1
|
Zhang N, Li Y. Receptor tyrosine kinases: biological functions and anticancer targeted therapy. MedComm (Beijing) 2023; 4:e446. [PMID: 38077251 PMCID: PMC10701465 DOI: 10.1002/mco2.446] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2023] [Revised: 11/23/2023] [Accepted: 11/24/2023] [Indexed: 10/16/2024] Open
Abstract
Receptor tyrosine kinases (RTKs) are a class of protein kinases that play crucial roles in various cellular processes, including cell migration, morphological differentiation, cell growth, and angiogenesis. In humans, 58 RTKs have been identified and categorized into 20 distinct families based on the composition of their extracellular regions. RTKs are primarily activated by specific ligands that bind to their extracellular region. They not only regulate tumor transformation, proliferation, metastasis, drug resistance, and angiogenesis, but also initiate and maintain the self-renewal and cloning ability of cancer stem cells. Accurate diagnosis and grading of tumors with dysregulated RTKs are essential in clinical practice. There is a growing body of evidence supporting the benefits of RTKs-targeted therapies for cancer patients, and researchers are actively exploring new targets and developing targeted agents. However, further optimization of RTK inhibitors is necessary to effectively target the diverse RTK alterations observed in human cancers. This review provides insights into the classification, structure, activation mechanisms, and expression of RTKs in tumors. It also highlights the research advances in RTKs targeted anticancer therapy and emphasizes their significance in optimizing cancer diagnosis and treatment strategies.
Collapse
Affiliation(s)
- Nan Zhang
- Chongqing University Cancer Hospital, School of MedicineChongqing UniversityChongqingChina
| | - Yongsheng Li
- Chongqing University Cancer Hospital, School of MedicineChongqing UniversityChongqingChina
- Department of Medical OncologyChongqing University Cancer HospitalChongqingChina
| |
Collapse
|
2
|
Schwarzkopf L, Büttner P, Scholtyssek K, Schröter T, Hiller R, Hindricks G, Bollmann A, Laufs U, Ueberham L. C-kit pos cells in the human left atrial appendage. Heliyon 2023; 9:e21268. [PMID: 37954289 PMCID: PMC10637945 DOI: 10.1016/j.heliyon.2023.e21268] [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: 07/14/2023] [Revised: 10/09/2023] [Accepted: 10/18/2023] [Indexed: 11/14/2023] Open
Abstract
Background Subpopulations of myocardial c-kitpos cells have the ability to stimulate regeneration in ischemic heart disease by paracrine effects. The left atrial appendage (LAA), which is easy accessible during cardiac surgery, may represent a perfect source for c-kitpos cell extraction for autologous cell therapies in the living human. So far, frequency and distribution of c-kitpos cells in LAA are unknown. Methods LAAs of patients who underwent cardiac surgery due to coronary artery disease (coronary artery bypass graft, CABG), valvular heart disease or both and of two body donors were examined. Tissue was fixed in 4 % paraformaldehyde, embedded in paraffin, dissected in consecutive sections and stained for c-kitpos cells. In parallel, grade of fibrosis, amount of fat per section and cells positive for mast cell tryptase were examined. Results We collected 27 LAAs (37.0 % female, mean left ventricular ejection fraction 50.4 %, 63.0 % persistent atrial fibrillation (AF)). Most of the patients underwent combined CABG and valve surgery (51.9 %). C-kitpos cells were detected in 3 different regions: A) Attached to the epicardial fat layer, B) close to vascular structures and C) between cardiomyocytes. C-kitpos cells ranged from 0.05 c-kitpos cells per mm2 to 67.5 c-kitpos cells per mm2. We found no association between number of c-kitpos cells and type of AF, amount of fibrosis or amount of fat. Up to 72 % of c-kitpos cells also showed a positive staining for mast cell tryptase. Conclusion C-kitpos cells are frequent in LAAs of cardiovascular patients with a rather homogenous distribution throughout the LAA. The LAA can therefore be considered as a source for extraction of a reasonable quantity of autologous cardiac progenitor cells in the living human patient.
