1
|
Lan T, Kaminsky S, Wu CC. Ploidy in cardiovascular development and regeneration. Semin Cell Dev Biol 2025; 172:103618. [PMID: 40398363 DOI: 10.1016/j.semcdb.2025.103618] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2024] [Revised: 04/01/2025] [Accepted: 05/05/2025] [Indexed: 05/23/2025]
Abstract
Somatic polyploidy, a non-inheritable form of genome multiplication, plays cell-type specific and context-dependent roles in organ development and regeneration. In the mammalian heart, embryonic cardiomyocytes are primarily diploid, which lose their ability to complete cell division and become polyploid as they mature. Unlike lower vertebrates like zebrafish, polyploid cardiomyocytes are commonly found across mammals, including humans. Intriguingly, the degree, timing, and modes of cardiomyocyte polyploidization vary greatly between species. In addition to the association with cardiomyocyte development and maturation, recent studies have established polyploidy as a barrier against cardiomyocyte proliferation and heart regeneration following cardiac injury. Hence, a thorough understanding of how and why cardiomyocyte become polyploid will provide insights into heart development and may help develop therapeutic strategies for heart regeneration. Here, we review the dynamics of cardiomyocyte polyploidization across species and how cardiomyocyte-intrinsic, -extrinsic, and environmental factors regulate this process as well as the impact of cardiomyocyte polyploidization on heart development and regeneration.
Collapse
Affiliation(s)
- Tian Lan
- Heidelberg University, Medical Faculty Mannheim, European Center for Angioscience, Mannheim, Germany; Helmholtz-Institute for Translational AngioCardioScience (HI-TAC) of the Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC) at Heidelberg University
| | - Sabrina Kaminsky
- Heidelberg University, Medical Faculty Mannheim, European Center for Angioscience, Mannheim, Germany; Faculty of Biosciences, Heidelberg University, Germany
| | - Chi-Chung Wu
- Heidelberg University, Medical Faculty Mannheim, European Center for Angioscience, Mannheim, Germany; Helmholtz-Institute for Translational AngioCardioScience (HI-TAC) of the Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC) at Heidelberg University.
| |
Collapse
|
2
|
Liu T, Guo X, Fu Y, Zhang W. SLAMF9 aggravates myocardial ischemia reperfusion injury through activating the hippo-yap pathway. Biochim Biophys Acta Gen Subj 2025; 1869:130821. [PMID: 40383314 DOI: 10.1016/j.bbagen.2025.130821] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2025] [Revised: 04/23/2025] [Accepted: 05/15/2025] [Indexed: 05/20/2025]
Abstract
BACKGROUND The objective was to investigate the impact of signaling lymphocyte activation molecule family member 9 (SLAMF9) on myocardial infarction (MI) and its mechanisms. METHODS SLAMF9 expression in MI rats was firstly measured. SLAMF9 effect on cardiac functions, myocardial fibrosis, cardiomyocyte hypertrophy, cardiomyocyte apoptosis and inflammation in MI rats was explored using echocardiography, HE staining, masson staining, wheat germ agglutinin staining, western blot, TUNEL staining and qRT-PCR. Meanwhile, SLAMF9 effect on the viability, apoptosis, and inflammation in H9C2 cells was investigated by CCK-8 assay, TUNEL staining and western blot. Moreover, the potential mechanisms of SLAMF9 were investigated using western blot, ELISA and TUNEL staining after different treatment. RESULTS SLAMF9 expression was upregulated in MI rats. SLAMF9 knockdown ameliorated heart damage, cardiomyocyte apoptosis and inflammatory response in MI rats. Similarly, SLAMF9 silencing in macrophages attenuated the apoptosis and inflammatory response in H/R-induced H9C2 cells. Moreover, SLAMF9 knockdown inhibited Hippo-Yap pathway in MI in vitro and in vivo. Besides, SLAMF9 knockdown in macrophages suppressed the activation of Hippo-Yap pathway in H9C2 cells by inhibiting TNF-α release. Additionally, LATS1 overexpression in H9C2 cells reversed the effect of SLAMF9 silencing on the apoptosis and inflammatory response in H/R-induced H9C2 cells. Meanwhile, PY-60 treatment in H9C2 cells reversed the effect of SLAMF9 overexpression on the apoptosis and inflammatory response in H/R-induced H9C2 cells. CONCLUSION The absence of SLAMF9 led to a reduction in TNF-α secretion in macrophages, consequently repressing Hippo-Yap pathway in cardiomyocytes, and ultimately ameliorating myocardial damage, cardiomyocyte apoptosis and inflammation in MI.
Collapse
Affiliation(s)
- Tingting Liu
- Cardiovascular Department, Yantaishan Hospital, Yantai 264003, Shandong, China
| | - Xiuli Guo
- Geriatrics Department, Yantaishan Hospital, Yantai 264003, Shandong, China
| | - Yanhua Fu
- Geriatrics Department, Yantaishan Hospital, Yantai 264003, Shandong, China
| | - Weili Zhang
- Geriatrics Department, Yantaishan Hospital, Yantai 264003, Shandong, China.
| |
Collapse
|
3
|
Wu B, Constanty F, Beisaw A. Cardiac regeneration: Unraveling the complex network of intercellular crosstalk. Semin Cell Dev Biol 2025; 171:103619. [PMID: 40367899 DOI: 10.1016/j.semcdb.2025.103619] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2024] [Revised: 03/13/2025] [Accepted: 05/06/2025] [Indexed: 05/16/2025]
Abstract
The heart is composed of multiple cell types, including cardiomyocytes, endothelial/endocardial cells, fibroblasts, resident immune cells and epicardium and crosstalk between these cell types is crucial for proper cardiac function and homeostasis. In response to cardiac injury or disease, cell-cell interactions and intercellular crosstalk contribute to remodeling to compensate reduced heart function. In some vertebrates, the heart can regenerate following cardiac injury. While cardiomyocytes play a crucial role in this process, additional cell types are necessary to create a pro-regenerative microenvironment in the injured heart. Here, we review recent literature regarding the importance of cellular crosstalk in promoting cardiac regeneration and provide insight into emerging technologies to investigate cell-cell interactions in vivo. Lastly, we explore recent studies highlighting the importance of inter-organ communication in response to injury and promotion of cardiac regeneration. Importantly, understanding how intercellular and inter-organ crosstalk promote cardiac regeneration is essential for the development of therapeutic strategies to stimulate regeneration in the human heart.
Collapse
Affiliation(s)
- Bailin Wu
- Institute of Experimental Cardiology, Heidelberg University, Heidelberg, Germany; German Centre for Cardiovascular Research (DZHK), Heidelberg/Mannheim partner site, Germany
| | - Florian Constanty
- Institute of Experimental Cardiology, Heidelberg University, Heidelberg, Germany; German Centre for Cardiovascular Research (DZHK), Heidelberg/Mannheim partner site, Germany; Helmholtz-Institute for Translational AngioCardioScience (HI-TAC) of the Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC) at Heidelberg University, Heidelberg 69117, Germany
| | - Arica Beisaw
- Institute of Experimental Cardiology, Heidelberg University, Heidelberg, Germany; German Centre for Cardiovascular Research (DZHK), Heidelberg/Mannheim partner site, Germany; Helmholtz-Institute for Translational AngioCardioScience (HI-TAC) of the Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC) at Heidelberg University, Heidelberg 69117, Germany.
| |
Collapse
|
4
|
Xu Q, Chen X, Zhao C, Liu Y, Wang J, Ao X, Ding W. Cell cycle arrest of cardiomyocytes in the context of cardiac regeneration. Front Cardiovasc Med 2025; 12:1538546. [PMID: 40357436 PMCID: PMC12066773 DOI: 10.3389/fcvm.2025.1538546] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2024] [Accepted: 04/14/2025] [Indexed: 05/15/2025] Open
Abstract
The limited capacity of adult mammalian cardiomyocytes to undergo cell division and proliferation is one of the key factors contributing to heart failure. In newborn mice, cardiac proliferation occurs during a brief window, but this proliferative capacity diminishes by 7 days after birth. Current studies on cardiac regeneration focused on elucidating changes in regulatory factors within the heart before and after this proliferative window, aiming to determine whether potential association between these factors and cell cycle arrest in cardiomyocytes. Facilitating the re-entry of cardiomyocytes into the cell cycle or reversing their exit from it represents a critical strategy for cardiac regeneration. This paper provides an overview of the role of cell cycle arrest in cardiac regeneration, briefly describes cardiomyocyte proliferation and cardiac regeneration, and systematically summarizes the regulation of the cell cycle arrest in cardiomyocytes, and the potential metabolic mechanisms underlying cardiomyocyte cycle arrest. Additionally, we highlight the development of cardiovascular disease drugs targeting cardiomyocyte cell cycle regulation and their status in clinical treatment. Our goal is to outline strategies for promoting cardiac regeneration and repair following cardiac injury, while also pointing toward future research directions that may offer new technologies and prospects for treating cardiovascular diseases, such as myocardial infarction, arrhythmia and heart failure.
Collapse
Affiliation(s)
- Qingling Xu
- Department of Comprehensive Internal Medicine, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
- School of Basic Medicine, Qingdao University, Qingdao, Shandong, China
| | - Xinhui Chen
- School of Basic Medicine, Qingdao University, Qingdao, Shandong, China
| | - Chunyige Zhao
- School of Basic Medicine, Qingdao University, Qingdao, Shandong, China
| | - Ying Liu
- Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, Qingdao Medical College, Qingdao University, Qingdao, Shandong, China
| | - Jianxun Wang
- School of Basic Medicine, Qingdao University, Qingdao, Shandong, China
| | - Xiang Ao
- Department of Comprehensive Internal Medicine, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
- School of Basic Medicine, Qingdao University, Qingdao, Shandong, China
| | - Wei Ding
- Department of Comprehensive Internal Medicine, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
| |
Collapse
|
5
|
Constanty F, Wu B, Wei KH, Lin IT, Dallmann J, Guenther S, Lautenschlaeger T, Priya R, Lai SL, Stainier DYR, Beisaw A. Border-zone cardiomyocytes and macrophages regulate extracellular matrix remodeling to promote cardiomyocyte protrusion during cardiac regeneration. Nat Commun 2025; 16:3823. [PMID: 40268967 PMCID: PMC12019606 DOI: 10.1038/s41467-025-59169-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2024] [Accepted: 04/14/2025] [Indexed: 04/25/2025] Open
Abstract
Despite numerous advances in our understanding of zebrafish cardiac regeneration, an aspect that remains less studied is how regenerating cardiomyocytes invade and replace the collagen-containing injured tissue. Here, we provide an in-depth analysis of the process of cardiomyocyte invasion. We observe close interactions between protruding border-zone cardiomyocytes and macrophages, and show that macrophages are essential for extracellular matrix remodeling at the wound border zone and cardiomyocyte protrusion into the injured area. Single-cell RNA-sequencing reveals the expression of mmp14b, encoding a membrane-anchored matrix metalloproteinase, in several cell types at the border zone. Genetic mmp14b mutation leads to decreased macrophage recruitment, collagen degradation, and subsequent cardiomyocyte protrusion into injured tissue. Furthermore, cardiomyocyte-specific overexpression of mmp14b is sufficient to enhance cardiomyocyte invasion into the injured tissue and along the apical surface of the wound. Altogether, our data provide important insights into the mechanisms underlying cardiomyocyte invasion of the collagen-containing injured tissue during cardiac regeneration.
Collapse
Affiliation(s)
- Florian Constanty
- Mechanisms of Cardiac Regeneration and Repair Lab, Institute of Experimental Cardiology, Heidelberg University, Heidelberg, Germany
- Helmholtz-Institute for Translational AngioCardioScience (HI-TAC) of the Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC) at Heidelberg University, Heidelberg, Germany
- DZHK (German Centre for Cardiovascular Research), Partner site Heidelberg/Mannheim, Heidelberg, Germany
| | - Bailin Wu
- Mechanisms of Cardiac Regeneration and Repair Lab, Institute of Experimental Cardiology, Heidelberg University, Heidelberg, Germany
- DZHK (German Centre for Cardiovascular Research), Partner site Heidelberg/Mannheim, Heidelberg, Germany
| | - Ke-Hsuan Wei
- Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan
| | - I-Ting Lin
- Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan
| | - Julia Dallmann
- Department of Developmental Genetics, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany
| | - Stefan Guenther
- Deep Sequencing Platform, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany
- DZHK (German Centre for Cardiovascular Research), Partner site Rhein/Main, Rhein/Main, Germany
| | - Till Lautenschlaeger
- Department of Developmental Genetics, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany
| | - Rashmi Priya
- Department of Developmental Genetics, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany
- Cardio-Pulmonary Institute, Bad Nauheim, Germany
- The Francis Crick Institute, London, UK
| | - Shih-Lei Lai
- Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan
| | - Didier Y R Stainier
- Department of Developmental Genetics, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany
- DZHK (German Centre for Cardiovascular Research), Partner site Rhein/Main, Rhein/Main, Germany
- Cardio-Pulmonary Institute, Bad Nauheim, Germany
| | - Arica Beisaw
- Mechanisms of Cardiac Regeneration and Repair Lab, Institute of Experimental Cardiology, Heidelberg University, Heidelberg, Germany.
- Helmholtz-Institute for Translational AngioCardioScience (HI-TAC) of the Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC) at Heidelberg University, Heidelberg, Germany.
- DZHK (German Centre for Cardiovascular Research), Partner site Heidelberg/Mannheim, Heidelberg, Germany.
| |
Collapse
|
6
|
Abou Nader N, Jakuc N, Meinsohn MC, Charrier L, Banville L, Brind’Amour J, Paquet M, St-Jean G, Boerboom D, Mao J, Pépin D, Breault DT, Zamberlam G, Boyer A. Hippo Signaling Is Essential for the Maintenance of Zona Glomerulosa Cell Fate in the Murine Adrenal Cortex. Endocrinology 2025; 166:bqaf077. [PMID: 40233139 PMCID: PMC12041920 DOI: 10.1210/endocr/bqaf077] [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: 12/05/2024] [Revised: 03/14/2025] [Accepted: 04/14/2025] [Indexed: 04/17/2025]
Abstract
Cells of the zona glomerulosa (zG), the outermost zone of the adrenal cortex, secrete aldosterone and transdifferentiate into glucocorticoid-producing cells of the zona fasciculata (zF) during adrenal homeostasis. However, our understanding of the signaling pathways mediating zG cell maintenance or their transdifferentiation into zF cells is incomplete. Hippo is a major pathway that regulates cell proliferation/differentiation during embryogenesis and postnatal tissue homeostasis. Hypothesizing that Hippo signaling could be involved in zG cell maintenance or transdifferentiation, we generated a mouse model in which the two main kinases of the Hippo signaling cascade large tumor suppressor homolog kinases 1/2 (Lats1 and Lats2) are specifically inactivated in zG cells. Here we show that loss of function of Lats1 and Lats2 impairs zG steroidogenesis and leads to zG cell transdifferentiation into cells sharing characteristics with chondroblasts/osteoblasts rather than zF cells. Furthermore, we demonstrate that this phenotype can be rescued by the concomitant inactivation of the transcriptional coactivators Yes-associated protein (Yap) and transcriptional coactivator with PDZ-binding motif (Taz) with Lats1 and Lats2. Finally, we show that expression of a constitutively active form of YAP (YAP5SA) in zG cells does not alter their fate as severely as the loss of Lats1 and Lats2 but leads to adrenal hyperplasia. Together, these findings highlight the critical role of Hippo signaling in maintaining zG cell fate and function and provide key insights into broader mechanisms underlying cellular differentiation.
Collapse
Affiliation(s)
- Nour Abou Nader
- Centre de Recherche en Reproduction et Fertilité, Faculté de Médecine Vétérinaire, Université de Montréal, Saint-Hyacinthe, QC J2S 2M2, Canada
- Département de Biomédecine Vétérinaire, Faculté de Médecine Vétérinaire, Université de Montréal, Saint-Hyacinthe, QC J2S 2M2, Canada
| | - Natalia Jakuc
- Centre de Recherche en Reproduction et Fertilité, Faculté de Médecine Vétérinaire, Université de Montréal, Saint-Hyacinthe, QC J2S 2M2, Canada
- Département de Biomédecine Vétérinaire, Faculté de Médecine Vétérinaire, Université de Montréal, Saint-Hyacinthe, QC J2S 2M2, Canada
| | | | - Laureline Charrier
- Centre de Recherche en Reproduction et Fertilité, Faculté de Médecine Vétérinaire, Université de Montréal, Saint-Hyacinthe, QC J2S 2M2, Canada
- Département de Biomédecine Vétérinaire, Faculté de Médecine Vétérinaire, Université de Montréal, Saint-Hyacinthe, QC J2S 2M2, Canada
| | - Laurence Banville
- Centre de Recherche en Reproduction et Fertilité, Faculté de Médecine Vétérinaire, Université de Montréal, Saint-Hyacinthe, QC J2S 2M2, Canada
- Département de Pathologie et de Microbiologie, Faculté de Médecine Vétérinaire, Université de Montréal, Saint-Hyacinthe, QC J2S 2M2, Canada
| | - Julie Brind’Amour
- Centre de Recherche en Reproduction et Fertilité, Faculté de Médecine Vétérinaire, Université de Montréal, Saint-Hyacinthe, QC J2S 2M2, Canada
- Département de Biomédecine Vétérinaire, Faculté de Médecine Vétérinaire, Université de Montréal, Saint-Hyacinthe, QC J2S 2M2, Canada
| | - Marilène Paquet
- Centre de Recherche en Reproduction et Fertilité, Faculté de Médecine Vétérinaire, Université de Montréal, Saint-Hyacinthe, QC J2S 2M2, Canada
- Département de Pathologie et de Microbiologie, Faculté de Médecine Vétérinaire, Université de Montréal, Saint-Hyacinthe, QC J2S 2M2, Canada
| | - Guillaume St-Jean
- Centre de Recherche en Reproduction et Fertilité, Faculté de Médecine Vétérinaire, Université de Montréal, Saint-Hyacinthe, QC J2S 2M2, Canada
- Département de Pathologie et de Microbiologie, Faculté de Médecine Vétérinaire, Université de Montréal, Saint-Hyacinthe, QC J2S 2M2, Canada
| | - Derek Boerboom
- Centre de Recherche en Reproduction et Fertilité, Faculté de Médecine Vétérinaire, Université de Montréal, Saint-Hyacinthe, QC J2S 2M2, Canada
- Département de Biomédecine Vétérinaire, Faculté de Médecine Vétérinaire, Université de Montréal, Saint-Hyacinthe, QC J2S 2M2, Canada
| | - Junhao Mao
- Department of Molecular, Cell and Cancer Biology, University of Massachusetts Chan Medical School, Worcester, MA 01655, USA
| | - David Pépin
- Pediatric Surgical Research Laboratories, Massachusetts General Hospital, Boston, MA 02114, USA
| | - David T Breault
- Division of Endocrinology, Boston Children’s Hospital, Harvard Medical School, Boston, MA 02115, USA
- Harvard Stem Cell Institute, Cambridge, MA 02138, USA
| | - Gustavo Zamberlam
- Centre de Recherche en Reproduction et Fertilité, Faculté de Médecine Vétérinaire, Université de Montréal, Saint-Hyacinthe, QC J2S 2M2, Canada
- Département de Biomédecine Vétérinaire, Faculté de Médecine Vétérinaire, Université de Montréal, Saint-Hyacinthe, QC J2S 2M2, Canada
| | - Alexandre Boyer
- Centre de Recherche en Reproduction et Fertilité, Faculté de Médecine Vétérinaire, Université de Montréal, Saint-Hyacinthe, QC J2S 2M2, Canada
- Département de Biomédecine Vétérinaire, Faculté de Médecine Vétérinaire, Université de Montréal, Saint-Hyacinthe, QC J2S 2M2, Canada
| |
Collapse
|
7
|
Koopmans T, van Rooij E. Molecular gatekeepers of endogenous adult mammalian cardiomyocyte proliferation. Nat Rev Cardiol 2025:10.1038/s41569-025-01145-y. [PMID: 40195566 DOI: 10.1038/s41569-025-01145-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 03/10/2025] [Indexed: 04/09/2025]
Abstract
Irreversible cardiac fibrosis, cardiomyocyte death and chronic cardiac dysfunction after myocardial infarction pose a substantial global health-care challenge, with no curative treatments available. To regenerate the injured heart, cardiomyocytes must proliferate to replace lost myocardial tissue - a capability that adult mammals have largely forfeited to adapt to the demanding conditions of life. Using various preclinical models, our understanding of cardiomyocyte proliferation has progressed remarkably, leading to the successful reactivation of cell cycle induction in adult animals, with functional recovery after cardiac injury. Central to this success is the targeting of key pathways and structures that drive cardiomyocyte maturation after birth - nucleation and ploidy, sarcomere structure, developmental signalling, chromatin and epigenetic regulation, the microenvironment and metabolic maturation - forming a complex regulatory framework that allows efficient cellular contraction but restricts cardiomyocyte proliferation. In this Review, we explore the molecular pathways underlying these core mechanisms and how their manipulation can reactivate the cell cycle in cardiomyocytes, potentially contributing to cardiac repair.
