HEART JOURNAL OPEN 2025; 5:oeaf031. [PMID: 40201591 PMCID: PMC11977460 DOI: 10.1093/ehjopen/oeaf031] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/14/2024] [Revised: 03/06/2025] [Accepted: 03/10/2025] [Indexed: 04/10/2025]
Abstract
Aims Natriuretic peptides (NPs) exert pleiotropic effects through the recruitment of cyclic guanosine monophosphate (cGMP) signalling pathways depending on their bioavailability, which is regulated by clearance receptors and peptidases. Here, we tested the hypothesis that increasing myocardial bioavailability of NP has a beneficial effect on heart failure. We studied the effects of a mutated NP, M-atrial natriuretic peptide (MANP), resistant to neprilysin in a model of diabetic cardiomyopathy characterized by marked myocardial fibrosis. Methods and results Natriuretic peptides as well as sacubitril were delivered via osmotic mini-pumps to high-fat/streptozotocin-induced Type 2 diabetic (T2D) rats. Cardiac function was evaluated by echocardiography. Myocardial remodelling was studied by histological approaches, collagen phenotype, and atrial natriuretic peptide (ANP)/cGMP concentrations. Live-cell cGMP biosensing was conducted on cultured rat cardiac fibroblasts to investigate the biological effects of NP. Cyclic guanosine monophosphate signalling pathway was studied using multiple antibody arrays and biochemical assays in cardiac tissue and cultured fibroblasts. M-atrial natriuretic peptide exhibits superior efficacy than ANP in reducing left ventricular dysfunction and myocardial fibrosis with less extracellular matrix deposition. In vitro, MANP and ANP similarly generated cGMP and activated the protein kinase G (PKG) signalling pathway in cardiac fibroblasts, attenuating Mothers against decapentaplegic homolog 2 (SMAD) activation, collagen secretion, and cell proliferation. Nevertheless, in vivo, MANP specifically enhanced cardiac cGMP accumulation and was more potent than ANP in activating myocardial cGMP/PKG signalling and inhibiting the profibrotic SMAD, extracellular signal-regulated kinases 1/2, and nuclear factor of activated T cells 3 pathways. Endopeptidase inhibition using sacubitril also led to cardiac ANP/cGMP accumulation and reduced myocardial fibrosis. Conclusion Myocardial bioavailability of ANP is a major determinant of peptide efficacy in reducing cardiac fibrosis and improving pump function during diabetic cardiomyopathy.
Collapse
Affiliation(s)
- Jules Joel Bakhos
- Laboratory of Research in Physiology and Pathophysiology, Faculty of Medicine, Saint-Joseph University of Beirut, 17-5208 - Mar Mikhaël, Beirut 1104 2020, Lebanon
| | - Youakim Saliba
- Laboratory of Research in Physiology and Pathophysiology, Faculty of Medicine, Saint-Joseph University of Beirut, 17-5208 - Mar Mikhaël, Beirut 1104 2020, Lebanon
| | - Joelle Hajal
- Laboratory of Research in Physiology and Pathophysiology, Faculty of Medicine, Saint-Joseph University of Beirut, 17-5208 - Mar Mikhaël, Beirut 1104 2020, Lebanon
| | - Guy Achkouty
- Laboratory of Research in Physiology and Pathophysiology, Faculty of Medicine, Saint-Joseph University of Beirut, 17-5208 - Mar Mikhaël, Beirut 1104 2020, Lebanon
| | - Hrag Oskaridjian
- Laboratory of Research in Physiology and Pathophysiology, Faculty of Medicine, Saint-Joseph University of Beirut, 17-5208 - Mar Mikhaël, Beirut 1104 2020, Lebanon
| | - Miguel Albuquerque
- INSERM, Centre de Recherche sur L'inflammation, UMR 1149, Université Paris-Cité, 45 Rue des Saints-Pères 75006 Paris, France
- Service d'Anatomie Pathologique, Hôpital Beaujon, Assistance Publique-Hôpitaux de Paris, 100 Bd du Général Leclerc, 92110 Clichy, France
| | - Chloé Azevedo
- Institute of Cardiometabolism and Nutrition, IHU ICAN, Sorbonne University, INSERM UMR_S1166 Pitié-Salpêtrière Hospital, 47-83 boulevard de l'Hôpital, 75013 Paris, France
| | - Albert Semaan
- Laboratory of Research in Physiology and Pathophysiology, Faculty of Medicine, Saint-Joseph University of Beirut, 17-5208 - Mar Mikhaël, Beirut 1104 2020, Lebanon
| | - Nadine Suffee
- Institute of Cardiometabolism and Nutrition, IHU ICAN, Sorbonne University, INSERM UMR_S1166 Pitié-Salpêtrière Hospital, 47-83 boulevard de l'Hôpital, 75013 Paris, France
| | - Elise Balse
- Institute of Cardiometabolism and Nutrition, IHU ICAN, Sorbonne University, INSERM UMR_S1166 Pitié-Salpêtrière Hospital, 47-83 boulevard de l'Hôpital, 75013 Paris, France
| | - Stéphane N Hatem
- Institute of Cardiometabolism and Nutrition, IHU ICAN, Sorbonne University, INSERM UMR_S1166 Pitié-Salpêtrière Hospital, 47-83 boulevard de l'Hôpital, 75013 Paris, France
| | - Nassim Fares
- Laboratory of Research in Physiology and Pathophysiology, Faculty of Medicine, Saint-Joseph University of Beirut, 17-5208 - Mar Mikhaël, Beirut 1104 2020, Lebanon
| |
Collapse
|
6
|
Johansen AKZ, Kasam RK, Vagnozzi RJ, Lin SCJ, Gomez-Arroyo JG, Shittu A, Bowers SL, Kuwabara Y, Grimes KM, Warrick K, Sargent MA, Baldwin TA, Quaggin SE, Barski A, Molkentin JD. Transcription Factor 21 Regulates Cardiac Myofibroblast Formation and Fibrosis. Circ Res 2025; 136:44-58. [PMID: 39629593 PMCID: PMC11740189 DOI: 10.1161/circresaha.124.325527] [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: 09/10/2024] [Revised: 11/07/2024] [Accepted: 11/24/2024] [Indexed: 01/04/2025]
Abstract
BACKGROUND TCF21 (transcription factor 21) is a bHLH (basic helix-loop-helix) protein required for the developmental specification of cardiac fibroblasts (CFs) from epicardial progenitor cells that surround the embryonic heart. In the adult heart, TCF21 is expressed in tissue-resident fibroblasts and is downregulated in response to injury or stimuli leading to myofibroblast differentiation. These findings led to the hypothesis that TCF21 regulates fibroblast differentiation in the adult mammalian heart to affect fibrosis. METHODS Tamoxifen-inducible Cre genetic mouse models were used to permit either Tcf21 gene deletion or its enforced expression in adult CFs. Histological and echocardiographic analyses were used, as well as transcriptomic analysis to determine the consequences of TCF21 gain-of-function and loss-of-function in vivo. Genomic Tcf21 occupancy was identified by chromatin immunoprecipitation and sequencing in CFs. Myocardial infarction and AngII (angiotensin II)/phenylephrine served as models of cardiac fibrosis. RESULTS Acute and long-term deletion of Tcf21 in CFs of the adult mouse heart does not alter fibroblast numbers, myofibroblast differentiation, or fibrosis. Fibroblast-specific Tcf21 gene-deleted mice demonstrate no significant alterations in cardiac function or scar formation in response to cardiac injury compared with control mice. In contrast, enforced expression of TCF21 in CFs inhibits myofibroblast differentiation and significantly reduces cardiac fibrosis and hypertrophy in response to 1 week of Ang II/phenylephrine infusion. Mechanistically, sustained TCF21 expression prevents the induction of genes associated with fibrosis and ECM (extracellular matrix) organization. CONCLUSIONS TCF21 expression is not required to maintain the cell state of CFs in the adult heart. However, preventing the normal downregulation of TCF21 expression with injury reduces myofibroblast formation, cardiac fibrosis, and the acute cardiac hypertrophic response following 1 week of Ang II/phenylephrine stimulation.