Collapse
Affiliation(s)
- Lea Schwarzkopf
- St. Elisabeth-Krankenhaus Leipzig, Department of Anaesthesiology, Leipzig, Germany
- German Heart Center Berlin, Department of Electrophysiology, Berlin, Germany
| | - Petra Büttner
- Heart Center Leipzig at University of Leipzig, Department of Cardiology, Leipzig, Germany
| | - Karl Scholtyssek
- German Heart Center Berlin, Department of Electrophysiology, Berlin, Germany
| | - Thomas Schröter
- Heart Center Leipzig at University of Leipzig, Department of Cardiac Surgery, Leipzig, Germany
| | - Ruth Hiller
- Insitut für Pathologie, University of Leipzig Medical Center, Leipzig, Germany
| | - Gerhard Hindricks
- German Heart Center Berlin, Department of Electrophysiology, Berlin, Germany
- Leipzig Heart Institute, Leipzig, Germany
| | - Andreas Bollmann
- German Heart Center Berlin, Department of Electrophysiology, Berlin, Germany
- Leipzig Heart Institute, Leipzig, Germany
| | - Ulrich Laufs
- Klinik und Poliklinik für Kardiologie, University of Leipzig Medical Center, Leipzig, Germany
| | - Laura Ueberham
- German Heart Center Berlin, Department of Electrophysiology, Berlin, Germany
- Leipzig Heart Institute, Leipzig, Germany
- Klinik und Poliklinik für Kardiologie, University of Leipzig Medical Center, Leipzig, Germany
| |
Collapse
|
3
|
Dergilev K, Tsokolaeva Z, Goltseva Y, Beloglazova I, Ratner E, Parfyonova Y. Urokinase-Type Plasminogen Activator Receptor Regulates Prosurvival and Angiogenic Properties of Cardiac Mesenchymal Stromal Cells. Int J Mol Sci 2023; 24:15554. [PMID: 37958542 PMCID: PMC10650341 DOI: 10.3390/ijms242115554] [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: 07/24/2023] [Revised: 09/29/2023] [Accepted: 10/21/2023] [Indexed: 11/15/2023] Open
Abstract
One of the largest challenges to the implementation of cardiac cell therapy is identifying selective reparative targets to enhance stem/progenitor cell therapeutic efficacy. In this work, we hypothesized that such a target could be an urokinase-type plasminogen activator receptor (uPAR)-a glycosyl-phosphatidyl-inositol-anchored membrane protein, interacting with urokinase. uPAR is able to form complexes with various transmembrane proteins such as integrins, activating intracellular signaling pathway and thus regulating multiple cell functions. We focused on studying the CD117+ population of cardiac mesenchymal progenitor cells (MPCs), expressing uPAR on their surface. It was found that the number of CD117+ MPCs in the heart of the uPAR-/- mice is lower, as well as their ability to proliferate in vitro compared with cells from wild-type animals. Knockdown of uPAR in CD117+ MPCs of wild-type animals was accompanied by a decrease in survival rate and Akt signaling pathway activity and by an increase in the level of caspase activity in these cells. That suggests the role of uPAR in supporting cell survival. After intramyocardial transplantation of uPAR(-) MPCs, reduced cell retention and angiogenesis stimulation were observed in mice with myocardial infarction model compared to uPAR(+) cells transplantation. Taken together, the present results appear to prove a novel mechanism of uPAR action in maintaining the survival and angiogenic properties of CD117+ MPCs. These results emphasize the importance of the uPAR as a potential pharmacological target for the regulation of reparative properties of myocardial mesenchymal progenitor cells.
Collapse
Affiliation(s)
- Konstantin Dergilev
- Institute of Experimental Cardiology Named after Academician V.N. Smirnov, Federal State Budgetary Institution National Medical Research Center of Cardiology Named after Academician E.I. Chazov, Ministry of Health of the Russian Federation, 121552 Moscow, Russia; (K.D.)
| | - Zoya Tsokolaeva
- Institute of Experimental Cardiology Named after Academician V.N. Smirnov, Federal State Budgetary Institution National Medical Research Center of Cardiology Named after Academician E.I. Chazov, Ministry of Health of the Russian Federation, 121552 Moscow, Russia; (K.D.)
- Federal Research and Clinical Center of Intensive Care Medicine and Rehabilitology, 107031 Moscow, Russia
| | - Yulia Goltseva
- Institute of Experimental Cardiology Named after Academician V.N. Smirnov, Federal State Budgetary Institution National Medical Research Center of Cardiology Named after Academician E.I. Chazov, Ministry of Health of the Russian Federation, 121552 Moscow, Russia; (K.D.)
| | - Irina Beloglazova
- Institute of Experimental Cardiology Named after Academician V.N. Smirnov, Federal State Budgetary Institution National Medical Research Center of Cardiology Named after Academician E.I. Chazov, Ministry of Health of the Russian Federation, 121552 Moscow, Russia; (K.D.)
| | - Elizaveta Ratner
- Institute of Experimental Cardiology Named after Academician V.N. Smirnov, Federal State Budgetary Institution National Medical Research Center of Cardiology Named after Academician E.I. Chazov, Ministry of Health of the Russian Federation, 121552 Moscow, Russia; (K.D.)
| | - Yelena Parfyonova
- Institute of Experimental Cardiology Named after Academician V.N. Smirnov, Federal State Budgetary Institution National Medical Research Center of Cardiology Named after Academician E.I. Chazov, Ministry of Health of the Russian Federation, 121552 Moscow, Russia; (K.D.)