Collapse
Affiliation(s)
- Tim Koopmans
- Hubrecht Institute, Royal Netherlands Academy of Arts and Sciences (KNAW) and University Medical Center Utrecht, Utrecht, Netherlands
| | - Eva van Rooij
- Hubrecht Institute, Royal Netherlands Academy of Arts and Sciences (KNAW) and University Medical Center Utrecht, Utrecht, Netherlands.
- Department of Cardiology, University Medical Center Utrecht, Utrecht, Netherlands.
| |
Collapse
|
8
|
Almeida M, Inácio JM, Vital CM, Rodrigues MR, Araújo BC, Belo JA. Cell Reprogramming, Transdifferentiation, and Dedifferentiation Approaches for Heart Repair. Int J Mol Sci 2025; 26:3063. [PMID: 40243729 PMCID: PMC11988544 DOI: 10.3390/ijms26073063] [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: 03/01/2025] [Revised: 03/22/2025] [Accepted: 03/24/2025] [Indexed: 04/18/2025] Open
Abstract
Cardiovascular disease (CVD) remains the leading cause of death globally, with myocardial infarction (MI) being a major contributor. The current therapeutic approaches are limited in effectively regenerating damaged cardiac tissue. Up-to-date strategies for heart regeneration/reconstitution aim at cardiac remodeling through repairing the damaged tissue with an external cell source or by stimulating the existing cells to proliferate and repopulate the compromised area. Cell reprogramming is addressed to this challenge as a promising solution, converting fibroblasts and other cell types into functional cardiomyocytes, either by reverting cells to a pluripotent state or by directly switching cell lineage. Several strategies such as gene editing and the application of miRNA and small molecules have been explored for their potential to enhance cardiac regeneration. Those strategies take advantage of cell plasticity by introducing reprogramming factors that regress cell maturity in vitro, allowing for their later differentiation and thus endorsing cell transplantation, or promote in situ cell proliferation, leveraged by scaffolds embedded with pro-regenerative factors promoting efficient heart restoration. Despite notable advancements, important challenges persist, including low reprogramming efficiency, cell maturation limitations, and safety concerns in clinical applications. Nonetheless, integrating these innovative approaches offers a promising alternative for restoring cardiac function and reducing the dependency on full heart transplants.
Collapse
Affiliation(s)
| | - José M. Inácio
- Stem Cells and Development Laboratory, iNOVA4Health, NOVA Medical School|Faculdade de Ciências Médicas, Universidade NOVA de Lisboa, 1169-056 Lisbon, Portugal; (M.A.); (C.M.V.); (M.R.R.); (B.C.A.)
| | | | | | | | - José A. Belo
- Stem Cells and Development Laboratory, iNOVA4Health, NOVA Medical School|Faculdade de Ciências Médicas, Universidade NOVA de Lisboa, 1169-056 Lisbon, Portugal; (M.A.); (C.M.V.); (M.R.R.); (B.C.A.)
| |
Collapse
|
9
|
Meng Y, Ding J, Wang Y, Wang J, Huang W, Jiang W, Li J, Lang X, Zhang S, Liu Y, Liu X, Li H, Sun B. The transcriptional repressor Ctbp2 as a metabolite sensor regulating cardiomyocytes proliferation and heart regeneration. Mol Med 2025; 31:119. [PMID: 40140769 PMCID: PMC11948641 DOI: 10.1186/s10020-025-01168-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2024] [Accepted: 03/13/2025] [Indexed: 03/28/2025] Open
Abstract
BACKGROUND C-terminal binding protein-2 (Ctbp2) is an evolutionarily conserved transcriptional repressor that regulates fundamental processes such as cell proliferation and apoptosis. However, the potential role of Ctbp2 in cardiomyocyte proliferation and heart regeneration remains unclear. In this study, we aim to explore the important role of Ctbp2 in cardiomyocyte proliferation and the regeneration of injured adult hearts. METHODS AND RESULTS In this study, we found that the expression of Ctbp2 in cardiomyocytes is downregulated after adulthood. Silencing Ctbp2 in cardiomyocytes on the post-natal day 1 (P1) reduced the proliferation ability of cardiomyocytes, whereas overexpressing Ctbp2 enhanced the proliferation ability of cardiomyocytes. Additionally, overexpressing Ctbp2 via adeno-associated virus-9 (AAV9) had no effect on the hearts of normal adult mice, but in the case of heart injury, overexpression of Ctbp2 in adult mice cardiomyocytes promoted cardiomyocyte proliferation. Mechanistically, the transcriptional repressor Ctbp2 acts as a metabolite sensor, and its regulation of cardiomyocyte proliferation is influenced by the metabolites NADH/NAD+ and fatty acyl-CoAs. Ctbp2 is activated by the intracellular accumulation of NADH during cardiomyocyte ischemia and hypoxia, inhibiting the transcriptional activity of the transcription factor FoxO1, thereby repressing the expression of the target genes and cell cycle negative regulators p21 and p27, allowing cardiomyocytes to re-enter the cell cycle. In contrast, normal adult cardiomyocytes mainly use fatty acid oxidation metabolism as their primary energy source, and the intracellular production of fatty acyl-CoAs inactivates Ctbp2, thus preventing it from inhibiting FoxO1 mediated cell cycle arrest. CONCLUSION In conclusion, this study demonstrates that the Ctbp2-FoxO1-p21/p27 axis can promote cardiomyocyte proliferation and heart regeneration. As a metabolite sensor, Ctbp2 is activated during cardiomyocyte ischemia and hypoxia, while it is inactivated under normal conditions. This controllable and transient regulation of cardiomyocyte proliferation can avoid the detrimental effects on cardiac function caused by long-term regulation of cardiomyocyte proliferation, such as hypertrophic cardiomyopathy or heart failure. This provides new targets and new ideas for addressing the issues of cardiomyocyte proliferation and heart regeneration.
Collapse
Affiliation(s)
- Yanting Meng
- Department of Neurobiology, School of Basic Medical Sciences, Harbin Medical University, 157 Health Road, Nangang District, Harbin, 150081, Heilongjiang, China
| | - Jianwen Ding
- Department of Neurobiology, School of Basic Medical Sciences, Harbin Medical University, 157 Health Road, Nangang District, Harbin, 150081, Heilongjiang, China
| | - Yanping Wang
- Department of Neurobiology, School of Basic Medical Sciences, Harbin Medical University, 157 Health Road, Nangang District, Harbin, 150081, Heilongjiang, China
| | - Jing Wang
- Department of Neurobiology, School of Basic Medical Sciences, Harbin Medical University, 157 Health Road, Nangang District, Harbin, 150081, Heilongjiang, China
| | - Wei Huang
- Department of Neurobiology, School of Basic Medical Sciences, Harbin Medical University, 157 Health Road, Nangang District, Harbin, 150081, Heilongjiang, China
| | - Wenkang Jiang
- Department of Neurobiology, School of Basic Medical Sciences, Harbin Medical University, 157 Health Road, Nangang District, Harbin, 150081, Heilongjiang, China
| | - Jiayi Li
- Department of Neurobiology, School of Basic Medical Sciences, Harbin Medical University, 157 Health Road, Nangang District, Harbin, 150081, Heilongjiang, China
| | - Xiujuan Lang
- Department of Neurobiology, School of Basic Medical Sciences, Harbin Medical University, 157 Health Road, Nangang District, Harbin, 150081, Heilongjiang, China
| | - Sifan Zhang
- Department of Neurobiology, School of Basic Medical Sciences, Harbin Medical University, 157 Health Road, Nangang District, Harbin, 150081, Heilongjiang, China
| | - Yumei Liu
- Department of Neurobiology, School of Basic Medical Sciences, Harbin Medical University, 157 Health Road, Nangang District, Harbin, 150081, Heilongjiang, China
| | - Xijun Liu
- Department of Neurobiology, School of Basic Medical Sciences, Harbin Medical University, 157 Health Road, Nangang District, Harbin, 150081, Heilongjiang, China
| | - Hulun Li
- Department of Neurobiology, School of Basic Medical Sciences, Harbin Medical University, 157 Health Road, Nangang District, Harbin, 150081, Heilongjiang, China
- The Key Laboratory of Myocardial Ischemia, Harbin Medical University, Ministry of Education, 157 Health Road, Nangang District, Harbin, 150081, Heilongjiang, China
| | - Bo Sun
- Department of Neurobiology, School of Basic Medical Sciences, Harbin Medical University, 157 Health Road, Nangang District, Harbin, 150081, Heilongjiang, China.
| |
Collapse
|
10
|
Dittrich A, Andersson SA, Busk M, Hansen K, Foldager CB, Palmfeldt J, Andersen A, Pedersen M, Vendelbo M, Nielsen KL, Lauridsen H. Metabolic changes during cardiac regeneration in the axolotl. Dev Dyn 2025. [PMID: 40119743 DOI: 10.1002/dvdy.70020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2024] [Revised: 01/11/2025] [Accepted: 02/26/2025] [Indexed: 03/24/2025] Open
Abstract
BACKGROUND The axolotl is a prominent model organism of heart regeneration due to its ability to anatomically and functionally repair the heart after an injury that mimics human myocardial infarction. In humans, such an injury leads to permanent scarring. Cardiac regeneration has been linked to metabolism and the oxygenation state, but so far, these factors remain to be detailed in the axolotl model. In this descriptive study, we have investigated metabolic changes that occurred during cardiac regeneration in the axolotl. RESULTS We describe systemic and local cardiac metabolic changes after injury involving an early upregulation of glucose uptake and nucleotide biosynthesis followed by a later increase in acetate uptake. We detect several promising factors and metabolites for future studies and show that, unlike other popular animal models capable of intrinsic regeneration, the axolotl maintains its cardiac regenerative ability under hyperoxic conditions. CONCLUSIONS Axolotls undergo dynamic metabolic changes during the process of heart regeneration and display a robust reparative response to cardiac cryo-injury, which is unaffected by hyperoxia.
Collapse
Affiliation(s)
- Anita Dittrich
- Comparative Medicine Lab, Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| | - Sofie Amalie Andersson
- Comparative Medicine Lab, Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| | - Morten Busk
- Department of Clinical Medicine, Experimental Clinical Oncology, Aarhus University, Aarhus, Denmark
| | - Kasper Hansen
- Department of Forensic Medicine, Aarhus University, Aarhus, Denmark
| | - Casper Bindzus Foldager
- Comparative Medicine Lab, Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
- Orthopaedic Research Lab, Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| | - Johan Palmfeldt
- Research Unit for Molecular Medicine, Department of Clinical Medicine, Aarhus University and Aarhus University Hospital, Aarhus, Denmark
| | - Asger Andersen
- Comparative Medicine Lab, Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
- Department of Cardiology, Aarhus University Hospital, Aarhus, Denmark
| | - Michael Pedersen
- Comparative Medicine Lab, Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| | - Mikkel Vendelbo
- Department of Nuclear Medicine and PET-Center, Aarhus University Hospital, Aarhus, Denmark
- Department of Biomedicine, Aarhus University, Aarhus, Denmark
| | | | - Henrik Lauridsen
- Comparative Medicine Lab, Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| |
Collapse
|
11
|
Kelebeev J, MacKeracher A, Miyake T, McDermott JC. TAZ interactome analysis using nanotrap-based affinity purification-mass spectrometry. J Cell Sci 2025; 138:jcs263527. [PMID: 39898439 PMCID: PMC11928053 DOI: 10.1242/jcs.263527] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2024] [Accepted: 01/15/2025] [Indexed: 02/04/2025] Open
Abstract
Characterization of protein-protein interactions (PPIs) is a fundamental goal in the post-genomic era. Here, we document a generally applicable approach to identify cellular protein interactomes using a combination of nanobody-based affinity purification (AP) coupled with liquid chromatography and tandem mass spectrometry (LC-MS/MS). The Hippo signaling regulator TAZ (also known as WWTR1) functions as a transcriptional co-repressor or activator depending on its PPI network; we therefore undertook an unbiased proteomic screen to identify TAZ PPIs in striated muscle cells. A GFP nanotrap-based AP approach coupled with protein identification through LC-MS/MS was used to document a comprehensive list of known and novel TAZ interactome components. Informatic analysis of the interactome documented known components of the Hippo signaling pathway and multiple epigenetic regulators such as the NuRD, FACT and SWI/SNF complexes and the pro-myogenic CARM1 methyltransferase. Hippo pathway reporter gene (HOP/HIP) analysis indicated that CARM1 represses TAZ transcriptional co-activator function, promoting TAZ Ser89 phosphorylation and TAZ cytoplasmic sequestration. MS analysis revealed that CARM1 dimethylates TAZ at Arg77 in a PGPR*LAGG consensus peptide, resulting in enhanced TAZ Ser89 phosphorylation. These studies underline the utility of a nanobody-based AP approach for interactome analysis.
Collapse
Affiliation(s)
- Jonathan Kelebeev
- Department of Biology, York University, Toronto, ON, M3J 1P3, Canada
- Muscle Health Research Centre (MHRC), York University, Toronto, ON, M3J 1P3, Canada
- Centre for Research in Biomolecular Interactions (CRBI), York University, Toronto, ON, M3J 1P3, Canada
| | - Anastasia MacKeracher
- Department of Biology, York University, Toronto, ON, M3J 1P3, Canada
- Muscle Health Research Centre (MHRC), York University, Toronto, ON, M3J 1P3, Canada
- Centre for Research in Biomolecular Interactions (CRBI), York University, Toronto, ON, M3J 1P3, Canada
| | - Tetsuaki Miyake
- Department of Biology, York University, Toronto, ON, M3J 1P3, Canada
- Muscle Health Research Centre (MHRC), York University, Toronto, ON, M3J 1P3, Canada
- Centre for Research in Biomolecular Interactions (CRBI), York University, Toronto, ON, M3J 1P3, Canada
| | - John C. McDermott
- Department of Biology, York University, Toronto, ON, M3J 1P3, Canada
- Muscle Health Research Centre (MHRC), York University, Toronto, ON, M3J 1P3, Canada
- Centre for Research in Biomolecular Interactions (CRBI), York University, Toronto, ON, M3J 1P3, Canada
| |
Collapse
|
12
|
Harris BN, Woo LA, Perry RN, Wallace AM, Civelek M, Wolf MJ, Saucerman JJ. Dynamic map illuminates Hippo-cMyc module crosstalk driving cardiomyocyte proliferation. Development 2025; 152:DEV204397. [PMID: 39866065 PMCID: PMC11883243 DOI: 10.1242/dev.204397] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2024] [Accepted: 01/17/2025] [Indexed: 01/28/2025]
Abstract
Numerous regulators of cardiomyocyte (CM) proliferation have been identified, yet how they coordinate during cardiac development or regeneration is poorly understood. Here, we developed a computational model of the CM proliferation regulatory network to obtain key regulators and systems-level understanding. The model defines five modules (DNA replication, mitosis, cytokinesis, growth factor, Hippo pathway) and integrates them into a network of 72 nodes and 88 reactions that correctly predicts 74 of 81 (91.35%) independent experiments from the literature. The model predicts that in response to YAP activation, the Hippo module crosstalks to the growth factor module via PI3K and cMyc to drive cell cycle activity. This predicted YAP-cMyc axis is validated experimentally in rat CMs and further supported by YAP-stimulated cMyc open chromatin and mRNA in mouse hearts. This validated computational model predicts how individual regulators and modules coordinate to control CM proliferation.
Collapse
Affiliation(s)
- Bryana N. Harris
- Department of Biomedical Engineering, University of Virginia, Charlottesville, VA 22908-0759, USA
| | - Laura A. Woo
- Department of Biomedical Engineering, University of Virginia, Charlottesville, VA 22908-0759, USA
| | - R. Noah Perry
- Department of Biomedical Engineering, University of Virginia, Charlottesville, VA 22908-0759, USA
| | - Alexia M. Wallace
- Department of Biomedical Engineering, University of Virginia, Charlottesville, VA 22908-0759, USA
| | - Mete Civelek
- Department of Biomedical Engineering, University of Virginia, Charlottesville, VA 22908-0759, USA
- Department of Biochemistry and Molecular Genetics, University of Virginia, Charlottesville, VA 22908-0759, USA
- Center for Public Health Genomics, University of Virginia, Charlottesville, VA 22908-0759, USA
- Robert M. Berne Cardiovascular Research Center, University of Virginia, Charlottesville, VA 22908-0759, USA
| | - Matthew J. Wolf
- Robert M. Berne Cardiovascular Research Center, University of Virginia, Charlottesville, VA 22908-0759, USA
- Division of Cardiovascular Medicine, University of Virginia, Charlottesville, VA 22908-0759,USA
| | - Jeffrey J. Saucerman
- Department of Biomedical Engineering, University of Virginia, Charlottesville, VA 22908-0759, USA
- Robert M. Berne Cardiovascular Research Center, University of Virginia, Charlottesville, VA 22908-0759, USA
- Division of Cardiovascular Medicine, University of Virginia, Charlottesville, VA 22908-0759,USA
| |
Collapse
|
13
|
Zhao HW, Liu ST, Wang XJ, Zhang XM, Ma X. Elucidating the Role of HIF-1α/YAP Signaling Pathway in Regulating Inflammation in Human Periodontal Stem Cells: An in vitro Study. J Inflamm Res 2025; 18:1875-1886. [PMID: 39931166 PMCID: PMC11809419 DOI: 10.2147/jir.s504793] [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: 11/20/2024] [Accepted: 01/20/2025] [Indexed: 02/13/2025] Open
Abstract
Background and Objective Periodontitis is a chronic inflammatory disease caused by dental plaque accumulation, leading to damage of periodontal tissues and potential tooth loss. Understanding the mechanisms of periodontitis, particularly the role of hypoxia in inflammation, is critical for identifying novel therapeutic strategies. This study investigated the effects of the prolyl hydroxylase (PHD) inhibitor DMOG on pro-inflammatory cytokine expression in human periodontal ligament stem cells (hPDLSCs) and examined the involvement of the HIF-1α/YAP signaling path ay in modulating inflammation. Materials and Methods hPDLSCs were cultured and treated with lipopolysaccharide (LPS) to induce inflammation, followed by DMOG treatment. Cell proliferation was assessed using the CCK-8 assay, while ELISA and RT-qPCR evaluated the expression levels of HIF-1α, IL-1β, TNF-α, and YAP. YAP expression was knocked down using siRNA transfection to examine its effects on inflammatory cytokines. Results DMOG significantly increased HIF-1α expression while reducing IL-1β and TNF-α levels in LPS-treated hPDLSCs. 0.1 mmol/L DMOG inhibited cell proliferation after 72 hours (P < 0.001). ELISA results showed that HIF-1α concentrations in the LPS + DMOG group were significantly higher than in the LPS group (P < 0.01), while IL-1β and TNF-α levels were significantly reduced (P < 0.01). RT-qPCR confirmed these trends, showing reduced mRNA levels of IL-1β and TNF-α and increased YAP expression in the LPS + DMOG group (P < 0.0001). YAP knockdown via siRNA transfection reversed these effects, increasing IL-1β and TNF-α levels (P < 0.01) while significantly reducing HIF-1α expression (P < 0.05). Conclusion This study demonstrated that DMOG reduces inflammatory cytokine expression in hPDLSCs by stabilizing HIF-1α and activating the YAP signaling pathway. These findings provide a mechanistic basis for targeting the HIF-1α/YAP axis to control periodontal inflammation and support the potential of PHD inhibitors as therapeutic agents for periodontitis.
