1
|
Cao C, Yang L, Song J, Liu Z, Li H, Li L, Fu J, Liu J. Cardiomyocyte regeneration after infarction: changes, opportunities and challenges. Mol Cell Biochem 2025:10.1007/s11010-025-05251-w. [PMID: 40097887 DOI: 10.1007/s11010-025-05251-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2024] [Accepted: 03/08/2025] [Indexed: 03/19/2025]
Abstract
Myocardial infarction is a cardiovascular disease that poses a serious threat to human health. The traditional view is that adult mammalian cardiomyocytes have almost no regenerative ability, but recent studies have shown that they have regenerative potential under specific conditions. This article comprehensively describes the research progress of post-infarction cardiomyocyte regeneration, including the characteristics of cardiomyocytes and post-infarction changes, regeneration mechanisms, influencing factors, potential therapeutic strategies, challenges and future development directions, and deeply discusses the specific pathways and targets included in the regeneration mechanism, aiming to provide new ideas and methods for the treatment of myocardial infarction.
Collapse
Affiliation(s)
- Ce Cao
- Beijing Key Laboratory of Chinese Materia Pharmacology, Institute of Basic Medical Sciences of Xiyuan Hospital, China Academy of Chinese Medical Sciences, National Clinical Research Center of Traditional Chinese Medicine for Cardiovascular Diseases, Beijing, 100091, China
| | - Lili Yang
- Beijing Key Laboratory of Chinese Materia Pharmacology, Institute of Basic Medical Sciences of Xiyuan Hospital, China Academy of Chinese Medical Sciences, National Clinical Research Center of Traditional Chinese Medicine for Cardiovascular Diseases, Beijing, 100091, China
| | - Jianshu Song
- Beijing Key Laboratory of Chinese Materia Pharmacology, Institute of Basic Medical Sciences of Xiyuan Hospital, China Academy of Chinese Medical Sciences, National Clinical Research Center of Traditional Chinese Medicine for Cardiovascular Diseases, Beijing, 100091, China
| | - Zixin Liu
- Beijing Key Laboratory of Chinese Materia Pharmacology, Institute of Basic Medical Sciences of Xiyuan Hospital, China Academy of Chinese Medical Sciences, National Clinical Research Center of Traditional Chinese Medicine for Cardiovascular Diseases, Beijing, 100091, China
| | - Haoran Li
- Beijing Key Laboratory of Chinese Materia Pharmacology, Institute of Basic Medical Sciences of Xiyuan Hospital, China Academy of Chinese Medical Sciences, National Clinical Research Center of Traditional Chinese Medicine for Cardiovascular Diseases, Beijing, 100091, China
| | - Lei Li
- Beijing Key Laboratory of Chinese Materia Pharmacology, Institute of Basic Medical Sciences of Xiyuan Hospital, China Academy of Chinese Medical Sciences, National Clinical Research Center of Traditional Chinese Medicine for Cardiovascular Diseases, Beijing, 100091, China
| | - Jianhua Fu
- Beijing Key Laboratory of Chinese Materia Pharmacology, Institute of Basic Medical Sciences of Xiyuan Hospital, China Academy of Chinese Medical Sciences, National Clinical Research Center of Traditional Chinese Medicine for Cardiovascular Diseases, Beijing, 100091, China
| | - Jianxun Liu
- Beijing Key Laboratory of Chinese Materia Pharmacology, Institute of Basic Medical Sciences of Xiyuan Hospital, China Academy of Chinese Medical Sciences, National Clinical Research Center of Traditional Chinese Medicine for Cardiovascular Diseases, Beijing, 100091, China.
| |
Collapse
|
2
|
Scalise M, Cianflone E, Quercia C, Pagano L, Chiefalo A, Stincelli A, Torella A, Puccio B, Santamaria G, Guzzi HP, Veltri P, De Angelis A, Urbanek K, Ellison-Hughes GM, Torella D, Marino F. Senolytics rejuvenate aging cardiomyopathy in human cardiac organoids. Mech Ageing Dev 2025; 223:112007. [PMID: 39622416 DOI: 10.1016/j.mad.2024.112007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2024] [Revised: 11/19/2024] [Accepted: 11/21/2024] [Indexed: 12/10/2024]
Abstract
BACKGROUND Human cardiac organoids closely replicate the architecture and function of the human heart, offering a potential accurate platform for studying cellular and molecular features of aging cardiomyopathy. Senolytics have shown potential in addressing age-related pathologies but their potential to reverse aging-related human cardiomyopathy remains largely unexplored. METHODS We employed human iPSC-derived cardiac organoids (hCOs/hCardioids) to model doxorubicin(DOXO)-induced cardiomyopathy in an aged context. hCardioids were treated with DOXO and subsequently with a combination of two senolytics: dasatinib (D) and quercetin (Q). RESULTS DOXO-treated hCardioids exhibited significantly increased oxidative stress, DNA damage (pH2AX), cellular senescence (p16INK4A) and decreased cell proliferation associated with a senescence-associated secretory phenotype (SASP). DOXO-treated hCardioids were considerably deprived of cardiac progenitors and displayed reduced cardiomyocyte proliferation as well as contractility. These distinctive aging-associated characteristics were confirmed by global RNA-sequencing analysis. Treatment with D+Q reversed these effects, reducing oxidative stress and senescence markers, alleviating SASP, and restoring hCardioids viability and function. Additionally, senolytics replenished cardiac progenitors and reversed the cardiomyocyte proliferation deficit. CONCLUSIONS Doxorubicin triggers an age-associated phenotype in hCardioids reliably modelling the main cellular and molecular features of aging cardiomyopathy. Senescence is a key mechanism of the aged-hCOs phenotype as senolytics rejuvenated aged-hCardioids restoring their structure and function while reverting the age-associated regenerative deficit.
Collapse
Affiliation(s)
- Mariangela Scalise
- Department of Experimental and Clinical Medicine, Magna Graecia University, Catanzaro 88100, Italy; Centre for Human and Applied Physiological, School of Basic and Medical Biosciences, Faculty of Life Sciences and Medicine, King's College London, London, UK
| | - Eleonora Cianflone
- Department of Medical and Surgical Sciences, Magna Graecia University, Catanzaro 88100, Italy.
| | - Claudia Quercia
- Department of Medical and Surgical Sciences, Magna Graecia University, Catanzaro 88100, Italy
| | - Loredana Pagano
- Department of Experimental and Clinical Medicine, Magna Graecia University, Catanzaro 88100, Italy
| | - Antonio Chiefalo
- Department of Experimental and Clinical Medicine, Magna Graecia University, Catanzaro 88100, Italy
| | - Antonio Stincelli
- Department of Medical and Surgical Sciences, Magna Graecia University, Catanzaro 88100, Italy
| | - Annalaura Torella
- Department of Experimental Medicine, University of Campania "L. Vanvitelli", Naples 80138, Italy
| | - Barbara Puccio
- Department of Medical and Surgical Sciences, Magna Graecia University, Catanzaro 88100, Italy
| | - Gianluca Santamaria
- Department of Experimental and Clinical Medicine, Magna Graecia University, Catanzaro 88100, Italy
| | - Hiram P Guzzi
- Department of Medical and Surgical Sciences, Magna Graecia University, Catanzaro 88100, Italy
| | - Pierangelo Veltri
- DIMES Department of Informatics, Modeling, Electronics and Systems, UNICAL, Rende, Cosenza, Italy
| | - Antonella De Angelis
- Department of Experimental Medicine, University of Campania "L. Vanvitelli", Naples 80138, Italy
| | - Konrad Urbanek
- Department of Molecular Medicine and Medical Biotechnologies, University of Naples "Federico II", and CEINGE-Advanced Biotechnologies, Naples 80131, Italy
| | - Georgina M Ellison-Hughes
- Centre for Human and Applied Physiological, School of Basic and Medical Biosciences, Faculty of Life Sciences and Medicine, King's College London, London, UK
| | - Daniele Torella
- Department of Experimental and Clinical Medicine, Magna Graecia University, Catanzaro 88100, Italy.
