1
|
Stougiannou TM, Christodoulou KC, Dimarakis I, Mikroulis D, Karangelis D. To Repair a Broken Heart: Stem Cells in Ischemic Heart Disease. Curr Issues Mol Biol 2024; 46:2181-2208. [PMID: 38534757 PMCID: PMC10969169 DOI: 10.3390/cimb46030141] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2024] [Revised: 02/26/2024] [Accepted: 03/04/2024] [Indexed: 03/28/2024] Open
Abstract
Despite improvements in contemporary medical and surgical therapies, cardiovascular disease (CVD) remains a significant cause of worldwide morbidity and mortality; more specifically, ischemic heart disease (IHD) may affect individuals as young as 20 years old. Typically managed with guideline-directed medical therapy, interventional or surgical methods, the incurred cardiomyocyte loss is not always completely reversible; however, recent research into various stem cell (SC) populations has highlighted their potential for the treatment and perhaps regeneration of injured cardiac tissue, either directly through cellular replacement or indirectly through local paracrine effects. Different stem cell (SC) types have been employed in studies of infarcted myocardium, both in animal models of myocardial infarction (MI) as well as in clinical studies of MI patients, including embryonic stem cells (ESCs) and induced pluripotent stem cells (iPSCs), Muse cells, multipotent stem cells such as bone marrow-derived cells, mesenchymal stem cells (MSCs) and cardiac stem and progenitor cells (CSC/CPCs). These have been delivered as is, in the form of cell therapies, or have been used to generate tissue-engineered (TE) constructs with variable results. In this text, we sought to perform a narrative review of experimental and clinical studies employing various stem cells (SC) for the treatment of infarcted myocardium within the last two decades, with an emphasis on therapies administered through thoracic incision or through percutaneous coronary interventions (PCI), to elucidate possible mechanisms of action and therapeutic effects of such cell therapies when employed in a surgical or interventional manner.
Collapse
Affiliation(s)
- Theodora M. Stougiannou
- Department of Cardiothoracic Surgery, University General Hospital of Alexandroupolis, Dragana, 68100 Alexandroupolis, Greece; (K.C.C.); (D.M.); (D.K.)
| | - Konstantinos C. Christodoulou
- Department of Cardiothoracic Surgery, University General Hospital of Alexandroupolis, Dragana, 68100 Alexandroupolis, Greece; (K.C.C.); (D.M.); (D.K.)
| | - Ioannis Dimarakis
- Division of Cardiothoracic Surgery, University of Washington Medical Center, Seattle, WA 98195, USA;
| | - Dimitrios Mikroulis
- Department of Cardiothoracic Surgery, University General Hospital of Alexandroupolis, Dragana, 68100 Alexandroupolis, Greece; (K.C.C.); (D.M.); (D.K.)
| | - Dimos Karangelis
- Department of Cardiothoracic Surgery, University General Hospital of Alexandroupolis, Dragana, 68100 Alexandroupolis, Greece; (K.C.C.); (D.M.); (D.K.)
| |
Collapse
|
2
|
Sjölin J, Jonsson M, Orback C, Oldfors A, Jeppsson A, Synnergren J, Rotter Sopasakis V, Vukusic K. Expression of Stem Cell Niche-Related Biomarkers at the Base of the Human Tricuspid Valve. Stem Cells Dev 2023; 32:140-151. [PMID: 36565027 PMCID: PMC9986114 DOI: 10.1089/scd.2022.0253] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Stem cell niches have been thoroughly investigated in tissue with high regenerative capacity but not in tissues where cell turnover is slow, such as the human heart. The left AtrioVentricular junction (AVj), the base of the mitral valve, has previously been proposed as a niche region for cardiac progenitors in the adult human heart. In the present study, we explore the right side of the human heart, the base of the tricuspid valve, to investigate the potential of this region as a progenitor niche. Paired biopsies from explanted human hearts were collected from multi-organ donors (N = 12). The lateral side of the AVj, right atria (RA), and right ventricle (RV) were compared for the expression of stem cell niche-related biomarkers using RNA sequencing. Gene expression data indicated upregulation of genes related to embryonic development and extracellular matrix (ECM) composition in the proposed niche region, that is, the AVj. In addition, immunohistochemistry showed high expression of the fetal cardiac markers MDR1, SSEA4, and WT1 within the same region. Nuclear expression of HIF1α was detected suggesting hypoxia. Rare cells were found with the co-staining of the proliferation marker PCNA and Ki67 with cardiomyocyte nuclei marker PCM1 and cardiac Troponin T (cTnT), indicating proliferation of small cardiomyocytes. WT1+/cTnT+ and SSEA4+/cTnT+ cells were also found, suggesting cardiomyocyte-specific progenitors. The expression of the stem cell markers gradually decreased with distance from the tricuspid valve. No expression of these markers was observed in the RV tissue. In summary, the base of the tricuspid valve is an ECM-rich region containing cells with expression of several stem cell niche-associated markers. Co-expression of stem cell markers with cTnT indicates cardiomyocyte-specific progenitors. We previously reported similar data from the base of the mitral valve and thus propose that human adult cardiomyocyte progenitors reside around both atrioventricular valves.
Collapse
Affiliation(s)
- Jacob Sjölin
- Department of Laboratory Medicine, Institute of Biomedicine, and Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden.,Department of Clinical Chemistry, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Marianne Jonsson
- Department of Laboratory Medicine, Institute of Biomedicine, and Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden.,Department of Clinical Chemistry, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Charlotta Orback
- Department of Laboratory Medicine, Institute of Biomedicine, and Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden.,Department of Clinical Chemistry, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Anders Oldfors
- Department of Laboratory Medicine, Institute of Biomedicine, and Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden.,Department of Pathology, and Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Anders Jeppsson
- Department of Cardiothoracic Surgery, Sahlgrenska University Hospital, Gothenburg, Sweden.,Department of Molecular and Clinical Medicine, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Jane Synnergren
- Department of Molecular and Clinical Medicine, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden.,Department of Biology and Bioinformatics, School of Bioscience, University of Skövde, Skövde, Sweden
| | - Victoria Rotter Sopasakis
- Department of Laboratory Medicine, Institute of Biomedicine, and Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden.,Department of Clinical Chemistry, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Kristina Vukusic
- Department of Laboratory Medicine, Institute of Biomedicine, and Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden.,Department of Clinical Chemistry, Sahlgrenska University Hospital, Gothenburg, Sweden
| |
Collapse
|
3
|
Wang C, Nistala R, Cao M, Li DP, Pan Y, Golzy M, Cui Y, Liu Z, Kang X. Repair of Limb Ischemia Is Dependent on Hematopoietic Stem Cell Specific-SHP-1 Regulation of TGF-β1. Arterioscler Thromb Vasc Biol 2023; 43:92-108. [PMID: 36412197 PMCID: PMC10037747 DOI: 10.1161/atvbaha.122.318205] [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] [Indexed: 11/23/2022]
Abstract
BACKGROUND Hematopoietic stem cell (HSC) therapy has shown promise for tissue regeneration after ischemia. Therefore, there is a need to understand mechanisms underlying endogenous HSCs activation in response to ischemic stress and coordination of angiogenesis and repair. SHP-1 plays important roles in HSC quiescence and differentiation by regulation of TGF-β1 signaling. TGF-β1 promotes angiogenesis by stimulating stem cells to secrete growth factors to initiate the formation of blood vessels and later aid in their maturation. We propose that SHP-1 responds to ischemia stress in HSC and progenitor cells (HSPC) via regulation of TGF-β1. METHODS A mouse hind limb ischemia model was used. Local blood perfusion in the limbs was determined using laser doppler perfusion imaging. The number of positive blood vessels per square millimeter, as well as blood vessel diameter (μm) and area (μm2), were calculated. Hematopoietic cells were analyzed using flow cytometry. The bone marrow transplantation assay was performed to measure HSC reconstitution. RESULTS After femoral artery ligation, TGF-β1 was initially decreased in the bone marrow by day 3 of ischemia, followed by an increase on day 7. This pattern was opposite to that in the peripheral blood, which is concordant with the response of HSC to ischemic stress. In contrast, SHP-1 deficiency in HSC is associated with irreversible activation of HSPCs in the bone marrow and increased circulating HSPCs in peripheral blood following limb ischemia. In addition, there was augmented auto-induction of TGF-β1 and sustained inactivation of SHP-1-Smad2 signaling, which impacted TGF-β1 expression in HSPCs in circulation. Importantly, restoration of normal T GF-β1 oscillations helped in the recovery of limb repair and function. CONCLUSIONS HSPC-SHP-1-mediated regulation of TGF-β1 in both bone marrow and peripheral blood is required for a normal response to ischemic stress.
Collapse
Affiliation(s)
- Chen Wang
- Center for Precision Medicine (C.W., R.N., M.C., D.-P.L., Y.P., Y.C., Z.L., X.K.), Department of Medicine, University of Missouri School of Medicine, Columbia
| | - Ravi Nistala
- Center for Precision Medicine (C.W., R.N., M.C., D.-P.L., Y.P., Y.C., Z.L., X.K.), Department of Medicine, University of Missouri School of Medicine, Columbia
- Division of Nephrology (R.N.), Department of Medicine, University of Missouri School of Medicine, Columbia
| | - Min Cao
- Center for Precision Medicine (C.W., R.N., M.C., D.-P.L., Y.P., Y.C., Z.L., X.K.), Department of Medicine, University of Missouri School of Medicine, Columbia
| | - De-Pei Li
- Center for Precision Medicine (C.W., R.N., M.C., D.-P.L., Y.P., Y.C., Z.L., X.K.), Department of Medicine, University of Missouri School of Medicine, Columbia
| | - Yi Pan
- Center for Precision Medicine (C.W., R.N., M.C., D.-P.L., Y.P., Y.C., Z.L., X.K.), Department of Medicine, University of Missouri School of Medicine, Columbia
| | - Mojgan Golzy
- Department of Family and Community Medicine - Biostatistics Unit, School of Medicine, University of Missouri, Columbia (M.G.)
| | - Yuqi Cui
- Center for Precision Medicine (C.W., R.N., M.C., D.-P.L., Y.P., Y.C., Z.L., X.K.), Department of Medicine, University of Missouri School of Medicine, Columbia
- Division of Cardiovascular Medicine (Y.C., Z.L.), Department of Medicine, University of Missouri School of Medicine, Columbia
| | - Zhenguo Liu
- Center for Precision Medicine (C.W., R.N., M.C., D.-P.L., Y.P., Y.C., Z.L., X.K.), Department of Medicine, University of Missouri School of Medicine, Columbia
- Division of Cardiovascular Medicine (Y.C., Z.L.), Department of Medicine, University of Missouri School of Medicine, Columbia
| | - XunLei Kang
- Center for Precision Medicine (C.W., R.N., M.C., D.-P.L., Y.P., Y.C., Z.L., X.K.), Department of Medicine, University of Missouri School of Medicine, Columbia
| |
Collapse
|
4
|
Pelliccia F, Zimarino M, De Luca G, Viceconte N, Tanzilli G, De Caterina R. Endothelial Progenitor Cells in Coronary Artery Disease: From Bench to Bedside. Stem Cells Transl Med 2022; 11:451-460. [PMID: 35365823 PMCID: PMC9154346 DOI: 10.1093/stcltm/szac010] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2021] [Accepted: 02/04/2022] [Indexed: 11/14/2022] Open
Abstract
Endothelial progenitor cells (EPCs) are a heterogeneous group of cells present in peripheral blood at various stages of endothelial differentiation. EPCs have been extensively investigated in patients with coronary artery disease (CAD), with controversial findings both on their role in atherosclerosis progression and in the process of neointimal growth after a percutaneous coronary intervention (PCI). Despite nearly 2 decades of experimental and clinical investigations, however, the significance of EPCs in clinical practice remains unclear and poorly understood. This review provides an update on the role of EPCs in the most common clinical scenarios that are experienced by cardiologists managing patients with CAD. We here summarize the main findings on the association of EPCs with cardiovascular risk factors, coronary atherosclerosis, and myocardial ischemia. We then discuss the potential effects of EPCs in post-PCI in-stent restenosis, as well as most recent findings with EPC-coated stents. Based on the mounting evidence of the relationship between levels of EPCs and several different adverse cardiovascular events, EPCs are emerging as novel predictive biomarkers of long-term outcomes in patients with CAD.
Collapse
Affiliation(s)
| | - Marco Zimarino
- Institute of Cardiology, “G. d’Annunzio” University, Chieti, Italy
- Cath Lab, SS. Annunziata Hospital, Chieti, Italy
| | - Giuseppe De Luca
- Division of Cardiology, Azienda Ospedaliero-Universitaria Maggiore della Carità, Università del Piemonte Orientale, Novara, Italy
| | - Nicola Viceconte
- Department of Cardiovascular Sciences, Sapienza University, Rome, Italy
| | - Gaetano Tanzilli
- Department of Cardiovascular Sciences, Sapienza University, Rome, Italy
| | | |
Collapse
|
5
|
Pelliccia F, Pasceri V, Zimarino M, De Luca G, De Caterina R, Mehran R, Dangas G. Endothelial progenitor cells in coronary atherosclerosis and percutaneous coronary intervention: A systematic review and meta-analysis. CARDIOVASCULAR REVASCULARIZATION MEDICINE 2022; 42:94-99. [DOI: 10.1016/j.carrev.2022.02.025] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2021] [Revised: 02/23/2022] [Accepted: 02/24/2022] [Indexed: 12/19/2022]
|
6
|
Nath AV, Ajit S, Sekar AJ, P R AK, Muthusamy S. MicroRNA-200c/429 mediated regulation of Zeb1 augments N-Cadherin in mouse cardiac mesenchymal cells. Cell Biol Int 2021; 46:222-233. [PMID: 34747544 DOI: 10.1002/cbin.11724] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2021] [Revised: 10/25/2021] [Accepted: 10/31/2021] [Indexed: 11/10/2022]
Abstract
Cardiac mesenchymal cells (CMCs) are a promising cell type that showed therapeutic potential in heart failure models. The analysis of the underlying mechanisms by which the CMCs improve cardiac function is on track. This study aimed to investigate the expression of N-Cadherin, a transmembrane protein that enhances cell adhesion, and recently gained attention for differentiation and augmentation of stem cell function. The mouse CMCs were isolated and analyzed for the mesenchymal markers using flow cytometry. Quantitative real-time polymerase chain reaction (qRT-PCR) and western blot analysis were used to assess the expression of N-Cadherin along with its counteracting molecule E-Cadherin and their regulator Zeb1 in CMCs and dermal fibroblast. The expression level of miR-200c and miR-429 was analyzed using miRNA assays. Transient transfection of miR-200c followed by qRT-PCR, western blot analysis, and immunostaining was done in CMCs to analyze the expression of Zeb1, N-Cadherin, and E-Cadherin. Flow cytometry analysis showed that CMCs possess mesenchymal markers and absence for hematopoietic and immune cell markers. Increased expression of N-Cadherin and Zeb1 in CMCs was observed in CMCs at both RNA and protein levels compared to fibroblast. We found significant downregulation of miR-200c and miR-429 in CMCs. The ectopic expression of miR-200c in CMCs significantly downregulated Zeb1 and N-Cadherin expression. Our findings suggest that the significant downregulation of miR-200c/429 in CMCs maintains the expression of N-Cadherin, which may be important for its functional integrity.
Collapse
Affiliation(s)
- Asha V Nath
- TIMED, Biomedical Technology Wing, Sree Chitra Tirunal Institute for Medical Sciences and Technology, Thiruvananthapuram, Kerala, India
| | - Shilpa Ajit
- Department of Applied Biology, Division of Tissue Culture, Biomedical Technology Wing, Sree Chitra Tirunal Institute for Medical Sciences and Technology, Thiruvananthapuram, Kerala, India
| | - Anupama J Sekar
- Department of Applied Biology, Division of Tissue Culture, Biomedical Technology Wing, Sree Chitra Tirunal Institute for Medical Sciences and Technology, Thiruvananthapuram, Kerala, India
| | - Anil Kumar P R
- Department of Applied Biology, Division of Tissue Culture, Biomedical Technology Wing, Sree Chitra Tirunal Institute for Medical Sciences and Technology, Thiruvananthapuram, Kerala, India
| | - Senthilkumar Muthusamy
- Department of Applied Biology, Division of Tissue Culture, Biomedical Technology Wing, Sree Chitra Tirunal Institute for Medical Sciences and Technology, Thiruvananthapuram, Kerala, India
| |
Collapse
|
7
|
Abstract
Each year 790,000 people in the United States suffer from a myocardial infarction. This results in the permanent loss of cardiomyocytes and an irreversible loss of cardiac function. Current therapies lower mortality rates, but do not address the core pathology, which opens a pathway to step-wise heart failure. Utilizing stem cells to regenerate the dead tissue is a potential method to reverse these devastating effects. Several clinical trials have already demonstrated the safety of stem cell therapy. In this review, we highlight clinical trials, which have utilized various stem cell lineages, and discuss areas for future research.
Collapse
|
8
|
Targeting fibroblast CD248 attenuates CCL17-expressing macrophages and tissue fibrosis. Sci Rep 2020; 10:16772. [PMID: 33033277 PMCID: PMC7544830 DOI: 10.1038/s41598-020-73194-x] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2020] [Accepted: 09/06/2020] [Indexed: 02/07/2023] Open
Abstract
The role of fibroblasts in tissue fibrosis has been extensively studied. Activated fibroblasts, namely myofibroblasts, produce pathological extracellular matrix. CD248, a type I transmembrane glycoprotein, is expressed in fibroblasts after birth. In human chronic kidney disease, upregulated CD248 in myofibroblasts is linked to poor renal survival. In this study, we demonstrated a novel interaction between CD248 and macrophages to be a key step in mediating tissue fibrosis. CD248 was upregulated in myofibroblasts in murine models of renal and peritoneal fibrosis. Cd248 knockout (Cd248–/–) could attenuate both renal and peritoneal fibrosis. By parabiosis of GFP reporter mice and Cd248–/– mice, we showed that attenuation of renal fibrosis was associated with a decrease of macrophage infiltration in Cd248–/– mice. Moreover, decrease of chemokine (C–C motif) ligand 17 and Ccl22 was found in macrophages isolated from the fibrotic kidneys of Cd248–/– mice. Because galectin-3-deficient macrophages showed decreased Ccl17 and Ccl22 in fibrotic kidneys, we further demonstrated that CD248 interacted specifically with galectin-3 of macrophages who then expressed CCL17 to activate collagen production in myofibroblasts. Mice with DNA vaccination targeting CD248 showed decreased fibrosis. We thus propose that CD248 targeting should be studied in the clinical tissue fibrosis setting.
Collapse
|
9
|
Exogenous biological renal support ameliorates renal pathology after ischemia reperfusion injury in elderly mice. Aging (Albany NY) 2020; 11:2031-2044. [PMID: 30978173 PMCID: PMC6503883 DOI: 10.18632/aging.101899] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2018] [Accepted: 03/31/2019] [Indexed: 12/28/2022]
Abstract
We established an exogenous biological renal support model through the generation of parabiotic mice. At 72 hours after ischemia reperfusion injury (IRI), the aged mice that received exogenous biological renal support showed significantly higher levels of renal cell proliferation and dedifferentiation, lower levels of renal tubular injury, improved renal function, and a lower mortality than those that did not receive exogenous biological renal support. Using the Quantibody Mouse Cytokine Antibody Array, we found that aged IRI mice that received exogenous biological renal support had an up-regulation of multiple inflammatory related cytokines compared to the group that did not receive exogenous biological renal support. We suggest that the exogenous biological renal support might promote renal tubular epithelial cell proliferation and dedifferentiation and improve the prognosis of aged IRI mice. Exogenous biological renal support may play an important role in the amelioration of renal IRI by regulating the expression of multiple cytokines.
