1
|
Chen X, Zhong X, Huang GN. Heart regeneration from the whole-organism perspective to single-cell resolution. NPJ Regen Med 2024; 9:34. [PMID: 39548113 PMCID: PMC11568173 DOI: 10.1038/s41536-024-00378-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2024] [Accepted: 10/29/2024] [Indexed: 11/17/2024] Open
Abstract
Cardiac regenerative potential in the animal kingdom displays striking divergence across ontogeny and phylogeny. Here we discuss several fundamental questions in heart regeneration and provide both a holistic view of heart regeneration in the organism as a whole, as well as a single-cell perspective on intercellular communication among diverse cardiac cell populations. We hope to provide valuable insights that advance our understanding of organ regeneration and future therapeutic strategies.
Collapse
Affiliation(s)
- Xiaoxin Chen
- Cardiovascular Research Institute and Department of Physiology, University of California, San Francisco, San Francisco, CA, USA
- Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California, San Francisco, San Francisco, CA, USA
- Bakar Aging Research Institute, University of California, San Francisco, San Francisco, CA, USA
| | - Xiaochen Zhong
- Cardiovascular Research Institute and Department of Physiology, University of California, San Francisco, San Francisco, CA, USA
- Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California, San Francisco, San Francisco, CA, USA
- Bakar Aging Research Institute, University of California, San Francisco, San Francisco, CA, USA
| | - Guo N Huang
- Cardiovascular Research Institute and Department of Physiology, University of California, San Francisco, San Francisco, CA, USA.
- Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California, San Francisco, San Francisco, CA, USA.
- Bakar Aging Research Institute, University of California, San Francisco, San Francisco, CA, USA.
| |
Collapse
|
2
|
Stairley RA, Trouten AM, Li S, Roddy PL, DeLeon-Pennell KY, Lee KH, Sucov HM, Liu C, Tao G. Anti-Ferroptotic Treatment Deteriorates Myocardial Infarction by Inhibiting Angiogenesis and Altering Immune Response. Antioxidants (Basel) 2024; 13:769. [PMID: 39061839 PMCID: PMC11273385 DOI: 10.3390/antiox13070769] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2024] [Revised: 06/16/2024] [Accepted: 06/20/2024] [Indexed: 07/28/2024] Open
Abstract
Mammalian cardiomyocytes have limited regenerative ability. Cardiac disease, such as congenital heart disease and myocardial infarction, causes an initial loss of cardiomyocytes through regulated cell death (RCD). Understanding the mechanisms that govern RCD in the injured myocardium is crucial for developing therapeutics to promote heart regeneration. We previously reported that ferroptosis, a non-apoptotic and iron-dependent form of RCD, is the main contributor to cardiomyocyte death in the injured heart. To investigate the mechanisms underlying the preference for ferroptosis in cardiomyocytes, we examined the effects of anti-ferroptotic reagents in infarcted mouse hearts. The results revealed that the anti-ferroptotic reagent did not improve neonatal heart regeneration, and further compromised the cardiac function of juvenile hearts. On the other hand, ferroptotic cardiomyocytes played a supportive role during wound healing by releasing pro-angiogenic factors. The inhibition of ferroptosis in the regenerating mouse heart altered the immune and angiogenic responses. Our study provides insights into the preference for ferroptosis over other types of RCD in stressed cardiomyocytes, and guidance for designing anti-cell-death therapies for treating heart disease.
Collapse
Affiliation(s)
- Rebecca A. Stairley
- Department of Regenerative Medicine and Cell Biology, Medical University of South Carolina, Charleston, SC 29425, USA; (R.A.S.); (A.M.T.); (S.L.); (P.L.R.); (H.M.S.)
| | - Allison M. Trouten
- Department of Regenerative Medicine and Cell Biology, Medical University of South Carolina, Charleston, SC 29425, USA; (R.A.S.); (A.M.T.); (S.L.); (P.L.R.); (H.M.S.)
| | - Shuang Li
- Department of Regenerative Medicine and Cell Biology, Medical University of South Carolina, Charleston, SC 29425, USA; (R.A.S.); (A.M.T.); (S.L.); (P.L.R.); (H.M.S.)
- Department of Pediatrics, Herman B Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Patrick L. Roddy
- Department of Regenerative Medicine and Cell Biology, Medical University of South Carolina, Charleston, SC 29425, USA; (R.A.S.); (A.M.T.); (S.L.); (P.L.R.); (H.M.S.)
| | - Kristine Y. DeLeon-Pennell
- Division of Cardiology, Department of Medicine, Medical University of South Carolina, Charleston, SC 29425, USA;
- Research Service, Ralph H. Johnson Veterans Affairs Medical Center, Charleston, SC 29401, USA
| | - Kyu-Ho Lee
- Department of Medicine Digestive Disease Research Core Center, Medical University of South Carolina, Charleston, SC 29425, USA;
| | - Henry M. Sucov
- Department of Regenerative Medicine and Cell Biology, Medical University of South Carolina, Charleston, SC 29425, USA; (R.A.S.); (A.M.T.); (S.L.); (P.L.R.); (H.M.S.)
- Division of Cardiology, Department of Medicine, Medical University of South Carolina, Charleston, SC 29425, USA;
| | - Chun Liu
- Department of Physiology, Medical College of Wisconsin, Milwaukee, WI 53226, USA;
| | - Ge Tao
- Department of Regenerative Medicine and Cell Biology, Medical University of South Carolina, Charleston, SC 29425, USA; (R.A.S.); (A.M.T.); (S.L.); (P.L.R.); (H.M.S.)
| |
Collapse
|
3
|
Javanshir E, Ebrahimi ZJ, Mirzohreh ST, Ghaffari S, Banisefid E, Alamdari NM, Roshanravan N. Disparity of gene expression in coronary artery disease: insights from MEIS1, HIRA, and Myocardin. Mol Biol Rep 2024; 51:712. [PMID: 38824221 DOI: 10.1007/s11033-024-09657-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2024] [Accepted: 05/20/2024] [Indexed: 06/03/2024]
Abstract
INTRODUCTION Coronary artery disease (CAD) in young adults can have devastating consequences. The cardiac developmental gene MEIS1 plays important roles in vascular networks and heart development. This gene effects on the regeneration capacity of the heart. Considering role of MEIS1 in cardiac tissue development and the progression of myocardial infarction this study investigated the expression levels of the MEIS1, HIRA, and Myocardin genes in premature CAD patients compared to healthy subjects and evaluated the relationships between these genes and possible inflammatory factors. METHODS AND RESULTS The study conducted a case-control design involving 35 CAD patients and 35 healthy individuals. Peripheral blood mononuclear cells (PBMCs) were collected, and gene expression analysis was performed using real-time PCR. Compared with control group, the number of PBMCs in the CAD group exhibited greater MEIS1 and HIRA gene expression, with fold changes of 2.45 and 3.6. The expression of MEIS1 exhibited a negative correlation with IL-10 (r= -0.312) expression and positive correlation with Interleukin (IL)-6 (r = 0.415) and tumor necrosis factor (TNF)-α (r = 0.534) gene expression. Moreover, there was an inverse correlation between the gene expression of HIRA and that of IL-10 (r= -0.326), and a positive correlation was revealed between the expression of this gene and that of the IL-6 (r = 0.453) and TNF-α (r = 0.572) genes. CONCLUSION This research demonstrated a disparity in expression levels of MEIS1, HIRA, and Myocardin, between CAD and healthy subjects. The results showed that, MEIS1 and HIRA play significant roles in regulating the synthesis of proinflammatory cytokines, namely, TNF-α and IL-6.
Collapse
Affiliation(s)
- Elnaz Javanshir
- Cardiovascular Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | | | | | - Samad Ghaffari
- Cardiovascular Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Erfan Banisefid
- Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran
| | | | - Neda Roshanravan
- Drug Applied Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.
| |
Collapse
|
4
|
Zhang YW, Pang X, Yang Y. Hydrogels containing KYNA promote angiogenesis and inhibit inflammation to improve the survival rate of multi-territory perforator flaps. Biomed Pharmacother 2024; 174:116454. [PMID: 38640710 DOI: 10.1016/j.biopha.2024.116454] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2023] [Revised: 03/08/2024] [Accepted: 03/15/2024] [Indexed: 04/21/2024] Open
Abstract
BACKGROUND A new spray adhesive (KYNA-PF127) was established through the combination of thermosensitive hydrogel (Pluronic F127) and KYNA, aimed to investigate the effect of KYNA-PF127 on multi-territory perforator flaps and its possible molecular mechanism. MATERIALS AND METHODS 36 SD male rats with 250-300 g were randomly divided into 3 groups (n = 12): control group, blank glue group and KYNA-PF127 group. KYNA-PF127 hydrogel was prepared and characterized for its morphology and properties using scanning electron microscopy. CCK-8 assay, scratch wound assay, transwell assay, tube formation assay and Ki67 staining were used to study the effect of KYNA-PF127 on the proliferation, migration, and tube formation of HUVECs. VEGF and FGF2 were measured by qPCR to evaluate the angiogenesis capacity of HUVECs in vitro. In vivo, the effect of each group on the survival area of the cross-zone perforator flap was evaluated, and angiogenesis was evaluated by HE and immunofluorescence (CD31 and MMP-9). The effect of inflammation on skin collagen fibers was assessed by Masson. Immunohistochemistry (SOD1, IL-1β, TNF-α) was used to evaluate the effects of oxidative stress and inflammatory factors on multi-territory flaps. RESULTS KYNA-PF127 has good sustained release and biocompatibility at 25% concentration. KYNA-PF127 promoted the proliferation, migration, and angiogenesis of HUVECs in vitro. In vivo, the survival area of multi-territory perforator flaps and angiogenic capability have increased after KYNA-PF127 intervention. KYNA-PF127 could effectively reduce the oxidative stress and inflammation of multi-territory perforator flaps. CONCLUSION KYNA-PF127 promotes angiogenesis through its antioxidant stress and anti-inflammatory effects, and shows potential clinical value in promoting the survival viability and drug delivery of multi-territory perforator flaps.
Collapse
Affiliation(s)
- Ya-Wei Zhang
- Department of Geriatric Surgery, The Second Xiangya Hospital of Central South University, Changsha, Hunan 410011, China
| | - Xiaoyang Pang
- Department of Orthopedics, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, Hunan 410008, China
| | - Yan Yang
- Department of Orthopedics, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, Hunan 410008, China.
| |
Collapse
|
5
|
Baccouche BM, Elde S, Wang H, Woo YJ. Structural, angiogenic, and immune responses influencing myocardial regeneration: a glimpse into the crucible. NPJ Regen Med 2024; 9:18. [PMID: 38688935 PMCID: PMC11061134 DOI: 10.1038/s41536-024-00357-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2023] [Accepted: 03/15/2024] [Indexed: 05/02/2024] Open
Abstract
Complete cardiac regeneration remains an elusive therapeutic goal. Although much attention has been focused on cardiomyocyte proliferation, especially in neonatal mammals, recent investigations have unearthed mechanisms by which non-cardiomyocytes, such as endothelial cells, fibroblasts, macrophages, and other immune cells, play critical roles in modulating the regenerative capacity of the injured heart. The degree to which each of these cell types influence cardiac regeneration, however, remains incompletely understood. This review highlights the roles of these non-cardiomyocytes and their respective contributions to cardiac regeneration, with emphasis on natural heart regeneration after cardiac injury during the neonatal period.
Collapse
Affiliation(s)
- Basil M Baccouche
- Stanford University Department of Cardiothoracic Surgery, Palo Alto, CA, USA
| | - Stefan Elde
- Stanford University Department of Cardiothoracic Surgery, Palo Alto, CA, USA
| | - Hanjay Wang
- Stanford University Department of Cardiothoracic Surgery, Palo Alto, CA, USA
| | - Y Joseph Woo
- Stanford University Department of Cardiothoracic Surgery, Palo Alto, CA, USA.
| |
Collapse
|
6
|
Lv J, Fu Z, Zheng H, Song Q. Global research trends and emerging opportunities for integrin adhesion complexes in cardiac repair: a scientometric analysis. Front Cardiovasc Med 2024; 11:1308763. [PMID: 38699584 PMCID: PMC11063371 DOI: 10.3389/fcvm.2024.1308763] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2023] [Accepted: 04/04/2024] [Indexed: 05/05/2024] Open
Abstract
Objective Cardiac regenerative medicine has gained significant attention in recent years, and integrins are known to play a critical role in mediating cardiac development and repair, especially after an injury from the myocardial infarction (MI). Given the extensive research history and interdisciplinary nature of this field, a quantitative retrospective analysis and visualization of related topics is necessary. Materials and methods We performed a scientometric analysis of published papers on cardiac integrin adhesion complexes (IACs), including analysis of annual publications, disciplinary evolution, keyword co-occurrence, and literature co-citation. Results A total of 2,664 publications were finally included in the past 20 years. The United States is the largest contributor to the study and is leading this area of research globally. The journal Circulation Research attracts the largest number of high-quality publications. The study of IACs in cardiac repair/regenerative therapies involves multiple disciplines, particularly in materials science and developmental biology. Keywords of research frontiers were represented by Tenasin-C (2019-2023) and inflammation (2020-2023). Conclusion Integrins are topics with ongoing enthusiasm in biological development and tissue regeneration. The rapidly emerging role of matricellular proteins and non-protein components of the extracellular matrix (ECM) in regulating matrix structure and function may be a further breakthrough point in the future; the emerging role of IACs and their downstream molecular signaling in cardiac repair are also of great interest, such as induction of cardiac proliferation, differentiation, maturation, and metabolism, fibroblast activation, and inflammatory modulation.
Collapse
Affiliation(s)
- Jiayu Lv
- Department of General Internal Medicine, Guang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
- China Academy of Chinese Medical Sciences, Beijing, China
| | - Zhenyue Fu
- Department of General Internal Medicine, Guang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
- College of Traditional Chinese Medicine, Beijing University of Traditional Chinese Medicine, Beijing, China
| | - Haoran Zheng
- Department of General Internal Medicine, Guang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
- College of Traditional Chinese Medicine, Beijing University of Traditional Chinese Medicine, Beijing, China
| | - Qingqiao Song
- Department of General Internal Medicine, Guang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| |
Collapse
|
7
|
Vanderslice EJ, Golding SGH, Jacot JG. Vascularization of PEGylated fibrin hydrogels increases the proliferation of human iPSC-cardiomyocytes. J Biomed Mater Res A 2024; 112:625-634. [PMID: 38155509 PMCID: PMC10922460 DOI: 10.1002/jbm.a.37662] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2023] [Revised: 12/13/2023] [Accepted: 12/15/2023] [Indexed: 12/30/2023]
Abstract
Studies have long sought to develop engineered heart tissue for the surgical correction of structural heart defects, as well as other applications and vascularization of this tissue has presented a challenge. Recent studies suggest that vascular cells and a vascular network may have regenerative effects on implanted cardiomyocytes (CM) and nearby heart tissue separate from perfusion of oxygen and nutrients. The goal of this study was to test whether vascular cells or a formed vascular network in a fibrin-based hydrogel would alter the proliferation of human iPSC-derived CM. First, vascular network formation in a slowly degrading PEGylated fibrin hydrogel was optimized by altering the cell ratio of human umbilical vein endothelial cells to human dermal fibroblasts, the inclusion of growth factors, and the total cell concentration. An endothelial to fibroblast ratio of 5:1 and a total cell concentration of 1.1 × 106 cells/mL without additional growth factors generated robust vascular networks while minimizing the number of cells required. Using this optimized system, human iPSC-derived CM were cultured on hydrogels without vascular cells, hydrogels with unorganized encapsulated vascular cells, or hydrogels with encapsulated vascular cells organized into networks for 7 days. CM proliferation and gene expression were assayed following 7 days of culture on the hydrogels. The presence of vascular cells in the hydrogel, whether unorganized or in vascular networks, significantly increased CM proliferation compared to an acellular hydrogel. Hydrogels with unorganized vascular cells resulted in lower CM maturity evidenced by decreased expression of cardiac troponin t (TNNT2), myosin light chain 7, and phospholamban compared to hydrogels without vascular cells and hydrogels with vascular networks. Altogether, this study details a robust method of forming rudimentary vascular networks in a fibrin-based hydrogel and shows that a hydrogel containing endothelial cells and fibroblasts can induce proliferation in adjacent CM, and these cells do not hinder CM gene expression when organized into a vascular network.