Collapse
Affiliation(s)
- Hui-Wei Zhao
- Department of Periodontology, The Affiliated Yantai Stomatological Hospital, Binzhou Medical University, Yantai, 264000, People’s Republic of China
| | - Shu-Tai Liu
- Department of Periodontology, Shenzhen Stomatology Hospital, Shenzhen, 518000, People’s Republic of China
| | - Xiang-Jin Wang
- Department of Periodontology, The Affiliated Yantai Stomatological Hospital, Binzhou Medical University, Yantai, 264000, People’s Republic of China
| | - Xue-Mei Zhang
- Department of Periodontology, The Affiliated Yantai Stomatological Hospital, Binzhou Medical University, Yantai, 264000, People’s Republic of China
| | - Xiang Ma
- Department of Periodontology, The Affiliated Yantai Stomatological Hospital, Binzhou Medical University, Yantai, 264000, People’s Republic of China
| |
Collapse
|
14
|
Mitra A, Mandal S, Banerjee K, Ganguly N, Sasmal P, Banerjee D, Gupta S. Cardiac Regeneration in Adult Zebrafish: A Review of Signaling and Metabolic Coordination. Curr Cardiol Rep 2025; 27:15. [PMID: 39792206 DOI: 10.1007/s11886-024-02162-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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 11/20/2024] [Indexed: 01/12/2025]
Abstract
PURPOSE OF REVIEW This review investigates how post-injury cellular signaling and energy metabolism are two pivotal points in zebrafish's cardiomyocyte cell cycle re-entry and proliferation. It seeks to highlight the probable mechanism of action in proliferative cardiomyocytes compared to mammals and identify gaps in the current understanding of metabolic regulation of cardiac regeneration. RECENT FINDINGS Metabolic substrate changes after birth correlate with reduced cardiomyocyte proliferation in mammals. Unlike adult mammalian hearts, zebrafish can regenerate cardiomyocytes by re-entering the cell cycle, characterized by a metabolic switch from oxidative metabolism to increased glycolysis. Zebrafish provide a valuable model for studying metabolic regulation during cell cycle re-entry and cardiac regeneration. Proliferative cardiomyocytes have upregulated Notch, hippo, and Wnt signaling and decreased ROS generation, DNA damage in different zebrafish cardiac regeneration models. Understanding the correlation between metabolic switches during cell cycle re-entry of already differentiated zebrafish cardiomyocytes is being increasingly recognized as a critical factor in heart regeneration. Zebrafish studies provide insights into metabolic adaptations during heart regeneration, emphasizing the importance of a metabolic switch. However, there are mechanistic gaps, and extensive studies are required to aid in formulating therapeutic strategies for cardiac regenerative medicine.
Collapse
Affiliation(s)
- Arkadeep Mitra
- Department of Zoology, City College, 102/1, Raja Rammohan Sarani, Kolkata, 700009, West Bengal, India
| | - Subhadeep Mandal
- Department of Zoology, Trivenidevi Bhalotia College (Affiliated to Kazi Nazrul University), College Para Rd, Raniganj, 713347, West Bengal, India
| | - Kalyan Banerjee
- Department of Zoology, Trivenidevi Bhalotia College (Affiliated to Kazi Nazrul University), College Para Rd, Raniganj, 713347, West Bengal, India
| | - Nilanjan Ganguly
- Department of Zoology, Trivenidevi Bhalotia College (Affiliated to Kazi Nazrul University), College Para Rd, Raniganj, 713347, West Bengal, India
| | - Pramit Sasmal
- Department of Zoology, Trivenidevi Bhalotia College (Affiliated to Kazi Nazrul University), College Para Rd, Raniganj, 713347, West Bengal, India
| | - Durba Banerjee
- Department of Anesthesiology and Pain Medicine, University of Washington, 850 Republican St, Seattle, WA, 98109, USA.
| | - Shreyasi Gupta
- Department of Zoology, Trivenidevi Bhalotia College (Affiliated to Kazi Nazrul University), College Para Rd, Raniganj, 713347, West Bengal, India.
| |
Collapse
|
15
|
Liu S, Deshmukh V, Meng F, Wang Y, Morikawa Y, Steimle JD, Li RG, Wang J, Martin JF. Microtubules Sequester Acetylated YAP in the Cytoplasm and Inhibit Heart Regeneration. Circulation 2025; 151:59-75. [PMID: 39185559 PMCID: PMC11671299 DOI: 10.1161/circulationaha.123.067646] [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: 10/17/2023] [Accepted: 07/30/2024] [Indexed: 08/27/2024]
Abstract
BACKGROUND The Hippo pathway effector YAP (Yes-associated protein) plays an essential role in cardiomyocyte proliferation and heart regeneration. In response to physiological changes, YAP moves in and out of the nucleus. The pathophysiological mechanisms regulating YAP subcellular localization after myocardial infarction remain poorly defined. METHODS We identified YAP acetylation at site K265 by in vitro acetylation followed by mass spectrometry analysis. We used adeno-associated virus to express YAP-containing mutations that either abolished acetylation (YAP-K265R) or mimicked acetylation (YAP-K265Q) and studied how acetylation regulates YAP subcellular localization in mouse hearts. We generated a cell line with YAP-K265R mutation and investigated the protein-protein interactors by YAP immunoprecipitation followed by mass spectrometry, then validated the YAP interaction in neonatal rat ventricular myocytes. We examined colocalization of YAP and TUBA4A (tubulin α 4A) by superresolution imaging. Furthermore, we developed YAP-K265R and αMHC-MerCreMer (MCM); Yap-loxP/K265R mutant mice to examine the pathophysiological role of YAP acetylation in cardiomyocytes during cardiac regeneration. RESULTS We found that YAP is acetylated at K265 by CBP (CREB-binding protein)/P300 (E1A-binding protein P300) and is deacetylated by nicotinamide phosphoribosyltransferase/nicotinamide adenine dinucleotide/sirtuins axis in cardiomyocytes. After myocardial infarction, YAP acetylation is increased, which promotes YAP cytoplasmic localization. Compared with controls, mice that were genetically engineered to express a K265R mutation that prevents YAP K265 acetylation showed improved cardiac regenerative ability and increased YAP nuclear localization. Mechanistically, YAP acetylation facilitates its interaction with TUBA4A, a component of the microtubule network that sequesters acetylated YAP in the cytoplasm. After myocardial infarction, the microtubule network increased in cardiomyocytes, resulting in the accumulation of YAP in the cytoplasm. CONCLUSIONS After myocardial infarction, decreased sirtuin activity enriches YAP acetylation at K265. The growing TUBA4A network sequesters acetylated YAP within the cytoplasm, which is detrimental to cardiac regeneration.
Collapse
Affiliation(s)
- Shijie Liu
- Cardiomyocyte Renewal Laboratory, Texas Heart Institute, Houston, (S.L., Y.M., R.G.L., J.W., J.F.M.)
- Division of Molecular Cardiovascular Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH (S.L.)
| | - Vaibhav Deshmukh
- Department of Integrative Physiology, Baylor College of Medicine, Houston, TX (V.D., F.M., J.D.S., J.F.M.)
- Department of Cell Biology & Physiology, Washington University School of Medicine, St Louis, MO (V.D.)
| | - Fansen Meng
- Department of Integrative Physiology, Baylor College of Medicine, Houston, TX (V.D., F.M., J.D.S., J.F.M.)
| | | | - Yuka Morikawa
- Cardiomyocyte Renewal Laboratory, Texas Heart Institute, Houston, (S.L., Y.M., R.G.L., J.W., J.F.M.)
| | - Jeffrey D Steimle
- Department of Integrative Physiology, Baylor College of Medicine, Houston, TX (V.D., F.M., J.D.S., J.F.M.)
| | - Rich Gang Li
- Cardiomyocyte Renewal Laboratory, Texas Heart Institute, Houston, (S.L., Y.M., R.G.L., J.W., J.F.M.)
| | - Jun Wang
- Cardiomyocyte Renewal Laboratory, Texas Heart Institute, Houston, (S.L., Y.M., R.G.L., J.W., J.F.M.)
| | - James F Martin
- Cardiomyocyte Renewal Laboratory, Texas Heart Institute, Houston, (S.L., Y.M., R.G.L., J.W., J.F.M.)
- Department of Integrative Physiology, Baylor College of Medicine, Houston, TX (V.D., F.M., J.D.S., J.F.M.)
| |
Collapse
|
16
|
Bouwman M, de Bakker DEM, Honkoop H, Giovou AE, Versteeg D, Boender AR, Nguyen PD, Slotboom M, Colquhoun D, Vigil-Garcia M, Kooijman L, Janssen R, Hooijkaas IB, Günthel M, Visser KJ, Klerk M, Zentilin L, Giacca M, Kaslin J, Boink GJJ, van Rooij E, Christoffels VM, Bakkers J. Cross-species comparison reveals that Hmga1 reduces H3K27me3 levels to promote cardiomyocyte proliferation and cardiac regeneration. NATURE CARDIOVASCULAR RESEARCH 2025; 4:64-82. [PMID: 39747457 PMCID: PMC11738996 DOI: 10.1038/s44161-024-00588-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/15/2023] [Accepted: 11/26/2024] [Indexed: 01/04/2025]
Abstract
In contrast to adult mammalian hearts, the adult zebrafish heart efficiently replaces cardiomyocytes lost after injury. Here we reveal shared and species-specific injury response pathways and a correlation between Hmga1, an architectural non-histone protein, and regenerative capacity, as Hmga1 is required and sufficient to induce cardiomyocyte proliferation and required for heart regeneration. In addition, Hmga1 was shown to reactivate developmentally silenced genes, likely through modulation of H3K27me3 levels, poising them for a pro-regenerative gene program. Furthermore, AAV-mediated Hmga1 expression in injured adult mouse hearts led to controlled cardiomyocyte proliferation in the border zone and enhanced heart function, without cardiomegaly and adverse remodeling. Histone modification mapping in mouse border zone cardiomyocytes revealed a similar modulation of H3K27me3 marks, consistent with findings in zebrafish. Our study demonstrates that Hmga1 mediates chromatin remodeling and drives a regenerative program, positioning it as a promising therapeutic target to enhance cardiac regeneration after injury.
Collapse
Affiliation(s)
- Mara Bouwman
- Hubrecht Institute, Royal Netherlands Academy of Arts and Sciences (KNAW) and University Medical Center Utrecht, Utrecht, The Netherlands
| | - Dennis E M de Bakker
- Hubrecht Institute, Royal Netherlands Academy of Arts and Sciences (KNAW) and University Medical Center Utrecht, Utrecht, The Netherlands
- Leibniz Institute on Aging, Fritz Lipmann Institute (FLI), Jena, Germany
| | - Hessel Honkoop
- Hubrecht Institute, Royal Netherlands Academy of Arts and Sciences (KNAW) and University Medical Center Utrecht, Utrecht, The Netherlands
| | - Alexandra E Giovou
- Department of Medical Biology, Amsterdam Cardiovascular Sciences, Amsterdam University Medical Centers, Amsterdam, The Netherlands
| | - Danielle Versteeg
- Hubrecht Institute, Royal Netherlands Academy of Arts and Sciences (KNAW) and University Medical Center Utrecht, Utrecht, The Netherlands
| | - Arie R Boender
- Department of Medical Biology, Amsterdam Cardiovascular Sciences, Amsterdam University Medical Centers, Amsterdam, The Netherlands
- PacingCure BV, Amsterdam, The Netherlands
| | - Phong D Nguyen
- Hubrecht Institute, Royal Netherlands Academy of Arts and Sciences (KNAW) and University Medical Center Utrecht, Utrecht, The Netherlands
- Institut Curie, Université PSL, CNRS UMR3215, INSERM U934, Paris, France
| | - Merel Slotboom
- Hubrecht Institute, Royal Netherlands Academy of Arts and Sciences (KNAW) and University Medical Center Utrecht, Utrecht, The Netherlands
| | - Daniel Colquhoun
- Australian Regenerative Medicine Institute, Monash University, Melbourne, VIC, Australia
| | - Marta Vigil-Garcia
- Hubrecht Institute, Royal Netherlands Academy of Arts and Sciences (KNAW) and University Medical Center Utrecht, Utrecht, The Netherlands
| | - Lieneke Kooijman
- Hubrecht Institute, Royal Netherlands Academy of Arts and Sciences (KNAW) and University Medical Center Utrecht, Utrecht, The Netherlands
| | - Rob Janssen
- Department of Medical Biology, Amsterdam Cardiovascular Sciences, Amsterdam University Medical Centers, Amsterdam, The Netherlands
| | - Ingeborg B Hooijkaas
- Department of Medical Biology, Amsterdam Cardiovascular Sciences, Amsterdam University Medical Centers, Amsterdam, The Netherlands
| | - Marie Günthel
- Department of Medical Biology, Amsterdam Cardiovascular Sciences, Amsterdam University Medical Centers, Amsterdam, The Netherlands
| | - Kimberly J Visser
- Hubrecht Institute, Royal Netherlands Academy of Arts and Sciences (KNAW) and University Medical Center Utrecht, Utrecht, The Netherlands
| | - Mischa Klerk
- Department of Medical Biology, Amsterdam Cardiovascular Sciences, Amsterdam University Medical Centers, Amsterdam, The Netherlands
| | - Lorena Zentilin
- International Centre for Genetic Engineering and Biotechnology (ICGEB), University of Trieste, Trieste, Italy
| | - Mauro Giacca
- International Centre for Genetic Engineering and Biotechnology (ICGEB), University of Trieste, Trieste, Italy
- School of Cardiovascular and Metabolic Medicine and Sciences, King's College London, London, UK
| | - Jan Kaslin
- Australian Regenerative Medicine Institute, Monash University, Melbourne, VIC, Australia
| | - Gerard J J Boink
- Department of Medical Biology, Amsterdam Cardiovascular Sciences, Amsterdam University Medical Centers, Amsterdam, The Netherlands
- PacingCure BV, Amsterdam, The Netherlands
- Department of Cardiology, Amsterdam Cardiovascular Sciences, Amsterdam University Medical Centers, Amsterdam, The Netherlands
| | - Eva van Rooij
- Hubrecht Institute, Royal Netherlands Academy of Arts and Sciences (KNAW) and University Medical Center Utrecht, Utrecht, The Netherlands
- Department of Cardiology, Division of Heart and Lungs, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Vincent M Christoffels
- Department of Medical Biology, Amsterdam Cardiovascular Sciences, Amsterdam University Medical Centers, Amsterdam, The Netherlands
| | - Jeroen Bakkers
- Hubrecht Institute, Royal Netherlands Academy of Arts and Sciences (KNAW) and University Medical Center Utrecht, Utrecht, The Netherlands.
- Department of Pediatric Cardiology, Division of Pediatrics, University Medical Center Utrecht, Utrecht, The Netherlands.
| |
Collapse
|
17
|
Hu C, Francisco J, Del Re DP, Sadoshima J. Decoding the Impact of the Hippo Pathway on Different Cell Types in Heart Failure. Circ J 2024; 89:6-15. [PMID: 38644191 DOI: 10.1253/circj.cj-24-0171] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 04/23/2024]
Abstract
The evolutionarily conserved Hippo pathway plays a pivotal role in governing a variety of biological processes. Heart failure (HF) is a major global health problem with a significant risk of mortality. This review provides a contemporary understanding of the Hippo pathway in regulating different cell types during HF. Through a systematic analysis of each component's regulatory mechanisms within the Hippo pathway, we elucidate their specific effects on cardiomyocytes, fibroblasts, endothelial cells, and macrophages in response to various cardiac injuries. Insights gleaned from both in vitro and in vivo studies highlight the therapeutic promise of targeting the Hippo pathway to address cardiovascular diseases, particularly HF.
Collapse
Affiliation(s)
- Chengchen Hu
- Department of Cell Biology and Molecular Medicine, Rutgers New Jersey Medical School
| | - Jamie Francisco
- Department of Cell Biology and Molecular Medicine, Rutgers New Jersey Medical School
| | - Dominic P Del Re
- Department of Cell Biology and Molecular Medicine, Rutgers New Jersey Medical School
| | - Junichi Sadoshima
- Department of Cell Biology and Molecular Medicine, Rutgers New Jersey Medical School
| |
Collapse
|
18
|
Wang Y, Chatterjee E, Li G, Xu J, Xiao J. Force-sensing protein expression in response to cardiovascular mechanotransduction. EBioMedicine 2024; 110:105412. [PMID: 39481337 PMCID: PMC11554632 DOI: 10.1016/j.ebiom.2024.105412] [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: 07/12/2024] [Revised: 10/01/2024] [Accepted: 10/07/2024] [Indexed: 11/02/2024] Open
Abstract
Force-sensing biophysical cues in microenvironment, including extracellular matrix performances, stretch-mediated mechanics, shear stress and flow-induced hemodynamics, have a significant influence in regulating vascular morphogenesis and cardiac remodeling by mechanotransduction. Once cells perceive these extracellular mechanical stimuli, Piezo activation promotes calcium influx by forming integrin-adhesion-coupling receptors. This induces robust contractility of cytoskeleton structures to further transmit biomechanical alternations into nuclei by regulating Hippo-Yes associated protein (YAP) signaling pathway between cytoplasmic and nuclear translocation. Although biomechanical stimuli are widely studied in cardiovascular diseases, the expression of force-sensing proteins in response to cardiovascular mechanotransduction has not been systematically concluded. Therefore, this review will summarize the force-sensing Piezo, cytoskeleton and YAP proteins to mediate extracellular mechanics, and also give the prominent emphasis on intrinsic connection of these mechanical proteins and cardiovascular mechanotransduction. Extensive insights into cardiovascular mechanics may provide some new strategies for cardiovascular clinical therapy.
Collapse
Affiliation(s)
- Yongtao Wang
- Cardiac Regeneration and Ageing Lab, Institute of Geriatrics (Shanghai University), Affiliated Nantong Hospital of Shanghai University (The Sixth People's Hospital of Nantong), School of Medicine, Shanghai University, Nantong 226011, China; Institute of Cardiovascular Sciences, Shanghai Engineering Research Center of Organ Repair, Joint International Research Laboratory of Biomaterials and Biotechnology in Organ Repair (Ministry of Education), School of Life Science, Shanghai University, Shanghai 200444, China
| | - Emeli Chatterjee
- Cardiovascular Division of the Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA
| | - Guoping Li
- Cardiovascular Division of the Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA
| | - Jiahong Xu
- Department of Cardiology, Shanghai Gongli Hospital, Shanghai 200135, China.
| | - Junjie Xiao
- Cardiac Regeneration and Ageing Lab, Institute of Geriatrics (Shanghai University), Affiliated Nantong Hospital of Shanghai University (The Sixth People's Hospital of Nantong), School of Medicine, Shanghai University, Nantong 226011, China; Institute of Cardiovascular Sciences, Shanghai Engineering Research Center of Organ Repair, Joint International Research Laboratory of Biomaterials and Biotechnology in Organ Repair (Ministry of Education), School of Life Science, Shanghai University, Shanghai 200444, China.
| |
Collapse
|
19
|
Zhang J. Non-coding RNAs and angiogenesis in cardiovascular diseases: a comprehensive review. Mol Cell Biochem 2024; 479:2921-2953. [PMID: 38306012 DOI: 10.1007/s11010-023-04919-5] [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: 10/27/2023] [Accepted: 12/18/2023] [Indexed: 02/03/2024]
Abstract
Non-coding RNAs (ncRNAs) have key roles in the etiology of many illnesses, including heart failure, myocardial infarction, stroke, and in physiological processes like angiogenesis. In transcriptional regulatory circuits that control heart growth, signaling, and stress response, as well as remodeling in cardiac disease, ncRNAs have become important players. Studies on ncRNAs and cardiovascular disease have made great progress recently. Here, we go through the functions of non-coding RNAs (ncRNAs) like circular RNAs (circRNAs), and microRNAs (miRNAs) as well as long non-coding RNAs (lncRNAs) in modulating cardiovascular disorders.
Collapse
Affiliation(s)
- Jie Zhang
- Medical School, Shandong University of Traditional Chinese Medicine, Jinan, 250355, China.
| |
Collapse
|
20
|
Fiorino E, Rossin D, Vanni R, Aubry M, Giachino C, Rastaldo R. Recent Insights into Endogenous Mammalian Cardiac Regeneration Post-Myocardial Infarction. Int J Mol Sci 2024; 25:11747. [PMID: 39519298 PMCID: PMC11546116 DOI: 10.3390/ijms252111747] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2024] [Revised: 10/29/2024] [Accepted: 10/30/2024] [Indexed: 11/16/2024] Open
Abstract
Myocardial infarction (MI) is a critical global health issue and a leading cause of heart failure. Indeed, while neonatal mammals can regenerate cardiac tissue mainly through cardiomyocyte proliferation, this ability is lost shortly after birth, resulting in the adult heart's inability to regenerate after injury effectively. In adult mammals, the adverse cardiac remodelling, which compensates for the loss of cardiac cells, impairs cardiac function due to the non-contractile nature of fibrotic tissue. Moreover, the neovascularisation after MI is inadequate to restore blood flow to the infarcted myocardium. This review aims to synthesise the most recent insights into the molecular and cellular players involved in endogenous myocardial and vascular regeneration, facilitating the identification of mechanisms that could be targeted to trigger cardiac regeneration, reduce fibrosis, and improve functional recovery post-MI. Reprogramming adult cardiomyocytes to regain their proliferative potential, along with the modulation of target cells responsible for neovascularisation, represents promising therapeutic strategies. An updated overview of endogenous mechanisms that regulate both myocardial and coronary vasculature regeneration-including stem and progenitor cells, growth factors, cell cycle regulators, and key signalling pathways-could help identify new critical intervention points for therapeutic applications.