Collapse
|
10
|
Heterogenetic parabiosis between healthy and dystrophic mice improve the histopathology in muscular dystrophy. Sci Rep 2020; 10:7075. [PMID: 32341395 PMCID: PMC7184587 DOI: 10.1038/s41598-020-64042-z] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2019] [Accepted: 04/09/2020] [Indexed: 11/10/2022] Open
Abstract
Duchenne muscular dystrophy (DMD) is a progressive muscle disease, characterized by mutations in the X-linked dystrophin, that has several therapeutic options but no curative treatment. Transplantation of muscle progenitor cells for treatment of DMD has been widely investigated; however, its application is hindered by limited cell survival due to the harmful dystrophic microenvironment. An alternative approach to utilize progenitor cells and circulatory factors and to improve the dystrophic muscle pathology and microenvironment is through parabiotic pairing, where mice are surgically sutured to create a joint circulatory system. Parabiotic mice were generated by surgically joining wild type (WT) mice expressing green fluorescent protein (GFP) with mdx mice. These mice developed a common circulation (approximately 50% green cells in the blood of mdx mice) 2-weeks after parabiotic pairing. We observed significantly improved dystrophic muscle pathology, including decreased inflammation, necrotic fibers and fibrosis in heterogenetic parabionts. Importantly, the GFP + cells isolated from the mdx mice (paired with GFP mice) underwent myogenic differentiation in vitro and expressed markers of mesenchymal stem cells and macrophages, which may potentially be involved in the improvement of dystrophic muscle pathology. These observations suggest that changing the dystrophic microenvironment can be a new approach to treat DMD.
Collapse
|
11
|
Xiao Y, Wang T, Song X, Yang D, Chu Q, Kang YJ. Copper promotion of myocardial regeneration. Exp Biol Med (Maywood) 2020; 245:911-921. [PMID: 32148090 DOI: 10.1177/1535370220911604] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
IMPACT STATEMENT Copper promotes angiogenesis, but the mechanistic insights have not been fully elucidated until recently. In addition, the significance of copper promotion of angiogenesis in myocardial regeneration was increasingly revealed. Copper critically participates in the regulation of hypoxia-inducible factor 1 (HIF-1) of angiogenic gene expression. Interestingly, myocardial ischemia causes copper efflux from the heart, leading to suppression of angiogenesis, although HIF-1α, the critical subunit of HIF-1, remains accumulated in the ischemic myocardium. Strategies targeting copper specific delivery to the ischemic myocardium lead to selective activation of HIF-1-regulated angiogenic gene expression. Vascularization of the ischemic myocardium re-establishes the tissue injury microenvironment, and rebuilds the conduit for communication between the tissue injury signals and the remote regenerative responses including stem cells. This process promotes myocardial regeneration. Thus, a simple and effective copper supplementation to the ischemic myocardium would become a novel therapeutic approach to the treatment of patients with ischemic heart diseases.
Collapse
Affiliation(s)
- Ying Xiao
- Regenerative Medicine Research Center, Sichuan University West China Hospital, Chengdu, Sichuan 610041, China
| | - Tao Wang
- Regenerative Medicine Research Center, Sichuan University West China Hospital, Chengdu, Sichuan 610041, China
| | - Xin Song
- Regenerative Medicine Research Center, Sichuan University West China Hospital, Chengdu, Sichuan 610041, China
| | - Dan Yang
- Regenerative Medicine Research Center, Sichuan University West China Hospital, Chengdu, Sichuan 610041, China
| | - Qing Chu
- Regenerative Medicine Research Center, Sichuan University West China Hospital, Chengdu, Sichuan 610041, China
| | - Y James Kang
- Regenerative Medicine Research Center, Sichuan University West China Hospital, Chengdu, Sichuan 610041, China
- Memphis Institute of Regenerative Medicine, University of Tennessee Health Science Center, Memphis, TN 38163, USA
| |
Collapse
|
12
|
Yellamilli A, Ren Y, McElmurry RT, Lambert JP, Gross P, Mohsin S, Houser SR, Elrod JW, Tolar J, Garry DJ, van Berlo JH. Abcg2-expressing side population cells contribute to cardiomyocyte renewal through fusion. FASEB J 2020; 34:5642-5657. [PMID: 32100368 DOI: 10.1096/fj.201902105r] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2019] [Revised: 02/10/2020] [Accepted: 02/13/2020] [Indexed: 12/15/2022]
Abstract
The adult mammalian heart has a limited regenerative capacity. Therefore, identification of endogenous cells and mechanisms that contribute to cardiac regeneration is essential for the development of targeted therapies. The side population (SP) phenotype has been used to enrich for stem cells throughout the body; however, SP cells isolated from the heart have been studied exclusively in cell culture or after transplantation, limiting our understanding of their function in vivo. We generated a new Abcg2-driven lineage-tracing mouse model with efficient labeling of SP cells. Labeled SP cells give rise to terminally differentiated cells in bone marrow and intestines. In the heart, labeled SP cells give rise to lineage-traced cardiomyocytes under homeostatic conditions with an increase in this contribution following cardiac injury. Instead of differentiating into cardiomyocytes like proposed cardiac progenitor cells, cardiac SP cells fuse with preexisting cardiomyocytes to stimulate cardiomyocyte cell cycle reentry. Our study is the first to show that fusion between cardiomyocytes and non-cardiomyocytes, identified by the SP phenotype, contribute to endogenous cardiac regeneration by triggering cardiomyocyte cell cycle reentry in the adult mammalian heart.
Collapse
Affiliation(s)
- Amritha Yellamilli
- Lillehei Heart Institute, University of Minnesota Medical School, Minneapolis, MN, USA.,Department of Integrative Biology and Physiology, University of Minnesota Medical School, Minneapolis, MN, USA.,Stem Cell Institute, University of Minnesota Medical School, Minneapolis, MN, USA
| | - Yi Ren
- Lillehei Heart Institute, University of Minnesota Medical School, Minneapolis, MN, USA
| | - Ron T McElmurry
- Stem Cell Institute, University of Minnesota Medical School, Minneapolis, MN, USA.,Department of Pediatrics, University of Minnesota Medical School, Minneapolis, MN, USA
| | - Jonathan P Lambert
- Center for Translational Medicine, Lewis Katz School of Medicine at Temple University, Philadelphia, PA, USA
| | - Polina Gross
- Cardiovascular Research Center, Lewis Katz School of Medicine at Temple University, Philadelphia, PA, USA
| | - Sadia Mohsin
- Cardiovascular Research Center, Lewis Katz School of Medicine at Temple University, Philadelphia, PA, USA
| | - Steven R Houser
- Cardiovascular Research Center, Lewis Katz School of Medicine at Temple University, Philadelphia, PA, USA
| | - John W Elrod
- Center for Translational Medicine, Lewis Katz School of Medicine at Temple University, Philadelphia, PA, USA
| | - Jakub Tolar
- Stem Cell Institute, University of Minnesota Medical School, Minneapolis, MN, USA.,Department of Pediatrics, University of Minnesota Medical School, Minneapolis, MN, USA
| | - Daniel J Garry
- Lillehei Heart Institute, University of Minnesota Medical School, Minneapolis, MN, USA
| | - Jop H van Berlo
- Lillehei Heart Institute, University of Minnesota Medical School, Minneapolis, MN, USA.,Department of Integrative Biology and Physiology, University of Minnesota Medical School, Minneapolis, MN, USA.,Stem Cell Institute, University of Minnesota Medical School, Minneapolis, MN, USA
| |
Collapse
|
13
|
Chen YT, Hsu H, Lin CC, Pan SY, Liu SY, Wu CF, Tsai PZ, Liao CT, Cheng HT, Chiang WC, Chen YM, Chu TS, Lin SL. Inflammatory macrophages switch to CCL17-expressing phenotype and promote peritoneal fibrosis. J Pathol 2019; 250:55-66. [PMID: 31579932 DOI: 10.1002/path.5350] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2019] [Revised: 09/10/2019] [Accepted: 09/19/2019] [Indexed: 12/30/2022]
Abstract
Peritoneal fibrosis remains a problem in kidney failure patients treated with peritoneal dialysis. Severe peritoneal fibrosis with encapsulation or encapsulating peritoneal sclerosis is devastating and life-threatening. Although submesothelial fibroblasts as the major precursor of scar-producing myofibroblasts in animal models and M2 macrophage (Mϕ)-derived chemokines in peritoneal effluents of patients before diagnosis of encapsulating peritoneal sclerosis have been identified, attenuation of peritoneal fibrosis is an unmet medical need partly because the mechanism for cross talk between Mϕs and fibroblasts remains unclear. We use a sodium hypochlorite-induced mouse model akin to clinical encapsulated peritoneal sclerosis to study how the peritoneal Mϕs activate fibroblasts and fibrosis. Sodium hypochlorite induces the disappearance of CD11bhigh F4/80high resident Mϕs but accumulation of CD11bint F4/80int inflammatory Mϕs (InfMϕs) through recruiting blood monocytes and activating local cell proliferation. InfMϕs switch to express chemokine (C-C motif) ligand 17 (CCL17), CCL22, and arginase-1 from day 2 after hypochlorite injury. More than 75% of InfMϕs undergo genetic recombination by Csf1r-driven Cre recombinase, providing the possibility to reduce myofibroblasts and fibrosis by diphtheria toxin-induced Mϕ ablation from day 2 after injury. Furthermore, administration of antibody against CCL17 can reduce Mϕs, myofibroblasts, fibrosis, and improve peritoneal function after injury. Mechanistically, CCL17 stimulates migration and collagen production of submesothelial fibroblasts in culture. By breeding mice that are induced to express red fluorescent protein in Mϕs and green fluorescence protein (GFP) in Col1a1-expressing cells, we confirmed that Mϕs do not produce collagen in peritoneum before and after injury. However, small numbers of fibrocytes are found in fibrotic peritoneum of chimeric mice with bone marrow from Col1a1-GFP reporter mice, but they do not contribute to myofibroblasts. These data demonstrate that InfMϕs switch to pro-fibrotic phenotype and activate peritoneal fibroblasts through CCL17 after injury. CCL17 blockade in patients with peritoneal fibrosis may provide a novel therapy. © 2019 Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.
Collapse
Affiliation(s)
- Yi-Ting Chen
- Department of Integrated Diagnostics and Therapeutics, National Taiwan University Hospital, Taipei, Taiwan.,Renal Division, Department of Internal Medicine, National Taiwan University Hospital, Taipei, Taiwan.,Graduate Institute of Physiology, National Taiwan University College of Medicine, Taipei, Taiwan.,Department of Internal Medicine, E-DA Hospital, I-Shou University, Kaohsiung, Taiwan
| | - Hao Hsu
- Graduate Institute of Physiology, National Taiwan University College of Medicine, Taipei, Taiwan
| | - Chi-Chun Lin
- Graduate Institute of Physiology, National Taiwan University College of Medicine, Taipei, Taiwan
| | - Szu-Yu Pan
- Renal Division, Department of Internal Medicine, National Taiwan University Hospital, Taipei, Taiwan.,Graduate Institute of Physiology, National Taiwan University College of Medicine, Taipei, Taiwan.,Renal Division, Department of Internal Medicine, Far Eastern Memorial Hospital, New Taipei City, Taiwan
| | - Shin-Yun Liu
- Graduate Institute of Physiology, National Taiwan University College of Medicine, Taipei, Taiwan
| | - Ching-Fang Wu
- Department of Internal Medicine, E-DA Hospital, I-Shou University, Kaohsiung, Taiwan
| | - Pei-Zhen Tsai
- Graduate Institute of Physiology, National Taiwan University College of Medicine, Taipei, Taiwan
| | - Chia-Te Liao
- Division of Infection and Immunity, Cardiff University School of Medicine, Cardiff, UK
| | - Hui-Teng Cheng
- Renal Division, Department of Internal Medicine, National Taiwan University Hospital, Hsin-Chu, Taiwan
| | - Wen-Chih Chiang
- Renal Division, Department of Internal Medicine, National Taiwan University Hospital, Taipei, Taiwan
| | - Yung-Ming Chen
- Renal Division, Department of Internal Medicine, National Taiwan University Hospital, Taipei, Taiwan
| | - Tzong-Shinn Chu
- Renal Division, Department of Internal Medicine, National Taiwan University Hospital, Taipei, Taiwan
| | - Shuei-Liong Lin
- Department of Integrated Diagnostics and Therapeutics, National Taiwan University Hospital, Taipei, Taiwan.,Renal Division, Department of Internal Medicine, National Taiwan University Hospital, Taipei, Taiwan.,Graduate Institute of Physiology, National Taiwan University College of Medicine, Taipei, Taiwan.,Research Center for Developmental Biology and Regenerative Medicine, National Taiwan University, Taipei, Taiwan
| |
Collapse
|
14
|
Deutsch MA, Doppler SA, Li X, Lahm H, Santamaria G, Cuda G, Eichhorn S, Ratschiller T, Dzilic E, Dreßen M, Eckart A, Stark K, Massberg S, Bartels A, Rischpler C, Gilsbach R, Hein L, Fleischmann BK, Wu SM, Lange R, Krane M. Reactivation of the Nkx2.5 cardiac enhancer after myocardial infarction does not presage myogenesis. Cardiovasc Res 2019; 114:1098-1114. [PMID: 29579159 DOI: 10.1093/cvr/cvy069] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/27/2017] [Accepted: 03/15/2018] [Indexed: 12/13/2022] Open
Abstract
Aims The contribution of resident stem or progenitor cells to cardiomyocyte renewal after injury in adult mammalian hearts remains a matter of considerable debate. We evaluated a cell population in the adult mouse heart induced by myocardial infarction (MI) and characterized by an activated Nkx2.5 enhancer element that is specific for multipotent cardiac progenitor cells (CPCs) during embryonic development. We hypothesized that these MI-induced cells (MICs) harbour cardiomyogenic properties similar to their embryonic counterparts. Methods and results MICs reside in the heart and mainly localize to the infarction area and border zone. Interestingly, gene expression profiling of purified MICs 1 week after infarction revealed increased expression of stem cell markers and embryonic cardiac transcription factors (TFs) in these cells as compared to the non-mycoyte cell fraction of adult hearts. A subsequent global transcriptome comparison with embryonic CPCs and fibroblasts and in vitro culture of MICs unveiled that (myo-)fibroblastic features predominated and that cardiac TFs were only expressed at background levels. Conclusions Adult injury-induced reactivation of a cardiac-specific Nkx2.5 enhancer element known to specifically mark myocardial progenitor cells during embryonic development does not reflect hypothesized embryonic cardiomyogenic properties. Our data suggest a decreasing plasticity of cardiac progenitor (-like) cell populations with increasing age. A re-expression of embryonic, stem or progenitor cell features in the adult heart must be interpreted very carefully with respect to the definition of cardiac resident progenitor cells. Albeit, the abundance of scar formation after cardiac injury suggests a potential to target predestinated activated profibrotic cells to push them towards cardiomyogenic differentiation to improve regeneration.
Collapse
Affiliation(s)
- Marcus-André Deutsch
- Department of Cardiovascular Surgery, German Heart Center Munich at the Technische Universität München, Lazarettstraße 36, 80636 Munich, Germany.,Department of Cardiovascular Surgery, German Heart Center, Insure (Institute for Translational Cardiac Surgery), Technische Universität München, Lothstraße 11, 80636 Munich, Germany.,DZHK (German Center for Cardiovascular Research), Partner Site Munich Heart Alliance, Munich, Germany
| | - Stefanie A Doppler
- Department of Cardiovascular Surgery, German Heart Center Munich at the Technische Universität München, Lazarettstraße 36, 80636 Munich, Germany.,Department of Cardiovascular Surgery, German Heart Center, Insure (Institute for Translational Cardiac Surgery), Technische Universität München, Lothstraße 11, 80636 Munich, Germany
| | - Xinghai Li
- Department of Cardiovascular Surgery, German Heart Center Munich at the Technische Universität München, Lazarettstraße 36, 80636 Munich, Germany.,Department of Cardiovascular Surgery, German Heart Center, Insure (Institute for Translational Cardiac Surgery), Technische Universität München, Lothstraße 11, 80636 Munich, Germany
| | - Harald Lahm
- Department of Cardiovascular Surgery, German Heart Center Munich at the Technische Universität München, Lazarettstraße 36, 80636 Munich, Germany.,Department of Cardiovascular Surgery, German Heart Center, Insure (Institute for Translational Cardiac Surgery), Technische Universität München, Lothstraße 11, 80636 Munich, Germany
| | - Gianluca Santamaria
- Stem Cell Laboratory, Department of Experimental and Clinical Medicine, Research Center of Advanced Biochemistry and Molecular Biology.,CIS (Centro Interdisciplinare Servizi), University 'Magna Graecia' of Catanzaro, Viale Europa, 88100 Catanzaro, Italy
| | - Giovanni Cuda
- Stem Cell Laboratory, Department of Experimental and Clinical Medicine, Research Center of Advanced Biochemistry and Molecular Biology
| | - Stefan Eichhorn
- Department of Cardiovascular Surgery, German Heart Center Munich at the Technische Universität München, Lazarettstraße 36, 80636 Munich, Germany.,Department of Cardiovascular Surgery, German Heart Center, Insure (Institute for Translational Cardiac Surgery), Technische Universität München, Lothstraße 11, 80636 Munich, Germany
| | - Thomas Ratschiller
- Department of Cardiothoracic and Vascular Surgery, Kepler University Hospital, 4021 Linz, Austria
| | - Elda Dzilic
- Department of Cardiovascular Surgery, German Heart Center Munich at the Technische Universität München, Lazarettstraße 36, 80636 Munich, Germany.,Department of Cardiovascular Surgery, German Heart Center, Insure (Institute for Translational Cardiac Surgery), Technische Universität München, Lothstraße 11, 80636 Munich, Germany
| | - Martina Dreßen
- Department of Cardiovascular Surgery, German Heart Center Munich at the Technische Universität München, Lazarettstraße 36, 80636 Munich, Germany.,Department of Cardiovascular Surgery, German Heart Center, Insure (Institute for Translational Cardiac Surgery), Technische Universität München, Lothstraße 11, 80636 Munich, Germany
| | - Annekathrin Eckart
- Medizinische Klinik und Poliklinik I, Ludwig-Maximilians-Universität, 81377 Munich, Germany
| | - Konstantin Stark
- DZHK (German Center for Cardiovascular Research), Partner Site Munich Heart Alliance, Munich, Germany.,Medizinische Klinik und Poliklinik I, Ludwig-Maximilians-Universität, 81377 Munich, Germany
| | - Steffen Massberg
- DZHK (German Center for Cardiovascular Research), Partner Site Munich Heart Alliance, Munich, Germany.,Medizinische Klinik und Poliklinik I, Ludwig-Maximilians-Universität, 81377 Munich, Germany
| | - Anna Bartels
- Nuklearmedizinische Klinik des Klinikums Rechts der Isar, Technische Universität München, 81675 Munich, Germany
| | - Christoph Rischpler
- DZHK (German Center for Cardiovascular Research), Partner Site Munich Heart Alliance, Munich, Germany.,Nuklearmedizinische Klinik des Klinikums Rechts der Isar, Technische Universität München, 81675 Munich, Germany
| | - Ralf Gilsbach
- Institute of Experimental and Clinical Pharmacology and Toxicology, Faculty of Medicine, University of Freiburg, Albertstraße 25, 79104 Freiburg, Germany
| | - Lutz Hein
- Institute of Experimental and Clinical Pharmacology and Toxicology, Faculty of Medicine, University of Freiburg, Albertstraße 25, 79104 Freiburg, Germany.,BIOSS Centre for Biological Signaling Studies, University of Freiburg, Schänzlestraße 18, 79104 Freiburg, Germany
| | - Bernd K Fleischmann
- Institute of Physiology I, Life and Brain Center, Medical Faculty, University of Bonn, 53105 Bonn, Germany
| | - Sean M Wu
- Division of Cardiovascular Medicine, Department of Medicine, Stanford Cardiovascular Institute, Institute for Stem Cell Biology and Regenerative Medicine, Stanford University, Stanford, CA 94305, USA
| | - Rüdiger Lange
- Department of Cardiovascular Surgery, German Heart Center Munich at the Technische Universität München, Lazarettstraße 36, 80636 Munich, Germany.,Department of Cardiovascular Surgery, German Heart Center, Insure (Institute for Translational Cardiac Surgery), Technische Universität München, Lothstraße 11, 80636 Munich, Germany.,DZHK (German Center for Cardiovascular Research), Partner Site Munich Heart Alliance, Munich, Germany
| | - Markus Krane
- Department of Cardiovascular Surgery, German Heart Center Munich at the Technische Universität München, Lazarettstraße 36, 80636 Munich, Germany.,Department of Cardiovascular Surgery, German Heart Center, Insure (Institute for Translational Cardiac Surgery), Technische Universität München, Lothstraße 11, 80636 Munich, Germany.,DZHK (German Center for Cardiovascular Research), Partner Site Munich Heart Alliance, Munich, Germany
| |
Collapse
|
15
|
Wei SY, Pan SY, Li B, Chen YM, Lin SL. Rejuvenation: Turning back the clock of aging kidney. J Formos Med Assoc 2019; 119:898-906. [PMID: 31202499 DOI: 10.1016/j.jfma.2019.05.020] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2019] [Revised: 05/14/2019] [Accepted: 05/24/2019] [Indexed: 12/11/2022] Open
Abstract
Aging is inevitable in life. It is defined as impaired adaptive capacity to environmental or internal stresses with growing rates of disease and death. Aging is also an important risk factor for various kidney diseases such as acute kidney injury and chronic kidney disease. Patients older than 65 years have nearly 28% risk of failing recovery of kidney function when suffering from acute kidney injury. It is reported that more than a third of population aged 65 years and older have chronic kidney disease in Taiwan, and the occurrence of multiple age-related disorders is predicted to increase in parallel. Renal aging is a complex, multifactorial process characterized by many anatomical and functional changes. Several factors are involved in renal aging, such as loss of telomeres, cell cycle arrest, chronic inflammation, activation of renin-angiotensin system, decreased klotho expression, and development of tertiary lymphoid tissues. These changes can also be observed in many other different types of renal injury. Recent studies suggested that young blood may rejuvenate aged organs, including the kidneys. In order to develop new therapeutic strategies for renal aging, the mechanisms underlying renal aging and by which young blood can halt or reverse aging process warrants further study.