| | - Fabiola Marino
- Department of Experimental and Clinical Medicine, Magna Graecia University, Catanzaro 88100, Italy
| |
Collapse
|
3
|
Deszcz I. Stem Cell-Based Therapy and Cell-Free Therapy as an Alternative Approach for Cardiac Regeneration. Stem Cells Int 2023; 2023:2729377. [PMID: 37954462 PMCID: PMC10635745 DOI: 10.1155/2023/2729377] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2023] [Revised: 06/21/2023] [Accepted: 10/10/2023] [Indexed: 11/14/2023] Open
Abstract
The World Health Organization reports that cardiovascular diseases (CVDs) represent 32% of all global deaths. The ineffectiveness of conventional therapies in CVDs encourages the development of novel, minimally invasive therapeutic strategies for the healing and regeneration of damaged tissue. The self-renewal capacity, multilineage differentiation, lack of immunogenicity, and immunosuppressive properties of mesenchymal stem cells (MSCs) make them a promising option for CVDs. However, growing evidence suggests that myocardial regeneration occurs through paracrine factors and extracellular vesicle (EV) secretion, rather than through differentiation into cardiomyocytes. Research shows that stem cells secrete or surface-shed into their culture media various cytokines, chemokines, growth factors, anti-inflammatory factors, and EVs, which constitute an MSC-conditioned medium (MSC-CM) or the secretome. The use of MSC-CM enhances cardiac repair through resident heart cell differentiation, proliferation, scar mass reduction, a decrease in infarct wall thickness, and cardiac function improvement comparable to MSCs without their side effects. This review highlights the limitations and benefits of therapies based on stem cells and their secretome as an innovative treatment of CVDs.
Collapse
Affiliation(s)
- Iwona Deszcz
- Department of Immunopathology and Molecular Biology, Wroclaw Medical University, Borowska 211, 50-556, Wroclaw, Poland
| |
Collapse
|
4
|
Torella D, Cianflone E. Science Is a Self-Correcting Discipline: Revisiting the Biological Potential of Adult Cardiac Progenitors. Tex Heart Inst J 2023; 50:e238241. [PMID: 37859618 PMCID: PMC10658147 DOI: 10.14503/thij-23-8241] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2023]
Affiliation(s)
- Daniele Torella
- Department of Experimental and Clinical Medicine, Magna Graecia University, Catanzaro, Italy
| | - Eleonora Cianflone
- Department of Medical and Surgical Sciences, Magna Graecia University, Catanzaro, Italy
| |
Collapse
|
5
|
Scalise M, Marino F, Salerno L, Amato N, Quercia C, Siracusa C, Filardo A, Chiefalo A, Pagano L, Misdea G, Salerno N, De Angelis A, Urbanek K, Viglietto G, Torella D, Cianflone E. Adult Multipotent Cardiac Progenitor-Derived Spheroids: A Reproducible Model of In Vitro Cardiomyocyte Commitment and Specification. Cells 2023; 12:1793. [PMID: 37443827 PMCID: PMC10341123 DOI: 10.3390/cells12131793] [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: 05/04/2023] [Revised: 06/16/2023] [Accepted: 07/03/2023] [Indexed: 07/15/2023] Open
Abstract
BACKGROUND Three-dimensional cell culture systems hold great promise for bridging the gap between in vitro cell-based model systems and small animal models to study tissue biology and disease. Among 3D cell culture systems, stem-cell-derived spheroids have attracted significant interest as a strategy to better mimic in vivo conditions. Cardiac stem cell/progenitor (CSC)-derived spheroids (CSs) provide a relevant platform for cardiac regeneration. METHODS We compared three different cell culture scaffold-free systems, (i) ultra-low attachment plates, (ii) hanging drops (both requiring a 2D/3D switch), and (iii) agarose micro-molds (entirely 3D), for CSC-derived CS formation and their cardiomyocyte commitment in vitro. RESULTS The switch from a 2D to a 3D culture microenvironment per se guides cell plasticity and myogenic differentiation within CS and is necessary for robust cardiomyocyte differentiation. On the contrary, 2D monolayer CSC cultures show a significant reduced cardiomyocyte differentiation potential compared to 3D CS culture. Forced aggregation into spheroids using hanging drop improves CS myogenic differentiation when compared to ultra-low attachment plates. Performing CS formation and myogenic differentiation exclusively in 3D culture using agarose micro-molds maximizes the cardiomyocyte yield. CONCLUSIONS A 3D culture system instructs CS myogenic differentiation, thus representing a valid model that can be used to study adult cardiac regenerative biology.
Collapse
Affiliation(s)
- Mariangela Scalise
- Department of Experimental and Clinical Medicine, Magna Graecia University, 88100 Catanzaro, Italy; (M.S.); (F.M.); (L.S.); (A.C.); (G.M.); (N.S.); (G.V.)
| | - Fabiola Marino
- Department of Experimental and Clinical Medicine, Magna Graecia University, 88100 Catanzaro, Italy; (M.S.); (F.M.); (L.S.); (A.C.); (G.M.); (N.S.); (G.V.)
| | - Luca Salerno
- Department of Experimental and Clinical Medicine, Magna Graecia University, 88100 Catanzaro, Italy; (M.S.); (F.M.); (L.S.); (A.C.); (G.M.); (N.S.); (G.V.)
| | - Nunzia Amato
- Department of Medical and Surgical Sciences, Magna Graecia University, 88100 Catanzaro, Italy; (N.A.); (C.Q.); (C.S.); (A.F.); (L.P.)
| | - Claudia Quercia
- Department of Medical and Surgical Sciences, Magna Graecia University, 88100 Catanzaro, Italy; (N.A.); (C.Q.); (C.S.); (A.F.); (L.P.)
| | - Chiara Siracusa
- Department of Medical and Surgical Sciences, Magna Graecia University, 88100 Catanzaro, Italy; (N.A.); (C.Q.); (C.S.); (A.F.); (L.P.)
| | - Andrea Filardo
- Department of Medical and Surgical Sciences, Magna Graecia University, 88100 Catanzaro, Italy; (N.A.); (C.Q.); (C.S.); (A.F.); (L.P.)
| | - Antonio Chiefalo
- Department of Experimental and Clinical Medicine, Magna Graecia University, 88100 Catanzaro, Italy; (M.S.); (F.M.); (L.S.); (A.C.); (G.M.); (N.S.); (G.V.)
| | - Loredana Pagano
- Department of Medical and Surgical Sciences, Magna Graecia University, 88100 Catanzaro, Italy; (N.A.); (C.Q.); (C.S.); (A.F.); (L.P.)
| | - Giuseppe Misdea
- Department of Experimental and Clinical Medicine, Magna Graecia University, 88100 Catanzaro, Italy; (M.S.); (F.M.); (L.S.); (A.C.); (G.M.); (N.S.); (G.V.)
| | - Nadia Salerno
- Department of Experimental and Clinical Medicine, Magna Graecia University, 88100 Catanzaro, Italy; (M.S.); (F.M.); (L.S.); (A.C.); (G.M.); (N.S.); (G.V.)
| | - Antonella De Angelis
- Department of Experimental Medicine, University of Campania “L. Vanvitelli”, 80138 Naples, Italy;
| | - Konrad Urbanek
- Department of Molecular Medicine and Medical Biotechnology, Federico II University, 88121 Naples, Italy;
| | - Giuseppe Viglietto
- Department of Experimental and Clinical Medicine, Magna Graecia University, 88100 Catanzaro, Italy; (M.S.); (F.M.); (L.S.); (A.C.); (G.M.); (N.S.); (G.V.)
| | - Daniele Torella
- Department of Experimental and Clinical Medicine, Magna Graecia University, 88100 Catanzaro, Italy; (M.S.); (F.M.); (L.S.); (A.C.); (G.M.); (N.S.); (G.V.)
| | - Eleonora Cianflone
- Department of Medical and Surgical Sciences, Magna Graecia University, 88100 Catanzaro, Italy; (N.A.); (C.Q.); (C.S.); (A.F.); (L.P.)