Collapse
Affiliation(s)
- Ethan J. Vanderslice
- Department of Bioengineering, University of Colorado Anschutz Medical Campus, Aurora, CO, USA 80045
| | - Staunton G. H. Golding
- Department of Bioengineering, University of Colorado Anschutz Medical Campus, Aurora, CO, USA 80045
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN, USA 37235
| | - Jeffrey G. Jacot
- Department of Bioengineering, University of Colorado Anschutz Medical Campus, Aurora, CO, USA 80045
- Department of Pediatrics, Children’s Hospital Colorado, Aurora, CO, USA 80045
| |
Collapse
|
8
|
Long H, Steimle JD, Grisanti Canozo FJ, Kim JH, Li X, Morikawa Y, Park M, Turaga D, Adachi I, Wythe JD, Samee MAH, Martin JF. Endothelial cells adopt a pro-reparative immune responsive signature during cardiac injury. Life Sci Alliance 2024; 7:e202201870. [PMID: 38012001 PMCID: PMC10681909 DOI: 10.26508/lsa.202201870] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2022] [Revised: 11/11/2023] [Accepted: 11/14/2023] [Indexed: 11/29/2023] Open
Abstract
Modulation of the heart's immune microenvironment is crucial for recovery after ischemic events such as myocardial infarction (MI). Endothelial cells (ECs) can have immune regulatory functions; however, interactions between ECs and the immune environment in the heart after MI remain poorly understood. We identified an EC-specific IFN responsive and immune regulatory gene signature in adult and pediatric heart failure (HF) tissues. Single-cell transcriptomic analysis of murine hearts subjected to MI uncovered an EC population (IFN-ECs) with immunologic gene signatures similar to those in human HF. IFN-ECs were enriched in regenerative-stage mouse hearts and expressed genes encoding immune responsive transcription factors (Irf7, Batf2, and Stat1). Single-cell chromatin accessibility studies revealed an enrichment of these TF motifs at IFN-EC signature genes. Expression of immune regulatory ligand genes by IFN-ECs suggests bidirectional signaling between IFN-ECs and macrophages in regenerative-stage hearts. Our data suggest that ECs may adopt immune regulatory signatures after cardiac injury to accompany the reparative response. The presence of these signatures in human HF and murine MI models suggests a potential role for EC-mediated immune regulation in responding to stress induced by acute injury in MI and chronic adverse remodeling in HF.
Collapse
Affiliation(s)
- Hali Long
- Interdepartmental Program in Integrative Molecular and Biomedical Sciences, Baylor College of Medicine, Houston, TX, USA
- Department of Integrative Physiology, Baylor College of Medicine, Houston, TX, USA
| | - Jeffrey D Steimle
- Department of Integrative Physiology, Baylor College of Medicine, Houston, TX, USA
| | | | - Jong Hwan Kim
- Department of Integrative Physiology, Baylor College of Medicine, Houston, TX, USA
- Cardiomyocyte Renewal Laboratory, The Texas Heart Institute, Houston, TX, USA
| | - Xiao Li
- Cardiomyocyte Renewal Laboratory, The Texas Heart Institute, Houston, TX, USA
| | - Yuka Morikawa
- Cardiomyocyte Renewal Laboratory, The Texas Heart Institute, Houston, TX, USA
| | - Minjun Park
- Department of Integrative Physiology, Baylor College of Medicine, Houston, TX, USA
| | - Diwakar Turaga
- Section of Critical Care Medicine, Department of Pediatrics, Baylor College of Medicine, Houston, TX, USA
| | - Iki Adachi
- Section of Cardiothoracic Surgery, Department of Surgery, Baylor College of Medicine, Houston, TX, USA
| | - Joshua D Wythe
- Department of Integrative Physiology, Baylor College of Medicine, Houston, TX, USA
- Cardiovascular Research Institute, Baylor College of Medicine, Houston, TX, USA
| | - Md Abul Hassan Samee
- Department of Integrative Physiology, Baylor College of Medicine, Houston, TX, USA
| | - James F Martin
- Interdepartmental Program in Integrative Molecular and Biomedical Sciences, Baylor College of Medicine, Houston, TX, USA
- Department of Integrative Physiology, Baylor College of Medicine, Houston, TX, USA
- Cardiomyocyte Renewal Laboratory, The Texas Heart Institute, Houston, TX, USA
- Cardiovascular Research Institute, Baylor College of Medicine, Houston, TX, USA
- Center for Organ Repair and Renewal, Baylor College of Medicine, Houston, TX, USA
| |
Collapse
|
9
|
Nguyen TM, Geng X, Wei Y, Ye L, Garry DJ, Zhang J. Single-cell RNA sequencing analysis identifies one subpopulation of endothelial cells that proliferates and another that undergoes the endothelial-mesenchymal transition in regenerating pig hearts. Front Bioeng Biotechnol 2024; 11:1257669. [PMID: 38288246 PMCID: PMC10823534 DOI: 10.3389/fbioe.2023.1257669] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2023] [Accepted: 12/04/2023] [Indexed: 01/31/2024] Open
Abstract
Background: In our previous work, we demonstrated that when newborn pigs undergo apical resection (AR) on postnatal day 1 (P1), the animals' hearts were completely recover from a myocardial infarction (MI) that occurs on postnatal day 28 (P28); single-nucleus RNA sequencing (snRNAseq) data suggested that this recovery was achieved by regeneration of pig cardiomyocyte subpopulations in response to MI. However, coronary vasculature also has a key role in promoting cardiac repair. Method: Thus, in this report, we used autoencoder algorithms to analyze snRNAseq data from endothelial cells (ECs) in the hearts of the same animals. Main results: Our results identified five EC clusters, three composed of vascular ECs (VEC1-3) and two containing lymphatic ECs (LEC1-2). Cells from VEC1 expressed elevated levels of each of five cell-cyclespecific markers (Aurora Kinase B [AURKB], Marker of Proliferation Ki-67 [MKI67], Inner Centromere Protein [INCENP], Survivin [BIRC5], and Borealin [CDCA8]), as well as a number of transcription factors that promote EC proliferation, while (VEC3 was enriched for genes that regulate intercellular junctions, participate in transforming growth factor β (TGFβ), bone morphogenic protein (BMP) signaling, and promote the endothelial mesenchymal transition (EndMT). The remaining VEC2 did not appear to participate directly in the angiogenic response to MI, but trajectory analyses indicated that it may serve as a reservoir for the generation of VEC1 and VEC3 ECs in response to MI. Notably, only the VEC3 cluster was more populous in regenerating (i.e., ARP1MIP28) than non-regenerating (i.e., MIP28) hearts during the 1-week period after MI induction, which suggests that further investigation of the VEC3 cluster could identify new targets for improving myocardial recovery after MI. Histological analysis of KI67 and EndMT marker PDGFRA demonstrated that while the expression of proliferation of endothelial cells was not significantly different, expression of EndMT markers was significantly higher among endothelial cells of ARP1MIP28 hearts compared to MIP28 hearts, which were consistent with snRNAseq analysis of clusters VEC1 and VEC3. Furthermore, upregulated secrete genes by VEC3 may promote cardiomyocyte proliferation via the Pi3k-Akt and ERBB signaling pathways, which directly contribute to cardiac muscle regeneration. Conclusion: In regenerative heart, endothelial cells may express EndMT markers, and this process could contribute to regeneration via a endothelial-cardiomyocyte crosstalk that supports cardiomyocyte proliferation.
Collapse
Affiliation(s)
- Thanh Minh Nguyen
- Department of Biomedical Engineering, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Xiaoxiao Geng
- Department of Biomedical Engineering, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Yuhua Wei
- Department of Biomedical Engineering, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Lei Ye
- Department of Biomedical Engineering, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Daniel J. Garry
- Department of Medicine, School of Medicine, University of Minnesota, Minneapolis, MN, United States
| | - Jianyi Zhang
- Department of Biomedical Engineering, University of Alabama at Birmingham, Birmingham, AL, United States
- Department of Medicine, Cardiovascular Diseases, University of Alabama at Birmingham, Birmingham, AL, United States
| |
Collapse
|
10
|
Beisaw A, Wu CC. Cardiomyocyte maturation and its reversal during cardiac regeneration. Dev Dyn 2024; 253:8-27. [PMID: 36502296 DOI: 10.1002/dvdy.557] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2022] [Revised: 12/03/2022] [Accepted: 12/03/2022] [Indexed: 12/14/2022] Open
Abstract
Cardiovascular disease is a leading cause of death worldwide. Due to the limited proliferative and regenerative capacity of adult cardiomyocytes, the lost myocardium is not replenished efficiently and is replaced by a fibrotic scar, which eventually leads to heart failure. Current therapies to cure or delay the progression of heart failure are limited; hence, there is a pressing need for regenerative approaches to support the failing heart. Cardiomyocytes undergo a series of transcriptional, structural, and metabolic changes after birth (collectively termed maturation), which is critical for their contractile function but limits the regenerative capacity of the heart. In regenerative organisms, cardiomyocytes revert from their terminally differentiated state into a less mature state (ie, dedifferentiation) to allow for proliferation and regeneration to occur. Importantly, stimulating adult cardiomyocyte dedifferentiation has been shown to promote morphological and functional improvement after myocardial infarction, further highlighting the importance of cardiomyocyte dedifferentiation in heart regeneration. Here, we review several hallmarks of cardiomyocyte maturation, and summarize how their reversal facilitates cardiomyocyte proliferation and heart regeneration. A detailed understanding of how cardiomyocyte dedifferentiation is regulated will provide insights into therapeutic options to promote cardiomyocyte de-maturation and proliferation, and ultimately heart regeneration in mammals.
Collapse
Affiliation(s)
- Arica Beisaw
- Institute of Experimental Cardiology, Heidelberg University, Heidelberg, Germany
| | - Chi-Chung Wu
- European Center for Angioscience (ECAS), Medical Faculty Mannheim, Heidelberg University, Heidelberg, Germany
| |
Collapse
|
11
|
Berkeley B, Tang MNH, Brittan M. Mechanisms regulating vascular and lymphatic regeneration in the heart after myocardial infarction. J Pathol 2023; 260:666-678. [PMID: 37272582 PMCID: PMC10953458 DOI: 10.1002/path.6093] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2023] [Revised: 04/14/2023] [Accepted: 04/27/2023] [Indexed: 06/06/2023]
Abstract
Myocardial infarction, caused by a thrombus or coronary vascular occlusion, leads to irreversible ischaemic injury. Advances in early reperfusion strategies have significantly reduced short-term mortality after myocardial infarction. However, survivors have an increased risk of developing heart failure, which confers a high risk of death at 1 year. The capacity of the injured neonatal mammalian heart to regenerate has stimulated extensive research into whether recapitulation of developmental regeneration programmes may be beneficial in adult cardiovascular disease. Restoration of functional blood and lymphatic vascular networks in the infarct and border regions via neovascularisation and lymphangiogenesis, respectively, is a key requirement to facilitate myocardial regeneration. An improved understanding of the endogenous mechanisms regulating coronary vascular and lymphatic expansion and function in development and in adult patients after myocardial infarction may inform future therapeutic strategies and improve translation from pre-clinical studies. In this review, we explore the underpinning research and key findings in the field of cardiovascular regeneration, with a focus on neovascularisation and lymphangiogenesis, and discuss the outcomes of therapeutic strategies employed to date. © 2023 The Authors. The Journal of Pathology published by John Wiley & Sons Ltd on behalf of The Pathological Society of Great Britain and Ireland.
Collapse
Affiliation(s)
- Bronwyn Berkeley
- Centre for Cardiovascular Science, The Queen's Medical Research InstituteUniversity of EdinburghEdinburghUK
| | - Michelle Nga Huen Tang
- Centre for Cardiovascular Science, The Queen's Medical Research InstituteUniversity of EdinburghEdinburghUK
| | - Mairi Brittan
- Centre for Cardiovascular Science, The Queen's Medical Research InstituteUniversity of EdinburghEdinburghUK
| |
Collapse
|
12
|
Li Z, Solomonidis EG, Berkeley B, Tang MNH, Stewart KR, Perez-Vicencio D, McCracken IR, Spiroski AM, Gray GA, Barton AK, Sellers SL, Riley PR, Baker AH, Brittan M. Multi-species meta-analysis identifies transcriptional signatures associated with cardiac endothelial responses in the ischaemic heart. Cardiovasc Res 2023; 119:136-154. [PMID: 36082978 PMCID: PMC10022865 DOI: 10.1093/cvr/cvac151] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/23/2022] [Revised: 07/04/2022] [Accepted: 08/10/2022] [Indexed: 11/12/2022] Open
Abstract
AIM Myocardial infarction remains the leading cause of heart failure. The adult human heart lacks the capacity to undergo endogenous regeneration. New blood vessel growth is integral to regenerative medicine necessitating a comprehensive understanding of the pathways that regulate vascular regeneration. We sought to define the transcriptomic dynamics of coronary endothelial cells following ischaemic injuries in the developing and adult mouse and human heart and to identify new mechanistic insights and targets for cardiovascular regeneration. METHODS AND RESULTS We carried out a comprehensive meta-analysis of integrated single-cell RNA-sequencing data of coronary vascular endothelial cells from the developing and adult mouse and human heart spanning healthy and acute and chronic ischaemic cardiac disease. We identified species-conserved gene regulatory pathways aligned to endogenous neovascularization. We annotated injury-associated temporal shifts of the endothelial transcriptome and validated four genes: VEGF-C, KLF4, EGR1, and ZFP36. Moreover, we showed that ZFP36 regulates human coronary endothelial cell proliferation and defined that VEGF-C administration in vivo enhances clonal expansion of the cardiac vasculature post-myocardial infarction. Finally, we constructed a coronary endothelial cell meta-atlas, CrescENDO, to empower future in-depth research to target pathways associated with coronary neovascularization. CONCLUSION We present a high-resolution single-cell meta-atlas of healthy and injured coronary endothelial cells in the mouse and human heart, revealing a suite of novel targets with great potential to promote vascular regeneration, and providing a rich resource for therapeutic development.
Collapse
Affiliation(s)
- Ziwen Li
- Centre for Cardiovascular Science, The Queen’s Medical Research Institute, University of Edinburgh, Edinburgh EH16 4TJ, UK
| | - Emmanouil G Solomonidis
- Centre for Cardiovascular Science, The Queen’s Medical Research Institute, University of Edinburgh, Edinburgh EH16 4TJ, UK
| | - Bronwyn Berkeley
- Centre for Cardiovascular Science, The Queen’s Medical Research Institute, University of Edinburgh, Edinburgh EH16 4TJ, UK
| | - Michelle Nga Huen Tang
- Centre for Cardiovascular Science, The Queen’s Medical Research Institute, University of Edinburgh, Edinburgh EH16 4TJ, UK
| | - Katherine Ross Stewart
- Centre for Cardiovascular Science, The Queen’s Medical Research Institute, University of Edinburgh, Edinburgh EH16 4TJ, UK
| | - Daniel Perez-Vicencio
- Centre for Cardiovascular Science, The Queen’s Medical Research Institute, University of Edinburgh, Edinburgh EH16 4TJ, UK
| | - Ian R McCracken
- Centre for Cardiovascular Science, The Queen’s Medical Research Institute, University of Edinburgh, Edinburgh EH16 4TJ, UK
| | - Ana-Mishel Spiroski
- Centre for Cardiovascular Science, The Queen’s Medical Research Institute, University of Edinburgh, Edinburgh EH16 4TJ, UK
| | - Gillian A Gray
- Centre for Cardiovascular Science, The Queen’s Medical Research Institute, University of Edinburgh, Edinburgh EH16 4TJ, UK
| | - Anna K Barton
- Centre for Cardiovascular Science, The Queen’s Medical Research Institute, University of Edinburgh, Edinburgh EH16 4TJ, UK
| | - Stephanie L Sellers
- Division of Cardiology, Centre for Heart Lung Innovation, Providence Research, University of British Columbia, Vancouver, Canada
| | - Paul R Riley
- Department of Physiology, Anatomy and Genetics, University of Oxford, South Parks Road, Oxford OX1 3PT, UK
| | - Andrew H Baker
- Centre for Cardiovascular Science, The Queen’s Medical Research Institute, University of Edinburgh, Edinburgh EH16 4TJ, UK
| | | |
Collapse
|
13
|
Qu X, Li J, Liu L, Zhang J, Hua Y, Suzuki K, Harada A, Ishida M, Yoshida N, Okuzaki D, Sakai Y, Sawa Y, Miyagawa S. ONO-1301 enhances post-transplantation survival of human induced pluripotent stem cell-derived cardiac tissue sheet by promoting angiogenesis. J Heart Lung Transplant 2023; 42:716-729. [PMID: 36964085 DOI: 10.1016/j.healun.2023.01.018] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2022] [Revised: 01/11/2023] [Accepted: 01/30/2023] [Indexed: 02/11/2023] Open
Abstract
BACKGROUND Transplanting human induced pluripotent stem cell-derived cardiomyocyte (hiPSC-CM) tissue sheets effectively treat ischemic cardiomyopathy. Cardiac functional recovery relies on graft survival in which angiogenesis played an important part. ONO-1301 is a synthetic prostacyclin analog with proangiogenic effects. We hypothesized that transplantation of hiPSC-CM tissue sheets with slow-release ONO-1301 scaffold could promote hostgraft angiogenesis, enhance tissue survival and therapeutic effect. METHODS We developed hiPSC-CM tissue sheets with ONO-1301 slow-release scaffold and evaluated their morphology, gene expression, and effects on angiogenesis. Three tissue sheet layers were transplanted into a rat myocardial infarction (MI) model. Left ventricular ejection fraction, gene expression in the MI border zone, and angiogenesis effects were investigated 4 weeks after transplantation. RESULTS In vitro assessment confirmed the slow-release of ONO-1301, and its pro-angiogenesis effects. In addition, in vivo data demonstrated that ONO-1301 administration positively correlated with graft survival. Cardiac tissue as thick as ∼900 μm was retained in the ONO (+) treated group. Additionally, left ventricular ejection fraction of the ONO (+) group was significantly enhanced, compared to ONO (-) group. The ONO (+) group also showed significantly improved interstitial fibrosis, higher capillary density, increased number of mature blood vessels, along with an enhanced supply of oxygen, and nutrients. CONCLUSIONS Slow-release ONO-1301 scaffold provided an efficient delivery method for thick hiPSC-CM tissue. ONO-1301 promotes angiogenesis between the host and graft and improves nutritional and oxygen supply, thereby enhancing the survival of transplanted cells, effectively improving ejection fraction, and therapeutic effects.
