1
|
Garmany A, Arrell DK, Yamada S, Jeon R, Behfar A, Park S, Terzic A. Decoded cardiopoietic cell secretome linkage to heart repair biosignature. Stem Cells Transl Med 2024; 13:1144-1159. [PMID: 39259666 PMCID: PMC11555478 DOI: 10.1093/stcltm/szae067] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2024] [Accepted: 08/03/2024] [Indexed: 09/13/2024] Open
Abstract
Cardiopoiesis-primed human stem cells exert sustained benefit in treating heart failure despite limited retention following myocardial delivery. To assess potential paracrine contribution, the secretome of cardiopoiesis conditioned versus naïve human mesenchymal stromal cells was decoded by directed proteomics augmented with machine learning and systems interrogation. Cardiopoiesis doubled cellular protein output generating a distinct secretome that segregated the conditioned state. Altering the expression of 1035 secreted proteins, cardiopoiesis reshaped the secretome across functional classes. The resolved differential cardiopoietic secretome was enriched in mesoderm development and cardiac progenitor signaling processes, yielding a cardiovasculogenic profile bolstered by upregulated cardiogenic proteins. In tandem, cardiopoiesis enhanced the secretion of immunomodulatory proteins associated with cytokine signaling, leukocyte migration, and chemotaxis. Network analysis integrated the differential secretome within an interactome of 1745 molecules featuring prioritized regenerative processes. Secretome contribution to the repair signature of cardiopoietic cell-treated infarcted hearts was assessed in a murine coronary ligation model. Intramyocardial delivery of cardiopoietic cells improved the performance of failing hearts, with undirected proteomics revealing 50 myocardial proteins responsive to cell therapy. Pathway analysis linked the secretome to cardiac proteome remodeling, pinpointing 17 cardiopoiesis-upregulated secretome proteins directly upstream of 44% of the cell therapy-responsive cardiac proteome. Knockout, in silico, of this 22-protein secretome-dependent myocardial ensemble eliminated indices of the repair signature. Accordingly, in vivo, cell therapy rendered the secretome-dependent myocardial proteome of an infarcted heart indiscernible from healthy counterparts. Thus, the secretagogue effect of cardiopoiesis transforms the human stem cell secretome, endows regenerative competency, and upregulates candidate paracrine effectors of cell therapy-mediated molecular restitution.
Collapse
Affiliation(s)
- Armin Garmany
- Department of Cardiovascular Medicine, Mayo Clinic, Rochester, MN, United States
- Center for Regenerative Biotherapeutics, Mayo Clinic, Rochester, MN, United States
- Marriott Heart Disease Research Program, Mayo Clinic, Rochester, MN, United States
- Mayo Clinic Alix School of Medicine, Regenerative Sciences Track, Mayo Clinic Graduate School of Biomedical Sciences, Mayo Clinic, Rochester, MN, United States
| | - D Kent Arrell
- Department of Cardiovascular Medicine, Mayo Clinic, Rochester, MN, United States
- Center for Regenerative Biotherapeutics, Mayo Clinic, Rochester, MN, United States
- Marriott Heart Disease Research Program, Mayo Clinic, Rochester, MN, United States
| | - Satsuki Yamada
- Department of Cardiovascular Medicine, Mayo Clinic, Rochester, MN, United States
- Center for Regenerative Biotherapeutics, Mayo Clinic, Rochester, MN, United States
- Marriott Heart Disease Research Program, Mayo Clinic, Rochester, MN, United States
- Section of Geriatric Medicine & Gerontology, Department of Medicine, Mayo Clinic, Rochester, MN, United States
| | - Ryounghoon Jeon
- Department of Cardiovascular Medicine, Mayo Clinic, Rochester, MN, United States
- Center for Regenerative Biotherapeutics, Mayo Clinic, Rochester, MN, United States
- Marriott Heart Disease Research Program, Mayo Clinic, Rochester, MN, United States
| | - Atta Behfar
- Department of Cardiovascular Medicine, Mayo Clinic, Rochester, MN, United States
- Center for Regenerative Biotherapeutics, Mayo Clinic, Rochester, MN, United States
- Van Cleve Cardiac Regenerative Medicine Program, Mayo Clinic, Rochester, MN, United States
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN, United States
| | - Sungjo Park
- Department of Cardiovascular Medicine, Mayo Clinic, Rochester, MN, United States
- Center for Regenerative Biotherapeutics, Mayo Clinic, Rochester, MN, United States
- Marriott Heart Disease Research Program, Mayo Clinic, Rochester, MN, United States
| | - Andre Terzic
- Department of Cardiovascular Medicine, Mayo Clinic, Rochester, MN, United States
- Center for Regenerative Biotherapeutics, Mayo Clinic, Rochester, MN, United States
- Marriott Heart Disease Research Program, Mayo Clinic, Rochester, MN, United States
- Department of Molecular Pharmacology & Experimental Therapeutics, Mayo Clinic, Rochester, MN, United States
- Department of Medical Genetics, Mayo Clinic, Rochester, MN, United States
| |
Collapse
|
2
|
Riebel LL, Wang ZJ, Martinez-Navarro H, Trovato C, Camps J, Berg LA, Zhou X, Doste R, Sachetto Oliveira R, Weber Dos Santos R, Biasetti J, Rodriguez B. In silico evaluation of cell therapy in acute versus chronic infarction: role of automaticity, heterogeneity and Purkinje in human. Sci Rep 2024; 14:21584. [PMID: 39284812 PMCID: PMC11405404 DOI: 10.1038/s41598-024-67951-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2024] [Accepted: 07/17/2024] [Indexed: 09/22/2024] Open
Abstract
Human-based modelling and simulation offer an ideal testbed for novel medical therapies to guide experimental and clinical studies. Myocardial infarction (MI) is a common cause of heart failure and mortality, for which novel therapies are urgently needed. Although cell therapy offers promise, electrophysiological heterogeneity raises pro-arrhythmic safety concerns, where underlying complex spatio-temporal dynamics cannot be investigated experimentally. Here, after demonstrating credibility of the modelling and simulation framework, we investigate cell therapy in acute versus chronic MI and the role of cell heterogeneity, scar size and the Purkinje system. Simulations agreed with experimental and clinical recordings from ionic to ECG dynamics in acute and chronic infarction. Following cell delivery, spontaneous beats were facilitated by heterogeneity in cell populations, chronic MI due to tissue depolarisation and slow sinus rhythm. Subsequent re-entrant arrhythmias occurred, in some instances with Purkinje involvement and their susceptibility was enhanced by impaired Purkinje-myocardium coupling, large scars and acute infarction. We conclude that homogeneity in injected ventricular-like cell populations minimises their spontaneous beating, which is enhanced by chronic MI, whereas a healthy Purkinje-myocardium coupling is key to prevent subsequent re-entrant arrhythmias, particularly for large scars.
Collapse
Affiliation(s)
| | | | | | - Cristian Trovato
- Department of Computer Science, University of Oxford, Oxford, UK
- Systems Medicine, Clinical Pharmacology & Safety Science, R&D, AstraZeneca, Cambridge, UK
| | - Julia Camps
- Department of Computer Science, University of Oxford, Oxford, UK
| | - Lucas Arantes Berg
- Department of Computer Science, University of Oxford, Oxford, UK
- Universidade Federal de Juiz de Fora, Juiz de Fora, Brazil
| | - Xin Zhou
- Department of Computer Science, University of Oxford, Oxford, UK
| | - Ruben Doste
- Department of Computer Science, University of Oxford, Oxford, UK
| | | | | | - Jacopo Biasetti
- Systems Medicine, Clinical Pharmacology & Safety Science, R&D, AstraZeneca, Gothenburg, Sweden
| | - Blanca Rodriguez
- Department of Computer Science, University of Oxford, Oxford, UK.
| |
Collapse
|
3
|
Xiao Y, Xia L, Jiang W, Qin J, Zhao L, Li Z, Huang L, Li K, Yu P, Wei L, Jiang X, Chen Z, Yu X. Cardiopulmonary progenitors facilitate cardiac repair via exosomal transfer of miR-27b-3p targeting the SIK1-CREB1 axis. Cell Prolif 2024; 57:e13593. [PMID: 38185757 PMCID: PMC11056695 DOI: 10.1111/cpr.13593] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2023] [Revised: 11/27/2023] [Accepted: 12/17/2023] [Indexed: 01/09/2024] Open
Abstract
Ischemic heart disease, especially myocardial infarction (MI), is one of the leading causes of death worldwide, and desperately needs effective treatments, such as cell therapy. Cardiopulmonary progenitors (CPPs) are stem cells for both heart and lung, but their repairing role in damaged heart is still unknown. Here, we obtained CPPs from E9.5 mouse embryos, maintained their stemness while expanding, and identified their characteristics by scRNA-seq, flow cytometry, quantitative reverse transcription-polymerase chain reaction, and differentiation assays. Moreover, we employed mouse MI model to investigate whether CPPs could repair the injured heart. Our data identified that CPPs exhibit hybrid fibroblastic, endothelial, and mesenchymal state, and they could differentiate into cell lineages within the cardiopulmonary system. Moreover, intramyocardial injection of CPPs improves cardiac function through CPPs exosomes (CPPs-Exo) by promotion of cardiomyocytic proliferation and vascularization. To uncover the underlying mechanism, we used miRNA-seq, bulk RNA-seq, and bioinformatic approaches, and found the highly expressed miR-27b-3p in CPPs-Exo and its target gene Sik1, which can influence the transcriptional activity of CREB1. Therefore, we postulate that CPPs facilitate cardiac repair partially through the SIK1-CREB1 axis via exosomal miR-27b-3p. Our study offers a novel insight into the role of CPPs-Exo in heart repair and highlights the potential of CPPs-Exo as a promising therapeutic strategy for MI.