Collapse
Affiliation(s)
- Anne Katrine Z. Johansen
- Department of Pediatrics, Division of Molecular Cardiovascular Biology, University of Cincinnati and Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio, USA
| | - Rajesh K. Kasam
- Department of Pediatrics, Division of Molecular Cardiovascular Biology, University of Cincinnati and Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio, USA
| | - Ronald J. Vagnozzi
- Department of Pediatrics, Division of Molecular Cardiovascular Biology, University of Cincinnati and Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio, USA
- Division of Cardiology, Consortium for Fibrosis Research and Translation, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Suh-Chin J. Lin
- Department of Pediatrics, Division of Molecular Cardiovascular Biology, University of Cincinnati and Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio, USA
| | - Jose G. Gomez-Arroyo
- Department of Pediatrics, Division of Pulmonary and Critical Care Medicine, University of Cincinnati and Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio, USA
| | - Adenike Shittu
- Department of Pediatrics, Division of Allergy and Immunology, University of Cincinnati and Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio, USA
- Department of Pediatrics, Division of Human Genetics, University of Cincinnati and Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio, USA
| | - Stephanie L.K. Bowers
- Department of Pediatrics, Division of Molecular Cardiovascular Biology, University of Cincinnati and Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio, USA
| | - Yasuhide Kuwabara
- Department of Pediatrics, Division of Molecular Cardiovascular Biology, University of Cincinnati and Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio, USA
| | - Kelly M. Grimes
- Department of Pediatrics, Division of Molecular Cardiovascular Biology, University of Cincinnati and Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio, USA
| | - Kathrynne Warrick
- Department of Pediatrics, Division of Molecular Cardiovascular Biology, University of Cincinnati and Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio, USA
| | - Michelle A. Sargent
- Department of Pediatrics, Division of Molecular Cardiovascular Biology, University of Cincinnati and Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio, USA
| | - Tanya A. Baldwin
- Department of Pediatrics, Division of Molecular Cardiovascular Biology, University of Cincinnati and Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio, USA
| | - Susan E. Quaggin
- Feinberg Cardiovascular Research and Renal Institute, Northwestern University, Chicago, Illinois, USA
| | - Artem Barski
- Department of Pediatrics, Division of Allergy and Immunology, University of Cincinnati and Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio, USA
- Department of Pediatrics, Division of Human Genetics, University of Cincinnati and Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio, USA
| | - Jeffery D. Molkentin
- Department of Pediatrics, Division of Molecular Cardiovascular Biology, University of Cincinnati and Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio, USA
| |
Collapse
|
7
|
Kimball TH, Gromova T, Gehred ND, Chapski DJ, Wang K, Vaseghi M, Fischer MA, Lefer DJ, Vondriska TM. Rapid onset fibrotic remodeling and ventricular dysfunction induced by phenylephrine involve targeted reprogramming of myocyte and fibroblast transcriptomes. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.10.11.617933. [PMID: 39464022 PMCID: PMC11507669 DOI: 10.1101/2024.10.11.617933] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 10/29/2024]
Abstract
Catecholamine dysregulation is a common feature of multiple acute and chronic cardiac conditions, including heart failure. To investigate the role of altered α-adrenergic stimulation on cardiac function, we developed a short-term exposure model, administering phenylephrine subcutaneously to mice for one week. Compared to vehicle-injected controls, phenylephrine-treated animals exhibited increased ejection fraction, decreased chamber size, diastolic dysfunction and ventricular hypertrophy in the absence of hypertension. Remarkably, these animals developed extensive fibrotic remodeling of the tissue that plateaued at 24 hours and myocyte hypertrophy localized to regions of fibrotic deposition after 3 days of treatment. Transcriptome analyses of purified myocyte and fibroblast populations from these hearts revealed an unexpected role for myocytes in the production of extracellular matrix. Comparison with other models of cardiac stress, including pressure overload hypertrophy and cytokine activation of fibroblasts, identified stimulus-specific transcriptional circuits associated with cardiac pathology. Given the rapid, robust fibrotic response that preceded myocyte hypertrophy, intercellular communication analyses were conducted to investigate fibroblast to myocyte signaling, identifying potential crosstalk between these cells. These studies thoroughly describe and phenotypically characterize a new model of short-term catecholamine stress and provide an atlas of transcriptional remodeling in myocytes and fibroblasts.
Collapse
Affiliation(s)
- Todd H. Kimball
- Departments of Anesthesiology & Perioperative Medicine, David Geffen School of Medicine
| | - Tatiana Gromova
- Departments of Anesthesiology & Perioperative Medicine, David Geffen School of Medicine
| | - Natalie D. Gehred
- Departments of Anesthesiology & Perioperative Medicine, David Geffen School of Medicine
| | - Douglas J. Chapski
- Departments of Anesthesiology & Perioperative Medicine, David Geffen School of Medicine
| | - Ke Wang
- Department of Cardiac Surgery, Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles
| | - Marmar Vaseghi
- Cardiac Arrhythmia Center, Division of Cardiology, Department of Medicine, University of California, Los Angeles
| | - Matthew A. Fischer
- Departments of Anesthesiology & Perioperative Medicine, David Geffen School of Medicine
| | - David J. Lefer
- Department of Cardiac Surgery, Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles
| | - Thomas M. Vondriska
- Departments of Anesthesiology & Perioperative Medicine, David Geffen School of Medicine
- Physiology, David Geffen School of Medicine
- Medicine, David Geffen School of Medicine
- Molecular Biology Institute, University of California, Los Angeles
| |
Collapse
|
8
|
Patrick R, Janbandhu V, Tallapragada V, Tan SSM, McKinna EE, Contreras O, Ghazanfar S, Humphreys DT, Murray NJ, Tran YTH, Hume RD, Chong JJH, Harvey RP. Integration mapping of cardiac fibroblast single-cell transcriptomes elucidates cellular principles of fibrosis in diverse pathologies. SCIENCE ADVANCES 2024; 10:eadk8501. [PMID: 38905342 PMCID: PMC11192082 DOI: 10.1126/sciadv.adk8501] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/16/2023] [Accepted: 05/14/2024] [Indexed: 06/23/2024]
Abstract
Single-cell technology has allowed researchers to probe tissue complexity and dynamics at unprecedented depth in health and disease. However, the generation of high-dimensionality single-cell atlases and virtual three-dimensional tissues requires integrated reference maps that harmonize disparate experimental designs, analytical pipelines, and taxonomies. Here, we present a comprehensive single-cell transcriptome integration map of cardiac fibrosis, which underpins pathophysiology in most cardiovascular diseases. Our findings reveal similarity between cardiac fibroblast (CF) identities and dynamics in ischemic versus pressure overload models of cardiomyopathy. We also describe timelines for commitment of activated CFs to proliferation and myofibrogenesis, profibrotic and antifibrotic polarization of myofibroblasts and matrifibrocytes, and CF conservation across mouse and human healthy and diseased hearts. These insights have the potential to inform knowledge-based therapies.