| |
Collapse
|
6
|
Molinaro C, Scalise M, Leo I, Salerno L, Sabatino J, Salerno N, De Rosa S, Torella D, Cianflone E, Marino F. Polarizing Macrophage Functional Phenotype to Foster Cardiac Regeneration. Int J Mol Sci 2023; 24:10747. [PMID: 37445929 DOI: 10.3390/ijms241310747] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2023] [Revised: 06/22/2023] [Accepted: 06/26/2023] [Indexed: 07/15/2023] Open
Abstract
There is an increasing interest in understanding the connection between the immune and cardiovascular systems, which are highly integrated and communicate through finely regulated cross-talking mechanisms. Recent evidence has demonstrated that the immune system does indeed have a key role in the response to cardiac injury and in cardiac regeneration. Among the immune cells, macrophages appear to have a prominent role in this context, with different subtypes described so far that each have a specific influence on cardiac remodeling and repair. Similarly, there are significant differences in how the innate and adaptive immune systems affect the response to cardiac damage. Understanding all these mechanisms may have relevant clinical implications. Several studies have already demonstrated that stem cell-based therapies support myocardial repair. However, the exact role that cardiac macrophages and their modulation may have in this setting is still unclear. The current need to decipher the dual role of immunity in boosting both heart injury and repair is due, at least for a significant part, to unresolved questions related to the complexity of cardiac macrophage phenotypes. The aim of this review is to provide an overview on the role of the immune system, and of macrophages in particular, in the response to cardiac injury and to outline, through the modulation of the immune response, potential novel therapeutic strategies for cardiac regeneration.
Collapse
Affiliation(s)
- Claudia Molinaro
- Department of Medical and Surgical Sciences, Magna Graecia University, 88100 Catanzaro, Italy
| | - Mariangela Scalise
- Department of Experimental and Clinical Medicine, Magna Graecia University, 88100 Catanzaro, Italy
| | - Isabella Leo
- Department of Experimental and Clinical Medicine, Magna Graecia University, 88100 Catanzaro, Italy
| | - Luca Salerno
- Department of Experimental and Clinical Medicine, Magna Graecia University, 88100 Catanzaro, Italy
| | - Jolanda Sabatino
- Department of Experimental and Clinical Medicine, Magna Graecia University, 88100 Catanzaro, Italy
| | - Nadia Salerno
- Department of Experimental and Clinical Medicine, Magna Graecia University, 88100 Catanzaro, Italy
| | - Salvatore De Rosa
- Department of Medical and Surgical Sciences, Magna Graecia University, 88100 Catanzaro, Italy
| | - Daniele Torella
- Department of Experimental and Clinical Medicine, Magna Graecia University, 88100 Catanzaro, Italy
| | - Eleonora Cianflone
- Department of Medical and Surgical Sciences, Magna Graecia University, 88100 Catanzaro, Italy
| | - Fabiola Marino
- Department of Experimental and Clinical Medicine, Magna Graecia University, 88100 Catanzaro, Italy
| |
Collapse
|
7
|
Salerno N, Scalise M, Marino F, Filardo A, Chiefalo A, Panuccio G, Torella M, De Angelis A, De Rosa S, Ellison-Hughes GM, Urbanek K, Viglietto G, Torella D, Cianflone E. A Mouse Model of Dilated Cardiomyopathy Produced by Isoproterenol Acute Exposure Followed by 5-Fluorouracil Administration. J Cardiovasc Dev Dis 2023; 10:225. [PMID: 37367390 DOI: 10.3390/jcdd10060225] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2023] [Revised: 05/19/2023] [Accepted: 05/20/2023] [Indexed: 06/28/2023] Open
Abstract
Appropriate dilated cardiomyopathy (DCM) animal models are highly desirable considering the pathophysiological and clinical heterogeneity of DCM. Genetically modified mice are the most widely and intensively utilized research animals for DCM. However, to translate discoveries from basic science into new and personalized medical applications, research in non-genetically based DCM models remains a key issue. Here, we characterized a mouse model of non-ischemic DCM induced by a stepwise pharmacologic regime of Isoproterenol (ISO) high dose bolus followed by a low dose systemic injection of the chemotherapy agent, 5-Fluorouracil (5-FU). C57BL/6J mice were injected with ISO and, 3 days after, were randomly assigned to saline or 5-FU. Echocardiography and a strain analysis show that ISO + 5FU in mice induces progressive left ventricular (LV) dilation and reduced systolic function, along with diastolic dysfunction and a persistent global cardiac contractility depression through 56 days. While mice treated with ISO alone recover anatomically and functionally, ISO + 5-FU causes persistent cardiomyocyte death, ensuing in cardiomyocyte hypertrophy through 56 days. ISO + 5-FU-dependent damage was accompanied by significant myocardial disarray and fibrosis along with exaggerated oxidative stress, tissue inflammation and premature cell senescence accumulation. In conclusions, a combination of ISO + 5FU produces anatomical, histological and functional cardiac alterations typical of DCM, representing a widely available, affordable, and reproducible mouse model of this cardiomyopathy.
Collapse
Affiliation(s)
- Nadia Salerno
- Department of Experimental and Clinical Medicine, Magna Graecia University, 88100 Catanzaro, Italy
| | - Mariangela Scalise
- Department of Experimental and Clinical Medicine, Magna Graecia University, 88100 Catanzaro, Italy
| | - Fabiola Marino
- Department of Experimental and Clinical Medicine, Magna Graecia University, 88100 Catanzaro, Italy
| | - Andrea Filardo
- Department of Medical and Surgical Sciences, Magna Graecia University, 88100 Catanzaro, Italy
| | - Antonio Chiefalo
- Department of Experimental and Clinical Medicine, Magna Graecia University, 88100 Catanzaro, Italy
| | - Giuseppe Panuccio
- Department of Medical and Surgical Sciences, Magna Graecia University, 88100 Catanzaro, Italy
| | - Michele Torella
- Department of Experimental Medicine, University of Campania "L. Vanvitelli", 80138 Naples, Italy
| | - Antonella De Angelis
- Department of Experimental Medicine, University of Campania "L. Vanvitelli", 80138 Naples, Italy
| | - Salvatore De Rosa
- Department of Medical and Surgical Sciences, Magna Graecia University, 88100 Catanzaro, Italy
| | - Georgina M Ellison-Hughes
- Centre for Human and Applied Physiological Sciences, School of Basic and Medical Biosciences, Faculty of Life Sciences & Medicine, King's College London, Guy's Campus, London SE1 1UL, UK
| | - Konrad Urbanek
- Department of Molecular Medicine and Medical Biotechnology, Federico II University, 88121 Naples, Italy
| | - Giuseppe Viglietto
- Department of Experimental and Clinical Medicine, Magna Graecia University, 88100 Catanzaro, Italy
| | - Daniele Torella
- Department of Experimental and Clinical Medicine, Magna Graecia University, 88100 Catanzaro, Italy
| | - Eleonora Cianflone
- Department of Medical and Surgical Sciences, Magna Graecia University, 88100 Catanzaro, Italy
| |
Collapse
|
8
|
Renikunta HV, Lazarow K, Gong Y, Shukla PC, Nageswaran V, Giral H, Kratzer A, Opitz L, Engel FB, Haghikia A, Costantino S, Paneni F, von Kries JP, Streckfuss-Bömeke K, Landmesser U, Jakob P. Large-scale microRNA functional high-throughput screening identifies miR-515-3p and miR-519e-3p as inducers of human cardiomyocyte proliferation. iScience 2023; 26:106593. [PMID: 37250320 PMCID: PMC10214393 DOI: 10.1016/j.isci.2023.106593] [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: 09/06/2022] [Revised: 01/07/2023] [Accepted: 03/31/2023] [Indexed: 05/31/2023] Open
Abstract
Ischemic cardiomyopathy, driven by loss of cardiomyocytes and inadequate proliferative response, persists to be a major global health problem. Using a functional high-throughput screening, we assessed differential proliferative potential of 2019 miRNAs after transient hypoxia by transfecting both miR-inhibitor and miR-mimic libraries in human iPSC-CM. Whereas miR-inhibitors failed to enhance EdU uptake, overexpression of 28 miRNAs substantially induced proliferative activity in hiPSC-CM, with an overrepresentation of miRNAs belonging to the primate-specific C19MC-cluster. Two of these miRNAs, miR-515-3p and miR-519e-3p, increased markers of early and late mitosis, indicative of cell division, and substantially alter signaling pathways relevant for cardiomyocyte proliferation in hiPSC-CM.