Collapse
Affiliation(s)
- Ying‐Ying Xiao
- Key Laboratory of Molecular Target & Clinical Pharmacology, the NMPA and State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences, The Fifth Affiliated Hospital & the First Affiliated HospitalGuangzhou Medical UniversityGuangzhouGuangdongChina
- Department of Pharmacy, The First Affiliated HospitalGuangzhou Medical UniversityGuangzhouGuangdongChina
| | - Luo‐Xing Xia
- Key Laboratory of Molecular Target & Clinical Pharmacology, the NMPA and State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences, The Fifth Affiliated Hospital & the First Affiliated HospitalGuangzhou Medical UniversityGuangzhouGuangdongChina
| | - Wen‐Jing Jiang
- Key Laboratory of Molecular Target & Clinical Pharmacology, the NMPA and State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences, The Fifth Affiliated Hospital & the First Affiliated HospitalGuangzhou Medical UniversityGuangzhouGuangdongChina
| | - Jian‐Feng Qin
- Key Laboratory of Molecular Target & Clinical Pharmacology, the NMPA and State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences, The Fifth Affiliated Hospital & the First Affiliated HospitalGuangzhou Medical UniversityGuangzhouGuangdongChina
| | - Li‐Xin Zhao
- Key Laboratory of Molecular Target & Clinical Pharmacology, the NMPA and State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences, The Fifth Affiliated Hospital & the First Affiliated HospitalGuangzhou Medical UniversityGuangzhouGuangdongChina
| | - Zhan Li
- Key Laboratory of Molecular Target & Clinical Pharmacology, the NMPA and State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences, The Fifth Affiliated Hospital & the First Affiliated HospitalGuangzhou Medical UniversityGuangzhouGuangdongChina
| | - Li‐Juan Huang
- Key Laboratory of Molecular Target & Clinical Pharmacology, the NMPA and State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences, The Fifth Affiliated Hospital & the First Affiliated HospitalGuangzhou Medical UniversityGuangzhouGuangdongChina
| | - Ke‐Xin Li
- Key Laboratory of Molecular Target & Clinical Pharmacology, the NMPA and State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences, The Fifth Affiliated Hospital & the First Affiliated HospitalGuangzhou Medical UniversityGuangzhouGuangdongChina
| | - Peng‐Jiu Yu
- Department of Pharmacy, The First Affiliated HospitalGuangzhou Medical UniversityGuangzhouGuangdongChina
| | - Li Wei
- Department of Pharmacy, The First Affiliated HospitalGuangzhou Medical UniversityGuangzhouGuangdongChina
| | - Xue‐Yan Jiang
- Key Laboratory of Molecular Target & Clinical Pharmacology, the NMPA and State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences, The Fifth Affiliated Hospital & the First Affiliated HospitalGuangzhou Medical UniversityGuangzhouGuangdongChina
| | - Zhe‐Sheng Chen
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, Institute for BiotechnologySt. John's UniversityQueensNew YorkUSA
| | - Xi‐Yong Yu
- Key Laboratory of Molecular Target & Clinical Pharmacology, the NMPA and State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences, The Fifth Affiliated Hospital & the First Affiliated HospitalGuangzhou Medical UniversityGuangzhouGuangdongChina
| |
Collapse
|
4
|
González-King H, Rodrigues PG, Albery T, Tangruksa B, Gurrapu R, Silva AM, Musa G, Kardasz D, Liu K, Kull B, Åvall K, Rydén-Markinhuhta K, Incitti T, Sharma N, Graneli C, Valadi H, Petkevicius K, Carracedo M, Tejedor S, Ivanova A, Heydarkhan-Hagvall S, Menasché P, Synnergren J, Dekker N, Wang QD, Jennbacken K. Head-to-head comparison of relevant cell sources of small extracellular vesicles for cardiac repair: Superiority of embryonic stem cells. J Extracell Vesicles 2024; 13:e12445. [PMID: 38711334 DOI: 10.1002/jev2.12445] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2023] [Accepted: 04/10/2024] [Indexed: 05/08/2024] Open
Abstract
Small extracellular vesicles (sEV) derived from various cell sources have been demonstrated to enhance cardiac function in preclinical models of myocardial infarction (MI). The aim of this study was to compare different sources of sEV for cardiac repair and determine the most effective one, which nowadays remains limited. We comprehensively assessed the efficacy of sEV obtained from human primary bone marrow mesenchymal stromal cells (BM-MSC), human immortalized MSC (hTERT-MSC), human embryonic stem cells (ESC), ESC-derived cardiac progenitor cells (CPC), human ESC-derived cardiomyocytes (CM), and human primary ventricular cardiac fibroblasts (VCF), in in vitro models of cardiac repair. ESC-derived sEV (ESC-sEV) exhibited the best pro-angiogenic and anti-fibrotic effects in vitro. Then, we evaluated the functionality of the sEV with the most promising performances in vitro, in a murine model of MI-reperfusion injury (IRI) and analysed their RNA and protein compositions. In vivo, ESC-sEV provided the most favourable outcome after MI by reducing adverse cardiac remodelling through down-regulating fibrosis and increasing angiogenesis. Furthermore, transcriptomic, and proteomic characterizations of sEV derived from hTERT-MSC, ESC, and CPC revealed factors in ESC-sEV that potentially drove the observed functions. In conclusion, ESC-sEV holds great promise as a cell-free treatment for promoting cardiac repair following MI.
Collapse
Affiliation(s)
- Hernán González-King
- Research and Early Development, Cardiovascular, Renal and Metabolism, BioPharmaceuticals R&D, AstraZeneca, Mölndal, Sweden
| | - Patricia G Rodrigues
- Research and Early Development, Cardiovascular, Renal and Metabolism, BioPharmaceuticals R&D, AstraZeneca, Mölndal, Sweden
| | - Tamsin Albery
- Research and Early Development, Cardiovascular, Renal and Metabolism, BioPharmaceuticals R&D, AstraZeneca, Mölndal, Sweden
| | - Benyapa Tangruksa
- Systems Biology Research Center, School of Bioscience, University of Skövde, Skövde, Sweden
- Department of Rheumatology and Inflammation Research, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Ramya Gurrapu
- AstraZeneca India Private Limited, Neville Tower 11th Floor, Ramanujan IT SEZ, Rajv Gandhi Salai (OMR), Taramani, Chennai, Tamil Nadu, India
| | - Andreia M Silva
- Discovery Sciences, Oligo Assay Development, BioPharmaceuticals R&D, AstraZeneca, Mölndal, Sweden
- Anjarium Biosciences AG, Schlieren, Switzerland
| | - Gentian Musa
- Research and Early Development, Cardiovascular, Renal and Metabolism, BioPharmaceuticals R&D, AstraZeneca, Mölndal, Sweden
| | - Dominika Kardasz
- Research and Early Development, Cardiovascular, Renal and Metabolism, BioPharmaceuticals R&D, AstraZeneca, Mölndal, Sweden
| | - Kai Liu
- Discovery Sciences, Oligo Assay Development, BioPharmaceuticals R&D, AstraZeneca, Mölndal, Sweden
- Pharmaceutical Sciences, Advanced Drug Delivery, BioPharmaceuticals R&D, AstraZeneca, Mölndal, Sweden
| | - Bengt Kull
- Research and Early Development, Cardiovascular, Renal and Metabolism, BioPharmaceuticals R&D, AstraZeneca, Mölndal, Sweden
| | - Karin Åvall
- Research and Early Development, Cardiovascular, Renal and Metabolism, BioPharmaceuticals R&D, AstraZeneca, Mölndal, Sweden
| | - Katarina Rydén-Markinhuhta
- Research and Early Development, Cardiovascular, Renal and Metabolism, BioPharmaceuticals R&D, AstraZeneca, Mölndal, Sweden
| | - Tania Incitti
- Research and Early Development, Cardiovascular, Renal and Metabolism, BioPharmaceuticals R&D, AstraZeneca, Mölndal, Sweden
| | - Nitin Sharma
- AstraZeneca India Private Limited, Neville Tower 11th Floor, Ramanujan IT SEZ, Rajv Gandhi Salai (OMR), Taramani, Chennai, Tamil Nadu, India
| | - Cecilia Graneli
- Research and Early Development, Cardiovascular, Renal and Metabolism, BioPharmaceuticals R&D, AstraZeneca, Mölndal, Sweden
| | - Hadi Valadi
- Department of Rheumatology and Inflammation Research, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Kasparas Petkevicius
- Research and Early Development, Cardiovascular, Renal and Metabolism, BioPharmaceuticals R&D, AstraZeneca, Mölndal, Sweden
| | - Miguel Carracedo
- Research and Early Development, Cardiovascular, Renal and Metabolism, BioPharmaceuticals R&D, AstraZeneca, Mölndal, Sweden
| | - Sandra Tejedor
- Research and Early Development, Cardiovascular, Renal and Metabolism, BioPharmaceuticals R&D, AstraZeneca, Mölndal, Sweden
- Systems Biology Research Center, School of Bioscience, University of Skövde, Skövde, Sweden
| | - Alena Ivanova
- Discovery Sciences, Oligo Assay Development, BioPharmaceuticals R&D, AstraZeneca, Mölndal, Sweden
| | - Sepideh Heydarkhan-Hagvall
- Systems Biology Research Center, School of Bioscience, University of Skövde, Skövde, Sweden
- Chief Medical Office, Global Patient Safety, AstraZeneca, Mölndal, Sweden
| | - Phillipe Menasché
- Department of Cardiovascular Surgery, Hôpital Européen Georges Pompidou, Université de Paris, PARCC, INSERM, Paris, France
| | - Jane Synnergren
- Systems Biology Research Center, School of Bioscience, University of Skövde, Skövde, Sweden
- Department of Molecular and Clinical Medicine, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Niek Dekker
- Discovery Sciences, Oligo Assay Development, BioPharmaceuticals R&D, AstraZeneca, Mölndal, Sweden
| | - Qing-Dong Wang
- Research and Early Development, Cardiovascular, Renal and Metabolism, BioPharmaceuticals R&D, AstraZeneca, Mölndal, Sweden
| | - Karin Jennbacken
- Research and Early Development, Cardiovascular, Renal and Metabolism, BioPharmaceuticals R&D, AstraZeneca, Mölndal, Sweden
| |
Collapse
|
5
|
Eschenhagen T, Weinberger F. Challenges and perspectives of heart repair with pluripotent stem cell-derived cardiomyocytes. NATURE CARDIOVASCULAR RESEARCH 2024; 3:515-524. [PMID: 39195938 DOI: 10.1038/s44161-024-00472-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/30/2023] [Accepted: 04/04/2024] [Indexed: 08/29/2024]
Abstract
Here we aim at providing a concise but comprehensive overview of the perspectives and challenges of heart repair with pluripotent stem cell-derived cardiomyocytes. This Review comes at a time when consensus has been reached about the lack of relevant proliferative capacity of adult mammalian cardiomyocytes and the lack of new heart muscle formation with autologous cell sources. While alternatives to cell-based approaches will be shortly summarized, the focus lies on pluripotent stem cell-derived cardiomyocyte repair, which entered first clinical trials just 2 years ago. In the view of the authors, these early trials are important but have to be viewed as early proof-of-concept trials in humans that will hopefully provide first answers on feasibility, safety and the survival of allogeneic pluripotent stem cell-derived cardiomyocyte in the human heart. Better approaches have to be developed to make this approach clinically applicable.