Collapse
Affiliation(s)
- Ralph Patrick
- Victor Chang Cardiac Research Institute, Darlinghurst, NSW 2010, Australia
- School of Clinical Medicine, UNSW Sydney, Kensington, NSW 2052, Australia
- Institute for Molecular Bioscience, The University of Queensland, St. Lucia, QLD 4072, Australia
| | - Vaibhao Janbandhu
- Victor Chang Cardiac Research Institute, Darlinghurst, NSW 2010, Australia
- School of Clinical Medicine, UNSW Sydney, Kensington, NSW 2052, Australia
| | | | - Shannon S. M. Tan
- Victor Chang Cardiac Research Institute, Darlinghurst, NSW 2010, Australia
| | - Emily E. McKinna
- Victor Chang Cardiac Research Institute, Darlinghurst, NSW 2010, Australia
- Westmead Institute for Medical Research, The University of Sydney, Westmead, NSW 2145, Australia
| | - Osvaldo Contreras
- Victor Chang Cardiac Research Institute, Darlinghurst, NSW 2010, Australia
- School of Clinical Medicine, UNSW Sydney, Kensington, NSW 2052, Australia
| | - Shila Ghazanfar
- School of Mathematics and Statistics, The University of Sydney, Camperdown, NSW 2006, Australia
- Charles Perkins Centre, The University of Sydney, Camperdown, NSW 2006, Australia
- Sydney Precision Data Science Centre, The University of Sydney, Camperdown, NSW 2006, Australia
| | - David T. Humphreys
- Victor Chang Cardiac Research Institute, Darlinghurst, NSW 2010, Australia
- School of Clinical Medicine, UNSW Sydney, Kensington, NSW 2052, Australia
| | - Nicholas J. Murray
- Victor Chang Cardiac Research Institute, Darlinghurst, NSW 2010, Australia
- School of Clinical Medicine, UNSW Sydney, Kensington, NSW 2052, Australia
| | - Yen T. H. Tran
- Victor Chang Cardiac Research Institute, Darlinghurst, NSW 2010, Australia
| | - Robert D. Hume
- Westmead Institute for Medical Research, The University of Sydney, Westmead, NSW 2145, Australia
- School of Medical Science, The University of Sydney, Camperdown, NSW 2006, Australia
- Centre for Heart Failure and Diseases of the Aorta, The Baird Institute, Sydney, NSW 2042, Australia
| | - James J. H. Chong
- Westmead Institute for Medical Research, The University of Sydney, Westmead, NSW 2145, Australia
- Department of Cardiology, Westmead Hospital, Westmead, NSW 2145, Australia
| | - Richard P. Harvey
- Victor Chang Cardiac Research Institute, Darlinghurst, NSW 2010, Australia
- School of Clinical Medicine, UNSW Sydney, Kensington, NSW 2052, Australia
- School of Biotechnology and Biomolecular Science, UNSW Sydney, Kensington, NSW 2052, Australia
| |
Collapse
|
9
|
Voronkov NS, Maslov LN, Vyshlov EV, Mukhomedzyanov AV, Ryabov VV, Derkachev IA, Kan A, Gusakova SV, Gombozhapova AE, Panteleev OO. Do platelets protect the heart against ischemia/reperfusion injury or exacerbate cardiac ischemia/reperfusion injury? The role of PDGF, VEGF, and PAF. Life Sci 2024; 347:122617. [PMID: 38608835 DOI: 10.1016/j.lfs.2024.122617] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2024] [Revised: 03/15/2024] [Accepted: 04/05/2024] [Indexed: 04/14/2024]
Abstract
BACKGROUND Acute myocardial infarction (AMI) is one of the main causes of death. It is quite obvious that there is an urgent need to develop new approaches for treatment of AMI. OBJECTIVE This review analyzes data on the role of platelets in the regulation of cardiac tolerance to ischemia/reperfusion (I/R). METHODS It was performed a search of topical articles using PubMed databases. FINDINGS Platelets activated by a cholesterol-enriched diet, thrombin, and myocardial ischemia exacerbate I/R injury of the heart. The P2Y12 receptor antagonists, remote ischemic postconditioning and conditioning alter the properties of platelets. Platelets acquire the ability to increase cardiac tolerance to I/R. Platelet-derived growth factors (PDGFs) increase tolerance of cardiomyocytes and endothelial cells to I/R. PDGF receptors (PDGFRs) were found in cardiomyocytes and endothelial cells. PDGFs decrease infarct size and partially abrogate adverse postinfarction remodeling. Protein kinase C, phosphoinositide 3-kinase, and Akt involved in the cytoprotective effect of PDGFs. Vascular endothelial growth factor increased cardiac tolerance to I/R and alleviated adverse postinfarction remodeling. The platelet-activating factor (PAF) receptor inhibitors increase cardiac tolerance to I/R in vivo. PAF enhances cardiac tolerance to I/R in vitro. It is possible that PAF receptor inhibitors could protect the heart by blocking PAF receptor localized outside the heart. PAF protects the heart through activation of PAF receptor localized in cardiomyocytes or endothelial cells. Reactive oxygen species and kinases are involved in the cardioprotective effect of PAF. CONCLUSION Platelets play an important role in the regulation of cardiac tolerance to I/R.
Collapse
Affiliation(s)
- Nikita S Voronkov
- Department of Emergency Cardiology and Laboratory of Experimental Cardiology, Cardiology Research Institute, Tomsk National Research Medical Center, Russian Academy of Sciences, 634012 Tomsk, Russia
| | - Leonid N Maslov
- Department of Emergency Cardiology and Laboratory of Experimental Cardiology, Cardiology Research Institute, Tomsk National Research Medical Center, Russian Academy of Sciences, 634012 Tomsk, Russia.
| | - Evgeniy V Vyshlov
- Department of Emergency Cardiology and Laboratory of Experimental Cardiology, Cardiology Research Institute, Tomsk National Research Medical Center, Russian Academy of Sciences, 634012 Tomsk, Russia
| | - Alexander V Mukhomedzyanov
- Department of Emergency Cardiology and Laboratory of Experimental Cardiology, Cardiology Research Institute, Tomsk National Research Medical Center, Russian Academy of Sciences, 634012 Tomsk, Russia
| | - Vyacheslav V Ryabov
- Department of Emergency Cardiology and Laboratory of Experimental Cardiology, Cardiology Research Institute, Tomsk National Research Medical Center, Russian Academy of Sciences, 634012 Tomsk, Russia
| | - Ivan A Derkachev
- Department of Emergency Cardiology and Laboratory of Experimental Cardiology, Cardiology Research Institute, Tomsk National Research Medical Center, Russian Academy of Sciences, 634012 Tomsk, Russia
| | - Artur Kan
- Department of Emergency Cardiology and Laboratory of Experimental Cardiology, Cardiology Research Institute, Tomsk National Research Medical Center, Russian Academy of Sciences, 634012 Tomsk, Russia
| | - Svetlana V Gusakova
- Department of Biophysics and Functional Diagnostics, Siberian State Medical University, 634050 Tomsk, Russia
| | - Alexandra E Gombozhapova
- Department of Emergency Cardiology and Laboratory of Experimental Cardiology, Cardiology Research Institute, Tomsk National Research Medical Center, Russian Academy of Sciences, 634012 Tomsk, Russia
| | - Oleg O Panteleev
- Department of Emergency Cardiology and Laboratory of Experimental Cardiology, Cardiology Research Institute, Tomsk National Research Medical Center, Russian Academy of Sciences, 634012 Tomsk, Russia
| |
Collapse
|
10
|
Sanches BDA, Teófilo FBS, Brunet MY, Villapun VM, Man K, Rocha LC, Neto JP, Matsumoto MR, Maldarine JS, Ciena AP, Cox SC, Carvalho HF. Telocytes: current methods of research, challenges and future perspectives. Cell Tissue Res 2024; 396:141-155. [PMID: 38539007 DOI: 10.1007/s00441-024-03888-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2023] [Accepted: 03/12/2024] [Indexed: 04/28/2024]
Abstract
Telocytes (TCs) are CD34-positive interstitial cells that have long cytoplasmic projections, called telopodes; they have been identified in several organs and in various species. These cells establish a complex communication network between different stromal and epithelial cell types, and there is growing evidence that they play a key role in physiology and pathology. In many tissues, TC network impairment has been implicated in the onset and progression of pathological conditions, which makes the study of TCs of great interest for the development of novel therapies. In this review, we summarise the main methods involved in the characterisation of these cells as well as their inherent difficulties and then discuss the functional assays that are used to uncover the role of TCs in normal and pathological conditions, from the most traditional to the most recent. Furthermore, we provide future perspectives in the study of TCs, especially regarding the establishment of more precise markers, commercial lineages and means for drug delivery and genetic editing that directly target TCs.