Collapse
Affiliation(s)
| | | | | | | | | | - Raffaella Rastaldo
- Department of Clinical and Biological Sciences, University of Turin, Regione Gonzole 10, 10043 Orbassano, Italy; (E.F.); (D.R.); (R.V.); (M.A.); (C.G.)
| |
Collapse
|
21
|
Bejar N, Xiao S, Iyer D, Muili A, Adeleye A, McConnell BK, Schwartz RJ. STEMIN and YAP5SA, the future of heart repair? Exp Biol Med (Maywood) 2024; 249:10246. [PMID: 39544432 PMCID: PMC11560420 DOI: 10.3389/ebm.2024.10246] [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: 05/17/2024] [Accepted: 09/30/2024] [Indexed: 11/17/2024] Open
Abstract
This review outlines some of the many approaches taken over a decade or more to repair damaged hearts. We showcase the recent breakthroughs in organ regeneration elicited by reprogramming factors OCT3/4, SOX2, KLF4, and C-MYC (OKSM). Transient OKSM transgene expression rejuvenated senescent organs in mice. OKSM transgenes also caused murine heart cell regeneration. A triplet alanine mutation of the N-terminus of Serum Response Factor's MADS box SRF153(A3), termed STEMIN, and the YAP mutant, YAP5SA synergized and activated OKSM and NANOG in adult rat cardiac myocytes; thus, causing rapid nuclear proliferation and blocked myocyte differentiation. In addition, ATAC seq showed induced expression of growth factor genes FGFs, BMPs, Notchs, IGFs, JAK, STATs and non-canonical Wnts. Injected STEMIN and YAP5SA synthetic modifying mRNA (mmRNA) into infarcted adult mouse hearts, brought damaged hearts back to near normal contractility without severe fibrosis. Thus, STEMIN and YAP5SA mmRNA may exert additional regenerative potential than OKSM alone for treating heart diseases.
Collapse
Affiliation(s)
- Nada Bejar
- Department of Biology and Biochemistry, University of Houston, Houston, TX, United States
| | - Siyu Xiao
- Department of Biology and Biochemistry, University of Houston, Houston, TX, United States
| | - Dinakar Iyer
- Department of Biology and Biochemistry, University of Houston, Houston, TX, United States
- Department of Pharmacological and Pharmaceutical Sciences, College of Pharmacy, University of Houston, Houston, TX, United States
| | - Azeez Muili
- Department of Biology and Biochemistry, University of Houston, Houston, TX, United States
| | - Adeniyi Adeleye
- Department of Biology and Biochemistry, University of Houston, Houston, TX, United States
| | - Bradley K. McConnell
- Department of Pharmacological and Pharmaceutical Sciences, College of Pharmacy, University of Houston, Houston, TX, United States
| | - Robert J. Schwartz
- Department of Biology and Biochemistry, University of Houston, Houston, TX, United States
| |
Collapse
|
22
|
Nelson VL, Eadie AL, Perez L, Madhu M, Platt M, Mercer A, Pulinilkunnil T, Kienesberger P, Simpson JA, Brunt KR. Yap Is a Nutrient Sensor Sensitive to the Amino Acid L-Isoleucine and Regulates the Expression of Ctgf in Cardiomyocytes. Biomolecules 2024; 14:1299. [PMID: 39456232 PMCID: PMC11506509 DOI: 10.3390/biom14101299] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2024] [Revised: 10/02/2024] [Accepted: 10/04/2024] [Indexed: 10/28/2024] Open
Abstract
Myocardial infarction and reperfusion constitute a complex injury consisting of many distinct molecular stress patterns that influence cardiomyocyte survival and adaptation. Cell signalling, which is essential to cardiac development, also presents potential disease-modifying opportunities to recover and limit myocardial injury or maladaptive remodelling. Here, we hypothesized that Yap signalling could be sensitive to one or more molecular stress patterns associated with early acute ischemia. We found that Yap, and not Taz, expression patterns differed in a post-myocardial infarct compared to a peri-infarct region of rat hearts post-myocardial infarction, suggesting cell specificity that would be challenging to resolve for causation in vivo. Using H9c2 ventricular myotubes in vitro as a model, Yap levels were determined to be more sensitive to nutrient deprivation than other stress patterns typified by ischemia within the first hour of stress. Moreover, this is mediated by amino acid availability, predominantly L-isoleucine, and influences the expression of connective tissue growth factor (Ctgf)-a major determinant of myocardial adaptation after injury. These findings present novel opportunities for future therapeutic development and risk assessment for myocardial injury and adaptation.
Collapse
Affiliation(s)
- Victoria L. Nelson
- Department of Pharmacology, Dalhousie University, Halifax, NS B3H 4R2, Canada
- Dalhousie Medicine New Brunswick, Faculty of Medicine, Dalhousie University, Saint John, NB E2L 4L5, Canada
- IMPART Investigator Team Canada, Saint John, NB E2L 4L5, Canada
| | - Ashley L. Eadie
- Department of Pharmacology, Dalhousie University, Halifax, NS B3H 4R2, Canada
- Dalhousie Medicine New Brunswick, Faculty of Medicine, Dalhousie University, Saint John, NB E2L 4L5, Canada
- IMPART Investigator Team Canada, Saint John, NB E2L 4L5, Canada
| | - Lester Perez
- Department of Pharmacology, Dalhousie University, Halifax, NS B3H 4R2, Canada
- Dalhousie Medicine New Brunswick, Faculty of Medicine, Dalhousie University, Saint John, NB E2L 4L5, Canada
- IMPART Investigator Team Canada, Saint John, NB E2L 4L5, Canada
| | - Malav Madhu
- Department of Pharmacology, Dalhousie University, Halifax, NS B3H 4R2, Canada
- Dalhousie Medicine New Brunswick, Faculty of Medicine, Dalhousie University, Saint John, NB E2L 4L5, Canada
- IMPART Investigator Team Canada, Saint John, NB E2L 4L5, Canada
| | - Mathew Platt
- IMPART Investigator Team Canada, Saint John, NB E2L 4L5, Canada
- Department of Human Health and Nutritional Sciences, University of Guelph, Guelph, ON N1G 2W1, Canada
| | - Angella Mercer
- Dalhousie Medicine New Brunswick, Faculty of Medicine, Dalhousie University, Saint John, NB E2L 4L5, Canada
- IMPART Investigator Team Canada, Saint John, NB E2L 4L5, Canada
- Department of Biochemistry and Molecular Biology, Dalhousie University, Halifax, NS B3H 4R2, Canada
| | - Thomas Pulinilkunnil
- Dalhousie Medicine New Brunswick, Faculty of Medicine, Dalhousie University, Saint John, NB E2L 4L5, Canada
- IMPART Investigator Team Canada, Saint John, NB E2L 4L5, Canada
- Department of Biochemistry and Molecular Biology, Dalhousie University, Halifax, NS B3H 4R2, Canada
| | - Petra Kienesberger
- Dalhousie Medicine New Brunswick, Faculty of Medicine, Dalhousie University, Saint John, NB E2L 4L5, Canada
- IMPART Investigator Team Canada, Saint John, NB E2L 4L5, Canada
- Department of Biochemistry and Molecular Biology, Dalhousie University, Halifax, NS B3H 4R2, Canada
| | - Jeremy A. Simpson
- IMPART Investigator Team Canada, Saint John, NB E2L 4L5, Canada
- Department of Human Health and Nutritional Sciences, University of Guelph, Guelph, ON N1G 2W1, Canada
| | - Keith R. Brunt
- Department of Pharmacology, Dalhousie University, Halifax, NS B3H 4R2, Canada
- Dalhousie Medicine New Brunswick, Faculty of Medicine, Dalhousie University, Saint John, NB E2L 4L5, Canada
- IMPART Investigator Team Canada, Saint John, NB E2L 4L5, Canada
| |
Collapse
|
23
|
Astone M, Tesoriero C, Schiavone M, Facchinello N, Tiso N, Argenton F, Vettori A. Wnt/β-Catenin Signaling Regulates Yap/Taz Activity during Embryonic Development in Zebrafish. Int J Mol Sci 2024; 25:10005. [PMID: 39337493 PMCID: PMC11432159 DOI: 10.3390/ijms251810005] [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: 05/31/2024] [Revised: 09/11/2024] [Accepted: 09/14/2024] [Indexed: 09/30/2024] Open
Abstract
Hippo-YAP/TAZ and Wnt/β-catenin signaling pathways, by controlling proliferation, migration, cell fate, stemness, and apoptosis, are crucial regulators of development and tissue homeostasis. We employed zebrafish embryos as a model system to elucidate in living reporter organisms the crosstalk between the two signaling pathways. Co-expression analysis between the Wnt/β-catenin Tg(7xTCF-Xla.Siam:GFP)ia4 and the Hippo-Yap/Taz Tg(Hsa.CTGF:nlsmCherry)ia49 zebrafish reporter lines revealed shared spatiotemporal expression profiles. These patterns were particularly evident in key developmental regions such as the midbrain-hindbrain boundary (MHB), epidermis, muscles, neural tube, notochord, floorplate, and otic vesicle. To investigate the relationship between the Wnt/β-catenin pathway and Hippo-Yap/Taz signaling in vivo, we conducted a series of experiments employing both pharmacological and genetic strategies. Modulation of the Wnt/β-catenin pathway with IWR-1, XAV939, or BIO resulted in a significant regulation of the Yap/Taz reporter signal, highlighting a clear correlation between β-catenin and Yap/Taz activities. Furthermore, genetic perturbation of the Wnt/β-catenin pathway, by APC inhibition or DKK1 upregulation, elicited evident and robust alteration of Yap/Taz activity. These findings revealed the intricate regulatory mechanisms underlying the crosstalk between the Wnt/β-catenin and Hippo-Yap/Taz signaling, shedding light on their roles in orchestrating developmental processes in vivo.
Collapse
Affiliation(s)
- Matteo Astone
- Department of Biology, University of Padua, 35131 Padua, Italy
| | - Chiara Tesoriero
- Department of Biotechnology, University of Verona, 37134 Verona, Italy
| | - Marco Schiavone
- Department of Molecular and Translational Medicine, University of Brescia, 25123 Brescia, Italy
| | - Nicola Facchinello
- Neuroscience Institute, Italian National Research Council (CNR), 35131 Padua, Italy
| | - Natascia Tiso
- Department of Biology, University of Padua, 35131 Padua, Italy
| | | | - Andrea Vettori
- Department of Biotechnology, University of Verona, 37134 Verona, Italy
| |
Collapse
|
24
|
Anwar I, Wang X, Pratt RE, Dzau VJ, Hodgkinson CP. The impact of aging on cardiac repair and regeneration. J Biol Chem 2024; 300:107682. [PMID: 39159819 PMCID: PMC11414664 DOI: 10.1016/j.jbc.2024.107682] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2024] [Revised: 07/10/2024] [Accepted: 08/02/2024] [Indexed: 08/21/2024] Open
Abstract
In contrast to neonates and lower organisms, the adult mammalian heart lacks any capacity to regenerate following injury. The vast majority of our understanding of cardiac regeneration is based on research in young animals. Research in aged individuals is rare. This is unfortunate as aging induces many changes in the heart. The first part of this review covers the main technologies being pursued in the cardiac regeneration field and how they are impacted by the aging processes. The second part of the review covers the significant amount of aging-related research that could be used to aid cardiac regeneration. Finally, a perspective is provided to suggest how cardiac regenerative technologies can be improved by addressing aging-related effects.
Collapse
Affiliation(s)
- Iqra Anwar
- Mandel Center for Heart and Vascular Research, Duke Cardiovascular Research Center, Duke University Medical Center, Durham, North Carolina, USA
| | - Xinghua Wang
- Mandel Center for Heart and Vascular Research, Duke Cardiovascular Research Center, Duke University Medical Center, Durham, North Carolina, USA
| | - Richard E Pratt
- Mandel Center for Heart and Vascular Research, Duke Cardiovascular Research Center, Duke University Medical Center, Durham, North Carolina, USA
| | - Victor J Dzau
- Mandel Center for Heart and Vascular Research, Duke Cardiovascular Research Center, Duke University Medical Center, Durham, North Carolina, USA
| | - Conrad P Hodgkinson
- Mandel Center for Heart and Vascular Research, Duke Cardiovascular Research Center, Duke University Medical Center, Durham, North Carolina, USA.
| |
Collapse
|
25
|
Zheng K, Hao Y, Xia C, Cheng S, Yu J, Chen Z, Li Y, Niu Y, Ran S, Wang S, Ye W, Luo Z, Li X, Zhao J, Li R, Zong J, Zhang H, Lai L, Huang P, Zhou C, Xia J, Zhang X, Wu J. Effects and mechanisms of the myocardial microenvironment on cardiomyocyte proliferation and regeneration. Front Cell Dev Biol 2024; 12:1429020. [PMID: 39050889 PMCID: PMC11266095 DOI: 10.3389/fcell.2024.1429020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2024] [Accepted: 06/20/2024] [Indexed: 07/27/2024] Open
Abstract
The adult mammalian cardiomyocyte has a limited capacity for self-renewal, which leads to the irreversible heart dysfunction and poses a significant threat to myocardial infarction patients. In the past decades, research efforts have been predominantly concentrated on the cardiomyocyte proliferation and heart regeneration. However, the heart is a complex organ that comprises not only cardiomyocytes but also numerous noncardiomyocyte cells, all playing integral roles in maintaining cardiac function. In addition, cardiomyocytes are exposed to a dynamically changing physical environment that includes oxygen saturation and mechanical forces. Recently, a growing number of studies on myocardial microenvironment in cardiomyocyte proliferation and heart regeneration is ongoing. In this review, we provide an overview of recent advances in myocardial microenvironment, which plays an important role in cardiomyocyte proliferation and heart regeneration.
Collapse
Affiliation(s)
- Kexiao Zheng
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yanglin Hao
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Chenkun Xia
- Department of Anesthesiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Shaoxian Cheng
- Jingshan Union Hospital, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jizhang Yu
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Zhang Chen
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yuan Li
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yuqing Niu
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Shuan Ran
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Song Wang
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Weicong Ye
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Zilong Luo
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xiaohan Li
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jiulu Zhao
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Ran Li
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Junjie Zong
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Han Zhang
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Longyong Lai
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Pinyan Huang
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Cheng Zhou
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jiahong Xia
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xi Zhang
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jie Wu
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
26
|
Stairley RA, Trouten AM, Li S, Roddy PL, DeLeon-Pennell KY, Lee KH, Sucov HM, Liu C, Tao G. Anti-Ferroptotic Treatment Deteriorates Myocardial Infarction by Inhibiting Angiogenesis and Altering Immune Response. Antioxidants (Basel) 2024; 13:769. [PMID: 39061839 PMCID: PMC11273385 DOI: 10.3390/antiox13070769] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2024] [Revised: 06/16/2024] [Accepted: 06/20/2024] [Indexed: 07/28/2024] Open
Abstract
Mammalian cardiomyocytes have limited regenerative ability. Cardiac disease, such as congenital heart disease and myocardial infarction, causes an initial loss of cardiomyocytes through regulated cell death (RCD). Understanding the mechanisms that govern RCD in the injured myocardium is crucial for developing therapeutics to promote heart regeneration. We previously reported that ferroptosis, a non-apoptotic and iron-dependent form of RCD, is the main contributor to cardiomyocyte death in the injured heart. To investigate the mechanisms underlying the preference for ferroptosis in cardiomyocytes, we examined the effects of anti-ferroptotic reagents in infarcted mouse hearts. The results revealed that the anti-ferroptotic reagent did not improve neonatal heart regeneration, and further compromised the cardiac function of juvenile hearts. On the other hand, ferroptotic cardiomyocytes played a supportive role during wound healing by releasing pro-angiogenic factors. The inhibition of ferroptosis in the regenerating mouse heart altered the immune and angiogenic responses. Our study provides insights into the preference for ferroptosis over other types of RCD in stressed cardiomyocytes, and guidance for designing anti-cell-death therapies for treating heart disease.
Collapse
Affiliation(s)
- Rebecca A. Stairley
- Department of Regenerative Medicine and Cell Biology, Medical University of South Carolina, Charleston, SC 29425, USA; (R.A.S.); (A.M.T.); (S.L.); (P.L.R.); (H.M.S.)
| | - Allison M. Trouten
- Department of Regenerative Medicine and Cell Biology, Medical University of South Carolina, Charleston, SC 29425, USA; (R.A.S.); (A.M.T.); (S.L.); (P.L.R.); (H.M.S.)
| | - Shuang Li
- Department of Regenerative Medicine and Cell Biology, Medical University of South Carolina, Charleston, SC 29425, USA; (R.A.S.); (A.M.T.); (S.L.); (P.L.R.); (H.M.S.)
- Department of Pediatrics, Herman B Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Patrick L. Roddy
- Department of Regenerative Medicine and Cell Biology, Medical University of South Carolina, Charleston, SC 29425, USA; (R.A.S.); (A.M.T.); (S.L.); (P.L.R.); (H.M.S.)
| | - Kristine Y. DeLeon-Pennell
- Division of Cardiology, Department of Medicine, Medical University of South Carolina, Charleston, SC 29425, USA;
- Research Service, Ralph H. Johnson Veterans Affairs Medical Center, Charleston, SC 29401, USA
| | - Kyu-Ho Lee
- Department of Medicine Digestive Disease Research Core Center, Medical University of South Carolina, Charleston, SC 29425, USA;
| | - Henry M. Sucov
- Department of Regenerative Medicine and Cell Biology, Medical University of South Carolina, Charleston, SC 29425, USA; (R.A.S.); (A.M.T.); (S.L.); (P.L.R.); (H.M.S.)
- Division of Cardiology, Department of Medicine, Medical University of South Carolina, Charleston, SC 29425, USA;
| | - Chun Liu
- Department of Physiology, Medical College of Wisconsin, Milwaukee, WI 53226, USA;
| | - Ge Tao
- Department of Regenerative Medicine and Cell Biology, Medical University of South Carolina, Charleston, SC 29425, USA; (R.A.S.); (A.M.T.); (S.L.); (P.L.R.); (H.M.S.)
| |
Collapse
|
27
|
Zhu Y, Ackers-Johnson M, Shanmugam MK, Pakkiri LS, Drum CL, Yanpu C, Kim J, Paltzer WG, Mahmoud AI, Wen Tan WL, Lee MCJ, Jianming J, Luu DAT, Ng SL, Li PYQ, Anhui W, Rong Q, Ong GJX, Ng Yu T, Haigh JJ, Tiang Z, Richards AM, Foo R. Asparagine Synthetase Marks a Distinct Dependency Threshold for Cardiomyocyte Dedifferentiation. Circulation 2024; 149:1833-1851. [PMID: 38586957 PMCID: PMC11147732 DOI: 10.1161/circulationaha.123.063965] [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: 03/05/2023] [Accepted: 01/23/2024] [Indexed: 04/09/2024]
Abstract
BACKGROUND Adult mammalian cardiomyocytes have limited proliferative capacity, but in specifically induced contexts they traverse through cell-cycle reentry, offering the potential for heart regeneration. Endogenous cardiomyocyte proliferation is preceded by cardiomyocyte dedifferentiation (CMDD), wherein adult cardiomyocytes revert to a less matured state that is distinct from the classical myocardial fetal stress gene response associated with heart failure. However, very little is known about CMDD as a defined cardiomyocyte cell state in transition. METHODS Here, we leveraged 2 models of in vitro cultured adult mouse cardiomyocytes and in vivo adeno-associated virus serotype 9 cardiomyocyte-targeted delivery of reprogramming factors (Oct4, Sox2, Klf4, and Myc) in adult mice to study CMDD. We profiled their transcriptomes using RNA sequencing, in combination with multiple published data sets, with the aim of identifying a common denominator for tracking CMDD. RESULTS RNA sequencing and integrated analysis identified Asparagine Synthetase (Asns) as a unique molecular marker gene well correlated with CMDD, required for increased asparagine and also for distinct fluxes in other amino acids. Although Asns overexpression in Oct4, Sox2, Klf4, and Myc cardiomyocytes augmented hallmarks of CMDD, Asns deficiency led to defective regeneration in the neonatal mouse myocardial infarction model, increased cell death of cultured adult cardiomyocytes, and reduced cell cycle in Oct4, Sox2, Klf4, and Myc cardiomyocytes, at least in part through disrupting the mammalian target of rapamycin complex 1 pathway. CONCLUSIONS We discovered a novel gene Asns as both a molecular marker and an essential mediator, marking a distinct threshold that appears in common for at least 4 models of CMDD, and revealing an Asns/mammalian target of rapamycin complex 1 axis dependency for dedifferentiating cardiomyocytes. Further study will be needed to extrapolate and assess its relevance to other cell state transitions as well as in heart regeneration.