Collapse
Affiliation(s)
- Harsha V. Renikunta
- Deutsches Herzzentrum der Charité, Department of Cardiology, Angiology and Intensive Care, Hindenburgdamm 30, 12203 Berlin, Germany
- Charité – Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Charitéplatz 1, 10117 Berlin, Germany
- DZHK (German Center for Cardiovascular Research), partner site Berlin, Berlin, Germany
| | - Katina Lazarow
- Leibniz-Institute for Molecular Pharmacology (FMP), Campus Berlin-Buch, Robert-Rössle-Strasse 10, 13125 Berlin, Germany
| | - Yiqi Gong
- Center for Translational and Experimental Cardiology (CTEC), Department of Cardiology, University Hospital Zurich, University of Zurich, Wagistrasse 12, 8952 Schlieren, Switzerland
| | - Praphulla Chandra Shukla
- Deutsches Herzzentrum der Charité, Department of Cardiology, Angiology and Intensive Care, Hindenburgdamm 30, 12203 Berlin, Germany
- Charité – Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Charitéplatz 1, 10117 Berlin, Germany
- School of Medical Science and Technology, Indian Institute of Technology, Kharagpur 721302, West Bengal, India
| | - Vanasa Nageswaran
- Deutsches Herzzentrum der Charité, Department of Cardiology, Angiology and Intensive Care, Hindenburgdamm 30, 12203 Berlin, Germany
- Charité – Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Charitéplatz 1, 10117 Berlin, Germany
- DZHK (German Center for Cardiovascular Research), partner site Berlin, Berlin, Germany
- Institute of Chemistry/Biochemistry, Thielallee 63, Freie Universität Berlin, 14195 Berlin, Germany
| | - Hector Giral
- Deutsches Herzzentrum der Charité, Department of Cardiology, Angiology and Intensive Care, Hindenburgdamm 30, 12203 Berlin, Germany
- Charité – Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Charitéplatz 1, 10117 Berlin, Germany
| | - Adelheid Kratzer
- Deutsches Herzzentrum der Charité, Department of Cardiology, Angiology and Intensive Care, Hindenburgdamm 30, 12203 Berlin, Germany
- Charité – Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Charitéplatz 1, 10117 Berlin, Germany
| | - Lennart Opitz
- Functional Genomics Center Zurich UZH/ETH, ETH Zurich and University of Zurich, 8057 Zurich, Switzerland
| | - Felix B. Engel
- Experimental Renal and Cardiovascular Research, Department of Nephropathology, Institute of Pathology, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Schwabachanlage 12 (TRC), 91054 Erlangen, Germany
| | - Arash Haghikia
- Deutsches Herzzentrum der Charité, Department of Cardiology, Angiology and Intensive Care, Hindenburgdamm 30, 12203 Berlin, Germany
- Charité – Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Charitéplatz 1, 10117 Berlin, Germany
- DZHK (German Center for Cardiovascular Research), partner site Berlin, Berlin, Germany
- Berlin Institute of Health (BIH) at Charité – Universitätsmedizin Berlin, Charitéplatz 1, 10117 Berlin, Germany
| | - Sarah Costantino
- Center for Translational and Experimental Cardiology (CTEC), Department of Cardiology, University Hospital Zurich, University of Zurich, Wagistrasse 12, 8952 Schlieren, Switzerland
| | - Francesco Paneni
- Center for Translational and Experimental Cardiology (CTEC), Department of Cardiology, University Hospital Zurich, University of Zurich, Wagistrasse 12, 8952 Schlieren, Switzerland
| | - Jens Peter von Kries
- Leibniz-Institute for Molecular Pharmacology (FMP), Campus Berlin-Buch, Robert-Rössle-Strasse 10, 13125 Berlin, Germany
| | - Katrin Streckfuss-Bömeke
- Clinic for Cardiology and Pneumology, University Medical Center Göttingen, Robert-Koch-Strasse 40, 37075 Göttingen, Germany
- DZHK (German Center for Cardiovascular Research), partner site Göttingen, Robert-Koch-Strasse 42a, 37075 Göttingen, Germany
- Institute of Pharmacology and Toxicology, University of Würzburg, Versbacher Str. 9, 97078 Würzburg, Germany
| | - Ulf Landmesser
- Deutsches Herzzentrum der Charité, Department of Cardiology, Angiology and Intensive Care, Hindenburgdamm 30, 12203 Berlin, Germany
- Charité – Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Charitéplatz 1, 10117 Berlin, Germany
- DZHK (German Center for Cardiovascular Research), partner site Berlin, Berlin, Germany
- Berlin Institute of Health (BIH) at Charité – Universitätsmedizin Berlin, Charitéplatz 1, 10117 Berlin, Germany
| | - Philipp Jakob
- Deutsches Herzzentrum der Charité, Department of Cardiology, Angiology and Intensive Care, Hindenburgdamm 30, 12203 Berlin, Germany
- Charité – Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Charitéplatz 1, 10117 Berlin, Germany
- DZHK (German Center for Cardiovascular Research), partner site Berlin, Berlin, Germany
- Center for Translational and Experimental Cardiology (CTEC), Department of Cardiology, University Hospital Zurich, University of Zurich, Wagistrasse 12, 8952 Schlieren, Switzerland
- Berlin Institute of Health (BIH) at Charité – Universitätsmedizin Berlin, Charitéplatz 1, 10117 Berlin, Germany
| |
Collapse
|
9
|
Marino F, Salerno N, Scalise M, Salerno L, Torella A, Molinaro C, Chiefalo A, Filardo A, Siracusa C, Panuccio G, Ferravante C, Giurato G, Rizzo F, Torella M, Donniacuo M, De Angelis A, Viglietto G, Urbanek K, Weisz A, Torella D, Cianflone E. Streptozotocin-Induced Type 1 and 2 Diabetes Mellitus Mouse Models Show Different Functional, Cellular and Molecular Patterns of Diabetic Cardiomyopathy. Int J Mol Sci 2023; 24:ijms24021132. [PMID: 36674648 PMCID: PMC9860590 DOI: 10.3390/ijms24021132] [Citation(s) in RCA: 26] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2022] [Revised: 01/01/2023] [Accepted: 01/03/2023] [Indexed: 01/11/2023] Open
Abstract
The main cause of morbidity and mortality in diabetes mellitus (DM) is cardiovascular complications. Diabetic cardiomyopathy (DCM) remains incompletely understood. Animal models have been crucial in exploring DCM pathophysiology while identifying potential therapeutic targets. Streptozotocin (STZ) has been widely used to produce experimental models of both type 1 and type 2 DM (T1DM and T2DM). Here, we compared these two models for their effects on cardiac structure, function and transcriptome. Different doses of STZ and diet chows were used to generate T1DM and T2DM in C57BL/6J mice. Normal euglycemic and nonobese sex- and age-matched mice served as controls (CTRL). Immunohistochemistry, RT-PCR and RNA-seq were employed to compare hearts from the three animal groups. STZ-induced T1DM and T2DM affected left ventricular function and myocardial performance differently. T1DM displayed exaggerated apoptotic cardiomyocyte (CM) death and reactive hypertrophy and fibrosis, along with increased cardiac oxidative stress, CM DNA damage and senescence, when compared to T2DM in mice. T1DM and T2DM affected the whole cardiac transcriptome differently. In conclusion, the STZ-induced T1DM and T2DM mouse models showed significant differences in cardiac remodeling, function and the whole transcriptome. These differences could be of key relevance when choosing an animal model to study specific features of DCM.