Collapse
Affiliation(s)
- Bruno D A Sanches
- Department of Structural and Functional Biology, Institute of Biology, State University of Campinas (UNICAMP), Carl von Linnaeus St. Bldg G1, Bertrand Russel Ave., Campinas, São Paulo, Brazil
| | - Francisco B S Teófilo
- Department of Structural and Functional Biology, Institute of Biology, State University of Campinas (UNICAMP), Carl von Linnaeus St. Bldg G1, Bertrand Russel Ave., Campinas, São Paulo, Brazil
| | - Mathieu Y Brunet
- School of Chemical Engineering, University of Birmingham, Edgbaston, Birmingham, B15 2TT, UK
| | - Victor M Villapun
- School of Chemical Engineering, University of Birmingham, Edgbaston, Birmingham, B15 2TT, UK
| | - Kenny Man
- Department of Oral and Maxillofacial Surgery & Special Dental Care, University Medical Center Utrecht, Utrecht University, Utrecht, 3508 GA, The Netherlands
- Regenerative Medicine Center Utrecht, Utrecht, 3584 CT, The Netherlands
| | - Lara C Rocha
- Laboratory of Morphology and Physical Activity (LAMAF), Institute of Biosciences, São Paulo State University (UNESP), 1515 24 A Ave., Rio Claro, São Paulo, Brazil
| | - Jurandyr Pimentel Neto
- Laboratory of Morphology and Physical Activity (LAMAF), Institute of Biosciences, São Paulo State University (UNESP), 1515 24 A Ave., Rio Claro, São Paulo, Brazil
| | - Marta R Matsumoto
- Department of Structural and Functional Biology, Institute of Biology, State University of Campinas (UNICAMP), Carl von Linnaeus St. Bldg G1, Bertrand Russel Ave., Campinas, São Paulo, Brazil
| | - Juliana S Maldarine
- Department of Structural and Functional Biology, Institute of Biology, State University of Campinas (UNICAMP), Carl von Linnaeus St. Bldg G1, Bertrand Russel Ave., Campinas, São Paulo, Brazil
| | - Adriano P Ciena
- Laboratory of Morphology and Physical Activity (LAMAF), Institute of Biosciences, São Paulo State University (UNESP), 1515 24 A Ave., Rio Claro, São Paulo, Brazil
| | - Sophie C Cox
- School of Chemical Engineering, University of Birmingham, Edgbaston, Birmingham, B15 2TT, UK
| | - Hernandes F Carvalho
- Department of Structural and Functional Biology, Institute of Biology, State University of Campinas (UNICAMP), Carl von Linnaeus St. Bldg G1, Bertrand Russel Ave., Campinas, São Paulo, Brazil.
| |
Collapse
|
11
|
Bengel F, Epstein JA, Gropler R, Haberkorn U, Kramann R, Lavine K, Leuschner F, Liu Y, Rosenthal N, Wu H. Linking immune modulation to cardiac fibrosis. NATURE CARDIOVASCULAR RESEARCH 2024; 3:414-419. [PMID: 39196217 DOI: 10.1038/s44161-024-00459-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/29/2024]
Affiliation(s)
| | | | - Robert Gropler
- Division of Radiological Sciences, Edward Mallinckrodt Institute of Radiology, Washington University School of Medicine, St Louis, MO, USA.
| | | | - Rafael Kramann
- Department of Medicine 2, RWTH Aachen University, Medical Faculty, Aachen, Germany
| | - Kory Lavine
- Center for Cardiovascular Research, Department of Medicine, Cardiovascular Division, Washington University School of Medicine, St Louis, MO, USA
- Department of Pathology and Immunology, Washington University School of Medicine, St Louis, MO, USA
- Department of Developmental Biology, Washington University School of Medicine, St Louis, MO, USA
| | - Florian Leuschner
- Department of Internal Medicine III (Cardiology, Angiology, and Pneumology), Heidelberg University Hospital, Heidelberg, Germany.
- DZHK (German Center for Cardiovascular Research), Partner Site Heidelberg/Mannheim, Heidelberg, Germany.
| | - Yongjian Liu
- Division of Radiological Sciences, Edward Mallinckrodt Institute of Radiology, Washington University School of Medicine, St Louis, MO, USA
| | - Nadia Rosenthal
- The Jackson Laboratory, Bar Harbor, ME, USA.
- National Heart and Lung Institute, Imperial College London, London, UK.
| | - Hao Wu
- University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| |
Collapse
|
12
|
Fu R, You N, Li R, Zhao X, Li Y, Li X, Jiang W. Renalase mediates macrophage-to-fibroblast crosstalk to attenuate pressure overload-induced pathological myocardial fibrosis. J Hypertens 2024; 42:629-643. [PMID: 38230609 DOI: 10.1097/hjh.0000000000003635] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2024]
Abstract
A potential antifibrotic mechanism in pathological myocardial remodeling is the recruitment of beneficial functional subpopulations of macrophages or the transformation of their phenotype. Macrophages are required to activate molecular cascades that regulate fibroblast behavior. Identifying mediators that activate the antifibrotic macrophage phenotype is tantamount to identifying the button that retards pathological remodeling of the myocardium; however, relevant studies are inadequate. Circulating renalase (RNLS) is mainly of renal origin, and cardiac myocytes also secrete it autonomously. Our previous studies revealed that RNLS delivers cell signaling to exert multiple cardiovascular protective effects, including the improvement of myocardial ischemia, and heart failure. Here, we further investigated the potential mechanism by which macrophage phenotypic transformation is targeted by RNLS to mediate stress load-induced myocardial fibrosis. Mice subjected to transverse aortic constriction (TAC) were used as a model of myocardial fibrosis. The co-incubation of macrophages and cardiac fibroblasts was used to study intercellular signaling. The results showed that RNLS co-localized with macrophages and reduced protein expression after cardiac pressure overload. TAC mice exhibited improved cardiac function and alleviated left ventricular fibrosis when exogenous RNLS was administered. Flow sorting showed that RNLS is essential for macrophage polarization towards a restorative phenotype (M2-like), thereby inhibiting myofibroblast activation, as proven by both mouse RAW264.7 and bone marrow-derived macrophage models. Mechanistically, we found that activated protein kinase B is a major pathway by which RNLS promotes M2 polarization in macrophages. RNLS may serve as a prognostic biomarker and a potential clinical candidate for the treatment of myocardial fibrosis.