Collapse
Affiliation(s)
- Shi-Yao Wei
- Graduate Institute of Physiology, College of Medicine, National Taiwan University, Taipei, Taiwan; Department of Nephrology, Second Affiliated Hospital of Harbin Medical University, Harbin, People's Republic of China
| | - Szu-Yu Pan
- Graduate Institute of Physiology, College of Medicine, National Taiwan University, Taipei, Taiwan; Department of Internal Medicine, Far Eastern Memorial Hospital, New Taipei City, Taiwan; Renal Division, Department of Internal Medicine, National Taiwan University Hospital, Taipei, Taiwan
| | - Bing Li
- Department of Nephrology, Second Affiliated Hospital of Harbin Medical University, Harbin, People's Republic of China
| | - Yung-Ming Chen
- Renal Division, Department of Internal Medicine, National Taiwan University Hospital, Taipei, Taiwan
| | - Shuei-Liong Lin
- Graduate Institute of Physiology, College of Medicine, National Taiwan University, Taipei, Taiwan; Renal Division, Department of Internal Medicine, National Taiwan University Hospital, Taipei, Taiwan; Department of Integrated Diagnostics & Therapeutics, National Taiwan University Hospital, Taipei, Taiwan; Research Center for Developmental Biology and Regenerative Medicine, National Taiwan University, Taipei, Taiwan.
| |
Collapse
|
16
|
Sung J, Sodhi CP, Voltaggio L, Hou X, Jia H, Zhou Q, Čiháková D, Hackam DJ. The recruitment of extra-intestinal cells to the injured mucosa promotes healing in radiation enteritis and chemical colitis in a mouse parabiosis model. Mucosal Immunol 2019; 12:503-517. [PMID: 30617302 PMCID: PMC6445662 DOI: 10.1038/s41385-018-0123-3] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2018] [Revised: 11/05/2018] [Accepted: 11/20/2018] [Indexed: 02/04/2023]
Abstract
Mucosal healing occurs through migration and proliferation of cells within injured epithelium, yet these processes may be inadequate for mucosal healing after significant injury where the mucosa is denuded. We hypothesize that extra-intestinal cells can contribute to mucosal healing after injury to the small and large intestine. We generated parabiotic pairs between wild-type and tdTomato mice, which were then subjected to radiation-induced enteritis and 2,4,6-trinitrobenzene sulfonic acid (TNBS)-induced colitis. We now show that as compared with singleton mice, mice with a parabiotic partner were protected against intestinal damage as revealed by significantly reduced weight loss, reduced expression of pro-inflammatory cytokines, reduced enterocyte apoptosis, and improved crypt proliferation. Donor cells expressed CD45-, Sca-1+, c-kit+, and CXCR4+ and accumulated around the injured crypts but did not transdifferentiate into epithelia, suggesting that extra-intestinal cells play a paracrine role in the healing response, while parabiotic pairings with Rag1-/- mice showed improved healing, indicating that adaptive immune cells were dispensable for mucosal healing. Strikingly, ablation of the bone marrow of the donor parabionts removed the protective effects. These findings reveal that the recruitment of extra-intestinal, bone marrow-derived cells into the injured intestinal mucosa can promote mucosal healing, suggesting novel therapeutic approaches for severe intestinal disease.
Collapse
Affiliation(s)
- J Sung
- Institute of Genetic Medicine, School of Medicine, Johns Hopkins University, Baltimore, MD, USA
| | - C P Sodhi
- Division of Pediatric Surgery, Johns Hopkins Children's Center and Department of Surgery, Baltimore, MD, USA
| | - L Voltaggio
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - X Hou
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD, USA
| | - H Jia
- Division of Pediatric Surgery, Johns Hopkins Children's Center and Department of Surgery, Baltimore, MD, USA
| | - Q Zhou
- Division of Pediatric Surgery, Johns Hopkins Children's Center and Department of Surgery, Baltimore, MD, USA
| | - D Čiháková
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - D J Hackam
- Institute of Genetic Medicine, School of Medicine, Johns Hopkins University, Baltimore, MD, USA.
- Division of Pediatric Surgery, Johns Hopkins Children's Center and Department of Surgery, Baltimore, MD, USA.
| |
Collapse
|
17
|
Doppler SA, Deutsch MA, Serpooshan V, Li G, Dzilic E, Lange R, Krane M, Wu SM. Mammalian Heart Regeneration: The Race to the Finish Line. Circ Res 2019; 120:630-632. [PMID: 28209796 DOI: 10.1161/circresaha.116.310051] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Affiliation(s)
- Stefanie A Doppler
- From the Division of Experimental Surgery, Department of Cardiovascular Surgery, German Heart Center Munich, Germany (S.A.D., M.-A.D., E.D., R.L., M.K.); Stanford Cardiovascular Institute (V.S., G.L., E.D., S.M.W.), Institute for Stem Cell and Regenerative Medicine (S.M.W.), and Division of Cardiovascular Medicine, Department of Medicine (S.M.W.), Stanford University School of Medicine, CA; DZHK (German Center for Cardiovascular Research), partner site Munich Heart Alliance, Germany (M.-A.D., M.K.)
| | - Marcus-Andre Deutsch
- From the Division of Experimental Surgery, Department of Cardiovascular Surgery, German Heart Center Munich, Germany (S.A.D., M.-A.D., E.D., R.L., M.K.); Stanford Cardiovascular Institute (V.S., G.L., E.D., S.M.W.), Institute for Stem Cell and Regenerative Medicine (S.M.W.), and Division of Cardiovascular Medicine, Department of Medicine (S.M.W.), Stanford University School of Medicine, CA; DZHK (German Center for Cardiovascular Research), partner site Munich Heart Alliance, Germany (M.-A.D., M.K.)
| | - Vahid Serpooshan
- From the Division of Experimental Surgery, Department of Cardiovascular Surgery, German Heart Center Munich, Germany (S.A.D., M.-A.D., E.D., R.L., M.K.); Stanford Cardiovascular Institute (V.S., G.L., E.D., S.M.W.), Institute for Stem Cell and Regenerative Medicine (S.M.W.), and Division of Cardiovascular Medicine, Department of Medicine (S.M.W.), Stanford University School of Medicine, CA; DZHK (German Center for Cardiovascular Research), partner site Munich Heart Alliance, Germany (M.-A.D., M.K.)
| | - Guang Li
- From the Division of Experimental Surgery, Department of Cardiovascular Surgery, German Heart Center Munich, Germany (S.A.D., M.-A.D., E.D., R.L., M.K.); Stanford Cardiovascular Institute (V.S., G.L., E.D., S.M.W.), Institute for Stem Cell and Regenerative Medicine (S.M.W.), and Division of Cardiovascular Medicine, Department of Medicine (S.M.W.), Stanford University School of Medicine, CA; DZHK (German Center for Cardiovascular Research), partner site Munich Heart Alliance, Germany (M.-A.D., M.K.)
| | - Elda Dzilic
- From the Division of Experimental Surgery, Department of Cardiovascular Surgery, German Heart Center Munich, Germany (S.A.D., M.-A.D., E.D., R.L., M.K.); Stanford Cardiovascular Institute (V.S., G.L., E.D., S.M.W.), Institute for Stem Cell and Regenerative Medicine (S.M.W.), and Division of Cardiovascular Medicine, Department of Medicine (S.M.W.), Stanford University School of Medicine, CA; DZHK (German Center for Cardiovascular Research), partner site Munich Heart Alliance, Germany (M.-A.D., M.K.)
| | - Rüdiger Lange
- From the Division of Experimental Surgery, Department of Cardiovascular Surgery, German Heart Center Munich, Germany (S.A.D., M.-A.D., E.D., R.L., M.K.); Stanford Cardiovascular Institute (V.S., G.L., E.D., S.M.W.), Institute for Stem Cell and Regenerative Medicine (S.M.W.), and Division of Cardiovascular Medicine, Department of Medicine (S.M.W.), Stanford University School of Medicine, CA; DZHK (German Center for Cardiovascular Research), partner site Munich Heart Alliance, Germany (M.-A.D., M.K.)
| | - Markus Krane
- From the Division of Experimental Surgery, Department of Cardiovascular Surgery, German Heart Center Munich, Germany (S.A.D., M.-A.D., E.D., R.L., M.K.); Stanford Cardiovascular Institute (V.S., G.L., E.D., S.M.W.), Institute for Stem Cell and Regenerative Medicine (S.M.W.), and Division of Cardiovascular Medicine, Department of Medicine (S.M.W.), Stanford University School of Medicine, CA; DZHK (German Center for Cardiovascular Research), partner site Munich Heart Alliance, Germany (M.-A.D., M.K.).
| | - Sean M Wu
- From the Division of Experimental Surgery, Department of Cardiovascular Surgery, German Heart Center Munich, Germany (S.A.D., M.-A.D., E.D., R.L., M.K.); Stanford Cardiovascular Institute (V.S., G.L., E.D., S.M.W.), Institute for Stem Cell and Regenerative Medicine (S.M.W.), and Division of Cardiovascular Medicine, Department of Medicine (S.M.W.), Stanford University School of Medicine, CA; DZHK (German Center for Cardiovascular Research), partner site Munich Heart Alliance, Germany (M.-A.D., M.K.).
| |
Collapse
|
18
|
Tang TW, Chen HC, Chen CY, Yen CY, Lin CJ, Prajnamitra RP, Chen LL, Ruan SC, Lin JH, Lin PJ, Lu HH, Kuo CW, Chang CM, Hall AD, Vivas EI, Shui JW, Chen P, Hacker TA, Rey FE, Kamp TJ, Hsieh PC. Loss of Gut Microbiota Alters Immune System Composition and Cripples Postinfarction Cardiac Repair. Circulation 2019; 139:647-659. [DOI: 10.1161/circulationaha.118.035235] [Citation(s) in RCA: 102] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Affiliation(s)
- Tony W.H. Tang
- Program in Molecular Medicine, National Yang Ming University and Academia Sinica, Taipei, Taiwan (T.W.H.T., P.C.C.H.)
- Institute of Biomedical Sciences (T.W.H.T., H.C.-C., C.-Y.C., C.Y.T.Y., C.-J.L., R.P.P., L.-L.C., S.-C.R., J.-H.L., P.-J.L., H.-H.L., J.-W.S., P.C.H.H.), Academia Sinica, Taipei, Taiwan
| | - Hung-Chih Chen
- Institute of Biomedical Sciences (T.W.H.T., H.C.-C., C.-Y.C., C.Y.T.Y., C.-J.L., R.P.P., L.-L.C., S.-C.R., J.-H.L., P.-J.L., H.-H.L., J.-W.S., P.C.H.H.), Academia Sinica, Taipei, Taiwan
| | - Chen-Yun Chen
- Institute of Biomedical Sciences (T.W.H.T., H.C.-C., C.-Y.C., C.Y.T.Y., C.-J.L., R.P.P., L.-L.C., S.-C.R., J.-H.L., P.-J.L., H.-H.L., J.-W.S., P.C.H.H.), Academia Sinica, Taipei, Taiwan
| | - Christopher Y.T. Yen
- Institute of Biomedical Sciences (T.W.H.T., H.C.-C., C.-Y.C., C.Y.T.Y., C.-J.L., R.P.P., L.-L.C., S.-C.R., J.-H.L., P.-J.L., H.-H.L., J.-W.S., P.C.H.H.), Academia Sinica, Taipei, Taiwan
| | - Chen-Ju Lin
- Institute of Biomedical Sciences (T.W.H.T., H.C.-C., C.-Y.C., C.Y.T.Y., C.-J.L., R.P.P., L.-L.C., S.-C.R., J.-H.L., P.-J.L., H.-H.L., J.-W.S., P.C.H.H.), Academia Sinica, Taipei, Taiwan
| | - Ray P. Prajnamitra
- Institute of Biomedical Sciences (T.W.H.T., H.C.-C., C.-Y.C., C.Y.T.Y., C.-J.L., R.P.P., L.-L.C., S.-C.R., J.-H.L., P.-J.L., H.-H.L., J.-W.S., P.C.H.H.), Academia Sinica, Taipei, Taiwan
| | - Li-Lun Chen
- Institute of Biomedical Sciences (T.W.H.T., H.C.-C., C.-Y.C., C.Y.T.Y., C.-J.L., R.P.P., L.-L.C., S.-C.R., J.-H.L., P.-J.L., H.-H.L., J.-W.S., P.C.H.H.), Academia Sinica, Taipei, Taiwan
| | - Shu-Chian Ruan
- Institute of Biomedical Sciences (T.W.H.T., H.C.-C., C.-Y.C., C.Y.T.Y., C.-J.L., R.P.P., L.-L.C., S.-C.R., J.-H.L., P.-J.L., H.-H.L., J.-W.S., P.C.H.H.), Academia Sinica, Taipei, Taiwan
| | - Jen-Hao Lin
- Institute of Biomedical Sciences (T.W.H.T., H.C.-C., C.-Y.C., C.Y.T.Y., C.-J.L., R.P.P., L.-L.C., S.-C.R., J.-H.L., P.-J.L., H.-H.L., J.-W.S., P.C.H.H.), Academia Sinica, Taipei, Taiwan
| | - Po-Ju Lin
- Institute of Biomedical Sciences (T.W.H.T., H.C.-C., C.-Y.C., C.Y.T.Y., C.-J.L., R.P.P., L.-L.C., S.-C.R., J.-H.L., P.-J.L., H.-H.L., J.-W.S., P.C.H.H.), Academia Sinica, Taipei, Taiwan
| | - Hsueh-Han Lu
- Institute of Biomedical Sciences (T.W.H.T., H.C.-C., C.-Y.C., C.Y.T.Y., C.-J.L., R.P.P., L.-L.C., S.-C.R., J.-H.L., P.-J.L., H.-H.L., J.-W.S., P.C.H.H.), Academia Sinica, Taipei, Taiwan
| | - Chiung-Wen Kuo
- Research Center for Applied Sciences (C.-W.K., P.C.), Academia Sinica, Taipei, Taiwan
| | - Cindy M. Chang
- NCKU Research and Development Foundation, Tainan, Taiwan (C.M.C.)
- Department of Medicine (C.M.C., A.D.H., T.A.H., T.J.K., P.C.H.H.), University of Wisconsin–Madison
| | - Alexander D. Hall
- Department of Medicine (C.M.C., A.D.H., T.A.H., T.J.K., P.C.H.H.), University of Wisconsin–Madison
| | - Eugenio I. Vivas
- Department of Bacteriology (E.I.V., F.E.R.), University of Wisconsin–Madison
| | - Jr-Wen Shui
- Institute of Biomedical Sciences (T.W.H.T., H.C.-C., C.-Y.C., C.Y.T.Y., C.-J.L., R.P.P., L.-L.C., S.-C.R., J.-H.L., P.-J.L., H.-H.L., J.-W.S., P.C.H.H.), Academia Sinica, Taipei, Taiwan
| | - Peilin Chen
- Research Center for Applied Sciences (C.-W.K., P.C.), Academia Sinica, Taipei, Taiwan
| | - Timothy A. Hacker
- Department of Medicine (C.M.C., A.D.H., T.A.H., T.J.K., P.C.H.H.), University of Wisconsin–Madison
| | - Federico E. Rey
- Department of Bacteriology (E.I.V., F.E.R.), University of Wisconsin–Madison
| | - Timothy J. Kamp
- Department of Medicine (C.M.C., A.D.H., T.A.H., T.J.K., P.C.H.H.), University of Wisconsin–Madison
- Stem Cell and Regenerative Medicine Center (T.J.K., P.C.H.H.), University of Wisconsin–Madison
| | - Patrick C.H. Hsieh
- Program in Molecular Medicine, National Yang Ming University and Academia Sinica, Taipei, Taiwan (T.W.H.T., P.C.C.H.)
- Institute of Biomedical Sciences (T.W.H.T., H.C.-C., C.-Y.C., C.Y.T.Y., C.-J.L., R.P.P., L.-L.C., S.-C.R., J.-H.L., P.-J.L., H.-H.L., J.-W.S., P.C.H.H.), Academia Sinica, Taipei, Taiwan
- Department of Medicine (C.M.C., A.D.H., T.A.H., T.J.K., P.C.H.H.), University of Wisconsin–Madison
- Stem Cell and Regenerative Medicine Center (T.J.K., P.C.H.H.), University of Wisconsin–Madison
| |
Collapse
|
19
|
Klyachkin YM, Idris A, Rodell CB, Tripathi H, Ye S, Nagareddy P, Asfour A, Gao E, Annabathula R, Ratajczak M, Burdick JA, Abdel-Latif A. Cathelicidin Related Antimicrobial Peptide (CRAMP) Enhances Bone Marrow Cell Retention and Attenuates Cardiac Dysfunction in a Mouse Model of Myocardial Infarction. Stem Cell Rev Rep 2018; 14:702-714. [PMID: 29948752 PMCID: PMC6119631 DOI: 10.1007/s12015-018-9833-x] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
BACKGROUND Acute myocardial infarction (MI) and the ensuing ischemic heart disease are approaching epidemic state. Unfortunately, no definitive therapies are available and human regenerative therapies have conflicting results. Limited stem cell retention following intracoronary administration has reduced the clinical efficacy of this novel therapy. Cathelicidin related antimicrobial peptides (CRAMPs) enhance chemotactic responsiveness of BMSPCs to low SDF-1 gradients, suggesting a potential role in BMSPCs engraftment. Here, we assessed the therapeutic efficacy of CRAMPs in the context of BMSPCs recruitment and retention via intracardiac delivery of CRAMP-treated BMSPCs or CRAMP-releasing hydrogels (HG) post-AMI. METHODS For cell transplantation experiments, mice were randomized into 3 groups: MI followed by injection of PBS, BMMNCs alone, and BMMNCs pre-incubated with CRAMP. During the in vivo HG studies, BM GFP chimera mice were randomized into 4 groups: MI followed by injection of HG alone, HG + SDF-1, HG + CRAMP, HG + SDF-1 + CRAMP. Changes in cardiac function at 5 weeks after MI were assessed using echocardiography. Angiogenesis was assessed using isolectin staining for capillary density. RESULTS Mice treated with BMMNCs pre-incubated with CRAMP had smaller scars, enhanced cardiac recovery and less adverse remodeling. Histologically, this group had higher capillary density. Similarly, sustained CRAMP release from hydrogels enhanced the therapeutic effect of SDF-1, leading to enhanced functional recovery, smaller scar size and higher capillary density. CONCLUSION Cathelicidins enhance BMMNC retention and recruitment after intramyocardial administration post-AMI resulting in improvements in heart physiology and recovery. Therapies employing these strategies may represent an attractive method for improving outcomes of regenerative therapies in human studies.