Collapse
Affiliation(s)
- Xiang Qu
- Department of Cardiovascular Surgery, Osaka University Graduate School of Medicine, Suita, Osaka, Japan
| | - Junjun Li
- Department of Cardiovascular Surgery, Osaka University Graduate School of Medicine, Suita, Osaka, Japan
| | - Li Liu
- Department of Cardiovascular Surgery, Osaka University Graduate School of Medicine, Suita, Osaka, Japan
| | - Jingbo Zhang
- Department of Cardiovascular Surgery, Osaka University Graduate School of Medicine, Suita, Osaka, Japan
| | - Ying Hua
- Department of Cardiovascular Surgery, Osaka University Graduate School of Medicine, Suita, Osaka, Japan
| | - Kota Suzuki
- Department of Cardiovascular Surgery, Osaka University Graduate School of Medicine, Suita, Osaka, Japan
| | - Akima Harada
- Department of Cardiovascular Surgery, Osaka University Graduate School of Medicine, Suita, Osaka, Japan
| | - Masako Ishida
- Department of Cardiovascular Surgery, Osaka University Graduate School of Medicine, Suita, Osaka, Japan
| | - Noriko Yoshida
- Department of Cardiovascular Surgery, Osaka University Graduate School of Medicine, Suita, Osaka, Japan
| | - Daisuke Okuzaki
- Laboratory of Human Immunology (Single Cell Genomics), WPI Immunology Research Center, Osaka University, Osaka, Japan; Genome Information Research Center, Research Institute for Microbial Diseases, Osaka University, Osaka, Japan
| | - Yoshiki Sakai
- Department of Cardiovascular Surgery, Osaka University Graduate School of Medicine, Suita, Osaka, Japan
| | - Yoshiki Sawa
- Department of Cardiovascular Surgery, Osaka University Graduate School of Medicine, Suita, Osaka, Japan
| | - Shigeru Miyagawa
- Department of Cardiovascular Surgery, Osaka University Graduate School of Medicine, Suita, Osaka, Japan.
| |
Collapse
|
14
|
Extracellular Matrix-Based Approaches in Cardiac Regeneration: Challenges and Opportunities. Int J Mol Sci 2022; 23:ijms232415783. [PMID: 36555424 PMCID: PMC9779713 DOI: 10.3390/ijms232415783] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2022] [Revised: 12/02/2022] [Accepted: 12/07/2022] [Indexed: 12/14/2022] Open
Abstract
Cardiac development is characterized by the active proliferation of different cardiac cell types, in particular cardiomyocytes and endothelial cells, that eventually build the beating heart. In mammals, these cells lose their regenerative potential early after birth, representing a major obstacle to our current capacity to restore the myocardial structure and function after an injury. Increasing evidence indicates that the cardiac extracellular matrix (ECM) actively regulates and orchestrates the proliferation, differentiation, and migration of cardiac cells within the heart, and that any change in either the composition of the ECM or its mechanical properties ultimately affect the behavior of these cells throughout one's life. Thus, understanding the role of ECMs' proteins and related signaling pathways on cardiac cell proliferation is essential to develop effective strategies fostering the regeneration of a damaged heart. This review provides an overview of the components of the ECM and its mechanical properties, whose function in cardiac regeneration has been elucidated, with a major focus on the strengths and weaknesses of the experimental models so far exploited to demonstrate the actual pro-regenerative capacity of the components of the ECM and to translate this knowledge into new therapies.
Collapse
|
15
|
Zhang D, Ning J, Ramprasath T, Yu C, Zheng X, Song P, Xie Z, Zou MH. Kynurenine promotes neonatal heart regeneration by stimulating cardiomyocyte proliferation and cardiac angiogenesis. Nat Commun 2022; 13:6371. [PMID: 36289221 PMCID: PMC9606021 DOI: 10.1038/s41467-022-33734-7] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2021] [Accepted: 09/29/2022] [Indexed: 12/25/2022] Open
Abstract
Indoleamine 2,3 dioxygenase-1 (IDO1) catalyzes tryptophan-kynurenine metabolism in many inflammatory and cancer diseases. Of note, acute inflammation that occurs immediately after heart injury is essential for neonatal cardiomyocyte proliferation and heart regeneration. However, the IDO1-catalyzed tryptophan metabolism during heart regeneration is largely unexplored. Here, we find that apical neonatal mouse heart resection surgery led to rapid and consistent increases in cardiac IDO1 expression and kynurenine accumulation. Cardiac deletion of Ido1 gene or chemical inhibition of IDO1 impairs heart regeneration. Mechanistically, elevated kynurenine triggers cardiomyocyte proliferation by activating the cytoplasmic aryl hydrocarbon receptor-SRC-YAP/ERK pathway. In addition, cardiomyocyte-derived kynurenine transports to endothelial cells and stimulates cardiac angiogenesis by promoting aryl hydrocarbon receptor nuclear translocation and enhancing vascular endothelial growth factor A expression. Notably, Ahr deletion prevents indoleamine 2,3 dioxygenase -kynurenine-associated heart regeneration. In summary, increasing indoleamine 2,3 dioxygenase-derived kynurenine level promotes cardiac regeneration by functioning as an endogenous regulator of cardiomyocyte proliferation and cardiac angiogenesis.
Collapse
Affiliation(s)
- Donghong Zhang
- Center for Molecular and Translational Medicine, Georgia State University, 157 Decatur Street North East, Atlanta, GA, 30303, USA
| | - Jinfeng Ning
- Center for Molecular and Translational Medicine, Georgia State University, 157 Decatur Street North East, Atlanta, GA, 30303, USA
| | - Tharmarajan Ramprasath
- Center for Molecular and Translational Medicine, Georgia State University, 157 Decatur Street North East, Atlanta, GA, 30303, USA
| | - Changjiang Yu
- Center for Molecular and Translational Medicine, Georgia State University, 157 Decatur Street North East, Atlanta, GA, 30303, USA
| | - Xiaoxu Zheng
- Center for Molecular and Translational Medicine, Georgia State University, 157 Decatur Street North East, Atlanta, GA, 30303, USA
| | - Ping Song
- Center for Molecular and Translational Medicine, Georgia State University, 157 Decatur Street North East, Atlanta, GA, 30303, USA
| | - Zhonglin Xie
- Center for Molecular and Translational Medicine, Georgia State University, 157 Decatur Street North East, Atlanta, GA, 30303, USA
| | - Ming-Hui Zou
- Center for Molecular and Translational Medicine, Georgia State University, 157 Decatur Street North East, Atlanta, GA, 30303, USA.
| |
Collapse
|
16
|
Singh S, Prakash S, Gupta SK. Angiogenesis: A critical determinant for cardiac regeneration. MOLECULAR THERAPY. NUCLEIC ACIDS 2022; 29:88-89. [PMID: 35795481 PMCID: PMC9249573 DOI: 10.1016/j.omtn.2022.06.007] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/01/2023]
Affiliation(s)
- Sandhya Singh
- Pharmacology Division, CSIR-Central Drug Research Institute, Sector 10, Jankipuram Extension, Lucknow 226031, India
| | - Shakti Prakash
- Pharmacology Division, CSIR-Central Drug Research Institute, Sector 10, Jankipuram Extension, Lucknow 226031, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Shashi Kumar Gupta
- Pharmacology Division, CSIR-Central Drug Research Institute, Sector 10, Jankipuram Extension, Lucknow 226031, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| |
Collapse
|
17
|
Long X, Qiu Z, Li C, Wang Y, Li J, Zhao R, Rong J, Gu N, Yuan J, Ge J, Shi B. CircERBB2IP promotes post-infarction revascularization via the miR-145a-5p/Smad5 axis. MOLECULAR THERAPY - NUCLEIC ACIDS 2022; 28:573-586. [PMID: 35592503 PMCID: PMC9096260 DOI: 10.1016/j.omtn.2022.04.006] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/20/2021] [Accepted: 04/15/2022] [Indexed: 10/27/2022]
|
18
|
Zhou H, Zhang F, Wu Y, Liu H, Duan R, Liu Y, Wang Y, He X, Zhang Y, Ma X, Guan Y, Liu Y, Liang D, Zhou L, Chen Y. LRP5 regulates cardiomyocyte proliferation and neonatal heart regeneration by the AKT/P21 pathway. J Cell Mol Med 2022; 26:2981-2994. [PMID: 35429093 PMCID: PMC9097834 DOI: 10.1111/jcmm.17311] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2021] [Revised: 03/02/2022] [Accepted: 03/16/2022] [Indexed: 12/22/2022] Open
Affiliation(s)
- Huixing Zhou
- Department of Cardiology Shanghai East Hospital Tongji University School of Medicine Shanghai China
- Key Laboratory of Arrhythmias of the Ministry of Education of China Shanghai East Hospital Tongji University School of Medicine Shanghai China
| | - Fulei Zhang
- Department of Cardiology Shanghai East Hospital Tongji University School of Medicine Shanghai China
- Key Laboratory of Arrhythmias of the Ministry of Education of China Shanghai East Hospital Tongji University School of Medicine Shanghai China
| | - Yahan Wu
- Department of Cardiology Shanghai East Hospital Tongji University School of Medicine Shanghai China
- Key Laboratory of Arrhythmias of the Ministry of Education of China Shanghai East Hospital Tongji University School of Medicine Shanghai China
| | - Hongyu Liu
- Department of Cardiology Shanghai East Hospital Tongji University School of Medicine Shanghai China
- Key Laboratory of Arrhythmias of the Ministry of Education of China Shanghai East Hospital Tongji University School of Medicine Shanghai China
| | - Ran Duan
- Department of Cardiology Shanghai East Hospital Tongji University School of Medicine Shanghai China
- Key Laboratory of Arrhythmias of the Ministry of Education of China Shanghai East Hospital Tongji University School of Medicine Shanghai China
| | - Yuanyuan Liu
- Key Laboratory of Arrhythmias of the Ministry of Education of China Shanghai East Hospital Tongji University School of Medicine Shanghai China
- Jinzhou Medical University Liaoning Jinzhou China
| | - Yan Wang
- Key Laboratory of Arrhythmias of the Ministry of Education of China Shanghai East Hospital Tongji University School of Medicine Shanghai China
- Jinzhou Medical University Liaoning Jinzhou China
| | - Xiaoyu He
- Department of Cardiology Shanghai East Hospital Tongji University School of Medicine Shanghai China
- Key Laboratory of Arrhythmias of the Ministry of Education of China Shanghai East Hospital Tongji University School of Medicine Shanghai China
| | - Yuemei Zhang
- Department of Cardiology Shanghai East Hospital Tongji University School of Medicine Shanghai China
- Key Laboratory of Arrhythmias of the Ministry of Education of China Shanghai East Hospital Tongji University School of Medicine Shanghai China
| | - Xiue Ma
- Department of Cardiology Shanghai East Hospital Tongji University School of Medicine Shanghai China
- Key Laboratory of Arrhythmias of the Ministry of Education of China Shanghai East Hospital Tongji University School of Medicine Shanghai China
| | - Yi Guan
- Department of Cardiology Shanghai East Hospital Tongji University School of Medicine Shanghai China
- Key Laboratory of Arrhythmias of the Ministry of Education of China Shanghai East Hospital Tongji University School of Medicine Shanghai China
| | - Yi Liu
- Department of Cardiology Shanghai East Hospital Tongji University School of Medicine Shanghai China
- Key Laboratory of Arrhythmias of the Ministry of Education of China Shanghai East Hospital Tongji University School of Medicine Shanghai China
| | - Dandan Liang
- Department of Cardiology Shanghai East Hospital Tongji University School of Medicine Shanghai China
- Key Laboratory of Arrhythmias of the Ministry of Education of China Shanghai East Hospital Tongji University School of Medicine Shanghai China
- Research Units of Origin and Regulation of Heart Rhythm Chinese Academy of Medical Sciences Shanghai China
| | - Liping Zhou
- Department of Cardiology Shanghai East Hospital Tongji University School of Medicine Shanghai China
- Key Laboratory of Arrhythmias of the Ministry of Education of China Shanghai East Hospital Tongji University School of Medicine Shanghai China
| | - Yi‐Han Chen
- Department of Cardiology Shanghai East Hospital Tongji University School of Medicine Shanghai China
- Key Laboratory of Arrhythmias of the Ministry of Education of China Shanghai East Hospital Tongji University School of Medicine Shanghai China
- Research Units of Origin and Regulation of Heart Rhythm Chinese Academy of Medical Sciences Shanghai China
- Department of Pathology and Pathophysiology Tongji University School of Medicine Shanghai China
| |
Collapse
|
19
|
Spadaccio C, Nenna A, Rose D, Piccirillo F, Nusca A, Grigioni F, Chello M, Vlahakes GJ. The Role of Angiogenesis and Arteriogenesisin Myocardial Infarction and Coronary Revascularization. J Cardiovasc Transl Res 2022; 15:1024-1048. [PMID: 35357670 DOI: 10.1007/s12265-022-10241-0] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/25/2021] [Accepted: 03/18/2022] [Indexed: 12/25/2022]
Abstract
Surgical myocardial revascularization is associated with long-term survival benefit in patients with multivessel coronary artery disease. However, the exact biological mechanisms underlying the clinical benefits of myocardial revascularization have not been elucidated yet. Angiogenesis and arteriogenesis biologically leading to vascular collateralization are considered one of the endogenous mechanisms to preserve myocardial viability during ischemia, and the presence of coronary collateralization has been regarded as one of the predictors of long-term survival in patients with coronary artery disease (CAD). Some experimental studies and indirect clinical evidence on chronic CAD confirmed an angiogenetic response induced by myocardial revascularization and suggested that revascularization procedures could constitute an angiogenetic trigger per se. In this review, the clinical and basic science evidence regarding arteriogenesis and angiogenesis in both CAD and coronary revascularization is analyzed with the aim to better elucidate their significance in the clinical arena and potential therapeutic use.
