151
|
Liu CF, Tang WW. Epigenetics in Cardiac Hypertrophy and Heart Failure. JACC Basic Transl Sci 2019; 4:976-993. [PMID: 31909304 PMCID: PMC6938823 DOI: 10.1016/j.jacbts.2019.05.011] [Citation(s) in RCA: 83] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/26/2019] [Revised: 05/29/2019] [Accepted: 05/30/2019] [Indexed: 12/14/2022]
Abstract
Heart failure (HF) is a complex syndrome affecting millions of people around the world. Over the past decade, the therapeutic potential of targeting epigenetic regulators in HF has been discussed extensively. Recent advances in next-generation sequencing techniques have contributed substantial progress in our understanding of the role of DNA methylation, post-translational modifications of histones, adenosine triphosphate (ATP)-dependent chromatin conformation and remodeling, and non-coding RNAs in HF pathophysiology. In this review, we summarize epigenomic studies on human and animal models in HF.
Collapse
Key Words
- BET, bromodomain
- EZH2, Enhancer of zeste homolog 2
- HAT, histone acetyltransferase
- HDAC, histone deacetylase
- HDM, histone demethylase
- HF, heart failure
- HMT, histone methyltransferase
- PRC2, polycomb repressor complex 2
- PTMs, post-translational modifications
- TAD, topologically associating domains
- TMAO, trimethylamine N-oxide
- cardiac hypertrophy
- epigenetics
- heart failure
- lnc-RNAs, long ncRNAs
Collapse
Affiliation(s)
- Chia-Feng Liu
- Department of Cardiovascular and Metabolic Sciences, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio
| | - W.H. Wilson Tang
- Department of Cardiovascular and Metabolic Sciences, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio
- Department of Cardiovascular Medicine, Heart and Vascular Institute, Cleveland Clinic, Cleveland, Ohio
| |
Collapse
|
152
|
Re-enforcing hypoxia-induced polyploid cardiomyocytes enter cytokinesis through activation of β-catenin. Sci Rep 2019; 9:17865. [PMID: 31780774 PMCID: PMC6883062 DOI: 10.1038/s41598-019-54334-4] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2019] [Accepted: 11/08/2019] [Indexed: 01/01/2023] Open
Abstract
Cardiomyocyte (CM) loss is a characteristic of various heart diseases, including ischaemic heart disease. Cardiac regeneration has been suggested as a promising strategy to address CM loss. Although many studies of regeneration have focused mainly on mononucleated or diploid CM, the limitations associated with the cytokinesis of polyploid and multinucleated CMs remain less well known. Here, we show that β-catenin, a key regulator in heart development, can increase cytokinesis in polyploid multinucleated CMs. The activation of β-catenin increases the expression of the cytokinesis-related factor epithelial cell transforming 2 (ECT2), which regulates the actomyosin ring and thus leads to the completion of cytokinesis in polyploid CMs. In addition, hypoxia can induce polyploid and multinucleated CMs by increasing factors related to the G1-S-anaphase of the cell cycle, but not those related to cytokinesis. Our study therefore reveals that the β-catenin can promote the cytokinesis of polyploid multinucleated CMs via upregulation of ECT2. These findings suggest a potential field of polyploid CM research that may be exploitable for cardiac regeneration therapy.
Collapse
|
153
|
Yifa O, Weisinger K, Bassat E, Li H, Kain D, Barr H, Kozer N, Genzelinakh A, Rajchman D, Eigler T, Umansky KB, Lendengolts D, Brener O, Bursac N, Tzahor E. The small molecule Chicago Sky Blue promotes heart repair following myocardial infarction in mice. JCI Insight 2019; 4:128025. [PMID: 31723055 DOI: 10.1172/jci.insight.128025] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2019] [Accepted: 10/03/2019] [Indexed: 12/11/2022] Open
Abstract
The adult mammalian heart regenerates poorly after injury and, as a result, ischemic heart diseases are among the leading causes of death worldwide. The recovery of the injured heart is dependent on orchestrated repair processes including inflammation, fibrosis, cardiomyocyte survival, proliferation, and contraction properties that could be modulated in patients. In this work we designed an automated high-throughput screening system for small molecules that induce cardiomyocyte proliferation in vitro and identified the small molecule Chicago Sky Blue 6B (CSB). Following induced myocardial infarction, CSB treatment reduced scar size and improved heart function of adult mice. Mechanistically, we show that although initially identified using in vitro screening for cardiomyocyte proliferation, in the adult mouse CSB promotes heart repair through (i) inhibition of CaMKII signaling, which improves cardiomyocyte contractility; and (ii) inhibition of neutrophil and macrophage activation, which attenuates the acute inflammatory response, thereby contributing to reduced scarring. In summary, we identified CSB as a potential therapeutic agent that enhances cardiac repair and function by suppressing postinjury detrimental processes, with no evidence for cardiomyocyte renewal.
Collapse
Affiliation(s)
- Oren Yifa
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot, Israel
| | - Karen Weisinger
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot, Israel
| | - Elad Bassat
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot, Israel
| | - Hanjun Li
- Department of Biomedical Engineering, Duke University, Durham, North Carolina, USA
| | - David Kain
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot, Israel
| | - Haim Barr
- HTS unit, The Nancy and Stephen Grand Israel National Center for Personalized Medicine (G-INCPM), and
| | - Noga Kozer
- HTS unit, The Nancy and Stephen Grand Israel National Center for Personalized Medicine (G-INCPM), and
| | - Alexander Genzelinakh
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot, Israel
| | - Dana Rajchman
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot, Israel
| | - Tamar Eigler
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot, Israel
| | - Kfir Baruch Umansky
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot, Israel
| | - Daria Lendengolts
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot, Israel
| | - Ori Brener
- Department of Veterinary Resources, Weizmann Institute of Science, Rehovot, Israel
| | - Nenad Bursac
- Department of Biomedical Engineering, Duke University, Durham, North Carolina, USA
| | - Eldad Tzahor
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot, Israel
| |
Collapse
|
154
|
Talman V, Teppo J, Pöhö P, Movahedi P, Vaikkinen A, Karhu ST, Trošt K, Suvitaival T, Heikkonen J, Pahikkala T, Kotiaho T, Kostiainen R, Varjosalo M, Ruskoaho H. Molecular Atlas of Postnatal Mouse Heart Development. J Am Heart Assoc 2019; 7:e010378. [PMID: 30371266 PMCID: PMC6474944 DOI: 10.1161/jaha.118.010378] [Citation(s) in RCA: 70] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Background The molecular mechanisms mediating postnatal loss of cardiac regeneration in mammals are not fully understood. We aimed to provide an integrated resource of mRNA, protein, and metabolite changes in the neonatal heart for identification of metabolism‐related mechanisms associated with cardiac regeneration. Methods and Results Mouse ventricular tissue samples taken on postnatal day 1 (P01), P04, P09, and P23 were analyzed with RNA sequencing and global proteomics and metabolomics. Gene ontology analysis, KEGG pathway analysis, and fuzzy c‐means clustering were used to identify up‐ or downregulated biological processes and metabolic pathways on all 3 levels, and Ingenuity pathway analysis (Qiagen) was used to identify upstream regulators. Differential expression was observed for 8547 mRNAs and for 1199 of 2285 quantified proteins. Furthermore, 151 metabolites with significant changes were identified. Differentially regulated metabolic pathways include branched chain amino acid degradation (upregulated at P23), fatty acid metabolism (upregulated at P04 and P09; downregulated at P23) as well as the HMGCS (HMG‐CoA [hydroxymethylglutaryl‐coenzyme A] synthase)–mediated mevalonate pathway and ketogenesis (transiently activated). Pharmacological inhibition of HMGCS in primary neonatal cardiomyocytes reduced the percentage of BrdU‐positive cardiomyocytes, providing evidence that the mevalonate and ketogenesis routes may participate in regulating the cardiomyocyte cell cycle. Conclusions This study is the first systems‐level resource combining data from genomewide transcriptomics with global quantitative proteomics and untargeted metabolomics analyses in the mouse heart throughout the early postnatal period. These integrated data of molecular changes associated with the loss of cardiac regeneration may open up new possibilities for the development of regenerative therapies.
Collapse
Affiliation(s)
- Virpi Talman
- 1 Drug Research Program and Division of Pharmacology and Pharmacotherapy Faculty of Pharmacy University of Helsinki Finland
| | - Jaakko Teppo
- 2 Drug Research Program and Division of Pharmaceutical Chemistry and Technology Faculty of Pharmacy University of Helsinki Finland.,3 Institute of Biotechnology and HiLIFE Helsinki Institute of Life Science University of Helsinki Finland
| | - Päivi Pöhö
- 2 Drug Research Program and Division of Pharmaceutical Chemistry and Technology Faculty of Pharmacy University of Helsinki Finland
| | - Parisa Movahedi
- 4 Department of Future Technologies Faculty of Mathematics and Natural Sciences University of Turku Finland
| | - Anu Vaikkinen
- 2 Drug Research Program and Division of Pharmaceutical Chemistry and Technology Faculty of Pharmacy University of Helsinki Finland
| | - S Tuuli Karhu
- 1 Drug Research Program and Division of Pharmacology and Pharmacotherapy Faculty of Pharmacy University of Helsinki Finland
| | | | | | - Jukka Heikkonen
- 4 Department of Future Technologies Faculty of Mathematics and Natural Sciences University of Turku Finland
| | - Tapio Pahikkala
- 4 Department of Future Technologies Faculty of Mathematics and Natural Sciences University of Turku Finland
| | - Tapio Kotiaho
- 2 Drug Research Program and Division of Pharmaceutical Chemistry and Technology Faculty of Pharmacy University of Helsinki Finland.,6 Department of Chemistry Faculty of Science University of Helsinki Finland
| | - Risto Kostiainen
- 2 Drug Research Program and Division of Pharmaceutical Chemistry and Technology Faculty of Pharmacy University of Helsinki Finland
| | - Markku Varjosalo
- 3 Institute of Biotechnology and HiLIFE Helsinki Institute of Life Science University of Helsinki Finland
| | - Heikki Ruskoaho
- 1 Drug Research Program and Division of Pharmacology and Pharmacotherapy Faculty of Pharmacy University of Helsinki Finland
| |
Collapse
|
155
|
Vujic A, Natarajan N, Lee RT. Molecular mechanisms of heart regeneration. Semin Cell Dev Biol 2019; 100:20-28. [PMID: 31587963 DOI: 10.1016/j.semcdb.2019.09.003] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2019] [Revised: 08/20/2019] [Accepted: 09/11/2019] [Indexed: 12/27/2022]
Abstract
The adult mammalian heart is incapable of clinically relevant regeneration. The regenerative deficit in adult mammalian heart contrasts with the fetal and neonatal heart, which demonstrate substantial regenerative capacity after injury. This deficiency in adult mammals is attributable to the lack of resident stem cells after birth, combined with an inability of pre-existing cardiomyocytes to complete cytokinesis. Studies of neonatal heart regeneration in mammals suggest that latent regenerative potential can be re-activated. Dissecting the cellular and molecular mechanisms that promote cardiomyocyte proliferation is key to stimulating true regeneration in adult humans. Here, we review recent advances in our understanding of cardiomyocyte proliferation that suggest molecular approaches to heart regeneration.
Collapse
Affiliation(s)
- Ana Vujic
- Department of Stem Cell and Regenerative Biology and the Harvard Stem Cell Institute, Harvard University, Cambridge, MA, 02138, USA
| | - Niranjana Natarajan
- Department of Stem Cell and Regenerative Biology and the Harvard Stem Cell Institute, Harvard University, Cambridge, MA, 02138, USA
| | - Richard T Lee
- Department of Stem Cell and Regenerative Biology and the Harvard Stem Cell Institute, Harvard University, Cambridge, MA, 02138, USA; Cardiovascular Division, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, 02115, USA.
| |
Collapse
|
156
|
Lavine KJ, Pinto AR, Epelman S, Kopecky BJ, Clemente-Casares X, Godwin J, Rosenthal N, Kovacic JC. The Macrophage in Cardiac Homeostasis and Disease: JACC Macrophage in CVD Series (Part 4). J Am Coll Cardiol 2019; 72:2213-2230. [PMID: 30360829 DOI: 10.1016/j.jacc.2018.08.2149] [Citation(s) in RCA: 166] [Impact Index Per Article: 27.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/07/2018] [Revised: 07/13/2018] [Accepted: 08/03/2018] [Indexed: 12/24/2022]
Abstract
Macrophages are integral components of cardiac tissue and exert profound effects on the healthy and diseased heart. Paradigm shifting studies using advanced molecular techniques have revealed significant complexity within these macrophage populations that reside in the heart. In this final of a 4-part review series covering the macrophage in cardiovascular disease, the authors review the origins, dynamics, cell surface markers, and respective functions of each cardiac macrophage subset identified to date, including in the specific scenarios of myocarditis and after myocardial infarction. Looking ahead, a deeper understanding of the diverse and often dichotomous functions of cardiac macrophages will be essential for the development of targeted therapies to mitigate injury and orchestrate recovery of the diseased heart. Moreover, as macrophages are critical for cardiac healing, they are an emerging focus for therapeutic strategies aimed at minimizing cardiomyocyte death, ameliorating pathological cardiac remodeling, and for treating heart failure and after myocardial infarction.
Collapse
Affiliation(s)
- Kory J Lavine
- Division of Cardiovascular Medicine, Department of Medicine, Washington University School of Medicine, St. Louis, Missouri; Center for Cardiovascular Research, Cardiovascular Division, Department of Medicine, Washington University School of Medicine, St. Louis, Missouri; Department of Developmental Biology, Washington University School of Medicine, St. Louis, Missouri; Department of Immunology and Pathology, Washington University School of Medicine, St. Louis, Missouri
| | - Alexander R Pinto
- Baker Heart and Diabetes Research Institute, Melbourne, Australia; Centre for Cardiovascular Biology and Disease Research, La Trobe University, Melbourne, Australia
| | - Slava Epelman
- Toronto General Hospital Research Institute, University Health Network, Toronto, Ontario, Canada; Ted Rogers Centre for Heart Research, Toronto, Ontario, Canada; University of Toronto, Department of Laboratory Medicine and Pathobiology, Toronto, Ontario, Canada; Department of Immunology, University of Toronto, Toronto, Ontario, Canada; Peter Munk Cardiac Centre, Toronto, Ontario, Canada
| | - Benjamin J Kopecky
- Division of Cardiovascular Medicine, Department of Medicine, Washington University School of Medicine, St. Louis, Missouri; Center for Cardiovascular Research, Cardiovascular Division, Department of Medicine, Washington University School of Medicine, St. Louis, Missouri
| | - Xavier Clemente-Casares
- Toronto General Hospital Research Institute, University Health Network, Toronto, Ontario, Canada; Department of Immunology, University of Toronto, Toronto, Ontario, Canada
| | - James Godwin
- The Jackson Laboratory, Bar Harbor, Maine; Mt. Desert Island Biological Laboratory, Bar Harbor, Maine
| | - Nadia Rosenthal
- The Jackson Laboratory, Bar Harbor, Maine; National Heart and Lung Institute, Imperial College London, London, United Kingdom
| | - Jason C Kovacic
- Zena and Michael A. Wiener Cardiovascular Institute, Icahn School of Medicine at Mount Sinai, New York, New York.
| |
Collapse
|
157
|
Forte E, Furtado MB, Rosenthal N. The interstitium in cardiac repair: role of the immune-stromal cell interplay. Nat Rev Cardiol 2019; 15:601-616. [PMID: 30181596 DOI: 10.1038/s41569-018-0077-x] [Citation(s) in RCA: 84] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Cardiac regeneration, that is, restoration of the original structure and function in a damaged heart, differs from tissue repair, in which collagen deposition and scar formation often lead to functional impairment. In both scenarios, the early-onset inflammatory response is essential to clear damaged cardiac cells and initiate organ repair, but the quality and extent of the immune response vary. Immune cells embedded in the damaged heart tissue sense and modulate inflammation through a dynamic interplay with stromal cells in the cardiac interstitium, which either leads to recapitulation of cardiac morphology by rebuilding functional scaffolds to support muscle regrowth in regenerative organisms or fails to resolve the inflammatory response and produces fibrotic scar tissue in adult mammals. Current investigation into the mechanistic basis of homeostasis and restoration of cardiac function has increasingly shifted focus away from stem cell-mediated cardiac repair towards a dynamic interplay of cells composing the less-studied interstitial compartment of the heart, offering unexpected insights into the immunoregulatory functions of cardiac interstitial components and the complex network of cell interactions that must be considered for clinical intervention in heart diseases.