Collapse
Affiliation(s)
- Yuri M. Klyachkin
- Gill Heart Institute and Division of Cardiovascular Medicine, University of Kentucky, Lexington, KY and VA Medical Center, Lexington, KY, USA
| | - Amr Idris
- Gill Heart Institute and Division of Cardiovascular Medicine, University of Kentucky, Lexington, KY and VA Medical Center, Lexington, KY, USA
| | | | - Himi Tripathi
- Gill Heart Institute and Division of Cardiovascular Medicine, University of Kentucky, Lexington, KY and VA Medical Center, Lexington, KY, USA
| | - Shaojing Ye
- Gill Heart Institute and Division of Cardiovascular Medicine, University of Kentucky, Lexington, KY and VA Medical Center, Lexington, KY, USA
| | - Prabha Nagareddy
- Gill Heart Institute and Division of Cardiovascular Medicine, University of Kentucky, Lexington, KY and VA Medical Center, Lexington, KY, USA
| | - Ahmed Asfour
- Gill Heart Institute and Division of Cardiovascular Medicine, University of Kentucky, Lexington, KY and VA Medical Center, Lexington, KY, USA
| | - Erhe Gao
- The Center for Translational Medicine, Temple University School of Medicine, Philadelphia, PA, USA
| | - Rahul Annabathula
- Gill Heart Institute and Division of Cardiovascular Medicine, University of Kentucky, Lexington, KY and VA Medical Center, Lexington, KY, USA
| | - Mariusz Ratajczak
- Stem Cell Biology Institute, James Graham Brown Cancer Center, University of Louisville, Louisville, KY, USA
| | - Jason A. Burdick
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, USA
| | - Ahmed Abdel-Latif
- Gill Heart Institute and Division of Cardiovascular Medicine, University of Kentucky, Lexington, KY and VA Medical Center, Lexington, KY, USA
| |
Collapse
|
20
|
Goldstone AB, Burnett CE, Cohen JE, Paulsen MJ, Eskandari A, Edwards BE, Ingason AB, Steele AN, Patel JB, MacArthur JW, Shizuru JA, Woo YJ. SDF 1-alpha Attenuates Myocardial Injury Without Altering the Direct Contribution of Circulating Cells. J Cardiovasc Transl Res 2018; 11:274-284. [PMID: 29468554 PMCID: PMC6103912 DOI: 10.1007/s12265-017-9772-y] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/23/2017] [Accepted: 11/03/2017] [Indexed: 12/29/2022]
Abstract
Stromal cell-derived factor 1-alpha (SDF) is a potent bone marrow chemokine capable of recruiting circulating progenitor populations to injured tissue. SDF has known angiogenic capabilities, but bone marrow-derived cellular contributions to tissue regeneration remain controversial. Bone marrow from DsRed-transgenic donors was transplanted into recipients to lineage-trace circulating cells after myocardial infarction (MI). SDF was delivered post-MI, and hearts were evaluated for recruitment and plasticity of bone marrow-derived populations. SDF treatment improved ventricular function, border zone vessel density, and CD31+ cell frequency post-MI. Bone marrow-derived endothelial cells were observed; these cells arose through both cell fusion and transdifferentiation. Circulating cells also adopted cardiomyocyte fates, but such events were exceedingly rare and almost exclusively resulted from cell fusion. SDF did not significantly alter the proportion of circulating cells that adopted non-hematopoietic fates. Mechanistic insight into the governance of circulating cells is essential to realizing the full potential of cytokine therapies.
Collapse
Affiliation(s)
- Andrew B Goldstone
- Department of Cardiothoracic Surgery, School of Medicine, Stanford University, Stanford, CA, USA
| | - Cassandra E Burnett
- Division of Blood and Marrow Transplantation, Department of Medicine, School of Medicine, Stanford University, Stanford, CA, USA
| | - Jeffery E Cohen
- Department of Cardiothoracic Surgery, School of Medicine, Stanford University, Stanford, CA, USA
| | - Michael J Paulsen
- Department of Cardiothoracic Surgery, School of Medicine, Stanford University, Stanford, CA, USA
| | - Anahita Eskandari
- Department of Cardiothoracic Surgery, School of Medicine, Stanford University, Stanford, CA, USA
| | - Bryan E Edwards
- Department of Cardiothoracic Surgery, School of Medicine, Stanford University, Stanford, CA, USA
| | - Arnar B Ingason
- Department of Cardiothoracic Surgery, School of Medicine, Stanford University, Stanford, CA, USA
| | - Amanda N Steele
- Department of Cardiothoracic Surgery, School of Medicine, Stanford University, Stanford, CA, USA
| | - Jay B Patel
- Department of Cardiothoracic Surgery, School of Medicine, Stanford University, Stanford, CA, USA
| | - John W MacArthur
- Department of Cardiothoracic Surgery, School of Medicine, Stanford University, Stanford, CA, USA
| | - Judith A Shizuru
- Division of Blood and Marrow Transplantation, Department of Medicine, School of Medicine, Stanford University, Stanford, CA, USA
| | - Y Joseph Woo
- Department of Cardiothoracic Surgery, School of Medicine, Stanford University, Stanford, CA, USA.
| |
Collapse
|
21
|
Günthel M, Barnett P, Christoffels VM. Development, Proliferation, and Growth of the Mammalian Heart. Mol Ther 2018; 26:1599-1609. [PMID: 29929790 PMCID: PMC6037201 DOI: 10.1016/j.ymthe.2018.05.022] [Citation(s) in RCA: 78] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2018] [Revised: 05/24/2018] [Accepted: 05/25/2018] [Indexed: 01/01/2023] Open
Abstract
During development, the embryonic heart grows by addition of cells from a highly proliferative progenitor pool and by subsequent precisely controlled waves of cardiomyocyte proliferation. In this period, the heart can compensate for cardiomyocyte loss by an increased proliferation rate of the remaining cardiomyocytes. This proliferative capacity is lost soon after birth, with heart growth continuing by an increase in cardiomyocyte volume. The failure of the injured adult heart to regenerate often leads to the development of heart failure, a major cause of death. With the recent observation of a small fraction of cardiomyocytes that appear to have retained the proliferative capacity within the adult heart, as well as the identification of developmental pathways such as the Hippo-signaling pathway that can invoke mature cardiomyocyte proliferation, more studies are taking a knowledge-based mechanistic approach to heart regeneration. A key question being asked is if this knowledge can be used therapeutically to reinitiate cardiomyocyte proliferation after injury such as myocardial infarction. In this respect, uncovering and understanding the mechanisms and conditions that give rise to a fully functional and adaptive heart in the developing embryo could provide us with the answers to many of the questions that are now being asked.
Collapse
Affiliation(s)
- Marie Günthel
- Department of Medical Biology, Amsterdam Cardiovascular Sciences, Amsterdam UMC, Amsterdam, the Netherlands
| | - Phil Barnett
- Department of Medical Biology, Amsterdam Cardiovascular Sciences, Amsterdam UMC, Amsterdam, the Netherlands
| | - Vincent M Christoffels
- Department of Medical Biology, Amsterdam Cardiovascular Sciences, Amsterdam UMC, Amsterdam, the Netherlands.
| |
Collapse
|
22
|
Kröpfl JM, Tschakert G, Stelzer I, Pekovits K, Zelzer S, Dohr G, Holasek S, Stojakovic T, Scharnagl H, Spengler CM, Hofmann P. Acute Exercise-Induced Circulating Haematopoietic Stem and Progenitor Cells in Cardiac Patients - A Case Series. Heart Lung Circ 2018; 28:e54-e58. [PMID: 29933914 DOI: 10.1016/j.hlc.2018.05.095] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2017] [Revised: 04/18/2018] [Accepted: 05/01/2018] [Indexed: 02/07/2023]
Abstract
BACKGROUND Exercise-induced circulating haematopoietic stem and progenitor cell (HPC) number has been discussed in the context of regeneration in heart disease patients. OBJECTIVE The aim of this pilot study was to compare the effect of different exercise protocols usually applied in cardiac rehabilitation on the number of acute, exercise-induced HPCs, related to potential mediators, e.g. biomarkers of sympathetic and oxidative stress, and inflammation. METHODS This is a case series comprising seven patients suffering from coronary heart disease (CHD) undertaken at the Center for Ambulant Cardiac Rehabilitation. Patients (n=6) performed two exercise modes (constant-load, CLE; high-intensity interval, HIIE) in randomised order. Venous blood was drawn before and immediately after each test to assess CD34+/CD45+ HPC number by flow cytometry and biomarkers in blood plasma. The primary outcome was the change in HPC number, the secondary outcomes were changes in sympathetic/oxidative stress and markers of inflammation. RESULTS Both exercise modes resulted in a non-significant increase in HPC number after exercise, even when the results of both tests were combined. Overall, free norepinephrine increased significantly and was positively related to exercise-induced HPC number (r=0.70, p<0.05). Markers of sympathetic activation (fNE), oxidative stress (myeloperoxidase) and inflammation (interleukin-6) significantly increased after CLE and HIIE with no difference between tests. CONCLUSIONS Interestingly, acute CLE and HIIE did not stimulate significant HPC mobilisation in CHD, although both exercise modes elevated circulating concentrations of sympathetic activation. Haematopoietic stem and progenitor cell mobilisation could be blunted due to disease-related bone-marrow exhaustion.
Collapse
Affiliation(s)
- J M Kröpfl
- Exercise Physiology Lab, Institute of Human Movement Sciences and Sport, ETH Zurich, Zurich, Switzerland; Institute of Biophysics, Medical University of Graz, Graz, Austria.
| | - G Tschakert
- Exercise Physiology and Training Research Group, Institute of Sports Science, University of Graz, Graz, Austria
| | - I Stelzer
- Institute of Medical and Chemical Laboratory Diagnostics, LKH Hochsteiermark, Leoben, Austria
| | - K Pekovits
- Department of Ophthalmology, Medical University Graz, Graz, Austria
| | - S Zelzer
- Clinical Institute of Medical and Chemical Laboratory Diagnostics, Medical University of Graz, Graz, Austria
| | - G Dohr
- Institute of Cell Biology, Histology and Embryology, Medical University of Graz, Graz, Austria
| | - S Holasek
- Institute for Pathophysiology and Immunology, Medical University of Graz, Graz, Austria
| | - T Stojakovic
- Clinical Institute of Medical and Chemical Laboratory Diagnostics, Medical University of Graz, Graz, Austria
| | - H Scharnagl
- Clinical Institute of Medical and Chemical Laboratory Diagnostics, Medical University of Graz, Graz, Austria
| | - C M Spengler
- Exercise Physiology Lab, Institute of Human Movement Sciences and Sport, ETH Zurich, Zurich, Switzerland
| | - P Hofmann
- Exercise Physiology and Training Research Group, Institute of Sports Science, University of Graz, Graz, Austria
| |
Collapse
|
23
|
Kramann R, Machado F, Wu H, Kusaba T, Hoeft K, Schneider RK, Humphreys BD. Parabiosis and single-cell RNA sequencing reveal a limited contribution of monocytes to myofibroblasts in kidney fibrosis. JCI Insight 2018; 3:99561. [PMID: 29720573 DOI: 10.1172/jci.insight.99561] [Citation(s) in RCA: 73] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2018] [Accepted: 04/04/2018] [Indexed: 12/18/2022] Open
Abstract
Fibrosis is the common final pathway of virtually all chronic injury to the kidney. While it is well accepted that myofibroblasts are the scar-producing cells in the kidney, their cellular origin is still hotly debated. The relative contribution of proximal tubular epithelium and circulating cells, including mesenchymal stem cells, macrophages, and fibrocytes, to the myofibroblast pool remains highly controversial. Using inducible genetic fate tracing of proximal tubular epithelium, we confirm that the proximal tubule does not contribute to the myofibroblast pool. However, in parabiosis models in which one parabiont is genetically labeled and the other is unlabeled and undergoes kidney fibrosis, we demonstrate that a small fraction of genetically labeled renal myofibroblasts derive from the circulation. Single-cell RNA sequencing confirms this finding but indicates that these cells are circulating monocytes, express few extracellular matrix or other myofibroblast genes, and express many proinflammatory cytokines. We conclude that this small circulating myofibroblast progenitor population contributes to renal fibrosis by paracrine rather than direct mechanisms.
Collapse
Affiliation(s)
- Rafael Kramann
- Division of Nephrology and Clinical Immunology, RWTH Aachen University, Aachen, Germany
| | - Flavia Machado
- Division of Nephrology, Department of Medicine and Department of Cell Biology, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Haojia Wu
- Division of Nephrology, Department of Medicine and Department of Cell Biology, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Tetsuro Kusaba
- Department of Nephrology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Konrad Hoeft
- Division of Nephrology and Clinical Immunology, RWTH Aachen University, Aachen, Germany
| | - Rebekka K Schneider
- Department of Hematology, Erasmus MC Cancer Institute, Rotterdam, Netherlands.,Division of Hematology, RWTH Aachen University, Aachen, Germany
| | - Benjamin D Humphreys
- Division of Nephrology, Department of Medicine and Department of Cell Biology, Washington University School of Medicine, St. Louis, Missouri, USA
| |
Collapse
|
24
|
Xiang Q, Liao Y, Chao H, Huang W, Liu J, Chen H, Hong D, Zou Z, Xiang AP, Li W. ISL1 overexpression enhances the survival of transplanted human mesenchymal stem cells in a murine myocardial infarction model. Stem Cell Res Ther 2018; 9:51. [PMID: 29482621 PMCID: PMC5828309 DOI: 10.1186/s13287-018-0803-7] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2017] [Revised: 02/08/2018] [Accepted: 02/08/2018] [Indexed: 12/17/2022] Open
Abstract
BACKGROUND The LIM-homeobox transcription factor islet-1 (ISL1) has been proposed as a marker for cardiovascular progenitor cells. This study investigated whether forced expression of ISL1 in human mesenchymal stem cells (hMSCs) improves myocardial infarction (MI) treatment outcomes. METHODS The lentiviral vector containing the human elongation factor 1α promoter, which drives the expression of ISL1 (EF1α-ISL1), was constructed using the Multisite Gateway System and used to transduce hMSCs. Flow cytometry, immunofluorescence, Western blotting, TUNEL assay, and RNA sequencing were performed to evaluate the function of ISL1-overexpressing hMSCs (ISL1-hMSCs). RESULTS The in vivo results showed that transplantation of ISL1-hMSCs improved cardiac function in a rat model of MI. Left ventricle ejection fraction and fractional shortening were greater in post-MI hearts after 4 weeks of treatment with ISL1-hMSCs compared with control hMSCs or phosphate-buffered saline. We also found that ISL1 overexpression increased angiogenesis and decreased apoptosis and inflammation. The greater potential of ISL1-hMSCs may be attributable to an increased number of surviving cells after transplantation. Conditioned medium from ISL1-hMSCs decreased the apoptotic effect of H2O2 on the cardiomyocyte cell line H9c2. To clarify the molecular basis of this finding, we employed RNA sequencing to compare the apoptotic-related gene expression profiles of control hMSCs and ISL1-hMSCs. The results showed that insulin-like growth factor binding protein 3 (IGFBP3) was the only gene in ISL1-hMSCs with a RPKM value higher than 100 and that the difference fold-change between ISL1-hMSCs and control hMSCs was greater than 3, suggesting that IGFBP3 might play an important role in the anti-apoptosis effect of ISL1-hMSCs through paracrine effects. Furthermore, the expression of IGFBP3 in the conditioned medium from ISL1-hMSCs was almost fourfold greater than that in conditioned medium from control hMSCs. Moreover, the IGFBP3 neutralization antibody reversed the apoptotic effect of ISL1-hMSCs-CM. CONCLUSIONS These results suggest that overexpression of ISL1 in hMSCs promotes cell survival in a model of MI and enhances their paracrine function to protect cardiomyocytes, which may be mediated through IGFBP3. ISL1 overexpression in hMSCs may represent a novel strategy for enhancing the effectiveness of stem cell therapy after MI.
Collapse
Affiliation(s)
- Qiuling Xiang
- Center for Stem Cell Biology and Tissue Engineering, Key Laboratory for Stem Cells and Tissue Engineering, Ministry of Education, Sun Yat-sen University, Guangzhou, People's Republic of China.,Zhongshan Medical School, Sun Yat-sen University, Guangzhou, People's Republic of China
| | - Yan Liao
- Center for Stem Cell Biology and Tissue Engineering, Key Laboratory for Stem Cells and Tissue Engineering, Ministry of Education, Sun Yat-sen University, Guangzhou, People's Republic of China.,Zhongshan Medical School, Sun Yat-sen University, Guangzhou, People's Republic of China
| | - Hua Chao
- Zhongshan Medical School, Sun Yat-sen University, Guangzhou, People's Republic of China
| | - Weijun Huang
- Center for Stem Cell Biology and Tissue Engineering, Key Laboratory for Stem Cells and Tissue Engineering, Ministry of Education, Sun Yat-sen University, Guangzhou, People's Republic of China.,Zhongshan Medical School, Sun Yat-sen University, Guangzhou, People's Republic of China
| | - Jia Liu
- Department of Cardiology, the Red Cross hospital of Guangzhou City, the Fourth Affiliated Hospital of Jinan University, Guangzhou, People's Republic of China
| | - Haixuan Chen
- The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, People's Republic of China
| | - Dongxi Hong
- Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, People's Republic of China
| | - Zhengwei Zou
- Center for Stem Cell Biology and Tissue Engineering, Key Laboratory for Stem Cells and Tissue Engineering, Ministry of Education, Sun Yat-sen University, Guangzhou, People's Republic of China.,Zhongshan Medical School, Sun Yat-sen University, Guangzhou, People's Republic of China
| | - Andy Peng Xiang
- Center for Stem Cell Biology and Tissue Engineering, Key Laboratory for Stem Cells and Tissue Engineering, Ministry of Education, Sun Yat-sen University, Guangzhou, People's Republic of China.,Zhongshan Medical School, Sun Yat-sen University, Guangzhou, People's Republic of China
| | - Weiqiang Li
- Center for Stem Cell Biology and Tissue Engineering, Key Laboratory for Stem Cells and Tissue Engineering, Ministry of Education, Sun Yat-sen University, Guangzhou, People's Republic of China. .,Zhongshan Medical School, Sun Yat-sen University, Guangzhou, People's Republic of China.
| |
Collapse
|
25
|
Cesselli D, Aleksova A, Mazzega E, Caragnano A, Beltrami AP. Cardiac stem cell aging and heart failure. Pharmacol Res 2018; 127:26-32. [DOI: 10.1016/j.phrs.2017.01.013] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/25/2016] [Revised: 01/07/2017] [Accepted: 01/11/2017] [Indexed: 12/11/2022]
|
26
|
Single-cell analysis of the fate of c-kit-positive bone marrow cells. NPJ Regen Med 2017; 2:27. [PMID: 29302361 PMCID: PMC5678002 DOI: 10.1038/s41536-017-0032-1] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2017] [Revised: 09/08/2017] [Accepted: 09/19/2017] [Indexed: 01/14/2023] Open
Abstract
The plasticity of c-kit-positive bone marrow cells (c-kit-BMCs) in tissues different from their organ of origin remains unclear. We tested the hypothesis that c-kit-BMCs are functionally heterogeneous and only a subgroup of these cells possesses cardiomyogenic potential. Population-based assays fall short of identifying the properties of individual stem cells, imposing on us the introduction of single cell-based approaches to track the fate of c-kit-BMCs in the injured heart; they included viral gene-tagging, multicolor clonal-marking and transcriptional profiling. Based on these strategies, we report that single mouse c-kit-BMCs expand clonally within the infarcted myocardium and differentiate into specialized cardiac cells. Newly-formed cardiomyocytes, endothelial cells, fibroblasts and c-kit-BMCs showed in their genome common sites of viral integration, providing strong evidence in favor of the plasticity of a subset of BMCs expressing the c-kit receptor. Similarly, individual c-kit-BMCs, which were infected with multicolor reporters and injected in infarcted hearts, formed cardiomyocytes and vascular cells organized in clusters of similarly colored cells. The uniform distribution of fluorescent proteins in groups of specialized cells documented the polyclonal nature of myocardial regeneration. The transcriptional profile of myogenic c-kit-BMCs and whole c-kit-BMCs was defined by RNA sequencing. Genes relevant for engraftment, survival, migration, and differentiation were enriched in myogenic c-kit-BMCs, a cell subtype which could not be assigned to a specific hematopoietic lineage. Collectively, our findings demonstrate that the bone marrow comprises a category of cardiomyogenic, vasculogenic and/or fibrogenic c-kit-positive cells and a category of c-kit-positive cells that retains an undifferentiated state within the damaged heart.