Collapse
Affiliation(s)
- Thomas Eschenhagen
- Institute of Experimental Pharmacology and Toxicology, University Medical Center Hamburg Eppendorf, Hamburg, Germany.
- German Centre for Cardiovascular Research (DZHK), partner site Hamburg/Kiel/Lübeck, Hamburg, Germany.
| | - Florian Weinberger
- Institute of Experimental Pharmacology and Toxicology, University Medical Center Hamburg Eppendorf, Hamburg, Germany
- German Centre for Cardiovascular Research (DZHK), partner site Hamburg/Kiel/Lübeck, Hamburg, Germany
- Cardiovascular Regeneration Program, Centro Nacional de Investigaciones Cardiovasculares, Madrid, Spain
| |
Collapse
|
6
|
Carey CM, Hollins HL, Schmid AV, Gagnon JA. Distinct features of the regenerating heart uncovered through comparative single-cell profiling. Biol Open 2024; 13:bio060156. [PMID: 38526188 PMCID: PMC11007736 DOI: 10.1242/bio.060156] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2023] [Accepted: 03/14/2024] [Indexed: 03/26/2024] Open
Abstract
Adult humans respond to heart injury by forming a permanent scar, yet other vertebrates are capable of robust and complete cardiac regeneration. Despite progress towards characterizing the mechanisms of cardiac regeneration in fish and amphibians, the large evolutionary gulf between mammals and regenerating vertebrates complicates deciphering which cellular and molecular features truly enable regeneration. To better define these features, we compared cardiac injury responses in zebrafish and medaka, two fish species that share similar heart anatomy and common teleost ancestry but differ in regenerative capability. We used single-cell transcriptional profiling to create a time-resolved comparative cell atlas of injury responses in all major cardiac cell types across both species. With this approach, we identified several key features that distinguish cardiac injury response in the non-regenerating medaka heart. By comparing immune responses to injury, we found altered cell recruitment and a distinct pro-inflammatory gene program in medaka leukocytes, and an absence of the injury-induced interferon response seen in zebrafish. In addition, we found a lack of pro-regenerative signals, including nrg1 and retinoic acid, from medaka endothelial and epicardial cells. Finally, we identified alterations in the myocardial structure in medaka, where they lack primordial layer cardiomyocytes and fail to employ a cardioprotective gene program shared by regenerating vertebrates. Our findings reveal notable variation in injury response across nearly all major cardiac cell types in zebrafish and medaka, demonstrating how evolutionary divergence influences the hidden cellular features underpinning regenerative potential in these seemingly similar vertebrates.
Collapse
Affiliation(s)
- Clayton M. Carey
- School of Biological Sciences, University of Utah, Salt Lake City, UT, 84112, USA
| | - Hailey L. Hollins
- School of Biological Sciences, University of Utah, Salt Lake City, UT, 84112, USA
| | - Alexis V. Schmid
- School of Biological Sciences, University of Utah, Salt Lake City, UT, 84112, USA
| | - James A. Gagnon
- School of Biological Sciences, University of Utah, Salt Lake City, UT, 84112, USA
- Henry Eyring Center for Cell & Genome Science, University of Utah, Salt Lake City, UT 84112, USA
| |
Collapse
|
7
|
Baron V, Sommer ST, Fiegle DJ, Pfeuffer AKM, Peyronnet R, Volk T, Seidel T. Effects of electro-mechanical uncouplers, hormonal stimulation and pacing rate on the stability and function of cultured rabbit myocardial slices. Front Bioeng Biotechnol 2024; 12:1363538. [PMID: 38646013 PMCID: PMC11026719 DOI: 10.3389/fbioe.2024.1363538] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2023] [Accepted: 03/18/2024] [Indexed: 04/23/2024] Open
Abstract
Introduction: Recent advances have enabled organotypic culture of beating human myocardial slices that are stable for weeks. However, human myocardial samples are rare, exhibit high variability and frequently originate from diseased hearts. Thus, there is a need to adapt long-term slice culture for animal myocardium. When applied to animal cardiac slices, studies in healthy or genetically modified myocardium will be possible. We present the culture of slices from rabbit hearts, which resemble the human heart in microstructure, electrophysiology and excitation-contraction coupling. Methods: Left ventricular myocardium from New Zealand White rabbits was cut using a vibratome and cultured in biomimetic chambers for up to 7 days (d). Electro-mechanical uncoupling agents 2,3-butanedione monoxime (BDM) and cytochalasin D (CytoD) were added during initiation of culture and effects on myocyte survival were quantified. We investigated pacing rates (0.5 Hz, 1 Hz, and 2 Hz) and hormonal supplements (cortisol, T3, catecholamines) at physiological plasma concentrations. T3 was buffered using BSA. Contractile force was recorded continuously. Glucose consumption and lactate production were measured. Whole-slice Ca2+ transients and action potentials were recorded. Effects of culture on microstructure were investigated with confocal microscopy and image analysis. Results: Protocols for human myocardial culture resulted in sustained contracture and myocyte death in rabbit slices within 24 h, which could be prevented by transient application of a combination of BDM and CytoD. Cortisol stabilized contraction amplitude and kinetics in culture. T3 and catecholaminergic stimulation did not further improve stability. T3 and higher pacing rates increased metabolic rate and lactate production. T3 stabilized the response to β-adrenergic stimulation over 7 d. Pacing rates above 1 Hz resulted in progredient decline in contraction force. Image analysis revealed no changes in volume fractions of cardiomyocytes or measures of fibrosis over 7 d. Ca2+ transient amplitudes and responsiveness to isoprenaline were comparable after 1 d and 7 d, while Ca2+ transient duration was prolonged after 7 d in culture. Conclusions: A workflow for rabbit myocardial culture has been established, preserving function for up to 7 d. This research underscores the importance of glucocorticoid signaling in maintaining tissue function and extending culture duration. Furthermore, BDM and CytoD appear to protect from tissue damage during the initiation phase of tissue culture.
Collapse
Affiliation(s)
- V. Baron
- Institute of Cellular and Molecular Physiology, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - S. T. Sommer
- Institute of Cellular and Molecular Physiology, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - D. J. Fiegle
- Institute of Cellular and Molecular Physiology, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - A.-K. M. Pfeuffer
- Institute of Cellular and Molecular Physiology, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - R. Peyronnet
- Institute for Experimental Cardiovascular Medicine, University Heart Center Freiburg, Bad Krozingen, Germany
- Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - T. Volk
- Institute of Cellular and Molecular Physiology, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - T. Seidel
- Institute of Cellular and Molecular Physiology, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| |
Collapse
|
8
|
Zhao J, Rui L, Ouyang W, Hao Y, Liu Y, Tang J, Ding Z, Teng Z, Liu X, Zhu H, Ding Z. Cardiac commitment driven by MyoD expression in pericardial stem cells. Front Cell Dev Biol 2024; 12:1369091. [PMID: 38601082 PMCID: PMC11004306 DOI: 10.3389/fcell.2024.1369091] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2024] [Accepted: 02/27/2024] [Indexed: 04/12/2024] Open
Abstract
Cellular therapy holds immense promise to remuscularize the damaged myocardium but is practically hindered by limited allogeneic sources of cardiac-committed cells that engraft stably in the recipient heart after transplantation. Here, we demonstrate that the pericardial tissue harbors myogenic stem cells (pSCs) that are activated in response to inflammatory signaling after myocardial infarction (MI). The pSCs derived from the MI rats (MI-pSCs) show in vivo and in vitro cardiac commitment characterized by cardiac-specific Tnnt2 expression and formation of rhythmic contraction in culture. Bulk RNA-seq analysis reveals significant upregulation of a panel of genes related to cardiac/myogenic differentiation, paracrine factors, and extracellular matrix in the activated pSCs compared to the control pSCs (Sham-pSCs). Notably, we define MyoD as a key factor that governs the process of cardiac commitment, as siRNA-mediated MyoD gene silencing results in a significant reduction of myogenic potential. Injection of the cardiac-committed cells into the infarcted rat heart leads to long-term survival and stable engraftment in the recipient myocardium. Therefore, these findings point to pericardial myogenic progenitors as an attractive candidate for cardiac cell-based therapy to remuscularize the damaged myocardium.
Collapse
Affiliation(s)
- Jianfeng Zhao
- Department of Cardiology, The People’s Hospital of Danyang Affiliated to Nantong University, Danyang, China
| | - Limei Rui
- Department of Cardiology, The People’s Hospital of Danyang Affiliated to Nantong University, Danyang, China
| | - Weili Ouyang
- Department of Cardiology, The People’s Hospital of Danyang Affiliated to Nantong University, Danyang, China
| | - Yingcai Hao
- Department of Cardiology, The People’s Hospital of Danyang Affiliated to Nantong University, Danyang, China
| | - Yusong Liu
- Department of Cardiology, The People’s Hospital of Danyang Affiliated to Nantong University, Danyang, China
| | - Jianfeng Tang
- Department of Cardiology, The People’s Hospital of Danyang Affiliated to Nantong University, Danyang, China
| | - Zheheng Ding
- Institute of Biochemistry and Molecular Biology II, Heinrich-Heine University of Düsseldorf, Düsseldorf, Germany
| | - Zenghui Teng
- Institute Neuro and Sensory Physiology, Heinrich-Heine University of Düsseldorf, Düsseldorf, Germany
| | - Xueqing Liu
- Department of Cardiology, The People’s Hospital of Danyang Affiliated to Nantong University, Danyang, China
| | - Hongtao Zhu
- Department of Cardiology, The People’s Hospital of Danyang Affiliated to Nantong University, Danyang, China
| | - Zhaoping Ding
- Institute of Molecular Cardiology, Heinrich-Heine University of Düsseldorf, Düsseldorf, Germany
| |
Collapse
|
9
|
Constanty F, Wu B, Wei KH, Lin IT, Dallmann J, Guenther S, Lautenschlaeger T, Priya R, Lai SL, Stainier DYR, Beisaw A. Border-zone cardiomyocytes and macrophages contribute to remodeling of the extracellular matrix to promote cardiomyocyte invasion during zebrafish cardiac regeneration. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.03.12.584570. [PMID: 38559277 PMCID: PMC10980021 DOI: 10.1101/2024.03.12.584570] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/04/2024]
Abstract
Despite numerous advances in our understanding of zebrafish cardiac regeneration, an aspect that remains less studied is how regenerating cardiomyocytes invade, and eventually replace, the collagen-containing fibrotic tissue following injury. Here, we provide an in-depth analysis of the process of cardiomyocyte invasion using live-imaging and histological approaches. We observed close interactions between protruding cardiomyocytes and macrophages at the wound border zone, and macrophage-deficient irf8 mutant zebrafish exhibited defects in extracellular matrix (ECM) remodeling and cardiomyocyte protrusion into the injured area. Using a resident macrophage ablation model, we show that defects in ECM remodeling at the border zone and subsequent cardiomyocyte protrusion can be partly attributed to a population of resident macrophages. Single-cell RNA-sequencing analysis of cells at the wound border revealed a population of cardiomyocytes and macrophages with fibroblast-like gene expression signatures, including the expression of genes encoding ECM structural proteins and ECM-remodeling proteins. The expression of mmp14b , which encodes a membrane-anchored matrix metalloproteinase, was restricted to cells in the border zone, including cardiomyocytes, macrophages, fibroblasts, and endocardial/endothelial cells. Genetic deletion of mmp14b led to a decrease in 1) macrophage recruitment to the border zone, 2) collagen degradation at the border zone, and 3) subsequent cardiomyocyte invasion. Furthermore, cardiomyocyte-specific overexpression of mmp14b was sufficient to enhance cardiomyocyte invasion into the injured tissue and along the apical surface of the wound. Altogether, our data shed important insights into the process of cardiomyocyte invasion of the collagen-containing injured tissue during cardiac regeneration. They further suggest that cardiomyocytes and resident macrophages contribute to ECM remodeling at the border zone to promote cardiomyocyte replenishment of the fibrotic injured tissue.