Collapse
Affiliation(s)
- Yike Zhu
- Cardiovascular Research Institute, Yong Loo Lin School of Medicine, National University of Singapore
- Cardiovascular Metabolic Disease Translational Research Programme, National University Health Systems, Singapore
| | - Matthew Ackers-Johnson
- Cardiovascular Research Institute, Yong Loo Lin School of Medicine, National University of Singapore
- Cardiovascular Metabolic Disease Translational Research Programme, National University Health Systems, Singapore
| | - Muthu K Shanmugam
- Cardiovascular Research Institute, Yong Loo Lin School of Medicine, National University of Singapore
- Cardiovascular Metabolic Disease Translational Research Programme, National University Health Systems, Singapore
| | - Leroy Sivappiragasam Pakkiri
- Cardiovascular Research Institute, Yong Loo Lin School of Medicine, National University of Singapore
- Cardiovascular Metabolic Disease Translational Research Programme, National University Health Systems, Singapore
| | - Chester Lee Drum
- Cardiovascular Research Institute, Yong Loo Lin School of Medicine, National University of Singapore
- Cardiovascular Metabolic Disease Translational Research Programme, National University Health Systems, Singapore
| | - Chen Yanpu
- Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany
| | - Johnny Kim
- Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany
- German Centre for Cardiovascular Research (DZHK), Partner site Rhein/Main, Bad Nauheim, Germany
| | - Wyatt G. Paltzer
- Department of Cell and Regenerative Biology, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, USA
| | - Ahmed I. Mahmoud
- Department of Cell and Regenerative Biology, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, USA
| | - Wilson Lek Wen Tan
- Cardiovascular Research Institute, Yong Loo Lin School of Medicine, National University of Singapore
- Cardiovascular Metabolic Disease Translational Research Programme, National University Health Systems, Singapore
| | - Mick Chang Jie Lee
- Cardiovascular Research Institute, Yong Loo Lin School of Medicine, National University of Singapore
- Cardiovascular Metabolic Disease Translational Research Programme, National University Health Systems, Singapore
| | - Jiang Jianming
- Cardiovascular Research Institute, Yong Loo Lin School of Medicine, National University of Singapore
- Cardiovascular Metabolic Disease Translational Research Programme, National University Health Systems, Singapore
| | - Danh Anh Tuan Luu
- Cardiovascular Research Institute, Yong Loo Lin School of Medicine, National University of Singapore
- Cardiovascular Metabolic Disease Translational Research Programme, National University Health Systems, Singapore
| | - Shi Ling Ng
- Cardiovascular Research Institute, Yong Loo Lin School of Medicine, National University of Singapore
- Cardiovascular Metabolic Disease Translational Research Programme, National University Health Systems, Singapore
| | - Peter Yi Qing Li
- Cardiovascular Research Institute, Yong Loo Lin School of Medicine, National University of Singapore
- Cardiovascular Metabolic Disease Translational Research Programme, National University Health Systems, Singapore
| | - Wang Anhui
- Department of Pharmacology, School of Basic Medical Sciences, Peking University Health Science Centre, Peking University
- State Key Laboratory of Vascular Homeostasis and Remodelling, Peking University
| | - Qi Rong
- Department of Pharmacology, School of Basic Medical Sciences, Peking University Health Science Centre, Peking University
- State Key Laboratory of Vascular Homeostasis and Remodelling, Peking University
| | - Gabriel Jing Xiang Ong
- Cardiovascular Research Institute, Yong Loo Lin School of Medicine, National University of Singapore
- Cardiovascular Metabolic Disease Translational Research Programme, National University Health Systems, Singapore
| | - Timothy Ng Yu
- Cardiovascular Research Institute, Yong Loo Lin School of Medicine, National University of Singapore
- Cardiovascular Metabolic Disease Translational Research Programme, National University Health Systems, Singapore
| | - Jody J. Haigh
- CancerCare Manitoba Research Institute, Department of Pharmacology and Therapeutics, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, Manitoba, Canada
- VIB, Flanders Institute for Biotechnology, Ghent University, Ghent, Belgium
| | - Zenia Tiang
- Cardiovascular Research Institute, Yong Loo Lin School of Medicine, National University of Singapore
- Cardiovascular Metabolic Disease Translational Research Programme, National University Health Systems, Singapore
| | - A. Mark Richards
- Cardiovascular Research Institute, Yong Loo Lin School of Medicine, National University of Singapore
- Cardiovascular Metabolic Disease Translational Research Programme, National University Health Systems, Singapore
- Christchurch Heart Institute, University of Otago, New Zealand
| | - Roger Foo
- Cardiovascular Research Institute, Yong Loo Lin School of Medicine, National University of Singapore
- Cardiovascular Metabolic Disease Translational Research Programme, National University Health Systems, Singapore
- Institute of Molecular and Cell Biology, A*STAR, Singapore
| |
Collapse
|
28
|
Liu S, Deshmukh V, Wang F, Liang J, Cusick J, Li X, Martin JF. Myocardial Infarction Suppresses Protein Synthesis and Causes Decoupling of Transcription and Translation. JACC Basic Transl Sci 2024; 9:792-807. [PMID: 39070274 PMCID: PMC11282883 DOI: 10.1016/j.jacbts.2024.02.014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/22/2024] [Revised: 02/23/2024] [Accepted: 02/23/2024] [Indexed: 07/30/2024]
Abstract
Gene expression involves transcription, translation, and mRNA and protein degradation. Advanced RNA sequencing measures mRNA levels for cell state assessment, but mRNA level does not fully reflect protein level. Identifying heart cell proteomes and their stress response is crucial. Using a cardiomyocyte-specific mouse model, we tracked protein synthesis after myocardial infarction. Our results showed that myocardial infarction suppresses protein synthesis and unveils a decoupling of translation and transcription regulation in cardiomyocytes.
Collapse
Affiliation(s)
- Shijie Liu
- Cardiomyocyte Renewal Laboratory, Texas Heart Institute, Houston, Texas, USA
- (currently) Division of Molecular Cardiovascular Biology, Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio, USA
- Division of Molecular Cardiovascular Biology, Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio, USA
| | - Vaibhav Deshmukh
- Department of Molecular Physiology and Biophysics, Baylor College of Medicine, Houston, Texas, USA
| | - Fangfei Wang
- Division of Molecular Cardiovascular Biology, Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio, USA
| | - Jie Liang
- Division of Molecular Cardiovascular Biology, Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio, USA
| | - Jenna Cusick
- Division of Molecular Cardiovascular Biology, Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio, USA
| | - Xiao Li
- Gene Editing Laboratory, Texas Heart Institute, Houston, Texas, USA
| | - James F. Martin
- Cardiomyocyte Renewal Laboratory, Texas Heart Institute, Houston, Texas, USA
- Department of Molecular Physiology and Biophysics, Baylor College of Medicine, Houston, Texas, USA
- Gene Editing Laboratory, Texas Heart Institute, Houston, Texas, USA
- Cardiovascular Research Institute, Baylor College of Medicine, Houston, Texas, USA
| |
Collapse
|
29
|
Zhu C, Yuan T, Krishnan J. Targeting cardiomyocyte cell cycle regulation in heart failure. Basic Res Cardiol 2024; 119:349-369. [PMID: 38683371 PMCID: PMC11142990 DOI: 10.1007/s00395-024-01049-x] [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: 11/21/2023] [Revised: 03/11/2024] [Accepted: 03/29/2024] [Indexed: 05/01/2024]
Abstract
Heart failure continues to be a significant global health concern, causing substantial morbidity and mortality. The limited ability of the adult heart to regenerate has posed challenges in finding effective treatments for cardiac pathologies. While various medications and surgical interventions have been used to improve cardiac function, they are not able to address the extensive loss of functioning cardiomyocytes that occurs during cardiac injury. As a result, there is growing interest in understanding how the cell cycle is regulated and exploring the potential for stimulating cardiomyocyte proliferation as a means of promoting heart regeneration. This review aims to provide an overview of current knowledge on cell cycle regulation and mechanisms underlying cardiomyocyte proliferation in cases of heart failure, while also highlighting established and novel therapeutic strategies targeting this area for treatment purposes.
Collapse
Affiliation(s)
- Chaonan Zhu
- Department of Medicine III, Cardiology/Angiology/Nephrology, Goethe University Hospital, 60590, Frankfurt am Main, Germany
- Institute for Cardiovascular Regeneration, Goethe University, 60590, Frankfurt am Main, Germany
| | - Ting Yuan
- Department of Medicine III, Cardiology/Angiology/Nephrology, Goethe University Hospital, 60590, Frankfurt am Main, Germany.
- Institute for Cardiovascular Regeneration, Goethe University, 60590, Frankfurt am Main, Germany.
- German Center for Cardiovascular Research, Partner Site Rhein-Main, 60590, Frankfurt am Main, Germany.
- Cardio-Pulmonary Institute, Goethe University Hospital, 60590, Frankfurt am Main, Germany.
| | - Jaya Krishnan
- Department of Medicine III, Cardiology/Angiology/Nephrology, Goethe University Hospital, 60590, Frankfurt am Main, Germany.
- Institute for Cardiovascular Regeneration, Goethe University, 60590, Frankfurt am Main, Germany.
- German Center for Cardiovascular Research, Partner Site Rhein-Main, 60590, Frankfurt am Main, Germany.
- Cardio-Pulmonary Institute, Goethe University Hospital, 60590, Frankfurt am Main, Germany.
| |
Collapse
|
30
|
Abou Nader N, Charrier L, Meisnsohn MC, Banville L, Deffrennes B, St-Jean G, Boerboom D, Zamberlam G, Brind'Amour J, Pépin D, Boyer A. Lats1 and Lats2 regulate YAP and TAZ activity to control the development of mouse Sertoli cells. FASEB J 2024; 38:e23633. [PMID: 38690712 DOI: 10.1096/fj.202400346r] [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: 02/19/2024] [Revised: 04/11/2024] [Accepted: 04/16/2024] [Indexed: 05/02/2024]
Abstract
Recent reports suggest that the Hippo signaling pathway regulates testis development, though its exact roles in Sertoli cell differentiation remain unknown. Here, we examined the functions of the main Hippo pathway kinases, large tumor suppressor homolog kinases 1 and 2 (Lats1 and Lats2) in developing mouse Sertoli cells. Conditional inactivation of Lats1/2 in Sertoli cells resulted in the disorganization and overgrowth of the testis cords, the induction of a testicular inflammatory response and germ cell apoptosis. Stimulated by retinoic acid 8 (STRA8) expression in germ cells additionally suggested that germ cells may have been preparing to enter meiosis prior to their loss. Gene expression analyses of the developing testes of conditional knockout animals further suggested impaired Sertoli cell differentiation, epithelial-to-mesenchymal transition, and the induction of a specific set of genes associated with Yes-associated protein (YAP) and transcriptional coactivator with PDZ-binding motif (TAZ)-mediated integrin signaling. Finally, the involvement of YAP/TAZ in Sertoli cell differentiation was confirmed by concomitantly inactivating Yap/Taz in Lats1/2 conditional knockout model, which resulted in a partial rescue of the testicular phenotypic changes. Taken together, these results identify Hippo signaling as a crucial pathway for Sertoli cell development and provide novel insight into Sertoli cell fate maintenance.
Collapse
Affiliation(s)
- Nour Abou Nader
- Centre de Recherche en Reproduction et Fertilité, Faculté de Médecine Vétérinaire, Université de Montréal, Saint-Hyacinthe, Quebec, Canada
| | - Laureline Charrier
- Centre de Recherche en Reproduction et Fertilité, Faculté de Médecine Vétérinaire, Université de Montréal, Saint-Hyacinthe, Quebec, Canada
| | - Marie-Charlotte Meisnsohn
- Pediatric Surgical Research Laboratories, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Laurence Banville
- Centre de Recherche en Reproduction et Fertilité, Faculté de Médecine Vétérinaire, Université de Montréal, Saint-Hyacinthe, Quebec, Canada
| | - Bérengère Deffrennes
- Centre de Recherche en Reproduction et Fertilité, Faculté de Médecine Vétérinaire, Université de Montréal, Saint-Hyacinthe, Quebec, Canada
- École Nationale Vétérinaire d'Alfort, Maisons-Alfort, France
| | - Guillaume St-Jean
- Centre de Recherche en Reproduction et Fertilité, Faculté de Médecine Vétérinaire, Université de Montréal, Saint-Hyacinthe, Quebec, Canada
| | - Derek Boerboom
- Centre de Recherche en Reproduction et Fertilité, Faculté de Médecine Vétérinaire, Université de Montréal, Saint-Hyacinthe, Quebec, Canada
| | - Gustavo Zamberlam
- Centre de Recherche en Reproduction et Fertilité, Faculté de Médecine Vétérinaire, Université de Montréal, Saint-Hyacinthe, Quebec, Canada
| | - Julie Brind'Amour
- Centre de Recherche en Reproduction et Fertilité, Faculté de Médecine Vétérinaire, Université de Montréal, Saint-Hyacinthe, Quebec, Canada
| | - David Pépin
- Pediatric Surgical Research Laboratories, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Alexandre Boyer
- Centre de Recherche en Reproduction et Fertilité, Faculté de Médecine Vétérinaire, Université de Montréal, Saint-Hyacinthe, Quebec, Canada
| |
Collapse
|
31
|
Namoto K, Baader C, Orsini V, Landshammer A, Breuer E, Dinh KT, Ungricht R, Pikiolek M, Laurent S, Lu B, Aebi A, Schönberger K, Vangrevelinghe E, Evrova O, Sun T, Annunziato S, Lachal J, Redmond E, Wang L, Wetzel K, Capodieci P, Turner J, Schutzius G, Unterreiner V, Trunzer M, Buschmann N, Behnke D, Machauer R, Scheufler C, Parker CN, Ferro M, Grevot A, Beyerbach A, Lu WY, Forbes SJ, Wagner J, Bouwmeester T, Liu J, Sohal B, Sahambi S, Greenbaum LE, Lohmann F, Hoppe P, Cong F, Sailer AW, Ruffner H, Glatthar R, Humar B, Clavien PA, Dill MT, George E, Maibaum J, Liberali P, Tchorz JS. NIBR-LTSi is a selective LATS kinase inhibitor activating YAP signaling and expanding tissue stem cells in vitro and in vivo. Cell Stem Cell 2024; 31:554-569.e17. [PMID: 38579685 DOI: 10.1016/j.stem.2024.03.003] [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: 07/26/2022] [Revised: 01/24/2024] [Accepted: 03/06/2024] [Indexed: 04/07/2024]
Abstract
The YAP/Hippo pathway is an organ growth and size regulation rheostat safeguarding multiple tissue stem cell compartments. LATS kinases phosphorylate and thereby inactivate YAP, thus representing a potential direct drug target for promoting tissue regeneration. Here, we report the identification and characterization of the selective small-molecule LATS kinase inhibitor NIBR-LTSi. NIBR-LTSi activates YAP signaling, shows good oral bioavailability, and expands organoids derived from several mouse and human tissues. In tissue stem cells, NIBR-LTSi promotes proliferation, maintains stemness, and blocks differentiation in vitro and in vivo. NIBR-LTSi accelerates liver regeneration following extended hepatectomy in mice. However, increased proliferation and cell dedifferentiation in multiple organs prevent prolonged systemic LATS inhibition, thus limiting potential therapeutic benefit. Together, we report a selective LATS kinase inhibitor agonizing YAP signaling and promoting tissue regeneration in vitro and in vivo, enabling future research on the regenerative potential of the YAP/Hippo pathway.
Collapse
Affiliation(s)
- Kenji Namoto
- Biomedical Research, Novartis Pharma AG, Basel, Switzerland.
| | - Clara Baader
- Friedrich Miescher Institute for Biomedical Research, Basel, Switzerland; University of Basel, Basel, Switzerland
| | - Vanessa Orsini
- Biomedical Research, Novartis Pharma AG, Basel, Switzerland
| | | | - Eva Breuer
- University Hospital Zurich (USZ), Zurich, Switzerland
| | - Kieu Trinh Dinh
- German Cancer Research Center (DKFZ) Heidelberg, Research Group Experimental Hepatology, Inflammation and Cancer, Heidelberg, Germany; Faculty of Biosciences, Heidelberg University, Heidelberg, Germany
| | | | | | | | - Bo Lu
- Biomedical Research, Novartis Pharma AG, Cambridge, MA, USA
| | - Alexandra Aebi
- Biomedical Research, Novartis Pharma AG, Basel, Switzerland
| | | | | | - Olivera Evrova
- Biomedical Research, Novartis Pharma AG, Basel, Switzerland
| | - Tianliang Sun
- Biomedical Research, Novartis Pharma AG, Basel, Switzerland; Division of Liver Diseases, Institute for Regenerative Medicine, Department of Cell, Developmental and Regenerative Biology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | | | - Julie Lachal
- Biomedical Research, Novartis Pharma AG, Basel, Switzerland
| | - Emily Redmond
- Biomedical Research, Novartis Pharma AG, Cambridge, MA, USA
| | - Louis Wang
- Biomedical Research, Novartis Pharma AG, Cambridge, MA, USA
| | - Kristie Wetzel
- Biomedical Research, Novartis Pharma AG, Cambridge, MA, USA
| | | | | | - Gabi Schutzius
- Biomedical Research, Novartis Pharma AG, Basel, Switzerland
| | | | - Markus Trunzer
- Biomedical Research, Novartis Pharma AG, Basel, Switzerland
| | | | - Dirk Behnke
- Biomedical Research, Novartis Pharma AG, Basel, Switzerland
| | | | | | | | - Magali Ferro
- Biomedical Research, Novartis Pharma AG, Basel, Switzerland
| | - Armelle Grevot
- Biomedical Research, Novartis Pharma AG, Basel, Switzerland
| | | | - Wei-Yu Lu
- University of Edinburgh, Center for Inflammation Research, Edinburgh, UK
| | - Stuart J Forbes
- University of Edinburgh, Center for Regenerative Medicine, Edinburgh, UK
| | - Jürgen Wagner
- Biomedical Research, Novartis Pharma AG, Basel, Switzerland
| | | | - Jun Liu
- Biomedical Research, Novartis Pharma AG, La Jolla, CA, USA
| | - Bindi Sohal
- Biomedical Research, Novartis Pharma AG, Basel, Switzerland
| | | | | | - Felix Lohmann
- Biomedical Research, Novartis Pharma AG, Basel, Switzerland
| | - Philipp Hoppe
- Biomedical Research, Novartis Pharma AG, Basel, Switzerland
| | - Feng Cong
- Biomedical Research, Novartis Pharma AG, Cambridge, MA, USA
| | | | - Heinz Ruffner
- Biomedical Research, Novartis Pharma AG, Basel, Switzerland
| | - Ralf Glatthar
- Biomedical Research, Novartis Pharma AG, Basel, Switzerland
| | - Bostjan Humar
- University Hospital Zurich (USZ), Zurich, Switzerland
| | | | - Michael T Dill
- German Cancer Research Center (DKFZ) Heidelberg, Research Group Experimental Hepatology, Inflammation and Cancer, Heidelberg, Germany; Department of Gastroenterology, Infectious Diseases and Intoxication, Heidelberg University Hospital, Heidelberg, Germany
| | | | - Jürgen Maibaum
- Biomedical Research, Novartis Pharma AG, Basel, Switzerland
| | - Prisca Liberali
- Friedrich Miescher Institute for Biomedical Research, Basel, Switzerland; University of Basel, Basel, Switzerland
| | - Jan S Tchorz
- Biomedical Research, Novartis Pharma AG, Basel, Switzerland.
| |
Collapse
|
32
|
Zhong Z, Jiao Z, Yu FX. The Hippo signaling pathway in development and regeneration. Cell Rep 2024; 43:113926. [PMID: 38457338 DOI: 10.1016/j.celrep.2024.113926] [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: 12/02/2023] [Revised: 02/05/2024] [Accepted: 02/20/2024] [Indexed: 03/10/2024] Open
Abstract
The Hippo signaling pathway is a central growth control mechanism in multicellular organisms. By integrating diverse mechanical, biochemical, and stress cues, the Hippo pathway orchestrates proliferation, survival, differentiation, and mechanics of cells, which in turn regulate organ development, homeostasis, and regeneration. A deep understanding of the regulation and function of the Hippo pathway therefore holds great promise for developing novel therapeutics in regenerative medicine. Here, we provide updates on the molecular organization of the mammalian Hippo signaling network, review the regulatory signals and functional outputs of the pathway, and discuss the roles of Hippo signaling in development and regeneration.