- Department of Biochemistry and Molecular Medicine, Faculty of Medicine, Lomonosov Moscow State University, 119192 Moscow, Russia
| |
Collapse
|
4
|
da Silva FJ, Carvalho de Azevedo J, Ralph ACL, Pinheiro JDJV, Freitas VM, Calcagno DQ. Salivary glands adenoid cystic carcinoma: a molecular profile update and potential implications. Front Oncol 2023; 13:1191218. [PMID: 37476370 PMCID: PMC10354556 DOI: 10.3389/fonc.2023.1191218] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2023] [Accepted: 06/01/2023] [Indexed: 07/22/2023] Open
Abstract
Adenoid cystic carcinoma (ACC) is an aggressive tumor with a high propensity for distant metastasis and perineural invasion. This tumor is more commonly found in regions of the head and neck, mainly the salivary glands. In general, the primary treatment modality for ACC is surgical resection and, in some cases, postoperative radiotherapy. However, no effective systemic treatment is available for patients with advanced disease. Furthermore, this tumor type is characterized by recurrent molecular alterations, especially rearrangements involving the MYB, MYBL1, and NFIB genes. In addition, they also reported copy number alterations (CNAs) that impact genes. One of them is C-KIT, mutations that affect signaling pathways such as NOTCH, PI3KCA, and PTEN, as well as alterations in chromatin remodeling genes. The identification of new molecular targets enables the development of specific therapies. Despite ongoing investigations into immunotherapy, tyrosine kinase inhibitors, and anti-angiogenics, no systemic therapy is approved by the FDA for ACC. In this review, we report the genetic and cytogenetic findings on head and neck ACC, highlighting possible targets for therapeutic interventions.
Collapse
Affiliation(s)
- Fernanda Jardim da Silva
- Núcleo de Pesquisas em Oncologia, Programa de Pós-Graduação em Oncologia e Ciências Médicas, Universidade Federal do Pará, Belém, Brazil
| | - Juscelino Carvalho de Azevedo
- Núcleo de Pesquisas em Oncologia, Programa de Pós-Graduação em Oncologia e Ciências Médicas, Universidade Federal do Pará, Belém, Brazil
- Hospital Universitário João de Barros Barreto, Programa de Residência Multiprofissional em Saúde (Oncologia), Universidade Federal do Pará, Belém, Brazil
| | - Ana Carolina Lima Ralph
- Faculdade de Farmácia, Faculdade Estácio, Carapicuíba, Brazil
- Instituto de Ciências da Saúde, Programa de Pós-Graduação em Odontologia, Universidade Federal do Pará, Belém, Brazil
| | - João de Jesus Viana Pinheiro
- Laboratório de Microambiente Tumoral, Instituto de Ciências Biomédicas, Universidade de São Paulo, São Paulo, Brazil
| | - Vanessa Morais Freitas
- Instituto de Ciências da Saúde, Programa de Pós-Graduação em Odontologia, Universidade Federal do Pará, Belém, Brazil
| | - Danielle Queiroz Calcagno
- Núcleo de Pesquisas em Oncologia, Programa de Pós-Graduação em Oncologia e Ciências Médicas, Universidade Federal do Pará, Belém, Brazil
- Hospital Universitário João de Barros Barreto, Programa de Residência Multiprofissional em Saúde (Oncologia), Universidade Federal do Pará, Belém, Brazil
- Laboratório de Microambiente Tumoral, Instituto de Ciências Biomédicas, Universidade de São Paulo, São Paulo, Brazil
| |
Collapse
|
5
|
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
|
6
|
Luo Y, Zhang H, Yu J, Wei L, Li M, Xu W. Stem cell factor/mast cell/CCL2/monocyte/macrophage axis promotes Coxsackievirus B3 myocarditis and cardiac fibrosis by increasing Ly6C high monocyte influx and fibrogenic mediators production. Immunology 2022; 167:590-605. [PMID: 36054617 DOI: 10.1111/imm.13556] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2021] [Accepted: 02/10/2022] [Indexed: 06/15/2023] Open
Abstract
Mast cells (MCs), central players in allergy and parasitic infections, play key roles in inflammation and fibrosis. Here, the impact of MCs on the progression of Coxsackievirus B3 (CVB3)-induced viral myocarditis (VMC) and fibrosis was investigated using MC-deficient KitW-sh mice. Viral titres, cellular infiltrates and heart pathologies were evaluated and compared with wild-type (WT) mice during acute CVB3 infection of C57BL/6 mice. CVB3 infection induced an increased accumulation and degranulation of MCs in the hearts of mice during acute infection. MC-deficient KitW-sh mice had slightly higher viral titres, decreased VMC and cardiac fibrosis and improved cardiac dysfunction compared to WT mice via decreasing cardiac influx of Ly6Chigh monocytes/macrophages (Mo/Mφ). While bone marrow-derived MC reconstitution decreased viral titre and worsened improved survival and VMC severity in Wsh mice. MC-fibroblasts co-culture revealed a cardiac MC-fibroblasts crosstalk during early infection: fibroblasts trigger MC degranulation and secretion of CCL2 and tumour necrosis factor alpha (TNF-α) via producing early stem cell factor (SCF); while MCs-fibrogenic mediators (TNF-α) stimulate fibroblasts to increase CCL2, α-smooth muscle actin (SMA), collagen and transforming growth factor beta(TGFβ) expression, thus aggravating cardiac fibrosis. MCs and fibroblast-derived CCL2s are both essential for cardiac Ly6Chigh Mo/Mφ influx. Administration of recombinant mouse SCF to CVB3-infected mice aggravates VMC via accelerating MCs accumulation and cardiac influx of Ly6Chi Mo/Mφ. Collectively, our data highlight an early MC-fibroblast crosstalk and SCF/MC/CCL2/Mo/Mφ axis as important mechanisms required for triggering VMC and myocardial fibrosis. This finding indicates critical roles of MCs in initiating and modulating cardiac innate response to CVB3 and has an implication in developing new and more effective treatments for VMC.