Collapse
|
10
|
Ye C, Yang C, Zhang H, Gao R, Liao Y, Zhang Y, Jie L, Zhang Y, Cheng T, Wang Y, Ren J. Canonical Wnt signaling directs the generation of functional human PSC-derived atrioventricular canal cardiomyocytes in bioprinted cardiac tissues. Cell Stem Cell 2024; 31:398-409.e5. [PMID: 38366588 DOI: 10.1016/j.stem.2024.01.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2023] [Revised: 12/13/2023] [Accepted: 01/24/2024] [Indexed: 02/18/2024]
Abstract
The creation of a functional 3D bioprinted human heart remains challenging, largely due to the lack of some crucial cardiac cell types, including the atrioventricular canal (AVC) cardiomyocytes, which are essential to slow down the electrical impulse between the atrium and ventricle. By utilizing single-cell RNA sequencing analysis and a 3D bioprinting technology, we discover that stage-specific activation of canonical Wnt signaling creates functional AVC cardiomyocytes derived from human pluripotent stem cells. These cardiomyocytes display morphological characteristics and express molecular markers of AVC cardiomyocytes, including transcription factors TBX2 and MSX2. When bioprinted in prefabricated cardiac tissues, these cardiomyocytes successfully delay the electrical impulse, demonstrating their capability of functioning as the AVC cardiomyocytes in vitro. Thus, these findings not only identify canonical Wnt signaling as a key regulator of the AVC cardiomyocyte differentiation in vitro, but, more importantly, provide a critical cellular source for the biofabrication of a functional human heart.
Collapse
Affiliation(s)
- Chenxi Ye
- Institute of Cardiovascular Diseases, Xiamen Cardiovascular Hospital, School of Medicine, Xiamen University, Xiamen 361006, Fujian, China
| | - Chuanlai Yang
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, National Institute of Diagnostics and Vaccine Development in Infectious Diseases, School of Life Sciences, School of Public Health, Xiamen University, Xiamen 361102, Fujian, China
| | - Heqiang Zhang
- Institute of Cardiovascular Diseases, Xiamen Cardiovascular Hospital, School of Medicine, Xiamen University, Xiamen 361006, Fujian, China
| | - Rui Gao
- Institute of Cardiovascular Diseases, Xiamen Cardiovascular Hospital, School of Medicine, Xiamen University, Xiamen 361006, Fujian, China
| | - Yingnan Liao
- Institute of Cardiovascular Diseases, Xiamen Cardiovascular Hospital, School of Medicine, Xiamen University, Xiamen 361006, Fujian, China
| | - Yali Zhang
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, National Institute of Diagnostics and Vaccine Development in Infectious Diseases, School of Life Sciences, School of Public Health, Xiamen University, Xiamen 361102, Fujian, China
| | - Lingjun Jie
- Institute of Cardiovascular Diseases, Xiamen Cardiovascular Hospital, School of Medicine, Xiamen University, Xiamen 361006, Fujian, China
| | - Yanhui Zhang
- Institute of Cardiovascular Diseases, Xiamen Cardiovascular Hospital, School of Medicine, Xiamen University, Xiamen 361006, Fujian, China
| | - Tong Cheng
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, National Institute of Diagnostics and Vaccine Development in Infectious Diseases, School of Life Sciences, School of Public Health, Xiamen University, Xiamen 361102, Fujian, China
| | - Yan Wang
- Institute of Cardiovascular Diseases, Xiamen Cardiovascular Hospital, School of Medicine, Xiamen University, Xiamen 361006, Fujian, China.
| | - Jie Ren
- Institute of Cardiovascular Diseases, Xiamen Cardiovascular Hospital, School of Medicine, Xiamen University, Xiamen 361006, Fujian, China.
| |
Collapse
|
11
|
Raniga K, Nasir A, Vo NTN, Vaidyanathan R, Dickerson S, Hilcove S, Mosqueira D, Mirams GR, Clements P, Hicks R, Pointon A, Stebbeds W, Francis J, Denning C. Strengthening cardiac therapy pipelines using human pluripotent stem cell-derived cardiomyocytes. Cell Stem Cell 2024; 31:292-311. [PMID: 38366587 DOI: 10.1016/j.stem.2024.01.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2023] [Revised: 11/27/2023] [Accepted: 01/19/2024] [Indexed: 02/18/2024]
Abstract
Advances in hiPSC isolation and reprogramming and hPSC-CM differentiation have prompted their therapeutic application and utilization for evaluating potential cardiovascular safety liabilities. In this perspective, we showcase key efforts toward the large-scale production of hiPSC-CMs, implementation of hiPSC-CMs in industry settings, and recent clinical applications of this technology. The key observations are a need for traceable gender and ethnically diverse hiPSC lines, approaches to reduce cost of scale-up, accessible clinical trial datasets, and transparent guidelines surrounding the safety and efficacy of hiPSC-based therapies.
Collapse
Affiliation(s)
- Kavita Raniga
- The Biodiscovery Institute, University of Nottingham, Nottingham NG7 2RD, UK; Pathology, Non-Clinical Safety, GlaxoSmithKline R&D, Stevenage SG1 2NY, UK.
| | - Aishah Nasir
- The Biodiscovery Institute, University of Nottingham, Nottingham NG7 2RD, UK
| | - Nguyen T N Vo
- The Biodiscovery Institute, University of Nottingham, Nottingham NG7 2RD, UK
| | | | | | | | - Diogo Mosqueira
- The Biodiscovery Institute, University of Nottingham, Nottingham NG7 2RD, UK
| | - Gary R Mirams
- Centre for Mathematical Medicine & Biology, School of Mathematical Sciences, University of Nottingham, Nottingham NG7 2RD, UK
| | - Peter Clements
- Pathology, Non-Clinical Safety, GlaxoSmithKline R&D, Stevenage SG1 2NY, UK
| | - Ryan Hicks
- BioPharmaceuticals R&D Cell Therapy Department, Research and Early Development, Cardiovascular, Renal, and Metabolism (CVRM), BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden; School of Cardiovascular and Metabolic Medicine & Sciences, King's College London, London WC2R 2LS, UK
| | - Amy Pointon
- Safety Sciences, Clinical Pharmacology and Safety Sciences, BioPharmaceuticals R&D, AstraZeneca, Cambridge CB2 0AA, UK
| | | | - Jo Francis
- Mechanstic Biology and Profiling, Discovery Sciences, BioPharmaceuticals R&D, AstraZeneca, Cambridge CB2 0AA, UK
| | - Chris Denning
- The Biodiscovery Institute, University of Nottingham, Nottingham NG7 2RD, UK.
| |
Collapse
|
12
|
Luo X, Wang R, Zhang X, Wen X, Xie W. Identification of key genes associated with heart failure based on bioinformatics analysis and screening of traditional Chinese medicines for the prevention and treatment of heart failure. Medicine (Baltimore) 2023; 102:e35959. [PMID: 38065888 PMCID: PMC10713177 DOI: 10.1097/md.0000000000035959] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/20/2023] [Accepted: 10/13/2023] [Indexed: 12/18/2023] Open
Abstract
Heart failure (HF) is the final stage of heart disease. An increasing number of experiments and clinical reports have shown that traditional Chinese medicine (TCM) has many therapeutic effects and advantages in treating HF. In this study, we used bioinformatics methods to screen key genes and predict the components of Chinese herbal medicines with preventive and therapeutic effects on HF. GSE120895 and GSE21610 HF chips were downloaded from the Gene Expression Omnibus database. We screened differentially expressed genes (DEGs). Weighted gene coexpression network analysis was performed to determine key modules. Genes in key modules were used for Gene Ontology and Kyoto Encyclopedia of Genes Genomes analysis to determine the biological functions. Finally, receiver operating characteristic curve analysis was used to screen out key genes, and single-sample GSEA was conducted to screen TCM compounds and effective ingredients of TCM compounds related to HF. We have selected a key module (MeTerquoise) and identified 489 DEGs, of which 357 are up regulated and 132 are down regulated. Gene Ontology and Kyoto Encyclopedia of Genes Genomes analyses indicated that the DEGs were associated with the extracellular matrix, fat metabolism and inflammatory response. We identified IL2, CXCR4, CCL5, THY1, CCN2, and IL7R as key genes. Single-sample GSEA showed that key genes were mainly related to energy metabolism, mitochondrial oxidative phosphorylation, extracellular matrix, and immunity. Finally, a total of 70 TCM compounds and 30 active ingredients of TCM compounds were identified. Bioinformatics methods were applied to preliminarily predict the key genes and TCM compounds involved in HF. These results provide theoretical support for the treatment of HF with TCM compounds and provide targets and research strategies for the development of related new Chinese medicines.