Collapse
Affiliation(s)
- Ru Fu
- Department of Cardiology, The Third Xiangya Hospital, Central South University, Changsha, China
| | | | | | | | | | | | | |
Collapse
|
13
|
Hastings MH, Castro C, Freeman R, Abdul Kadir A, Lerchenmüller C, Li H, Rhee J, Roh JD, Roh K, Singh AP, Wu C, Xia P, Zhou Q, Xiao J, Rosenzweig A. Intrinsic and Extrinsic Contributors to the Cardiac Benefits of Exercise. JACC Basic Transl Sci 2024; 9:535-552. [PMID: 38680954 PMCID: PMC11055208 DOI: 10.1016/j.jacbts.2023.07.011] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/06/2023] [Revised: 07/06/2023] [Accepted: 07/20/2023] [Indexed: 05/01/2024]
Abstract
Among its many cardiovascular benefits, exercise training improves heart function and protects the heart against age-related decline, pathological stress, and injury. Here, we focus on cardiac benefits with an emphasis on more recent updates to our understanding. While the cardiomyocyte continues to play a central role as both a target and effector of exercise's benefits, there is a growing recognition of the important roles of other, noncardiomyocyte lineages and pathways, including some that lie outside the heart itself. We review what is known about mediators of exercise's benefits-both those intrinsic to the heart (at the level of cardiomyocytes, fibroblasts, or vascular cells) and those that are systemic (including metabolism, inflammation, the microbiome, and aging)-highlighting what is known about the molecular mechanisms responsible.
Collapse
Affiliation(s)
- Margaret H. Hastings
- Institute for Heart and Brain Health, University of Michigan Medical Center, Ann Arbor, Michigan, USA
| | - Claire Castro
- Cardiovascular Research Center, Division of Cardiology, Corrigan Minehan Heart Center, Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Rebecca Freeman
- Cardiovascular Research Center, Division of Cardiology, Corrigan Minehan Heart Center, Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Azrul Abdul Kadir
- Cardiovascular Research Center, Division of Cardiology, Corrigan Minehan Heart Center, Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Carolin Lerchenmüller
- Department of Cardiology, University Hospital Heidelberg, German Center for Cardiovascular Research (DZHK), Partner Site Heidelberg/Mannheim, Heidelberg, Germany
| | - Haobo Li
- Cardiovascular Research Center, Division of Cardiology, Corrigan Minehan Heart Center, Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - James Rhee
- Cardiovascular Research Center, Division of Cardiology, Corrigan Minehan Heart Center, Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
- Department of Anesthesiology and Critical Care, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Jason D. Roh
- Cardiovascular Research Center, Division of Cardiology, Corrigan Minehan Heart Center, Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Kangsan Roh
- Cardiovascular Research Center, Division of Cardiology, Corrigan Minehan Heart Center, Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
- Department of Anesthesiology and Critical Care, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Anand P. Singh
- Institute for Heart and Brain Health, University of Michigan Medical Center, Ann Arbor, Michigan, USA
| | - Chao Wu
- Institute for Heart and Brain Health, University of Michigan Medical Center, Ann Arbor, Michigan, USA
| | - Peng Xia
- Cardiovascular Research Center, Division of Cardiology, Corrigan Minehan Heart Center, Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Qiulian Zhou
- Institute for Heart and Brain Health, University of Michigan Medical Center, Ann Arbor, Michigan, USA
| | - Junjie Xiao
- Cardiac Regeneration and Ageing Lab, Institute of Cardiovascular Sciences, School of Life Science, Shanghai University, Shanghai, China
| | - Anthony Rosenzweig
- Institute for Heart and Brain Health, University of Michigan Medical Center, Ann Arbor, Michigan, USA
| |
Collapse
|
14
|
Mohr ME, Li S, Trouten AM, Stairley RA, Roddy PL, Liu C, Zhang M, Sucov HM, Tao G. Cardiomyocyte-fibroblast interaction regulates ferroptosis and fibrosis after myocardial injury. iScience 2024; 27:109219. [PMID: 38469561 PMCID: PMC10926204 DOI: 10.1016/j.isci.2024.109219] [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: 09/27/2023] [Revised: 12/11/2023] [Accepted: 02/07/2024] [Indexed: 03/13/2024] Open
Abstract
Neonatal mouse hearts have transient renewal capacity, which is lost in juvenile and adult stages. In neonatal mouse hearts, myocardial infarction (MI) causes an initial loss of cardiomyocytes. However, it is unclear which type of regulated cell death (RCD) occurs in stressed cardiomyocytes. In the current studies, we induced MI in neonatal and juvenile mouse hearts and showed that ischemic cardiomyocytes primarily undergo ferroptosis, a non-apoptotic and iron-dependent form of RCD. We demonstrated that cardiac fibroblasts (CFs) protect cardiomyocytes from ferroptosis through paracrine effects and direct cell-cell interaction. CFs show strong resistance to ferroptosis due to high ferritin expression. The fibrogenic activity of CFs, typically considered detrimental to heart function, is negatively regulated by paired-like homeodomain 2 (Pitx2) signaling from cardiomyocytes. In addition, Pitx2 prevents ferroptosis in cardiomyocytes by regulating ferroptotic genes. Understanding the regulatory mechanisms of cardiomyocyte survival and death can identify potentially translatable therapeutic strategies for MI.
Collapse
Affiliation(s)
- Mary E. Mohr
- Department of Regenerative Medicine and Cell Biology, Medical University of South Carolina, Charleston, SC 29425, USA
| | - Shuang Li
- Department of Regenerative Medicine and Cell Biology, Medical University of South Carolina, Charleston, SC 29425, USA
| | - Allison M. Trouten
- Department of Regenerative Medicine and Cell Biology, Medical University of South Carolina, Charleston, SC 29425, USA
| | - Rebecca A. Stairley
- Department of Regenerative Medicine and Cell Biology, Medical University of South Carolina, Charleston, SC 29425, USA
| | - Patrick L. Roddy
- Department of Regenerative Medicine and Cell Biology, Medical University of South Carolina, Charleston, SC 29425, USA
| | - Chun Liu
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA 94305, USA
- Department of Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Min Zhang
- Pediatric Translational Medicine Institute, Shanghai Children’s Medical Center, Shanghai Jiao Tong University School of Medicine, Shanghai 200127, China
| | - Henry M. Sucov
- Department of Regenerative Medicine and Cell Biology, Medical University of South Carolina, Charleston, SC 29425, USA
- Department of Medicine, Division of Cardiology, Medical University of South Carolina, Charleston, SC 29425, USA
| | - Ge Tao
- Department of Regenerative Medicine and Cell Biology, Medical University of South Carolina, Charleston, SC 29425, USA
| |
Collapse
|
15
|
Liu D, Wang M, Murthy V, McNamara DM, Nguyen TTL, Philips TJ, Vyas H, Gao H, Sahni J, Starling RC, Cooper LT, Skime MK, Batzler A, Jenkins GD, Barlera S, Pileggi S, Mestroni L, Merlo M, Sinagra G, Pinet F, Krejčí J, Chaloupka A, Miller JD, de Groote P, Tschumperlin DJ, Weinshilboum RM, Pereira NL. Myocardial Recovery in Recent Onset Dilated Cardiomyopathy: Role of CDCP1 and Cardiac Fibrosis. Circ Res 2023; 133:810-825. [PMID: 37800334 PMCID: PMC10746262 DOI: 10.1161/circresaha.123.323200] [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: 06/25/2023] [Accepted: 09/22/2023] [Indexed: 10/07/2023]
Abstract
BACKGROUND Dilated cardiomyopathy (DCM) is a major cause of heart failure and carries a high mortality rate. Myocardial recovery in DCM-related heart failure patients is highly variable, with some patients having little or no response to standard drug therapy. A genome-wide association study may agnostically identify biomarkers and provide novel insight into the biology of myocardial recovery in DCM. METHODS A genome-wide association study for change in left ventricular ejection fraction was performed in 686 White subjects with recent-onset DCM who received standard pharmacotherapy. Genome-wide association study signals were subsequently functionally validated and studied in relevant cellular models to understand molecular mechanisms that may have contributed to the change in left ventricular ejection fraction. RESULTS The genome-wide association study identified a highly suggestive locus that mapped to the 5'-flanking region of the CDCP1 (CUB [complement C1r/C1s, Uegf, and Bmp1] domain containing protein 1) gene (rs6773435; P=7.12×10-7). The variant allele was associated with improved cardiac function and decreased CDCP1 transcription. CDCP1 expression was significantly upregulated in human cardiac fibroblasts (HCFs) in response to the PDGF (platelet-derived growth factor) signaling, and knockdown of CDCP1 significantly repressed HCF proliferation and decreased AKT (protein kinase B) phosphorylation. Transcriptomic profiling after CDCP1 knockdown in HCFs supported the conclusion that CDCP1 regulates HCF proliferation and mitosis. In addition, CDCP1 knockdown in HCFs resulted in significantly decreased expression of soluble ST2 (suppression of tumorigenicity-2), a prognostic biomarker for heart failure and inductor of cardiac fibrosis. CONCLUSIONS CDCP1 may play an important role in myocardial recovery in recent-onset DCM and mediates its effect primarily by attenuating cardiac fibrosis.