Collapse
Affiliation(s)
| | | | - Nadia Rosenthal
- The Jackson Laboratory, Bar Harbor, ME, USA. .,National Heart and Lung Institute, Imperial College London, Faculty of Medicine, Imperial Centre for Translational and Experimental Medicine, London, UK.
| |
Collapse
|
158
|
Echeagaray O, Sussman MA. Transcribing the heart: faithfully interpreting cardiac transcriptional insights. Regen Med 2019; 14:805-810. [PMID: 31464566 PMCID: PMC6770408 DOI: 10.2217/rme-2019-0063] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2019] [Accepted: 08/07/2019] [Indexed: 12/12/2022] Open
Abstract
Transcriptional profiling continues to produce phenotypical data essential for understanding of basic cardiac biology and required to improve efficiency of cardiac regenerative and therapeutic approaches after injury. Accurate interpretation of cardiac transcriptional data comes with the unique challenges of heart biology including cardiomyocyte morphology, cryopreservation of limited samples and adequate selection of transcriptional platform at a single-cell resolution. Consequently, development and implementation of novel transcriptional platforms and creative bioinformatic analysis are essential to resolve standing questions in the field of cardiac regenerative medicine. Targeted bioinformatic approaches, advancing technological access, increase technical availability and fostering communication between interdisciplinary groups is critical to improve therapeutic approaches and to overcome challenges inherent to transcriptomic cardiac research.
Collapse
Affiliation(s)
- Oscar Echeagaray
- San Diego Heart Research Institute and Integrated Regenerative Research Institute, San Diego State University, San Diego, CA 92182-4650, USA
| | - Mark A Sussman
- San Diego Heart Research Institute and Integrated Regenerative Research Institute, San Diego State University, San Diego, CA 92182-4650, USA
| |
Collapse
|
159
|
Abstract
Cardiac ageing manifests as a decline in function leading to heart failure. At the cellular level, ageing entails decreased replicative capacity and dysregulation of cellular processes in myocardial and nonmyocyte cells. Various extrinsic parameters, such as lifestyle and environment, integrate important signalling pathways, such as those involving inflammation and oxidative stress, with intrinsic molecular mechanisms underlying resistance versus progression to cellular senescence. Mitigation of cardiac functional decline in an ageing organism requires the activation of enhanced maintenance and reparative capacity, thereby overcoming inherent endogenous limitations to retaining a youthful phenotype. Deciphering the molecular mechanisms underlying dysregulation of cellular function and renewal reveals potential interventional targets to attenuate degenerative processes at the cellular and systemic levels to improve quality of life for our ageing population. In this Review, we discuss the roles of extrinsic and intrinsic factors in cardiac ageing. Animal models of cardiac ageing are summarized, followed by an overview of the current and possible future treatments to mitigate the deleterious effects of cardiac ageing.
Collapse
|
160
|
Mechanistic basis of neonatal heart regeneration revealed by transcriptome and histone modification profiling. Proc Natl Acad Sci U S A 2019; 116:18455-18465. [PMID: 31451669 DOI: 10.1073/pnas.1905824116] [Citation(s) in RCA: 89] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
The adult mammalian heart has limited capacity for regeneration following injury, whereas the neonatal heart can readily regenerate within a short period after birth. To uncover the molecular mechanisms underlying neonatal heart regeneration, we compared the transcriptomes and epigenomes of regenerative and nonregenerative mouse hearts over a 7-d time period following myocardial infarction injury. By integrating gene expression profiles with histone marks associated with active or repressed chromatin, we identified transcriptional programs underlying neonatal heart regeneration, and the blockade to regeneration in later life. Our results reveal a unique immune response in regenerative hearts and a retained embryonic cardiogenic gene program that is active during neonatal heart regeneration. Among the unique immune factors and embryonic genes associated with cardiac regeneration, we identified Ccl24, which encodes a cytokine, and Igf2bp3, which encodes an RNA-binding protein, as previously unrecognized regulators of cardiomyocyte proliferation. Our data provide insights into the molecular basis of neonatal heart regeneration and identify genes that can be modulated to promote heart regeneration.
Collapse
|
161
|
Vegfc/d-dependent regulation of the lymphatic vasculature during cardiac regeneration is influenced by injury context. NPJ Regen Med 2019; 4:18. [PMID: 31452940 PMCID: PMC6706389 DOI: 10.1038/s41536-019-0079-2] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2018] [Accepted: 07/30/2019] [Indexed: 12/18/2022] Open
Abstract
The lymphatic vasculature mediates essential physiological functions including fluid homeostasis, lipid and hormone transport, and immune cell trafficking. Recent studies have suggested that promoting lymphangiogenesis enhances cardiac repair following injury, but it is unknown whether lymphangiogenesis is required for cardiac regeneration. Here, we describe the anatomical distribution, regulation, and function of the cardiac lymphatic network in a highly regenerative zebrafish model system using transgenic reporter lines and loss-of-function approaches. We show that zebrafish lacking functional vegfc and vegfd signaling are devoid of a cardiac lymphatic network and display cardiac hypertrophy in the absence of injury, suggesting a role for these vessels in cardiac tissue homeostasis. Using two different cardiac injury models, we report a robust lymphangiogenic response following cryoinjury, but not following apical resection injury. Although the majority of mutants lacking functional vegfc and vegfd signaling were able to mount a full regenerative response even in the complete absence of a cardiac lymphatic vasculature, cardiac regeneration was severely impaired in a subset of mutants, which was associated with heightened pro-inflammatory cytokine signaling. These findings reveal a context-dependent requirement for the lymphatic vasculature during cardiac growth and regeneration.
Collapse
|
162
|
Iyer LM, Nagarajan S, Woelfer M, Schoger E, Khadjeh S, Zafiriou MP, Kari V, Herting J, Pang ST, Weber T, Rathjens FS, Fischer TH, Toischer K, Hasenfuss G, Noack C, Johnsen SA, Zelarayán LC. A context-specific cardiac β-catenin and GATA4 interaction influences TCF7L2 occupancy and remodels chromatin driving disease progression in the adult heart. Nucleic Acids Res 2019; 46:2850-2867. [PMID: 29394407 PMCID: PMC5887416 DOI: 10.1093/nar/gky049] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2017] [Accepted: 01/18/2018] [Indexed: 12/17/2022] Open
Abstract
Chromatin remodelling precedes transcriptional and structural changes in heart failure. A body of work suggests roles for the developmental Wnt signalling pathway in cardiac remodelling. Hitherto, there is no evidence supporting a direct role of Wnt nuclear components in regulating chromatin landscapes in this process. We show that transcriptionally active, nuclear, phosphorylated(p)Ser675-β-catenin and TCF7L2 are upregulated in diseased murine and human cardiac ventricles. We report that inducible cardiomyocytes (CM)-specific pSer675-β-catenin accumulation mimics the disease situation by triggering TCF7L2 expression. This enhances active chromatin, characterized by increased H3K27ac and TCF7L2 occupancies to cardiac developmental and remodelling genes in vivo. Accordingly, transcriptomic analysis of β-catenin stabilized hearts shows a strong recapitulation of cardiac developmental processes like cell cycling and cytoskeletal remodelling. Mechanistically, TCF7L2 co-occupies distal genomic regions with cardiac transcription factors NKX2–5 and GATA4 in stabilized-β-catenin hearts. Validation assays revealed a previously unrecognized function of GATA4 as a cardiac repressor of the TCF7L2/β-catenin complex in vivo, thereby defining a transcriptional switch controlling disease progression. Conversely, preventing β-catenin activation post-pressure-overload results in a downregulation of these novel TCF7L2-targets and rescues cardiac function. Thus, we present a novel role for TCF7L2/β-catenin in CMs-specific chromatin modulation, which could be exploited for manipulating the ubiquitous Wnt pathway.
Collapse
Affiliation(s)
- Lavanya M Iyer
- Institute of Pharmacology and Toxicology, University Medical Center Goettingen, Georg-August University, Goettingen 37075, Germany.,German Center for Cardiovascular Research (DZHK) partner site Goettingen, Goettingen 37075, Germany
| | - Sankari Nagarajan
- Clinic for General, Visceral and Pediatric Surgery, University Medical Center Goettingen, Georg-August University, Goettingen 37075, Germany.,Cancer Research UK (CRUK-CI), Cambridge CB2 0RE, UK
| | - Monique Woelfer
- Institute of Pharmacology and Toxicology, University Medical Center Goettingen, Georg-August University, Goettingen 37075, Germany.,German Center for Cardiovascular Research (DZHK) partner site Goettingen, Goettingen 37075, Germany
| | - Eric Schoger
- Institute of Pharmacology and Toxicology, University Medical Center Goettingen, Georg-August University, Goettingen 37075, Germany.,German Center for Cardiovascular Research (DZHK) partner site Goettingen, Goettingen 37075, Germany
| | - Sara Khadjeh
- German Center for Cardiovascular Research (DZHK) partner site Goettingen, Goettingen 37075, Germany.,Department of Cardiology and Pneumology, University Medical Center Goettingen, Georg-August University, Goettingen 37075, Germany
| | - Maria Patapia Zafiriou
- Institute of Pharmacology and Toxicology, University Medical Center Goettingen, Georg-August University, Goettingen 37075, Germany.,German Center for Cardiovascular Research (DZHK) partner site Goettingen, Goettingen 37075, Germany
| | - Vijayalakshmi Kari
- Clinic for General, Visceral and Pediatric Surgery, University Medical Center Goettingen, Georg-August University, Goettingen 37075, Germany
| | - Jonas Herting
- German Center for Cardiovascular Research (DZHK) partner site Goettingen, Goettingen 37075, Germany.,Department of Cardiology and Pneumology, University Medical Center Goettingen, Georg-August University, Goettingen 37075, Germany
| | - Sze Ting Pang
- Institute of Pharmacology and Toxicology, University Medical Center Goettingen, Georg-August University, Goettingen 37075, Germany.,German Center for Cardiovascular Research (DZHK) partner site Goettingen, Goettingen 37075, Germany
| | - Tobias Weber
- Institute of Pharmacology and Toxicology, University Medical Center Goettingen, Georg-August University, Goettingen 37075, Germany.,German Center for Cardiovascular Research (DZHK) partner site Goettingen, Goettingen 37075, Germany
| | - Franziska S Rathjens
- Institute of Pharmacology and Toxicology, University Medical Center Goettingen, Georg-August University, Goettingen 37075, Germany.,German Center for Cardiovascular Research (DZHK) partner site Goettingen, Goettingen 37075, Germany
| | - Thomas H Fischer
- German Center for Cardiovascular Research (DZHK) partner site Goettingen, Goettingen 37075, Germany.,Department of Cardiology and Pneumology, University Medical Center Goettingen, Georg-August University, Goettingen 37075, Germany
| | - Karl Toischer
- German Center for Cardiovascular Research (DZHK) partner site Goettingen, Goettingen 37075, Germany.,Department of Cardiology and Pneumology, University Medical Center Goettingen, Georg-August University, Goettingen 37075, Germany
| | - Gerd Hasenfuss
- German Center for Cardiovascular Research (DZHK) partner site Goettingen, Goettingen 37075, Germany.,Department of Cardiology and Pneumology, University Medical Center Goettingen, Georg-August University, Goettingen 37075, Germany
| | - Claudia Noack
- Institute of Pharmacology and Toxicology, University Medical Center Goettingen, Georg-August University, Goettingen 37075, Germany.,German Center for Cardiovascular Research (DZHK) partner site Goettingen, Goettingen 37075, Germany
| | - Steven A Johnsen
- Clinic for General, Visceral and Pediatric Surgery, University Medical Center Goettingen, Georg-August University, Goettingen 37075, Germany
| | - Laura C Zelarayán
- Institute of Pharmacology and Toxicology, University Medical Center Goettingen, Georg-August University, Goettingen 37075, Germany.,German Center for Cardiovascular Research (DZHK) partner site Goettingen, Goettingen 37075, Germany
| |
Collapse
|
163
|
Malek Mohammadi M, Abouissa A, Azizah I, Xie Y, Cordero J, Shirvani A, Gigina A, Engelhardt M, Trogisch FA, Geffers R, Dobreva G, Bauersachs J, Heineke J. Induction of cardiomyocyte proliferation and angiogenesis protects neonatal mice from pressure overload-associated maladaptation. JCI Insight 2019; 5:128336. [PMID: 31335322 DOI: 10.1172/jci.insight.128336] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
Cardiac pressure overload (for example due to aortic stenosis) induces irreversible myocardial dysfunction, cardiomyocyte hypertrophy and interstitial fibrosis in patients. In contrast to adult, neonatal mice can efficiently regenerate the heart after injury in the first week after birth. To decipher whether insufficient cardiac regeneration contributes to the progression of pressure overload dependent disease, we established a transverse aortic constriction protocol in neonatal mice (nTAC). nTAC in the non-regenerative stage (at postnatal day P7) induced cardiac dysfunction, myocardial fibrosis and cardiomyocyte hypertrophy. In contrast, nTAC in the regenerative stage (at P1) largely prevented these maladaptive responses and was in particular associated with enhanced myocardial angiogenesis and increased cardiomyocyte proliferation, which both supported adaptation during nTAC. A comparative transcriptomic analysis between hearts after regenerative versus non-regenerative nTAC suggested the transcription factor GATA4 as master regulator of the regenerative gene-program. Indeed, cardiomyocyte specific deletion of GATA4 converted the regenerative nTAC into a non-regenerative, maladaptive response. Our new nTAC model can be used to identify mediators of adaptation during pressure overload and to discover novel potential therapeutic strategies.