Collapse
Affiliation(s)
- Zhenxing Zhong
- Institute of Pediatrics, Children's Hospital of Fudan University, and the Shanghai Key Laboratory of Medical Epigenetics, International Co-laboratory of Medical Epigenetics and Metabolism, State Key Laboratory of Genetic Engineering, Institutes of Biomedical Sciences, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Zhihan Jiao
- Institute of Pediatrics, Children's Hospital of Fudan University, and the Shanghai Key Laboratory of Medical Epigenetics, International Co-laboratory of Medical Epigenetics and Metabolism, State Key Laboratory of Genetic Engineering, Institutes of Biomedical Sciences, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Fa-Xing Yu
- Institute of Pediatrics, Children's Hospital of Fudan University, and the Shanghai Key Laboratory of Medical Epigenetics, International Co-laboratory of Medical Epigenetics and Metabolism, State Key Laboratory of Genetic Engineering, Institutes of Biomedical Sciences, Shanghai Medical College, Fudan University, Shanghai 200032, China.
| |
Collapse
|
33
|
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
|
34
|
Constanty F, Wu B, Wei KH, Lin IT, Dallmann J, Guenther S, Lautenschlaeger T, Priya R, Lai SL, Stainier DYR, Beisaw A. Border-zone cardiomyocytes and macrophages contribute to remodeling of the extracellular matrix to promote cardiomyocyte invasion during zebrafish cardiac regeneration. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.03.12.584570. [PMID: 38559277 PMCID: PMC10980021 DOI: 10.1101/2024.03.12.584570] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/04/2024]
Abstract
Despite numerous advances in our understanding of zebrafish cardiac regeneration, an aspect that remains less studied is how regenerating cardiomyocytes invade, and eventually replace, the collagen-containing fibrotic tissue following injury. Here, we provide an in-depth analysis of the process of cardiomyocyte invasion using live-imaging and histological approaches. We observed close interactions between protruding cardiomyocytes and macrophages at the wound border zone, and macrophage-deficient irf8 mutant zebrafish exhibited defects in extracellular matrix (ECM) remodeling and cardiomyocyte protrusion into the injured area. Using a resident macrophage ablation model, we show that defects in ECM remodeling at the border zone and subsequent cardiomyocyte protrusion can be partly attributed to a population of resident macrophages. Single-cell RNA-sequencing analysis of cells at the wound border revealed a population of cardiomyocytes and macrophages with fibroblast-like gene expression signatures, including the expression of genes encoding ECM structural proteins and ECM-remodeling proteins. The expression of mmp14b , which encodes a membrane-anchored matrix metalloproteinase, was restricted to cells in the border zone, including cardiomyocytes, macrophages, fibroblasts, and endocardial/endothelial cells. Genetic deletion of mmp14b led to a decrease in 1) macrophage recruitment to the border zone, 2) collagen degradation at the border zone, and 3) subsequent cardiomyocyte invasion. Furthermore, cardiomyocyte-specific overexpression of mmp14b was sufficient to enhance cardiomyocyte invasion into the injured tissue and along the apical surface of the wound. Altogether, our data shed important insights into the process of cardiomyocyte invasion of the collagen-containing injured tissue during cardiac regeneration. They further suggest that cardiomyocytes and resident macrophages contribute to ECM remodeling at the border zone to promote cardiomyocyte replenishment of the fibrotic injured tissue.
Collapse
|
35
|
Tan FH, Bronner ME. Regenerative loss in the animal kingdom as viewed from the mouse digit tip and heart. Dev Biol 2024; 507:44-63. [PMID: 38145727 PMCID: PMC10922877 DOI: 10.1016/j.ydbio.2023.12.008] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2023] [Revised: 11/30/2023] [Accepted: 12/19/2023] [Indexed: 12/27/2023]
Abstract
The myriad regenerative abilities across the animal kingdom have fascinated us for centuries. Recent advances in developmental, molecular, and cellular biology have allowed us to unearth a surprising diversity of mechanisms through which these processes occur. Developing an all-encompassing theory of animal regeneration has thus proved a complex endeavor. In this chapter, we frame the evolution and loss of animal regeneration within the broad developmental constraints that may physiologically inhibit regenerative ability across animal phylogeny. We then examine the mouse as a model of regeneration loss, specifically the experimental systems of the digit tip and heart. We discuss the digit tip and heart as a positionally-limited system of regeneration and a temporally-limited system of regeneration, respectively. We delve into the physiological processes involved in both forms of regeneration, and how each phase of the healing and regenerative process may be affected by various molecular signals, systemic changes, or microenvironmental cues. Lastly, we also discuss the various approaches and interventions used to induce or improve the regenerative response in both contexts, and the implications they have for our understanding regenerative ability more broadly.
Collapse
Affiliation(s)
- Fayth Hui Tan
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA 91125, USA
| | - Marianne E Bronner
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA 91125, USA.
| |
Collapse
|
36
|
Rehmani T, Dias AP, Kamal M, Salih M, Tuana BS. Deletion of Sarcolemmal Membrane-Associated Protein Isoform 3 (SLMAP3) in Cardiac Progenitors Delays Embryonic Growth of Myocardium without Affecting Hippo Pathway. Int J Mol Sci 2024; 25:2888. [PMID: 38474134 DOI: 10.3390/ijms25052888] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2023] [Revised: 02/18/2024] [Accepted: 02/21/2024] [Indexed: 03/14/2024] Open
Abstract
The slmap gene is alternatively spliced to generate many isoforms that are abundant in developing myocardium. The largest protein isoform SLMAP3 is ubiquitously expressed and has been linked to cardiomyopathy, Brugada syndrome and Hippo signaling. To examine any role in cardiogenesis, mice homozygous for floxed slmap allele were crossed with Nkx2.5-cre mice to nullify its expression in cardiac progenitors. Targeted deletion of the slmap gene resulted in the specific knockout (KO) of the SLMAP3 (~91 KDa) isoform without any changes in the expression of the SLMAP2 (~43 kDa) or the SLMAP1 (~35 kDa) isoforms which continued to accumulate to similar levels as seen in Wt embryonic hearts. The loss of SLMAP3 from cardiac progenitors resulted in decreased size of the developing embryonic hearts evident at E9.5 to E16.5 with four small chambers and significantly thinner left ventricles. The proliferative capacity assessed with the phosphorylation of histone 3 or with Ki67 in E12.5 hearts was not significantly altered due to SLMAP3 deficiency. The size of embryonic cardiomyocytes, marked with anti-Troponin C, revealed significantly smaller cells, but their hypertrophic response (AKT1 and MTOR1) was not significantly affected by the specific loss of SLMAP3 protein. Further, no changes in phosphorylation of MST1/2 or YAP were detected in SLMAP3-KO embryonic myocardium, ruling out any impact on Hippo signaling. Rat embryonic cardiomyocytes express the three SLMAP isoforms and their knockdown (KD) with sh-RNA, resulted in decreased proliferation and enhanced senescence but without any impact on Hippo signaling. Collectively, these data show that SLMAP is critical for normal cardiac development with potential for the various isoforms to serve compensatory roles. Our data imply novel mechanisms for SLMAP action in cardiac growth independent of Hippo signaling.
Collapse
Affiliation(s)
- Taha Rehmani
- Department of Cellular and Molecular Medicine, Faculty of Medicine, University of Ottawa, 451 Smyth Road, Ottawa, ON K1H 8M5, Canada
| | - Ana Paula Dias
- Department of Cellular and Molecular Medicine, Faculty of Medicine, University of Ottawa, 451 Smyth Road, Ottawa, ON K1H 8M5, Canada
| | - Marsel Kamal
- Department of Cellular and Molecular Medicine, Faculty of Medicine, University of Ottawa, 451 Smyth Road, Ottawa, ON K1H 8M5, Canada
| | - Maysoon Salih
- Department of Cellular and Molecular Medicine, Faculty of Medicine, University of Ottawa, 451 Smyth Road, Ottawa, ON K1H 8M5, Canada
| | - Balwant S Tuana
- Department of Cellular and Molecular Medicine, Faculty of Medicine, University of Ottawa, 451 Smyth Road, Ottawa, ON K1H 8M5, Canada
| |
Collapse
|
37
|
Li RG, Li X, Morikawa Y, Grisanti-Canozo FJ, Meng F, Tsai CR, Zhao Y, Liu L, Kim J, Xie B, Klysik E, Liu S, Samee MAH, Martin JF. YAP induces a neonatal-like pro-renewal niche in the adult heart. NATURE CARDIOVASCULAR RESEARCH 2024; 3:283-300. [PMID: 38510108 PMCID: PMC10954255 DOI: 10.1038/s44161-024-00428-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/18/2022] [Accepted: 01/12/2024] [Indexed: 03/22/2024]
Abstract
After myocardial infarction (MI), mammalian hearts do not regenerate, and the microenvironment is disrupted. Hippo signaling loss of function with activation of transcriptional co-factor YAP induces heart renewal and rebuilds the post-MI microenvironment. In this study, we investigated adult renewal-competent mouse hearts expressing an active version of YAP, called YAP5SA, in cardiomyocytes (CMs). Spatial transcriptomics and single-cell RNA sequencing revealed a conserved, renewal-competent CM cell state called adult (a)CM2 with high YAP activity. aCM2 co-localized with cardiac fibroblasts (CFs) expressing complement pathway component C3 and macrophages (MPs) expressing C3ar1 receptor to form a cellular triad in YAP5SA hearts and renewal-competent neonatal hearts. Although aCM2 was detected in adult mouse and human hearts, the cellular triad failed to co-localize in these non-renewing hearts. C3 and C3ar1 loss-of-function experiments indicated that C3a signaling between MPs and CFs was required to assemble the pro-renewal aCM2, C3+ CF and C3ar1+ MP cellular triad.
Collapse
Affiliation(s)
- Rich Gang Li
- Cardiomyocyte Renewal Laboratory, Texas Heart Institute, Houston, TX, USA
- McGill Gene Editing Laboratory, Texas Heart Institute, Houston, TX, USA
- These authors contributed equally: Rich Gang Li, Xiao Li
| | - Xiao Li
- McGill Gene Editing Laboratory, Texas Heart Institute, Houston, TX, USA
- These authors contributed equally: Rich Gang Li, Xiao Li
| | - Yuka Morikawa
- Cardiomyocyte Renewal Laboratory, Texas Heart Institute, Houston, TX, USA
| | - Francisco J. Grisanti-Canozo
- Cardiomyocyte Renewal Laboratory, Texas Heart Institute, Houston, TX, USA
- Department of Integrative Physiology, Baylor College of Medicine, Houston, TX, USA
| | - Fansen Meng
- Department of Integrative Physiology, Baylor College of Medicine, Houston, TX, USA
| | - Chang-Ru Tsai
- Department of Integrative Physiology, Baylor College of Medicine, Houston, TX, USA
| | - Yi Zhao
- McGill Gene Editing Laboratory, Texas Heart Institute, Houston, TX, USA
| | - Lin Liu
- McGill Gene Editing Laboratory, Texas Heart Institute, Houston, TX, USA
| | - Jong Kim
- Cardiomyocyte Renewal Laboratory, Texas Heart Institute, Houston, TX, USA
| | - Bing Xie
- Department of Integrative Physiology, Baylor College of Medicine, Houston, TX, USA
| | - Elzbieta Klysik
- Department of Integrative Physiology, Baylor College of Medicine, Houston, TX, USA
| | - Shijie Liu
- Cardiomyocyte Renewal Laboratory, Texas Heart Institute, Houston, TX, USA
| | - Md Abul Hassan Samee
- Department of Integrative Physiology, Baylor College of Medicine, Houston, TX, USA
| | - James F. Martin
- Cardiomyocyte Renewal Laboratory, Texas Heart Institute, Houston, TX, USA
- McGill Gene Editing Laboratory, Texas Heart Institute, Houston, TX, USA
- Department of Integrative Physiology, Baylor College of Medicine, Houston, TX, USA
- Center for Organ Repair and Renewal, Baylor College of Medicine, Houston, TX, USA
| |
Collapse
|
38
|
Zhang F, Zhou H, Xue J, Zhang Y, Zhou L, Leng J, Fang G, Liu Y, Wang Y, Liu H, Wu Y, Qi L, Duan R, He X, Wang Y, Liu Y, Li L, Yang J, Liang D, Chen YH. Deficiency of Transcription Factor Sp1 Contributes to Hypertrophic Cardiomyopathy. Circ Res 2024; 134:290-306. [PMID: 38197258 DOI: 10.1161/circresaha.123.323272] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/28/2023] [Accepted: 01/02/2024] [Indexed: 01/11/2024]
Abstract
BACKGROUND Hypertrophic cardiomyopathy (HCM) is the most prevalent monogenic heart disorder. However, the pathogenesis of HCM, especially its nongenetic mechanisms, remains largely unclear. Transcription factors are known to be involved in various biological processes including cell growth. We hypothesized that SP1 (specificity protein 1), the first purified TF in mammals, plays a role in the cardiomyocyte growth and cardiac hypertrophy of HCM. METHODS Cardiac-specific conditional knockout of Sp1 mice were constructed to investigate the role of SP1 in the heart. The echocardiography, histochemical experiment, and transmission electron microscope were performed to analyze the cardiac phenotypes of cardiac-specific conditional knockout of Sp1 mice. RNA sequencing, chromatin immunoprecipitation sequencing, and adeno-associated virus experiments in vivo were performed to explore the downstream molecules of SP1. To examine the therapeutic effect of SP1 on HCM, an SP1 overexpression vector was constructed and injected into the mutant allele of Myh6 R404Q/+ (Myh6 c. 1211C>T) HCM mice. The human induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs) from a patient with HCM were used to detect the potential therapeutic effects of SP1 in human HCM. RESULTS The cardiac-specific conditional knockout of Sp1 mice developed a typical HCM phenotype, displaying overt myocardial hypertrophy, interstitial fibrosis, and disordered myofilament. In addition, Sp1 knockdown dramatically increased the cell area of hiPSC-CMs and caused intracellular myofibrillar disorganization, which was similar to the hypertrophic cardiomyocytes of HCM. Mechanistically, Tuft1 was identified as the key target gene of SP1. The hypertrophic phenotypes induced by Sp1 knockdown in both hiPSC-CMs and mice could be rescued by TUFT1 (tuftelin 1) overexpression. Furthermore, SP1 overexpression suppressed the development of HCM in the mutant allele of Myh6 R404Q/+ mice and also reversed the hypertrophic phenotype of HCM hiPSC-CMs. CONCLUSIONS Our study demonstrates that SP1 deficiency leads to HCM. SP1 overexpression exhibits significant therapeutic effects on both HCM mice and HCM hiPSC-CMs, suggesting that SP1 could be a potential intervention target for HCM.