Collapse
Affiliation(s)
- Yuan Luo
- Jiangsu Provincial Key Laboratory of Infection and Immunity, Institutes of Biology and Medical Sciences, Soochow University, Suzhou, China
| | - Hongkai Zhang
- Jiangsu Provincial Key Laboratory of Infection and Immunity, Institutes of Biology and Medical Sciences, Soochow University, Suzhou, China
| | - Jie Yu
- Jiangsu Provincial Key Laboratory of Infection and Immunity, Institutes of Biology and Medical Sciences, Soochow University, Suzhou, China
| | - Lin Wei
- Jiangsu Provincial Key Laboratory of Infection and Immunity, Institutes of Biology and Medical Sciences, Soochow University, Suzhou, China
| | - Min Li
- Jiangsu Provincial Key Laboratory of Infection and Immunity, Institutes of Biology and Medical Sciences, Soochow University, Suzhou, China
| | - Wei Xu
- Jiangsu Provincial Key Laboratory of Infection and Immunity, Institutes of Biology and Medical Sciences, Soochow University, Suzhou, China
| |
Collapse
|
7
|
Saheli M, Khoramipour K, Vosough M, Piryaei A, Rahmati M, Suzuki K. Athletes' Mesenchymal Stem Cells Could Be the Best Choice for Cell Therapy in Omicron-Infected Patients. Cells 2022; 11:1926. [PMID: 35741055 PMCID: PMC9221912 DOI: 10.3390/cells11121926] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2022] [Revised: 06/08/2022] [Accepted: 06/12/2022] [Indexed: 02/05/2023] Open
Abstract
New severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) variant, Omicron, contains 32 mutations that have caused a high incidence of breakthrough infections or re-infections. These mutations have reduced vaccine protection against Omicron and other new emerging variants. This highlights the need to find effective treatment, which is suggested to be stem cell-based therapy. Stem cells could support respiratory epithelial cells and they could restore alveolar bioenergetics. In addition, they can increase the secretion of immunomodulatory cytokines. However, after transplantation, cell survival and growth rate are low because of an inappropriate microenvironment, and stem cells face ischemia, inflammation, and oxidative stress in the transplantation niche which reduces the cells' survival and growth. Exercise-training can upregulate antioxidant, anti-inflammatory, and anti-apoptotic defense mechanisms and increase growth signaling, thereby improving transplanted cells' survival and growth. Hence, using athletes' stem cells may increase stem-cell therapy outcomes in Omicron-affected patients.