Collapse
Affiliation(s)
- Mona Malek Mohammadi
- Department of Cardiovascular Research, European Center for Angioscience, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany.,Department of Cardiology and Angiology, Hannover Medical School, Hannover, Germany.,German Center for Cardiovascular Research (DZHK) partner site Heidelberg/Mannheim, Heidelberg, Germany
| | - Aya Abouissa
- Department of Cardiovascular Research, European Center for Angioscience, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Isyatul Azizah
- Department of Cardiovascular Research, European Center for Angioscience, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Yinuo Xie
- Department of Cardiovascular Research, European Center for Angioscience, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Julio Cordero
- Department of Anatomy and Developmental Biology, Center for Biomedicine and Medical Technology Mannheim, European Center for Angioscience, and
| | - Amir Shirvani
- Department of Cardiology and Angiology, Hannover Medical School, Hannover, Germany
| | - Anna Gigina
- Department of Cardiology and Angiology, Hannover Medical School, Hannover, Germany
| | - Maren Engelhardt
- Institute for Neuroanatomy, Center for Biomedicine and Medical Technology Mannheim, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Felix A Trogisch
- Department of Cardiovascular Research, European Center for Angioscience, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany.,German Center for Cardiovascular Research (DZHK) partner site Heidelberg/Mannheim, Heidelberg, Germany
| | - Robert Geffers
- Department Genome Analytics, Helmholtz-Center for Infection Research GmbH, Braunschweig, Germany
| | - Gergana Dobreva
- German Center for Cardiovascular Research (DZHK) partner site Heidelberg/Mannheim, Heidelberg, Germany.,Department of Anatomy and Developmental Biology, Center for Biomedicine and Medical Technology Mannheim, European Center for Angioscience, and
| | - Johann Bauersachs
- Department of Cardiology and Angiology, Hannover Medical School, Hannover, Germany
| | - Joerg Heineke
- Department of Cardiovascular Research, European Center for Angioscience, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany.,Department of Cardiology and Angiology, Hannover Medical School, Hannover, Germany.,German Center for Cardiovascular Research (DZHK) partner site Heidelberg/Mannheim, Heidelberg, Germany
| |
Collapse
|
164
|
Placek K, Schultze JL, Aschenbrenner AC. Epigenetic reprogramming of immune cells in injury, repair, and resolution. J Clin Invest 2019; 129:2994-3005. [PMID: 31329166 DOI: 10.1172/jci124619] [Citation(s) in RCA: 62] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Immune cells are pivotal in the reaction to injury, whereupon, under ideal conditions, repair and resolution phases restore homeostasis following initial acute inflammation. Immune cell activation and reprogramming require transcriptional changes that can only be initiated if epigenetic alterations occur. Recently, accelerated deciphering of epigenetic mechanisms has extended knowledge of epigenetic regulation, including long-distance chromatin remodeling, DNA methylation, posttranslational histone modifications, and involvement of small and long noncoding RNAs. Epigenetic changes have been linked to aspects of immune cell development, activation, and differentiation. Furthermore, genome-wide epigenetic landscapes have been established for some immune cells, including tissue-resident macrophages, and blood-derived cells including T cells. The epigenetic mechanisms underlying developmental steps from hematopoietic stem cells to fully differentiated immune cells led to development of epigenetic technologies and insights into general rules of epigenetic regulation. Compared with more advanced research areas, epigenetic reprogramming of immune cells in injury remains in its infancy. While the early epigenetic mechanisms supporting activation of the immune response to injury have been studied, less is known about resolution and repair phases and cell type-specific changes. We review prominent recent findings concerning injury-mediated epigenetic reprogramming, particularly in stroke and myocardial infarction. Lastly, we illustrate how single-cell technologies will be crucial to understanding epigenetic reprogramming in the complex sequential processes following injury.
Collapse
Affiliation(s)
- Katarzyna Placek
- Immunology and Metabolism, LIMES Institute, University of Bonn, Bonn, Germany
| | - Joachim L Schultze
- Platform for Single Cell Genomics and Epigenomics at the German Center for Neurodegenerative Diseases and the University of Bonn, Bonn, Germany.,Genomics and Immunoregulation, LIMES Institute, University of Bonn, Bonn, Germany
| | - Anna C Aschenbrenner
- Genomics and Immunoregulation, LIMES Institute, University of Bonn, Bonn, Germany
| |
Collapse
|
165
|
van Duijvenboden K, de Bakker DEM, Man JCK, Janssen R, Günthel M, Hill MC, Hooijkaas IB, van der Made I, van der Kraak PH, Vink A, Creemers EE, Martin JF, Barnett P, Bakkers J, Christoffels VM. Conserved NPPB+ Border Zone Switches From MEF2- to AP-1-Driven Gene Program. Circulation 2019; 140:864-879. [PMID: 31259610 DOI: 10.1161/circulationaha.118.038944] [Citation(s) in RCA: 72] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
BACKGROUND Surviving cells in the postinfarction border zone are subjected to intense fluctuations of their microenvironment. Recently, border zone cardiomyocytes have been specifically implicated in cardiac regeneration. Here, we defined their unique transcriptional and regulatory properties, and comprehensively validated new molecular markers, including Nppb, encoding B-type natriuretic peptide, after infarction. METHODS Transgenic reporter mice were used to identify the Nppb-positive border zone after myocardial infarction. Transcriptome analysis of remote, border, and infarct zones and of purified cardiomyocyte nuclei was performed using RNA-sequencing. Top candidate genes displaying border zone spatial specificity were histologically validated in ischemic human hearts. Mice in which Nppb was deleted by genome editing were subjected to myocardial infarction. Chromatin accessibility landscapes of border zone and control cardiomyocyte nuclei were assessed by using assay for transposase-accessible chromatin using sequencing. RESULTS We identified the border zone as a spatially confined region transcriptionally distinct from the remote myocardium. The transcriptional response of the border zone was much stronger than that of the remote ventricular wall, involving acute downregulation of mitochondrial oxidative phosphorylation, fatty acid metabolism, calcium handling, and sarcomere function, and the activation of a stress-response program. Analysis of infarcted human hearts revealed that the transcriptionally discrete border zone is conserved in humans, and led to the identification of novel conserved border zone markers including NPPB, ANKRD1, DES, UCHL1, JUN, and FOXP1. Homozygous Nppb mutant mice developed acute and lethal heart failure after myocardial infarction, indicating that B-type natriuretic peptide is required to preserve postinfarct heart function. Assay for transposase-accessible chromatin using sequencing revealed thousands of cardiomyocyte lineage-specific MEF2-occupied regulatory elements that lost accessibility in the border zone. Putative injury-responsive enhancers that gained accessibility were highly associated with AP-1 (activator protein 1) binding sites. Nuclear c-Jun, a component of AP-1, was observed specifically in border zone cardiomyocytes. CONCLUSIONS Cardiomyocytes in a discrete zone bordering the infarct switch from a MEF2-driven homeostatic lineage-specific to an AP-1-driven injury-induced gene expression program. This program is conserved between mouse and human, and includes Nppb expression, which is required to prevent acute heart failure after infarction.
Collapse
Affiliation(s)
- Karel van Duijvenboden
- Departments of Medical Biology, Amsterdam Cardiovascular Sciences (K.v.D., J.C.K.M., R.J., M.G., I.B.H., P.B., V.M.C.), Academic Medical Center, Amsterdam, The Netherlands
| | - Dennis E M de Bakker
- Hubrecht Institute (D.E.M.d.B., J.B.), University Medical Centre Utrecht, The Netherlands
| | - Joyce C K Man
- Departments of Medical Biology, Amsterdam Cardiovascular Sciences (K.v.D., J.C.K.M., R.J., M.G., I.B.H., P.B., V.M.C.), Academic Medical Center, Amsterdam, The Netherlands
| | - Rob Janssen
- Departments of Medical Biology, Amsterdam Cardiovascular Sciences (K.v.D., J.C.K.M., R.J., M.G., I.B.H., P.B., V.M.C.), Academic Medical Center, Amsterdam, The Netherlands
| | - Marie Günthel
- Departments of Medical Biology, Amsterdam Cardiovascular Sciences (K.v.D., J.C.K.M., R.J., M.G., I.B.H., P.B., V.M.C.), Academic Medical Center, Amsterdam, The Netherlands
| | - Matthew C Hill
- Program in Developmental Biology (M.C.H., J.F.M.), Baylor College of Medicine, Houston, TX
| | - Ingeborg B Hooijkaas
- Departments of Medical Biology, Amsterdam Cardiovascular Sciences (K.v.D., J.C.K.M., R.J., M.G., I.B.H., P.B., V.M.C.), Academic Medical Center, Amsterdam, The Netherlands
| | - Ingeborg van der Made
- Experimental Cardiology (I.v.d.M., E.E.C.), Academic Medical Center, Amsterdam, The Netherlands
| | - Petra H van der Kraak
- Department of Pathology (P.H.v.d.K., A.V.), University Medical Centre Utrecht, The Netherlands
| | - Aryan Vink
- Department of Pathology (P.H.v.d.K., A.V.), University Medical Centre Utrecht, The Netherlands
| | - Esther E Creemers
- Experimental Cardiology (I.v.d.M., E.E.C.), Academic Medical Center, Amsterdam, The Netherlands
| | - James F Martin
- Program in Developmental Biology (M.C.H., J.F.M.), Baylor College of Medicine, Houston, TX.,Department of Molecular Physiology and Biophysics (J.F.M.), Baylor College of Medicine, Houston, TX
| | - Phil Barnett
- Departments of Medical Biology, Amsterdam Cardiovascular Sciences (K.v.D., J.C.K.M., R.J., M.G., I.B.H., P.B., V.M.C.), Academic Medical Center, Amsterdam, The Netherlands
| | - Jeroen Bakkers
- Hubrecht Institute (D.E.M.d.B., J.B.), University Medical Centre Utrecht, The Netherlands
| | - Vincent M Christoffels
- Departments of Medical Biology, Amsterdam Cardiovascular Sciences (K.v.D., J.C.K.M., R.J., M.G., I.B.H., P.B., V.M.C.), Academic Medical Center, Amsterdam, The Netherlands
| |
Collapse
|
166
|
Borden A, Kurian J, Nickoloff E, Yang Y, Troupes CD, Ibetti J, Lucchese AM, Gao E, Mohsin S, Koch WJ, Houser SR, Kishore R, Khan M. Transient Introduction of miR-294 in the Heart Promotes Cardiomyocyte Cell Cycle Reentry After Injury. Circ Res 2019; 125:14-25. [PMID: 30964391 PMCID: PMC6586499 DOI: 10.1161/circresaha.118.314223] [Citation(s) in RCA: 79] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/06/2018] [Accepted: 04/05/2019] [Indexed: 02/06/2023]
Abstract
RATIONALE Embryonic heart is characterized of rapidly dividing cardiomyocytes required to build a working myocardium. Cardiomyocytes retain some proliferative capacity in the neonates but lose it in adulthood. Consequently, a number of signaling hubs including microRNAs are altered during cardiac development that adversely impacts regenerative potential of cardiac tissue. Embryonic stem cell cycle miRs are a class of microRNAs exclusively expressed during developmental stages; however, their effect on cardiomyocyte proliferation and heart function in adult myocardium has not been studied previously. OBJECTIVE To determine whether transient reintroduction of embryonic stem cell cycle miR-294 promotes cardiomyocyte cell cycle reentry enhancing cardiac repair after myocardial injury. METHODS AND RESULTS miR-294 is expressed in the heart during development, prenatal stages, lost in the neonate, and adult heart confirmed by qRT-PCR and in situ hybridization. Neonatal ventricular myocytes treated with miR-294 showed elevated expression of Ki67, p-histone H3, and Aurora B confirmed by immunocytochemistry compared with control cells. miR-294 enhanced oxidative phosphorylation and glycolysis in Neonatal ventricular myocytes measured by seahorse assay. Mechanistically, miR-294 represses Wee1 leading to increased activity of the cyclin B1/CDK1 complex confirmed by qRT-PCR and immunoblot analysis. Next, a doxycycline-inducible AAV9-miR-294 vector was delivered to mice for activating miR-294 in myocytes for 14 days continuously after myocardial infarction. miR-294-treated mice significantly improved left ventricular functions together with decreased infarct size and apoptosis 8 weeks after MI. Myocyte cell cycle reentry increased in miR-294 hearts analyzed by Ki67, pH3, and AurB (Aurora B kinase) expression parallel to increased small myocyte number in the heart. Isolated adult myocytes from miR-294 hearts showed increased 5-ethynyl-2'-deoxyuridine+ cells and upregulation of cell cycle markers and miR-294 targets 8 weeks after MI. CONCLUSIONS Ectopic transient expression of miR-294 recapitulates developmental signaling and phenotype in cardiomyocytes promoting cell cycle reentry that leads to augmented cardiac function in mice after myocardial infarction.
Collapse
Affiliation(s)
- Austin Borden
- Center for Metabolic Disease Research (CMDR), Temple University
| | - Justin Kurian
- Center for Metabolic Disease Research (CMDR), Temple University
| | - Emily Nickoloff
- Center for Translational Medicine (CTM), LKSOM, LKSOM, Temple University
| | - Yijun Yang
- Cardiovascular Research Institute (CVRC), Temple University
| | | | - Jessica Ibetti
- Center for Translational Medicine (CTM), LKSOM, LKSOM, Temple University
| | | | - Erhe Gao
- Center for Translational Medicine (CTM), LKSOM, LKSOM, Temple University
| | - Sadia Mohsin
- Cardiovascular Research Institute (CVRC), Temple University
- Department of Pharmacology, LKSOM, Temple University, LKSOM, Temple University
| | - Walter J Koch
- Center for Translational Medicine (CTM), LKSOM, LKSOM, Temple University
- Department of Pharmacology, LKSOM, Temple University, LKSOM, Temple University
| | - Steven R Houser
- Cardiovascular Research Institute (CVRC), Temple University
- Department of Physiology, LKSOM, Temple University
| | - Raj Kishore
- Center for Translational Medicine (CTM), LKSOM, LKSOM, Temple University
- Department of Pharmacology, LKSOM, Temple University, LKSOM, Temple University
| | - Mohsin Khan
- Center for Metabolic Disease Research (CMDR), Temple University
- Department of Physiology, LKSOM, Temple University
| |
Collapse
|
167
|
Affiliation(s)
- Mark A Sussman
- Department of Biology & Integrated Regenerative Research Institute, San Diego State University, San Diego, CA 92182, USA
| |
Collapse
|
168
|
Bagchi RA, Weeks KL. Histone deacetylases in cardiovascular and metabolic diseases. J Mol Cell Cardiol 2019; 130:151-159. [PMID: 30978343 DOI: 10.1016/j.yjmcc.2019.04.003] [Citation(s) in RCA: 74] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/30/2018] [Revised: 03/29/2019] [Accepted: 04/06/2019] [Indexed: 12/13/2022]
Abstract
Histone deacetylases (HDACs) regulate gene transcription by catalyzing the removal of acetyl groups from key lysine residues in nucleosomal histones and via the recruitment of other epigenetic regulators to DNA promoter/enhancer regions. Over the past two decades, HDACs have been implicated in multiple processes pertinent to cardiovascular and metabolic diseases, including cardiac hypertrophy and remodeling, fibrosis, calcium handling, inflammation and energy metabolism. The development of small molecule HDAC inhibitors and genetically modified loss- and gain-of-function mouse models has allowed interrogation of the roles of specific HDAC isoforms in these processes. Isoform-selective HDAC inhibitors may prove to be powerful therapeutic agents for the treatment of cardiovascular diseases, obesity and diabetes.