Collapse
Affiliation(s)
- Fulei Zhang
- State Key Laboratory of Cardiology (F.Z., H.Z., Y.Z., L.Z., J.L., G.F., H.L., Y. Wu, R.D., X.H., Yi Liu, L.L., J.Y., D.L., Y.-H.C.), Shanghai East Hospital, Tongji University School of Medicine, China
- Shanghai Arrhythmias Research Center (F.Z., H.Z., Y.Z., L.Z., J.L., G.F., Yuanyuan Liu, Y. Wang, H.L., Y. Wu, R.D., X.H., Yi Liu, L.L., J.Y., D.L., Y.-H.C.), Shanghai East Hospital, Tongji University School of Medicine, China
- Department of Cardiology (F.Z., H.Z., Y.Z., L.Z., J.L., G.F., H.L., Y. Wu, R.D., X.H., L.L., J.Y., D.L., Y.-H.C.), Shanghai East Hospital, Tongji University School of Medicine, China
| | - Huixing Zhou
- State Key Laboratory of Cardiology (F.Z., H.Z., Y.Z., L.Z., J.L., G.F., H.L., Y. Wu, R.D., X.H., Yi Liu, L.L., J.Y., D.L., Y.-H.C.), Shanghai East Hospital, Tongji University School of Medicine, China
- Shanghai Arrhythmias Research Center (F.Z., H.Z., Y.Z., L.Z., J.L., G.F., Yuanyuan Liu, Y. Wang, H.L., Y. Wu, R.D., X.H., Yi Liu, L.L., J.Y., D.L., Y.-H.C.), Shanghai East Hospital, Tongji University School of Medicine, China
- Department of Cardiology (F.Z., H.Z., Y.Z., L.Z., J.L., G.F., H.L., Y. Wu, R.D., X.H., L.L., J.Y., D.L., Y.-H.C.), Shanghai East Hospital, Tongji University School of Medicine, China
| | - Jinfeng Xue
- Department of Regenerative Medicine (J.X., L.Q.), Tongji University School of Medicine, Shanghai, China
| | - Yuemei Zhang
- State Key Laboratory of Cardiology (F.Z., H.Z., Y.Z., L.Z., J.L., G.F., H.L., Y. Wu, R.D., X.H., Yi Liu, L.L., J.Y., D.L., Y.-H.C.), Shanghai East Hospital, Tongji University School of Medicine, China
- Shanghai Arrhythmias Research Center (F.Z., H.Z., Y.Z., L.Z., J.L., G.F., Yuanyuan Liu, Y. Wang, H.L., Y. Wu, R.D., X.H., Yi Liu, L.L., J.Y., D.L., Y.-H.C.), Shanghai East Hospital, Tongji University School of Medicine, China
- Department of Cardiology (F.Z., H.Z., Y.Z., L.Z., J.L., G.F., H.L., Y. Wu, R.D., X.H., L.L., J.Y., D.L., Y.-H.C.), Shanghai East Hospital, Tongji University School of Medicine, China
| | - Liping Zhou
- State Key Laboratory of Cardiology (F.Z., H.Z., Y.Z., L.Z., J.L., G.F., H.L., Y. Wu, R.D., X.H., Yi Liu, L.L., J.Y., D.L., Y.-H.C.), Shanghai East Hospital, Tongji University School of Medicine, China
- Shanghai Arrhythmias Research Center (F.Z., H.Z., Y.Z., L.Z., J.L., G.F., Yuanyuan Liu, Y. Wang, H.L., Y. Wu, R.D., X.H., Yi Liu, L.L., J.Y., D.L., Y.-H.C.), Shanghai East Hospital, Tongji University School of Medicine, China
- Department of Cardiology (F.Z., H.Z., Y.Z., L.Z., J.L., G.F., H.L., Y. Wu, R.D., X.H., L.L., J.Y., D.L., Y.-H.C.), Shanghai East Hospital, Tongji University School of Medicine, China
| | - Junwei Leng
- State Key Laboratory of Cardiology (F.Z., H.Z., Y.Z., L.Z., J.L., G.F., H.L., Y. Wu, R.D., X.H., Yi Liu, L.L., J.Y., D.L., Y.-H.C.), Shanghai East Hospital, Tongji University School of Medicine, China
- Shanghai Arrhythmias Research Center (F.Z., H.Z., Y.Z., L.Z., J.L., G.F., Yuanyuan Liu, Y. Wang, H.L., Y. Wu, R.D., X.H., Yi Liu, L.L., J.Y., D.L., Y.-H.C.), Shanghai East Hospital, Tongji University School of Medicine, China
- Department of Cardiology (F.Z., H.Z., Y.Z., L.Z., J.L., G.F., H.L., Y. Wu, R.D., X.H., L.L., J.Y., D.L., Y.-H.C.), Shanghai East Hospital, Tongji University School of Medicine, China
| | - Guojian Fang
- State Key Laboratory of Cardiology (F.Z., H.Z., Y.Z., L.Z., J.L., G.F., H.L., Y. Wu, R.D., X.H., Yi Liu, L.L., J.Y., D.L., Y.-H.C.), Shanghai East Hospital, Tongji University School of Medicine, China
- Shanghai Arrhythmias Research Center (F.Z., H.Z., Y.Z., L.Z., J.L., G.F., Yuanyuan Liu, Y. Wang, H.L., Y. Wu, R.D., X.H., Yi Liu, L.L., J.Y., D.L., Y.-H.C.), Shanghai East Hospital, Tongji University School of Medicine, China
- Department of Cardiology (F.Z., H.Z., Y.Z., L.Z., J.L., G.F., H.L., Y. Wu, R.D., X.H., L.L., J.Y., D.L., Y.-H.C.), Shanghai East Hospital, Tongji University School of Medicine, China
| | - Yuanyuan Liu
- Shanghai Arrhythmias Research Center (F.Z., H.Z., Y.Z., L.Z., J.L., G.F., Yuanyuan Liu, Y. Wang, H.L., Y. Wu, R.D., X.H., Yi Liu, L.L., J.Y., D.L., Y.-H.C.), Shanghai East Hospital, Tongji University School of Medicine, China
- Jinzhou Medical University, China (Yuanyuan Liu, Y. Wang, Yan Wang)
| | - Yan Wang
- Shanghai Arrhythmias Research Center (F.Z., H.Z., Y.Z., L.Z., J.L., G.F., Yuanyuan Liu, Y. Wang, H.L., Y. Wu, R.D., X.H., Yi Liu, L.L., J.Y., D.L., Y.-H.C.), Shanghai East Hospital, Tongji University School of Medicine, China
- Jinzhou Medical University, China (Yuanyuan Liu, Y. Wang, Yan Wang)
| | - Hongyu Liu
- State Key Laboratory of Cardiology (F.Z., H.Z., Y.Z., L.Z., J.L., G.F., H.L., Y. Wu, R.D., X.H., Yi Liu, L.L., J.Y., D.L., Y.-H.C.), Shanghai East Hospital, Tongji University School of Medicine, China
- Shanghai Arrhythmias Research Center (F.Z., H.Z., Y.Z., L.Z., J.L., G.F., Yuanyuan Liu, Y. Wang, H.L., Y. Wu, R.D., X.H., Yi Liu, L.L., J.Y., D.L., Y.-H.C.), Shanghai East Hospital, Tongji University School of Medicine, China
- Department of Cardiology (F.Z., H.Z., Y.Z., L.Z., J.L., G.F., H.L., Y. Wu, R.D., X.H., L.L., J.Y., D.L., Y.-H.C.), Shanghai East Hospital, Tongji University School of Medicine, China
| | - Yahan Wu
- State Key Laboratory of Cardiology (F.Z., H.Z., Y.Z., L.Z., J.L., G.F., H.L., Y. Wu, R.D., X.H., Yi Liu, L.L., J.Y., D.L., Y.-H.C.), Shanghai East Hospital, Tongji University School of Medicine, China
- Shanghai Arrhythmias Research Center (F.Z., H.Z., Y.Z., L.Z., J.L., G.F., Yuanyuan Liu, Y. Wang, H.L., Y. Wu, R.D., X.H., Yi Liu, L.L., J.Y., D.L., Y.-H.C.), Shanghai East Hospital, Tongji University School of Medicine, China
- Department of Cardiology (F.Z., H.Z., Y.Z., L.Z., J.L., G.F., H.L., Y. Wu, R.D., X.H., L.L., J.Y., D.L., Y.-H.C.), Shanghai East Hospital, Tongji University School of Medicine, China
| | - Lingbin Qi
- Department of Regenerative Medicine (J.X., L.Q.), Tongji University School of Medicine, Shanghai, China
| | - Ran Duan
- State Key Laboratory of Cardiology (F.Z., H.Z., Y.Z., L.Z., J.L., G.F., H.L., Y. Wu, R.D., X.H., Yi Liu, L.L., J.Y., D.L., Y.-H.C.), Shanghai East Hospital, Tongji University School of Medicine, China
- Shanghai Arrhythmias Research Center (F.Z., H.Z., Y.Z., L.Z., J.L., G.F., Yuanyuan Liu, Y. Wang, H.L., Y. Wu, R.D., X.H., Yi Liu, L.L., J.Y., D.L., Y.-H.C.), Shanghai East Hospital, Tongji University School of Medicine, China
- Department of Cardiology (F.Z., H.Z., Y.Z., L.Z., J.L., G.F., H.L., Y. Wu, R.D., X.H., L.L., J.Y., D.L., Y.-H.C.), Shanghai East Hospital, Tongji University School of Medicine, China
| | - Xiaoyu He
- State Key Laboratory of Cardiology (F.Z., H.Z., Y.Z., L.Z., J.L., G.F., H.L., Y. Wu, R.D., X.H., Yi Liu, L.L., J.Y., D.L., Y.-H.C.), Shanghai East Hospital, Tongji University School of Medicine, China
- Shanghai Arrhythmias Research Center (F.Z., H.Z., Y.Z., L.Z., J.L., G.F., Yuanyuan Liu, Y. Wang, H.L., Y. Wu, R.D., X.H., Yi Liu, L.L., J.Y., D.L., Y.-H.C.), Shanghai East Hospital, Tongji University School of Medicine, China
- Department of Cardiology (F.Z., H.Z., Y.Z., L.Z., J.L., G.F., H.L., Y. Wu, R.D., X.H., L.L., J.Y., D.L., Y.-H.C.), Shanghai East Hospital, Tongji University School of Medicine, China
| | - Yan Wang
- Jinzhou Medical University, China (Yuanyuan Liu, Y. Wang, Yan Wang)
| | - Yi Liu
- State Key Laboratory of Cardiology (F.Z., H.Z., Y.Z., L.Z., J.L., G.F., H.L., Y. Wu, R.D., X.H., Yi Liu, L.L., J.Y., D.L., Y.-H.C.), Shanghai East Hospital, Tongji University School of Medicine, China
- Shanghai Arrhythmias Research Center (F.Z., H.Z., Y.Z., L.Z., J.L., G.F., Yuanyuan Liu, Y. Wang, H.L., Y. Wu, R.D., X.H., Yi Liu, L.L., J.Y., D.L., Y.-H.C.), Shanghai East Hospital, Tongji University School of Medicine, China
| | - Li Li
- State Key Laboratory of Cardiology (F.Z., H.Z., Y.Z., L.Z., J.L., G.F., H.L., Y. Wu, R.D., X.H., Yi Liu, L.L., J.Y., D.L., Y.-H.C.), Shanghai East Hospital, Tongji University School of Medicine, China
- Shanghai Arrhythmias Research Center (F.Z., H.Z., Y.Z., L.Z., J.L., G.F., Yuanyuan Liu, Y. Wang, H.L., Y. Wu, R.D., X.H., Yi Liu, L.L., J.Y., D.L., Y.-H.C.), Shanghai East Hospital, Tongji University School of Medicine, China
- Department of Cardiology (F.Z., H.Z., Y.Z., L.Z., J.L., G.F., H.L., Y. Wu, R.D., X.H., L.L., J.Y., D.L., Y.-H.C.), Shanghai East Hospital, Tongji University School of Medicine, China
- Department of Pathology and Pathophysiology (L.L., J.Y., Y.-H.C.), Tongji University School of Medicine, Shanghai, China
- Research Units of Origin and Regulation of Heart Rhythm, Chinese Academy of Medical Sciences, Shanghai, China (L.L., J.Y., D.L., Y.-H.C.)
| | - Jian Yang
- State Key Laboratory of Cardiology (F.Z., H.Z., Y.Z., L.Z., J.L., G.F., H.L., Y. Wu, R.D., X.H., Yi Liu, L.L., J.Y., D.L., Y.-H.C.), Shanghai East Hospital, Tongji University School of Medicine, China
- Shanghai Arrhythmias Research Center (F.Z., H.Z., Y.Z., L.Z., J.L., G.F., Yuanyuan Liu, Y. Wang, H.L., Y. Wu, R.D., X.H., Yi Liu, L.L., J.Y., D.L., Y.-H.C.), Shanghai East Hospital, Tongji University School of Medicine, China
- Department of Cardiology (F.Z., H.Z., Y.Z., L.Z., J.L., G.F., H.L., Y. Wu, R.D., X.H., L.L., J.Y., D.L., Y.-H.C.), Shanghai East Hospital, Tongji University School of Medicine, China
- Department of Pathology and Pathophysiology (L.L., J.Y., Y.-H.C.), Tongji University School of Medicine, Shanghai, China
- Research Units of Origin and Regulation of Heart Rhythm, Chinese Academy of Medical Sciences, Shanghai, China (L.L., J.Y., D.L., Y.-H.C.)
| | - Dandan Liang
- State Key Laboratory of Cardiology (F.Z., H.Z., Y.Z., L.Z., J.L., G.F., H.L., Y. Wu, R.D., X.H., Yi Liu, L.L., J.Y., D.L., Y.-H.C.), Shanghai East Hospital, Tongji University School of Medicine, China
- Shanghai Arrhythmias Research Center (F.Z., H.Z., Y.Z., L.Z., J.L., G.F., Yuanyuan Liu, Y. Wang, H.L., Y. Wu, R.D., X.H., Yi Liu, L.L., J.Y., D.L., Y.-H.C.), Shanghai East Hospital, Tongji University School of Medicine, China
- Department of Cardiology (F.Z., H.Z., Y.Z., L.Z., J.L., G.F., H.L., Y. Wu, R.D., X.H., L.L., J.Y., D.L., Y.-H.C.), Shanghai East Hospital, Tongji University School of Medicine, China
- Research Units of Origin and Regulation of Heart Rhythm, Chinese Academy of Medical Sciences, Shanghai, China (L.L., J.Y., D.L., Y.-H.C.)
| | - Yi-Han Chen
- State Key Laboratory of Cardiology (F.Z., H.Z., Y.Z., L.Z., J.L., G.F., H.L., Y. Wu, R.D., X.H., Yi Liu, L.L., J.Y., D.L., Y.-H.C.), Shanghai East Hospital, Tongji University School of Medicine, China
- Shanghai Arrhythmias Research Center (F.Z., H.Z., Y.Z., L.Z., J.L., G.F., Yuanyuan Liu, Y. Wang, H.L., Y. Wu, R.D., X.H., Yi Liu, L.L., J.Y., D.L., Y.-H.C.), Shanghai East Hospital, Tongji University School of Medicine, China
- Department of Cardiology (F.Z., H.Z., Y.Z., L.Z., J.L., G.F., H.L., Y. Wu, R.D., X.H., L.L., J.Y., D.L., Y.-H.C.), Shanghai East Hospital, Tongji University School of Medicine, China
- Department of Pathology and Pathophysiology (L.L., J.Y., Y.-H.C.), Tongji University School of Medicine, Shanghai, China
- Research Units of Origin and Regulation of Heart Rhythm, Chinese Academy of Medical Sciences, Shanghai, China (L.L., J.Y., D.L., Y.-H.C.)
| |
Collapse
|
39
|
Sakamoto T, Kelly DP. Cardiac maturation. J Mol Cell Cardiol 2024; 187:38-50. [PMID: 38160640 PMCID: PMC10923079 DOI: 10.1016/j.yjmcc.2023.12.008] [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: 10/31/2023] [Revised: 12/12/2023] [Accepted: 12/21/2023] [Indexed: 01/03/2024]
Abstract
The heart undergoes a dynamic maturation process following birth, in response to a wide range of stimuli, including both physiological and pathological cues. This process entails substantial re-programming of mitochondrial energy metabolism coincident with the emergence of specialized structural and contractile machinery to meet the demands of the adult heart. Many components of this program revert to a more "fetal" format during development of pathological cardiac hypertrophy and heart failure. In this review, emphasis is placed on recent progress in our understanding of the transcriptional control of cardiac maturation, encompassing the results of studies spanning from in vivo models to cardiomyocytes derived from human stem cells. The potential applications of this current state of knowledge to new translational avenues aimed at the treatment of heart failure is also addressed.
Collapse
Affiliation(s)
- Tomoya Sakamoto
- Cardiovascular Institute, Department of Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Daniel P Kelly
- Cardiovascular Institute, Department of Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA.
| |
Collapse
|
40
|
Vora N, Patel P, Gajjar A, Ladani P, Konat A, Bhanderi D, Gadam S, Prajjwal P, Sharma K, Arunachalam SP. Gene therapy for heart failure: A novel treatment for the age old disease. Dis Mon 2024; 70:101636. [PMID: 37734966 DOI: 10.1016/j.disamonth.2023.101636] [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] [Indexed: 09/23/2023]
Abstract
Across the globe, cardiovascular disease (CVD) is the leading cause of mortality. According to reports, around 6.2 million people in the United states have heart failure. Current standards of care for heart failure can delay but not prevent progression of disease. Gene therapy is one of the novel treatment modalities that promises to fill this limitation in the current standard of care for Heart Failure. In this paper we performed an extensive search of the literature on various advances made in gene therapy for heart failure till date. We review the delivery methods, targets, current applications, trials, limitations and feasibility of gene therapy for heart failure. Various methods have been employed till date for administering gene therapies including but not limited to arterial and venous infusion, direct myocardial injection and pericardial injection. Various strategies such as AC6 expression, S100A1 protein upregulation, VEGF-B and SDF-1 gene therapy have shown promise in recent preclinical trials. Furthermore, few studies even show that stimulation of cardiomyocyte proliferation such as through cyclin A2 overexpression is a realistic avenue. However, a considerable number of obstacles need to be overcome for gene therapy to be part of standard treatment of care such as definitive choice of gene, gene delivery systems and a suitable method for preclinical trials and clinical trials on patients. Considering the challenges and taking into account the recent advances in gene therapy research, there are encouraging signs to indicate gene therapy for heart failure to be a promising treatment modality for the future. However, the time and feasibility of this option remains in a situation of balance.
Collapse
Affiliation(s)
- Neel Vora
- B. J. Medical College, Ahmedabad, India
| | - Parth Patel
- Pramukhswami Medical College, Karamsad, India
| | | | | | - Ashwati Konat
- University School of Sciences, Gujarat University, Ahmedabad, India
| | | | | | | | - Kamal Sharma
- U. N. Mehta Institute of Cardiology and Research Centre, Ahmedabad, India.
| | | |
Collapse
|
41
|
Leng J, Wang C, Liang Z, Qiu F, Zhang S, Yang Y. An updated review of YAP: A promising therapeutic target against cardiac aging? Int J Biol Macromol 2024; 254:127670. [PMID: 37913886 DOI: 10.1016/j.ijbiomac.2023.127670] [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: 07/11/2023] [Revised: 09/05/2023] [Accepted: 10/23/2023] [Indexed: 11/03/2023]
Abstract
The transcriptional co-activator Yes-associated protein (YAP) functions as a downstream effector of the Hippo signaling pathway and plays a crucial role in cardiomyocyte survival. In its non-phosphorylated activated state, YAP binds to transcription factors, activating the transcription of downstream target genes. It also regulates cell proliferation and survival by selectively binding to enhancers and activating target genes. However, the upregulation of the Hippo pathway in human heart failure inhibits cardiac regeneration and disrupts astrogenesis, thus preventing the nuclear translocation of YAP. Existing literature indicates that the Hippo/YAP axis contributes to inflammation and fibrosis, potentially playing a role in the development of cardiac, vascular and renal injuries. Moreover, it is a key mediator of myofibroblast differentiation and fibrosis in the infarcted heart. Given these insights, can we harness YAP's regenerative potential in a targeted manner? In this review, we provide a detailed discussion of the Hippo signaling pathway and consolidate concepts for the development and intervention of cardiac anti-aging drugs to leverage YAP signaling as a pivotal target.
Collapse
Affiliation(s)
- Jingzhi Leng
- Cancer Institute, The Affiliated Hospital of Qingdao University, Qingdao, China; School of Physical Education, Qingdao University, China
| | - Chuanzhi Wang
- College of Sports Science, South China Normal University, Guangzhou, China
| | - Zhide Liang
- Cancer Institute, The Affiliated Hospital of Qingdao University, Qingdao, China; Qingdao Cancer Institute, Qingdao University, Qingdao, China
| | - Fanghui Qiu
- School of Physical Education, Qingdao University, China
| | - Shuangshuang Zhang
- Cancer Institute, The Affiliated Hospital of Qingdao University, Qingdao, China; Qingdao Cancer Institute, Qingdao University, Qingdao, China; School of Physical Education, Qingdao University, China.
| | - Yuan Yang
- Cancer Institute, The Affiliated Hospital of Qingdao University, Qingdao, China; Qingdao Cancer Institute, Qingdao University, Qingdao, China; School of Physical Education, Qingdao University, China.
| |
Collapse
|
42
|
Beisaw A, Wu CC. Cardiomyocyte maturation and its reversal during cardiac regeneration. Dev Dyn 2024; 253:8-27. [PMID: 36502296 DOI: 10.1002/dvdy.557] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2022] [Revised: 12/03/2022] [Accepted: 12/03/2022] [Indexed: 12/14/2022] Open
Abstract
Cardiovascular disease is a leading cause of death worldwide. Due to the limited proliferative and regenerative capacity of adult cardiomyocytes, the lost myocardium is not replenished efficiently and is replaced by a fibrotic scar, which eventually leads to heart failure. Current therapies to cure or delay the progression of heart failure are limited; hence, there is a pressing need for regenerative approaches to support the failing heart. Cardiomyocytes undergo a series of transcriptional, structural, and metabolic changes after birth (collectively termed maturation), which is critical for their contractile function but limits the regenerative capacity of the heart. In regenerative organisms, cardiomyocytes revert from their terminally differentiated state into a less mature state (ie, dedifferentiation) to allow for proliferation and regeneration to occur. Importantly, stimulating adult cardiomyocyte dedifferentiation has been shown to promote morphological and functional improvement after myocardial infarction, further highlighting the importance of cardiomyocyte dedifferentiation in heart regeneration. Here, we review several hallmarks of cardiomyocyte maturation, and summarize how their reversal facilitates cardiomyocyte proliferation and heart regeneration. A detailed understanding of how cardiomyocyte dedifferentiation is regulated will provide insights into therapeutic options to promote cardiomyocyte de-maturation and proliferation, and ultimately heart regeneration in mammals.
Collapse
Affiliation(s)
- Arica Beisaw
- Institute of Experimental Cardiology, Heidelberg University, Heidelberg, Germany
| | - Chi-Chung Wu
- European Center for Angioscience (ECAS), Medical Faculty Mannheim, Heidelberg University, Heidelberg, Germany
| |
Collapse
|
43
|
Huang H, Huang GN, Payumo AY. Two decades of heart regeneration research: Cardiomyocyte proliferation and beyond. WIREs Mech Dis 2024; 16:e1629. [PMID: 37700522 PMCID: PMC10840678 DOI: 10.1002/wsbm.1629] [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: 05/15/2023] [Revised: 08/03/2023] [Accepted: 08/09/2023] [Indexed: 09/14/2023]
Abstract
Interest in vertebrate cardiac regeneration has exploded over the past two decades since the discovery that adult zebrafish are capable of complete heart regeneration, contrasting the limited regenerative potential typically observed in adult mammalian hearts. Undercovering the mechanisms that both support and limit cardiac regeneration across the animal kingdom may provide unique insights in how we may unlock this capacity in adult humans. In this review, we discuss key discoveries in the heart regeneration field over the last 20 years. Initially, seminal findings revealed that pre-existing cardiomyocytes are the major source of regenerated cardiac muscle, drawing interest into the intrinsic mechanisms regulating cardiomyocyte proliferation. Moreover, recent studies have identified the importance of intercellular interactions and physiological adaptations, which highlight the vast complexity of the cardiac regenerative process. Finally, we compare strategies that have been tested to increase the regenerative capacity of the adult mammalian heart. This article is categorized under: Cardiovascular Diseases > Stem Cells and Development.