Collapse
Affiliation(s)
- Duan Liu
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, MN, USA
| | - Min Wang
- Department of Cardiovascular Medicine, Mayo Clinic, Rochester, MN, USA
| | - Vishakantha Murthy
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, MN, USA
- Department of Cardiovascular Medicine, Mayo Clinic, Rochester, MN, USA
- Department of Medicine. Mayo Clinic, Rochester, MN, USA
| | | | | | - Thanh Thanh L. Nguyen
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, MN, USA
| | - Trudy J. Philips
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, MN, USA
| | - Hridyanshu Vyas
- Department of Cardiovascular Medicine, Mayo Clinic, Rochester, MN, USA
| | - Huanyao Gao
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, MN, USA
| | - Jyotan Sahni
- Department of Cardiovascular Medicine, Mayo Clinic, Rochester, MN, USA
| | | | - Leslie T. Cooper
- Department of Cardiovascular Medicine, Mayo Clinic, Jacksonville, FL, USA
| | - Michelle K. Skime
- Department of Psychiatry and Psychology, Mayo Clinic, Rochester, MN, USA
| | - Anthony Batzler
- Department of Health Sciences Research, Mayo Clinic, Rochester, MN, USA
| | | | - Simona Barlera
- Department of Cardiovascular Research, Istituto di Ricovero e Cura a Carattere Scientifico–Istituto di Ricerche Farmacologiche Mario Negri, Milan, Italy
| | - Silvana Pileggi
- Department of Cardiovascular Research, Istituto di Ricovero e Cura a Carattere Scientifico–Istituto di Ricerche Farmacologiche Mario Negri, Milan, Italy
| | - Luisa Mestroni
- Cardiovascular Institute, University of Colorado School of Medicine, Aurora, CO, USA
| | - Marco Merlo
- Cardiothoracovascular Department, Azienda Sanitaria Universitaria Giuliano Isontina (ASUGI), University of Trieste, Italy
| | - Gianfranco Sinagra
- Cardiothoracovascular Department, Azienda Sanitaria Universitaria Giuliano Isontina (ASUGI), University of Trieste, Italy
| | - Florence Pinet
- Univ. Lille, Inserm, CHU Lille, Institut Pasteur de Lille, U1167, Lille, France
| | - Jan Krejčí
- St. Anne’s University Hospital and Masaryk University, Brno, Czech Republic
| | - Anna Chaloupka
- St. Anne’s University Hospital and Masaryk University, Brno, Czech Republic
| | - Jordan D. Miller
- Department of Cardiovascular Surgery, Mayo Clinic, Rochester, MN, USA
| | - Pascal de Groote
- Univ. Lille, Inserm, CHU Lille, Institut Pasteur de Lille, U1167, Lille, France
- CHU Lille, Service de Cardiologie, Lille, France
| | | | - Richard M. Weinshilboum
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, MN, USA
| | - Naveen L. Pereira
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, MN, USA
- Department of Cardiovascular Medicine, Mayo Clinic, Rochester, MN, USA
| |
Collapse
|
16
|
Matsiukevich D, Kovacs A, Li T, Kokkonen-Simon K, Matkovich SJ, Oladipupo SS, Ornitz DM. Characterization of a robust mouse model of heart failure with preserved ejection fraction. Am J Physiol Heart Circ Physiol 2023; 325:H203-H231. [PMID: 37204871 PMCID: PMC11932539 DOI: 10.1152/ajpheart.00038.2023] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/23/2023] [Revised: 05/12/2023] [Accepted: 05/12/2023] [Indexed: 05/21/2023]
Abstract
Heart failure (HF) is a leading cause of morbidity and mortality particularly in older adults and patients with multiple metabolic comorbidities. Heart failure with preserved ejection fraction (HFpEF) is a clinical syndrome with multisystem organ dysfunction in which patients develop symptoms of HF as a result of high left ventricular (LV) diastolic pressure in the context of normal or near normal LV ejection fraction (LVEF; ≥50%). Challenges to create and reproduce a robust rodent phenotype that recapitulates the multiple comorbidities that exist in this syndrome explain the presence of various animal models that fail to satisfy all the criteria of HFpEF. Using a continuous infusion of angiotensin II and phenylephrine (ANG II/PE), we demonstrate a strong HFpEF phenotype satisfying major clinically relevant manifestations and criteria of this pathology, including exercise intolerance, pulmonary edema, concentric myocardial hypertrophy, diastolic dysfunction, histological signs of microvascular impairment, and fibrosis. Conventional echocardiographic analysis of diastolic dysfunction identified early stages of HFpEF development and speckle tracking echocardiography analysis including the left atrium (LA) identified strain abnormalities indicative of contraction-relaxation cycle impairment. Diastolic dysfunction was validated by retrograde cardiac catheterization and analysis of LV end-diastolic pressure (LVEDP). Among mice that developed HFpEF, two major subgroups were identified with predominantly perivascular fibrosis and interstitial myocardial fibrosis. In addition to major phenotypic criteria of HFpEF that were evident at early stages of this model (3 and 10 days), accompanying RNAseq data demonstrate activation of pathways associated with myocardial metabolic changes, inflammation, activation of extracellular matrix (ECM) deposition, microvascular rarefaction, and pressure- and volume-related myocardial stress.NEW & NOTEWORTHY Heart failure with preserved ejection fraction (HFpEF) is an emerging epidemic affecting up to half of patients with heart failure. Here we used a chronic angiotensin II/phenylephrine (ANG II/PE) infusion model and instituted an updated algorithm for HFpEF assessment. Given the simplicity in generating this model, it may become a useful tool for investigating pathogenic mechanisms, identification of diagnostic markers, and for drug discovery aimed at both prevention and treatment of HFpEF.