Collapse
Affiliation(s)
- Fabiola Marino
- Department of Experimental and Clinical Medicine, Magna Graecia University, 88100 Catanzaro, Italy
| | - Nadia Salerno
- Department of Medical and Surgical Sciences, Magna Graecia University, 88100 Catanzaro, Italy
| | - Mariangela Scalise
- Department of Experimental and Clinical Medicine, Magna Graecia University, 88100 Catanzaro, Italy
| | - Luca Salerno
- Department of Experimental and Clinical Medicine, Magna Graecia University, 88100 Catanzaro, Italy
| | - Annalaura Torella
- Department of Precision Medicine, University of Campania “Luigi Vanvitelli”, 80138 Naples, Italy
| | - Claudia Molinaro
- Department of Medical and Surgical Sciences, Magna Graecia University, 88100 Catanzaro, Italy
| | - Antonio Chiefalo
- Department of Experimental and Clinical Medicine, Magna Graecia University, 88100 Catanzaro, Italy
| | - Andrea Filardo
- Department of Medical and Surgical Sciences, Magna Graecia University, 88100 Catanzaro, Italy
| | - Chiara Siracusa
- Department of Medical and Surgical Sciences, Magna Graecia University, 88100 Catanzaro, Italy
| | - Giuseppe Panuccio
- Department of Medical and Surgical Sciences, Magna Graecia University, 88100 Catanzaro, Italy
| | - Carlo Ferravante
- Department of Medicine, Surgery and Dentistry ‘Scuola Medica Salernitana′, University of Salerno, 84081 Salerno, Italy
| | - Giorgio Giurato
- Department of Medicine, Surgery and Dentistry ‘Scuola Medica Salernitana′, University of Salerno, 84081 Salerno, Italy
| | - Francesca Rizzo
- Department of Medicine, Surgery and Dentistry ‘Scuola Medica Salernitana′, University of Salerno, 84081 Salerno, Italy
| | - Michele Torella
- Department of Translational Medical Science, University of Campania “L. Vanvitelli”, 80138 Naples, Italy
| | - Maria Donniacuo
- Department of Experimental Medicine, University of Campania “L. Vanvitelli”, 80138 Naples, Italy
| | - Antonella De Angelis
- Department of Experimental Medicine, University of Campania “L. Vanvitelli”, 80138 Naples, Italy
| | - Giuseppe Viglietto
- Department of Experimental and Clinical Medicine, Magna Graecia University, 88100 Catanzaro, Italy
| | - Konrad Urbanek
- Department of Molecular Medicine and Medical Biotechnology, Federico II University, 88121 Naples, Italy
| | - Alessandro Weisz
- Department of Medicine, Surgery and Dentistry ‘Scuola Medica Salernitana′, University of Salerno, 84081 Salerno, Italy
| | - Daniele Torella
- Department of Experimental and Clinical Medicine, Magna Graecia University, 88100 Catanzaro, Italy
- Correspondence: (D.T.); (E.C.); Tel.: +39-0961369-7564 (D.T.); +39-0961369-4185 (E.C.)
| | - Eleonora Cianflone
- Department of Medical and Surgical Sciences, Magna Graecia University, 88100 Catanzaro, Italy
- Correspondence: (D.T.); (E.C.); Tel.: +39-0961369-7564 (D.T.); +39-0961369-4185 (E.C.)
| |
Collapse
|
10
|
Azizidoost S, Farzaneh M. MicroRNAs as a Novel Player for Differentiation of Mesenchymal Stem Cells into Cardiomyocytes. Curr Stem Cell Res Ther 2023; 18:27-34. [PMID: 35466882 DOI: 10.2174/1574888x17666220422094150] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2021] [Revised: 02/14/2022] [Accepted: 03/02/2022] [Indexed: 11/22/2022]
Abstract
Cardiovascular disease (CVD) is defined as a class of disorders affecting the heart and blood vessels. Cardiomyocytes and endothelial cells play important roles in cardiac regeneration and heart repair. However, the proliferating capacity of cardiomyocytes is limited. To overcome this issue, mesenchymal stem cells (MSCs) have emerged as an alternative strategy for CVD therapy. MSCs can proliferate and differentiate (or trans-differentiate) into cardiomyocytes. Several in vitro and in vivo differentiation protocols have been used to obtain MSCs-derived cardiomyocytes. It was recently investigated that microRNAs (miRNAs) by targeting several signaling pathways, including STAT3, Wnt/β-catenin, Notch, and TBX5, play a crucial role in regulating cardiomyocytes' differentiation of MSCs. In this review, we focused on the role of miRNAs in the differentiation of MSCs into cardiomyocytes.
Collapse
Affiliation(s)
- Shirin Azizidoost
- Atherosclerosis Research Center, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Maryam Farzaneh
- Fertility, Infertility and Perinatology Research Center, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran.,Cellular and Molecular Research Center, Medical Basic Sciences Research Institute, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| |
Collapse
|
11
|
Salerno N, Marino F, Scalise M, Salerno L, Molinaro C, Filardo A, Chiefalo A, Panuccio G, De Angelis A, Urbanek K, Torella D, Cianflone E. Pharmacological clearance of senescent cells improves cardiac remodeling and function after myocardial infarction in female aged mice. Mech Ageing Dev 2022; 208:111740. [PMID: 36150603 DOI: 10.1016/j.mad.2022.111740] [Citation(s) in RCA: 33] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2022] [Revised: 09/09/2022] [Accepted: 09/18/2022] [Indexed: 12/30/2022]
Abstract
Cardiovascular diseases (CVD) are predominantly an aging disease. Important sex-specific differences exist and the mechanism(s) by which this sex-by-age interaction influences CVD development and progression remains elusive. Accordingly, it is still unknown whether cell senescence, a main feature of cardiac male aging, is a significant feature also of the female aged mouse heart and whether senolytics, senescence-clearing compounds, promote myocardial repair and regeneration after myocardial infarction (MI) in aged female mice. To this aim, the combination of two senolytics, dasatinib and quercetin (D+Q) or just their vehicle was administered to 22-24 months old C57BL/6 female mice after MI. D+Q improved global left ventricle function and myocardial performance after MI whereby female cardiac aging is characterized by accumulation of cardiac senescent cells that are further increased by MI. Despite their terminal differentiation nature, also cardiomyocytes acquire a senescent phenotype with age in females. D+Q removed senescent cardiac non-myocyte and myocyte cells ameliorating cardiac remodeling and regeneration. Senolytics removed aged dysfunctional cardiac stem/progenitor cells (CSCs), relieving healthy CSCs with normal proliferative and cardiomyogenic differentiation potential. In conclusions, cardiac senescent cells accumulate in the aged female hearts. Removing senescent cells is a key therapeutic target for efficient repair of the aged female heart.
Collapse
Affiliation(s)
- Nadia Salerno
- Department of Medical and Surgical Sciences, Magna Graecia University, 88100 Catanzaro, Italy
| | - Fabiola Marino
- Department of Experimental and Clinical Medicine, Magna Graecia University, 88100 Catanzaro, Italy
| | - Mariangela Scalise
- Department of Experimental and Clinical Medicine, Magna Graecia University, 88100 Catanzaro, Italy
| | - Luca Salerno
- Department of Experimental and Clinical Medicine, Magna Graecia University, 88100 Catanzaro, Italy
| | - Claudia Molinaro
- Department of Medical and Surgical Sciences, Magna Graecia University, 88100 Catanzaro, Italy
| | - Andrea Filardo
- Department of Medical and Surgical Sciences, Magna Graecia University, 88100 Catanzaro, Italy
| | - Antonio Chiefalo
- Department of Experimental and Clinical Medicine, Magna Graecia University, 88100 Catanzaro, Italy
| | - Giuseppe Panuccio
- Department of Medical and Surgical Sciences, Magna Graecia University, 88100 Catanzaro, Italy
| | - Antonella De Angelis
- Department of Experimental Medicine, University of Campania "L. Vanvitelli", 80138 Naples, Italy
| | - Konrad Urbanek
- Department of Molecular Medicine and Medical Biotechnology, Federico II University, 88121, Naples, Italy
| | - Daniele Torella
- Department of Experimental and Clinical Medicine, Magna Graecia University, 88100 Catanzaro, Italy.