Collapse
|
27
|
Eschenhagen T, Bolli R, Braun T, Field LJ, Fleischmann BK, Frisén J, Giacca M, Hare JM, Houser S, Lee RT, Marbán E, Martin JF, Molkentin JD, Murry CE, Riley PR, Ruiz-Lozano P, Sadek HA, Sussman MA, Hill JA. Cardiomyocyte Regeneration: A Consensus Statement. Circulation 2017; 136:680-686. [PMID: 28684531 PMCID: PMC5557671 DOI: 10.1161/circulationaha.117.029343] [Citation(s) in RCA: 377] [Impact Index Per Article: 47.1] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Affiliation(s)
- Thomas Eschenhagen
- From Department of Experimental Pharmacology and Toxicology, University Medical Center Hamburg Eppendorf, Hamburg, Germany (T.E.); DZHK (German Center for Cardiovascular Research), partner site Hamburg/Kiel/Lübeck, Hamburg, Germany (T.E.) and partner site Rhein/Main, Bad Nauheim, Germany (T.B.); Institute of Molecular Cardiology, University of Louisville, Louisville, KY (R.B.); Max-Planck-Institute for Heart and Lung Research, Bad Nauheim, Germany (T.B.); Department of Internal Medicine II, University of Giessen, Germany (T.B.); German Center for Lung Research (DZHL), Giessen/Marburg Bad Nauheim, Bad Nauheim, Germany (T.B.); Krannert Institute of Cardiology and Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis (L.J.F.); Institute of Physiology I, Life and Brain Center, Medical Faculty, University of Bonn, Germany (B.K.F.); Department of Cell and Molecular Biology, Karolinska Institute, Stockholm, Sweden (J.F.); International Centre for Genetic Engineering and Biotechnology (ICGEB), Trieste, Italy (M.G.); Donald Soffer Endowed Program in Regenerative Medicine, Miller School of Medicine, Miami, FL (J.M.H.); Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD (J.M.H.); Department of Physiology, Lewis Katz School of Medicine, Temple University, Philadelphia, PA (S.H.); Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA (R.T.L.); Cedars-Sinai Heart Institute, Los Angeles, CA (E.M.); Cardiomyocyte Renewal Laboratory, Texas Heart Institute, Houston (J.F.M.); Department of Molecular Physiology and Biophysics, Baylor College of Medicine, Houston, TX (J.F.M.); Department of Pediatrics, Cincinnati Children's Hospital Medical Center, University of Cincinnati, OH (J.D.M.); Departments of Pathology, Bioengineering, and Medicine/Cardiology, Institute for Stem Cell and Regenerative Medicine, and Center for Cardiovascular Biology, University of Washington, Seattle (C.E.M.); University of Oxford, Department of Physiology, Anatomy and Genetics, United Kingdom (P.R.R.) Regencor, Inc, Los Altos, CA (P.R.-L.); Departments of Internal Medicine (Division of Cardiology) and Molecular Biology, UT Southwestern Medical Center, Dallas, TX (H.A.S., J.A.H.); and Heart Institute, Integrated Regenerative Research Institute, and Biology Department, San Diego State University, CA (M.S.A.).
| | - Roberto Bolli
- From Department of Experimental Pharmacology and Toxicology, University Medical Center Hamburg Eppendorf, Hamburg, Germany (T.E.); DZHK (German Center for Cardiovascular Research), partner site Hamburg/Kiel/Lübeck, Hamburg, Germany (T.E.) and partner site Rhein/Main, Bad Nauheim, Germany (T.B.); Institute of Molecular Cardiology, University of Louisville, Louisville, KY (R.B.); Max-Planck-Institute for Heart and Lung Research, Bad Nauheim, Germany (T.B.); Department of Internal Medicine II, University of Giessen, Germany (T.B.); German Center for Lung Research (DZHL), Giessen/Marburg Bad Nauheim, Bad Nauheim, Germany (T.B.); Krannert Institute of Cardiology and Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis (L.J.F.); Institute of Physiology I, Life and Brain Center, Medical Faculty, University of Bonn, Germany (B.K.F.); Department of Cell and Molecular Biology, Karolinska Institute, Stockholm, Sweden (J.F.); International Centre for Genetic Engineering and Biotechnology (ICGEB), Trieste, Italy (M.G.); Donald Soffer Endowed Program in Regenerative Medicine, Miller School of Medicine, Miami, FL (J.M.H.); Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD (J.M.H.); Department of Physiology, Lewis Katz School of Medicine, Temple University, Philadelphia, PA (S.H.); Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA (R.T.L.); Cedars-Sinai Heart Institute, Los Angeles, CA (E.M.); Cardiomyocyte Renewal Laboratory, Texas Heart Institute, Houston (J.F.M.); Department of Molecular Physiology and Biophysics, Baylor College of Medicine, Houston, TX (J.F.M.); Department of Pediatrics, Cincinnati Children's Hospital Medical Center, University of Cincinnati, OH (J.D.M.); Departments of Pathology, Bioengineering, and Medicine/Cardiology, Institute for Stem Cell and Regenerative Medicine, and Center for Cardiovascular Biology, University of Washington, Seattle (C.E.M.); University of Oxford, Department of Physiology, Anatomy and Genetics, United Kingdom (P.R.R.) Regencor, Inc, Los Altos, CA (P.R.-L.); Departments of Internal Medicine (Division of Cardiology) and Molecular Biology, UT Southwestern Medical Center, Dallas, TX (H.A.S., J.A.H.); and Heart Institute, Integrated Regenerative Research Institute, and Biology Department, San Diego State University, CA (M.S.A.)
| | - Thomas Braun
- From Department of Experimental Pharmacology and Toxicology, University Medical Center Hamburg Eppendorf, Hamburg, Germany (T.E.); DZHK (German Center for Cardiovascular Research), partner site Hamburg/Kiel/Lübeck, Hamburg, Germany (T.E.) and partner site Rhein/Main, Bad Nauheim, Germany (T.B.); Institute of Molecular Cardiology, University of Louisville, Louisville, KY (R.B.); Max-Planck-Institute for Heart and Lung Research, Bad Nauheim, Germany (T.B.); Department of Internal Medicine II, University of Giessen, Germany (T.B.); German Center for Lung Research (DZHL), Giessen/Marburg Bad Nauheim, Bad Nauheim, Germany (T.B.); Krannert Institute of Cardiology and Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis (L.J.F.); Institute of Physiology I, Life and Brain Center, Medical Faculty, University of Bonn, Germany (B.K.F.); Department of Cell and Molecular Biology, Karolinska Institute, Stockholm, Sweden (J.F.); International Centre for Genetic Engineering and Biotechnology (ICGEB), Trieste, Italy (M.G.); Donald Soffer Endowed Program in Regenerative Medicine, Miller School of Medicine, Miami, FL (J.M.H.); Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD (J.M.H.); Department of Physiology, Lewis Katz School of Medicine, Temple University, Philadelphia, PA (S.H.); Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA (R.T.L.); Cedars-Sinai Heart Institute, Los Angeles, CA (E.M.); Cardiomyocyte Renewal Laboratory, Texas Heart Institute, Houston (J.F.M.); Department of Molecular Physiology and Biophysics, Baylor College of Medicine, Houston, TX (J.F.M.); Department of Pediatrics, Cincinnati Children's Hospital Medical Center, University of Cincinnati, OH (J.D.M.); Departments of Pathology, Bioengineering, and Medicine/Cardiology, Institute for Stem Cell and Regenerative Medicine, and Center for Cardiovascular Biology, University of Washington, Seattle (C.E.M.); University of Oxford, Department of Physiology, Anatomy and Genetics, United Kingdom (P.R.R.) Regencor, Inc, Los Altos, CA (P.R.-L.); Departments of Internal Medicine (Division of Cardiology) and Molecular Biology, UT Southwestern Medical Center, Dallas, TX (H.A.S., J.A.H.); and Heart Institute, Integrated Regenerative Research Institute, and Biology Department, San Diego State University, CA (M.S.A.)
| | - Loren J Field
- From Department of Experimental Pharmacology and Toxicology, University Medical Center Hamburg Eppendorf, Hamburg, Germany (T.E.); DZHK (German Center for Cardiovascular Research), partner site Hamburg/Kiel/Lübeck, Hamburg, Germany (T.E.) and partner site Rhein/Main, Bad Nauheim, Germany (T.B.); Institute of Molecular Cardiology, University of Louisville, Louisville, KY (R.B.); Max-Planck-Institute for Heart and Lung Research, Bad Nauheim, Germany (T.B.); Department of Internal Medicine II, University of Giessen, Germany (T.B.); German Center for Lung Research (DZHL), Giessen/Marburg Bad Nauheim, Bad Nauheim, Germany (T.B.); Krannert Institute of Cardiology and Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis (L.J.F.); Institute of Physiology I, Life and Brain Center, Medical Faculty, University of Bonn, Germany (B.K.F.); Department of Cell and Molecular Biology, Karolinska Institute, Stockholm, Sweden (J.F.); International Centre for Genetic Engineering and Biotechnology (ICGEB), Trieste, Italy (M.G.); Donald Soffer Endowed Program in Regenerative Medicine, Miller School of Medicine, Miami, FL (J.M.H.); Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD (J.M.H.); Department of Physiology, Lewis Katz School of Medicine, Temple University, Philadelphia, PA (S.H.); Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA (R.T.L.); Cedars-Sinai Heart Institute, Los Angeles, CA (E.M.); Cardiomyocyte Renewal Laboratory, Texas Heart Institute, Houston (J.F.M.); Department of Molecular Physiology and Biophysics, Baylor College of Medicine, Houston, TX (J.F.M.); Department of Pediatrics, Cincinnati Children's Hospital Medical Center, University of Cincinnati, OH (J.D.M.); Departments of Pathology, Bioengineering, and Medicine/Cardiology, Institute for Stem Cell and Regenerative Medicine, and Center for Cardiovascular Biology, University of Washington, Seattle (C.E.M.); University of Oxford, Department of Physiology, Anatomy and Genetics, United Kingdom (P.R.R.) Regencor, Inc, Los Altos, CA (P.R.-L.); Departments of Internal Medicine (Division of Cardiology) and Molecular Biology, UT Southwestern Medical Center, Dallas, TX (H.A.S., J.A.H.); and Heart Institute, Integrated Regenerative Research Institute, and Biology Department, San Diego State University, CA (M.S.A.)
| | - Bernd K Fleischmann
- From Department of Experimental Pharmacology and Toxicology, University Medical Center Hamburg Eppendorf, Hamburg, Germany (T.E.); DZHK (German Center for Cardiovascular Research), partner site Hamburg/Kiel/Lübeck, Hamburg, Germany (T.E.) and partner site Rhein/Main, Bad Nauheim, Germany (T.B.); Institute of Molecular Cardiology, University of Louisville, Louisville, KY (R.B.); Max-Planck-Institute for Heart and Lung Research, Bad Nauheim, Germany (T.B.); Department of Internal Medicine II, University of Giessen, Germany (T.B.); German Center for Lung Research (DZHL), Giessen/Marburg Bad Nauheim, Bad Nauheim, Germany (T.B.); Krannert Institute of Cardiology and Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis (L.J.F.); Institute of Physiology I, Life and Brain Center, Medical Faculty, University of Bonn, Germany (B.K.F.); Department of Cell and Molecular Biology, Karolinska Institute, Stockholm, Sweden (J.F.); International Centre for Genetic Engineering and Biotechnology (ICGEB), Trieste, Italy (M.G.); Donald Soffer Endowed Program in Regenerative Medicine, Miller School of Medicine, Miami, FL (J.M.H.); Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD (J.M.H.); Department of Physiology, Lewis Katz School of Medicine, Temple University, Philadelphia, PA (S.H.); Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA (R.T.L.); Cedars-Sinai Heart Institute, Los Angeles, CA (E.M.); Cardiomyocyte Renewal Laboratory, Texas Heart Institute, Houston (J.F.M.); Department of Molecular Physiology and Biophysics, Baylor College of Medicine, Houston, TX (J.F.M.); Department of Pediatrics, Cincinnati Children's Hospital Medical Center, University of Cincinnati, OH (J.D.M.); Departments of Pathology, Bioengineering, and Medicine/Cardiology, Institute for Stem Cell and Regenerative Medicine, and Center for Cardiovascular Biology, University of Washington, Seattle (C.E.M.); University of Oxford, Department of Physiology, Anatomy and Genetics, United Kingdom (P.R.R.) Regencor, Inc, Los Altos, CA (P.R.-L.); Departments of Internal Medicine (Division of Cardiology) and Molecular Biology, UT Southwestern Medical Center, Dallas, TX (H.A.S., J.A.H.); and Heart Institute, Integrated Regenerative Research Institute, and Biology Department, San Diego State University, CA (M.S.A.)
| | - Jonas Frisén
- From Department of Experimental Pharmacology and Toxicology, University Medical Center Hamburg Eppendorf, Hamburg, Germany (T.E.); DZHK (German Center for Cardiovascular Research), partner site Hamburg/Kiel/Lübeck, Hamburg, Germany (T.E.) and partner site Rhein/Main, Bad Nauheim, Germany (T.B.); Institute of Molecular Cardiology, University of Louisville, Louisville, KY (R.B.); Max-Planck-Institute for Heart and Lung Research, Bad Nauheim, Germany (T.B.); Department of Internal Medicine II, University of Giessen, Germany (T.B.); German Center for Lung Research (DZHL), Giessen/Marburg Bad Nauheim, Bad Nauheim, Germany (T.B.); Krannert Institute of Cardiology and Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis (L.J.F.); Institute of Physiology I, Life and Brain Center, Medical Faculty, University of Bonn, Germany (B.K.F.); Department of Cell and Molecular Biology, Karolinska Institute, Stockholm, Sweden (J.F.); International Centre for Genetic Engineering and Biotechnology (ICGEB), Trieste, Italy (M.G.); Donald Soffer Endowed Program in Regenerative Medicine, Miller School of Medicine, Miami, FL (J.M.H.); Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD (J.M.H.); Department of Physiology, Lewis Katz School of Medicine, Temple University, Philadelphia, PA (S.H.); Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA (R.T.L.); Cedars-Sinai Heart Institute, Los Angeles, CA (E.M.); Cardiomyocyte Renewal Laboratory, Texas Heart Institute, Houston (J.F.M.); Department of Molecular Physiology and Biophysics, Baylor College of Medicine, Houston, TX (J.F.M.); Department of Pediatrics, Cincinnati Children's Hospital Medical Center, University of Cincinnati, OH (J.D.M.); Departments of Pathology, Bioengineering, and Medicine/Cardiology, Institute for Stem Cell and Regenerative Medicine, and Center for Cardiovascular Biology, University of Washington, Seattle (C.E.M.); University of Oxford, Department of Physiology, Anatomy and Genetics, United Kingdom (P.R.R.) Regencor, Inc, Los Altos, CA (P.R.-L.); Departments of Internal Medicine (Division of Cardiology) and Molecular Biology, UT Southwestern Medical Center, Dallas, TX (H.A.S., J.A.H.); and Heart Institute, Integrated Regenerative Research Institute, and Biology Department, San Diego State University, CA (M.S.A.)
| | - Mauro Giacca
- From Department of Experimental Pharmacology and Toxicology, University Medical Center Hamburg Eppendorf, Hamburg, Germany (T.E.); DZHK (German Center for Cardiovascular Research), partner site Hamburg/Kiel/Lübeck, Hamburg, Germany (T.E.) and partner site Rhein/Main, Bad Nauheim, Germany (T.B.); Institute of Molecular Cardiology, University of Louisville, Louisville, KY (R.B.); Max-Planck-Institute for Heart and Lung Research, Bad Nauheim, Germany (T.B.); Department of Internal Medicine II, University of Giessen, Germany (T.B.); German Center for Lung Research (DZHL), Giessen/Marburg Bad Nauheim, Bad Nauheim, Germany (T.B.); Krannert Institute of Cardiology and Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis (L.J.F.); Institute of Physiology I, Life and Brain Center, Medical Faculty, University of Bonn, Germany (B.K.F.); Department of Cell and Molecular Biology, Karolinska Institute, Stockholm, Sweden (J.F.); International Centre for Genetic Engineering and Biotechnology (ICGEB), Trieste, Italy (M.G.); Donald Soffer Endowed Program in Regenerative Medicine, Miller School of Medicine, Miami, FL (J.M.H.); Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD (J.M.H.); Department of Physiology, Lewis Katz School of Medicine, Temple University, Philadelphia, PA (S.H.); Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA (R.T.L.); Cedars-Sinai Heart Institute, Los Angeles, CA (E.M.); Cardiomyocyte Renewal Laboratory, Texas Heart Institute, Houston (J.F.M.); Department of Molecular Physiology and Biophysics, Baylor College of Medicine, Houston, TX (J.F.M.); Department of Pediatrics, Cincinnati Children's Hospital Medical Center, University of Cincinnati, OH (J.D.M.); Departments of Pathology, Bioengineering, and Medicine/Cardiology, Institute for Stem Cell and Regenerative Medicine, and Center for Cardiovascular Biology, University of Washington, Seattle (C.E.M.); University of Oxford, Department of Physiology, Anatomy and Genetics, United Kingdom (P.R.R.) Regencor, Inc, Los Altos, CA (P.R.-L.); Departments of Internal Medicine (Division of Cardiology) and Molecular Biology, UT Southwestern Medical Center, Dallas, TX (H.A.S., J.A.H.); and Heart Institute, Integrated Regenerative Research Institute, and Biology Department, San Diego State University, CA (M.S.A.)
| | - Joshua M Hare
- From Department of Experimental Pharmacology and Toxicology, University Medical Center Hamburg Eppendorf, Hamburg, Germany (T.E.); DZHK (German Center for Cardiovascular Research), partner site Hamburg/Kiel/Lübeck, Hamburg, Germany (T.E.) and partner site Rhein/Main, Bad Nauheim, Germany (T.B.); Institute of Molecular Cardiology, University of Louisville, Louisville, KY (R.B.); Max-Planck-Institute for Heart and Lung Research, Bad Nauheim, Germany (T.B.); Department of Internal Medicine II, University of Giessen, Germany (T.B.); German Center for Lung Research (DZHL), Giessen/Marburg Bad Nauheim, Bad Nauheim, Germany (T.B.); Krannert Institute of Cardiology and Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis (L.J.F.); Institute of Physiology I, Life and Brain Center, Medical Faculty, University of Bonn, Germany (B.K.F.); Department of Cell and Molecular Biology, Karolinska Institute, Stockholm, Sweden (J.F.); International Centre for Genetic Engineering and Biotechnology (ICGEB), Trieste, Italy (M.G.); Donald Soffer Endowed Program in Regenerative Medicine, Miller School of Medicine, Miami, FL (J.M.H.); Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD (J.M.H.); Department of Physiology, Lewis Katz School of Medicine, Temple University, Philadelphia, PA (S.H.); Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA (R.T.L.); Cedars-Sinai Heart Institute, Los Angeles, CA (E.M.); Cardiomyocyte Renewal Laboratory, Texas Heart Institute, Houston (J.F.M.); Department of Molecular Physiology and Biophysics, Baylor College of Medicine, Houston, TX (J.F.M.); Department of Pediatrics, Cincinnati Children's Hospital Medical Center, University of Cincinnati, OH (J.D.M.); Departments of Pathology, Bioengineering, and Medicine/Cardiology, Institute for Stem Cell and Regenerative Medicine, and Center for Cardiovascular Biology, University of Washington, Seattle (C.E.M.); University of Oxford, Department of Physiology, Anatomy and Genetics, United Kingdom (P.R.R.) Regencor, Inc, Los Altos, CA (P.R.-L.); Departments of Internal Medicine (Division of Cardiology) and Molecular Biology, UT Southwestern Medical Center, Dallas, TX (H.A.S., J.A.H.); and Heart Institute, Integrated Regenerative Research Institute, and Biology Department, San Diego State University, CA (M.S.A.)