Collapse
Affiliation(s)
- Xu Luo
- Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, China
| | - Rui Wang
- Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, China
| | - Xin Zhang
- Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, China
| | - Xin Wen
- Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, China
| | - Wen Xie
- Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, China
- Department of Cardiology, Affiliated Hospital of Chengdu University of Traditional Chinese Medicine (Traditional Chinese Medicine Hospital of Sichuan), Chengdu, Sichuan, China
| |
Collapse
|
13
|
Lancaster JJ, Grijalva A, Fink J, Ref J, Daugherty S, Whitman S, Fox K, Gorman G, Lancaster LD, Avery R, Acharya T, McArthur A, Strom J, Pierce MK, Moukabary T, Borgstrom M, Benson D, Mangiola M, Pandey AC, Zile MR, Bradshaw A, Koevary JW, Goldman S. Biologically derived epicardial patch induces macrophage mediated pathophysiologic repair in chronically infarcted swine hearts. Commun Biol 2023; 6:1203. [PMID: 38007534 PMCID: PMC10676365 DOI: 10.1038/s42003-023-05564-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2023] [Accepted: 11/09/2023] [Indexed: 11/27/2023] Open
Abstract
There are nearly 65 million people with chronic heart failure (CHF) globally, with no treatment directed at the pathologic cause of the disease, the loss of functioning cardiomyocytes. We have an allogeneic cardiac patch comprised of cardiomyocytes and human fibroblasts on a bioresorbable matrix. This patch increases blood flow to the damaged heart and improves left ventricular (LV) function in an immune competent rat model of ischemic CHF. After 6 months of treatment in an immune competent Yucatan mini swine ischemic CHF model, this patch restores LV contractility without constrictive physiology, partially reversing maladaptive LV and right ventricular remodeling, increases exercise tolerance, without inducing any cardiac arrhythmias or a change in myocardial oxygen consumption. Digital spatial profiling in mice with patch placement 3 weeks after a myocardial infarction shows that the patch induces a CD45pos immune cell response that results in an infiltration of dendritic cells and macrophages with high expression of macrophages polarization to the anti-inflammatory reparative M2 phenotype. Leveraging the host native immune system allows for the potential use of immunomodulatory therapies for treatment of chronic inflammatory diseases not limited to ischemic CHF.
Collapse
Affiliation(s)
- J J Lancaster
- Sarver Heart Center, Department of Medicine, University of Arizona, 1501 North Campbell Avenue, Tucson, AZ, 85724, USA
| | - A Grijalva
- Sarver Heart Center, Department of Medicine, University of Arizona, 1501 North Campbell Avenue, Tucson, AZ, 85724, USA
| | - J Fink
- Division of Blood & Marrow Transplant & Cellular Therapy, Department of Pediatrics, Masonic Cancer Center, University of Minnesota, Minneapolis, MN, 55455, USA
| | - J Ref
- Sarver Heart Center, Department of Medicine, University of Arizona, 1501 North Campbell Avenue, Tucson, AZ, 85724, USA
| | - S Daugherty
- Sarver Heart Center, Department of Medicine, University of Arizona, 1501 North Campbell Avenue, Tucson, AZ, 85724, USA
| | - S Whitman
- Sarver Heart Center, Department of Medicine, University of Arizona, 1501 North Campbell Avenue, Tucson, AZ, 85724, USA
| | - K Fox
- Sarver Heart Center, Department of Medicine, University of Arizona, 1501 North Campbell Avenue, Tucson, AZ, 85724, USA
- Division of Cardiothoracic Surgery, Department of Surgery, University of Arizona, 1501 North Campbell Avenue, Tucson, AZ, 85724, USA
| | - G Gorman
- Sarver Heart Center, Department of Medicine, University of Arizona, 1501 North Campbell Avenue, Tucson, AZ, 85724, USA
| | - L D Lancaster
- Sarver Heart Center, Department of Medicine, University of Arizona, 1501 North Campbell Avenue, Tucson, AZ, 85724, USA
| | - R Avery
- Sarver Heart Center, Department of Medicine, University of Arizona, 1501 North Campbell Avenue, Tucson, AZ, 85724, USA
| | - T Acharya
- Sarver Heart Center, Department of Medicine, University of Arizona, 1501 North Campbell Avenue, Tucson, AZ, 85724, USA
| | - A McArthur
- Sarver Heart Center, Department of Medicine, University of Arizona, 1501 North Campbell Avenue, Tucson, AZ, 85724, USA
| | - J Strom
- Sarver Heart Center, Department of Medicine, University of Arizona, 1501 North Campbell Avenue, Tucson, AZ, 85724, USA
| | - M K Pierce
- Sarver Heart Center, Department of Medicine, University of Arizona, 1501 North Campbell Avenue, Tucson, AZ, 85724, USA
| | - T Moukabary
- Sarver Heart Center, Department of Medicine, University of Arizona, 1501 North Campbell Avenue, Tucson, AZ, 85724, USA
| | - M Borgstrom
- Research & Discovery Tech, Research Computing Specialist, Principal, University of Arizona, 1501 North Campbell Avenue, Tucson, AZ, 85724, USA
| | - D Benson
- Sarver Heart Center, Department of Medicine, University of Arizona, 1501 North Campbell Avenue, Tucson, AZ, 85724, USA
| | - M Mangiola
- Department of Pathology, NYU Grossman School of Medicine, New York City, NY, 11016, USA
| | - A C Pandey
- Section of Cardiology, Tulane University Heart and Vascular Institute, John W. Deming Department of Medicine, Section of Cardiology, Department of Medicine, Southeast Louisiana Veterans Healthcare System, Tulane University School of Medicine, New Orleans, LA, 70122, USA
| | - M R Zile
- Ralph H. Johnson VA Medical Center, Division of Cardiology, Medical University of South Carolina, Thurmond/Gazes Building, 30 Courtenay Drive, Charleston, SC, 29425, USA
| | - A Bradshaw
- Ralph H. Johnson VA Medical Center, Division of Cardiology, Medical University of South Carolina, Thurmond/Gazes Building, 30 Courtenay Drive, Charleston, SC, 29425, USA
| | - J W Koevary
- Sarver Heart Center, Department of Medicine, University of Arizona, 1501 North Campbell Avenue, Tucson, AZ, 85724, USA
- Biomedical Engineering, College of Engineering, University of Arizona, 1127 E. James E. Rogers Way, Tucson, AZ, 85721, USA
| | - S Goldman
- Sarver Heart Center, Department of Medicine, University of Arizona, 1501 North Campbell Avenue, Tucson, AZ, 85724, USA.
| |
Collapse
|
14
|
Wu R, Hu X, Wang J. Current optimized strategies for stem cell-derived extracellular vesicle/exosomes in cardiac repair. J Mol Cell Cardiol 2023; 184:13-25. [PMID: 37801756 DOI: 10.1016/j.yjmcc.2023.09.006] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/04/2023] [Revised: 09/10/2023] [Accepted: 09/20/2023] [Indexed: 10/08/2023]
Abstract
Ischemic heart diseases remain the leading cause of death globally, and stem cell-based therapy has been investigated as a potential approach for cardiac repair. Due to poor survival and engraftment in the cardiac ischemic milieu post transplantation, the predominant therapeutic effects of stem cells act via paracrine actions, by secreting extracellular vesicles (EVs) and/or other factors. Exosomes are nano-sized EVs of endosomal origin, and now viewed as a major contributor in facilitating myocardial repair and regeneration. However, EV/exosome therapy has major obstacles before entering clinical settings, such as limited production yield, unstable biological activity, poor homing efficiency, and low tissue retention. This review aims to provide an overview of the biogenesis and mechanisms of stem cell-derived EV/exosomes in the process of cardiac repair and discuss the current advancements in different optimized strategies to produce high-yield EV/exosomes with higher bioactivity, or engineer them with improved homing efficiency and therapeutic potency. In particular, we outline recent findings toward preclinical and clinical translation of EV/exosome therapy in ischemic heart diseases, and discuss the potential barriers in regard to clinical translation of EV/exosome therapy.
Collapse
Affiliation(s)
- Rongrong Wu
- Department of Cardiology, Second Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou 310009, PR China; State Key Laboratory of Transvascular Implantation Devices, Hangzhou 310009, PR China; Cardiovascular Key Laboratory of Zhejiang Province, Hangzhou 310009, PR China
| | - Xinyang Hu
- Department of Cardiology, Second Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou 310009, PR China; State Key Laboratory of Transvascular Implantation Devices, Hangzhou 310009, PR China; Cardiovascular Key Laboratory of Zhejiang Province, Hangzhou 310009, PR China; Research Center for Life Science and Human Health, Binjiang Institute of Zhejiang University, Hangzhou 310053, PR China.
| | - Jian'an Wang
- Department of Cardiology, Second Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou 310009, PR China; State Key Laboratory of Transvascular Implantation Devices, Hangzhou 310009, PR China; Cardiovascular Key Laboratory of Zhejiang Province, Hangzhou 310009, PR China; Research Center for Life Science and Human Health, Binjiang Institute of Zhejiang University, Hangzhou 310053, PR China.
| |
Collapse
|
15
|
Velichkova G, Dobreva G. Human pluripotent stem cell-based models of heart development and disease. Cells Dev 2023; 175:203857. [PMID: 37257755 DOI: 10.1016/j.cdev.2023.203857] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2023] [Revised: 04/16/2023] [Accepted: 05/25/2023] [Indexed: 06/02/2023]
Abstract
The heart is a complex organ composed of distinct cell types, such as cardiomyocytes, cardiac fibroblasts, endothelial cells, smooth muscle cells, neuronal cells and immune cells. All these cell types contribute to the structural, electrical and mechanical properties of the heart. Genetic manipulation and lineage tracing studies in mice have been instrumental in gaining critical insights into the networks regulating cardiac cell lineage specification, cell fate and plasticity. Such knowledge has been of fundamental importance for the development of efficient protocols for the directed differentiation of pluripotent stem cells (PSCs) in highly specialized cardiac cell types. In this review, we summarize the evolution and current advances in protocols for cardiac subtype specification, maturation, and assembly in cardiac microtissues and organoids.