Collapse
Affiliation(s)
- Rushita A Bagchi
- Department of Medicine, Division of Cardiology, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, United States of America
| | - Kate L Weeks
- Baker Heart and Diabetes Institute, Melbourne, VIC 3004, Australia; Department of Diabetes, Central Clinical School, Monash University, Clayton, VIC 3800, Australia.
| |
Collapse
|
169
|
Psarras S, Beis D, Nikouli S, Tsikitis M, Capetanaki Y. Three in a Box: Understanding Cardiomyocyte, Fibroblast, and Innate Immune Cell Interactions to Orchestrate Cardiac Repair Processes. Front Cardiovasc Med 2019; 6:32. [PMID: 31001541 PMCID: PMC6454035 DOI: 10.3389/fcvm.2019.00032] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2018] [Accepted: 03/11/2019] [Indexed: 12/11/2022] Open
Abstract
Following an insult by both intrinsic and extrinsic pathways, complex cellular, and molecular interactions determine a successful recovery or inadequate repair of damaged tissue. The efficiency of this process is particularly important in the heart, an organ characterized by very limited regenerative and repair capacity in higher adult vertebrates. Cardiac insult is characteristically associated with fibrosis and heart failure, as a result of cardiomyocyte death, myocardial degeneration, and adverse remodeling. Recent evidence implies that resident non-cardiomyocytes, fibroblasts but also macrophages -pillars of the innate immunity- form part of the inflammatory response and decisively affect the repair process following a cardiac insult. Multiple studies in model organisms (mouse, zebrafish) of various developmental stages (adult and neonatal) combined with genetically engineered cell plasticity and differentiation intervention protocols -mainly targeting cardiac fibroblasts or progenitor cells-reveal particular roles of resident and recruited innate immune cells and their secretome in the coordination of cardiac repair. The interplay of innate immune cells with cardiac fibroblasts and cardiomyocytes is emerging as a crucial platform to help our understanding and, importantly, to allow the development of effective interventions sufficient to minimize cardiac damage and dysfunction after injury.
Collapse
Affiliation(s)
- Stelios Psarras
- Center of Basic Research, Biomedical Research Foundation of the Academy of Athens, Athens, Greece
| | - Dimitris Beis
- Center of Clinical, Experimental Surgery & Translational Research, Biomedical Research Foundation of the Academy of Athens, Athens, Greece
| | - Sofia Nikouli
- Center of Basic Research, Biomedical Research Foundation of the Academy of Athens, Athens, Greece
| | - Mary Tsikitis
- Center of Basic Research, Biomedical Research Foundation of the Academy of Athens, Athens, Greece
| | - Yassemi Capetanaki
- Center of Basic Research, Biomedical Research Foundation of the Academy of Athens, Athens, Greece
| |
Collapse
|
170
|
Lai SL, Marín-Juez R, Stainier DYR. Immune responses in cardiac repair and regeneration: a comparative point of view. Cell Mol Life Sci 2019; 76:1365-1380. [PMID: 30578442 PMCID: PMC6420886 DOI: 10.1007/s00018-018-2995-5] [Citation(s) in RCA: 68] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2018] [Revised: 11/26/2018] [Accepted: 12/10/2018] [Indexed: 12/13/2022]
Abstract
Immediately after cardiac injury, the immune system plays major roles in repair and regeneration as it becomes involved in a number of processes including damage-associated signaling, inflammation, revascularization, cardiomyocyte dedifferentiation and replenishment, and fibrotic scar formation/resolution. Recent studies have revealed that different immune responses occur in the various experimental models capable or incapable of cardiac regeneration, and that harnessing these immune responses might improve cardiac repair. In light of this concept, this review analyzes current knowledge about the immune responses to cardiac injury from a comparative perspective. Insights gained from such comparative analyses may provide ways to modulate the immune response as a potential therapeutic strategy for cardiac disease.
Collapse
Affiliation(s)
- Shih-Lei Lai
- Department of Developmental Genetics, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany.
- Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan.
| | - Rubén Marín-Juez
- Department of Developmental Genetics, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany
| | - Didier Y R Stainier
- Department of Developmental Genetics, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany.
| |
Collapse
|
171
|
Drug Screening in Human PSC-Cardiac Organoids Identifies Pro-proliferative Compounds Acting via the Mevalonate Pathway. Cell Stem Cell 2019; 24:895-907.e6. [PMID: 30930147 DOI: 10.1016/j.stem.2019.03.009] [Citation(s) in RCA: 197] [Impact Index Per Article: 32.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2018] [Revised: 12/05/2018] [Accepted: 03/07/2019] [Indexed: 12/28/2022]
Abstract
We have previously developed a high-throughput bioengineered human cardiac organoid (hCO) platform, which provides functional contractile tissue with biological properties similar to native heart tissue, including mature, cell-cycle-arrested cardiomyocytes. In this study, we perform functional screening of 105 small molecules with pro-regenerative potential. Our findings reveal surprising discordance between our hCO system and traditional 2D assays. In addition, functional analyses uncovered detrimental effects of many hit compounds. Two pro-proliferative small molecules without detrimental impacts on cardiac function were identified. High-throughput proteomics in hCO revealed synergistic activation of the mevalonate pathway and a cell-cycle network by the pro-proliferative compounds. Cell-cycle reentry in hCO and in vivo required the mevalonate pathway as inhibition of the mevalonate pathway with a statin attenuated pro-proliferative effects. This study highlights the utility of human cardiac organoids for pro-regenerative drug development, including identification of underlying biological mechanisms and minimization of adverse side effects.
Collapse
|
172
|
Farbehi N, Patrick R, Dorison A, Xaymardan M, Janbandhu V, Wystub-Lis K, Ho JW, Nordon RE, Harvey RP. Single-cell expression profiling reveals dynamic flux of cardiac stromal, vascular and immune cells in health and injury. eLife 2019; 8:43882. [PMID: 30912746 PMCID: PMC6459677 DOI: 10.7554/elife.43882] [Citation(s) in RCA: 358] [Impact Index Per Article: 59.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2018] [Accepted: 03/25/2019] [Indexed: 12/11/2022] Open
Abstract
Besides cardiomyocytes (CM), the heart contains numerous interstitial cell types which play key roles in heart repair, regeneration and disease, including fibroblast, vascular and immune cells. However, a comprehensive understanding of this interactive cell community is lacking. We performed single-cell RNA-sequencing of the total non-CM fraction and enriched (Pdgfra-GFP+) fibroblast lineage cells from murine hearts at days 3 and 7 post-sham or myocardial infarction (MI) surgery. Clustering of >30,000 single cells identified >30 populations representing nine cell lineages, including a previously undescribed fibroblast lineage trajectory present in both sham and MI hearts leading to a uniquely activated cell state defined in part by a strong anti-WNT transcriptome signature. We also uncovered novel myofibroblast subtypes expressing either pro-fibrotic or anti-fibrotic signatures. Our data highlight non-linear dynamics in myeloid and fibroblast lineages after cardiac injury, and provide an entry point for deeper analysis of cardiac homeostasis, inflammation, fibrosis, repair and regeneration. In our bodies, heart attacks lead to cell death and inflammation. This is then followed by a healing phase where the organ repairs itself. There are many types of heart cells, from muscle and pacemaker cells that help to create the beating motion, to so-called fibroblasts that act as a supporting network. Yet, it is still unclear how individual cells participate in the heart's response to injury. All cells possess the same genetic information, but they turn on or off different genes depending on the specific tasks that they need to perform. Spotting which genes are activated in individual cells can therefore provide clues about their exact roles in the body. Until recently, technological limitations meant that this information was difficult to access, because it was only possible to capture the global response of a group of cells in a sample. A new method called single-cell RNA sequencing is now allowing researchers to study the activities of many genes in thousands of individual cells at the same time. Here, Farbehi, Patrick et al. performed single-cell RNA sequencing on over 30,000 individual cells from healthy and injured mouse hearts. Computational approaches were then used to cluster cells into groups according to the activities of their genes. The experiments identified over 30 distinct sub-types of cell, including several that were previously unknown. For example, a group of fibroblasts that express a gene called Wif1 was discovered. Previous genetic studies have shown that Wif1 is essential for the heart's response to injury. Further experiments by Farbehi, Patrick et al. indicated that this new sub-type of cells may control the timing of the different aspects of heart repair after damage. Tens of millions of people around the world suffer from heart attacks and other heart diseases. Knowing how different types of heart cells participate in repair mechanisms may help to find new targets for drugs and other treatments.
Collapse
Affiliation(s)
- Nona Farbehi
- Victor Chang Cardiac Research Institute, Darlinghurst, Australia.,Stem Cells Australia, Melbourne Brain Centre, University of Melbourne, Victoria, Australia.,Garvan Weizmann Centre for Cellular Genomics, Garvan Institute of Medical Research, Sydney, Australia.,Graduate School of Biomedical Engineering, UNSW Sydney, Kensington, Australia
| | - Ralph Patrick
- Victor Chang Cardiac Research Institute, Darlinghurst, Australia.,Stem Cells Australia, Melbourne Brain Centre, University of Melbourne, Victoria, Australia.,St. Vincent's Clinical School, UNSW Sydney, Kensington, Australia
| | - Aude Dorison
- Victor Chang Cardiac Research Institute, Darlinghurst, Australia.,Stem Cells Australia, Melbourne Brain Centre, University of Melbourne, Victoria, Australia
| | - Munira Xaymardan
- Victor Chang Cardiac Research Institute, Darlinghurst, Australia.,Stem Cells Australia, Melbourne Brain Centre, University of Melbourne, Victoria, Australia.,School of Dentistry, Faculty of Medicine and Health, University of Sydney, Westmead Hospital, Westmead, Australia
| | - Vaibhao Janbandhu
- Victor Chang Cardiac Research Institute, Darlinghurst, Australia.,Stem Cells Australia, Melbourne Brain Centre, University of Melbourne, Victoria, Australia.,St. Vincent's Clinical School, UNSW Sydney, Kensington, Australia
| | | | - Joshua Wk Ho
- Victor Chang Cardiac Research Institute, Darlinghurst, Australia.,St. Vincent's Clinical School, UNSW Sydney, Kensington, Australia
| | - Robert E Nordon
- Stem Cells Australia, Melbourne Brain Centre, University of Melbourne, Victoria, Australia.,Graduate School of Biomedical Engineering, UNSW Sydney, Kensington, Australia
| | - Richard P Harvey
- Victor Chang Cardiac Research Institute, Darlinghurst, Australia.,Stem Cells Australia, Melbourne Brain Centre, University of Melbourne, Victoria, Australia.,School of Biotechnology and Biomolecular Science, UNSW Sydney, Kensington, Australia
| |
Collapse
|
173
|
Wang Z, Long DW, Huang Y, Chen WCW, Kim K, Wang Y. Decellularized neonatal cardiac extracellular matrix prevents widespread ventricular remodeling in adult mammals after myocardial infarction. Acta Biomater 2019; 87:140-151. [PMID: 30710713 DOI: 10.1016/j.actbio.2019.01.062] [Citation(s) in RCA: 51] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2018] [Revised: 01/28/2019] [Accepted: 01/30/2019] [Indexed: 12/15/2022]
Abstract
Heart disease remains a leading killer in western society and irreversibly impacts the lives of millions of patients annually. While adult mammals do not possess the ability to regenerate functional cardiac tissue, neonatal mammals are capable of robust cardiomyocyte proliferation and regeneration within a week of birth. Given this change in regenerative function through development, the extracellular matrix (ECM) from adult tissues may not be conducive to promoting cardiac regeneration, although conventional ECM therapies rely exclusively on adult-derived tissues. Therefore the potential of ECM derived from neonatal mouse hearts (nmECM) to prevent adverse ventricular remodeling in adults was investigated using an in vivo model of acute myocardial infarction (MI). Following a single administration of nmECM, we observed a significant improvement in heart function while adult heart-derived ECM (amECM) did not improve these parameters. Treatment with nmECM limits scar expansion in the left ventricle and promotes revascularization of the injured region. Furthermore, nmECM induced expression of the ErbB2 receptor, simulating a neonatal-like environment and promoting neuregulin-1 associated cardiac function. Inhibition of the ErbB2 receptor effectively prevents these actions, suggesting its role in the context of nmECM as a therapy. This study shows the potential of a neonatal-derived biological material in vivo, diverting from the conventional use of adult-derived ECM therapies in research and the clinic. STATEMENT OF SIGNIFICANCE: The of use extracellular matrix biomaterials to aid tissue repair has been previously reported in many forms of injury. The majority of ECM studies to date utilized ECM derived from adult tissues that are not able to fully regenerate functional tissue. In contrast, this study tests the ability of ECM derived from a regenerative organ, the neonatal heart, to stimulate functional cardiac repair after MI. This study is the first to test its potential in vivo. Our results indicate that extracellular factors present in the neonatal environment can be used to alter the healing response in adults, and we have identified the role of ErbB2 in neonatal ECM-based cardiac repair.
Collapse
Affiliation(s)
- Zhouguang Wang
- Department of Bioengineering, Swanson School of Engineering, University of Pittsburgh, Pittsburgh, PA 15261, USA; Molecular Pharmacology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325100, China
| | - Daniel W Long
- Department of Bioengineering, Swanson School of Engineering, University of Pittsburgh, Pittsburgh, PA 15261, USA
| | - Yan Huang
- Department of Bioengineering, Swanson School of Engineering, University of Pittsburgh, Pittsburgh, PA 15261, USA; Molecular Pharmacology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325100, China
| | - William C W Chen
- Department of Bioengineering, Swanson School of Engineering, University of Pittsburgh, Pittsburgh, PA 15261, USA
| | - Kang Kim
- Department of Bioengineering, Swanson School of Engineering, University of Pittsburgh, Pittsburgh, PA 15261, USA; Center for Ultrasound Molecular Imaging and Therapeutics, Department of Medicine and Heart and Vascular Institute, University of Pittsburgh School of Medicine and University of Pittsburgh Medical Center (UPMC), Pittsburgh, PA 15260, USA; Division of Cardiology, Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA; McGowan Institute for Regenerative Medicine, University of Pittsburgh and UPMC, Pittsburgh, PA 15260, USA
| | - Yadong Wang
- Department of Bioengineering, Swanson School of Engineering, University of Pittsburgh, Pittsburgh, PA 15261, USA; McGowan Institute for Regenerative Medicine, University of Pittsburgh and UPMC, Pittsburgh, PA 15260, USA.
| |
Collapse
|
174
|
Mills RJ, Hudson JE. Bioengineering adult human heart tissue: How close are we? APL Bioeng 2019; 3:010901. [PMID: 31069330 PMCID: PMC6481734 DOI: 10.1063/1.5070106] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2018] [Accepted: 02/04/2019] [Indexed: 12/22/2022] Open
Abstract
Human pluripotent stem cells (hPSCs) have extensive applications in fundamental biology, regenerative medicine, disease modelling, and drug discovery/toxicology. Whilst large numbers of cardiomyocytes can be generated from hPSCs, extensive characterization has revealed that they have immature cardiac properties. This has raised potential concerns over their usefulness for many applications and has led to the pursuit of driving maturation of hPSC-cardiomyocytes. Currently, the best approach for driving maturity is the use of tissue engineering to generate highly functional three-dimensional heart tissue. Although we have made significant progress in this area, we have still not generated heart tissue that fully recapitulates all the properties of an adult heart. Deciphering the processes driving cardiomyocyte maturation will be instrumental in uncovering the mechanisms that govern optimal heart function and identifying new therapeutic targets for heart disease.