| | - Steven Houser
- From Department of Experimental Pharmacology and Toxicology, University Medical Center Hamburg Eppendorf, Hamburg, Germany (T.E.); DZHK (German Center for Cardiovascular Research), partner site Hamburg/Kiel/Lübeck, Hamburg, Germany (T.E.) and partner site Rhein/Main, Bad Nauheim, Germany (T.B.); Institute of Molecular Cardiology, University of Louisville, Louisville, KY (R.B.); Max-Planck-Institute for Heart and Lung Research, Bad Nauheim, Germany (T.B.); Department of Internal Medicine II, University of Giessen, Germany (T.B.); German Center for Lung Research (DZHL), Giessen/Marburg Bad Nauheim, Bad Nauheim, Germany (T.B.); Krannert Institute of Cardiology and Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis (L.J.F.); Institute of Physiology I, Life and Brain Center, Medical Faculty, University of Bonn, Germany (B.K.F.); Department of Cell and Molecular Biology, Karolinska Institute, Stockholm, Sweden (J.F.); International Centre for Genetic Engineering and Biotechnology (ICGEB), Trieste, Italy (M.G.); Donald Soffer Endowed Program in Regenerative Medicine, Miller School of Medicine, Miami, FL (J.M.H.); Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD (J.M.H.); Department of Physiology, Lewis Katz School of Medicine, Temple University, Philadelphia, PA (S.H.); Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA (R.T.L.); Cedars-Sinai Heart Institute, Los Angeles, CA (E.M.); Cardiomyocyte Renewal Laboratory, Texas Heart Institute, Houston (J.F.M.); Department of Molecular Physiology and Biophysics, Baylor College of Medicine, Houston, TX (J.F.M.); Department of Pediatrics, Cincinnati Children's Hospital Medical Center, University of Cincinnati, OH (J.D.M.); Departments of Pathology, Bioengineering, and Medicine/Cardiology, Institute for Stem Cell and Regenerative Medicine, and Center for Cardiovascular Biology, University of Washington, Seattle (C.E.M.); University of Oxford, Department of Physiology, Anatomy and Genetics, United Kingdom (P.R.R.) Regencor, Inc, Los Altos, CA (P.R.-L.); Departments of Internal Medicine (Division of Cardiology) and Molecular Biology, UT Southwestern Medical Center, Dallas, TX (H.A.S., J.A.H.); and Heart Institute, Integrated Regenerative Research Institute, and Biology Department, San Diego State University, CA (M.S.A.)
| | - Richard T Lee
- From Department of Experimental Pharmacology and Toxicology, University Medical Center Hamburg Eppendorf, Hamburg, Germany (T.E.); DZHK (German Center for Cardiovascular Research), partner site Hamburg/Kiel/Lübeck, Hamburg, Germany (T.E.) and partner site Rhein/Main, Bad Nauheim, Germany (T.B.); Institute of Molecular Cardiology, University of Louisville, Louisville, KY (R.B.); Max-Planck-Institute for Heart and Lung Research, Bad Nauheim, Germany (T.B.); Department of Internal Medicine II, University of Giessen, Germany (T.B.); German Center for Lung Research (DZHL), Giessen/Marburg Bad Nauheim, Bad Nauheim, Germany (T.B.); Krannert Institute of Cardiology and Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis (L.J.F.); Institute of Physiology I, Life and Brain Center, Medical Faculty, University of Bonn, Germany (B.K.F.); Department of Cell and Molecular Biology, Karolinska Institute, Stockholm, Sweden (J.F.); International Centre for Genetic Engineering and Biotechnology (ICGEB), Trieste, Italy (M.G.); Donald Soffer Endowed Program in Regenerative Medicine, Miller School of Medicine, Miami, FL (J.M.H.); Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD (J.M.H.); Department of Physiology, Lewis Katz School of Medicine, Temple University, Philadelphia, PA (S.H.); Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA (R.T.L.); Cedars-Sinai Heart Institute, Los Angeles, CA (E.M.); Cardiomyocyte Renewal Laboratory, Texas Heart Institute, Houston (J.F.M.); Department of Molecular Physiology and Biophysics, Baylor College of Medicine, Houston, TX (J.F.M.); Department of Pediatrics, Cincinnati Children's Hospital Medical Center, University of Cincinnati, OH (J.D.M.); Departments of Pathology, Bioengineering, and Medicine/Cardiology, Institute for Stem Cell and Regenerative Medicine, and Center for Cardiovascular Biology, University of Washington, Seattle (C.E.M.); University of Oxford, Department of Physiology, Anatomy and Genetics, United Kingdom (P.R.R.) Regencor, Inc, Los Altos, CA (P.R.-L.); Departments of Internal Medicine (Division of Cardiology) and Molecular Biology, UT Southwestern Medical Center, Dallas, TX (H.A.S., J.A.H.); and Heart Institute, Integrated Regenerative Research Institute, and Biology Department, San Diego State University, CA (M.S.A.)
| | - Eduardo Marbán
- From Department of Experimental Pharmacology and Toxicology, University Medical Center Hamburg Eppendorf, Hamburg, Germany (T.E.); DZHK (German Center for Cardiovascular Research), partner site Hamburg/Kiel/Lübeck, Hamburg, Germany (T.E.) and partner site Rhein/Main, Bad Nauheim, Germany (T.B.); Institute of Molecular Cardiology, University of Louisville, Louisville, KY (R.B.); Max-Planck-Institute for Heart and Lung Research, Bad Nauheim, Germany (T.B.); Department of Internal Medicine II, University of Giessen, Germany (T.B.); German Center for Lung Research (DZHL), Giessen/Marburg Bad Nauheim, Bad Nauheim, Germany (T.B.); Krannert Institute of Cardiology and Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis (L.J.F.); Institute of Physiology I, Life and Brain Center, Medical Faculty, University of Bonn, Germany (B.K.F.); Department of Cell and Molecular Biology, Karolinska Institute, Stockholm, Sweden (J.F.); International Centre for Genetic Engineering and Biotechnology (ICGEB), Trieste, Italy (M.G.); Donald Soffer Endowed Program in Regenerative Medicine, Miller School of Medicine, Miami, FL (J.M.H.); Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD (J.M.H.); Department of Physiology, Lewis Katz School of Medicine, Temple University, Philadelphia, PA (S.H.); Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA (R.T.L.); Cedars-Sinai Heart Institute, Los Angeles, CA (E.M.); Cardiomyocyte Renewal Laboratory, Texas Heart Institute, Houston (J.F.M.); Department of Molecular Physiology and Biophysics, Baylor College of Medicine, Houston, TX (J.F.M.); Department of Pediatrics, Cincinnati Children's Hospital Medical Center, University of Cincinnati, OH (J.D.M.); Departments of Pathology, Bioengineering, and Medicine/Cardiology, Institute for Stem Cell and Regenerative Medicine, and Center for Cardiovascular Biology, University of Washington, Seattle (C.E.M.); University of Oxford, Department of Physiology, Anatomy and Genetics, United Kingdom (P.R.R.) Regencor, Inc, Los Altos, CA (P.R.-L.); Departments of Internal Medicine (Division of Cardiology) and Molecular Biology, UT Southwestern Medical Center, Dallas, TX (H.A.S., J.A.H.); and Heart Institute, Integrated Regenerative Research Institute, and Biology Department, San Diego State University, CA (M.S.A.)
| | - James F Martin
- From Department of Experimental Pharmacology and Toxicology, University Medical Center Hamburg Eppendorf, Hamburg, Germany (T.E.); DZHK (German Center for Cardiovascular Research), partner site Hamburg/Kiel/Lübeck, Hamburg, Germany (T.E.) and partner site Rhein/Main, Bad Nauheim, Germany (T.B.); Institute of Molecular Cardiology, University of Louisville, Louisville, KY (R.B.); Max-Planck-Institute for Heart and Lung Research, Bad Nauheim, Germany (T.B.); Department of Internal Medicine II, University of Giessen, Germany (T.B.); German Center for Lung Research (DZHL), Giessen/Marburg Bad Nauheim, Bad Nauheim, Germany (T.B.); Krannert Institute of Cardiology and Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis (L.J.F.); Institute of Physiology I, Life and Brain Center, Medical Faculty, University of Bonn, Germany (B.K.F.); Department of Cell and Molecular Biology, Karolinska Institute, Stockholm, Sweden (J.F.); International Centre for Genetic Engineering and Biotechnology (ICGEB), Trieste, Italy (M.G.); Donald Soffer Endowed Program in Regenerative Medicine, Miller School of Medicine, Miami, FL (J.M.H.); Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD (J.M.H.); Department of Physiology, Lewis Katz School of Medicine, Temple University, Philadelphia, PA (S.H.); Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA (R.T.L.); Cedars-Sinai Heart Institute, Los Angeles, CA (E.M.); Cardiomyocyte Renewal Laboratory, Texas Heart Institute, Houston (J.F.M.); Department of Molecular Physiology and Biophysics, Baylor College of Medicine, Houston, TX (J.F.M.); Department of Pediatrics, Cincinnati Children's Hospital Medical Center, University of Cincinnati, OH (J.D.M.); Departments of Pathology, Bioengineering, and Medicine/Cardiology, Institute for Stem Cell and Regenerative Medicine, and Center for Cardiovascular Biology, University of Washington, Seattle (C.E.M.); University of Oxford, Department of Physiology, Anatomy and Genetics, United Kingdom (P.R.R.) Regencor, Inc, Los Altos, CA (P.R.-L.); Departments of Internal Medicine (Division of Cardiology) and Molecular Biology, UT Southwestern Medical Center, Dallas, TX (H.A.S., J.A.H.); and Heart Institute, Integrated Regenerative Research Institute, and Biology Department, San Diego State University, CA (M.S.A.)
| | - Jeffery D Molkentin
- From Department of Experimental Pharmacology and Toxicology, University Medical Center Hamburg Eppendorf, Hamburg, Germany (T.E.); DZHK (German Center for Cardiovascular Research), partner site Hamburg/Kiel/Lübeck, Hamburg, Germany (T.E.) and partner site Rhein/Main, Bad Nauheim, Germany (T.B.); Institute of Molecular Cardiology, University of Louisville, Louisville, KY (R.B.); Max-Planck-Institute for Heart and Lung Research, Bad Nauheim, Germany (T.B.); Department of Internal Medicine II, University of Giessen, Germany (T.B.); German Center for Lung Research (DZHL), Giessen/Marburg Bad Nauheim, Bad Nauheim, Germany (T.B.); Krannert Institute of Cardiology and Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis (L.J.F.); Institute of Physiology I, Life and Brain Center, Medical Faculty, University of Bonn, Germany (B.K.F.); Department of Cell and Molecular Biology, Karolinska Institute, Stockholm, Sweden (J.F.); International Centre for Genetic Engineering and Biotechnology (ICGEB), Trieste, Italy (M.G.); Donald Soffer Endowed Program in Regenerative Medicine, Miller School of Medicine, Miami, FL (J.M.H.); Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD (J.M.H.); Department of Physiology, Lewis Katz School of Medicine, Temple University, Philadelphia, PA (S.H.); Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA (R.T.L.); Cedars-Sinai Heart Institute, Los Angeles, CA (E.M.); Cardiomyocyte Renewal Laboratory, Texas Heart Institute, Houston (J.F.M.); Department of Molecular Physiology and Biophysics, Baylor College of Medicine, Houston, TX (J.F.M.); Department of Pediatrics, Cincinnati Children's Hospital Medical Center, University of Cincinnati, OH (J.D.M.); Departments of Pathology, Bioengineering, and Medicine/Cardiology, Institute for Stem Cell and Regenerative Medicine, and Center for Cardiovascular Biology, University of Washington, Seattle (C.E.M.); University of Oxford, Department of Physiology, Anatomy and Genetics, United Kingdom (P.R.R.) Regencor, Inc, Los Altos, CA (P.R.-L.); Departments of Internal Medicine (Division of Cardiology) and Molecular Biology, UT Southwestern Medical Center, Dallas, TX (H.A.S., J.A.H.); and Heart Institute, Integrated Regenerative Research Institute, and Biology Department, San Diego State University, CA (M.S.A.)
| | - Charles E Murry
- From Department of Experimental Pharmacology and Toxicology, University Medical Center Hamburg Eppendorf, Hamburg, Germany (T.E.); DZHK (German Center for Cardiovascular Research), partner site Hamburg/Kiel/Lübeck, Hamburg, Germany (T.E.) and partner site Rhein/Main, Bad Nauheim, Germany (T.B.); Institute of Molecular Cardiology, University of Louisville, Louisville, KY (R.B.); Max-Planck-Institute for Heart and Lung Research, Bad Nauheim, Germany (T.B.); Department of Internal Medicine II, University of Giessen, Germany (T.B.); German Center for Lung Research (DZHL), Giessen/Marburg Bad Nauheim, Bad Nauheim, Germany (T.B.); Krannert Institute of Cardiology and Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis (L.J.F.); Institute of Physiology I, Life and Brain Center, Medical Faculty, University of Bonn, Germany (B.K.F.); Department of Cell and Molecular Biology, Karolinska Institute, Stockholm, Sweden (J.F.); International Centre for Genetic Engineering and Biotechnology (ICGEB), Trieste, Italy (M.G.); Donald Soffer Endowed Program in Regenerative Medicine, Miller School of Medicine, Miami, FL (J.M.H.); Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD (J.M.H.); Department of Physiology, Lewis Katz School of Medicine, Temple University, Philadelphia, PA (S.H.); Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA (R.T.L.); Cedars-Sinai Heart Institute, Los Angeles, CA (E.M.); Cardiomyocyte Renewal Laboratory, Texas Heart Institute, Houston (J.F.M.); Department of Molecular Physiology and Biophysics, Baylor College of Medicine, Houston, TX (J.F.M.); Department of Pediatrics, Cincinnati Children's Hospital Medical Center, University of Cincinnati, OH (J.D.M.); Departments of Pathology, Bioengineering, and Medicine/Cardiology, Institute for Stem Cell and Regenerative Medicine, and Center for Cardiovascular Biology, University of Washington, Seattle (C.E.M.); University of Oxford, Department of Physiology, Anatomy and Genetics, United Kingdom (P.R.R.) Regencor, Inc, Los Altos, CA (P.R.-L.); Departments of Internal Medicine (Division of Cardiology) and Molecular Biology, UT Southwestern Medical Center, Dallas, TX (H.A.S., J.A.H.); and Heart Institute, Integrated Regenerative Research Institute, and Biology Department, San Diego State University, CA (M.S.A.)
| | - Paul R Riley
- From Department of Experimental Pharmacology and Toxicology, University Medical Center Hamburg Eppendorf, Hamburg, Germany (T.E.); DZHK (German Center for Cardiovascular Research), partner site Hamburg/Kiel/Lübeck, Hamburg, Germany (T.E.) and partner site Rhein/Main, Bad Nauheim, Germany (T.B.); Institute of Molecular Cardiology, University of Louisville, Louisville, KY (R.B.); Max-Planck-Institute for Heart and Lung Research, Bad Nauheim, Germany (T.B.); Department of Internal Medicine II, University of Giessen, Germany (T.B.); German Center for Lung Research (DZHL), Giessen/Marburg Bad Nauheim, Bad Nauheim, Germany (T.B.); Krannert Institute of Cardiology and Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis (L.J.F.); Institute of Physiology I, Life and Brain Center, Medical Faculty, University of Bonn, Germany (B.K.F.); Department of Cell and Molecular Biology, Karolinska Institute, Stockholm, Sweden (J.F.); International Centre for Genetic Engineering and Biotechnology (ICGEB), Trieste, Italy (M.G.); Donald Soffer Endowed Program in Regenerative Medicine, Miller School of Medicine, Miami, FL (J.M.H.); Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD (J.M.H.); Department of Physiology, Lewis Katz School of Medicine, Temple University, Philadelphia, PA (S.H.); Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA (R.T.L.); Cedars-Sinai Heart Institute, Los Angeles, CA (E.M.); Cardiomyocyte Renewal Laboratory, Texas Heart Institute, Houston (J.F.M.); Department of Molecular Physiology and Biophysics, Baylor College of Medicine, Houston, TX (J.F.M.); Department of Pediatrics, Cincinnati Children's Hospital Medical Center, University of Cincinnati, OH (J.D.M.); Departments of Pathology, Bioengineering, and Medicine/Cardiology, Institute for Stem Cell and Regenerative Medicine, and Center for Cardiovascular Biology, University of Washington, Seattle (C.E.M.); University of Oxford, Department of Physiology, Anatomy and Genetics, United Kingdom (P.R.R.) Regencor, Inc, Los Altos, CA (P.R.-L.); Departments of Internal Medicine (Division of Cardiology) and Molecular Biology, UT Southwestern Medical Center, Dallas, TX (H.A.S., J.A.H.); and Heart Institute, Integrated Regenerative Research Institute, and Biology Department, San Diego State University, CA (M.S.A.)
| | - Pilar Ruiz-Lozano
- From Department of Experimental Pharmacology and Toxicology, University Medical Center Hamburg Eppendorf, Hamburg, Germany (T.E.); DZHK (German Center for Cardiovascular Research), partner site Hamburg/Kiel/Lübeck, Hamburg, Germany (T.E.) and partner site Rhein/Main, Bad Nauheim, Germany (T.B.); Institute of Molecular Cardiology, University of Louisville, Louisville, KY (R.B.); Max-Planck-Institute for Heart and Lung Research, Bad Nauheim, Germany (T.B.); Department of Internal Medicine II, University of Giessen, Germany (T.B.); German Center for Lung Research (DZHL), Giessen/Marburg Bad Nauheim, Bad Nauheim, Germany (T.B.); Krannert Institute of Cardiology and Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis (L.J.F.); Institute of Physiology I, Life and Brain Center, Medical Faculty, University of Bonn, Germany (B.K.F.); Department of Cell and Molecular Biology, Karolinska Institute, Stockholm, Sweden (J.F.); International Centre for Genetic Engineering and Biotechnology (ICGEB), Trieste, Italy (M.G.); Donald Soffer Endowed Program in Regenerative Medicine, Miller School of Medicine, Miami, FL (J.M.H.); Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD (J.M.H.); Department of Physiology, Lewis Katz School of Medicine, Temple University, Philadelphia, PA (S.H.); Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA (R.T.L.); Cedars-Sinai Heart Institute, Los Angeles, CA (E.M.); Cardiomyocyte Renewal Laboratory, Texas Heart Institute, Houston (J.F.M.); Department of Molecular Physiology and Biophysics, Baylor College of Medicine, Houston, TX (J.F.M.); Department of Pediatrics, Cincinnati Children's Hospital Medical Center, University of Cincinnati, OH (J.D.M.); Departments of Pathology, Bioengineering, and Medicine/Cardiology, Institute for Stem Cell and Regenerative Medicine, and Center for Cardiovascular Biology, University of Washington, Seattle (C.E.M.); University of Oxford, Department of Physiology, Anatomy and Genetics, United Kingdom (P.R.R.) Regencor, Inc, Los Altos, CA (P.R.-L.); Departments of Internal Medicine (Division of Cardiology) and Molecular Biology, UT Southwestern Medical Center, Dallas, TX (H.A.S., J.A.H.); and Heart Institute, Integrated Regenerative Research Institute, and Biology Department, San Diego State University, CA (M.S.A.)
| | - Hesham A Sadek
- From Department of Experimental Pharmacology and Toxicology, University Medical Center Hamburg Eppendorf, Hamburg, Germany (T.E.); DZHK (German Center for Cardiovascular Research), partner site Hamburg/Kiel/Lübeck, Hamburg, Germany (T.E.) and partner site Rhein/Main, Bad Nauheim, Germany (T.B.); Institute of Molecular Cardiology, University of Louisville, Louisville, KY (R.B.); Max-Planck-Institute for Heart and Lung Research, Bad Nauheim, Germany (T.B.); Department of Internal Medicine II, University of Giessen, Germany (T.B.); German Center for Lung Research (DZHL), Giessen/Marburg Bad Nauheim, Bad Nauheim, Germany (T.B.); Krannert Institute of Cardiology and Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis (L.J.F.); Institute of Physiology I, Life and Brain Center, Medical Faculty, University of Bonn, Germany (B.K.F.); Department of Cell and Molecular Biology, Karolinska Institute, Stockholm, Sweden (J.F.); International Centre for Genetic Engineering and Biotechnology (ICGEB), Trieste, Italy (M.G.); Donald Soffer Endowed Program in Regenerative Medicine, Miller School of Medicine, Miami, FL (J.M.H.); Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD (J.M.H.); Department of Physiology, Lewis Katz School of Medicine, Temple University, Philadelphia, PA (S.H.); Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA (R.T.L.); Cedars-Sinai Heart Institute, Los Angeles, CA (E.M.); Cardiomyocyte Renewal Laboratory, Texas Heart Institute, Houston (J.F.M.); Department of Molecular Physiology and Biophysics, Baylor College of Medicine, Houston, TX (J.F.M.); Department of Pediatrics, Cincinnati Children's Hospital Medical Center, University of Cincinnati, OH (J.D.M.); Departments of Pathology, Bioengineering, and Medicine/Cardiology, Institute for Stem Cell and Regenerative Medicine, and Center for Cardiovascular Biology, University of Washington, Seattle (C.E.M.); University of Oxford, Department of Physiology, Anatomy and Genetics, United Kingdom (P.R.R.) Regencor, Inc, Los Altos, CA (P.R.-L.); Departments of Internal Medicine (Division of Cardiology) and Molecular Biology, UT Southwestern Medical Center, Dallas, TX (H.A.S., J.A.H.); and Heart Institute, Integrated Regenerative Research Institute, and Biology Department, San Diego State University, CA (M.S.A.)