Collapse
Affiliation(s)
- Cristiano Spadaccio
- Cardiac Surgery, Massachusetts General Hospital & Harvard Medical School, Boston, USA. .,Cardiac Surgery, Golden Jubilee National Hospital & University of Glasgow, Glasgow, UK.
| | - Antonio Nenna
- Cardiac Surgery, Università Campus Bio-Medico di Roma, Rome, Italy
| | - David Rose
- Cardiac Surgery, Lancashire Cardiac Centre, Blackpool Victoria Hospital, Blackpool, UK
| | | | | | | | - Massimo Chello
- Cardiac Surgery, Università Campus Bio-Medico di Roma, Rome, Italy
| | - Gus J Vlahakes
- Cardiac Surgery, Massachusetts General Hospital & Harvard Medical School, Boston, USA
| |
Collapse
|
20
|
Hasmad HN, Bt Hj Idrus R, Sulaiman N, Lokanathan Y. Electrospun Fiber-Coated Human Amniotic Membrane: A Potential Angioinductive Scaffold for Ischemic Tissue Repair. Int J Mol Sci 2022; 23:ijms23031743. [PMID: 35163664 PMCID: PMC8836161 DOI: 10.3390/ijms23031743] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2021] [Revised: 01/24/2022] [Accepted: 01/27/2022] [Indexed: 12/10/2022] Open
Abstract
Cardiac patch implantation helps maximize the paracrine function of grafted cells and serves as a reservoir of soluble proangiogenic factors required for the neovascularization of infarcted hearts. We have previously fabricated a cardiac patch, EF-HAM, composed of a human amniotic membrane (HAM) coated with aligned PLGA electrospun fibers (EF). In this study, we aimed to evaluate the biocompatibility and angiogenic effects of EF-HAM scaffolds with varying fiber thicknesses on the paracrine behavior of skeletal muscle cells (SkM). Conditioned media (CM) obtained from SkM-seeded HAM and EF-HAM scaffolds were subjected to multiplex analysis of angiogenic factors and tested on HUVECs for endothelial cell viability, migration, and tube formation analyses. All three different groups of EF-HAM scaffolds demonstrated excellent biocompatibility with SkM. CM derived from SkM-seeded EF-HAM 7 min scaffolds contained significantly elevated levels of proangiogenic factors, including angiopoietin-1, IL-8, and VEGF-C compared to plain CM, which was obtained from SkM cultured on the plain surface. CM obtained from all SkM-seeded EF-HAM scaffolds significantly increased the viability of HUVECs compared to plain CM after five days of culture. However, only EF-HAM 7 min CM induced a higher migration capacity in HUVECs and formed a longer and more elaborate capillary-like network on Matrigel compared with plain CM. Surface roughness and wettability of EF-HAM 7 min scaffolds might have influenced the proportion of skeletal myoblasts and fibroblasts growing on the scaffolds and subsequently potentiated the angiogenic paracrine function of SkM. This study demonstrated the angioinductive properties of EF-HAM composite scaffold and its potential applications in the repair and regeneration of ischemic tissues.
Collapse
|
21
|
Naqvi N, Iismaa SE, Graham RM, Husain A. Mechanism-Based Cardiac Regeneration Strategies in Mammals. Front Cell Dev Biol 2021; 9:747842. [PMID: 34708043 PMCID: PMC8542766 DOI: 10.3389/fcell.2021.747842] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2021] [Accepted: 09/06/2021] [Indexed: 12/11/2022] Open
Abstract
Heart failure in adults is a leading cause of morbidity and mortality worldwide. It can arise from a variety of diseases, with most resulting in a loss of cardiomyocytes that cannot be replaced due to their inability to replicate, as well as to a lack of resident cardiomyocyte progenitor cells in the adult heart. Identifying and exploiting mechanisms underlying loss of developmental cardiomyocyte replicative capacity has proved to be useful in developing therapeutics to effect adult cardiac regeneration. Of course, effective regeneration of myocardium after injury requires not just expansion of cardiomyocytes, but also neovascularization to allow appropriate perfusion and resolution of injury-induced inflammation and interstitial fibrosis, but also reversal of adverse left ventricular remodeling. In addition to overcoming these challenges, a regenerative therapy needs to be safe and easily translatable. Failure to address these critical issues will delay the translation of regenerative approaches. This review critically analyzes current regenerative approaches while also providing a framework for future experimental studies aimed at enhancing success in regenerating the injured heart.
Collapse
Affiliation(s)
- Nawazish Naqvi
- Division of Cardiology, Department of Medicine, Emory University School of Medicine, Atlanta, GA, United States
| | - Siiri E Iismaa
- Victor Chang Cardiac Research Institute, Darlinghurst, NSW, Australia
| | - Robert M Graham
- Victor Chang Cardiac Research Institute, Darlinghurst, NSW, Australia
| | - Ahsan Husain
- Division of Cardiology, Department of Medicine, Emory University School of Medicine, Atlanta, GA, United States
| |
Collapse
|
22
|
Bongiovanni C, Sacchi F, Da Pra S, Pantano E, Miano C, Morelli MB, D'Uva G. Reawakening the Intrinsic Cardiac Regenerative Potential: Molecular Strategies to Boost Dedifferentiation and Proliferation of Endogenous Cardiomyocytes. Front Cardiovasc Med 2021; 8:750604. [PMID: 34692797 PMCID: PMC8531484 DOI: 10.3389/fcvm.2021.750604] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2021] [Accepted: 09/13/2021] [Indexed: 12/27/2022] Open
Abstract
Despite considerable efforts carried out to develop stem/progenitor cell-based technologies aiming at replacing and restoring the cardiac tissue following severe damages, thus far no strategies based on adult stem cell transplantation have been demonstrated to efficiently generate new cardiac muscle cells. Intriguingly, dedifferentiation, and proliferation of pre-existing cardiomyocytes and not stem cell differentiation represent the preponderant cellular mechanism by which lower vertebrates spontaneously regenerate the injured heart. Mammals can also regenerate their heart up to the early neonatal period, even in this case by activating the proliferation of endogenous cardiomyocytes. However, the mammalian cardiac regenerative potential is dramatically reduced soon after birth, when most cardiomyocytes exit from the cell cycle, undergo further maturation, and continue to grow in size. Although a slow rate of cardiomyocyte turnover has also been documented in adult mammals, both in mice and humans, this is not enough to sustain a robust regenerative process. Nevertheless, these remarkable findings opened the door to a branch of novel regenerative approaches aiming at reactivating the endogenous cardiac regenerative potential by triggering a partial dedifferentiation process and cell cycle re-entry in endogenous cardiomyocytes. Several adaptations from intrauterine to extrauterine life starting at birth and continuing in the immediate neonatal period concur to the loss of the mammalian cardiac regenerative ability. A wide range of systemic and microenvironmental factors or cell-intrinsic molecular players proved to regulate cardiomyocyte proliferation and their manipulation has been explored as a therapeutic strategy to boost cardiac function after injuries. We here review the scientific knowledge gained thus far in this novel and flourishing field of research, elucidating the key biological and molecular mechanisms whose modulation may represent a viable approach for regenerating the human damaged myocardium.
Collapse
Affiliation(s)
- Chiara Bongiovanni
- Department of Experimental, Diagnostic and Specialty Medicine (DIMES), University of Bologna, Bologna, Italy.,Centre for Applied Biomedical Research (CRBA), University of Bologna, Bologna, Italy.,National Laboratory of Molecular Biology and Stem Cell Engineering, National Institute of Biostructures and Biosystems (INBB), Bologna, Italy
| | - Francesca Sacchi
- National Laboratory of Molecular Biology and Stem Cell Engineering, National Institute of Biostructures and Biosystems (INBB), Bologna, Italy
| | - Silvia Da Pra
- Department of Experimental, Diagnostic and Specialty Medicine (DIMES), University of Bologna, Bologna, Italy.,Centre for Applied Biomedical Research (CRBA), University of Bologna, Bologna, Italy
| | - Elvira Pantano
- Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) MultiMedica, Milan, Italy
| | - Carmen Miano
- National Laboratory of Molecular Biology and Stem Cell Engineering, National Institute of Biostructures and Biosystems (INBB), Bologna, Italy
| | - Marco Bruno Morelli
- Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) MultiMedica, Milan, Italy
| | - Gabriele D'Uva
- Department of Experimental, Diagnostic and Specialty Medicine (DIMES), University of Bologna, Bologna, Italy.,Centre for Applied Biomedical Research (CRBA), University of Bologna, Bologna, Italy.,National Laboratory of Molecular Biology and Stem Cell Engineering, National Institute of Biostructures and Biosystems (INBB), Bologna, Italy
| |
Collapse
|
23
|
Cui M, Atmanli A, Morales MG, Tan W, Chen K, Xiao X, Xu L, Liu N, Bassel-Duby R, Olson EN. Nrf1 promotes heart regeneration and repair by regulating proteostasis and redox balance. Nat Commun 2021; 12:5270. [PMID: 34489413 PMCID: PMC8421386 DOI: 10.1038/s41467-021-25653-w] [Citation(s) in RCA: 65] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2021] [Accepted: 08/20/2021] [Indexed: 12/13/2022] Open
Abstract
Following injury, cells in regenerative tissues have the ability to regrow. The mechanisms whereby regenerating cells adapt to injury-induced stress conditions and activate the regenerative program remain to be defined. Here, using the mammalian neonatal heart regeneration model, we show that Nrf1, a stress-responsive transcription factor encoded by the Nuclear Factor Erythroid 2 Like 1 (Nfe2l1) gene, is activated in regenerating cardiomyocytes. Genetic deletion of Nrf1 prevented regenerating cardiomyocytes from activating a transcriptional program required for heart regeneration. Conversely, Nrf1 overexpression protected the adult mouse heart from ischemia/reperfusion (I/R) injury. Nrf1 also protected human induced pluripotent stem cell-derived cardiomyocytes from doxorubicin-induced cardiotoxicity and other cardiotoxins. The protective function of Nrf1 is mediated by a dual stress response mechanism involving activation of the proteasome and redox balance. Our findings reveal that the adaptive stress response mechanism mediated by Nrf1 is required for neonatal heart regeneration and confers cardioprotection in the adult heart.
Collapse
Affiliation(s)
- Miao Cui
- Department of Molecular Biology, the Hamon Center for Regenerative Science and Medicine, University of Texas Southwestern Medical Center, Dallas, TX, USA
- Senator Paul D. Wellstone Muscular Dystrophy Specialized Research Center, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Ayhan Atmanli
- Department of Molecular Biology, the Hamon Center for Regenerative Science and Medicine, University of Texas Southwestern Medical Center, Dallas, TX, USA
- Senator Paul D. Wellstone Muscular Dystrophy Specialized Research Center, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Maria Gabriela Morales
- Department of Molecular Biology, the Hamon Center for Regenerative Science and Medicine, University of Texas Southwestern Medical Center, Dallas, TX, USA
- Senator Paul D. Wellstone Muscular Dystrophy Specialized Research Center, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Wei Tan
- Department of Molecular Biology, the Hamon Center for Regenerative Science and Medicine, University of Texas Southwestern Medical Center, Dallas, TX, USA
- Senator Paul D. Wellstone Muscular Dystrophy Specialized Research Center, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Kenian Chen
- Quantitative Biomedical Research Center, Department of Population & Data Sciences and Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Xue Xiao
- Quantitative Biomedical Research Center, Department of Population & Data Sciences and Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Lin Xu
- Quantitative Biomedical Research Center, Department of Population & Data Sciences and Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Ning Liu
- Department of Molecular Biology, the Hamon Center for Regenerative Science and Medicine, University of Texas Southwestern Medical Center, Dallas, TX, USA
- Senator Paul D. Wellstone Muscular Dystrophy Specialized Research Center, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Rhonda Bassel-Duby
- Department of Molecular Biology, the Hamon Center for Regenerative Science and Medicine, University of Texas Southwestern Medical Center, Dallas, TX, USA
- Senator Paul D. Wellstone Muscular Dystrophy Specialized Research Center, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Eric N Olson
- Department of Molecular Biology, the Hamon Center for Regenerative Science and Medicine, University of Texas Southwestern Medical Center, Dallas, TX, USA.
- Senator Paul D. Wellstone Muscular Dystrophy Specialized Research Center, University of Texas Southwestern Medical Center, Dallas, TX, USA.
| |
Collapse
|
24
|
Lu P, Wang Y, Liu Y, Wang Y, Wu B, Zheng D, Harvey RP, Zhou B. Perinatal angiogenesis from pre-existing coronary vessels via DLL4-NOTCH1 signalling. Nat Cell Biol 2021; 23:967-977. [PMID: 34497373 DOI: 10.1038/s41556-021-00747-1] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2020] [Accepted: 07/27/2021] [Indexed: 12/13/2022]
Abstract
New coronary vessels are added to the heart around birth to support postnatal cardiac growth. Here we show that, in late fetal development, the embryonic coronary plexus at the inner myocardium of the ventricles expresses the angiogenic signalling factors VEGFR3 and DLL4 and generates new coronary vessels in neonates. Contrary to a previous model in which the formation of new coronary vessels in neonates from ventricular endocardial cells was proposed, we find that late fetal and neonatal ventricular endocardial cells lack angiogenic potential and do not contribute to new coronary vessels. Instead, we show using lineage-tracing as well as gain- and loss-of-function experiments that the pre-existing embryonic coronary plexus at the inner myocardium undergoes angiogenic expansion through the DLL4-NOTCH1 signalling pathway to vascularize the expanding myocardium. We also show that the pre-existing coronary plexus revascularizes the regenerating neonatal heart through a similar mechanism. These findings provide a different model of neonatal coronary angiogenesis and regeneration, potentially informing cardiovascular medicine.
Collapse
Affiliation(s)
- Pengfei Lu
- Department of Genetics, Albert Einstein College of Medicine, New York, NY, USA
| | - Yidong Wang
- Cardiovascular Research Center, School of Basic Medical Sciences, Key Laboratory of Environment and Genes Related to Diseases of Ministry of Education, Xi'an Jiaotong University Health Science Center, Xi'an, China
| | - Yang Liu
- Department of Genetics, Albert Einstein College of Medicine, New York, NY, USA
| | - Yifeng Wang
- Department of Cardiology, First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Bingruo Wu
- Department of Genetics, Albert Einstein College of Medicine, New York, NY, USA
| | - Deyou Zheng
- Department of Genetics, Albert Einstein College of Medicine, New York, NY, USA
- Departments of Neurology and Neuroscience, Albert Einstein College of Medicine, New York, NY, USA
| | - Richard P Harvey
- Developmental and Stem Cell Biology Division, Victor Chang Cardiac Research Institute, Sydney, New South Wales, Australia
- St Vincent's Clinical School, and School of Biotechnology and Biomolecular Science, University of New South Wales, Sydney, New South Wales, Australia
| | - Bin Zhou
- Department of Genetics, Albert Einstein College of Medicine, New York, NY, USA.
- Departments of Pediatrics (Pediatric Genetic Medicine) and Medicine (Cardiology), Albert Einstein College of Medicine, New York, NY, USA.
- Wilf Family Cardiovascular Research Institute and Einstein Institute for Aging Research, Albert Einstein College of Medicine, New York, NY, USA.
| |
Collapse
|
25
|
Kim Y, Zharkinbekov Z, Sarsenova M, Yeltay G, Saparov A. Recent Advances in Gene Therapy for Cardiac Tissue Regeneration. Int J Mol Sci 2021; 22:9206. [PMID: 34502115 PMCID: PMC8431496 DOI: 10.3390/ijms22179206] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2021] [Revised: 08/16/2021] [Accepted: 08/16/2021] [Indexed: 12/12/2022] Open
Abstract
Cardiovascular diseases (CVDs) are responsible for enormous socio-economic impact and the highest mortality globally. The standard of care for CVDs, which includes medications and surgical interventions, in most cases, can delay but not prevent the progression of disease. Gene therapy has been considered as a potential therapy to improve the outcomes of CVDs as it targets the molecular mechanisms implicated in heart failure. Cardiac reprogramming, therapeutic angiogenesis using growth factors, antioxidant, and anti-apoptotic therapies are the modalities of cardiac gene therapy that have led to promising results in preclinical studies. Despite the benefits observed in animal studies, the attempts to translate them to humans have been inconsistent so far. Low concentration of the gene product at the target site, incomplete understanding of the molecular pathways of the disease, selected gene delivery method, difference between animal models and humans among others are probable causes of the inconsistent results in clinics. In this review, we discuss the most recent applications of the aforementioned gene therapy strategies to improve cardiac tissue regeneration in preclinical and clinical studies as well as the challenges associated with them. In addition, we consider ongoing gene therapy clinical trials focused on cardiac regeneration in CVDs.