| | - Eleonora Cianflone
- Department of Medical and Surgical Sciences, Magna Graecia University, 88100 Catanzaro, Italy.
| |
Collapse
|
12
|
Bonavida V, Ghassemi K, Ung G, Inouye K, Thankam FG, Agrawal DK. Novel Approaches to Program Cells to Differentiate into Cardiomyocytes in Myocardial Regeneration. Rev Cardiovasc Med 2022; 23:392. [PMID: 39076655 PMCID: PMC11270456 DOI: 10.31083/j.rcm2312392] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2022] [Revised: 10/17/2022] [Accepted: 10/31/2022] [Indexed: 07/31/2024] Open
Abstract
With heart failure (HF) being one of the leading causes of hospitalization and death worldwide, multiple stem cell therapies have been attempted to accelerate the regeneration of the infarct zone. Versatile strategies have emerged to establish the cell candidates of cardiomyocyte lineage for regenerative cardiology. This article illustrates critical insights into the emerging technologies, current approaches, and translational promises on the programming of diverse cell types for cardiac regeneration.
Collapse
Affiliation(s)
- Victor Bonavida
- Department of Translational Research, Western University of Health Sciences, Pomona, CA 91766, USA
| | - Kaitlyn Ghassemi
- Department of Translational Research, Western University of Health Sciences, Pomona, CA 91766, USA
| | - Gwendolyn Ung
- Department of Translational Research, Western University of Health Sciences, Pomona, CA 91766, USA
| | - Keiko Inouye
- Department of Translational Research, Western University of Health Sciences, Pomona, CA 91766, USA
| | - Finosh G Thankam
- Department of Translational Research, Western University of Health Sciences, Pomona, CA 91766, USA
| | - Devendra K Agrawal
- Department of Translational Research, Western University of Health Sciences, Pomona, CA 91766, USA
| |
Collapse
|
13
|
The negative regulation of gene expression by microRNAs as key driver of inducers and repressors of cardiomyocyte differentiation. Clin Sci (Lond) 2022; 136:1179-1203. [PMID: 35979890 PMCID: PMC9411751 DOI: 10.1042/cs20220391] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2022] [Revised: 07/29/2022] [Accepted: 08/02/2022] [Indexed: 11/28/2022]
Abstract
Cardiac muscle damage-induced loss of cardiomyocytes (CMs) and dysfunction of the remaining ones leads to heart failure, which nowadays is the number one killer worldwide. Therapies fostering effective cardiac regeneration are the holy grail of cardiovascular research to stop the heart failure epidemic. The main goal of most myocardial regeneration protocols is the generation of new functional CMs through the differentiation of endogenous or exogenous cardiomyogenic cells. Understanding the cellular and molecular basis of cardiomyocyte commitment, specification, differentiation and maturation is needed to devise innovative approaches to replace the CMs lost after injury in the adult heart. The transcriptional regulation of CM differentiation is a highly conserved process that require sequential activation and/or repression of different genetic programs. Therefore, CM differentiation and specification have been depicted as a step-wise specific chemical and mechanical stimuli inducing complete myogenic commitment and cell-cycle exit. Yet, the demonstration that some microRNAs are sufficient to direct ESC differentiation into CMs and that four specific miRNAs reprogram fibroblasts into CMs show that CM differentiation must also involve negative regulatory instructions. Here, we review the mechanisms of CM differentiation during development and from regenerative stem cells with a focus on the involvement of microRNAs in the process, putting in perspective their negative gene regulation as a main modifier of effective CM regeneration in the adult heart.
Collapse
|
14
|
Salerno N, Salerno L, Marino F, Scalise M, Chiefalo A, Panuccio G, De Angelis A, Cianflone E, Urbanek K, Torella D. Myocardial regeneration protocols towards the routine clinical scenario: An unseemly path from bench to bedside. EClinicalMedicine 2022; 50:101530. [PMID: 35799845 PMCID: PMC9253597 DOI: 10.1016/j.eclinm.2022.101530] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/10/2022] [Revised: 05/31/2022] [Accepted: 06/06/2022] [Indexed: 12/11/2022] Open
Abstract
UNLABELLED Heart failure secondary to cardiomyocyte loss and/or dysfunction is the number one killer worldwide. The field of myocardial regeneration with its far-reaching primary goal of cardiac remuscularization and its hard-to-accomplish translation from bench to bedside, has been filled with ups and downs, steps forward and steps backward, controversies galore and, unfortunately, scientific scandals. Despite the present morass in which cardiac remuscularization is stuck in, the search for clinically effective regenerative approaches remains keenly active. Starting with a concise overview of the still highly debated regenerative capacity of the adult mammalian heart, we focus on the main interventions, that have reached or are close to clinical use, critically discussing key findings, successes, and failures. Finally, some promising and innovative approaches for myocardial repair/regeneration still at the pre-clinical stage are discussed to offer a holistic view on the future of myocardial repair/regeneration for the prevention/management of heart failure in the clinical scenario. FUNDING This research was funded by Grants from the Ministry of University and Research PRIN2015 2015ZTT5KB_004; PRIN2017NKB2N4_005; PON-AIM - 1829805-2.
Collapse
Affiliation(s)
- Nadia Salerno
- Department of Medical and Surgical Sciences, Magna Graecia University, 88100, Catanzaro, Italy
| | - Luca Salerno
- Department of Experimental and Clinical Medicine, Magna Graecia University, 88100, Catanzaro, Italy
| | - Fabiola Marino
- Department of Experimental and Clinical Medicine, Magna Graecia University, 88100, Catanzaro, Italy
| | - Mariangela Scalise
- Department of Experimental and Clinical Medicine, Magna Graecia University, 88100, Catanzaro, Italy
| | - Antonio Chiefalo
- Department of Experimental and Clinical Medicine, Magna Graecia University, 88100, Catanzaro, Italy
| | - Giuseppe Panuccio
- Department of Medical and Surgical Sciences, Magna Graecia University, 88100, Catanzaro, Italy
| | - Antonella De Angelis
- Department of Experimental Medicine, University of Campania “L. Vanvitelli”, 80138 Naples, Italy
| | - Eleonora Cianflone
- Department of Medical and Surgical Sciences, Magna Graecia University, 88100, Catanzaro, Italy
| | - Konrad Urbanek
- Department of Experimental and Clinical Medicine, Magna Graecia University, 88100, Catanzaro, Italy
- Department of Molecular Medicine and Medical Biotechnologies, University of Naples “Federico II”, 80125, Naples, Italy
| | - Daniele Torella
- Department of Experimental and Clinical Medicine, Magna Graecia University, 88100, Catanzaro, Italy
- Corresponding author.