Collapse
Affiliation(s)
- Herman Huang
- Department of Biological Sciences, San Jose State University, San Jose, CA 95192, USA
| | - Guo N. Huang
- Cardiovascular Research Institute & Department of Physiology, University of California, San Francisco, San Francisco, CA, 94158, USA
- Eli and Edythe Broad Center for Regeneration Medicine and Stem Cell Research, University of California, San Francisco, San Francisco, CA, 94158, USA
| | - Alexander Y. Payumo
- Department of Biological Sciences, San Jose State University, San Jose, CA 95192, USA
| |
Collapse
|
44
|
Bai W, Zhu T, Zuo J, Li Y, Huang X, Li G. Delivery of SAV-siRNA via Exosomes from Adipose-Derived Stem Cells for the Treatment of Myocardial Infarction. Tissue Eng Regen Med 2023; 20:1063-1077. [PMID: 37801227 PMCID: PMC10645647 DOI: 10.1007/s13770-023-00588-z] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2023] [Revised: 07/24/2023] [Accepted: 08/07/2023] [Indexed: 10/07/2023] Open
Abstract
BACKGROUND Myocardial infarction (MI) leads to cardiomyocyte death, poor cardiac remodeling, and heart failure, making it a major cause of mortality and morbidity. To restore cardiac pumping function, induction of cardiomyocyte regeneration has become a focus of academic interest. The Hippo pathway is known to regulate cardiomyocyte proliferation and heart size, and its inactivation allows adult cardiomyocytes to re-enter the cell cycle. METHODS In this study, we investigated whether exosomes from adipose-derived stem cells (ADSCs) could effectively transfer siRNA for the Hippo pathway regulator Salvador (SAV) into cardiomyocytes to induce cardiomyocyte regeneration in a mouse model of MI. RESULTS Our results showed that exosomes loaded with SAV-siRNA effectively transferred siRNA into cardiomyocytes and induced cardiomyocyte re-entry into the cell cycle, while retaining the previously demonstrated therapeutic efficacy of ADSC-derived exosomes to improve post-infarction cardiac function through anti-fibrotic, pro-angiogenic, and other effects. CONCLUSIONS Our findings suggest that siRNA delivery via ADSC-derived exosomes may be a promising approach for the treatment of MI.
Collapse
Affiliation(s)
- Weizhe Bai
- Department of Cardiac Surgery, The Fifth Affiliated Hospital of Sun Yat-Sen University, No. 52, Meihua East Road, Zhuhai, Guangdong, People's Republic of China
| | - Tianchuan Zhu
- Center for Infection and Immunity, Guangdong Provincial Key Laboratory of Biomedical Imaging, The Fifth Affiliated Hospital of Sun Yat-Sen University, No. 52, Meihua East Road, Zhuhai, Guangdong, People's Republic of China
| | - Jiebin Zuo
- Department of Cardiac Surgery, The Fifth Affiliated Hospital of Sun Yat-Sen University, No. 52, Meihua East Road, Zhuhai, Guangdong, People's Republic of China
| | - Yang Li
- Department of Cardiac Surgery, The Fifth Affiliated Hospital of Sun Yat-Sen University, No. 52, Meihua East Road, Zhuhai, Guangdong, People's Republic of China
| | - Xi Huang
- Center for Infection and Immunity, Guangdong Provincial Key Laboratory of Biomedical Imaging, The Fifth Affiliated Hospital of Sun Yat-Sen University, No. 52, Meihua East Road, Zhuhai, Guangdong, People's Republic of China.
| | - Gang Li
- Department of Cardiac Surgery, The Fifth Affiliated Hospital of Sun Yat-Sen University, No. 52, Meihua East Road, Zhuhai, Guangdong, People's Republic of China.
| |
Collapse
|
45
|
Driskill JH, Pan D. Control of stem cell renewal and fate by YAP and TAZ. Nat Rev Mol Cell Biol 2023; 24:895-911. [PMID: 37626124 DOI: 10.1038/s41580-023-00644-5] [Citation(s) in RCA: 55] [Impact Index Per Article: 27.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/14/2023] [Indexed: 08/27/2023]
Abstract
Complex physiological processes control whether stem cells self-renew, differentiate or remain quiescent. Two decades of research have placed the Hippo pathway, a highly conserved kinase signalling cascade, and its downstream molecular effectors YAP and TAZ at the nexus of this decision. YAP and TAZ translate complex biological cues acting on stem cells - from mechanical forces to cellular metabolism - into genome-wide effects to mediate stem cell functions. While aberrant YAP/TAZ activity drives stem cell dysfunction in ageing, tumorigenesis and disease, therapeutic targeting of Hippo signalling and YAP/TAZ can boost stem cell activity to enhance regeneration. In this Review, we discuss how YAP/TAZ control the self-renewal, fate and plasticity of stem cells in different contexts, how dysregulation of YAP/TAZ in stem cells leads to disease, and how therapeutic modalities targeting YAP/TAZ may benefit regenerative medicine and cancer therapy.
Collapse
Affiliation(s)
- Jordan H Driskill
- Department of Physiology, Howard Hughes Medical Institute, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Duojia Pan
- Department of Physiology, Howard Hughes Medical Institute, University of Texas Southwestern Medical Center, Dallas, TX, USA.
| |
Collapse
|
46
|
Zlatanova I, Sun F, Wu RS, Chen X, Lau BH, Colombier P, Sinha T, Celona B, Xu SM, Materna SC, Huang GN, Black BL. An injury-responsive mmp14b enhancer is required for heart regeneration. SCIENCE ADVANCES 2023; 9:eadh5313. [PMID: 38019918 PMCID: PMC10686572 DOI: 10.1126/sciadv.adh5313] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/09/2023] [Accepted: 10/27/2023] [Indexed: 12/01/2023]
Abstract
Mammals have limited capacity for heart regeneration, whereas zebrafish have extraordinary regeneration abilities. During zebrafish heart regeneration, endothelial cells promote cardiomyocyte cell cycle reentry and myocardial repair, but the mechanisms responsible for promoting an injury microenvironment conducive to regeneration remain incompletely defined. Here, we identify the matrix metalloproteinase Mmp14b as an essential regulator of heart regeneration. We identify a TEAD-dependent mmp14b endothelial enhancer induced by heart injury in zebrafish and mice, and we show that the enhancer is required for regeneration, supporting a role for Hippo signaling upstream of mmp14b. Last, we show that MMP-14 function in mice is important for the accumulation of Agrin, an essential regulator of neonatal mouse heart regeneration. These findings reveal mechanisms for extracellular matrix remodeling that promote heart regeneration.
Collapse
Affiliation(s)
- Ivana Zlatanova
- Cardiovascular Research Institute, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Fei Sun
- Duke Regeneration Center, Department of Cell Biology, Duke University School of Medicine, Durham, NC 27710, USA
| | - Roland S. Wu
- Cardiovascular Research Institute, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Xiaoxin Chen
- Cardiovascular Research Institute, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Bryan H. Lau
- Cardiovascular Research Institute, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Pauline Colombier
- Cardiovascular Research Institute, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Tanvi Sinha
- Cardiovascular Research Institute, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Barbara Celona
- Cardiovascular Research Institute, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Shan-Mei Xu
- Cardiovascular Research Institute, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Stefan C. Materna
- Cardiovascular Research Institute, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Guo N. Huang
- Cardiovascular Research Institute, University of California, San Francisco, San Francisco, CA 94143, USA
- Department of Physiology, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Brian L. Black
- Cardiovascular Research Institute, University of California, San Francisco, San Francisco, CA 94143, USA
- Department of Biochemistry and Biophysics, University of California, San Francisco, San Francisco, CA 94143, USA
| |
Collapse
|
47
|
Liu C, Liu X, He Z, Zhang J, Tan X, Yang W, Zhang Y, Yu T, Liao S, Dai L, Xu Z, Li F, Huang Y, Zhao J. Proenkephalin-A secreted by renal proximal tubules functions as a brake in kidney regeneration. Nat Commun 2023; 14:7167. [PMID: 37935684 PMCID: PMC10630464 DOI: 10.1038/s41467-023-42929-5] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2022] [Accepted: 10/26/2023] [Indexed: 11/09/2023] Open
Abstract
Organ regeneration necessitates precise coordination of accelerators and brakes to restore organ function. However, the mechanisms underlying this intricate molecular crosstalk remain elusive. In this study, the level of proenkephalin-A (PENK-A), expressed by renal proximal tubular epithelial cells, decreases significantly with the loss of renal proximal tubules and increased at the termination phase of zebrafish kidney regeneration. Notably, this change contrasts with the role of hydrogen peroxide (H2O2), which acts as an accelerator in kidney regeneration. Through experiments with penka mutants and pharmaceutical treatments, we demonstrate that PENK-A inhibits H2O2 production in a dose-dependent manner, suggesting its involvement in regulating the rate and termination of regeneration. Furthermore, H2O2 influences the expression of tcf21, a vital factor in the formation of renal progenitor cell aggregates, by remodeling H3K4me3 in renal cells. Overall, our findings highlight the regulatory role of PENK-A as a brake in kidney regeneration.
Collapse
Affiliation(s)
- Chi Liu
- Department of Nephrology, the Key Laboratory for the Prevention and Treatment of Chronic Kidney Disease of Chongqing, Chongqing Clinical Research Center of Kidney and Urology Diseases, Xinqiao Hospital, Army Medical University (Third Military Medical University), 400037, Chongqing, P.R. China.
| | - Xiaoliang Liu
- Department of Nephrology, the Key Laboratory for the Prevention and Treatment of Chronic Kidney Disease of Chongqing, Chongqing Clinical Research Center of Kidney and Urology Diseases, Xinqiao Hospital, Army Medical University (Third Military Medical University), 400037, Chongqing, P.R. China
| | - Zhongwei He
- Department of Nephrology, the Key Laboratory for the Prevention and Treatment of Chronic Kidney Disease of Chongqing, Chongqing Clinical Research Center of Kidney and Urology Diseases, Xinqiao Hospital, Army Medical University (Third Military Medical University), 400037, Chongqing, P.R. China
| | - Jiangping Zhang
- Department of Nephrology, the Key Laboratory for the Prevention and Treatment of Chronic Kidney Disease of Chongqing, Chongqing Clinical Research Center of Kidney and Urology Diseases, Xinqiao Hospital, Army Medical University (Third Military Medical University), 400037, Chongqing, P.R. China
| | - Xiaoqin Tan
- Department of Nephrology, the Key Laboratory for the Prevention and Treatment of Chronic Kidney Disease of Chongqing, Chongqing Clinical Research Center of Kidney and Urology Diseases, Xinqiao Hospital, Army Medical University (Third Military Medical University), 400037, Chongqing, P.R. China
| | - Wenmin Yang
- Department of Nephrology, the Key Laboratory for the Prevention and Treatment of Chronic Kidney Disease of Chongqing, Chongqing Clinical Research Center of Kidney and Urology Diseases, Xinqiao Hospital, Army Medical University (Third Military Medical University), 400037, Chongqing, P.R. China
| | - Yunfeng Zhang
- Department of Nephrology, the Key Laboratory for the Prevention and Treatment of Chronic Kidney Disease of Chongqing, Chongqing Clinical Research Center of Kidney and Urology Diseases, Xinqiao Hospital, Army Medical University (Third Military Medical University), 400037, Chongqing, P.R. China
| | - Ting Yu
- Department of Respiratory Medicine, Xinqiao Hospital, Army Medical University (Third Military Medical University), 400037, Chongqing, P.R. China
| | - Shuyi Liao
- Department of Nephrology, the Key Laboratory for the Prevention and Treatment of Chronic Kidney Disease of Chongqing, Chongqing Clinical Research Center of Kidney and Urology Diseases, Xinqiao Hospital, Army Medical University (Third Military Medical University), 400037, Chongqing, P.R. China
| | - Lu Dai
- Department of Nephrology, the Key Laboratory for the Prevention and Treatment of Chronic Kidney Disease of Chongqing, Chongqing Clinical Research Center of Kidney and Urology Diseases, Xinqiao Hospital, Army Medical University (Third Military Medical University), 400037, Chongqing, P.R. China
| | - Zhi Xu
- Department of Respiratory Medicine, Xinqiao Hospital, Army Medical University (Third Military Medical University), 400037, Chongqing, P.R. China
| | - Furong Li
- Department of Nephrology, the Key Laboratory for the Prevention and Treatment of Chronic Kidney Disease of Chongqing, Chongqing Clinical Research Center of Kidney and Urology Diseases, Xinqiao Hospital, Army Medical University (Third Military Medical University), 400037, Chongqing, P.R. China
| | - Yinghui Huang
- Department of Nephrology, the Key Laboratory for the Prevention and Treatment of Chronic Kidney Disease of Chongqing, Chongqing Clinical Research Center of Kidney and Urology Diseases, Xinqiao Hospital, Army Medical University (Third Military Medical University), 400037, Chongqing, P.R. China.
| | - Jinghong Zhao
- Department of Nephrology, the Key Laboratory for the Prevention and Treatment of Chronic Kidney Disease of Chongqing, Chongqing Clinical Research Center of Kidney and Urology Diseases, Xinqiao Hospital, Army Medical University (Third Military Medical University), 400037, Chongqing, P.R. China.
| |
Collapse
|
48
|
Chen C, Wang J, Liu C, Hu J, Liu L. Pioneering therapies for post-infarction angiogenesis: Insight into molecular mechanisms and preclinical studies. Biomed Pharmacother 2023; 166:115306. [PMID: 37572633 DOI: 10.1016/j.biopha.2023.115306] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2023] [Revised: 08/01/2023] [Accepted: 08/07/2023] [Indexed: 08/14/2023] Open
Abstract
Acute myocardial infarction (MI), despite significant progress in its treatment, remains a leading cause of chronic heart failure and cardiovascular events such as cardiac arrest. Promoting angiogenesis in the myocardial tissue after MI to restore blood flow in the ischemic and hypoxic tissue is considered an effective treatment strategy. The repair of the myocardial tissue post-MI involves a robust angiogenic response, with mechanisms involved including endothelial cell proliferation and migration, capillary growth, changes in the extracellular matrix, and stabilization of pericytes for neovascularization. In this review, we provide a detailed overview of six key pathways in angiogenesis post-MI: the PI3K/Akt/mTOR signaling pathway, the Notch signaling pathway, the Wnt/β-catenin signaling pathway, the Hippo signaling pathway, the Sonic Hedgehog signaling pathway, and the JAK/STAT signaling pathway. We also discuss novel therapeutic approaches targeting these pathways, including drug therapy, gene therapy, protein therapy, cell therapy, and extracellular vesicle therapy. A comprehensive understanding of these key pathways and their targeted therapies will aid in our understanding of the pathological and physiological mechanisms of angiogenesis after MI and the development and application of new treatment strategies.
Collapse
Affiliation(s)
- Cong Chen
- Guang'anmen Hospital, China Academy of Chinese Medicine Sciences, Beijing 100053, China
| | - Jie Wang
- Guang'anmen Hospital, China Academy of Chinese Medicine Sciences, Beijing 100053, China.
| | - Chao Liu
- Guang'anmen Hospital, China Academy of Chinese Medicine Sciences, Beijing 100053, China
| | - Jun Hu
- Guang'anmen Hospital, China Academy of Chinese Medicine Sciences, Beijing 100053, China
| | - Lanchun Liu
- Guang'anmen Hospital, China Academy of Chinese Medicine Sciences, Beijing 100053, China
| |
Collapse
|
49
|
Burgon PG, Weldrick JJ, Talab OMSA, Nadeer M, Nomikos M, Megeney LA. Regulatory Mechanisms That Guide the Fetal to Postnatal Transition of Cardiomyocytes. Cells 2023; 12:2324. [PMID: 37759546 PMCID: PMC10528641 DOI: 10.3390/cells12182324] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2023] [Revised: 09/16/2023] [Accepted: 09/19/2023] [Indexed: 09/29/2023] Open
Abstract
Heart disease remains a global leading cause of death and disability, necessitating a comprehensive understanding of the heart's development, repair, and dysfunction. This review surveys recent discoveries that explore the developmental transition of proliferative fetal cardiomyocytes into hypertrophic postnatal cardiomyocytes, a process yet to be well-defined. This transition is key to the heart's growth and has promising therapeutic potential, particularly for congenital or acquired heart damage, such as myocardial infarctions. Although significant progress has been made, much work is needed to unravel the complex interplay of signaling pathways that regulate cardiomyocyte proliferation and hypertrophy. This review provides a detailed perspective for future research directions aimed at the potential therapeutic harnessing of the perinatal heart transitions.
Collapse
Affiliation(s)
- Patrick G. Burgon
- Department of Chemistry and Earth Sciences, College of Arts and Sciences, Qatar University, Doha P.O. Box 2713, Qatar
| | - Jonathan J. Weldrick
- Department of Medicine, Department of Cellular and Molecular Medicine, Faculty of Medicine, University of Ottawa, Ottawa, ON K1H 8M5, Canada; (J.J.W.); (L.A.M.)
| | | | - Muhammad Nadeer
- College of Medicine, QU Health, Qatar University, Doha P.O. Box 2713, Qatar; (O.M.S.A.T.)
| | - Michail Nomikos
- College of Medicine, QU Health, Qatar University, Doha P.O. Box 2713, Qatar; (O.M.S.A.T.)
| | - Lynn A. Megeney
- Department of Medicine, Department of Cellular and Molecular Medicine, Faculty of Medicine, University of Ottawa, Ottawa, ON K1H 8M5, Canada; (J.J.W.); (L.A.M.)
- Sprott Centre for Stem Cell Research, Ottawa Hospital Research Institute, Ottawa, ON K1H 8L6, Canada
| |
Collapse
|
50
|
DeFoor N, Paul S, Li S, Basso EKG, Stevenson V, Browning JL, Prater AK, Brindley S, Tao G, Pickrell AM. Remdesivir increases mtDNA copy number causing mild alterations to oxidative phosphorylation. Sci Rep 2023; 13:15339. [PMID: 37714940 PMCID: PMC10504289 DOI: 10.1038/s41598-023-42704-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: 05/29/2023] [Accepted: 09/13/2023] [Indexed: 09/17/2023] Open
Abstract
SARS-CoV-2 causes the severe respiratory disease COVID-19. Remdesivir (RDV) was the first fast-tracked FDA approved treatment drug for COVID-19. RDV acts as an antiviral ribonucleoside (adenosine) analogue that becomes active once it accumulates intracellularly. It then diffuses into the host cell and terminates viral RNA transcription. Previous studies have shown that certain nucleoside analogues unintentionally inhibit mitochondrial RNA or DNA polymerases or cause mutational changes to mitochondrial DNA (mtDNA). These past findings on the mitochondrial toxicity of ribonucleoside analogues motivated us to investigate what effects RDV may have on mitochondrial function. Using in vitro and in vivo rodent models treated with RDV, we observed increases in mtDNA copy number in Mv1Lu cells (35.26% increase ± 11.33%) and liver (100.27% increase ± 32.73%) upon treatment. However, these increases only resulted in mild changes to mitochondrial function. Surprisingly, skeletal muscle and heart were extremely resistant to RDV treatment, tissues that have preferentially been affected by other nucleoside analogues. Although our data suggest that RDV does not greatly impact mitochondrial function, these data are insightful for the treatment of RDV for individuals with mitochondrial disease.
Collapse
Affiliation(s)
- Nicole DeFoor
- School of Neuroscience, Virginia Tech, Life Science I Room 217, 970 Washington Street SW, Blacksburg, VA, 24061, USA
| | - Swagatika Paul
- Graduate Program in Biomedical and Veterinary Sciences, Virginia-Maryland College of Veterinary Medicine, Blacksburg, VA, 24061, USA
| | - Shuang Li
- Department of Regenerative Medicine and Cell Biology, Medical University of South Carolina, Charleston, SC, 29425, USA
| | - Erwin K Gudenschwager Basso
- Department of Biomedical Sciences and Pathobiology, Virginia-Maryland College of Veterinary Medicine, Blacksburg, VA, 24061, USA
| | - Valentina Stevenson
- Virginia Tech Animal Laboratory Services, Virginia-Maryland College of Veterinary Medicine, Blacksburg, VA, 24061, USA
| | - Jack L Browning
- School of Neuroscience, Virginia Tech, Life Science I Room 217, 970 Washington Street SW, Blacksburg, VA, 24061, USA
| | - Anna K Prater
- School of Neuroscience, Virginia Tech, Life Science I Room 217, 970 Washington Street SW, Blacksburg, VA, 24061, USA
| | - Samantha Brindley
- School of Neuroscience, Virginia Tech, Life Science I Room 217, 970 Washington Street SW, Blacksburg, VA, 24061, USA
| | - Ge Tao
- Department of Regenerative Medicine and Cell Biology, Medical University of South Carolina, Charleston, SC, 29425, USA
| | - Alicia M Pickrell
- School of Neuroscience, Virginia Tech, Life Science I Room 217, 970 Washington Street SW, Blacksburg, VA, 24061, USA.
| |
Collapse
|