Collapse
Affiliation(s)
- Mona Saheli
- Department of Anatomical Sciences, and Pathology and Stem Cell Research Centre, Afzalipour Faculty of Medicine, Kerman University of Medical Sciences, Kerman 7616914115, Iran;
| | - Kayvan Khoramipour
- Neuroscience Research Center, Institute of Neuropharmacology, and Department of Physiology and Pharmacology, Afzalipour School of Medicine, Kerman University of Medical Sciences, Kerman 7616914115, Iran
- Student Research Committee, Kerman University of Medical Sciences, Kerman 7619813159, Iran
| | - Massoud Vosough
- Department of Regenerative Medicine, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran 1665659911, Iran;
- Experimental Cancer Medicine, Institution for Laboratory Medicine, Karolinska Institute, 17177 Stockholm, Sweden
| | - Abbas Piryaei
- Department of Biology and Anatomical Sciences, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran 1985717443, Iran;
- Department of Tissue Engineering and Applied Cell Sciences, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran 1434875451, Iran
| | - Masoud Rahmati
- Department of Physical Education and Sport Sciences, Faculty of Literature and Human Sciences, Lorestan University, Khoramabad 6815144316, Iran;
| | - Katsuhiko Suzuki
- Faculty of Sport Sciences, Waseda University, Tokorozawa 359-1192, Saitama, Japan
| |
Collapse
|
8
|
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: 41] [Impact Index Per Article: 13.7] [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
|
9
|
Garbern JC, Lee RT. Heart regeneration: 20 years of progress and renewed optimism. Dev Cell 2022; 57:424-439. [PMID: 35231426 PMCID: PMC8896288 DOI: 10.1016/j.devcel.2022.01.012] [Citation(s) in RCA: 41] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2021] [Revised: 01/10/2022] [Accepted: 01/18/2022] [Indexed: 02/06/2023]
Abstract
Cardiovascular disease is a leading cause of death worldwide, and thus there remains great interest in regenerative approaches to treat heart failure. In the past 20 years, the field of heart regeneration has entered a renaissance period with remarkable progress in the understanding of endogenous heart regeneration, stem cell differentiation for exogenous cell therapy, and cell-delivery methods. In this review, we highlight how this new understanding can lead to viable strategies for human therapy. For the near term, drugs, electrical and mechanical devices, and heart transplantation will remain mainstays of cardiac therapies, but eventually regenerative therapies based on fundamental regenerative biology may offer more permanent solutions for patients with heart failure.
Collapse
Affiliation(s)
- Jessica C. Garbern
- Department of Stem Cell and Regenerative Biology and the Harvard Stem Cell Institute, Harvard University, 7 Divinity Ave, Cambridge, MA 02138, USA,Department of Cardiology, Boston Children’s Hospital, 300 Longwood Ave, Boston, MA 02115, USA
| | - Richard T. Lee
- Department of Stem Cell and Regenerative Biology and the Harvard Stem Cell Institute, Harvard University, 7 Divinity Ave, Cambridge, MA 02138, USA,Division of Cardiovascular Medicine, Department of Medicine, Brigham and Women’s Hospital and Harvard Medical School, 75 Francis St, Boston, MA 02115, USA,Corresponding author and lead contact: Richard T. Lee, Department of Stem Cell and Regenerative Biology, Harvard University, 7 Divinity Ave, Cambridge, MA 02138, Phone: 617-496-5394, Fax: 617-496-8351,
| |
Collapse
|
10
|
Taghizadeh S, Keyhanmanesh R, Rahbarghazi R, Rezaie J, Delkhosh A, Hassanpour M, Heiran H, Ghaffari-Nasab A, Ahmadi M. Systemic administration of c-Kit + cells diminished pulmonary and vascular inflammation in rat model of chronic asthma. BMC Mol Cell Biol 2022; 23:11. [PMID: 35209844 PMCID: PMC8876378 DOI: 10.1186/s12860-022-00410-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2021] [Accepted: 02/09/2022] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND To circumvent some pitfalls related to acute status, chronic model of asthma is conceived to be more suitable approach to guarantee the conditions which are similar to human pulmonary disease. Here, possible therapeutic mechanisms were monitored by which c-kit+ bone marrow cells can attenuate vascular inflammation in rat model of chronic asthma. RESULTS Data revealed c-Kit+ cells could significantly reduce pathological injures in asthmatic rats via modulating the expression of IL-4, INF-γ, ICAM-1 and VCAM-1 in lung tissues and TNF-α, IL-1β and NO levels in BALF (p < 0.001 to p < 0.05). Besides, c-Kit+ cells reduced increased levels of VCAM-1 evaluated by immunohistochemistry staining. In contrast to c-Kit+ cells, c-Kit- cells could not exert beneficial effects in the asthmatic conditions. CONCLUSION Overall, we found that systemic administration of C-kit positive cells can diminish pulmonary and vascular inflammation of chronic asthmatic changes in a rat model. These cells are eligible to suppress inflammation and nitrosative stress in lung tissue coincides with the reduction of pathological changes. These data indicate that C-kit positive cells be used as an alternative cell source for the amelioration of asthmatic changes.
Collapse
Affiliation(s)
- Sajjad Taghizadeh
- Department of Physiology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
- Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Rana Keyhanmanesh
- Drug Applied Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.
| | - Reza Rahbarghazi
- Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
- Department of Applied Cell Sciences, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Jafar Rezaie
- Solid Tumor Research Center, Cellular and Molecular Medicine Institute, Urmia University of Medical Sciences, Urmia, Iran
| | - Aref Delkhosh
- Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mehdi Hassanpour
- Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
- Department of Clinical Biochemistry and Laboratory Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Hossein Heiran
- Department of Physiology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
- Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran
| | | | - Mahdi Ahmadi
- Drug Applied Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.