Collapse
Affiliation(s)
- Gabriel Velichkova
- Department of Cardiovascular Genomics and Epigenomics, European Center for Angioscience (ECAS), Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Gergana Dobreva
- Department of Cardiovascular Genomics and Epigenomics, European Center for Angioscience (ECAS), Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany; German Centre for Cardiovascular Research (DZHK), Germany.
| |
Collapse
|
16
|
Pfeuffer AKM, Küpfer LK, Shankar TS, Drakos SG, Volk T, Seidel T. Ryanodine Receptor Staining Identifies Viable Cardiomyocytes in Human and Rabbit Cardiac Tissue Slices. Int J Mol Sci 2023; 24:13514. [PMID: 37686327 PMCID: PMC10488113 DOI: 10.3390/ijms241713514] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2023] [Revised: 08/26/2023] [Accepted: 08/29/2023] [Indexed: 09/10/2023] Open
Abstract
In terms of preserving multicellularity and myocardial function in vitro, the cultivation of beating myocardial slices is an emerging technique in basic and translational cardiac research. It can be used, for example, for drug screening or to study pathomechanisms. Here, we describe staining for viable cardiomyocytes based on the immunofluorescence of ryanodine receptors (RyRs) in human and rabbit myocardial slices. Biomimetic chambers were used for culture and measurements of contractile force. Fixable fluorophore-conjugated dextran, entering cells with a permeable membrane, was used for death staining. RyRs, nuclei and the extracellular matrix, including the t-system, were additionally stained and analyzed by confocal microscopy and image processing. We found the mutual exclusion of the RyR and dextran signals in cultivated slices. T-System density and nucleus size were reduced in RyR-negative/dextran-positive myocytes. The fraction of RyR-positive myocytes and pixels correlated with the contractile force. In RyR-positive/dextran-positive myocytes, we found irregular RyR clusters and SERCA distribution patterns, confirmed by an altered power spectrum. We conclude that RyR immunofluorescence indicates viable cardiomyocytes in vibratome-cut myocardial slices, facilitating the detection and differential structural analysis of living vs. dead or dying myocytes. We suggest the loss of sarcoplasmic reticulum integrity as an early event during cardiomyocyte death.
Collapse
Affiliation(s)
- Ann-Katrin M. Pfeuffer
- Institute of Cellular and Molecular Physiology, Friedrich-Alexander University (FAU) Erlangen-Nuremberg, 91054 Erlangen, Germany; (A.-K.M.P.); (L.K.K.); (T.V.)
| | - Linda K. Küpfer
- Institute of Cellular and Molecular Physiology, Friedrich-Alexander University (FAU) Erlangen-Nuremberg, 91054 Erlangen, Germany; (A.-K.M.P.); (L.K.K.); (T.V.)
| | - Thirupura S. Shankar
- Nora Eccles Harrison Cardiovascular Research and Training Institute, University of Utah, Salt Lake City, UT 84112, USA; (T.S.S.); (S.G.D.)
| | - Stavros G. Drakos
- Nora Eccles Harrison Cardiovascular Research and Training Institute, University of Utah, Salt Lake City, UT 84112, USA; (T.S.S.); (S.G.D.)
| | - Tilmann Volk
- Institute of Cellular and Molecular Physiology, Friedrich-Alexander University (FAU) Erlangen-Nuremberg, 91054 Erlangen, Germany; (A.-K.M.P.); (L.K.K.); (T.V.)
| | - Thomas Seidel
- Institute of Cellular and Molecular Physiology, Friedrich-Alexander University (FAU) Erlangen-Nuremberg, 91054 Erlangen, Germany; (A.-K.M.P.); (L.K.K.); (T.V.)
| |
Collapse
|
17
|
Chabanovska O, Lemcke H, Lang H, Vollmar B, Dohmen PM, David R, Etz C, Neßelmann C. Sarcomeric network analysis of ex vivo cultivated human atrial appendage tissue using super-resolution microscopy. Sci Rep 2023; 13:13041. [PMID: 37563225 PMCID: PMC10415305 DOI: 10.1038/s41598-023-39962-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2023] [Accepted: 08/02/2023] [Indexed: 08/12/2023] Open
Abstract
Investigating native human cardiac tissue with preserved 3D macro- and microarchitecture is fundamental for clinical and basic research. Unfortunately, the low accessibility of the human myocardium continues to limit scientific progress. To overcome this issue, utilizing atrial appendages of the human heart may become highly beneficial. Atrial appendages are often removed during open-heart surgery and can be preserved ex vivo as living tissue with varying durability depending on the culture method. In this study, we prepared living thin myocardial slices from left atrial appendages that were cultured using an air-liquid interface system for overall 10 days. Metabolic activity of the cultured slices was assessed using a conventional methyl thiazolyl tetrazolium (MTT) assay. To monitor the structural integrity of cardiomyocytes within the tissue, we implemented our recently described super-resolution microscopy approach that allows both qualitative and quantitative in-depth evaluation of sarcomere network based on parameters such as overall sarcomere content, filament size and orientation. Additionally, expression of mRNAs coding for key structural and functional proteins was analyzed by real-time reverse transcription polymerase chain reaction (qRT-PCR). Our findings demonstrate highly significant disassembly of contractile apparatus represented by degradation of [Formula: see text]-actinin filaments detected after three days in culture, while metabolic activity was constantly rising and remained high for up to seven days. However, gene expression of crucial cardiac markers strongly decreased after the first day in culture indicating an early destructive response to ex vivo conditions. Therefore, we suggest static cultivation of living myocardial slices derived from left atrial appendage and prepared according to our protocol only for short-termed experiments (e.g. medicinal drug testing), while introduction of electro-mechanical stimulation protocols may offer the possibility for long-term integrity of such constructs.
Collapse
Affiliation(s)
- Oleksandra Chabanovska
- Reference and Translation Center for Cardiac Stem Cell therapy (RTC), Department of Cardiac Surgery, Rostock University Medical Center, 18057, Rostock, Germany
- Department of Life, Light, and Matter of the Interdisciplinary Faculty, Rostock University, 18059, Rostock, Germany
- Department of Operative Dentistry and Periodontology, Rostock University Medical Center, 18059, Rostock, Germany
| | - Heiko Lemcke
- Reference and Translation Center for Cardiac Stem Cell therapy (RTC), Department of Cardiac Surgery, Rostock University Medical Center, 18057, Rostock, Germany
- Department of Life, Light, and Matter of the Interdisciplinary Faculty, Rostock University, 18059, Rostock, Germany
| | - Hermann Lang
- Department of Operative Dentistry and Periodontology, Rostock University Medical Center, 18059, Rostock, Germany
| | - Brigitte Vollmar
- Rudolf-Zenker-Institute of Experimental Surgery, Rostock University Medical Center, 18059, Rostock, Germany
| | - Pascal M Dohmen
- Department of Cardiac Surgery, Rostock University Medical Center, 18059, Rostock, Germany
- Department of Cardiothoracic Surgery, Faculty of Health Science, University of the Free State, Bloemfontein, 9301, South Africa
| | - Robert David
- Reference and Translation Center for Cardiac Stem Cell therapy (RTC), Department of Cardiac Surgery, Rostock University Medical Center, 18057, Rostock, Germany.
- Department of Life, Light, and Matter of the Interdisciplinary Faculty, Rostock University, 18059, Rostock, Germany.
| | - Christian Etz
- Department of Cardiac Surgery, Rostock University Medical Center, 18059, Rostock, Germany
| | - Catharina Neßelmann
- Department of Cardiac Surgery, Rostock University Medical Center, 18059, Rostock, Germany
| |
Collapse
|
18
|
Bhosle VK, Tan JM, Li T, Hua R, Kwon H, Li Z, Patel S, Tessier-Lavigne M, Robinson LA, Kim PK, Brumell JH. SLIT2/ROBO1 signaling suppresses mTORC1 for organelle control and bacterial killing. Life Sci Alliance 2023; 6:e202301964. [PMID: 37311584 PMCID: PMC10264968 DOI: 10.26508/lsa.202301964] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2023] [Revised: 05/31/2023] [Accepted: 05/31/2023] [Indexed: 06/15/2023] Open
Abstract
SLIT/ROBO signaling impacts many aspects of tissue development and homeostasis, in part, through the regulation of cell growth and proliferation. Recent studies have also linked SLIT/ROBO signaling to the regulation of diverse phagocyte functions. However, the mechanisms by which SLIT/ROBO signaling acts at the nexus of cellular growth control and innate immunity remain enigmatic. Here, we show that SLIT2-mediated activation of ROBO1 leads to inhibition of mTORC1 kinase activity in macrophages, leading to dephosphorylation of its downstream targets, including transcription factor EB and ULK1. Consequently, SLIT2 augments lysosome biogenesis, potently induces autophagy, and robustly promotes the killing of bacteria within phagosomes. Concordant with these results, we demonstrate decreased lysosomal content and accumulated peroxisomes in the spinal cords of embryos from Robo1 -/- , Robo2 -/- double knockout mice. We also show that impediment of auto/paracrine SLIT-ROBO signaling axis in cancer cells leads to hyperactivation of mTORC1 and inhibition of autophagy. Together, these findings elucidate a central role of chemorepellent SLIT2 in the regulation of mTORC1 activity with important implications for innate immunity and cancer cell survival.