| | - Mark A Sussman
- From Department of Experimental Pharmacology and Toxicology, University Medical Center Hamburg Eppendorf, Hamburg, Germany (T.E.); DZHK (German Center for Cardiovascular Research), partner site Hamburg/Kiel/Lübeck, Hamburg, Germany (T.E.) and partner site Rhein/Main, Bad Nauheim, Germany (T.B.); Institute of Molecular Cardiology, University of Louisville, Louisville, KY (R.B.); Max-Planck-Institute for Heart and Lung Research, Bad Nauheim, Germany (T.B.); Department of Internal Medicine II, University of Giessen, Germany (T.B.); German Center for Lung Research (DZHL), Giessen/Marburg Bad Nauheim, Bad Nauheim, Germany (T.B.); Krannert Institute of Cardiology and Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis (L.J.F.); Institute of Physiology I, Life and Brain Center, Medical Faculty, University of Bonn, Germany (B.K.F.); Department of Cell and Molecular Biology, Karolinska Institute, Stockholm, Sweden (J.F.); International Centre for Genetic Engineering and Biotechnology (ICGEB), Trieste, Italy (M.G.); Donald Soffer Endowed Program in Regenerative Medicine, Miller School of Medicine, Miami, FL (J.M.H.); Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD (J.M.H.); Department of Physiology, Lewis Katz School of Medicine, Temple University, Philadelphia, PA (S.H.); Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA (R.T.L.); Cedars-Sinai Heart Institute, Los Angeles, CA (E.M.); Cardiomyocyte Renewal Laboratory, Texas Heart Institute, Houston (J.F.M.); Department of Molecular Physiology and Biophysics, Baylor College of Medicine, Houston, TX (J.F.M.); Department of Pediatrics, Cincinnati Children's Hospital Medical Center, University of Cincinnati, OH (J.D.M.); Departments of Pathology, Bioengineering, and Medicine/Cardiology, Institute for Stem Cell and Regenerative Medicine, and Center for Cardiovascular Biology, University of Washington, Seattle (C.E.M.); University of Oxford, Department of Physiology, Anatomy and Genetics, United Kingdom (P.R.R.) Regencor, Inc, Los Altos, CA (P.R.-L.); Departments of Internal Medicine (Division of Cardiology) and Molecular Biology, UT Southwestern Medical Center, Dallas, TX (H.A.S., J.A.H.); and Heart Institute, Integrated Regenerative Research Institute, and Biology Department, San Diego State University, CA (M.S.A.)
| | - Joseph A Hill
- From Department of Experimental Pharmacology and Toxicology, University Medical Center Hamburg Eppendorf, Hamburg, Germany (T.E.); DZHK (German Center for Cardiovascular Research), partner site Hamburg/Kiel/Lübeck, Hamburg, Germany (T.E.) and partner site Rhein/Main, Bad Nauheim, Germany (T.B.); Institute of Molecular Cardiology, University of Louisville, Louisville, KY (R.B.); Max-Planck-Institute for Heart and Lung Research, Bad Nauheim, Germany (T.B.); Department of Internal Medicine II, University of Giessen, Germany (T.B.); German Center for Lung Research (DZHL), Giessen/Marburg Bad Nauheim, Bad Nauheim, Germany (T.B.); Krannert Institute of Cardiology and Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis (L.J.F.); Institute of Physiology I, Life and Brain Center, Medical Faculty, University of Bonn, Germany (B.K.F.); Department of Cell and Molecular Biology, Karolinska Institute, Stockholm, Sweden (J.F.); International Centre for Genetic Engineering and Biotechnology (ICGEB), Trieste, Italy (M.G.); Donald Soffer Endowed Program in Regenerative Medicine, Miller School of Medicine, Miami, FL (J.M.H.); Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD (J.M.H.); Department of Physiology, Lewis Katz School of Medicine, Temple University, Philadelphia, PA (S.H.); Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA (R.T.L.); Cedars-Sinai Heart Institute, Los Angeles, CA (E.M.); Cardiomyocyte Renewal Laboratory, Texas Heart Institute, Houston (J.F.M.); Department of Molecular Physiology and Biophysics, Baylor College of Medicine, Houston, TX (J.F.M.); Department of Pediatrics, Cincinnati Children's Hospital Medical Center, University of Cincinnati, OH (J.D.M.); Departments of Pathology, Bioengineering, and Medicine/Cardiology, Institute for Stem Cell and Regenerative Medicine, and Center for Cardiovascular Biology, University of Washington, Seattle (C.E.M.); University of Oxford, Department of Physiology, Anatomy and Genetics, United Kingdom (P.R.R.) Regencor, Inc, Los Altos, CA (P.R.-L.); Departments of Internal Medicine (Division of Cardiology) and Molecular Biology, UT Southwestern Medical Center, Dallas, TX (H.A.S., J.A.H.); and Heart Institute, Integrated Regenerative Research Institute, and Biology Department, San Diego State University, CA (M.S.A.).
| |
Collapse
|
28
|
Patterson M, Barske L, Van Handel B, Rau CD, Gan P, Sharma A, Parikh S, Denholtz M, Huang Y, Yamaguchi Y, Shen H, Allayee H, Crump JG, Force TI, Lien CL, Makita T, Lusis AJ, Kumar SR, Sucov HM. Frequency of mononuclear diploid cardiomyocytes underlies natural variation in heart regeneration. Nat Genet 2017; 49:1346-1353. [PMID: 28783163 DOI: 10.1038/ng.3929] [Citation(s) in RCA: 252] [Impact Index Per Article: 31.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2017] [Accepted: 07/11/2017] [Indexed: 12/16/2022]
Abstract
Adult mammalian cardiomyocyte regeneration after injury is thought to be minimal. Mononuclear diploid cardiomyocytes (MNDCMs), a relatively small subpopulation in the adult heart, may account for the observed degree of regeneration, but this has not been tested. We surveyed 120 inbred mouse strains and found that the frequency of adult mononuclear cardiomyocytes was surprisingly variable (>7-fold). Cardiomyocyte proliferation and heart functional recovery after coronary artery ligation both correlated with pre-injury MNDCM content. Using genome-wide association, we identified Tnni3k as one gene that influences variation in this composition and demonstrated that Tnni3k knockout resulted in elevated MNDCM content and increased cardiomyocyte proliferation after injury. Reciprocally, overexpression of Tnni3k in zebrafish promoted cardiomyocyte polyploidization and compromised heart regeneration. Our results corroborate the relevance of MNDCMs in heart regeneration. Moreover, they imply that intrinsic heart regeneration is not limited nor uniform in all individuals, but rather is a variable trait influenced by multiple genes.
Collapse
Affiliation(s)
- Michaela Patterson
- Department of Stem Cell Biology and Regenerative Medicine, Keck School of Medicine, University of Southern California, Los Angeles, California, USA
| | - Lindsey Barske
- Department of Stem Cell Biology and Regenerative Medicine, Keck School of Medicine, University of Southern California, Los Angeles, California, USA
| | - Ben Van Handel
- Department of Stem Cell Biology and Regenerative Medicine, Keck School of Medicine, University of Southern California, Los Angeles, California, USA
| | - Christoph D Rau
- Department of Human Genetics, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California, USA
| | - Peiheng Gan
- Department of Stem Cell Biology and Regenerative Medicine, Keck School of Medicine, University of Southern California, Los Angeles, California, USA
| | - Avneesh Sharma
- Department of Stem Cell Biology and Regenerative Medicine, Keck School of Medicine, University of Southern California, Los Angeles, California, USA
| | - Shan Parikh
- Division of Cardiovascular Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Matt Denholtz
- Department of Biological Chemistry, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California, USA
| | - Ying Huang
- Program of Developmental Biology and Regenerative Medicine, The Saban Research Institute, Children's Hospital Los Angeles, Los Angeles, California, USA
| | - Yukiko Yamaguchi
- Department of Stem Cell Biology and Regenerative Medicine, Keck School of Medicine, University of Southern California, Los Angeles, California, USA
| | - Hua Shen
- Department of Stem Cell Biology and Regenerative Medicine, Keck School of Medicine, University of Southern California, Los Angeles, California, USA
| | - Hooman Allayee
- Department of Preventive Medicine, Keck School of Medicine, University of Southern California, Los Angeles, California, USA
| | - J Gage Crump
- Department of Stem Cell Biology and Regenerative Medicine, Keck School of Medicine, University of Southern California, Los Angeles, California, USA
| | - Thomas I Force
- Division of Cardiovascular Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Ching-Ling Lien
- Program of Developmental Biology and Regenerative Medicine, The Saban Research Institute, Children's Hospital Los Angeles, Los Angeles, California, USA.,Department of Surgery, Keck School of Medicine, University of Southern California, Los Angeles, California, USA
| | - Takako Makita
- Developmental Neuroscience Program, The Saban Research Institute, Children's Hospital Los Angeles, Los Angeles, California, USA.,Department of Pediatrics, Keck School of Medicine, University of Southern California, Los Angeles, California, USA
| | - Aldons J Lusis
- Department of Human Genetics, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California, USA
| | - S Ram Kumar
- Department of Surgery, Keck School of Medicine, University of Southern California, Los Angeles, California, USA
| | - Henry M Sucov
- Department of Stem Cell Biology and Regenerative Medicine, Keck School of Medicine, University of Southern California, Los Angeles, California, USA
| |
Collapse
|
29
|
Doppler SA, Lange R, Laugwitz KL, Krane M. Cardiac development: from current understanding to new regenerative concepts. J Thorac Dis 2017; 9:S1-S4. [PMID: 28446962 DOI: 10.21037/jtd.2017.03.131] [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)
- Stefanie A Doppler
- Department of Cardiovascular Surgery, Division of Experimental Surgery, German Heart Center Munich, Technische Universität München, Munich, Germany
| | - Rüdiger Lange
- Department of Cardiovascular Surgery, Division of Experimental Surgery, German Heart Center Munich, Technische Universität München, Munich, Germany.,DZHK (German Center for Cardiovascular Research), partner site Munich Heart Alliance, Munich, Germany
| | - Karl-Ludwig Laugwitz
- DZHK (German Center for Cardiovascular Research), partner site Munich Heart Alliance, Munich, Germany.,I. Department of Medicine (Cardiology), Klinikum rechts der Isar der Technischen Universität München, Munich, Germany
| | - Markus Krane
- Department of Cardiovascular Surgery, Division of Experimental Surgery, German Heart Center Munich, Technische Universität München, Munich, Germany.,DZHK (German Center for Cardiovascular Research), partner site Munich Heart Alliance, Munich, Germany
| |
Collapse
|
30
|
Hao M, Wang R, Wang W. Cell Therapies in Cardiomyopathy: Current Status of Clinical Trials. Anal Cell Pathol (Amst) 2017; 2017:9404057. [PMID: 28194324 PMCID: PMC5282433 DOI: 10.1155/2017/9404057] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2016] [Revised: 12/06/2016] [Accepted: 12/08/2016] [Indexed: 12/28/2022] Open
Abstract
Because the human heart has limited potential for regeneration, the loss of cardiomyocytes during cardiac myopathy and ischaemic injury can result in heart failure and death. Stem cell therapy has emerged as a promising strategy for the treatment of dead myocardium, directly or indirectly, and seems to offer functional benefits to patients. The ideal candidate donor cell for myocardial reconstitution is a stem-like cell that can be easily obtained, has a robust proliferation capacity and a low risk of tumour formation and immune rejection, differentiates into functionally normal cardiomyocytes, and is suitable for minimally invasive clinical transplantation. The ultimate goal of cardiac repair is to regenerate functionally viable myocardium after myocardial infarction (MI) to prevent or heal heart failure. This review provides a comprehensive overview of treatment with stem-like cells in preclinical and clinical studies to assess the feasibility and efficacy of this novel therapeutic strategy in ischaemic cardiomyopathy.
Collapse
Affiliation(s)
- Ming Hao
- Cellular Biomedicine Group, 333 Guiping Road, Shanghai 200233, China
- Cellular Biomedicine Group, 19925 Stevens Creek Blvd, Suite 100, Cupertino, CA 95014, USA
| | - Richard Wang
- Cellular Biomedicine Group, 333 Guiping Road, Shanghai 200233, China
- Cellular Biomedicine Group, 19925 Stevens Creek Blvd, Suite 100, Cupertino, CA 95014, USA
| | - Wen Wang
- Cellular Biomedicine Group, 333 Guiping Road, Shanghai 200233, China
- Cellular Biomedicine Group, 19925 Stevens Creek Blvd, Suite 100, Cupertino, CA 95014, USA
| |
Collapse
|
31
|
Cheng B, Chen HC, Chou IW, Tang TWH, Hsieh PCH. Harnessing the early post-injury inflammatory responses for cardiac regeneration. J Biomed Sci 2017; 24:7. [PMID: 28086885 PMCID: PMC5237143 DOI: 10.1186/s12929-017-0315-2] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2016] [Accepted: 01/04/2017] [Indexed: 12/13/2022] Open
Abstract
Cardiac inflammation is considered by many as the main driving force in prolonging the pathological condition in the heart after myocardial infarction. Immediately after cardiac ischemic injury, neutrophils are the first innate immune cells recruited to the ischemic myocardium within the first 24 h. Once they have infiltrated the injured myocardium, neutrophils would then secret proteases that promote cardiac remodeling and chemokines that enhance the recruitment of monocytes from the spleen, in which the recruitment peaks at 72 h after myocardial infarction. Monocytes would transdifferentiate into macrophages after transmigrating into the infarct area. Both neutrophils and monocytes-derived macrophages are known to release proteases and cytokines that are detrimental to the surviving cardiomyocytes. Paradoxically, these inflammatory cells also play critical roles in repairing the injured myocardium. Depletion of either neutrophils or monocytes do not improve overall cardiac function after myocardial infarction. Instead, the left ventricular function is further impaired and cardiac fibrosis persists. Moreover, the inflammatory microenvironment created by the infiltrated neutrophils and monocytes-derived macrophages is essential for the recruitment of cardiac progenitor cells. Recent studies also suggest that treatment with anti-inflammatory drugs may cause cardiac dysfunction after injury. Indeed, clinical studies have shown that traditional ant-inflammatory strategies are ineffective to improve cardiac function after infarction. Thus, the focus should be on how to harness these inflammatory events to either improve the efficacy of the delivered drugs or to favor the recruitment of cardiac progenitor cells.
Collapse
Affiliation(s)
- Bill Cheng
- Institute of Biomedical Sciences, Academia Sinica, 128 Academia Road, Sec. 2, Nankang District, Taipei, 115, Taiwan
| | - H C Chen
- Institute of Biomedical Sciences, Academia Sinica, 128 Academia Road, Sec. 2, Nankang District, Taipei, 115, Taiwan
| | - I W Chou
- Institute of Biomedical Sciences, Academia Sinica, 128 Academia Road, Sec. 2, Nankang District, Taipei, 115, Taiwan.,Graduate Institute of Life Sciences, National Defence Medical Center, Taipei, 114, Taiwan
| | - Tony W H Tang
- Institute of Biomedical Sciences, Academia Sinica, 128 Academia Road, Sec. 2, Nankang District, Taipei, 115, Taiwan.,Program in Molecular Medicine, National Yang Ming University, Taipei, 112, Taiwan
| | - Patrick C H Hsieh
- Institute of Biomedical Sciences, Academia Sinica, 128 Academia Road, Sec. 2, Nankang District, Taipei, 115, Taiwan. .,Graduate Institute of Life Sciences, National Defence Medical Center, Taipei, 114, Taiwan. .,Program in Molecular Medicine, National Yang Ming University, Taipei, 112, Taiwan. .,Graduate Institute of Medical Genomics and Proteomics, and Institute of Clinical Medicine, College of Medicine, National Taiwan University, Taipei, 100, Taiwan. .,Department of Surgery, National Taiwan University Hospital, Taipei, 100, Taiwan.
| |
Collapse
|
32
|
Yucel D, Kocabas F. Developments in Hematopoietic Stem Cell Expansion and Gene Editing Technologies. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2017; 1079:103-125. [DOI: 10.1007/5584_2017_114] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
|
33
|
Xiang MSW, Kikuchi K. Endogenous Mechanisms of Cardiac Regeneration. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2016; 326:67-131. [PMID: 27572127 DOI: 10.1016/bs.ircmb.2016.04.002] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Zebrafish possess a remarkable capacity for cardiac regeneration throughout their lifetime, providing a model for investigating endogenous cellular and molecular mechanisms regulating myocardial regeneration. By contrast, adult mammals have an extremely limited capacity for cardiac regeneration, contributing to mortality and morbidity from cardiac diseases such as myocardial infarction and heart failure. However, the viewpoint of the mammalian heart as a postmitotic organ was recently revised based on findings that the mammalian heart contains multiple undifferentiated cell types with cardiogenic potential as well as a robust regenerative capacity during a short period early in life. Although it occurs at an extremely low level, continuous cardiomyocyte turnover has been detected in adult mouse and human hearts, which could potentially be enhanced to restore lost myocardium in damaged human hearts. This review summarizes and discusses recent advances in the understanding of endogenous mechanisms of cardiac regeneration.
Collapse
Affiliation(s)
- M S W Xiang
- Developmental and Stem Cell Biology Division, Victor Chang Cardiac Research Institute, Darlinghurst NSW, Australia
| | - K Kikuchi
- Developmental and Stem Cell Biology Division, Victor Chang Cardiac Research Institute, Darlinghurst NSW, Australia; St. Vincent's Clinical School, University of New South Wales, Kensington NSW, Australia.
| |
Collapse
|
34
|
Golpanian S, Wolf A, Hatzistergos KE, Hare JM. Rebuilding the Damaged Heart: Mesenchymal Stem Cells, Cell-Based Therapy, and Engineered Heart Tissue. Physiol Rev 2016; 96:1127-68. [PMID: 27335447 PMCID: PMC6345247 DOI: 10.1152/physrev.00019.2015] [Citation(s) in RCA: 250] [Impact Index Per Article: 27.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Mesenchymal stem cells (MSCs) are broadly distributed cells that retain postnatal capacity for self-renewal and multilineage differentiation. MSCs evade immune detection, secrete an array of anti-inflammatory and anti-fibrotic mediators, and very importantly activate resident precursors. These properties form the basis for the strategy of clinical application of cell-based therapeutics for inflammatory and fibrotic conditions. In cardiovascular medicine, administration of autologous or allogeneic MSCs in patients with ischemic and nonischemic cardiomyopathy holds significant promise. Numerous preclinical studies of ischemic and nonischemic cardiomyopathy employing MSC-based therapy have demonstrated that the properties of reducing fibrosis, stimulating angiogenesis, and cardiomyogenesis have led to improvements in the structure and function of remodeled ventricles. Further attempts have been made to augment MSCs' effects through genetic modification and cell preconditioning. Progression of MSC therapy to early clinical trials has supported their role in improving cardiac structure and function, functional capacity, and patient quality of life. Emerging data have supported larger clinical trials that have been either completed or are currently underway. Mechanistically, MSC therapy is thought to benefit the heart by stimulating innate anti-fibrotic and regenerative responses. The mechanisms of action involve paracrine signaling, cell-cell interactions, and fusion with resident cells. Trans-differentiation of MSCs to bona fide cardiomyocytes and coronary vessels is also thought to occur, although at a nonphysiological level. Recently, MSC-based tissue engineering for cardiovascular disease has been examined with quite encouraging results. This review discusses MSCs from their basic biological characteristics to their role as a promising therapeutic strategy for clinical cardiovascular disease.