Collapse
Affiliation(s)
- Dzmitry Matsiukevich
- Department of Developmental Biology, Washington University in St. Louis School of Medicine, St. Louis, Missouri, United States
- Department of Pediatrics, Washington University in St. Louis School of Medicine, St. Louis, Missouri, United States
| | - Attila Kovacs
- Department of Medicine, Washington University in St. Louis School of Medicine, St. Louis, Missouri, United States
| | - Tiandao Li
- Department of Developmental Biology, Washington University in St. Louis School of Medicine, St. Louis, Missouri, United States
| | | | - Scot J Matkovich
- Eli Lilly and Company, Lilly Corporate Center, Indianapolis, Indiana, United States
| | - Sunday S Oladipupo
- Eli Lilly and Company, Lilly Corporate Center, Indianapolis, Indiana, United States
| | - David M Ornitz
- Department of Developmental Biology, Washington University in St. Louis School of Medicine, St. Louis, Missouri, United States
| |
Collapse
|
17
|
Abstract
PURPOSE OF REVIEW The intricate interplay between inflammatory and reparative responses in the context of heart injury is central to the pathogenesis of heart failure. Recent clinical studies have shown the therapeutic benefits of anti-inflammatory strategies in the treatment of cardiovascular diseases. This review provides a comprehensive overview of the cross-talk between immune cells and fibroblasts in the diseased heart. RECENT FINDINGS The role of inflammatory cells in fibroblast activation after cardiac injury is well-documented, but recent single-cell transcriptomics studies have identified putative pro-inflammatory fibroblasts in the infarcted heart, suggesting that fibroblasts, in turn, can modify inflammatory cell behavior. Furthermore, anti-inflammatory immune cells and fibroblasts have been described. The use of spatial and temporal-omics analyses may provide additional insights toward a better understanding of disease-specific microenvironments, where activated fibroblasts and inflammatory cells are in proximity. Recent studies focused on the interplay between fibroblasts and immune cells have brought us closer to the identification of cell type-specific targets for intervention. Further exploration of these intercellular communications will provide deeper insights toward the development of novel therapeutics.
Collapse
Affiliation(s)
- Akitoshi Hara
- Center for Cardiovascular Research, University of Hawaii at Manoa, Honolulu, HI, 96825, USA.
| | - Michelle D Tallquist
- Center for Cardiovascular Research, University of Hawaii at Manoa, Honolulu, HI, 96825, USA
| |
Collapse
|
18
|
Chhabra Y, Weeraratna AT. Fibroblasts in cancer: Unity in heterogeneity. Cell 2023; 186:1580-1609. [PMID: 37059066 PMCID: PMC11422789 DOI: 10.1016/j.cell.2023.03.016] [Citation(s) in RCA: 166] [Impact Index Per Article: 83.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2023] [Revised: 03/13/2023] [Accepted: 03/15/2023] [Indexed: 04/16/2023]
Abstract
Tumor cells do not exist in isolation in vivo, and carcinogenesis depends on the surrounding tumor microenvironment (TME), composed of a myriad of cell types and biophysical and biochemical components. Fibroblasts are integral in maintaining tissue homeostasis. However, even before a tumor develops, pro-tumorigenic fibroblasts in close proximity can provide the fertile 'soil' to the cancer 'seed' and are known as cancer-associated fibroblasts (CAFs). In response to intrinsic and extrinsic stressors, CAFs reorganize the TME enabling metastasis, therapeutic resistance, dormancy and reactivation by secreting cellular and acellular factors. In this review, we summarize the recent discoveries on CAF-mediated cancer progression with a particular focus on fibroblast heterogeneity and plasticity.
Collapse
Affiliation(s)
- Yash Chhabra
- Department of Biochemistry and Molecular Biology, Bloomberg School of Public Health, Department of Oncology, Sidney Kimmel Cancer Center, School of Medicine, Johns Hopkins University, Baltimore, MD 21205, USA.
| | - Ashani T Weeraratna
- Department of Biochemistry and Molecular Biology, Bloomberg School of Public Health, Department of Oncology, Sidney Kimmel Cancer Center, School of Medicine, Johns Hopkins University, Baltimore, MD 21205, USA.
| |
Collapse
|
19
|
Uscategui Calderon M, Gonzalez BA, Yutzey KE. Cardiomyocyte-fibroblast crosstalk in the postnatal heart. Front Cell Dev Biol 2023; 11:1163331. [PMID: 37077417 PMCID: PMC10106698 DOI: 10.3389/fcell.2023.1163331] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2023] [Accepted: 03/22/2023] [Indexed: 04/05/2023] Open
Abstract
During the postnatal period in mammals, the heart undergoes significant remodeling in response to increased circulatory demands. In the days after birth, cardiac cells, including cardiomyocytes and fibroblasts, progressively lose embryonic characteristics concomitant with the loss of the heart’s ability to regenerate. Moreover, postnatal cardiomyocytes undergo binucleation and cell cycle arrest with induction of hypertrophic growth, while cardiac fibroblasts proliferate and produce extracellular matrix (ECM) that transitions from components that support cellular maturation to production of the mature fibrous skeleton of the heart. Recent studies have implicated interactions of cardiac fibroblasts and cardiomyocytes within the maturing ECM environment to promote heart maturation in the postnatal period. Here, we review the relationships of different cardiac cell types and the ECM as the heart undergoes both structural and functional changes during development. Recent advances in the field, particularly in several recently published transcriptomic datasets, have highlighted specific signaling mechanisms that underlie cellular maturation and demonstrated the biomechanical interdependence of cardiac fibroblast and cardiomyocyte maturation. There is increasing evidence that postnatal heart development in mammals is dependent on particular ECM components and that resulting changes in biomechanics influence cell maturation. These advances, in definition of cardiac fibroblast heterogeneity and function in relation to cardiomyocyte maturation and the extracellular environment provide, support for complex cell crosstalk in the postnatal heart with implications for heart regeneration and disease mechanisms.
Collapse
Affiliation(s)
- Maria Uscategui Calderon
- Division of Molecular Cardiovascular Biology, The Heart Institute, Cincinnati Children’s Medical Center, Cincinnati, OH, United States
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, United States
| | - Brittany A. Gonzalez
- Division of Molecular Cardiovascular Biology, The Heart Institute, Cincinnati Children’s Medical Center, Cincinnati, OH, United States
| | - Katherine E. Yutzey
- Division of Molecular Cardiovascular Biology, The Heart Institute, Cincinnati Children’s Medical Center, Cincinnati, OH, United States
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, United States
- *Correspondence: Katherine E. Yutzey,
| |
Collapse
|
20
|
Yin X, Yin X, Pan X, Zhang J, Fan X, Li J, Zhai X, Jiang L, Hao P, Wang J, Chen Y. Post-myocardial infarction fibrosis: Pathophysiology, examination, and intervention. Front Pharmacol 2023; 14:1070973. [PMID: 37056987 PMCID: PMC10086160 DOI: 10.3389/fphar.2023.1070973] [Citation(s) in RCA: 27] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2022] [Accepted: 03/13/2023] [Indexed: 03/30/2023] Open
Abstract
Cardiac fibrosis plays an indispensable role in cardiac tissue homeostasis and repair after myocardial infarction (MI). The cardiac fibroblast-to-myofibroblast differentiation and extracellular matrix collagen deposition are the hallmarks of cardiac fibrosis, which are modulated by multiple signaling pathways and various types of cells in time-dependent manners. Our understanding of the development of cardiac fibrosis after MI has evolved in basic and clinical researches, and the regulation of fibrotic remodeling may facilitate novel diagnostic and therapeutic strategies, and finally improve outcomes. Here, we aim to elaborate pathophysiology, examination and intervention of cardiac fibrosis after MI.