| |
Collapse
|
15
|
Marino F, Scalise M, Salerno N, Salerno L, Molinaro C, Cappetta D, Torella M, Greco M, Foti D, Sasso FC, Mastroroberto P, De Angelis A, Ellison-Hughes GM, Sampaolesi M, Rota M, Rossi F, Urbanek K, Nadal-Ginard B, Torella D, Cianflone E. Diabetes-Induced Cellular Senescence and Senescence-Associated Secretory Phenotype Impair Cardiac Regeneration and Function Independently of Age. Diabetes 2022; 71:1081-1098. [PMID: 35108360 PMCID: PMC9490451 DOI: 10.2337/db21-0536] [Citation(s) in RCA: 43] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/23/2021] [Accepted: 01/30/2022] [Indexed: 11/13/2022]
Abstract
Diabetes mellitus (DM) affects the biology of multipotent cardiac stem/progenitor cells (CSCs) and adult myocardial regeneration. We assessed the hypothesis that senescence and senescence-associated secretory phenotype (SASP) are main mechanisms of cardiac degenerative defect in DM. Accordingly, we tested whether ablation of senescent CSCs would rescue the cardiac regenerative/reparative defect imposed by DM. We obtained cardiac tissue from nonaged (50- to 64-year-old) patients with type 2 diabetes mellitus (T2DM) and without DM (NDM) and postinfarct cardiomyopathy undergoing cardiac surgery. A higher reactive oxygen species production in T2DM was associated with an increased number of senescent/dysfunctional T2DM-human CSCs (hCSCs) with reduced proliferation, clonogenesis/spherogenesis, and myogenic differentiation versus NDM-hCSCs in vitro. T2DM-hCSCs showed a defined pathologic SASP. A combination of two senolytics, dasatinib (D) and quercetin (Q), cleared senescent T2DM-hCSCs in vitro, restoring their expansion and myogenic differentiation capacities. In a T2DM model in young mice, diabetic status per se (independently of ischemia and age) caused CSC senescence coupled with myocardial pathologic remodeling and cardiac dysfunction. D + Q treatment efficiently eliminated senescent cells, rescuing CSC function, which resulted in functional myocardial repair/regeneration, improving cardiac function in murine DM. In conclusion, DM hampers CSC biology, inhibiting CSCs' regenerative potential through the induction of cellular senescence and SASP independently from aging. Senolytics clear senescence, abrogating the SASP and restoring a fully proliferative/differentiation-competent hCSC pool in T2DM with normalization of cardiac function.
Collapse
Affiliation(s)
- Fabiola Marino
- Department of Experimental and Clinical Medicine, Magna Græcia University, Catanzaro, Italy
- Translational Cardiomyology Laboratory, Stem Cell Biology and Embryology, Department of Development and Regeneration, KU Leuven, Leuven, Belgium
| | - Mariangela Scalise
- Department of Experimental and Clinical Medicine, Magna Græcia University, Catanzaro, Italy
| | - Nadia Salerno
- Department of Medical and Surgical Sciences, Magna Græcia University, Catanzaro, Italy
| | - Luca Salerno
- Department of Experimental and Clinical Medicine, Magna Græcia University, Catanzaro, Italy
| | - Claudia Molinaro
- Department of Medical and Surgical Sciences, Magna Græcia University, Catanzaro, Italy
| | - Donato Cappetta
- Department of Experimental Medicine, Section of Pharmacology, University of Campania “L. Vanvitelli,” Naples, Italy
| | - Michele Torella
- Department of Translational Medicine, University of Campania “L. Vanvitelli,” Naples, Italy
| | - Marta Greco
- Department of Health Sciences, Magna Græcia University, Catanzaro, Italy
| | - Daniela Foti
- Department of Experimental and Clinical Medicine, Magna Græcia University, Catanzaro, Italy
| | - Ferdinando C. Sasso
- Department of Translational Medicine, University of Campania “L. Vanvitelli,” Naples, Italy
| | - Pasquale Mastroroberto
- Department of Experimental and Clinical Medicine, Magna Græcia University, Catanzaro, Italy
| | - Antonella De Angelis
- Department of Experimental Medicine, Section of Pharmacology, University of Campania “L. Vanvitelli,” Naples, Italy
| | - Georgina M. Ellison-Hughes
- Centre for Human and Applied Physiological Sciences and Centre for Stem Cells and Regenerative Medicine, School of Basic and Medical Biosciences, Faculty of Life Sciences and Medicine, King’s College London, London, U.K
| | - Maurilio Sampaolesi
- Translational Cardiomyology Laboratory, Stem Cell Biology and Embryology, Department of Development and Regeneration, KU Leuven, Leuven, Belgium
| | - Marcello Rota
- Department of Physiology, New York Medical College, Valhalla, NY
| | - Francesco Rossi
- Department of Experimental Medicine, Section of Pharmacology, University of Campania “L. Vanvitelli,” Naples, Italy
| | - Konrad Urbanek
- Department of Experimental and Clinical Medicine, Magna Græcia University, Catanzaro, Italy
| | - Bernardo Nadal-Ginard
- Department of Medical and Surgical Sciences, Magna Græcia University, Catanzaro, Italy
| | - Daniele Torella
- Department of Experimental and Clinical Medicine, Magna Græcia University, Catanzaro, Italy
- Corresponding authors: Daniele Torella, , and Eleonora Cianflone,
| | - Eleonora Cianflone
- Department of Medical and Surgical Sciences, Magna Græcia University, Catanzaro, Italy
- Department of Physiology, New York Medical College, Valhalla, NY
- Corresponding authors: Daniele Torella, , and Eleonora Cianflone,
| |
Collapse
|
16
|
Mehanna RA, Essawy MM, Barkat MA, Awaad AK, Thabet EH, Hamed HA, Elkafrawy H, Khalil NA, Sallam A, Kholief MA, Ibrahim SS, Mourad GM. Cardiac stem cells: Current knowledge and future prospects. World J Stem Cells 2022; 14:1-40. [PMID: 35126826 PMCID: PMC8788183 DOI: 10.4252/wjsc.v14.i1.1] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/26/2021] [Revised: 07/02/2021] [Accepted: 01/06/2022] [Indexed: 02/06/2023] Open
Abstract
Regenerative medicine is the field concerned with the repair and restoration of the integrity of damaged human tissues as well as whole organs. Since the inception of the field several decades ago, regenerative medicine therapies, namely stem cells, have received significant attention in preclinical studies and clinical trials. Apart from their known potential for differentiation into the various body cells, stem cells enhance the organ's intrinsic regenerative capacity by altering its environment, whether by exogenous injection or introducing their products that modulate endogenous stem cell function and fate for the sake of regeneration. Recently, research in cardiology has highlighted the evidence for the existence of cardiac stem and progenitor cells (CSCs/CPCs). The global burden of cardiovascular diseases' morbidity and mortality has demanded an in-depth understanding of the biology of CSCs/CPCs aiming at improving the outcome for an innovative therapeutic strategy. This review will discuss the nature of each of the CSCs/CPCs, their environment, their interplay with other cells, and their metabolism. In addition, important issues are tackled concerning the potency of CSCs/CPCs in relation to their secretome for mediating the ability to influence other cells. Moreover, the review will throw the light on the clinical trials and the preclinical studies using CSCs/CPCs and combined therapy for cardiac regeneration. Finally, the novel role of nanotechnology in cardiac regeneration will be explored.
Collapse
Affiliation(s)
- Radwa A Mehanna
- Medical Physiology Department/Center of Excellence for Research in Regenerative Medicine and Applications, Faculty of Medicine, Alexandria University, Alexandria 21500, Egypt
| | - Marwa M Essawy
- Oral Pathology Department, Faculty of Dentistry/Center of Excellence for Research in Regenerative Medicine and Applications, Faculty of Medicine, Alexandria University, Alexandria 21500, Egypt
| | - Mona A Barkat
- Human Anatomy and Embryology Department/Center of Excellence for Research in Regenerative Medicine and Applications, Faculty of Medicine, Alexandria University, Alexandria 21500, Egypt
| | - Ashraf K Awaad
- Center of Excellence for Research in Regenerative Medicine and Applications, Faculty of Medicine, Alexandria University, Alexandria 21500, Egypt
| | - Eman H Thabet
- Medical Physiology Department/Center of Excellence for Research in Regenerative Medicine and Applications, Faculty of Medicine, Alexandria University, Alexandria 21500, Egypt
| | - Heba A Hamed
- Histology and Cell Biology Department/Center of Excellence for Research in Regenerative Medicine and Applications, Faculty of Medicine, Alexandria University, Alexandria 21500, Egypt
| | - Hagar Elkafrawy
- Medical Biochemistry Department/Center of Excellence for Research in Regenerative Medicine and Applications, Faculty of Medicine, Alexandria University, Alexandria 21500, Egypt
| | - Nehal A Khalil
- Medical Biochemistry Department/Center of Excellence for Research in Regenerative Medicine and Applications, Faculty of Medicine, Alexandria University, Alexandria 21500, Egypt
| | - Abeer Sallam
- Medical Physiology Department/Center of Excellence for Research in Regenerative Medicine and Applications, Faculty of Medicine, Alexandria University, Alexandria 21500, Egypt
| | - Marwa A Kholief
- Forensic Medicine and Clinical toxicology Department/Center of Excellence for Research in Regenerative Medicine and Applications, Faculty of Medicine, Alexandria University, Alexandria 21500, Egypt
| | - Samar S Ibrahim
- Center of Excellence for Research in Regenerative Medicine and Applications, Faculty of Medicine, Alexandria University, Alexandria 21500, Egypt
| | - Ghada M Mourad
- Histology and Cell Biology Department/Center of Excellence for Research in Regenerative Medicine and Applications, Faculty of Medicine, Alexandria University, Alexandria 21500, Egypt.