Collapse
Affiliation(s)
- Richard J Mills
- QIMR Berghofer Medical Research Institute, Brisbane, Queensland 4006, Australia
| | - James E Hudson
- QIMR Berghofer Medical Research Institute, Brisbane, Queensland 4006, Australia
| |
Collapse
|
175
|
Lock MC, Darby JRT, Soo JY, Brooks DA, Perumal SR, Selvanayagam JB, Seed M, Macgowan CK, Porrello ER, Tellam RL, Morrison JL. Differential Response to Injury in Fetal and Adolescent Sheep Hearts in the Immediate Post-myocardial Infarction Period. Front Physiol 2019; 10:208. [PMID: 30890961 PMCID: PMC6412108 DOI: 10.3389/fphys.2019.00208] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2018] [Accepted: 02/18/2019] [Indexed: 12/11/2022] Open
Abstract
Aim: Characterizing the response to myocardial infarction (MI) in the regenerative sheep fetus heart compared to the post-natal non-regenerative adolescent heart may reveal key morphological and molecular differences that equate to the response to MI in humans. We hypothesized that the immediate response to injury in (a) infarct compared with sham, and (b) infarct, border, and remote tissue, in the fetal sheep heart would be fundamentally different to the adolescent, allowing for repair after damage. Methods: We used a sheep model of MI induced by ligating the left anterior descending coronary artery. Surgery was performed on fetuses (105 days) and adolescent sheep (6 months). Sheep were randomly separated into MI (n = 5) or Sham (n = 5) surgery groups at both ages. We used magnetic resonance imaging (MRI), histological/immunohistochemical staining, and qRT-PCR to assess the morphological and molecular differences between the different age groups in response to infarction. Results: Magnetic resonance imaging showed no difference in fetuses for key functional parameters; however there was a significant decrease in left ventricular ejection fraction and cardiac output in the adolescent sheep heart at 3 days post-infarction. There was no significant difference in functional parameters between MRI sessions at Day 0 and Day 3 after surgery. Expression of genes involved in glucose transport and fatty acid metabolism, inflammatory cytokines as well as growth factors and cell cycle regulators remained largely unchanged in the infarcted compared to sham ventricular tissue in the fetus, but were significantly dysregulated in the adolescent sheep. Different cardiac tissue region-specific gene expression profiles were observed between the fetal and adolescent sheep. Conclusion: Fetuses demonstrated a resistance to cardiac damage not observed in the adolescent animals. The manipulation of specific gene expression profiles to a fetal-like state may provide a therapeutic strategy to treat patients following an infarction.
Collapse
Affiliation(s)
- Mitchell C Lock
- Early Origins of Adult Health Research Group, School of Pharmacy and Medical Sciences, University of South Australia, Adelaide, SA, Australia
| | - Jack R T Darby
- Early Origins of Adult Health Research Group, School of Pharmacy and Medical Sciences, University of South Australia, Adelaide, SA, Australia
| | - Jia Yin Soo
- Early Origins of Adult Health Research Group, School of Pharmacy and Medical Sciences, University of South Australia, Adelaide, SA, Australia
| | - Doug A Brooks
- Mechanisms in Cell Biology and Disease Research Group, School of Pharmacy and Medical Sciences, University of South Australia, Adelaide, SA, Australia
| | - Sunthara Rajan Perumal
- Preclinical, Imaging and Research Laboratories, South Australian Health and Medical Research Institute, Adelaide, SA, Australia
| | - Joseph B Selvanayagam
- Cardiac Imaging Research Group, Department of Heart Health, South Australian Health and Medical Research Institute, Flinders University, Adelaide, SA, Australia
| | - Mike Seed
- The Hospital for Sick Children, Division of Cardiology, Toronto, ON, Canada
| | | | - Enzo R Porrello
- Murdoch Children's Research Institute, The Royal Children's Hospital, Parkville, VIC, Australia.,Department of Physiology, School of Biomedical Sciences, University of Melbourne, Parkville, VIC, Australia
| | - Ross L Tellam
- Early Origins of Adult Health Research Group, School of Pharmacy and Medical Sciences, University of South Australia, Adelaide, SA, Australia
| | - Janna L Morrison
- Early Origins of Adult Health Research Group, School of Pharmacy and Medical Sciences, University of South Australia, Adelaide, SA, Australia
| |
Collapse
|
176
|
Senyo SE. Price check on A(i)s(l)e 13: investigation of interleukin-13 activity in cardiac regeneration. Am J Physiol Heart Circ Physiol 2019; 316:H262-H264. [PMID: 30462551 DOI: 10.1152/ajpheart.00718.2018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
|
177
|
Epigenetics, cardiovascular disease, and cellular reprogramming. J Mol Cell Cardiol 2019; 128:129-133. [PMID: 30690032 DOI: 10.1016/j.yjmcc.2019.01.019] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/15/2019] [Accepted: 01/22/2019] [Indexed: 12/29/2022]
Abstract
Under the seeming disorder of "junk" sequences the last decade has seen developments in our understanding of non-coding RNA's (ncRNAs). It's a complex revised order and nowhere is this more relevant than in the developing heart whereby old rules have been set aside to make room for new ones. The development of the mammalian heart has been studied at the genetic and cellular level for several decades because these areas were considered ideal control points. As such, detailed mechanisms governing cell lineages are well described. Emerging evidence suggests a complex new order regulated by epigenetic mechanisms mark cardiac cell lineage. Indeed, molecular cardiologists are in the process of shedding light on the roles played by ncRNAs, nucleic acid methylation and histone/chromatin modifications in specific pathologies of the heart. The aim of this article is to discuss some of the recent advances in the field of cardiovascular epigenetics that are related to direct cell reprogramming and repair. As such, we explore ncRNAs as nodes regulating signaling networks and attempt to make sense of regulatory disorder by reinforcing the importance of epigenetic components in the developmental program.
Collapse
|
178
|
Das S, Goldstone AB, Wang H, Farry J, D'Amato G, Paulsen MJ, Eskandari A, Hironaka CE, Phansalkar R, Sharma B, Rhee S, Shamskhou EA, Agalliu D, de Jesus Perez V, Woo YJ, Red-Horse K. A Unique Collateral Artery Development Program Promotes Neonatal Heart Regeneration. Cell 2019; 176:1128-1142.e18. [PMID: 30686582 DOI: 10.1016/j.cell.2018.12.023] [Citation(s) in RCA: 167] [Impact Index Per Article: 27.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2018] [Revised: 11/13/2018] [Accepted: 12/14/2018] [Indexed: 10/27/2022]
Abstract
Collateral arteries are an uncommon vessel subtype that can provide alternate blood flow to preserve tissue following vascular occlusion. Some patients with heart disease develop collateral coronary arteries, and this correlates with increased survival. However, it is not known how these collaterals develop or how to stimulate them. We demonstrate that neonatal mouse hearts use a novel mechanism to build collateral arteries in response to injury. Arterial endothelial cells (ECs) migrated away from arteries along existing capillaries and reassembled into collateral arteries, which we termed "artery reassembly". Artery ECs expressed CXCR4, and following injury, capillary ECs induced its ligand, CXCL12. CXCL12 or CXCR4 deletion impaired collateral artery formation and neonatal heart regeneration. Artery reassembly was nearly absent in adults but was induced by exogenous CXCL12. Thus, understanding neonatal regenerative mechanisms can identify pathways that restore these processes in adults and identify potentially translatable therapeutic strategies for ischemic heart disease.
Collapse
Affiliation(s)
- Soumyashree Das
- Department of Biology, Stanford University, Stanford, CA 94305, USA
| | - Andrew B Goldstone
- Department of Cardiothoracic Surgery, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Hanjay Wang
- Department of Cardiothoracic Surgery, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Justin Farry
- Department of Cardiothoracic Surgery, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Gaetano D'Amato
- Department of Biology, Stanford University, Stanford, CA 94305, USA
| | - Michael J Paulsen
- Department of Cardiothoracic Surgery, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Anahita Eskandari
- Department of Cardiothoracic Surgery, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Camille E Hironaka
- Department of Cardiothoracic Surgery, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Ragini Phansalkar
- Department of Biology, Stanford University, Stanford, CA 94305, USA; Department of Genetics, Stanford University, Stanford, CA 94305, USA
| | - Bikram Sharma
- Department of Biology, Ball State University, Muncie, IN 47306, USA
| | - Siyeon Rhee
- Department of Biology, Stanford University, Stanford, CA 94305, USA
| | - Elya Ali Shamskhou
- Department of Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Dritan Agalliu
- Departments of Neurology, Pathology and Cell Biology, and Pharmacology, Columbia University Irving Medical Center, New York, NY 10032, USA
| | | | - Y Joseph Woo
- Department of Cardiothoracic Surgery, Stanford University School of Medicine, Stanford, CA 94305, USA.
| | - Kristy Red-Horse
- Department of Biology, Stanford University, Stanford, CA 94305, USA; Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA.
| |
Collapse
|
179
|
Weeks KL. HDAC inhibitors and cardioprotection: Homing in on a mechanism of action. EBioMedicine 2019; 40:21-22. [PMID: 30639419 PMCID: PMC6413300 DOI: 10.1016/j.ebiom.2019.01.015] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2019] [Accepted: 01/08/2019] [Indexed: 11/17/2022] Open
Affiliation(s)
- Kate L Weeks
- Baker Heart and Diabetes Institute, Melbourne, VIC 3004, Australia; Department of Diabetes, Central Clinical School, Monash University, Clayton, VIC 3800, Australia.
| |
Collapse
|
180
|
Begeman IJ, Kang J. Transcriptional Programs and Regeneration Enhancers Underlying Heart Regeneration. J Cardiovasc Dev Dis 2018; 6:jcdd6010002. [PMID: 30583498 PMCID: PMC6463103 DOI: 10.3390/jcdd6010002] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2018] [Revised: 12/10/2018] [Accepted: 12/12/2018] [Indexed: 12/31/2022] Open
Abstract
The heart plays the vital role of propelling blood to the entire body, which is essential to life. While maintaining heart function is critical, adult mammalian hearts poorly regenerate damaged cardiac tissue upon injury and form scar tissue instead. Unlike adult mammals, adult zebrafish can regenerate injured hearts with no sign of scarring, making zebrafish an ideal model system with which to study the molecular mechanisms underlying heart regeneration. Investigation of heart regeneration in zebrafish together with mice has revealed multiple cardiac regeneration genes that are induced by injury to facilitate heart regeneration. Altered expression of these regeneration genes in adult mammals is one of the main causes of heart regeneration failure. Previous studies have focused on the roles of these regeneration genes, yet the regulatory mechanisms by which the expression of cardiac regeneration genes is precisely controlled are largely unknown. In this review, we will discuss the importance of differential gene expression for heart regeneration, the recent discovery of cardiac injury or regeneration enhancers, and their impact on heart regeneration.
Collapse
Affiliation(s)
- Ian J Begeman
- Department of Cell and Regenerative Biology, University of Wisconsin School of Medicine and Public Health, University of Wisconsin⁻Madison, Madison, WI 53705, USA.
| | - Junsu Kang
- Department of Cell and Regenerative Biology, University of Wisconsin School of Medicine and Public Health, University of Wisconsin⁻Madison, Madison, WI 53705, USA.
| |
Collapse
|
181
|
Cui M, Wang Z, Bassel-Duby R, Olson EN. Genetic and epigenetic regulation of cardiomyocytes in development, regeneration and disease. Development 2018; 145:145/24/dev171983. [PMID: 30573475 DOI: 10.1242/dev.171983] [Citation(s) in RCA: 57] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Embryonic and postnatal life depend on the uninterrupted function of cardiac muscle cells. These cells, termed cardiomyocytes, display many fascinating behaviors, including complex morphogenic movements, interactions with other cell types of the heart, persistent contractility and quiescence after birth. Each of these behaviors depends on complex interactions between both cardiac-restricted and widely expressed transcription factors, as well as on epigenetic modifications. Here, we review recent advances in our understanding of the genetic and epigenetic control of cardiomyocyte differentiation and proliferation during heart development, regeneration and disease. We focus on those regulators that are required for both heart development and disease, and highlight the regenerative principles that might be manipulated to restore function to the injured adult heart.
Collapse
Affiliation(s)
- Miao Cui
- Department of Molecular Biology, Hamon Center for Regenerative Science and Medicine, and Senator Paul D. Wellstone Muscular Dystrophy Cooperative Research Center, University of Texas Southwestern Medical Center, 5323 Harry Hines Boulevard, Dallas, TX 75390, USA
| | - Zhaoning Wang
- Department of Molecular Biology, Hamon Center for Regenerative Science and Medicine, and Senator Paul D. Wellstone Muscular Dystrophy Cooperative Research Center, University of Texas Southwestern Medical Center, 5323 Harry Hines Boulevard, Dallas, TX 75390, USA
| | - Rhonda Bassel-Duby
- Department of Molecular Biology, Hamon Center for Regenerative Science and Medicine, and Senator Paul D. Wellstone Muscular Dystrophy Cooperative Research Center, University of Texas Southwestern Medical Center, 5323 Harry Hines Boulevard, Dallas, TX 75390, USA
| | - Eric N Olson
- Department of Molecular Biology, Hamon Center for Regenerative Science and Medicine, and Senator Paul D. Wellstone Muscular Dystrophy Cooperative Research Center, University of Texas Southwestern Medical Center, 5323 Harry Hines Boulevard, Dallas, TX 75390, USA
| |
Collapse
|
182
|
Cardiomyocyte cell cycle dynamics and proliferation revealed through cardiac-specific transgenesis of fluorescent ubiquitinated cell cycle indicator (FUCCI). J Mol Cell Cardiol 2018; 127:154-164. [PMID: 30571978 DOI: 10.1016/j.yjmcc.2018.12.007] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/29/2018] [Revised: 11/21/2018] [Accepted: 12/17/2018] [Indexed: 01/11/2023]
Abstract
RATIONALE Understanding and manipulating the cardiomyocyte cell cycle has been the focus of decades of research, however the ultimate goal of activating mitotic activity in adult mammalian cardiomyocytes remains elusive and controversial. The relentless pursuit of controlling cardiomyocyte mitosis has been complicated and obfuscated by a multitude of indices used as evidence of cardiomyocyte cell cycle activity that lack clear identification of cardiomyocyte "proliferation" versus cell cycle progression, endoreplication, endomitosis, and even DNA damage. Unambiguous appreciation of the complexity of cardiomyocyte replication that avoids oversimplification and misinterpretation is desperately needed. OBJECTIVE Track cardiomyocyte cell cycle activity and authenticate fidelity of proliferation markers as indicators of de novo cardiomyogenesis in post-mitotic cardiomyocytes. METHODS AND RESULTS Cardiomyocytes expressing the FUCCI construct driven by the α-myosin heavy chain promoter were readily and uniformly detected through the myocardium of transgenic mice. Cardiomyocyte cell cycle activity peaks at postnatal day 2 and rapidly declines thereafter with almost all cardiomyocytes arrested at the G1/S cell cycle transition. Myocardial infarction injury in adult hearts prompts transient small increases in myocytes progressing through cell cycle without concurrent mitotic activity, indicating lack of cardiomyogenesis. In comparison, cardiomyogenic activity during early postnatal development correlated with coincidence of FUCCI and cKit+ cells that were undetectable in the adult myocardium. CONCLUSIONS Cardiomyocyte-specific expression of Fluorescence Ubiquitination-based Cell Cycle Indicators (FUCCI) reveals previously unappreciated aspects of cardiomyocyte cell cycle arrest and biological activity in postnatal development and in response to pathologic damage. Compared to many other methods and model systems, the FUCCI transgenic (FUCCI-Tg) mouse represents a valuable tool to unambiguously track cell cycle and proliferation of the entire cardiomyocyte population in the adult murine heart. FUCCI-Tg provides a desperately needed novel approach in the armamentarium of tools to validate cardiomyocyte proliferative activity that will reveal cell cycle progression, discriminate between cycle progression, DNA replication, and proliferation, and provide important insight for enhancing cardiomyocyte proliferation in the context of adult myocardial tissue.