| |
Collapse
|
11
|
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: 5] [Impact Index Per Article: 1.7] [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
|
12
|
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
|
13
|
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
|
14
|
Mirershadi F, Ahmadi M, Rahbarghazi R, Heiran H, Keyhanmanesh R. C-Kit + cells can modulate asthmatic condition via differentiation into pneumocyte-like cells and alteration of inflammatory responses via ERK/NF-ƙB pathway. IRANIAN JOURNAL OF BASIC MEDICAL SCIENCES 2022; 25:96-102. [PMID: 35656445 PMCID: PMC9118279 DOI: 10.22038/ijbms.2021.59946.13293] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/26/2021] [Accepted: 11/21/2021] [Indexed: 11/06/2022]
Abstract
Objectives The exact role of the progenitor cell types in the dynamic healing of asthmatic lungs is lacking. This investigation was proposed to evaluate the effect of intratracheally administered rat bone marrow-derived c-kit+ cells on ovalbumin-induced sensitized male rats. Materials and Methods Forty rats were randomly divided into 4 groups; healthy rats received phosphate-buffered saline (PBS) (C); sensitized rats received PBS (S); PBS containing C-kit- cells (S+C-kit-); and PBS containing C-kit+ cells (S+C-kit+). After two weeks, circulatory CD4+/CD8+ T-cell counts and pulmonary ERK/NF-ƙB signaling pathway as well as the probability of cellular differentiation were assessed. Results The results showed that transplanted C-Kit+ cells were engrafted into pulmonary tissue and differentiated into epithelial cells. C-Kit+ cells could increase the number of CD4+ cells in comparison with the S group (P<0.001); however, they diminished the level of CD8+ cells (P<0.01). Moreover, data demonstrated increased p-ERK/ERK ratio (P<0.001) and NF-ƙB level (P<0.05) in sensitized rats compared with the C group. The administration of C-kit+, but not C-Kit-, decreased p-ERK/ERK ratio and NF-ƙB level compared with those of the S group (P<0.05). Conclusion The study revealed that C-Kit+ cells engrafted into pulmonary tissue reduced the NF-ƙB protein level and diminished p-ERK/ERK ratio, leading to suppression of inflammatory response in asthmatic lungs.
Collapse
Affiliation(s)
- Fatemeh Mirershadi
- Department of Physiology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran, Department of Physiology, Ardabil Branch, Islamic Azad University, Ardabil, Iran, Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mahdi Ahmadi
- Department of Physiology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran, Tuberculosis and Lung Disease Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Reza Rahbarghazi
- Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran, Department of Applied Cell Sciences, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran,Corresponding authors: Rana Keyhanmanesh. Department of Physiology, Tabriz University of Medical Sciences, Tabriz, Iran. Tel/Fax: +98-4133364664; ; and Reza Rahbarghazi. Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.
| | - Hossein Heiran
- Department of Physiology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Rana Keyhanmanesh
- Department of Physiology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran, Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran, Drug Applied Research Center, Tabriz University of Medical Sciences, Tabriz, Iran,Corresponding authors: Rana Keyhanmanesh. Department of Physiology, Tabriz University of Medical Sciences, Tabriz, Iran. Tel/Fax: +98-4133364664; ; and Reza Rahbarghazi. Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.
| |
Collapse
|
15
|
Rodríguez JMM, Fonfara S, Hetzel U, Kipar A. Feline hypertrophic cardiomyopathy: reduced microvascular density and involvement of CD34+ interstitial cells. Vet Pathol 2021; 59:269-283. [PMID: 34955067 PMCID: PMC8928422 DOI: 10.1177/03009858211062631] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The sequence of pathological events in feline hypertrophic cardiomyopathy (fHCM) is still largely unknown, although we know that fHCM is characterized by interstitial remodeling in a macrophage-driven pro-inflammatory environment and that myocardial ischemia might contribute to its progression. This study aimed to gain further insights into the structural changes associated with interstitial remodeling in fHCM with special focus on the myocardial microvasculature and the phenotype of the interstitial cells. Twenty-eight hearts (16 hearts with fHCM and 12 without cardiac disease) were evaluated in the current study, with immunohistochemistry, RNA-in situ hybridization, and transmission electron microscopy. Morphometrical evaluations revealed a statistically significant lower microvascular density in fHCM. This was associated with structural alterations in capillaries that go along with a widening of the interstitium due to the accumulation of edema fluid, collagen fibers, and mononuclear cells that also proliferated locally. The interstitial cells were mainly of fibroblastic or vascular phenotype, with a substantial contribution of predominantly resident macrophages. A large proportion expressed CD34 mRNA, which suggests a progenitor cell potential. Our results indicate that microvascular alterations are key events in the pathogenesis of fHCM and that myocardial interstitial cell populations with CD34+ phenotype play a role in the pathogenesis of the disease.