Collapse
Affiliation(s)
| | | | | | | | - Arman Saparov
- Department of Medicine, School of Medicine, Nazarbayev University, Nur-Sultan 010000, Kazakhstan; (Y.K.); (Z.Z.); (M.S.); (G.Y.)
| |
Collapse
|
26
|
Proteomic Analysis of Estrogen-Mediated Enhancement of Mesenchymal Stem Cell-Induced Angiogenesis In Vivo. Cells 2021; 10:cells10092181. [PMID: 34571830 PMCID: PMC8468955 DOI: 10.3390/cells10092181] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Revised: 08/10/2021] [Accepted: 08/17/2021] [Indexed: 12/21/2022] Open
Abstract
Therapeutic use of mesenchymal stem cells (MSCs) for tissue repair has great potential. MSCs from multiple sources, including those derived from human umbilical matrix, namely Wharton’s jelly, may serve as a resource for obtaining MSCs. However, low in vivo engraftment efficacy of MSCs remains a challenging limitation. To improve clinical outcomes using MSCs, an in-depth understanding of the mechanisms and factors involved in successful engraftment is required. We recently demonstrated that 17β-estradiol (E2) improves MSCs in vitro proliferation, directed migration and engraftment in murine heart slices. Here, using a proteomics approach, we investigated the angiogenic potential of MSCs in vivo and the modulatory actions of E2 on mechanisms involved in tissue repair. Specifically, using a Matrigel® plug assay, we evaluated the effects of E2 on MSCs-induced angiogenesis in ovariectomized (OVX) mice. Moreover, using proteomics we investigated the potential pro-repair processes, pathways, and co-mechanisms possibly modified by the treatment of MSCs with E2. Using RT-qPCR, we evaluated mRNA expression of pro-angiogenic molecules, including endoglin, Tie-2, ANG, and VEGF. Hemoglobin levels, a marker for blood vessel formation, were increased in plugs treated with E2 + MSCs, suggesting increased capillary formation. This conclusion was confirmed by the histological analysis of capillary numbers in the Matrigel® plugs treated with E2 + MSC. The LC-MS screening of proteins obtained from the excised Matrigel® plugs revealed 71 proteins that were significantly altered following E2 exposure, 57 up-regulated proteins and 14 down-regulated proteins. A major result was the association of over 100 microRNA molecules (miRNAs) involved in cellular communication, vesicle transport, and metabolic and energy processes, and the high percentage of approximately 25% of genes involved in unknown biological processes. Together, these data provide evidence for increased angiogenesis by MSCs treated with the sex hormone E2. In conclusion, E2 treatment may increase the engraftment and repair potential of MSCs into tissue, and may promote MSC-induced angiogenesis after tissue injury.
Collapse
|
27
|
Wang H, Hironaka CE, Mullis DM, Lucian HJ, Shin HS, Tran NA, Thakore AD, Anilkumar S, Wu MA, Paulsen MJ, Zhu Y, Baker SW, Woo YJ. A neonatal leporine model of age-dependent natural heart regeneration after myocardial infarction. J Thorac Cardiovasc Surg 2021; 164:e389-e405. [PMID: 34649718 DOI: 10.1016/j.jtcvs.2021.08.013] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/22/2020] [Revised: 07/13/2021] [Accepted: 08/01/2021] [Indexed: 11/30/2022]
Abstract
OBJECTIVES Neonatal rodents and piglets naturally regenerate the injured heart after myocardial infarction. We hypothesized that neonatal rabbits also exhibit natural heart regeneration after myocardial infarction. METHODS New Zealand white rabbit kits underwent sham surgery or left coronary ligation on postnatal day 1 (n = 94), postnatal day 4 (n = 11), or postnatal day 7 (n = 52). Hearts were explanted 1 day postsurgery to confirm ischemic injury, at 1 week postsurgery to assess cardiomyocyte proliferation, and at 3 weeks postsurgery to assess left ventricular ejection fraction and scar size. Data are presented as mean ± standard deviation. RESULTS Size of ischemic injury as a percentage of left ventricular area was similar after myocardial infarction on postnatal day 1 versus on postnatal day 7 (42.3% ± 5.4% vs 42.3% ± 4.7%, P = .9984). Echocardiography confirmed severely reduced ejection fraction at 1 day after postnatal day 1 myocardial infarction (33.7% ± 5.3% vs 65.2% ± 5.5% for postnatal day 1 sham, P = .0001), but no difference at 3 weeks after postnatal day 1 myocardial infarction (56.0% ± 4.0% vs 58.0% ± 3.3% for postnatal day 1 sham, P = .2198). Ejection fraction failed to recover after postnatal day 4 myocardial infarction (49.2% ± 1.8% vs 58.5% ± 5.8% for postnatal day 4 sham, P = .0109) and postnatal day 7 myocardial infarction (39.0% ± 7.8% vs 60.2% ± 5.0% for postnatal day 7 sham, P < .0001). At 3 weeks after infarction, fibrotic scar represented 5.3% ± 1.9%, 14.3% ± 4.9%, and 25.4% ± 13.3% of the left ventricle area in the postnatal day 1, postnatal day 4, and postnatal day 7 groups, respectively. An increased proportion of peri-infarct cardiomyocytes expressed Ki67 (15.9% ± 1.8% vs 10.2% ± 0.8%, P = .0039) and aurora B kinase (4.0% ± 0.9% vs 1.5% ± 0.6%, P = .0088) after postnatal day 1 myocardial infarction compared with sham, but no increase was observed after postnatal day 7 myocardial infarction. CONCLUSIONS A neonatal leporine myocardial infarction model reveals that newborn rabbits are capable of age-dependent natural heart regeneration.
Collapse
Affiliation(s)
- Hanjay Wang
- Department of Cardiothoracic Surgery, Stanford University, Stanford, Calif; Stanford Cardiovascular Institute, Stanford University, Stanford, Calif
| | - Camille E Hironaka
- Department of Cardiothoracic Surgery, Stanford University, Stanford, Calif
| | - Danielle M Mullis
- Department of Cardiothoracic Surgery, Stanford University, Stanford, Calif
| | - Haley J Lucian
- Department of Cardiothoracic Surgery, Stanford University, Stanford, Calif
| | - Hye Sook Shin
- Department of Cardiothoracic Surgery, Stanford University, Stanford, Calif
| | - Nicholas A Tran
- Department of Cardiothoracic Surgery, Stanford University, Stanford, Calif
| | - Akshara D Thakore
- Department of Cardiothoracic Surgery, Stanford University, Stanford, Calif
| | - Shreya Anilkumar
- Department of Cardiothoracic Surgery, Stanford University, Stanford, Calif
| | - Matthew A Wu
- Department of Cardiothoracic Surgery, Stanford University, Stanford, Calif
| | - Michael J Paulsen
- Department of Cardiothoracic Surgery, Stanford University, Stanford, Calif
| | - Yuanjia Zhu
- Department of Cardiothoracic Surgery, Stanford University, Stanford, Calif; Department of Bioengineering, Stanford University, Stanford, Calif
| | - Sam W Baker
- Department of Comparative Medicine, Stanford University, Stanford, Calif
| | - Y Joseph Woo
- Department of Cardiothoracic Surgery, Stanford University, Stanford, Calif; Stanford Cardiovascular Institute, Stanford University, Stanford, Calif; Department of Bioengineering, Stanford University, Stanford, Calif.
| |
Collapse
|
28
|
Elde S, Wang H, Woo YJ. The Expanding Armamentarium of Innovative Bioengineered Strategies to Augment Cardiovascular Repair and Regeneration. Front Bioeng Biotechnol 2021; 9:674172. [PMID: 34141702 PMCID: PMC8205517 DOI: 10.3389/fbioe.2021.674172] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2021] [Accepted: 04/13/2021] [Indexed: 11/27/2022] Open
Abstract
Cardiovascular disease remains the leading cause of death worldwide. While clinical trials of cell therapy have demonstrated largely neutral results, recent investigations into the mechanisms of natural myocardial regeneration have demonstrated promising new intersections between molecular, cellular, tissue, biomaterial, and biomechanical engineering solutions. New insight into the crucial role of inflammation in natural regenerative processes may explain why previous efforts have yielded only modest degrees of regeneration. Furthermore, the new understanding of the interdependent relationship of inflammation and myocardial regeneration have catalyzed the emergence of promising new areas of investigation at the intersection of many fields.
Collapse
Affiliation(s)
- Stefan Elde
- Department of Cardiothoracic Surgery, Stanford University, Stanford, CA, United States
| | - Hanjay Wang
- Department of Cardiothoracic Surgery, Stanford University, Stanford, CA, United States.,Stanford Cardiovascular Institute, Stanford University, Stanford, CA, United States
| | - Y Joseph Woo
- Department of Cardiothoracic Surgery, Stanford University, Stanford, CA, United States.,Stanford Cardiovascular Institute, Stanford University, Stanford, CA, United States.,Department of Bioengineering, Stanford University, Stanford, CA, United States
| |
Collapse
|
29
|
Elde S, Wang H, Woo YJ. Navigating the Crossroads of Cell Therapy and Natural Heart Regeneration. Front Cell Dev Biol 2021; 9:674180. [PMID: 34046410 PMCID: PMC8148343 DOI: 10.3389/fcell.2021.674180] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2021] [Accepted: 04/15/2021] [Indexed: 01/14/2023] Open
Abstract
Cardiovascular disease remains the leading cause of death worldwide despite significant advances in our understanding of the disease and its treatment. Consequently, the therapeutic potential of cell therapy and induction of natural myocardial regeneration have stimulated a recent surge of research and clinical trials aimed at addressing this challenge. Recent developments in the field have shed new light on the intricate relationship between inflammation and natural regeneration, an intersection that warrants further investigation.
Collapse
Affiliation(s)
- Stefan Elde
- Department of Cardiothoracic Surgery, Stanford University, Stanford, CA, United States
| | - Hanjay Wang
- Department of Cardiothoracic Surgery, Stanford University, Stanford, CA, United States.,Stanford Cardiovascular Institute, Stanford University, Stanford, CA, United States
| | - Y Joseph Woo
- Department of Cardiothoracic Surgery, Stanford University, Stanford, CA, United States.,Stanford Cardiovascular Institute, Stanford University, Stanford, CA, United States.,Department of Bioengineering, Stanford University, Stanford, CA, United States
| |
Collapse
|
30
|
Zhao M, Nakada Y, Wei Y, Bian W, Chu Y, Borovjagin AV, Xie M, Zhu W, Nguyen T, Zhou Y, Serpooshan V, Walcott GP, Zhang J. Cyclin D2 Overexpression Enhances the Efficacy of Human Induced Pluripotent Stem Cell-Derived Cardiomyocytes for Myocardial Repair in a Swine Model of Myocardial Infarction. Circulation 2021; 144:210-228. [PMID: 33951921 DOI: 10.1161/circulationaha.120.049497] [Citation(s) in RCA: 71] [Impact Index Per Article: 17.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
BACKGROUND Human induced pluripotent stem cells with normal (wild-type) or upregulated (overexpressed) levels of CCND2 (cyclin D2) expression were differentiated into cardiomyocytes (CCND2WTCMs or CCND2OECMs, respectively) and injected into infarcted pig hearts. METHODS Acute myocardial infarction was induced by a 60-minute occlusion of the left anterior descending coronary artery. Immediately after reperfusion, CCND2WTCMs or CCND2OECMs (3×107 cells each) or an equivalent volume of the delivery vehicle was injected around the infarct border zone area. RESULTS The number of the engrafted CCND2OECMs exceeded that of the engrafted CCND2WTCMs from 6- to 8-fold, rising from 1 week to 4 weeks after implantation. In contrast to the treatment with the CCND2WTCMs or the delivery vehicle, the administration of CCND2OECM was associated with significantly improved left ventricular function, as revealed by magnetic resonance imaging. This correlated with reduction of infarct size, fibrosis, ventricular hypertrophy, and cardiomyocyte apoptosis, and increase of vascular density and arterial density, as per histologic analysis of the treated hearts. Expression of cell proliferation markers (eg, Ki67, phosphorylated histone 3, and Aurora B kinase) was also significantly upregulated in the recipient cardiomyocytes from the CCND2OECM-treated than from the CCND2WTCM-treated pigs. The cell proliferation rate and the hypoxia tolerance measured in cultured human induced pluripotent stem cell cardiomyocytes were significantly greater after treatment with exosomes isolated from the CCND2OECMs (CCND2OEExos) than from the CCND2WTCMs (CCND2WTExos). As demonstrated by our study, CCND2OEExos can also promote the proliferation activity of postnatal rat and adult mouse cardiomyocytes. A bulk miRNA sequencing analysis of CCND2OEExos versus CCND2WTExos identified 206 and 91 miRNAs that were significantly upregulated and downregulated, respectively. Gene ontology enrichment analysis identified significant differences in the expression profiles of miRNAs from various functional categories and pathways, including miRNAs implicated in cell-cycle checkpoints (G2/M and G1/S transitions), or the mechanism of cytokinesis. CONCLUSIONS We demonstrated that enhanced potency of CCND2OECMs promoted myocyte proliferation in both grafts and recipient tissue in a large mammal acute myocardial infarction model. These results suggest that CCND2OECMs transplantation may be a potential therapeutic strategy for the repair of infarcted hearts.
Collapse
Affiliation(s)
- Meng Zhao
- Department of Biomedical Engineering (M.Z., Y.N., Y.W., W.B., A.V.B., Y.Z., G.P.W., J.Z.), the University of Alabama at Birmingham
| | - Yuji Nakada
- Department of Biomedical Engineering (M.Z., Y.N., Y.W., W.B., A.V.B., Y.Z., G.P.W., J.Z.), the University of Alabama at Birmingham
| | - Yuhua Wei
- Department of Biomedical Engineering (M.Z., Y.N., Y.W., W.B., A.V.B., Y.Z., G.P.W., J.Z.), the University of Alabama at Birmingham
| | - Weihua Bian
- Department of Biomedical Engineering (M.Z., Y.N., Y.W., W.B., A.V.B., Y.Z., G.P.W., J.Z.), the University of Alabama at Birmingham
| | - Yuxin Chu
- Division of Cardiology, Department of Medicine (Y.C., M.X., G.P.W., J.Z.), the University of Alabama at Birmingham
| | - Anton V Borovjagin
- Department of Biomedical Engineering (M.Z., Y.N., Y.W., W.B., A.V.B., Y.Z., G.P.W., J.Z.), the University of Alabama at Birmingham
| | - Min Xie
- Division of Cardiology, Department of Medicine (Y.C., M.X., G.P.W., J.Z.), the University of Alabama at Birmingham
| | - Wuqiang Zhu
- Department of Cardiovascular Diseases, Physiology and Biomedical Engineering, Mayo Clinic Arizona, Scottsdale (W.Z.)
| | - Thanh Nguyen
- School of Medicine and School of Engineering, and Informatics Institute (T.N.), the University of Alabama at Birmingham
| | - Yang Zhou
- Department of Biomedical Engineering (M.Z., Y.N., Y.W., W.B., A.V.B., Y.Z., G.P.W., J.Z.), the University of Alabama at Birmingham
| | - Vahid Serpooshan
- Wallace H. Coulter Department of Biomedical Engineering, Department of Pediatrics, Emory University and Georgia Institute of Technology, Atlanta (V.S.)
| | - Gregory P Walcott
- Department of Biomedical Engineering (M.Z., Y.N., Y.W., W.B., A.V.B., Y.Z., G.P.W., J.Z.), the University of Alabama at Birmingham.,Division of Cardiology, Department of Medicine (Y.C., M.X., G.P.W., J.Z.), the University of Alabama at Birmingham
| | - Jianyi Zhang
- Department of Biomedical Engineering (M.Z., Y.N., Y.W., W.B., A.V.B., Y.Z., G.P.W., J.Z.), the University of Alabama at Birmingham.,Division of Cardiology, Department of Medicine (Y.C., M.X., G.P.W., J.Z.), the University of Alabama at Birmingham
| |
Collapse
|
31
|
Galeano-Otero I, Del Toro R, Khatib AM, Rosado JA, Ordóñez-Fernández A, Smani T. SARAF and Orai1 Contribute to Endothelial Cell Activation and Angiogenesis. Front Cell Dev Biol 2021; 9:639952. [PMID: 33748129 PMCID: PMC7970240 DOI: 10.3389/fcell.2021.639952] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2020] [Accepted: 02/01/2021] [Indexed: 12/23/2022] Open
Abstract
Angiogenesis is a multistep process that controls endothelial cells (ECs) functioning to form new blood vessels from preexisting vascular beds. This process is tightly regulated by pro-angiogenic factors, such as vascular endothelial growth factor (VEGF), which promote signaling pathways involving the increase in the intracellular Ca2+ concentration ([Ca2+]i). Recent evidence suggests that store-operated calcium entry (SOCE) might play a role in angiogenesis. However, little is known regarding the role of SARAF, SOCE-associated regulatory factor, and Orai1, the pore-forming subunit of the store-operated calcium channel (SOCC), in angiogenesis. Here, we show that SOCE inhibition with GSK-7975A blocks aorta sprouting, as well as human umbilical vein endothelial cell (HUVEC) tube formation and migration. The intraperitoneal injection of GSK-7975A also delays the development of retinal vasculature assessed at postnatal day 6 in mice, since it reduces vessel length and the number of junctions, while it increases lacunarity. Moreover, we find that SARAF and Orai1 are involved in VEGF-mediated [Ca2+]i increase, and their knockdown using siRNA impairs HUVEC tube formation, proliferation, and migration. Finally, immunostaining and in situ proximity ligation assays indicate that SARAF likely interacts with Orai1 in HUVECs. Therefore, these findings show for the first time a functional interaction between SARAF and Orai1 in ECs and highlight their essential role in different steps of the angiogenesis process.