Collapse
|
183
|
Ye L, D’Agostino G, Loo SJ, Wang CX, Su LP, Tan SH, Tee GZ, Pua CJ, Pena EM, Cheng RB, Chen WC, Abdurrachim D, Lalic J, Tan RS, Lee TH, Zhang J, Cook SA. Early Regenerative Capacity in the Porcine Heart. Circulation 2018; 138:2798-2808. [DOI: 10.1161/circulationaha.117.031542] [Citation(s) in RCA: 141] [Impact Index Per Article: 20.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Affiliation(s)
- Lei Ye
- National Heart Research Institute Singapore, National Heart Centre Singapore (L.Y., S.J.L., C.X.W., L.P.S., S.H.T., G.Z.T., C.J.P., R.S.T., S.A.C.)
| | - Giuseppe D’Agostino
- Programme in Cardiovascular & Metabolic Disorders, Duke-National University of Singapore (G.D., S.A.C.)
| | - Sze Jie Loo
- National Heart Research Institute Singapore, National Heart Centre Singapore (L.Y., S.J.L., C.X.W., L.P.S., S.H.T., G.Z.T., C.J.P., R.S.T., S.A.C.)
| | - Chen Xu Wang
- National Heart Research Institute Singapore, National Heart Centre Singapore (L.Y., S.J.L., C.X.W., L.P.S., S.H.T., G.Z.T., C.J.P., R.S.T., S.A.C.)
| | - Li Ping Su
- National Heart Research Institute Singapore, National Heart Centre Singapore (L.Y., S.J.L., C.X.W., L.P.S., S.H.T., G.Z.T., C.J.P., R.S.T., S.A.C.)
| | - Shi Hua Tan
- National Heart Research Institute Singapore, National Heart Centre Singapore (L.Y., S.J.L., C.X.W., L.P.S., S.H.T., G.Z.T., C.J.P., R.S.T., S.A.C.)
| | - Gui Zhen Tee
- National Heart Research Institute Singapore, National Heart Centre Singapore (L.Y., S.J.L., C.X.W., L.P.S., S.H.T., G.Z.T., C.J.P., R.S.T., S.A.C.)
| | - Chee Jian Pua
- National Heart Research Institute Singapore, National Heart Centre Singapore (L.Y., S.J.L., C.X.W., L.P.S., S.H.T., G.Z.T., C.J.P., R.S.T., S.A.C.)
| | - Edgar Macabe Pena
- SingHealth Experimental Medicine Centre, Singapore Health Services Pte Ltd (E.M.P., R.B.C.)
| | - Redmond Belen Cheng
- SingHealth Experimental Medicine Centre, Singapore Health Services Pte Ltd (E.M.P., R.B.C.)
| | - Way Cherng Chen
- Singapore Bioimaging Consortium, A*STAR (W.C.C., D.A., J.L., T.H.L.)
| | | | - Janise Lalic
- Singapore Bioimaging Consortium, A*STAR (W.C.C., D.A., J.L., T.H.L.)
| | - Ru San Tan
- National Heart Research Institute Singapore, National Heart Centre Singapore (L.Y., S.J.L., C.X.W., L.P.S., S.H.T., G.Z.T., C.J.P., R.S.T., S.A.C.)
| | - Teck Hock Lee
- Singapore Bioimaging Consortium, A*STAR (W.C.C., D.A., J.L., T.H.L.)
| | - JianYi Zhang
- Department of Biomedical Engineering, University of Alabama at Birmingham (J.Y.Z.)
| | - Stuart Alexander Cook
- National Heart Research Institute Singapore, National Heart Centre Singapore (L.Y., S.J.L., C.X.W., L.P.S., S.H.T., G.Z.T., C.J.P., R.S.T., S.A.C.)
- Programme in Cardiovascular & Metabolic Disorders, Duke-National University of Singapore (G.D., S.A.C.)
- National Heart and Lung Institute, Imperial College, London, United Kingdom (S.A.C.)
| |
Collapse
|
184
|
Affiliation(s)
- Ahmed I Mahmoud
- Department of Cell and Regenerative Biology, University of Wisconsin-Madison School of Medicine and Public Health (A.I.M.)
| | - Enzo R Porrello
- Murdoch Children's Research Institute, The Royal Children's Hospital, Parkville, Victoria, Australia (E.R.P.)
- Department of Physiology, School of Biomedical Sciences, The University of Melbourne, Parkville, Victoria, Australia (E.R.P.)
| |
Collapse
|
185
|
Zhu W, Zhang E, Zhao M, Chong Z, Fan C, Tang Y, Hunter JD, Borovjagin AV, Walcott GP, Chen JY, Qin G, Zhang J. Regenerative Potential of Neonatal Porcine Hearts. Circulation 2018; 138:2809-2816. [PMID: 30030418 PMCID: PMC6301098 DOI: 10.1161/circulationaha.118.034886] [Citation(s) in RCA: 201] [Impact Index Per Article: 28.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/14/2018] [Accepted: 06/29/2018] [Indexed: 11/16/2022]
Abstract
BACKGROUND Rodent hearts can regenerate myocardium lost to apical resection or myocardial infarction for up to 7 days after birth, but whether a similar window for myocardial regeneration also exists in large mammals is unknown. METHODS Acute myocardial infarction (AMI) was surgically induced in neonatal pigs on postnatal days 1, 2, 3, 7, and 14 (ie, the P1, P2, P3, P7, and P14 groups, respectively). Cardiac systolic function was evaluated before AMI and at 30 days post-AMI via transthoracic echocardiography. Cardiomyocyte cell cycle activity was assessed via immunostaining for proliferation and mitosis markers, infarct size was evaluated histologically, and telomerase activity was measured by quantitative polymerase chain reaction. RESULTS Systolic function at day 30 post-AMI was largely restored in P1 animals and partially restored in P2 animals, but significantly impaired when AMI was induced on postnatal day 3 or later. Hearts of P1 animals showed little evidence of scar formation or wall thinning on day 30 after AMI, with increased measures of cell-cycle activity seen 6 days after AMI (ie, postnatal day 7) compared with postnatal day 7 in noninfarcted hearts. CONCLUSIONS The neonatal porcine heart is capable of regeneration after AMI during the first 2 days of life. This phenomenon is associated with induction of cardiomyocyte proliferation and is lost when cardiomyocytes exit the cell cycle shortly after birth.
Collapse
Affiliation(s)
- Wuqiang Zhu
- School of Medicine and School of Engineering, the University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Eric Zhang
- School of Medicine and School of Engineering, the University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Meng Zhao
- School of Medicine and School of Engineering, the University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Zechen Chong
- Department of Genetics, School of Medicine, University of Alabama at Birmingham, AL 35294, USA
| | - Chengming Fan
- School of Medicine and School of Engineering, the University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Yawen Tang
- School of Medicine and School of Engineering, the University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Jervaughn D. Hunter
- School of Medicine and School of Engineering, the University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Anton V. Borovjagin
- School of Medicine and School of Engineering, the University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Gregory P. Walcott
- School of Medicine and School of Engineering, the University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Jake Y. Chen
- Informatics Institute, School of Medicine, University of Alabama at Birmingham, AL 35294, USA
| | - Gangjian Qin
- School of Medicine and School of Engineering, the University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Jianyi Zhang
- School of Medicine and School of Engineering, the University of Alabama at Birmingham, Birmingham, AL 35294, USA
| |
Collapse
|
186
|
Bassaneze V, Lee RT. Revealing Pathways of Cardiac Regeneration. CIRCULATION-GENOMIC AND PRECISION MEDICINE 2018; 11:e002053. [PMID: 30520316 DOI: 10.1161/circgen.117.002053] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Affiliation(s)
- Vinícius Bassaneze
- From the Cardiovascular Division, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA and the Department of Stem Cell and Regenerative Biology and the Harvard Stem Cell Institute, Harvard University, Cambridge, MA
| | - Richard T Lee
- From the Cardiovascular Division, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA and the Department of Stem Cell and Regenerative Biology and the Harvard Stem Cell Institute, Harvard University, Cambridge, MA
| |
Collapse
|
187
|
Farber G, Parks MM, Lustgarten Guahmich N, Zhang Y, Monette S, Blanchard SC, Di Lorenzo A, Blobel CP. ADAM10 controls the differentiation of the coronary arterial endothelium. Angiogenesis 2018; 22:237-250. [PMID: 30446855 DOI: 10.1007/s10456-018-9653-2] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2018] [Accepted: 11/08/2018] [Indexed: 12/12/2022]
Abstract
The coronary vasculature is crucial for normal heart function, yet much remains to be learned about its development, especially the maturation of coronary arterial endothelium. Here, we show that endothelial inactivation of ADAM10, a key regulator of Notch signaling, leads to defects in coronary arterial differentiation, as evidenced by dysregulated genes related to Notch signaling and arterial identity. Moreover, transcriptome analysis indicated reduced EGFR signaling in A10ΔEC coronary endothelium. Further analysis revealed that A10ΔEC mice have enlarged dysfunctional hearts with abnormal myocardial compaction, and increased expression of venous and immature endothelium markers. These findings provide the first evidence for a potential role for endothelial ADAM10 in cardioprotective homeostatic EGFR signaling and implicate ADAM10/Notch signaling in coronary arterial cell specification, which is vital for normal heart development and function. The ADAM10/Notch signaling pathway thus emerges as a potential therapeutic target for improving the regenerative capacity and maturation of the coronary vasculature.
Collapse
Affiliation(s)
- Gregory Farber
- Department of Physiology, Biophysics and Systems Biology, Weill Cornell Medicine, New York, NY, USA
| | - Matthew M Parks
- Department of Physiology, Biophysics and Systems Biology, Weill Cornell Medicine, New York, NY, USA
| | | | - Yi Zhang
- Center for Vascular Biology, Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, NY, USA
| | - Sébastien Monette
- Laboratory of Comparative Pathology, Hospital for Special Surgery, Memorial Sloan Kettering Cancer Center, The Rockefeller University, Weill Cornell Medicine, New York, NY, USA
| | - Scott C Blanchard
- Department of Physiology, Biophysics and Systems Biology, Weill Cornell Medicine, New York, NY, USA.,Tri-Institutional Training Program in Chemical Biology, Weill Cornell Medicine, New York, NY, USA
| | - Annarita Di Lorenzo
- Center for Vascular Biology, Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, NY, USA
| | - Carl P Blobel
- Department of Physiology, Biophysics and Systems Biology, Weill Cornell Medicine, New York, NY, USA. .,Arthritis and Tissue Degeneration Program, Hospital for Special Surgery, S-Building, Room 702, 535 East 70th Street, New York, NY, 10021, USA. .,Institute for Advanced Study, Technical University Munich, Munich, Germany.
| |
Collapse
|
188
|
Nguyen MN, Ziemann M, Kiriazis H, Su Y, Thomas Z, Lu Q, Donner DG, Zhao WB, Rafehi H, Sadoshima J, McMullen JR, El-Osta A, Du XJ. Galectin-3 deficiency ameliorates fibrosis and remodeling in dilated cardiomyopathy mice with enhanced Mst1 signaling. Am J Physiol Heart Circ Physiol 2018; 316:H45-H60. [PMID: 30387702 DOI: 10.1152/ajpheart.00609.2018] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
Dilated cardiomyopathy (DCM) is a major cause of heart failure without effective therapy. Fibrogenesis plays a key role in the development of DCM, but little is known of the expression of the profibrotic factor galectin-3 (Gal-3) and its role in DCM pathophysiology. In a mouse DCM model with transgenic (TG) overexpression of mammalian sterile 20-like kinase 1 (Mst1), we studied Gal-3 expression and effects of the Gal-3 inhibitor modified citrus pectin (MCP) or Gal-3 gene knockout (KO). Gal-3 deletion in TG mice (TG/KO) was achieved by crossbreeding Mst1-TG mice with Gal-3 KO mice. The DCM phenotype was assessed by echocardiography and micromanometry. Cardiac expression of Gal-3 and fibrosis were determined. The cardiac transcriptome was profiled by RNA sequencing. Mst1-TG mice at 3-8 mo of age exhibited upregulated expression of Gal-3 by ~40-fold. TG mice had dilatation of cardiac chambers, suppressed left ventricular (LV) ejection fraction, poor LV contractility and relaxation, a threefold increase in LV collagen content, and upregulated fibrotic genes. Four-month treatment with MCP showed no beneficial effects. Gal-3 deletion in Mst1-TG mice attenuated chamber dilatation, organ congestion, and fibrogenesis. RNA sequencing identified profound disturbances by Mst1 overexpression in the cardiac transcriptome, which largely remained in TG/KO hearts. Gal-3 deletion in Mst1-TG mice, however, partially reversed the dysregulated transcriptional signaling involving extracellular matrix remodeling and collagen formation. We conclude that cardiac Mst1 activation leads to marked Gal-3 upregulation and transcriptome disturbances in the heart. Gal-3 deficiency attenuated cardiac remodeling and fibrotic signaling. NEW & NOTEWORTHY We found in a transgenic mouse dilated cardiomyopathy (DCM) model a pronounced upregulation of galectin-3 in cardiomyocytes. Galectin-3 gene deletion reduced cardiac fibrosis and fibrotic gene profiles and ameliorated cardiac remodeling and dysfunction. These benefits of galectin-3 deletion were in contrast to the lack of effect of treatment with the galectin-3 inhibitor modified citrus pectin. Our study suggests that suppression of galectin-3 mRNA expression could be used to treat DCM with high cardiac galectin-3 content.