| |
Collapse
|
17
|
Mehanna RA, Essawy MM, Barkat MA, Awaad AK, Thabet EH, Hamed HA, Elkafrawy H, Khalil NA, Sallam A, Kholief MA, Ibrahim SS, Mourad GM. Cardiac stem cells: Current knowledge and future prospects. World J Stem Cells 2022. [PMID: 35126826 DOI: 10.4252/wjsc.v14.i1.1]] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Regenerative medicine is the field concerned with the repair and restoration of the integrity of damaged human tissues as well as whole organs. Since the inception of the field several decades ago, regenerative medicine therapies, namely stem cells, have received significant attention in preclinical studies and clinical trials. Apart from their known potential for differentiation into the various body cells, stem cells enhance the organ's intrinsic regenerative capacity by altering its environment, whether by exogenous injection or introducing their products that modulate endogenous stem cell function and fate for the sake of regeneration. Recently, research in cardiology has highlighted the evidence for the existence of cardiac stem and progenitor cells (CSCs/CPCs). The global burden of cardiovascular diseases' morbidity and mortality has demanded an in-depth understanding of the biology of CSCs/CPCs aiming at improving the outcome for an innovative therapeutic strategy. This review will discuss the nature of each of the CSCs/CPCs, their environment, their interplay with other cells, and their metabolism. In addition, important issues are tackled concerning the potency of CSCs/CPCs in relation to their secretome for mediating the ability to influence other cells. Moreover, the review will throw the light on the clinical trials and the preclinical studies using CSCs/CPCs and combined therapy for cardiac regeneration. Finally, the novel role of nanotechnology in cardiac regeneration will be explored.
Collapse
Affiliation(s)
- Radwa A Mehanna
- Medical Physiology Department/Center of Excellence for Research in Regenerative Medicine and Applications, Faculty of Medicine, Alexandria University, Alexandria 21500, Egypt
| | - Marwa M Essawy
- Oral Pathology Department, Faculty of Dentistry/Center of Excellence for Research in Regenerative Medicine and Applications, Faculty of Medicine, Alexandria University, Alexandria 21500, Egypt
| | - Mona A Barkat
- Human Anatomy and Embryology Department/Center of Excellence for Research in Regenerative Medicine and Applications, Faculty of Medicine, Alexandria University, Alexandria 21500, Egypt
| | - Ashraf K Awaad
- Center of Excellence for Research in Regenerative Medicine and Applications, Faculty of Medicine, Alexandria University, Alexandria 21500, Egypt
| | - Eman H Thabet
- Medical Physiology Department/Center of Excellence for Research in Regenerative Medicine and Applications, Faculty of Medicine, Alexandria University, Alexandria 21500, Egypt
| | - Heba A Hamed
- Histology and Cell Biology Department/Center of Excellence for Research in Regenerative Medicine and Applications, Faculty of Medicine, Alexandria University, Alexandria 21500, Egypt
| | - Hagar Elkafrawy
- Medical Biochemistry Department/Center of Excellence for Research in Regenerative Medicine and Applications, Faculty of Medicine, Alexandria University, Alexandria 21500, Egypt
| | - Nehal A Khalil
- Medical Biochemistry Department/Center of Excellence for Research in Regenerative Medicine and Applications, Faculty of Medicine, Alexandria University, Alexandria 21500, Egypt
| | - Abeer Sallam
- Medical Physiology Department/Center of Excellence for Research in Regenerative Medicine and Applications, Faculty of Medicine, Alexandria University, Alexandria 21500, Egypt
| | - Marwa A Kholief
- Forensic Medicine and Clinical toxicology Department/Center of Excellence for Research in Regenerative Medicine and Applications, Faculty of Medicine, Alexandria University, Alexandria 21500, Egypt
| | - Samar S Ibrahim
- Center of Excellence for Research in Regenerative Medicine and Applications, Faculty of Medicine, Alexandria University, Alexandria 21500, Egypt
| | - Ghada M Mourad
- Histology and Cell Biology Department/Center of Excellence for Research in Regenerative Medicine and Applications, Faculty of Medicine, Alexandria University, Alexandria 21500, Egypt.
| |
Collapse
|
18
|
Unraveling and Targeting Myocardial Regeneration Deficit in Diabetes. Antioxidants (Basel) 2022; 11:antiox11020208. [PMID: 35204091 PMCID: PMC8868283 DOI: 10.3390/antiox11020208] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2021] [Revised: 01/13/2022] [Accepted: 01/20/2022] [Indexed: 02/04/2023] Open
Abstract
Cardiomyopathy is a common complication in diabetic patients. Ventricular dysfunction without coronary atherosclerosis and hypertension is driven by hyperglycemia, hyperinsulinemia and impaired insulin signaling. Cardiomyocyte death, hypertrophy, fibrosis, and cell signaling defects underlie cardiomyopathy. Notably, detrimental effects of the diabetic milieu are not limited to cardiomyocytes and vascular cells. The diabetic heart acquires a senescent phenotype and also suffers from altered cellular homeostasis and the insufficient replacement of dying cells. Chronic inflammation, oxidative stress, and metabolic dysregulation damage the population of endogenous cardiac stem cells, which contribute to myocardial cell turnover and repair after injury. Therefore, deficient myocardial repair and the progressive senescence and dysfunction of stem cells in the diabetic heart can represent potential therapeutic targets. While our knowledge of the effects of diabetes on stem cells is growing, several strategies to preserve, activate or restore cardiac stem cell compartments await to be tested in diabetic cardiomyopathy.
Collapse
|
19
|
From Spheroids to Organoids: The Next Generation of Model Systems of Human Cardiac Regeneration in a Dish. Int J Mol Sci 2021; 22:ijms222413180. [PMID: 34947977 PMCID: PMC8708686 DOI: 10.3390/ijms222413180] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2021] [Revised: 12/02/2021] [Accepted: 12/05/2021] [Indexed: 12/12/2022] Open
Abstract
Organoids are tiny, self-organized, three-dimensional tissue cultures that are derived from the differentiation of stem cells. The growing interest in the use of organoids arises from their ability to mimic the biology and physiology of specific tissue structures in vitro. Organoids indeed represent promising systems for the in vitro modeling of tissue morphogenesis and organogenesis, regenerative medicine and tissue engineering, drug therapy testing, toxicology screening, and disease modeling. Although 2D cell cultures have been used for more than 50 years, even for their simplicity and low-cost maintenance, recent years have witnessed a steep rise in the availability of organoid model systems. Exploiting the ability of cells to re-aggregate and reconstruct the original architecture of an organ makes it possible to overcome many limitations of 2D cell culture systems. In vitro replication of the cellular micro-environment of a specific tissue leads to reproducing the molecular, biochemical, and biomechanical mechanisms that directly influence cell behavior and fate within that specific tissue. Lineage-specific self-organizing organoids have now been generated for many organs. Currently, growing cardiac organoid (cardioids) from pluripotent stem cells and cardiac stem/progenitor cells remains an open challenge due to the complexity of the spreading, differentiation, and migration of cardiac muscle and vascular layers. Here, we summarize the evolution of biological model systems from the generation of 2D spheroids to 3D organoids by focusing on the generation of cardioids based on the currently available laboratory technologies and outline their high potential for cardiovascular research.
Collapse
|