Collapse
Affiliation(s)
- Isabel Galeano-Otero
- Department of Medical Physiology and Biophysics, University of Seville, Seville, Spain.,Group of Cardiovascular Pathophysiology, Institute of Biomedicine of Seville, University Hospital of Virgen del Rocío/University of Seville/CSIC, Seville, Spain.,CIBERCV, Madrid, Spain
| | - Raquel Del Toro
- Department of Medical Physiology and Biophysics, University of Seville, Seville, Spain.,Group of Cardiovascular Pathophysiology, Institute of Biomedicine of Seville, University Hospital of Virgen del Rocío/University of Seville/CSIC, Seville, Spain.,CIBERCV, Madrid, Spain
| | | | | | - Antonio Ordóñez-Fernández
- Group of Cardiovascular Pathophysiology, Institute of Biomedicine of Seville, University Hospital of Virgen del Rocío/University of Seville/CSIC, Seville, Spain.,CIBERCV, Madrid, Spain.,Department of Surgery, University of Seville, Seville, Spain
| | - Tarik Smani
- Department of Medical Physiology and Biophysics, University of Seville, Seville, Spain.,Group of Cardiovascular Pathophysiology, Institute of Biomedicine of Seville, University Hospital of Virgen del Rocío/University of Seville/CSIC, Seville, Spain.,CIBERCV, Madrid, Spain
| |
Collapse
|
32
|
Santos F, Correia M, Nóbrega-Pereira S, Bernardes de Jesus B. Age-Related Pathways in Cardiac Regeneration: A Role for lncRNAs? Front Physiol 2021; 11:583191. [PMID: 33551829 PMCID: PMC7855957 DOI: 10.3389/fphys.2020.583191] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2020] [Accepted: 12/16/2020] [Indexed: 12/13/2022] Open
Abstract
Aging imposes a barrier for tissue regeneration. In the heart, aging leads to a severe rearrangement of the cardiac structure and function and to a subsequent increased risk of heart failure. An intricate network of distinct pathways contributes to age-related alterations during healthy heart aging and account for a higher susceptibility of heart disease. Our understanding of the systemic aging process has already led to the design of anti-aging strategies or to the adoption of protective interventions. Nevertheless, our understanding of the molecular determinants operating during cardiac aging or repair remains limited. Here, we will summarize the molecular and physiological alterations that occur during aging of the heart, highlighting the potential role for long non-coding RNAs (lncRNAs) as novel and valuable targets in cardiac regeneration/repair.
Collapse
Affiliation(s)
- Francisco Santos
- Department of Medical Sciences and Institute of Biomedicine - iBiMED, University of Aveiro, Aveiro, Portugal
| | - Magda Correia
- Department of Medical Sciences and Institute of Biomedicine - iBiMED, University of Aveiro, Aveiro, Portugal
| | - Sandrina Nóbrega-Pereira
- Department of Medical Sciences and Institute of Biomedicine - iBiMED, University of Aveiro, Aveiro, Portugal
- Faculdade de Medicina, Instituto de Medicina Molecular João Lobo Antunes, Universidade de Lisboa, Lisboa, Portugal
| | - Bruno Bernardes de Jesus
- Department of Medical Sciences and Institute of Biomedicine - iBiMED, University of Aveiro, Aveiro, Portugal
| |
Collapse
|
33
|
Derivation of proliferative islet1-positive cells during metamorphosis and wound response in Xenopus. Histochem Cell Biol 2020; 155:133-143. [PMID: 33070205 DOI: 10.1007/s00418-020-01929-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/01/2020] [Indexed: 10/22/2022]
Abstract
In mammalian hearts, cardiomyocytes retain a transient capacity to proliferate and regenerate following injury before birth, whereas they lose proliferative capacity immediately after birth. It has also been known that cardiac progenitor cells including islet1-positive cells do not contribute to the cardiac repair and regeneration in mammals. In contrast, hearts of zebrafish, amphibians and reptiles maintain a regenerative ability throughout life. Here, we analyzed proliferative capacity of cardiac cells during cardiac development and post-ventricular resection using Xenopus laevis, especially focusing on islet1. Immunohistochemical examination showed that islet1-positive cells were present in a wide range of the ventricle and maintained high dividing ability after metamorphosis. Interestingly, the islet1-positive cells were preserved even at 1 year after metamorphosis, some of which showed tropomyosin expression. To assess the possibility of islet1-positive cells as a cellular resource, islet1 response to cardiac resection was analyzed, using adult hearts of 3 months after metamorphosis. Transient gene activation of islet1 in apical region was detected within 1 day after amputation. Histological analyses revealed that islet1-positive cells appeared in the vicinity of resection plane at 1 day post-amputation (dpa) and increased at 3 dpa in both tropomyosin-positive and tropomyosin-negative regions. Vascular labeling analysis by biotinylated dextran amine (BDA) indicated that the islet1-positive cells in a tropomyosin-negative region were closely associated with cardiac vessels. Moreover, dividing ability at this time point was peaked. The resected region was healed with tropomyosin-positive cardiomyocytes until 3 months post-amputation. These results suggest a role of islet1-positive cells as a cellular resource for vascularization and cardiogenesis in Xenopus laevis.
Collapse
|
34
|
de Wit L, Fang J, Neef K, Xiao J, A. Doevendans P, Schiffelers RM, Lei Z, Sluijter JP. Cellular and Molecular Mechanism of Cardiac Regeneration: A Comparison of Newts, Zebrafish, and Mammals. Biomolecules 2020; 10:biom10091204. [PMID: 32825069 PMCID: PMC7564143 DOI: 10.3390/biom10091204] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2020] [Revised: 08/06/2020] [Accepted: 08/17/2020] [Indexed: 12/13/2022] Open
Abstract
Cardiovascular disease is the leading cause of death worldwide. Current palliative treatments can slow the progression of heart failure, but ultimately, the only curative treatment for end-stage heart failure is heart transplantation, which is only available for a minority of patients due to lack of donors' hearts. Explorative research has shown the replacement of the damaged and lost myocardium by inducing cardiac regeneration from preexisting myocardial cells. Lower vertebrates, such as the newt and zebrafish, can regenerate lost myocardium through cardiomyocyte proliferation. The preexisting adult cardiomyocytes replace the lost cells through subsequent dedifferentiation, proliferation, migration, and re-differentiation. Similarly, neonatal mice show complete cardiac regeneration post-injury; however, this regenerative capacity is remarkably diminished one week after birth. In contrast, the adult mammalian heart presents a fibrotic rather than a regenerative response and only shows signs of partial pathological cardiomyocyte dedifferentiation after injury. In this review, we explore the cellular and molecular responses to myocardial insults in different adult species to give insights for future interventional directions by which one can promote or activate cardiac regeneration in mammals.
Collapse
Affiliation(s)
- Lousanne de Wit
- Department of Cardiology, Experimental Cardiology Laboratory, UMC Utrecht, 3584CX Utrecht, The Netherlands; (L.d.W.); (J.F.); (K.N.); (P.A.D.)
| | - Juntao Fang
- Department of Cardiology, Experimental Cardiology Laboratory, UMC Utrecht, 3584CX Utrecht, The Netherlands; (L.d.W.); (J.F.); (K.N.); (P.A.D.)
| | - Klaus Neef
- Department of Cardiology, Experimental Cardiology Laboratory, UMC Utrecht, 3584CX Utrecht, The Netherlands; (L.d.W.); (J.F.); (K.N.); (P.A.D.)
- UMC Utrecht RM Center, Circulatory Health Laboratory, 3584CT Utrecht, The Netherlands
| | - Junjie Xiao
- Institute of Cardiovascular Sciences, Shanghai University, Shanghai 200444, China;
| | - Pieter A. Doevendans
- Department of Cardiology, Experimental Cardiology Laboratory, UMC Utrecht, 3584CX Utrecht, The Netherlands; (L.d.W.); (J.F.); (K.N.); (P.A.D.)
- Utrecht University, 3584CS Utrecht, The Netherlands
- Netherlands Heart Institute (NHI), Central Military Hospital (CMH), 3511EP Utrecht, The Netherlands
| | | | - Zhiyong Lei
- Department of Cardiology, Experimental Cardiology Laboratory, UMC Utrecht, 3584CX Utrecht, The Netherlands; (L.d.W.); (J.F.); (K.N.); (P.A.D.)
- Division LAB, CDL Research, UMC Utrecht, 3584CX Utrecht, The Netherlands;
- Correspondence: (Z.L.); (J.P.G.S.)
| | - Joost P.G. Sluijter
- Department of Cardiology, Experimental Cardiology Laboratory, UMC Utrecht, 3584CX Utrecht, The Netherlands; (L.d.W.); (J.F.); (K.N.); (P.A.D.)
- UMC Utrecht RM Center, Circulatory Health Laboratory, 3584CT Utrecht, The Netherlands
- Utrecht University, 3584CS Utrecht, The Netherlands
- Correspondence: (Z.L.); (J.P.G.S.)
| |
Collapse
|
35
|
Castellan RF, Vitiello M, Vidmar M, Johnstone S, Iacobazzi D, Mellis D, Cathcart B, Thomson A, Ruhrberg C, Caputo M, Newby DE, Gray GA, Baker AH, Caporali A, Meloni M. miR-96 and miR-183 differentially regulate neonatal and adult postinfarct neovascularization. JCI Insight 2020; 5:134888. [PMID: 32544097 PMCID: PMC7453899 DOI: 10.1172/jci.insight.134888] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2019] [Accepted: 06/10/2020] [Indexed: 12/18/2022] Open
Abstract
Following myocardial infarction (MI), the adult heart has minimal regenerative potential. Conversely, the neonatal heart can undergo extensive regeneration, and neovascularization capacity was hypothesized to contribute to this difference. Here, we demonstrate the higher angiogenic potential of neonatal compared with adult mouse cardiac endothelial cells (MCECs) in vitro and use this difference to identify candidate microRNAs (miRs) regulating cardiac angiogenesis after MI. miR expression profiling revealed miR-96 and miR-183 upregulation in adult compared with neonatal MCECs. Their overexpression decreased the angiogenic potential of neonatal MCECs in vitro and prevented scar resolution and neovascularization in neonatal mice after MI. Inversely, their inhibition improved the angiogenic potential of adult MCECs, and miR-96/miR-183–KO mice had increased peri-infarct neovascularization. In silico analyses identified anillin (ANLN) as a direct target of miR-96 and miR-183. In agreement, Anln expression declined following their overexpression and increased after their inhibition in vitro. Moreover, ANLN expression inversely correlated with miR-96 expression and age in cardiac ECs of cardiovascular patients. In vivo, ANLN+ vessels were enriched in the peri-infarct area of miR-96/miR-183–KO mice. These findings identify miR-96 and miR-183 as regulators of neovascularization following MI and miR-regulated genes, such as anillin, as potential therapeutic targets for cardiovascular disease. MiR-96 and miR-183 act as molecular switches to regulate endothelial cells angiogenic potential and differentially regulate neovascularization following myocardial infarction in neonatal and adult mice.
Collapse
Affiliation(s)
- Raphael Fp Castellan
- British Heart Foundation Centre for Cardiovascular Science, Queen's Medical Research Institute, University of Edinburgh, Edinburgh, United Kingdom.,UCL Institute of Ophthalmology, London, United Kingdom
| | - Milena Vitiello
- British Heart Foundation Centre for Cardiovascular Science, Queen's Medical Research Institute, University of Edinburgh, Edinburgh, United Kingdom
| | - Martina Vidmar
- British Heart Foundation Centre for Cardiovascular Science, Queen's Medical Research Institute, University of Edinburgh, Edinburgh, United Kingdom
| | - Steven Johnstone
- Institute of Cardiovascular and Medical Sciences, British Heart Foundation Glasgow Cardiovascular Research Centre, University of Glasgow, Glasgow, United Kingdom
| | - Dominga Iacobazzi
- Bristol Heart Institute, School of Clinical Sciences, University of Bristol, Bristol, United Kingdom
| | - David Mellis
- British Heart Foundation Centre for Cardiovascular Science, Queen's Medical Research Institute, University of Edinburgh, Edinburgh, United Kingdom
| | - Benjamin Cathcart
- British Heart Foundation Centre for Cardiovascular Science, Queen's Medical Research Institute, University of Edinburgh, Edinburgh, United Kingdom
| | - Adrian Thomson
- British Heart Foundation Centre for Cardiovascular Science, Queen's Medical Research Institute, University of Edinburgh, Edinburgh, United Kingdom
| | | | - Massimo Caputo
- Bristol Heart Institute, School of Clinical Sciences, University of Bristol, Bristol, United Kingdom
| | - David E Newby
- British Heart Foundation Centre for Cardiovascular Science, Queen's Medical Research Institute, University of Edinburgh, Edinburgh, United Kingdom
| | - Gillian A Gray
- British Heart Foundation Centre for Cardiovascular Science, Queen's Medical Research Institute, University of Edinburgh, Edinburgh, United Kingdom
| | - Andrew H Baker
- British Heart Foundation Centre for Cardiovascular Science, Queen's Medical Research Institute, University of Edinburgh, Edinburgh, United Kingdom
| | - Andrea Caporali
- British Heart Foundation Centre for Cardiovascular Science, Queen's Medical Research Institute, University of Edinburgh, Edinburgh, United Kingdom
| | - Marco Meloni
- British Heart Foundation Centre for Cardiovascular Science, Queen's Medical Research Institute, University of Edinburgh, Edinburgh, United Kingdom
| |
Collapse
|
36
|
Bergeron A, Brezai A, Shukr R, Villeneuve L, Allen BG, Qureshi WMS, Hentges KE, Calderone A. Filamentous nestin and nonmuscle myosin IIB are associated with a migratory phenotype in neonatal rat cardiomyocytes. J Cell Physiol 2020; 236:1281-1294. [PMID: 32654195 DOI: 10.1002/jcp.29934] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2020] [Revised: 06/28/2020] [Accepted: 07/01/2020] [Indexed: 12/13/2022]
Abstract
Cardiomyocyte migration represents a requisite event of cardiogenesis and the regenerative response of the injured adult zebrafish and neonatal rodent heart. The present study tested the hypothesis that the appearance of the intermediate filament protein nestin in neonatal rat ventricular cardiomyocytes (NNVMs) was associated in part with the acquisition of a migratory phenotype. The cotreatment of NNVMs with phorbol 12,13-dibutyrate (PDBu) and the p38α/β mitogen-activated protein kinase inhibitor SB203580 led to the de novo synthesis of nestin. The intermediate filament protein was subsequently reorganized into a filamentous pattern and redistributed to the leading edge of elongated membrane protrusions translating to significant lengthening of NNVMs. PDBu/SB203580 treatment concomitantly promoted the reorganization of nonmuscle myosin IIB (NMIIB) located predominantly at the periphery of the plasma membrane of NNVMs to a filamentous phenotype extending to the leading edge of elongated membrane protrusions. Coimmunoprecipitation assay revealed a physical interaction between NMIIB and nestin after PDBu/SB203580 treatment of NNVMs. In wild-type and heterozygous NMIIB embryonic hearts at E11.5-E14.5 days, nestin immunoreactivity was identified in a subpopulation of cardiomyocytes elongating perpendicular to the compact myocardium, at the leading edge of nascent trabeculae and during thickening of the compact myocardium. In mutant embryonic hearts lacking NMIIB protein expression, trabeculae formation was reduced, the compact myocardium significantly thinner and nestin immunoreactivity undetectable in cardiomyocytes at E14.5 days. These data suggest that NMIIB and nestin may act in a coordinated fashion to facilitate the acquisition of a migratory phenotype in neonatal and embryonic cardiomyocytes.