Collapse
Affiliation(s)
- My-Nhan Nguyen
- Baker Heart and Diabetes Institute , Melbourne, Victoria , Australia.,Central Clinical School, Monash University , Melbourne, Victoria , Australia
| | - Mark Ziemann
- Department of Diabetes, Monash University , Melbourne, Victoria , Australia
| | - Helen Kiriazis
- Baker Heart and Diabetes Institute , Melbourne, Victoria , Australia
| | - Yidan Su
- Baker Heart and Diabetes Institute , Melbourne, Victoria , Australia
| | - Zara Thomas
- Baker Heart and Diabetes Institute , Melbourne, Victoria , Australia
| | - Qun Lu
- Baker Heart and Diabetes Institute , Melbourne, Victoria , Australia.,Health Science Center, Xian Jiaotong University , Xian , People's Republic of China
| | - Daniel G Donner
- Baker Heart and Diabetes Institute , Melbourne, Victoria , Australia
| | - Wei-Bo Zhao
- Baker Heart and Diabetes Institute , Melbourne, Victoria , Australia
| | - Haloom Rafehi
- Department of Diabetes, Monash University , Melbourne, Victoria , Australia
| | - Junichi Sadoshima
- Department of Cell Biology and Molecular Medicine, Rutgers New Jersey Medical School , Newark, New Jersey
| | - Julie R McMullen
- Baker Heart and Diabetes Institute , Melbourne, Victoria , Australia.,Central Clinical School, Monash University , Melbourne, Victoria , Australia
| | - Assam El-Osta
- Department of Diabetes, Monash University , Melbourne, Victoria , Australia.,Prince of Wales Hospital, The Chinese University of Hong Kong , Shatin, Hong Kong Special Administrative Region , People's Republic of China
| | - Xiao-Jun Du
- Baker Heart and Diabetes Institute , Melbourne, Victoria , Australia.,Central Clinical School, Monash University , Melbourne, Victoria , Australia.,Health Science Center, Xian Jiaotong University , Xian , People's Republic of China
| |
Collapse
|
189
|
Friedman CE, Nguyen Q, Lukowski SW, Helfer A, Chiu HS, Miklas J, Levy S, Suo S, Han JDJ, Osteil P, Peng G, Jing N, Baillie GJ, Senabouth A, Christ AN, Bruxner TJ, Murry CE, Wong ES, Ding J, Wang Y, Hudson J, Ruohola-Baker H, Bar-Joseph Z, Tam PPL, Powell JE, Palpant NJ. Single-Cell Transcriptomic Analysis of Cardiac Differentiation from Human PSCs Reveals HOPX-Dependent Cardiomyocyte Maturation. Cell Stem Cell 2018; 23:586-598.e8. [PMID: 30290179 PMCID: PMC6220122 DOI: 10.1016/j.stem.2018.09.009] [Citation(s) in RCA: 190] [Impact Index Per Article: 27.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2017] [Revised: 05/30/2018] [Accepted: 09/13/2018] [Indexed: 11/25/2022]
Abstract
Cardiac differentiation of human pluripotent stem cells (hPSCs) requires orchestration of dynamic gene regulatory networks during stepwise fate transitions but often generates immature cell types that do not fully recapitulate properties of their adult counterparts, suggesting incomplete activation of key transcriptional networks. We performed extensive single-cell transcriptomic analyses to map fate choices and gene expression programs during cardiac differentiation of hPSCs and identified strategies to improve in vitro cardiomyocyte differentiation. Utilizing genetic gain- and loss-of-function approaches, we found that hypertrophic signaling is not effectively activated during monolayer-based cardiac differentiation, thereby preventing expression of HOPX and its activation of downstream genes that govern late stages of cardiomyocyte maturation. This study therefore provides a key transcriptional roadmap of in vitro cardiac differentiation at single-cell resolution, revealing fundamental mechanisms underlying heart development and differentiation of hPSC-derived cardiomyocytes.
Collapse
Affiliation(s)
- Clayton E Friedman
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, QLD 4072, Australia; Centre for Cardiac and Vascular Biology, The University of Queensland, Brisbane, QLD 4072, Australia
| | - Quan Nguyen
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, QLD 4072, Australia
| | - Samuel W Lukowski
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, QLD 4072, Australia
| | - Abbigail Helfer
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, QLD 4072, Australia; Centre for Cardiac and Vascular Biology, The University of Queensland, Brisbane, QLD 4072, Australia
| | - Han Sheng Chiu
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, QLD 4072, Australia; Centre for Cardiac and Vascular Biology, The University of Queensland, Brisbane, QLD 4072, Australia
| | - Jason Miklas
- Departments of Pathology, Biochemistry, Bioengineering and Medicine/Cardiology, Institute for Stem Cell and Regenerative Medicine, The University of Washington, Seattle, WA 98195, USA
| | - Shiri Levy
- Departments of Pathology, Biochemistry, Bioengineering and Medicine/Cardiology, Institute for Stem Cell and Regenerative Medicine, The University of Washington, Seattle, WA 98195, USA
| | - Shengbao Suo
- Key Laboratory of Computational Biology, Chinese Academy of Sciences-Max Planck Partner Institute for Computational Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, 320 Yue Yang Road, Shanghai 200031, China
| | - Jing-Dong Jackie Han
- Key Laboratory of Computational Biology, Chinese Academy of Sciences-Max Planck Partner Institute for Computational Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, 320 Yue Yang Road, Shanghai 200031, China
| | - Pierre Osteil
- Embryology Unit, Children's Medical Research Institute, The University of Sydney, Westmead, NSW 2145, Australia
| | - Guangdun Peng
- State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences; University of Chinese Academy of Sciences, 320 Yue Yang Road, Shanghai 200031, China
| | - Naihe Jing
- State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences; University of Chinese Academy of Sciences, 320 Yue Yang Road, Shanghai 200031, China; School of Life Science and Technology, ShanghaiTech University, 100 Haike Road, Shanghai 201210, China
| | - Greg J Baillie
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, QLD 4072, Australia
| | - Anne Senabouth
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, QLD 4072, Australia
| | - Angelika N Christ
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, QLD 4072, Australia
| | - Timothy J Bruxner
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, QLD 4072, Australia
| | - Charles E Murry
- Departments of Pathology, Biochemistry, Bioengineering and Medicine/Cardiology, Institute for Stem Cell and Regenerative Medicine, The University of Washington, Seattle, WA 98195, USA
| | - Emily S Wong
- School of Biomedical Sciences, The University of Queensland, Brisbane, QLD 4072, Australia
| | - Jun Ding
- Computational Biology Department, School of Computer Science, Carnegie Mellon University, Pittsburgh, PA 15213, USA
| | - Yuliang Wang
- Departments of Pathology, Biochemistry, Bioengineering and Medicine/Cardiology, Institute for Stem Cell and Regenerative Medicine, The University of Washington, Seattle, WA 98195, USA; Paul G. Allen School of Computer Science & Engineering, University of Washington, Seattle, WA 98195, USA
| | - James Hudson
- Queensland Institute for Medical Research, Brisbane, QLD 4006, Australia
| | - Hannele Ruohola-Baker
- Departments of Pathology, Biochemistry, Bioengineering and Medicine/Cardiology, Institute for Stem Cell and Regenerative Medicine, The University of Washington, Seattle, WA 98195, USA
| | - Ziv Bar-Joseph
- Computational Biology Department, School of Computer Science, Carnegie Mellon University, Pittsburgh, PA 15213, USA
| | - Patrick P L Tam
- Embryology Unit, Children's Medical Research Institute, The University of Sydney, Westmead, NSW 2145, Australia; School of Medical Sciences, Sydney Medical School, University of Sydney, NSW 2006, Australia
| | - Joseph E Powell
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, QLD 4072, Australia; Garvan-Weizmann Centre for Cellular Genomics, Garvan Institute for Medical Research, Sydney, NSW 2010, Australia.
| | - Nathan J Palpant
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, QLD 4072, Australia; Centre for Cardiac and Vascular Biology, The University of Queensland, Brisbane, QLD 4072, Australia; School of Biomedical Sciences, The University of Queensland, Brisbane, QLD 4072, Australia.
| |
Collapse
|
190
|
Abstract
Ischaemic heart disease is a leading cause of death worldwide. Injury to the heart is followed by loss of the damaged cardiomyocytes, which are replaced with fibrotic scar tissue. Depletion of cardiomyocytes results in decreased cardiac contraction, which leads to pathological cardiac dilatation, additional cardiomyocyte loss, and mechanical dysfunction, culminating in heart failure. This sequential reaction is defined as cardiac remodelling. Many therapies have focused on preventing the progressive process of cardiac remodelling to heart failure. However, after patients have developed end-stage heart failure, intervention is limited to heart transplantation. One of the main reasons for the dramatic injurious effect of cardiomyocyte loss is that the adult human heart has minimal regenerative capacity. In the past 2 decades, several strategies to repair the injured heart and improve heart function have been pursued, including cellular and noncellular therapies. In this Review, we discuss current therapeutic approaches for cardiac repair and regeneration, describing outcomes, limitations, and future prospects of preclinical and clinical trials of heart regeneration. Substantial progress has been made towards understanding the cellular and molecular mechanisms regulating heart regeneration, offering the potential to control cardiac remodelling and redirect the adult heart to a regenerative state.
Collapse
Affiliation(s)
- Hisayuki Hashimoto
- Department of Molecular Biology, Hamon Center for Regenerative Science and Medicine, University of Texas Southwestern Medical Center, Dallas, TX, USA
- Senator Paul D. Wellstone Muscular Dystrophy Cooperative Research Center, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Eric N Olson
- Department of Molecular Biology, Hamon Center for Regenerative Science and Medicine, University of Texas Southwestern Medical Center, Dallas, TX, USA.
- Senator Paul D. Wellstone Muscular Dystrophy Cooperative Research Center, University of Texas Southwestern Medical Center, Dallas, TX, USA.
| | - Rhonda Bassel-Duby
- Department of Molecular Biology, Hamon Center for Regenerative Science and Medicine, University of Texas Southwestern Medical Center, Dallas, TX, USA
- Senator Paul D. Wellstone Muscular Dystrophy Cooperative Research Center, University of Texas Southwestern Medical Center, Dallas, TX, USA
| |
Collapse
|
191
|
Talman V, Kivelä R. Cardiomyocyte-Endothelial Cell Interactions in Cardiac Remodeling and Regeneration. Front Cardiovasc Med 2018; 5:101. [PMID: 30175102 PMCID: PMC6108380 DOI: 10.3389/fcvm.2018.00101] [Citation(s) in RCA: 109] [Impact Index Per Article: 15.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2018] [Accepted: 07/05/2018] [Indexed: 12/14/2022] Open
Abstract
The heart is a complex organ consisting of various cell types, each of which plays an important role in both physiological and pathophysiological conditions. The cells communicate with each other through direct cell-cell interactions and paracrine signaling, and both homotypic and heterotypic cell interactions contribute to the organized structure and proper function of the heart. Cardiomyocytes (CMs) and endothelial cells (ECs) are two of the most abundant cardiac cell types and they also play central roles in both cardiac remodeling and regeneration. The postnatal cell cycle withdrawal of CMs, which takes place within days or weeks after birth, represents the major barrier for regeneration in adult mammalian hearts, as adult CMs exhibit a very low proliferative capacity. Recent evidence highlights the importance of ECs not only as the most abundant cell type in the heart but also as key players in post-infarction remodeling and regeneration. In this MiniReview, we focus on blood vascular ECs and CMs and their roles and interactions in cardiac physiology and pathologies, with a special emphasis on cardiac regeneration. We summarize the known mediators of the bidirectional CM-EC interactions and discuss the related recent advances in the development of therapies aiming to promote heart repair and regeneration targeting these two cell types.
Collapse
Affiliation(s)
- Virpi Talman
- Drug Research Program and Division of Pharmacology and Pharmacotherapy, Faculty of Pharmacy, University of Helsinki, Helsinki, Finland
| | - Riikka Kivelä
- Wihuri Research Institute and Translational Cancer Biology Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| |
Collapse
|
192
|
Marques FZ, Chu PY, Ziemann M, Kaspi A, Kiriazis H, Du XJ, El-Osta A, Kaye DM. Age-Related Differential Structural and Transcriptomic Responses in the Hypertensive Heart. Front Physiol 2018; 9:817. [PMID: 30038575 PMCID: PMC6046461 DOI: 10.3389/fphys.2018.00817] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2018] [Accepted: 06/11/2018] [Indexed: 01/20/2023] Open
Abstract
While aging is a critical risk factor for heart failure, it remains uncertain whether the aging heart responds differentially to a hypertensive stimuli. Here we investigated phenotypic and transcriptomic differences between the young and aging heart using a mineralocorticoid-excess model of hypertension. Ten-week (“young”) and 36-week (“aging”) mice underwent a unilateral uninephrectomy with deoxycorticosterone acetate (DOCA) pellet implantation (n = 6–8/group) and were followed for 6 weeks. Cardiac structure and function, blood pressure (BP) and the cardiac transcriptome were subsequently examined. Young and aging DOCA mice had high BP, increased cardiac mass, cardiac hypertrophy, and fibrosis. Left ventricular end-diastolic pressure increased in aging DOCA-treated mice in contrast to young DOCA mice. Interstitial and perivascular fibrosis occurred in response to DOCA, but perivascular fibrosis was greater in aging mice. Transcriptomic analysis showed that young mice had features of higher oxidative stress, likely due to activation of the respiratory electron transport chain. In contrast, aging mice showed up-regulation of collagen formation in association with activation of innate immunity together with markers of inflammation including cytokine and platelet signaling. In comparison to younger mice, aging mice demonstrated different phenotypic and molecular responses to hypertensive stress. These findings have potential implications for the pathogenesis of age-related forms of cardiovascular disease, particularly heart failure.