Collapse
Affiliation(s)
- Samuel Golpanian
- Interdisciplinary Stem Cell Institute, Department of Medicine, and Department of Surgery, University of Miami Miller School of Medicine, Miami, Florida
| | - Ariel Wolf
- Interdisciplinary Stem Cell Institute, Department of Medicine, and Department of Surgery, University of Miami Miller School of Medicine, Miami, Florida
| | - Konstantinos E Hatzistergos
- Interdisciplinary Stem Cell Institute, Department of Medicine, and Department of Surgery, University of Miami Miller School of Medicine, Miami, Florida
| | - Joshua M Hare
- Interdisciplinary Stem Cell Institute, Department of Medicine, and Department of Surgery, University of Miami Miller School of Medicine, Miami, Florida
| |
Collapse
|
35
|
Hatzistergos KE, Hare JM. Murine Models Demonstrate Distinct Vasculogenic and Cardiomyogenic cKit+ Lineages in the Heart. Circ Res 2016; 118:382-7. [PMID: 26846638 DOI: 10.1161/circresaha.115.308061] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
After 2 recent genetic studies in mice addressing the developmental origins and regenerative activity of cardiac cKit+ cells, 2 additional reports by Sultana et al and Liu et al provide further information on the expression of cKit in the embryonic and adult hearts. Here, we synthesize the findings from the 4 distinct cKit models to gain insights into the biology of this important cell type.
Collapse
Affiliation(s)
- Konstantinos E Hatzistergos
- From the Interdisciplinary Stem Cell Institute (K.E.H.), and Department of Medicine, Division of Cardiology and Department of Molecular and Cellular Pharmacology (J.M.H.), Leonard M. Miller School of Medicine, University of Miami, FL
| | - Joshua M Hare
- From the Interdisciplinary Stem Cell Institute (K.E.H.), and Department of Medicine, Division of Cardiology and Department of Molecular and Cellular Pharmacology (J.M.H.), Leonard M. Miller School of Medicine, University of Miami, FL.
| |
Collapse
|
36
|
Williams R, The Editors. Circulation Research “In This Issue” Anthology. Circ Res 2016. [DOI: 10.1161/res.0000000000000108] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
|
37
|
Zaglia T, Di Bona A, Chioato T, Basso C, Ausoni S, Mongillo M. Optimized protocol for immunostaining of experimental GFP-expressing and human hearts. Histochem Cell Biol 2016; 146:407-19. [PMID: 27311322 DOI: 10.1007/s00418-016-1456-1] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/07/2016] [Indexed: 02/07/2023]
Abstract
Morphological and histochemical analysis of the heart is fundamental for the understanding of cardiac physiology and pathology. The accurate detection of different myocardial cell populations, as well as the high-resolution imaging of protein expression and distribution, within the diverse intracellular compartments, is essential for basic research on disease mechanisms and for the translatability of the results to human pathophysiology. While enormous progress has been made on the imaging hardware and methods and on biotechnological tools [e.g., use of green fluorescent protein (GFP), viral-mediated gene transduction] to investigate heart cell structure and function, most of the protocols to prepare heart tissue samples for analysis have remained almost identical for decades. We here provide a detailed description of a novel protocol of heart processing, tailored to the simultaneous detection of tissue morphology, immunofluorescence markers and native emission of fluorescent proteins (i.e., GFP). We compared a variety of procedures of fixation, antigen unmasking and tissue permeabilization, to identify the best combination for preservation of myocardial morphology and native GFP fluorescence, while simultaneously allowing detection of antibody staining toward sarcomeric, membrane, cytosolic and nuclear markers. Furthermore, with minimal variations, we implemented such protocol for the study of human heart samples, including those already fixed and stored with conventional procedures, in tissue archives or bio-banks. In conclusion, a procedure is here presented for the laboratory investigation of the heart, in both rodents and humans, which accrues from the same tissue section information that would normally require the time-consuming and tissue-wasting observation of multiple serial sections.
Collapse
Affiliation(s)
- Tania Zaglia
- Department of Biomedical Sciences, University of Padova, Via Ugo Bassi 58/b, 35133, Padua, Italy. .,Venetian Institute of Molecular Medicine (VIMM), Via Orus 2, 35129, Padua, Italy.
| | - Anna Di Bona
- Venetian Institute of Molecular Medicine (VIMM), Via Orus 2, 35129, Padua, Italy.,Department of Cardiac, Thoracic and Vascular Sciences, University of Padua, Via A. Gabelli, 61, 35121, Padua, Italy
| | | | - Cristina Basso
- Department of Cardiac, Thoracic and Vascular Sciences, University of Padua, Via A. Gabelli, 61, 35121, Padua, Italy
| | - Simonetta Ausoni
- Department of Biomedical Sciences, University of Padova, Via Ugo Bassi 58/b, 35133, Padua, Italy
| | - Marco Mongillo
- Department of Biomedical Sciences, University of Padova, Via Ugo Bassi 58/b, 35133, Padua, Italy.,Venetian Institute of Molecular Medicine (VIMM), Via Orus 2, 35129, Padua, Italy.,CNR Institute of Neuroscience, Viale G. Colombo 3, 35121, Padua, Italy
| |
Collapse
|
38
|
Kanda M, Nagai T, Takahashi T, Liu ML, Kondou N, Naito AT, Akazawa H, Sashida G, Iwama A, Komuro I, Kobayashi Y. Leukemia Inhibitory Factor Enhances Endogenous Cardiomyocyte Regeneration after Myocardial Infarction. PLoS One 2016; 11:e0156562. [PMID: 27227407 PMCID: PMC4881916 DOI: 10.1371/journal.pone.0156562] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2016] [Accepted: 04/22/2016] [Indexed: 12/20/2022] Open
Abstract
Cardiac stem cells or precursor cells regenerate cardiomyocytes; however, the mechanism underlying this effect remains unclear. We generated CreLacZ mice in which more than 99.9% of the cardiomyocytes in the left ventricular field were positive for 5-bromo-4-chloro-3-indolyl-β-d-galactoside (X-gal) staining immediately after tamoxifen injection. Three months after myocardial infarction (MI), the MI mice had more X-gal-negative (newly generated) cells than the control mice (3.04 ± 0.38/mm2, MI; 0.47 ± 0.16/mm2, sham; p < 0.05). The cardiac side population (CSP) cell fraction contained label-retaining cells, which differentiated into X-gal-negative cardiomyocytes after MI. We injected a leukemia inhibitory factor (LIF)-expression construct at the time of MI and identified a significant functional improvement in the LIF-treated group. At 1 month after MI, in the MI border and scar area, the LIF-injected mice had 31.41 ± 5.83 X-gal-negative cardiomyocytes/mm2, whereas the control mice had 12.34 ± 2.56 X-gal-negative cardiomyocytes/mm2 (p < 0.05). Using 5-ethynyl-2'-deoxyurinide (EdU) administration after MI, the percentages of EdU-positive CSP cells in the LIF-treated and control mice were 29.4 ± 2.7% and 10.6 ± 3.7%, respectively, which suggests that LIF influenced CSP proliferation. Moreover, LIF activated the Janus kinase (JAK)signal transducer and activator of transcription (STAT), mitogen-activated protein kinase/extracellular signal-regulated (MEK)extracellular signal-regulated kinase (ERK), and phosphatidylinositol 3-kinase (PI3K)–AKT pathways in CSPs in vivo and in vitro. The enhanced green fluorescent protein (EGFP)-bone marrow-chimeric CreLacZ mouse results indicated that LIF did not stimulate cardiogenesis via circulating bone marrow-derived cells during the 4 weeks following MI. Thus, LIF stimulates, in part, stem cell-derived cardiomyocyte regeneration by activating cardiac stem or precursor cells. This approach may represent a novel therapeutic strategy for cardiogenesis.
Collapse
Affiliation(s)
- Masato Kanda
- Department of Cardiovascular Medicine, Chiba University Graduate School of Medicine, Chiba, Japan
| | - Toshio Nagai
- Department of Cardiovascular Medicine, Chiba University Graduate School of Medicine, Chiba, Japan
- * E-mail:
| | - Toshinao Takahashi
- Department of Cardiovascular Medicine, Chiba University Graduate School of Medicine, Chiba, Japan
| | - Mei Lan Liu
- Department of Cardiovascular Medicine, Chiba University Graduate School of Medicine, Chiba, Japan
| | - Naomichi Kondou
- Department of Cardiovascular Medicine, Chiba University Graduate School of Medicine, Chiba, Japan
| | - Atsuhiko T. Naito
- Department of Cardiovascular Medicine, The University of Tokyo Graduate School of Medicine, Tokyo, Japan
| | - Hiroshi Akazawa
- Department of Cardiovascular Medicine, The University of Tokyo Graduate School of Medicine, Tokyo, Japan
| | - Goro Sashida
- Department of Cellular and Molecular Medicine, Chiba University Graduate School of Medicine, Chiba, Japan
| | - Atsushi Iwama
- Department of Cellular and Molecular Medicine, Chiba University Graduate School of Medicine, Chiba, Japan
| | - Issei Komuro
- Department of Cardiovascular Medicine, The University of Tokyo Graduate School of Medicine, Tokyo, Japan
| | - Yoshio Kobayashi
- Department of Cardiovascular Medicine, Chiba University Graduate School of Medicine, Chiba, Japan
| |
Collapse
|
39
|
RhoA determines lineage fate of mesenchymal stem cells by modulating CTGF-VEGF complex in extracellular matrix. Nat Commun 2016; 7:11455. [PMID: 27126736 PMCID: PMC4855537 DOI: 10.1038/ncomms11455] [Citation(s) in RCA: 59] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2015] [Accepted: 03/21/2016] [Indexed: 12/26/2022] Open
Abstract
Mesenchymal stem cells (MSCs) participate in the repair/remodelling of many tissues, where MSCs commit to different lineages dependent on the cues in the local microenvironment. Here we show that TGFβ-activated RhoA/ROCK signalling functions as a molecular switch regarding the fate of MSCs in arterial repair/remodelling after injury. MSCs differentiate into myofibroblasts when RhoA/ROCK is turned on, endothelial cells when turned off. The former is pathophysiologic resulting in intimal hyperplasia, whereas the latter is physiological leading to endothelial repair. Further analysis revealed that MSC RhoA activation promotes formation of an extracellular matrix (ECM) complex consisting of connective tissue growth factor (CTGF) and vascular endothelial growth factor (VEGF). Inactivation of RhoA/ROCK in MSCs induces matrix metalloproteinase-3-mediated CTGF cleavage, resulting in VEGF release and MSC endothelial differentiation. Our findings uncover a novel mechanism by which cell–ECM interactions determine stem cell lineage specificity and offer additional molecular targets to manipulate MSC-involved tissue repair/regeneration. It is unclear what regulates the fate of mesenchymal stem cells (MSCs) in arterial repair following injury. Here, the authors show that MSC differentiation following injury is triggered by RhoA which in turn stimulates the release of connective tissue growth factor and vascular endothelial growth factor.
Collapse
|
40
|
Affiliation(s)
- Konstantinos E Hatzistergos
- From the Department of Medicine, Division of Cardiology (J.M.H.), Department of Molecular and Cellular Pharmacology (J.M.H.), and Interdisciplinary Stem Cell Institute (K.E.H., J.M.H.), Leonard M. Miller School of Medicine, University of Miami, FL
| | - Joshua M Hare
- From the Department of Medicine, Division of Cardiology (J.M.H.), Department of Molecular and Cellular Pharmacology (J.M.H.), and Interdisciplinary Stem Cell Institute (K.E.H., J.M.H.), Leonard M. Miller School of Medicine, University of Miami, FL.
| |
Collapse
|
41
|
Finan A, Richard S. Stimulating endogenous cardiac repair. Front Cell Dev Biol 2015; 3:57. [PMID: 26484341 PMCID: PMC4586501 DOI: 10.3389/fcell.2015.00057] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2015] [Accepted: 09/08/2015] [Indexed: 01/10/2023] Open
Abstract
The healthy adult heart has a low turnover of cardiac myocytes. The renewal capacity, however, is augmented after cardiac injury. Participants in cardiac regeneration include cardiac myocytes themselves, cardiac progenitor cells, and peripheral stem cells, particularly from the bone marrow compartment. Cardiac progenitor cells and bone marrow stem cells are augmented after cardiac injury, migrate to the myocardium, and support regeneration. Depletion studies of these populations have demonstrated their necessary role in cardiac repair. However, the potential of these cells to completely regenerate the heart is limited. Efforts are now being focused on ways to augment these natural pathways to improve cardiac healing, primarily after ischemic injury but in other cardiac pathologies as well. Cell and gene therapy or pharmacological interventions are proposed mechanisms. Cell therapy has demonstrated modest results and has passed into clinical trials. However, the beneficial effects of cell therapy have primarily been their ability to produce paracrine effects on the cardiac tissue and recruit endogenous stem cell populations as opposed to direct cardiac regeneration. Gene therapy efforts have focused on prolonging or reactivating natural signaling pathways. Positive results have been demonstrated to activate the endogenous stem cell populations and are currently being tested in clinical trials. A potential new avenue may be to refine pharmacological treatments that are currently in place in the clinic. Evidence is mounting that drugs such as statins or beta blockers may alter endogenous stem cell activity. Understanding the effects of these drugs on stem cell repair while keeping in mind their primary function may strike a balance in myocardial healing. To maximize endogenous cardiac regeneration, a combination of these approaches could ameliorate the overall repair process to incorporate the participation of multiple cellular players.
Collapse
Affiliation(s)
- Amanda Finan
- Centre National de la Recherche Scientifique United Medical Resource 9214, Institut National de la Santé et de la Recherche Médicale U1046, Physiology and Experimental Medicine of the Heart and Muscles, University of Montpellier Montpellier, France
| | - Sylvain Richard
- Centre National de la Recherche Scientifique United Medical Resource 9214, Institut National de la Santé et de la Recherche Médicale U1046, Physiology and Experimental Medicine of the Heart and Muscles, University of Montpellier Montpellier, France
| |
Collapse
|
42
|
Quijada P, Salunga HT, Hariharan N, Cubillo JD, El-Sayed FG, Moshref M, Bala KM, Emathinger JM, De La Torre A, Ormachea L, Alvarez R, Gude NA, Sussman MA. Cardiac Stem Cell Hybrids Enhance Myocardial Repair. Circ Res 2015; 117:695-706. [PMID: 26228030 DOI: 10.1161/circresaha.115.306838] [Citation(s) in RCA: 67] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/12/2015] [Accepted: 07/29/2015] [Indexed: 02/07/2023]
Abstract
RATIONALE Dual cell transplantation of cardiac progenitor cells (CPCs) and mesenchymal stem cells (MSCs) after infarction improves myocardial repair and performance in large animal models relative to delivery of either cell population. OBJECTIVE To demonstrate that CardioChimeras (CCs) formed by fusion between CPCs and MSCs have enhanced reparative potential in a mouse model of myocardial infarction relative to individual stem cells or combined cell delivery. METHODS AND RESULTS Two distinct and clonally derived CCs, CC1 and CC2, were used for this study. CCs improved left ventricular anterior wall thickness at 4 weeks post injury, but only CC1 treatment preserved anterior wall thickness at 18 weeks. Ejection fraction was enhanced at 6 weeks in CCs, and functional improvements were maintained in CCs and CPC+MSC groups at 18 weeks. Infarct size was decreased in CCs, whereas CPC+MSC and CPC parent groups remained unchanged at 12 weeks. CCs exhibited increased persistence, engraftment, and expression of early commitment markers within the border zone relative to combinatorial and individual cell population-injected groups. CCs increased capillary density and preserved cardiomyocyte size in the infarcted regions suggesting CCs role in protective paracrine secretion. CONCLUSIONS CCs merge the application of distinct cells into a single entity for cellular therapeutic intervention in the progression of heart failure. CCs are a novel cell therapy that improves on combinatorial cell approaches to support myocardial regeneration.
Collapse
Affiliation(s)
- Pearl Quijada
- From the Integrated Regenerative Research Institute, Department of Biology, San Diego State University, CA (P.Q., H.T.S., J.D.C., F.G.E.-S., M.M., K.M.B., J.M.E., A.D.L.T., L.O., R.A., N.A.G., M.A.S.); and Department of Pharmacology, University of California at Davis (N.H.)
| | - Hazel T Salunga
- From the Integrated Regenerative Research Institute, Department of Biology, San Diego State University, CA (P.Q., H.T.S., J.D.C., F.G.E.-S., M.M., K.M.B., J.M.E., A.D.L.T., L.O., R.A., N.A.G., M.A.S.); and Department of Pharmacology, University of California at Davis (N.H.)
| | - Nirmala Hariharan
- From the Integrated Regenerative Research Institute, Department of Biology, San Diego State University, CA (P.Q., H.T.S., J.D.C., F.G.E.-S., M.M., K.M.B., J.M.E., A.D.L.T., L.O., R.A., N.A.G., M.A.S.); and Department of Pharmacology, University of California at Davis (N.H.)
| | - Jonathan D Cubillo
- From the Integrated Regenerative Research Institute, Department of Biology, San Diego State University, CA (P.Q., H.T.S., J.D.C., F.G.E.-S., M.M., K.M.B., J.M.E., A.D.L.T., L.O., R.A., N.A.G., M.A.S.); and Department of Pharmacology, University of California at Davis (N.H.)
| | - Farid G El-Sayed
- From the Integrated Regenerative Research Institute, Department of Biology, San Diego State University, CA (P.Q., H.T.S., J.D.C., F.G.E.-S., M.M., K.M.B., J.M.E., A.D.L.T., L.O., R.A., N.A.G., M.A.S.); and Department of Pharmacology, University of California at Davis (N.H.)
| | - Maryam Moshref
- From the Integrated Regenerative Research Institute, Department of Biology, San Diego State University, CA (P.Q., H.T.S., J.D.C., F.G.E.-S., M.M., K.M.B., J.M.E., A.D.L.T., L.O., R.A., N.A.G., M.A.S.); and Department of Pharmacology, University of California at Davis (N.H.)
| | - Kristin M Bala
- From the Integrated Regenerative Research Institute, Department of Biology, San Diego State University, CA (P.Q., H.T.S., J.D.C., F.G.E.-S., M.M., K.M.B., J.M.E., A.D.L.T., L.O., R.A., N.A.G., M.A.S.); and Department of Pharmacology, University of California at Davis (N.H.)
| | - Jacqueline M Emathinger
- From the Integrated Regenerative Research Institute, Department of Biology, San Diego State University, CA (P.Q., H.T.S., J.D.C., F.G.E.-S., M.M., K.M.B., J.M.E., A.D.L.T., L.O., R.A., N.A.G., M.A.S.); and Department of Pharmacology, University of California at Davis (N.H.)
| | - Andrea De La Torre
- From the Integrated Regenerative Research Institute, Department of Biology, San Diego State University, CA (P.Q., H.T.S., J.D.C., F.G.E.-S., M.M., K.M.B., J.M.E., A.D.L.T., L.O., R.A., N.A.G., M.A.S.); and Department of Pharmacology, University of California at Davis (N.H.)
| | - Lucia Ormachea
- From the Integrated Regenerative Research Institute, Department of Biology, San Diego State University, CA (P.Q., H.T.S., J.D.C., F.G.E.-S., M.M., K.M.B., J.M.E., A.D.L.T., L.O., R.A., N.A.G., M.A.S.); and Department of Pharmacology, University of California at Davis (N.H.)
| | - Roberto Alvarez
- From the Integrated Regenerative Research Institute, Department of Biology, San Diego State University, CA (P.Q., H.T.S., J.D.C., F.G.E.-S., M.M., K.M.B., J.M.E., A.D.L.T., L.O., R.A., N.A.G., M.A.S.); and Department of Pharmacology, University of California at Davis (N.H.)
| | - Natalie A Gude
- From the Integrated Regenerative Research Institute, Department of Biology, San Diego State University, CA (P.Q., H.T.S., J.D.C., F.G.E.-S., M.M., K.M.B., J.M.E., A.D.L.T., L.O., R.A., N.A.G., M.A.S.); and Department of Pharmacology, University of California at Davis (N.H.)
| | - Mark A Sussman
- From the Integrated Regenerative Research Institute, Department of Biology, San Diego State University, CA (P.Q., H.T.S., J.D.C., F.G.E.-S., M.M., K.M.B., J.M.E., A.D.L.T., L.O., R.A., N.A.G., M.A.S.); and Department of Pharmacology, University of California at Davis (N.H.).
| |
Collapse
|
43
|
Affiliation(s)
- Pearl Quijada
- From the Department of Biology, The Integrated Regenerative Research Institute, San Diego State University, CA
| | - Mark A Sussman
- From the Department of Biology, The Integrated Regenerative Research Institute, San Diego State University, CA.
| |
Collapse
|