Collapse
Affiliation(s)
- Xiaoying Yin
- Department of Emergency and Chest Pain Center, Qilu Hospital of Shandong University, Jinan, China
- Clinical Research Center for Emergency and Critical Care Medicine of Shandong Province, Institute of Emergency and Critical Care Medicine of Shandong University, Qilu Hospital of Shandong University, Jinan, China
- Key Laboratory of Emergency and Critical Care Medicine of Shandong Province, Qilu Hospital of Shandong University, Jinan, China
- Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese Ministry of Health and Chinese Academy of Medical Sciences, The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Qilu Hospital of Shandong University, Jinan, China
| | - Xinxin Yin
- Department of Emergency and Chest Pain Center, Qilu Hospital of Shandong University, Jinan, China
- Clinical Research Center for Emergency and Critical Care Medicine of Shandong Province, Institute of Emergency and Critical Care Medicine of Shandong University, Qilu Hospital of Shandong University, Jinan, China
- Key Laboratory of Emergency and Critical Care Medicine of Shandong Province, Qilu Hospital of Shandong University, Jinan, China
- Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese Ministry of Health and Chinese Academy of Medical Sciences, The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Qilu Hospital of Shandong University, Jinan, China
| | - Xin Pan
- Department of Emergency and Chest Pain Center, Qilu Hospital of Shandong University, Jinan, China
- Clinical Research Center for Emergency and Critical Care Medicine of Shandong Province, Institute of Emergency and Critical Care Medicine of Shandong University, Qilu Hospital of Shandong University, Jinan, China
- Key Laboratory of Emergency and Critical Care Medicine of Shandong Province, Qilu Hospital of Shandong University, Jinan, China
- Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese Ministry of Health and Chinese Academy of Medical Sciences, The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Qilu Hospital of Shandong University, Jinan, China
| | - Jingyu Zhang
- Department of Emergency and Chest Pain Center, Qilu Hospital of Shandong University, Jinan, China
- Clinical Research Center for Emergency and Critical Care Medicine of Shandong Province, Institute of Emergency and Critical Care Medicine of Shandong University, Qilu Hospital of Shandong University, Jinan, China
- Key Laboratory of Emergency and Critical Care Medicine of Shandong Province, Qilu Hospital of Shandong University, Jinan, China
- Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese Ministry of Health and Chinese Academy of Medical Sciences, The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Qilu Hospital of Shandong University, Jinan, China
| | - Xinhui Fan
- Department of Emergency and Chest Pain Center, Qilu Hospital of Shandong University, Jinan, China
- Clinical Research Center for Emergency and Critical Care Medicine of Shandong Province, Institute of Emergency and Critical Care Medicine of Shandong University, Qilu Hospital of Shandong University, Jinan, China
- Key Laboratory of Emergency and Critical Care Medicine of Shandong Province, Qilu Hospital of Shandong University, Jinan, China
- Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese Ministry of Health and Chinese Academy of Medical Sciences, The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Qilu Hospital of Shandong University, Jinan, China
| | - Jiaxin Li
- Department of Emergency and Chest Pain Center, Qilu Hospital of Shandong University, Jinan, China
- Clinical Research Center for Emergency and Critical Care Medicine of Shandong Province, Institute of Emergency and Critical Care Medicine of Shandong University, Qilu Hospital of Shandong University, Jinan, China
- Key Laboratory of Emergency and Critical Care Medicine of Shandong Province, Qilu Hospital of Shandong University, Jinan, China
- Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese Ministry of Health and Chinese Academy of Medical Sciences, The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Qilu Hospital of Shandong University, Jinan, China
| | - Xiaoxuan Zhai
- Department of Emergency and Chest Pain Center, Qilu Hospital of Shandong University, Jinan, China
- Clinical Research Center for Emergency and Critical Care Medicine of Shandong Province, Institute of Emergency and Critical Care Medicine of Shandong University, Qilu Hospital of Shandong University, Jinan, China
- Key Laboratory of Emergency and Critical Care Medicine of Shandong Province, Qilu Hospital of Shandong University, Jinan, China
- Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese Ministry of Health and Chinese Academy of Medical Sciences, The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Qilu Hospital of Shandong University, Jinan, China
| | - Lijun Jiang
- Department of Emergency and Chest Pain Center, Qilu Hospital of Shandong University, Jinan, China
- Clinical Research Center for Emergency and Critical Care Medicine of Shandong Province, Institute of Emergency and Critical Care Medicine of Shandong University, Qilu Hospital of Shandong University, Jinan, China
- Key Laboratory of Emergency and Critical Care Medicine of Shandong Province, Qilu Hospital of Shandong University, Jinan, China
- Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese Ministry of Health and Chinese Academy of Medical Sciences, The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Qilu Hospital of Shandong University, Jinan, China
| | - Panpan Hao
- Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese Ministry of Health and Chinese Academy of Medical Sciences, The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Qilu Hospital of Shandong University, Jinan, China
| | - Jiali Wang
- Department of Emergency and Chest Pain Center, Qilu Hospital of Shandong University, Jinan, China
- Clinical Research Center for Emergency and Critical Care Medicine of Shandong Province, Institute of Emergency and Critical Care Medicine of Shandong University, Qilu Hospital of Shandong University, Jinan, China
- Key Laboratory of Emergency and Critical Care Medicine of Shandong Province, Qilu Hospital of Shandong University, Jinan, China
- Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese Ministry of Health and Chinese Academy of Medical Sciences, The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Qilu Hospital of Shandong University, Jinan, China
| | - Yuguo Chen
- Department of Emergency and Chest Pain Center, Qilu Hospital of Shandong University, Jinan, China
- Clinical Research Center for Emergency and Critical Care Medicine of Shandong Province, Institute of Emergency and Critical Care Medicine of Shandong University, Qilu Hospital of Shandong University, Jinan, China
- Key Laboratory of Emergency and Critical Care Medicine of Shandong Province, Qilu Hospital of Shandong University, Jinan, China
- Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese Ministry of Health and Chinese Academy of Medical Sciences, The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Qilu Hospital of Shandong University, Jinan, China
| |
Collapse
|
21
|
Bretherton RC, Reichardt IM, Zabrecky KA, Goldstein AJ, Bailey LRJ, Bugg D, McMillen TS, Kooiker KB, Flint GV, Martinson A, Gunaje J, Koser F, Plaster E, Linke WA, Regnier M, Moussavi-Harami F, Sniadecki NJ, DeForest CA, Davis J. Correcting dilated cardiomyopathy with fibroblast-targeted p38 deficiency. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.01.23.523684. [PMID: 36747691 PMCID: PMC9900749 DOI: 10.1101/2023.01.23.523684] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
Inherited mutations in contractile and structural genes, which decrease cardiomyocyte tension generation, are principal drivers of dilated cardiomyopathy (DCM)- the leading cause of heart failure 1,2 . Progress towards developing precision therapeutics for and defining the underlying determinants of DCM has been cardiomyocyte centric with negligible attention directed towards fibroblasts despite their role in regulating the best predictor of DCM severity, cardiac fibrosis 3,4 . Given that failure to reverse fibrosis is a major limitation of both standard of care and first in class precision therapeutics for DCM, this study examined whether cardiac fibroblast-mediated regulation of the heart's material properties is essential for the DCM phenotype. Here we report in a mouse model of inherited DCM that prior to the onset of fibrosis and dilated myocardial remodeling both the myocardium and extracellular matrix (ECM) stiffen from switches in titin isoform expression, enhanced collagen fiber alignment, and expansion of the cardiac fibroblast population, which we blocked by genetically suppressing p38α in cardiac fibroblasts. This fibroblast-targeted intervention unexpectedly improved the primary cardiomyocyte defect in contractile function and reversed ECM and dilated myocardial remodeling. Together these findings challenge the long-standing paradigm that ECM remodeling is a secondary complication to inherited defects in cardiomyocyte contractile function and instead demonstrate cardiac fibroblasts are essential contributors to the DCM phenotype, thus suggesting DCM-specific therapeutics will require fibroblast-specific strategies.
Collapse
|