Collapse
Affiliation(s)
- Francine Z Marques
- Heart Failure Research Group, Baker Heart and Diabetes Institute, Melbourne, VIC, Australia.,Department of Pharmacology, Faculty of Medicine Nursing and Health Sciences, Monash University, Melbourne, VIC, Australia
| | - Po-Yin Chu
- Heart Failure Research Group, Baker Heart and Diabetes Institute, Melbourne, VIC, Australia
| | - Mark Ziemann
- Epigenetics in Human Health and Disease, Department of Diabetes, Monash University, Melbourne, VIC, Australia
| | - Antony Kaspi
- Epigenetics in Human Health and Disease, Department of Diabetes, Monash University, Melbourne, VIC, Australia
| | - Helen Kiriazis
- Experimental Cardiology Laboratory, Baker Heart and Diabetes Institute, Melbourne, VIC, Australia
| | - Xiao-Jun Du
- Experimental Cardiology Laboratory, Baker Heart and Diabetes Institute, Melbourne, VIC, Australia.,Central Clinical School, Faculty of Medicine Nursing and Health Sciences, Monash University, Melbourne, VIC, Australia
| | - Assam El-Osta
- Epigenetics in Human Health and Disease, Department of Diabetes, Monash University, Melbourne, VIC, Australia.,Hong Kong Institute of Diabetes and Obesity, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong, Hong Kong.,Department of Pathology, The University of Melbourne, Melbourne, VIC, Australia
| | - David M Kaye
- Heart Failure Research Group, Baker Heart and Diabetes Institute, Melbourne, VIC, Australia.,Central Clinical School, Faculty of Medicine Nursing and Health Sciences, Monash University, Melbourne, VIC, Australia.,Heart Centre, Alfred Hospital, Melbourne, VIC, Australia
| |
Collapse
|
193
|
Heart muscle regeneration: the wonder of a Cardio-Cocktail. Cell Res 2018; 28:503-504. [PMID: 29674740 DOI: 10.1038/s41422-018-0035-5] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2018] [Accepted: 03/28/2018] [Indexed: 02/03/2023] Open
|
194
|
Golan-Lagziel T, Lewis YE, Shkedi O, Douvdevany G, Caspi LH, Kehat I. Analysis of rat cardiac myocytes and fibroblasts identifies combinatorial enhancer organization and transcription factor families. J Mol Cell Cardiol 2018; 116:91-105. [DOI: 10.1016/j.yjmcc.2018.02.003] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/09/2017] [Revised: 01/31/2018] [Accepted: 02/02/2018] [Indexed: 12/20/2022]
|
195
|
Ulmer BM, Stoehr A, Schulze ML, Patel S, Gucek M, Mannhardt I, Funcke S, Murphy E, Eschenhagen T, Hansen A. Contractile Work Contributes to Maturation of Energy Metabolism in hiPSC-Derived Cardiomyocytes. Stem Cell Reports 2018; 10:834-847. [PMID: 29503093 PMCID: PMC5919410 DOI: 10.1016/j.stemcr.2018.01.039] [Citation(s) in RCA: 136] [Impact Index Per Article: 19.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2017] [Revised: 01/29/2018] [Accepted: 01/30/2018] [Indexed: 02/07/2023] Open
Abstract
Energy metabolism is a key aspect of cardiomyocyte biology. Human induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs) are a promising tool for biomedical application, but they are immature and have not undergone metabolic maturation related to early postnatal development. To assess whether cultivation of hiPSC-CMs in 3D engineered heart tissue format leads to maturation of energy metabolism, we analyzed the mitochondrial and metabolic state of 3D hiPSC-CMs and compared it with 2D culture. 3D hiPSC-CMs showed increased mitochondrial mass, DNA content, and protein abundance (proteome). While hiPSC-CMs exhibited the principal ability to use glucose, lactate, and fatty acids as energy substrates irrespective of culture format, hiPSC-CMs in 3D performed more oxidation of glucose, lactate, and fatty acid and less anaerobic glycolysis. The increase in mitochondrial mass and DNA in 3D was diminished by pharmacological reduction of contractile force. In conclusion, contractile work contributes to metabolic maturation of hiPSC-CMs. Higher mitochondrial mass, protein, and DNA content in 3D versus 2D hiPSC-CMs Similarity of mitochondrial proteomes between 3D hiPSC-CMs and adult human heart Preference for oxidative metabolism in favor of anaerobic glycolysis in 3D hiPSC-CMs
Collapse
Affiliation(s)
- Bärbel M Ulmer
- University Medical Center Hamburg Eppendorf, Department of Experimental Pharmacology and Toxicology, 20246 Hamburg, Germany; German Center for Heart Research (DZHK), Partner Site Hamburg/Kiel/Lübeck, Hamburg, Germany.
| | - Andrea Stoehr
- Systems Biology Center, National Heart Lung and Blood Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Mirja L Schulze
- University Medical Center Hamburg Eppendorf, Department of Experimental Pharmacology and Toxicology, 20246 Hamburg, Germany; German Center for Heart Research (DZHK), Partner Site Hamburg/Kiel/Lübeck, Hamburg, Germany
| | - Sajni Patel
- Proteomics Core Facility, National Heart Lung and Blood Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Marjan Gucek
- Proteomics Core Facility, National Heart Lung and Blood Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Ingra Mannhardt
- University Medical Center Hamburg Eppendorf, Department of Experimental Pharmacology and Toxicology, 20246 Hamburg, Germany; German Center for Heart Research (DZHK), Partner Site Hamburg/Kiel/Lübeck, Hamburg, Germany
| | - Sandra Funcke
- University Medical Center Hamburg Eppendorf, Department of Experimental Pharmacology and Toxicology, 20246 Hamburg, Germany; German Center for Heart Research (DZHK), Partner Site Hamburg/Kiel/Lübeck, Hamburg, Germany
| | - Elizabeth Murphy
- Systems Biology Center, National Heart Lung and Blood Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Thomas Eschenhagen
- University Medical Center Hamburg Eppendorf, Department of Experimental Pharmacology and Toxicology, 20246 Hamburg, Germany; German Center for Heart Research (DZHK), Partner Site Hamburg/Kiel/Lübeck, Hamburg, Germany
| | - Arne Hansen
- University Medical Center Hamburg Eppendorf, Department of Experimental Pharmacology and Toxicology, 20246 Hamburg, Germany; German Center for Heart Research (DZHK), Partner Site Hamburg/Kiel/Lübeck, Hamburg, Germany.
| |
Collapse
|
196
|
Gross JC, Zelarayán LC. The Mingle-Mangle of Wnt Signaling and Extracellular Vesicles: Functional Implications for Heart Research. Front Cardiovasc Med 2018; 5:10. [PMID: 29564334 PMCID: PMC5850280 DOI: 10.3389/fcvm.2018.00010] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2017] [Accepted: 01/29/2018] [Indexed: 12/21/2022] Open
Abstract
Wnt signaling is an important pathway in health and disease and a key regulator of stem cell maintenance, differentiation, and proliferation. During heart development, Wnt signaling controls specification, proliferation and differentiation of cardiovascular cells. In this regard, the role of activated Wnt signaling in cardiogenesis is well defined. However, the knowledge about signaling transmission has been challenged. Recently, the packaging of hydrophobic Wnt proteins on extracellular vesicles (EVs) has emerged as a mechanism to facilitate their extracellular spreading and their functioning as morphogens. EVs spread systemically and therefore can have pleiotropic effects on very different cell types. They are heavily studied in tumor biology where they affect tumor growth and vascularization and can serve as biomarkers in liquid biopsies. In this review we will highlight recent discoveries of factors involved in the release of Wnts on EVs and its potential implications in the communication between physiological and pathological heart cells.
Collapse
Affiliation(s)
- Julia Christina Gross
- Hematology and Oncology, University Medical Center Göttingen, Göttingen, Germany.,Developmental Biochemistry, University Medical Center Göttingen, Göttingen, Germany
| | - Laura Cecilia Zelarayán
- Institute of Pharmacology and Toxicology, University Medical Center Göttingen, Göttingen, Germany.,Partner Site Göttingen, German Centre for Cardiovascular Research (DZHK), Göttingen, Germany
| |
Collapse
|
197
|
Ussher JR, Campbell JE, Mulvihill EE, Baggio LL, Bates HE, McLean BA, Gopal K, Capozzi M, Yusta B, Cao X, Ali S, Kim M, Kabir MG, Seino Y, Suzuki J, Drucker DJ. Inactivation of the Glucose-Dependent Insulinotropic Polypeptide Receptor Improves Outcomes following Experimental Myocardial Infarction. Cell Metab 2018; 27:450-460.e6. [PMID: 29275960 DOI: 10.1016/j.cmet.2017.11.003] [Citation(s) in RCA: 62] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/02/2017] [Revised: 10/15/2017] [Accepted: 11/15/2017] [Indexed: 12/15/2022]
Abstract
Incretin hormones exert pleiotropic metabolic actions beyond the pancreas. Although the heart expresses both incretin receptors, the cardiac biology of GIP receptor (GIPR) action remains incompletely understood. Here we show that GIPR agonism did not impair the response to cardiac ischemia. In contrast, genetic elimination of the Gipr reduced myocardial infarction (MI)-induced ventricular injury and enhanced survival associated with reduced hormone sensitive lipase (HSL) phosphorylation; it also increased myocardial triacylglycerol (TAG) stores. Conversely, direct GIPR agonism in the isolated heart reduced myocardial TAG stores and increased fatty acid oxidation. The cardioprotective phenotype in Gipr-/- mice was partially reversed by pharmacological activation or genetic overexpression of HSL. Selective Gipr inactivation in cardiomyocytes phenocopied Gipr-/- mice, resulting in improved survival and reduced adverse remodeling following experimental MI. Hence, the cardiomyocyte GIPR regulates fatty acid metabolism and the adaptive response to ischemic cardiac injury. These findings have translational relevance for developing GIPR-based therapeutics.
Collapse
Affiliation(s)
- John R Ussher
- Lunenfeld Tanenbaum Research Institute, Mount Sinai Hospital, University of Toronto, Toronto, ON M5G 1X5, Canada; Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton, AB T6G 2H7, Canada
| | - Jonathan E Campbell
- Lunenfeld Tanenbaum Research Institute, Mount Sinai Hospital, University of Toronto, Toronto, ON M5G 1X5, Canada; Duke Molecular Physiology Institute, Duke University Medical Center, Durham, NC 27701, USA
| | - Erin E Mulvihill
- Lunenfeld Tanenbaum Research Institute, Mount Sinai Hospital, University of Toronto, Toronto, ON M5G 1X5, Canada
| | - Laurie L Baggio
- Lunenfeld Tanenbaum Research Institute, Mount Sinai Hospital, University of Toronto, Toronto, ON M5G 1X5, Canada
| | - Holly E Bates
- Lunenfeld Tanenbaum Research Institute, Mount Sinai Hospital, University of Toronto, Toronto, ON M5G 1X5, Canada
| | - Brent A McLean
- Lunenfeld Tanenbaum Research Institute, Mount Sinai Hospital, University of Toronto, Toronto, ON M5G 1X5, Canada
| | - Keshav Gopal
- Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton, AB T6G 2H7, Canada
| | - Megan Capozzi
- Duke Molecular Physiology Institute, Duke University Medical Center, Durham, NC 27701, USA
| | - Bernardo Yusta
- Lunenfeld Tanenbaum Research Institute, Mount Sinai Hospital, University of Toronto, Toronto, ON M5G 1X5, Canada
| | - Xiemin Cao
- Lunenfeld Tanenbaum Research Institute, Mount Sinai Hospital, University of Toronto, Toronto, ON M5G 1X5, Canada
| | - Safina Ali
- Lunenfeld Tanenbaum Research Institute, Mount Sinai Hospital, University of Toronto, Toronto, ON M5G 1X5, Canada
| | - Minsuk Kim
- Lunenfeld Tanenbaum Research Institute, Mount Sinai Hospital, University of Toronto, Toronto, ON M5G 1X5, Canada
| | - M Golam Kabir
- Lunenfeld Tanenbaum Research Institute, Mount Sinai Hospital, University of Toronto, Toronto, ON M5G 1X5, Canada
| | - Yutaka Seino
- Kansai Electric Power Hospital and Medical Research Institute, 2-1-7 Fukushima-ku, Osaka 553-0003, Japan
| | - Jinya Suzuki
- Faculty of Medical Sciences, University of Fukui, Fukui 910-1193, Japan
| | - Daniel J Drucker
- Lunenfeld Tanenbaum Research Institute, Mount Sinai Hospital, University of Toronto, Toronto, ON M5G 1X5, Canada.
| |
Collapse
|
198
|
Castellan RFP, Meloni M. Mechanisms and Therapeutic Targets of Cardiac Regeneration: Closing the Age Gap. Front Cardiovasc Med 2018; 5:7. [PMID: 29459901 PMCID: PMC5807373 DOI: 10.3389/fcvm.2018.00007] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2017] [Accepted: 01/17/2018] [Indexed: 12/12/2022] Open
Abstract
While a regenerative response is limited in the mammalian adult heart, it has been recently shown that the neonatal mammalian heart possesses a marked but transient capacity for regeneration after cardiac injury, including myocardial infarction. These findings evidence that the mammalian heart still retains a regenerative capacity and highlights the concept that the expression of distinct molecular switches (that activate or inhibit cellular mechanisms regulating tissue development and regeneration) vary during different stages of life, indicating that cardiac regeneration is an age-dependent process. Thus, understanding the mechanisms underpinning regeneration in the neonatal-infarcted heart is crucial to develop new treatments aimed at improving cardiovascular regeneration in the adult. The present review summarizes the current knowledge on the pathways and factors that are known to determine cardiac regeneration in the neonatal-infarcted heart. In particular, we will focus on the effects of microRNA manipulation in regulating cardiomyocyte proliferation and regeneration, as well as on the role of the Hippo signaling pathway and Meis1 in the regenerative response of the neonatal-infarcted heart. We will also briefly comment on the role of macrophages in scar formation of the adult-infarcted heart or their contribution for scar-free regeneration of the neonatal mouse heart after myocardial infarction. Although additional research is needed in order to identify other factors that regulate cardiovascular regeneration, these pathways represent potential therapeutic targets for rejuvenation of aging hearts and for improving regeneration of the adult-infarcted heart.
Collapse
Affiliation(s)
- Raphael F. P. Castellan
- British Heart Foundation and University of Edinburgh Centre for Cardiovascular Science, Queen’s Medical Research Institute, Edinburgh, United Kingdom
| | - Marco Meloni
- British Heart Foundation and University of Edinburgh Centre for Cardiovascular Science, Queen’s Medical Research Institute, Edinburgh, United Kingdom
| |
Collapse
|
199
|
Du X. Post-infarct cardiac injury, protection and repair: roles of non-cardiomyocyte multicellular and acellular components. SCIENCE CHINA-LIFE SCIENCES 2018; 61:266-276. [DOI: 10.1007/s11427-017-9223-x] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/26/2017] [Accepted: 10/23/2017] [Indexed: 02/06/2023]
|
200
|
Therapeutic effect of a novel Wnt pathway inhibitor on cardiac regeneration after myocardial infarction. Clin Sci (Lond) 2017; 131:2919-2932. [PMID: 29162747 DOI: 10.1042/cs20171256] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2017] [Revised: 11/10/2017] [Accepted: 11/20/2017] [Indexed: 12/30/2022]
Abstract
After myocardial infarction (MI), the heart is difficult to repair because of great loss of cardiomyoctyes and lack of cardiac regeneration. Novel drug candidates that aim at reducing pathological remodeling and stimulating cardiac regeneration are highly desirable. In the present study, we identified if and how a novel porcupine inhibitor CGX1321 influenced MI and cardiac regeneration. Permanent ligation of left anterior descending (LAD) coronary artery was performed in mice to induce MI injury. Cardiac function was measured by echocardiography, infarct size was examined by TTC staining. Fibrosis was evaluated with Masson's trichrome staining and vimentin staining. As a result, CGX1321 administration blocked the secretion of Wnt proteins, and inhibited both canonical and non-canonical Wnt signaling pathways. CGX1321 improved cardiac function, reduced myocardial infarct size, and fibrosis of post-MI hearts. CGX1321 significantly increased newly formed cardiomyocytes in infarct border zone of post-MI hearts, evidenced by the increased EdU+ cardiomyocytes. Meanwhile, CGX1321 increased Ki67+ and phosphohistone H3 (PH3+) cardiomyocytes in culture, indicating enhanced cardiomyocyte proliferation. The mRNA microarray showed that CGX1321 up-regulated cell cycle regulating genes such as Ccnb1 and Ccne1 CGX1321 did not alter YAP protein phosphorylation and nuclear translocation in cardiomyocytes. In conclusion, porcupine inhibitor CGX1321 reduces MI injury by limiting fibrosis and promoting regeneration. It promotes cardiomyocyte proliferation by stimulating cell cycle regulating genes with a Hippo/YAP-independent pathway.
Collapse
|