Collapse
Affiliation(s)
- Vikrant K Bhosle
- Cell Biology Program, The Hospital for Sick Children, Toronto, Canada
| | - Joel Mj Tan
- Cell Biology Program, The Hospital for Sick Children, Toronto, Canada
| | - Taoyingnan Li
- Cell Biology Program, The Hospital for Sick Children, Toronto, Canada
- Department of Molecular Genetics, University of Toronto, Toronto, Canada
| | - Rong Hua
- Cell Biology Program, The Hospital for Sick Children, Toronto, Canada
| | - Hyunwoo Kwon
- Cell Biology Program, The Hospital for Sick Children, Toronto, Canada
- Department of Internal Medicine, The Ohio State University, Columbus, OH, USA
| | - Zhubing Li
- Cell Biology Program, The Hospital for Sick Children, Toronto, Canada
| | - Sajedabanu Patel
- Cell Biology Program, The Hospital for Sick Children, Toronto, Canada
| | - Marc Tessier-Lavigne
- Laboratory of Brain Development and Repair, Rockefeller University, New York, NY, USA
- Department of Biology, Stanford University, Stanford, CA, USA
| | - Lisa A Robinson
- Cell Biology Program, The Hospital for Sick Children, Toronto, Canada
- Institute of Medical Science, University of Toronto, Toronto, Canada
- Division of Nephrology, The Hospital for Sick Children, Toronto, Canada
- Department of Paediatrics, Faculty of Medicine, University of Toronto, Toronto, Canada
| | - Peter K Kim
- Cell Biology Program, The Hospital for Sick Children, Toronto, Canada
- Department of Biochemistry, University of Toronto, Toronto, Canada
| | - John H Brumell
- Cell Biology Program, The Hospital for Sick Children, Toronto, Canada
- Department of Molecular Genetics, University of Toronto, Toronto, Canada
- Institute of Medical Science, University of Toronto, Toronto, Canada
- SickKids IBD Centre, Hospital for Sick Children, Toronto, Canada
| |
Collapse
|
19
|
Carey CM, Hollins HL, Schmid AV, Gagnon JA. Distinct features of the regenerating heart uncovered through comparative single-cell profiling. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.07.04.547574. [PMID: 37461520 PMCID: PMC10349989 DOI: 10.1101/2023.07.04.547574] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 07/28/2023]
Abstract
Adult humans respond to heart injury by forming a permanent scar, yet other vertebrates are capable of robust and complete cardiac regeneration. Despite progress towards characterizing the mechanisms of cardiac regeneration in fish and amphibians, the large evolutionary gulf between mammals and regenerating vertebrates complicates deciphering which cellular and molecular features truly enable regeneration. To better define these features, we compared cardiac injury responses in zebrafish and medaka, two fish species that share similar heart anatomy and common teleost ancestry but differ in regenerative capability. We used single-cell transcriptional profiling to create a time-resolved comparative cell atlas of injury responses in all major cardiac cell types across both species. With this approach, we identified several key features that distinguish cardiac injury response in the non-regenerating medaka heart. By comparing immune responses to injury, we found altered cell recruitment and a distinct pro-inflammatory gene program in medaka leukocytes, and an absence of the injury-induced interferon response seen in zebrafish. In addition, we found a lack of pro-regenerative signals, including nrg1 and retinoic acid, from medaka endothelial and epicardial cells. Finally, we identified alterations in the myocardial structure in medaka, where they lack embryonic-like primordial layer cardiomyocytes, and fail to employ a cardioprotective gene program shared by regenerating vertebrates. Our findings reveal notable variation in injury response across nearly all major cardiac cell types in zebrafish and medaka, demonstrating how evolutionary divergence influences the hidden cellular features underpinning regenerative potential in these seemingly similar vertebrates.
Collapse
Affiliation(s)
- Clayton M. Carey
- School of Biological Sciences, University of Utah, Salt Lake City, UT, 84112, USA
| | - Hailey L. Hollins
- School of Biological Sciences, University of Utah, Salt Lake City, UT, 84112, USA
| | - Alexis V. Schmid
- School of Biological Sciences, University of Utah, Salt Lake City, UT, 84112, USA
| | - James A. Gagnon
- School of Biological Sciences, University of Utah, Salt Lake City, UT, 84112, USA
- Henry Eyring Center for Cell & Genome Science, University of Utah, Salt Lake City, UT 84112, USA
| |
Collapse
|
20
|
Poch CM, Dendorfer A, Laugwitz KL, Moretti A. Ex vivo Culture and Contractile Force Measurements of Non-human Primate Heart Slices. Bio Protoc 2023; 13:e4750. [PMID: 37456341 PMCID: PMC10338344 DOI: 10.21769/bioprotoc.4750] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2022] [Revised: 03/15/2023] [Accepted: 05/22/2023] [Indexed: 07/18/2023] Open
Abstract
Cardiovascular diseases are the leading cause of death and morbidity worldwide. Patient mortality has been successfully reduced by nearly half in the last four decades, mainly due to advances in minimally invasive surgery techniques and interventional cardiology methods. However, a major hurdle is still the translational gap between preclinical findings and the conversion into effective therapies, which is partly due to the use of model systems that fail to recapitulate key aspects of human physiology and disease. Large animal models such as pigs and non-human primates are highly valuable because they closely resemble humans but are costly and time intensive. Here, we provide a method for long-term ex vivo culture of non-human primate (NHP) myocardial tissue that offers a powerful alternative for a wide range of applications including electrophysiology studies, drug screening, and gene function analyses.
Collapse
Affiliation(s)
- Christine M. Poch
- First Department of Medicine, Cardiology, Klinikum rechts der Isar, Technical University of Munich, School of Medicine and Health, Munich, Germany
| | - Andreas Dendorfer
- Walter-Brendel-Centre of Experimental Medicine, University Hospital, LMU Munich, Germany
- DZHK (German Centre of Cardiovascular Research) – Munich Heart Alliance, Munich, Germany
| | - Karl-Ludwig Laugwitz
- First Department of Medicine, Cardiology, Klinikum rechts der Isar, Technical University of Munich, School of Medicine and Health, Munich, Germany
- DZHK (German Centre of Cardiovascular Research) – Munich Heart Alliance, Munich, Germany
| | - Alessandra Moretti
- First Department of Medicine, Cardiology, Klinikum rechts der Isar, Technical University of Munich, School of Medicine and Health, Munich, Germany
- DZHK (German Centre of Cardiovascular Research) – Munich Heart Alliance, Munich, Germany
| |
Collapse
|
21
|
Yap L, Chong LY, Tan C, Adusumalli S, Seow M, Guo J, Cai Z, Loo SJ, Lim E, Tan RS, Grishina E, Soong PL, Lath N, Ye L, Petretto E, Tryggvason K. Pluripotent stem cell-derived committed cardiac progenitors remuscularize damaged ischemic hearts and improve their function in pigs. NPJ Regen Med 2023; 8:26. [PMID: 37236990 DOI: 10.1038/s41536-023-00302-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2022] [Accepted: 05/16/2023] [Indexed: 05/28/2023] Open
Abstract
Ischemic heart disease, which is often associated with irreversibly damaged heart muscle, is a major global health burden. Here, we report the potential of stem cell-derived committed cardiac progenitors (CCPs) have in regenerative cardiology. Human pluripotent embryonic stem cells were differentiated to CCPs on a laminin 521 + 221 matrix, characterized with bulk and single-cell RNA sequencing, and transplanted into infarcted pig hearts. CCPs differentiated for eleven days expressed a set of genes showing higher expression than cells differentiated for seven days. Functional heart studies revealed significant improvement in left ventricular ejection fraction at four and twelve weeks following transplantation. We also observed significant improvements in ventricular wall thickness and a reduction in infarction size after CCP transplantation (p-value < 0.05). Immunohistology analyses revealed in vivo maturation of the CCPs into cardiomyocytes (CM). We observed temporary episodes of ventricular tachyarrhythmia (VT) in four pigs and persistent VT in one pig, but the remaining five pigs exhibited normal sinus rhythm. Importantly, all pigs survived without the formation of any tumors or VT-related abnormalities. We conclude that pluripotent stem cell-derived CCPs constitute a promising possibility for myocardial infarction treatment and that they may positively impact regenerative cardiology.
Collapse
Affiliation(s)
- Lynn Yap
- Cardiovascular & Metabolic Disorders Program, Duke-NUS Medical School, National University of Singapore, Singapore, 169857, Singapore.
- Lee Kong Chian School of Medicine, Nanyang Technological University, 11 Mandalay Road, Singapore, 308232, Singapore.
| | - Li Yen Chong
- Cardiovascular & Metabolic Disorders Program, Duke-NUS Medical School, National University of Singapore, Singapore, 169857, Singapore
| | - Clarissa Tan
- Cardiovascular & Metabolic Disorders Program, Duke-NUS Medical School, National University of Singapore, Singapore, 169857, Singapore
| | - Swarnaseetha Adusumalli
- Cardiovascular & Metabolic Disorders Program, Duke-NUS Medical School, National University of Singapore, Singapore, 169857, Singapore
| | - Millie Seow
- Cardiovascular & Metabolic Disorders Program, Duke-NUS Medical School, National University of Singapore, Singapore, 169857, Singapore
| | - Jing Guo
- Cardiovascular & Metabolic Disorders Program, Duke-NUS Medical School, National University of Singapore, Singapore, 169857, Singapore
| | - Zuhua Cai
- Cardiovascular & Metabolic Disorders Program, Duke-NUS Medical School, National University of Singapore, Singapore, 169857, Singapore
| | - Sze Jie Loo
- National Heart Research Institute Singapore, National Heart Centre Singapore, Singapore, 169609, Singapore
| | - Eric Lim
- National Heart Research Institute Singapore, National Heart Centre Singapore, Singapore, 169609, Singapore
| | - Ru San Tan
- National Heart Research Institute Singapore, National Heart Centre Singapore, Singapore, 169609, Singapore
| | | | - Poh Loong Soong
- Ternion Biosciences, Singapore, 574329, Singapore
- Cardiovascular Disease Translational Research Program, Yong Loo Lin School of Medicine, NUS, Singapore, 169609, Singapore
| | - Narayan Lath
- National Heart Research Institute Singapore, National Heart Centre Singapore, Singapore, 169609, Singapore
| | - Lei Ye
- Department of Biomedical Engineering, University of Alabama, Birmingham, 35233, England
| | - Enrico Petretto
- Cardiovascular & Metabolic Disorders Program, Duke-NUS Medical School, National University of Singapore, Singapore, 169857, Singapore
| | - Karl Tryggvason
- Cardiovascular & Metabolic Disorders Program, Duke-NUS Medical School, National University of Singapore, Singapore, 169857, Singapore.
- Department of Medicine Duke University, Durham, NC, 27710, USA.
- Department of Medical Biochemistry and Biophysics, Karolinska Institute, 171 77, Stockholm, Sweden.
| |
Collapse
|
22
|
Rawat H, Kornherr J, Zawada D, Bakhshiyeva S, Kupatt C, Laugwitz KL, Bähr A, Dorn T, Moretti A, Nowak-Imialek M. Recapitulating porcine cardiac development in vitro: from expanded potential stem cell to embryo culture models. Front Cell Dev Biol 2023; 11:1111684. [PMID: 37261075 PMCID: PMC10227949 DOI: 10.3389/fcell.2023.1111684] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2022] [Accepted: 04/21/2023] [Indexed: 06/02/2023] Open
Abstract
Domestic pigs (Sus scrofa) share many genetic, anatomical, and physiological traits with humans and therefore constitute an excellent preclinical animal model. Fundamental understanding of the cellular and molecular processes governing early porcine cardiogenesis is critical for developing advanced porcine models used for the study of heart diseases and new regenerative therapies. Here, we provide a detailed characterization of porcine cardiogenesis based on fetal porcine hearts at various developmental stages and cardiac cells derived from porcine expanded pluripotent stem cells (pEPSCs), i.e., stem cells having the potential to give rise to both embryonic and extraembryonic tissue. We notably demonstrate for the first time that pEPSCs can differentiate into cardiovascular progenitor cells (CPCs), functional cardiomyocytes (CMs), epicardial cells and epicardial-derived cells (EPDCs) in vitro. Furthermore, we present an enhanced system for whole-embryo culture which allows continuous ex utero development of porcine post-implantation embryos from the cardiac crescent stage (ED14) up to the cardiac looping (ED17) stage. These new techniques provide a versatile platform for studying porcine cardiac development and disease modeling.