Collapse
Affiliation(s)
- Josep M Monné Rodríguez
- The Veterinary Cardiac Pathophysiology Consortium.,University of Zurich, Zurich, Switzerland.,University of Bern, Bern, Switzerland
| | - Sonja Fonfara
- The Veterinary Cardiac Pathophysiology Consortium.,University of Guelph, Guelph, Ontario, Canada
| | - Udo Hetzel
- The Veterinary Cardiac Pathophysiology Consortium.,University of Zurich, Zurich, Switzerland
| | - Anja Kipar
- The Veterinary Cardiac Pathophysiology Consortium.,University of Zurich, Zurich, Switzerland
| |
Collapse
|
16
|
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: 30] [Impact Index Per Article: 7.5] [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
|
17
|
Pathania S, Pentikäinen OT, Singh PK. A holistic view on c-Kit in cancer: Structure, signaling, pathophysiology and its inhibitors. Biochim Biophys Acta Rev Cancer 2021; 1876:188631. [PMID: 34606974 DOI: 10.1016/j.bbcan.2021.188631] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2021] [Revised: 09/08/2021] [Accepted: 09/28/2021] [Indexed: 11/18/2022]
Abstract
Receptor tyrosine kinases play an important role in many cellular processes, and their dysregulation leads to diseases, most importantly cancer. One such receptor tyrosine kinase is c-Kit, a type-III receptor tyrosine kinase, which is involved in various intracellular signaling pathways. The role of different mutant isoforms of c-Kit has been established in several types of cancers. Accordingly, promising c-Kit inhibition results have been reported for the treatment of different cancers (e.g., gastrointestinal stromal tumors, melanoma, acute myeloid leukemia, and other tumors). Therefore, lots of effort has been put to target c-Kit for the treatment of cancer. Here, we provide a comprehensive compilation to provide an insight into c-Kit inhibitor discovery. This compilation provides key information regarding the structure, signaling pathways related to c-Kit, and, more importantly, pharmacophores, binding modes, and SAR analysis for almost all small-molecule heterocycles reported for their c-Kit inhibitory activity. This work could be used as a guide in understanding the basic requirements for targeting c-Kit, and how the selectivity and efficacy of the molecules have been achieved till today.
Collapse
Affiliation(s)
- Shelly Pathania
- Department of Pharmaceutical Chemistry, ISF College of Pharmacy, Ghal Kalan, Ferozpur G.T. Road, Moga 142001, Punjab, India
| | - Olli T Pentikäinen
- Integrative Physiology and Pharmacology, Institute of Biomedicine, Faculty of Medicine, University of Turku, FI-20520 Turku, Finland
| | - Pankaj Kumar Singh
- Integrative Physiology and Pharmacology, Institute of Biomedicine, Faculty of Medicine, University of Turku, FI-20520 Turku, Finland.
| |
Collapse
|
18
|
In vitro CSC-derived cardiomyocytes exhibit the typical microRNA-mRNA blueprint of endogenous cardiomyocytes. Commun Biol 2021; 4:1146. [PMID: 34593953 PMCID: PMC8484596 DOI: 10.1038/s42003-021-02677-y] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2021] [Accepted: 09/15/2021] [Indexed: 02/08/2023] Open
Abstract
miRNAs modulate cardiomyocyte specification by targeting mRNAs of cell cycle regulators and acting in cardiac muscle lineage gene regulatory loops. It is unknown if or to-what-extent these miRNA/mRNA networks are operative during cardiomyocyte differentiation of adult cardiac stem/progenitor cells (CSCs). Clonally-derived mouse CSCs differentiated into contracting cardiomyocytes in vitro (iCMs). Comparison of "CSCs vs. iCMs" mRNome and microRNome showed a balanced up-regulation of CM-related mRNAs together with a down-regulation of cell cycle and DNA replication mRNAs. The down-regulation of cell cycle genes and the up-regulation of the mature myofilament genes in iCMs reached intermediate levels between those of fetal and neonatal cardiomyocytes. Cardiomyo-miRs were up-regulated in iCMs. The specific networks of miRNA/mRNAs operative in iCMs closely resembled those of adult CMs (aCMs). miR-1 and miR-499 enhanced myogenic commitment toward terminal differentiation of iCMs. In conclusions, CSC specification/differentiation into contracting iCMs follows known cardiomyo-MiR-dependent developmental cardiomyocyte differentiation trajectories and iCMs transcriptome/miRNome resembles that of CMs.