Collapse
Affiliation(s)
- Alexandre Bergeron
- Research Center, Montreal Heart Institute, Université de Montréal, Montréal, Québec, Canada
| | - Andra Brezai
- Research Center, Montreal Heart Institute, Université de Montréal, Montréal, Québec, Canada
| | - Rami Shukr
- Research Center, Montreal Heart Institute, Université de Montréal, Montréal, Québec, Canada
| | - Louis Villeneuve
- Research Center, Montreal Heart Institute, Université de Montréal, Montréal, Québec, Canada
| | - Bruce G Allen
- Research Center, Montreal Heart Institute, Université de Montréal, Montréal, Québec, Canada
| | - Wasay M S Qureshi
- Division of Evolution and Genomic Sciences, Faculty of Biology, Medicine and Health, School of Biological Sciences, Manchester Academic Health Science Centre, The University of Manchester, Manchester, UK
| | - Kathryn E Hentges
- Division of Evolution and Genomic Sciences, Faculty of Biology, Medicine and Health, School of Biological Sciences, Manchester Academic Health Science Centre, The University of Manchester, Manchester, UK
| | - Angelino Calderone
- Research Center, Montreal Heart Institute, Université de Montréal, Montréal, Québec, Canada.,Department of Pharmacology and Physiology, Université de Montréal, Montréal, Québec, Canada
| |
Collapse
|
37
|
Wang H, Bennett-Kennett R, Paulsen MJ, Hironaka CE, Thakore AD, Farry JM, Eskandari A, Lucian HJ, Shin HS, Wu MA, Imbrie-Moore AM, Steele AN, Stapleton LM, Zhu Y, Dauskardt RH, Woo YJ. Multiaxial Lenticular Stress-Strain Relationship of Native Myocardium is Preserved by Infarct-Induced Natural Heart Regeneration in Neonatal Mice. Sci Rep 2020; 10:7319. [PMID: 32355240 PMCID: PMC7193551 DOI: 10.1038/s41598-020-63324-w] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2019] [Accepted: 03/13/2020] [Indexed: 12/16/2022] Open
Abstract
Neonatal mice exhibit natural heart regeneration after myocardial infarction (MI) on postnatal day 1 (P1), but this ability is lost by postnatal day 7 (P7). Cardiac biomechanics intricately affect long-term heart function, but whether regenerated cardiac muscle is biomechanically similar to native myocardium remains unknown. We hypothesized that neonatal heart regeneration preserves native left ventricular (LV) biomechanical properties after MI. C57BL/6J mice underwent sham surgery or left anterior descending coronary artery ligation at age P1 or P7. Echocardiography performed 4 weeks post-MI showed that P1 MI and sham mice (n = 22, each) had similar LV wall thickness, diameter, and ejection fraction (59.6% vs 60.7%, p = 0.6514). Compared to P7 shams (n = 20), P7 MI mice (n = 20) had significant LV wall thinning, chamber enlargement, and depressed ejection fraction (32.6% vs 61.8%, p < 0.0001). Afterward, the LV was explanted and pressurized ex vivo, and the multiaxial lenticular stress-strain relationship was tracked. While LV tissue modulus for P1 MI and sham mice were similar (341.9 kPa vs 363.4 kPa, p = 0.6140), the modulus for P7 MI mice was significantly greater than that for P7 shams (691.6 kPa vs 429.2 kPa, p = 0.0194). We conclude that, in neonatal mice, regenerated LV muscle has similar biomechanical properties as native LV myocardium.
Collapse
Affiliation(s)
- Hanjay Wang
- Department of Cardiothoracic Surgery, Stanford University, Stanford, CA, USA
- Stanford Cardiovascular Institute, Stanford University, Stanford, CA, USA
| | - Ross Bennett-Kennett
- Department of Materials Science and Engineering, Stanford University, Stanford, CA, USA
| | - Michael J Paulsen
- Department of Cardiothoracic Surgery, Stanford University, Stanford, CA, USA
| | - Camille E Hironaka
- Department of Cardiothoracic Surgery, Stanford University, Stanford, CA, USA
| | - Akshara D Thakore
- Department of Cardiothoracic Surgery, Stanford University, Stanford, CA, USA
| | - Justin M Farry
- Department of Cardiothoracic Surgery, Stanford University, Stanford, CA, USA
| | - Anahita Eskandari
- Department of Cardiothoracic Surgery, Stanford University, Stanford, CA, USA
| | - Haley J Lucian
- Department of Cardiothoracic Surgery, Stanford University, Stanford, CA, USA
| | - Hye Sook Shin
- Department of Cardiothoracic Surgery, Stanford University, Stanford, CA, USA
| | - Matthew A Wu
- Department of Cardiothoracic Surgery, Stanford University, Stanford, CA, USA
| | - Annabel M Imbrie-Moore
- Department of Cardiothoracic Surgery, Stanford University, Stanford, CA, USA
- Department of Mechanical Engineering, Stanford University, Stanford, CA, USA
| | - Amanda N Steele
- Department of Cardiothoracic Surgery, Stanford University, Stanford, CA, USA
- Department of Bioengineering, Stanford University, Stanford, CA, USA
| | - Lyndsay M Stapleton
- Department of Cardiothoracic Surgery, Stanford University, Stanford, CA, USA
- Department of Bioengineering, Stanford University, Stanford, CA, USA
| | - Yuanjia Zhu
- Department of Cardiothoracic Surgery, Stanford University, Stanford, CA, USA
- Department of Bioengineering, Stanford University, Stanford, CA, USA
| | - Reinhold H Dauskardt
- Department of Materials Science and Engineering, Stanford University, Stanford, CA, USA
| | - Y Joseph Woo
- Department of Cardiothoracic Surgery, Stanford University, Stanford, CA, USA.
- Stanford Cardiovascular Institute, Stanford University, Stanford, CA, USA.
- Department of Bioengineering, Stanford University, Stanford, CA, USA.
| |
Collapse
|
38
|
Chávez MN, Morales RA, López-Crisosto C, Roa JC, Allende ML, Lavandero S. Autophagy Activation in Zebrafish Heart Regeneration. Sci Rep 2020; 10:2191. [PMID: 32042056 PMCID: PMC7010704 DOI: 10.1038/s41598-020-59106-z] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2019] [Accepted: 01/23/2020] [Indexed: 02/06/2023] Open
Abstract
Autophagy is an evolutionarily conserved process that plays a key role in the maintenance of overall cellular health. While it has been suggested that autophagy may elicit cardioprotective and pro-survival modulating functions, excessive activation of autophagy can also be detrimental. In this regard, the zebrafish is considered a hallmark model for vertebrate regeneration, since contrary to adult mammals, it is able to faithfully regenerate cardiac tissue. Interestingly, the role that autophagy may play in zebrafish heart regeneration has not been studied yet. In the present work, we hypothesize that, in the context of a well-established injury model of ventricular apex resection, autophagy plays a critical role during cardiac regeneration and its regulation can directly affect the zebrafish regenerative potential. We studied the autophagy events occurring upon injury using electron microscopy, in vivo tracking of autophagy markers, and protein analysis. Additionally, using pharmacological tools, we investigated how rapamycin, an inducer of autophagy, affects regeneration relevant processes. Our results show that a tightly regulated autophagic response is triggered upon injury and during the early stages of the regeneration process. Furthermore, treatment with rapamycin caused an impairment in the cardiac regeneration outcome. These findings are reminiscent of the pathophysiological description of an injured human heart and hence put forward the zebrafish as a model to study the poorly understood double-sword effect that autophagy has in cardiac homeostasis.
Collapse
Affiliation(s)
- Myra N Chávez
- Advanced Center for Chronic Diseases (ACCDiS) & Corporación Centro de Estudios Científicos de las Enfermedades Crónicas (CECEC), Faculty of Chemical and Pharmaceutical Sciences & Faculty of Medicine, Universidad de Chile, Santiago, Chile.,Center for Genome Regulation (CGR), Department of Biology, Faculty of Sciences, University of Chile, Santiago, Chile
| | - Rodrigo A Morales
- Center for Genome Regulation (CGR), Department of Biology, Faculty of Sciences, University of Chile, Santiago, Chile
| | - Camila López-Crisosto
- Advanced Center for Chronic Diseases (ACCDiS) & Corporación Centro de Estudios Científicos de las Enfermedades Crónicas (CECEC), Faculty of Chemical and Pharmaceutical Sciences & Faculty of Medicine, Universidad de Chile, Santiago, Chile
| | - Juan Carlos Roa
- Department of Pathology, School of Medicine, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Miguel L Allende
- Center for Genome Regulation (CGR), Department of Biology, Faculty of Sciences, University of Chile, Santiago, Chile.
| | - Sergio Lavandero
- Advanced Center for Chronic Diseases (ACCDiS) & Corporación Centro de Estudios Científicos de las Enfermedades Crónicas (CECEC), Faculty of Chemical and Pharmaceutical Sciences & Faculty of Medicine, Universidad de Chile, Santiago, Chile. .,Department of Internal Medicine (Cardiology Division), University of Texas Southwestern Medical Center, Dallas, USA.
| |
Collapse
|
39
|
Natural Heart Regeneration in a Neonatal Rat Myocardial Infarction Model. Cells 2020; 9:cells9010229. [PMID: 31963369 PMCID: PMC7017245 DOI: 10.3390/cells9010229] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2019] [Revised: 01/06/2020] [Accepted: 01/13/2020] [Indexed: 01/09/2023] Open
Abstract
Newborn mice and piglets exhibit natural heart regeneration after myocardial infarction (MI). Discovering other mammals with this ability would provide evidence that neonatal cardiac regeneration after MI may be a conserved phenotype, which if activated in adults could open new options for treating ischemic cardiomyopathy in humans. Here, we hypothesized that newborn rats undergo natural heart regeneration after MI. Using a neonatal rat MI model, we performed left anterior descending coronary artery ligation or sham surgery in one-day-old rats under hypothermic circulatory arrest (n = 74). Operative survival was 97.3%. At 1 day post-surgery, rats in the MI group exhibited significantly reduced ejection fraction (EF) compared to shams (87.1% vs. 53.0%, p < 0.0001). At 3 weeks post-surgery, rats in the sham and MI groups demonstrated no difference in EF (71.1% vs. 69.2%, respectively, p = 0.2511), left ventricular wall thickness (p = 0.9458), or chamber diameter (p = 0.7801). Masson's trichome and picrosirius red staining revealed minimal collagen scar after MI. Increased numbers of cardiomyocytes positive for 5-ethynyl-2'-deoxyuridine (p = 0.0072), Ki-67 (p = 0.0340), and aurora B kinase (p = 0.0430) were observed within the peri-infarct region after MI, indicating ischemia-induced cardiomyocyte proliferation. Overall, we present a neonatal rat MI model and demonstrate that newborn rats are capable of endogenous neocardiomyogenesis after MI.
Collapse
|
40
|
TRPC Channels: Dysregulation and Ca 2+ Mishandling in Ischemic Heart Disease. Cells 2020; 9:cells9010173. [PMID: 31936700 PMCID: PMC7017417 DOI: 10.3390/cells9010173] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2019] [Revised: 01/03/2020] [Accepted: 01/08/2020] [Indexed: 12/17/2022] Open
Abstract
Transient receptor potential canonical (TRPC) channels are ubiquitously expressed in excitable and non-excitable cardiac cells where they sense and respond to a wide variety of physical and chemical stimuli. As other TRP channels, TRPC channels may form homo or heterotetrameric ion channels, and they can associate with other membrane receptors and ion channels to regulate intracellular calcium concentration. Dysfunctions of TRPC channels are involved in many types of cardiovascular diseases. Significant increase in the expression of different TRPC isoforms was observed in different animal models of heart infarcts and in vitro experimental models of ischemia and reperfusion. TRPC channel-mediated increase of the intracellular Ca2+ concentration seems to be required for the activation of the signaling pathway that plays minor roles in the healthy heart, but they are more relevant for cardiac responses to ischemia, such as the activation of different factors of transcription and cardiac hypertrophy, fibrosis, and angiogenesis. In this review, we highlight the current knowledge regarding TRPC implication in different cellular processes related to ischemia and reperfusion and to heart infarction.
Collapse
|
41
|
Cardiac Regeneration and Repair: From Mechanisms to Therapeutic Strategies. CONCEPTS AND APPLICATIONS OF STEM CELL BIOLOGY 2020. [DOI: 10.1007/978-3-030-43939-2_10] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
42
|
Avila-Medina J, Mayoral-González I, Galeano-Otero I, Redondo PC, Rosado JA, Smani T. Pathophysiological Significance of Store-Operated Calcium Entry in Cardiovascular and Skeletal Muscle Disorders and Angiogenesis. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2020; 1131:489-504. [PMID: 31646522 DOI: 10.1007/978-3-030-12457-1_19] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Store-Operated Ca2+ Entry (SOCE) is an important Ca2+ influx pathway expressed by several excitable and non-excitable cell types. SOCE is recognized as relevant signaling pathway not only for physiological process, but also for its involvement in different pathologies. In fact, independent studies demonstrated the implication of essential protein regulating SOCE, such as STIM, Orai and TRPCs, in different pathogenesis and cell disorders, including cardiovascular disease, muscular dystrophies and angiogenesis. Compelling evidence showed that dysregulation in the function and/or expression of isoforms of STIM, Orai or TRPC play pivotal roles in cardiac hypertrophy and heart failure, vascular remodeling and hypertension, skeletal myopathies, and angiogenesis. In this chapter, we summarized the current knowledge concerning the mechanisms underlying abnormal SOCE and its involvement in some diseases, as well as, we discussed the significance of STIM, Orai and TRPC isoforms as possible therapeutic targets for the treatment of angiogenesis, cardiovascular and skeletal muscle diseases.
Collapse
Affiliation(s)
- Javier Avila-Medina
- Department of Medical Physiology and Biophysics, University of Seville, Sevilla, Spain
- Institute of Biomedicine of Seville (IBiS), University Hospital of Virgen del Rocío/CSIC/University of Seville, Sevilla, Spain
| | - Isabel Mayoral-González
- Department of Medical Physiology and Biophysics, University of Seville, Sevilla, Spain
- Institute of Biomedicine of Seville (IBiS), University Hospital of Virgen del Rocío/CSIC/University of Seville, Sevilla, Spain
- Department of Surgery, University of Seville, Sevilla, Spain
| | - Isabel Galeano-Otero
- Department of Medical Physiology and Biophysics, University of Seville, Sevilla, Spain
- Institute of Biomedicine of Seville (IBiS), University Hospital of Virgen del Rocío/CSIC/University of Seville, Sevilla, Spain
| | - Pedro C Redondo
- Department of Physiology, Cell Physiology Research Group and Institute of Molecular Pathology Biomarkers, University of Extremadura, Cáceres, Spain
| | - Juan A Rosado
- Department of Physiology, Cell Physiology Research Group and Institute of Molecular Pathology Biomarkers, University of Extremadura, Cáceres, Spain
| | - Tarik Smani
- Department of Medical Physiology and Biophysics, University of Seville, Sevilla, Spain.
- Institute of Biomedicine of Seville (IBiS), University Hospital of Virgen del Rocío/CSIC/University of Seville, Sevilla, Spain.
- CIBERCV, Madrid, Spain.
| |
Collapse
|
43
|
Grigorian Shamagian L, Madonna R, Taylor D, Climent AM, Prosper F, Bras-Rosario L, Bayes-Genis A, Ferdinandy P, Fernández-Avilés F, Izpisua Belmonte JC, Fuster V, Bolli R. Perspectives on Directions and Priorities for Future Preclinical Studies in Regenerative Medicine. Circ Res 2019; 124:938-951. [PMID: 30870121 DOI: 10.1161/circresaha.118.313795] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
The myocardium consists of numerous cell types embedded in organized layers of ECM (extracellular matrix) and requires an intricate network of blood and lymphatic vessels and nerves to provide nutrients and electrical coupling to the cells. Although much of the focus has been on cardiomyocytes, these cells make up <40% of cells within a healthy adult heart. Therefore, repairing or regenerating cardiac tissue by merely reconstituting cardiomyocytes is a simplistic and ineffective approach. In fact, when an injury occurs, cardiac tissue organization is disrupted at the level of the cells, the tissue architecture, and the coordinated interaction among the cells. Thus, reconstitution of a functional tissue must reestablish electrical and mechanical communication between cardiomyocytes and restore their surrounding environment. It is also essential to restore distinctive myocardial features, such as vascular patency and pump function. In this article, we review the current status, challenges, and future priorities in cardiac regenerative or reparative medicine. In the first part, we provide an overview of our current understanding of heart repair and comment on the main contributors and mechanisms involved in innate regeneration. A brief section is dedicated to the novel concept of rejuvenation or regeneration, which we think may impact future development in the field. The last section describes regenerative therapies, where the most advanced and disruptive strategies used for myocardial repair are discussed. Our recommendations for priority areas in studies of cardiac regeneration or repair are summarized in Tables 1 and 2 .