Collapse
Affiliation(s)
- Hilansi Rawat
- First Department of Medicine, Cardiology, Klinikum Rechts der Isar, School of Medicine and Health, Technical University of Munich, Munich, Germany
- German Center for Cardiovascular Research (DZHK), Munich Heart Alliance, Munich, Germany
- Regenerative Medicine in Cardiovascular Diseases, First Department of Medicine, Klinikum Rechts der Isar, School of Medicine and Health, Technical University of Munich, Munich, Germany
| | - Jessica Kornherr
- First Department of Medicine, Cardiology, Klinikum Rechts der Isar, School of Medicine and Health, Technical University of Munich, Munich, Germany
- German Center for Cardiovascular Research (DZHK), Munich Heart Alliance, Munich, Germany
- Regenerative Medicine in Cardiovascular Diseases, First Department of Medicine, Klinikum Rechts der Isar, School of Medicine and Health, Technical University of Munich, Munich, Germany
| | - Dorota Zawada
- First Department of Medicine, Cardiology, Klinikum Rechts der Isar, School of Medicine and Health, Technical University of Munich, Munich, Germany
- German Center for Cardiovascular Research (DZHK), Munich Heart Alliance, Munich, Germany
- Regenerative Medicine in Cardiovascular Diseases, First Department of Medicine, Klinikum Rechts der Isar, School of Medicine and Health, Technical University of Munich, Munich, Germany
| | - Sara Bakhshiyeva
- First Department of Medicine, Cardiology, Klinikum Rechts der Isar, School of Medicine and Health, Technical University of Munich, Munich, Germany
- German Center for Cardiovascular Research (DZHK), Munich Heart Alliance, Munich, Germany
- Regenerative Medicine in Cardiovascular Diseases, First Department of Medicine, Klinikum Rechts der Isar, School of Medicine and Health, Technical University of Munich, Munich, Germany
| | - Christian Kupatt
- First Department of Medicine, Cardiology, Klinikum Rechts der Isar, School of Medicine and Health, Technical University of Munich, Munich, Germany
| | - Karl-Ludwig Laugwitz
- First Department of Medicine, Cardiology, Klinikum Rechts der Isar, School of Medicine and Health, Technical University of Munich, Munich, Germany
| | - Andrea Bähr
- First Department of Medicine, Cardiology, Klinikum Rechts der Isar, School of Medicine and Health, Technical University of Munich, Munich, Germany
| | - Tatjana Dorn
- First Department of Medicine, Cardiology, Klinikum Rechts der Isar, School of Medicine and Health, Technical University of Munich, Munich, Germany
- German Center for Cardiovascular Research (DZHK), Munich Heart Alliance, Munich, Germany
- Regenerative Medicine in Cardiovascular Diseases, First Department of Medicine, Klinikum Rechts der Isar, School of Medicine and Health, Technical University of Munich, Munich, Germany
| | - Alessandra Moretti
- First Department of Medicine, Cardiology, Klinikum Rechts der Isar, School of Medicine and Health, Technical University of Munich, Munich, Germany
- German Center for Cardiovascular Research (DZHK), Munich Heart Alliance, Munich, Germany
- Regenerative Medicine in Cardiovascular Diseases, First Department of Medicine, Klinikum Rechts der Isar, School of Medicine and Health, Technical University of Munich, Munich, Germany
- Department of Surgery, Yale University School of Medicine, New Haven, CT, United States
| | - Monika Nowak-Imialek
- First Department of Medicine, Cardiology, Klinikum Rechts der Isar, School of Medicine and Health, Technical University of Munich, Munich, Germany
- German Center for Cardiovascular Research (DZHK), Munich Heart Alliance, Munich, Germany
- Regenerative Medicine in Cardiovascular Diseases, First Department of Medicine, Klinikum Rechts der Isar, School of Medicine and Health, Technical University of Munich, Munich, Germany
| |
Collapse
|
23
|
Secco I, Giacca M. Regulation of endogenous cardiomyocyte proliferation: The known unknowns. J Mol Cell Cardiol 2023; 179:80-89. [PMID: 37030487 PMCID: PMC10390341 DOI: 10.1016/j.yjmcc.2023.04.001] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/15/2023] [Revised: 03/29/2023] [Accepted: 04/04/2023] [Indexed: 04/10/2023]
Abstract
Myocardial regeneration in patients with cardiac damage is a long-sought goal of clinical medicine. In animal species in which regeneration occurs spontaneously, as well as in neonatal mammals, regeneration occurs through the proliferation of differentiated cardiomyocytes, which re-enter the cell cycle and proliferate. Hence, the reprogramming of the replicative potential of cardiomyocytes is an achievable goal, provided that the mechanisms that regulate this process are understood. Cardiomyocyte proliferation is under the control of a series of signal transduction pathways that connect extracellular cues to the activation of specific gene transcriptional programmes, eventually leading to the activation of the cell cycle. Both coding and non-coding RNAs (in particular, microRNAs) are involved in this regulation. The available information can be exploited for therapeutic purposes, provided that a series of conceptual and technical barriers are overcome. A major obstacle remains the delivery of pro-regenerative factors specifically to the heart. Improvements in the design of AAV vectors to enhance their cardiotropism and efficacy or, alternatively, the development of non-viral methods for nucleic acid delivery in cardiomyocytes are among the challenges ahead to progress cardiac regenerative therapies towards clinical application.
Collapse
Affiliation(s)
- Ilaria Secco
- School of Cardiovascular and Metabolic Medicine & Sciences and British Heart Foundation Centre of Research Excellence, King's College London, London, United Kingdom
| | - Mauro Giacca
- School of Cardiovascular and Metabolic Medicine & Sciences and British Heart Foundation Centre of Research Excellence, King's College London, London, United Kingdom.
| |
Collapse
|
24
|
Tekieli Ł, Szot W, Kwiecień E, Mazurek A, Borkowska E, Czyż Ł, Dąbrowski M, Kozynacka A, Skubera M, Podolec P, Majka M, Kostkiewicz M, Musiałek P. Single-photon emission computed tomography as a fundamental tool in evaluation of myocardial reparation and regeneration therapies. ADVANCES IN INTERVENTIONAL CARDIOLOGY 2022; 18:326-339. [PMID: 36967839 PMCID: PMC10031666 DOI: 10.5114/aic.2023.124403] [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: 03/19/2022] [Accepted: 06/14/2022] [Indexed: 01/26/2023] Open
Abstract
Despite unquestionable progress in interventional and pharmacologic therapies of ischemic heart disease, the number of patients with chronic ischemic heart failure is increasing and the prognosis remains poor. Repair/restoration of functional myocardium through progenitor cell-mediated (PCs) healing and renovation of injured myocardium is one of the pivotal directions in biomedical research. PCs release numerous pro-angiogenic and anti-apoptotic factors. Moreover, they have self-renewal capability and may differentiate into specialized cells that include endothelial cells and cardiomyocytes. Uptake and homing of PCs in the zone(s) of ischaemic injury (i.e., their effective transplantation to the target zone) is an essential pre-requisite for any potential therapeutic effect; thus effective cell tracking is fundamental in pre-clinical and early clinical studies. Another crucial requirement in rigorous research is quantification of the infarct zone, including the amount of non-perfused and hypo-perfused myocardium. Quantitative and reproducible evaluation of global and regional myocardial contractility and left ventricular remodeling is particularly relevant in clinical studies. Using SPECT, our earlier work has addressed several critical questions in cardiac regenerative medicine including optimizing transcoronary cell delivery, determination of the zone(s) of myocardial cell uptake, and late functional improvement in relation to the magnitude of cell uptake. Here, we review the role of single-photon emission computed tomography (SPECT), a technique that offers high-sensitivity, quantitative cell tracking on top of its ability to evaluate myocardial perfusion and function on both cross-sectional and longitudinal bases. SPECT, with its direct relevance to routine clinical practice, is a fundamental tool in evaluation of myocardial reparation and regeneration therapies.
Collapse
Affiliation(s)
- Łukasz Tekieli
- Department of Cardiac and Vascular Diseases, John Paul II Hospital, Jagiellonian University, Krakow, Poland
- Department of Interventional Cardiology, John Paul II Hospital, Jagiellonian University, Krakow, Poland
| | - Wojciech Szot
- Department of Radiology, John Paul II Hospital, Krakow, Poland
| | - Ewa Kwiecień
- Department of Cardiac and Vascular Diseases, John Paul II Hospital, Jagiellonian University, Krakow, Poland
| | - Adam Mazurek
- Department of Cardiac and Vascular Diseases, John Paul II Hospital, Jagiellonian University, Krakow, Poland
| | - Eliza Borkowska
- Department of Radiology, John Paul II Hospital, Krakow, Poland
| | - Łukasz Czyż
- Department of Cardiac and Vascular Diseases, John Paul II Hospital, Jagiellonian University, Krakow, Poland
| | - Maciej Dąbrowski
- Department of Interventional Cardiology and Angiology, National Institute of Cardiology, Warsaw, Poland
| | - Anna Kozynacka
- Department of Coronary Artery Disease and Heart Failure, John Paul II Hospital, Jagiellonian University, Krakow, Poland
| | - Maciej Skubera
- Department of Cardiac and Vascular Diseases, John Paul II Hospital, Jagiellonian University, Krakow, Poland
| | - Piotr Podolec
- Department of Cardiac and Vascular Diseases, John Paul II Hospital, Jagiellonian University, Krakow, Poland
| | - Marcin Majka
- Department of Transplantation, Jagiellonian University, Krakow, Poland
| | | | - Piotr Musiałek
- Department of Cardiac and Vascular Diseases, John Paul II Hospital, Jagiellonian University, Krakow, Poland
| |
Collapse
|
25
|
|