Collapse
Affiliation(s)
- Lilian Grigorian Shamagian
- From the Hospital Gregorio Marañón, Instituto de Investigación Sanitaria Gregorio Marañón, Universidad Complutense, Madrid, Spain (L.G.S., A.M.C., F.F.-A.).,CIBERCV, ISCIII, Madrid, Spain (L.G.S., A.M.C., A.B.-G., F.F.-A., V.F.)
| | - Rosalinda Madonna
- Center of Aging Sciences and Translational Medicine (CESI-MeT), Institute of Cardiology, G. d'Annunzio University, Chieti, Italy (R.M.).,Department of Internal Medicine, the University of Texas Health Science Center at Houston (R.M., )
| | | | - Andreu M Climent
- From the Hospital Gregorio Marañón, Instituto de Investigación Sanitaria Gregorio Marañón, Universidad Complutense, Madrid, Spain (L.G.S., A.M.C., F.F.-A.).,CIBERCV, ISCIII, Madrid, Spain (L.G.S., A.M.C., A.B.-G., F.F.-A., V.F.)
| | | | - Luis Bras-Rosario
- Cardiology Department, Santa Maria University Hospital (CHLN), Lisbon Academic Medical Centre and Cardiovascular Centre of the University of Lisbon, Faculty of Medicine, Portugal (L.B.-R.)
| | - Antoni Bayes-Genis
- CIBERCV, ISCIII, Madrid, Spain (L.G.S., A.M.C., A.B.-G., F.F.-A., V.F.).,Hospital Germans Trias i Pujol, Badalona, Spain (A.B.-G.)
| | - Péter Ferdinandy
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary (P.F.).,Pharmahungary Group, Szeged, Hungary (P.F.)
| | - Francisco Fernández-Avilés
- From the Hospital Gregorio Marañón, Instituto de Investigación Sanitaria Gregorio Marañón, Universidad Complutense, Madrid, Spain (L.G.S., A.M.C., F.F.-A.).,CIBERCV, ISCIII, Madrid, Spain (L.G.S., A.M.C., A.B.-G., F.F.-A., V.F.)
| | | | - Valentin Fuster
- CIBERCV, ISCIII, Madrid, Spain (L.G.S., A.M.C., A.B.-G., F.F.-A., V.F.).,The Mount Sinai Hospital, New York, NY (V.F.).,Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), Madrid, Spain (V.F.)
| | | |
Collapse
|
44
|
Fujita A, Ueno K, Saito T, Yanagihara M, Kurazumi H, Suzuki R, Mikamo A, Hamano K. Hypoxic-conditioned cardiosphere-derived cell sheet transplantation for chronic myocardial infarction. Eur J Cardiothorac Surg 2019; 56:1062-1074. [PMID: 31329857 DOI: 10.1093/ejcts/ezz122] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/03/2018] [Revised: 03/16/2019] [Accepted: 03/20/2019] [Indexed: 01/07/2023] Open
Abstract
OBJECTIVES Cell therapy provides a suitable environment for regeneration through paracrine effects such as secretion of growth factors. Cardiosphere-derived cells (CDCs) have a high capacity for growth factor secretion and are an attractive target for clinical applications. In particular, a cell sheet technique was reported to have clinical advantages by covering a specific region. Here, we examined the effect of the hypoxic-conditioned (HC) autologous CDC sheet therapy on a rabbit chronic myocardial infarction model. METHODS CDC sheet function was assessed by the enzyme-linked immunosorbent assay and quantified by polymerase chain reaction in vitro (days 1-3 of conditioning). The rabbit chronic myocardial infarction model was established by left coronary ligation. Autologous CDCs were isolated from the left atrial specimen; CDC sheets with or without 2-day HC were transplanted onto the infarcted hearts at 4 weeks. The cardiac function was assessed by an echocardiography at 0, 4 and 8 weeks. A histological analysis of the host hearts was performed by tomato lectin staining at 8 weeks. RESULTS The optimal HC duration was 48 h. HC significantly increased the mRNA expression levels of VEGF and ANG2 on day 2 compared to the normoxic-conditioned (NC) group. The HC group showed significant improvement in the left ventricular ejection fraction (64.4% vs 58.8% and 53.4% in the NC and control) and a greater lectin-positive area in the ischaemic region (HC:NC:control = 13:8:2). CONCLUSIONS HC enhances the paracrine effect of a CDC sheet on angiogenesis to improve cardiac function in the chronic myocardial infarction model, which is essential for cardiomyocyte proliferation during cardiac regeneration.
Collapse
Affiliation(s)
- Akira Fujita
- Department of Surgery and Clinical Science, Graduate School of Medicine, Yamaguchi University, Ube, Yamaguchi, Japan
| | - Koji Ueno
- Department of Surgery and Clinical Science, Graduate School of Medicine, Yamaguchi University, Ube, Yamaguchi, Japan
| | - Toshiro Saito
- Department of Surgery and Clinical Science, Graduate School of Medicine, Yamaguchi University, Ube, Yamaguchi, Japan
| | - Masashi Yanagihara
- Department of Surgery and Clinical Science, Graduate School of Medicine, Yamaguchi University, Ube, Yamaguchi, Japan
| | - Hiroshi Kurazumi
- Department of Surgery and Clinical Science, Graduate School of Medicine, Yamaguchi University, Ube, Yamaguchi, Japan
| | - Ryo Suzuki
- Department of Surgery and Clinical Science, Graduate School of Medicine, Yamaguchi University, Ube, Yamaguchi, Japan
| | - Akihito Mikamo
- Department of Surgery and Clinical Science, Graduate School of Medicine, Yamaguchi University, Ube, Yamaguchi, Japan
| | - Kimikazu Hamano
- Department of Surgery and Clinical Science, Graduate School of Medicine, Yamaguchi University, Ube, Yamaguchi, Japan
| |
Collapse
|
45
|
Shi J, Fan C, Zhuang Y, Sun J, Hou X, Chen B, Xiao Z, Chen Y, Zhan Z, Zhao Y, Dai J. Heparan sulfate proteoglycan promotes fibroblast growth factor-2 function for ischemic heart repair. Biomater Sci 2019; 7:5438-5450. [PMID: 31642823 DOI: 10.1039/c9bm01336a] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
It is well known that the basic fibroblast growth factor (bFGF) promotes angiogenesis after myocardial infarction (MI), but its biological functions decrease in the event of diffusion, enzymolysis, and weak binding with co-receptors in vivo. Heparan sulfate proteoglycans (HSPG) are a major component of extracellular matrices and have been shown to regulate a wide range of cellular functions and bioprocesses by acting as a co-receptor for bFGF and affecting its bioactivities. However, the influence of HSPG on the function of bFGF after myocardial infarction is unknown. Here, exogenous HSPG along with bFGF was injected into the hearts of rats to deliver the angiogenic growth factor for ischemic heart repair following induced MI. The specific binding of HSPG with bFGF protein was demonstrated, which was about 6-fold stronger than the binding of bFGF with heparin. The biological mechanisms of HSPG binding with bFGF were further studied by cell adhesion assay, and assays of bFGF and matrix metalloproteinase 2 (MMP2) activities demonstrated that HSPG enhances cell adhesion, promotes the bioactivity of bFGF in angiogenesis, and protects bFGF from enzymolysis. Our results indicate that HSPG has potential clinical utility as a delivery agent for heparin-binding growth factors. Additionally, HSPG shows high binding affinities with different ECM proteins which also help to anchor bFGF to heart tissue. Therefore, extracellular proteins that mimic the bio-scaffold of the extracellular matrix could promote the activities of bFGF to facilitate ischemic heart repair.
Collapse
Affiliation(s)
- Jiajia Shi
- School of Nano Technology and Nano Bionics, University of Science and Technology of China, Hefei 230026, China and Key Laboratory for Nano-Bio Interface Research, Division of Nanobiomedicine, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences, Suzhou 215123, China
| | - Caixia Fan
- Key Laboratory for Nano-Bio Interface Research, Division of Nanobiomedicine, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences, Suzhou 215123, China
| | - Yan Zhuang
- Key Laboratory for Nano-Bio Interface Research, Division of Nanobiomedicine, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences, Suzhou 215123, China
| | - Jie Sun
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, Orthopedic Institute, Soochow University, 708 Renmin Road, Suzhou, Jiangsu 215007, P.R. China
| | - Xianglin Hou
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China.
| | - Bing Chen
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China.
| | - Zhifeng Xiao
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China.
| | - Yanyan Chen
- Key Laboratory for Nano-Bio Interface Research, Division of Nanobiomedicine, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences, Suzhou 215123, China
| | - Zixuan Zhan
- Department of Thoracic and Cardiovascular Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, 210008, Nanjing, China
| | - Yannan Zhao
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China.
| | - Jianwu Dai
- School of Nano Technology and Nano Bionics, University of Science and Technology of China, Hefei 230026, China and Key Laboratory for Nano-Bio Interface Research, Division of Nanobiomedicine, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences, Suzhou 215123, China and State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China.
| |
Collapse
|
46
|
Paul S, Zhang X, He JQ. Homeobox gene Meis1 modulates cardiovascular regeneration. Semin Cell Dev Biol 2019; 100:52-61. [PMID: 31623926 DOI: 10.1016/j.semcdb.2019.10.003] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2019] [Revised: 09/30/2019] [Accepted: 10/04/2019] [Indexed: 12/20/2022]
Abstract
Regeneration of cardiomyocytes, endothelial cells and vascular smooth muscle cells (three major lineages of cardiac tissues) following myocardial infarction is the critical step to recover the function of the damaged heart. Myeloid ecotropic viral integration site 1 (Meis1) was first discovered in leukemic mice in 1995 and its biological function has been extensively studied in leukemia, hematopoiesis, the embryonic pattering of body axis, eye development and various genetic diseases, such as restless leg syndrome. It was found that Meis1 is highly associated with Hox genes and their cofactors to exert its regulatory effects on multiple intracellular signaling pathways. Recently with the advent of bioinformatics, biochemical methods and advanced genetic engineering tools, new function of Meis1 has been found to be involved in the cell cycle regulation of cardiomyocytes and endothelial cells. For example, inhibition of Meis1 expression increases the proliferative capacity of neonatal mouse cardiomyocytes, whereas overexpression of Meis1 results in the reduction in the length of cardiomyocyte proliferative window. Interestingly, downregulation of one of the circular RNAs, which acts downstream of Meis1 in the cardiomyocytes, promotes angiogenesis and restores the myocardial blood supply, thus reinforcing better regeneration of the damaged heart. It appears that Meis1 may play double roles in modulating proliferation and regeneration of cardiomyocytes and endothelial cells post-myocardial infarction. In this review, we propose to summarize the major findings of Meis1 in modulating fetal development and adult abnormalities, especially focusing on the recent discoveries of Meis1 in controlling the fate of cardiomyocytes and endothelial cells.
Collapse
Affiliation(s)
- Swagatika Paul
- Department of Biomedical Sciences & Pathobiology, College of Veterinary Medicine, Virginia Tech, Blacksburg, VA 24061, USA
| | - Xiaonan Zhang
- Beijing Yulong Shengshi Biotechnology, Haidian District, Beijing, 100085, China
| | - Jia-Qiang He
- Department of Biomedical Sciences & Pathobiology, College of Veterinary Medicine, Virginia Tech, Blacksburg, VA 24061, USA.
| |
Collapse
|
47
|
Hertig V, Brezai A, Bergeron A, Villeneuve L, Gillis MA, Calderone A. p38α MAPK inhibition translates to cell cycle re-entry of neonatal rat ventricular cardiomyocytes and de novo nestin expression in response to thrombin and after apex resection. Sci Rep 2019; 9:8203. [PMID: 31160695 PMCID: PMC6547723 DOI: 10.1038/s41598-019-44712-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2019] [Accepted: 05/20/2019] [Indexed: 12/18/2022] Open
Abstract
The present study tested the hypothesis that p38α MAPK inhibition leads to cell cycle re-entry of neonatal ventricular cardiomyocytes (NNVMs) and de novo nestin expression in response to thrombin and after apex resection of the neonatal rat heart. Thrombin (1 U/ml) treatment of 1-day old NNVMs did not induce cell cycle re-entry or nestin expression. Acute exposure of NNVMs to thrombin increased p38α MAPK and HSP27 phosphorylation and p38α/β MAPK inhibitor SB203580 abrogated HSP27 phosphorylation. Thrombin and SB203580 co-treatment of NNVMs led to bromodeoxyuridine incorporation and nestin expression. SB203580 (5 mg/kg) administration immediately after apex resection of 1-day old neonatal rat hearts and continued for two additional days shortened the fibrin clot length sealing the exposed left ventricular chamber. SB203580-treatment increased the density of troponin-T(+)-NNVMs that incorporated bromodeoxyuridine and expressed nuclear phosphohistone-3. Nestin(+)-NNVMs were selectively detected at the border of the fibrin clot and SB203580 potentiated the density that re-entered the cell cycle. These data suggest that the greater density of ventricular cardiomyocytes and nestin(+)-ventricular cardiomyocytes that re-entered the cell cycle after SB203580 treatment of the apex-resected neonatal rat heart during the acute phase of fibrin clot formation may be attributed in part to inhibition of thrombin-mediated p38α MAPK signalling.
Collapse
Affiliation(s)
- Vanessa Hertig
- Montreal Heart Institute, Université de Montréal, Montréal, Québec, Canada
| | - Andra Brezai
- Montreal Heart Institute, Université de Montréal, Montréal, Québec, Canada
| | - Alexandre Bergeron
- Montreal Heart Institute, Université de Montréal, Montréal, Québec, Canada
| | - Louis Villeneuve
- Montreal Heart Institute, Université de Montréal, Montréal, Québec, Canada
| | | | - Angelino Calderone
- Montreal Heart Institute, Université de Montréal, Montréal, Québec, Canada.
- Département de Pharmacologie et Physiologie, Université de Montréal, Montréal, Québec, Canada.
| |
Collapse
|
48
|
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: 159] [Impact Index Per Article: 26.5] [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
|
49
|
Robich MP, Ryzhov S, Sawyer DB. Successful rebuilding after disaster, even in the heart, starts with infrastructure. J Thorac Dis 2019; 10:S4165-S4167. [PMID: 30631583 DOI: 10.21037/jtd.2018.10.95] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Affiliation(s)
- Michael P Robich
- Maine Medical Center, Cardiovascular Institute, Portland, ME, USA.,Maine Medical Center Research Institute, Scarborough, ME, USA
| | - Sergey Ryzhov
- Maine Medical Center, Cardiovascular Institute, Portland, ME, USA
| | - Douglas B Sawyer
- Maine Medical Center, Cardiovascular Institute, Portland, ME, USA.,Maine Medical Center Research Institute, Scarborough, ME, USA
| |
Collapse
|
50
|
Kapuria S, Yoshida T, Lien CL. Coronary Vasculature in Cardiac Development and Regeneration. J Cardiovasc Dev Dis 2018; 5:E59. [PMID: 30563016 PMCID: PMC6306797 DOI: 10.3390/jcdd5040059] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2018] [Revised: 11/27/2018] [Accepted: 11/30/2018] [Indexed: 12/20/2022] Open
Abstract
Functional coronary circulation is essential for a healthy heart in warm-blooded vertebrates, and coronary diseases can have a fatal consequence. Despite the growing interest, the knowledge about the coronary vessel development and the roles of new coronary vessel formation during heart regeneration is still limited. It is demonstrated that early revascularization is required for efficient heart regeneration. In this comprehensive review, we first describe the coronary vessel formation from an evolutionary perspective. We further discuss the cell origins of coronary endothelial cells and perivascular cells and summarize the critical signaling pathways regulating coronary vessel development. Lastly, we focus on the current knowledge about the molecular mechanisms regulating heart regeneration in zebrafish, a genetically tractable vertebrate model with a regenerative adult heart and well-developed coronary system.
Collapse
Affiliation(s)
- Subir Kapuria
- The Saban Research Institute of Children's Hospital Los Angeles, Los Angeles, CA 90027, USA.
| | - Tyler Yoshida
- The Saban Research Institute of Children's Hospital Los Angeles, Los Angeles, CA 90027, USA.
- Department of Biological Sciences, Dornsife College of Letters, Arts and Sciences, University of Southern California, Los Angeles, CA 90007, USA.
| | - Ching-Ling Lien
- The Saban Research Institute of Children's Hospital Los Angeles, Los Angeles, CA 90027, USA.
- Department of Surgery, University of Southern California, Los Angeles, CA 90033, USA.
- Department of Biochemistry & Molecular Biology, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA.
| |
Collapse
|