Collapse
|
19
|
Kalidasan V, Ng WH, Ishola OA, Ravichantar N, Tan JJ, Das KT. A guide in lentiviral vector production for hard-to-transfect cells, using cardiac-derived c-kit expressing cells as a model system. Sci Rep 2021; 11:19265. [PMID: 34584147 PMCID: PMC8478948 DOI: 10.1038/s41598-021-98657-7] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2021] [Accepted: 09/09/2021] [Indexed: 02/08/2023] Open
Abstract
Gene therapy revolves around modifying genetic makeup by inserting foreign nucleic acids into targeted cells via gene delivery methods to treat a particular disease. While the genes targeted play a key role in gene therapy, the gene delivery system used is also of utmost importance as it determines the success of gene therapy. As primary cells and stem cells are often the target cells for gene therapy in clinical trials, the delivery system would need to be robust, and viral-based entries such as lentiviral vectors work best at transporting the transgene into the cells. However, even within lentiviral vectors, several parameters can affect the functionality of the delivery system. Using cardiac-derived c-kit expressing cells (CCs) as a model system, this study aims to optimize lentiviral production by investigating various experimental factors such as the generation of the lentiviral system, concentration method, and type of selection marker. Our findings showed that the 2nd generation system with pCMV-dR8.2 dvpr as the packaging plasmid produced a 7.3-fold higher yield of lentiviral production compared to psPAX2. Concentrating the virus with ultracentrifuge produced a higher viral titer at greater than 5 × 105 infectious unit values/ml (IFU/ml). And lastly, the minimum inhibitory concentration (MIC) of puromycin selection marker was 10 μg/mL and 7 μg/mL for HEK293T and CCs, demonstrating the suitability of antibiotic selection for all cell types. This encouraging data can be extrapolated and applied to other difficult-to-transfect cells, such as different types of stem cells or primary cells.
Collapse
Affiliation(s)
- V. Kalidasan
- grid.11875.3a0000 0001 2294 3534Infectomics Cluster, Advanced Medical and Dental Institute, Universiti Sains Malaysia, Kepala Batas, Malaysia
| | - Wai Hoe Ng
- grid.11875.3a0000 0001 2294 3534Regenerative Medicine Cluster, Advanced Medical and Dental Institute, Universiti Sains Malaysia, Kepala Batas, Malaysia
| | - Oluwaseun Ayodeji Ishola
- grid.11875.3a0000 0001 2294 3534Infectomics Cluster, Advanced Medical and Dental Institute, Universiti Sains Malaysia, Kepala Batas, Malaysia ,Helmholtz Research Zentrum, Munich, Germany
| | - Nithya Ravichantar
- grid.11875.3a0000 0001 2294 3534Infectomics Cluster, Advanced Medical and Dental Institute, Universiti Sains Malaysia, Kepala Batas, Malaysia
| | - Jun Jie Tan
- grid.11875.3a0000 0001 2294 3534Regenerative Medicine Cluster, Advanced Medical and Dental Institute, Universiti Sains Malaysia, Kepala Batas, Malaysia
| | - Kumitaa Theva Das
- grid.11875.3a0000 0001 2294 3534Infectomics Cluster, Advanced Medical and Dental Institute, Universiti Sains Malaysia, Kepala Batas, Malaysia
| |
Collapse
|
20
|
Mannino G, Russo C, Maugeri G, Musumeci G, Vicario N, Tibullo D, Giuffrida R, Parenti R, Lo Furno D. Adult stem cell niches for tissue homeostasis. J Cell Physiol 2021; 237:239-257. [PMID: 34435361 PMCID: PMC9291197 DOI: 10.1002/jcp.30562] [Citation(s) in RCA: 55] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2021] [Revised: 07/26/2021] [Accepted: 08/09/2021] [Indexed: 12/13/2022]
Abstract
Adult stem cells are fundamental to maintain tissue homeostasis, growth, and regeneration. They reside in specialized environments called niches. Following activating signals, they proliferate and differentiate into functional cells that are able to preserve tissue physiology, either to guarantee normal turnover or to counteract tissue damage caused by injury or disease. Multiple interactions occur within the niche between stem cell‐intrinsic factors, supporting cells, the extracellular matrix, and signaling pathways. Altogether, these interactions govern cell fate, preserving the stem cell pool, and regulating stem cell proliferation and differentiation. Based on their response to body needs, tissues can be largely classified into three main categories: tissues that even in normal conditions are characterized by an impressive turnover to replace rapidly exhausting cells (blood, epidermis, or intestinal epithelium); tissues that normally require only a basal cell replacement, though able to efficiently respond to increased tissue needs, injury, or disease (skeletal muscle); tissues that are equipped with less powerful stem cell niches, whose repairing ability is not able to overcome severe damage (heart or nervous tissue). The purpose of this review is to describe the main characteristics of stem cell niches in these different tissues, highlighting the various components influencing stem cell activity. Although much has been done, more work is needed to further increase our knowledge of niche interactions. This would be important not only to shed light on this fundamental chapter of human physiology but also to help the development of cell‐based strategies for clinical therapeutic applications, especially when other approaches fail.
Collapse
Affiliation(s)
- Giuliana Mannino
- Department of Biomedical and Biotechnological Sciences, University of Catania, Catania, Italy
| | - Cristina Russo
- Department of Biomedical and Biotechnological Sciences, University of Catania, Catania, Italy
| | - Grazia Maugeri
- Department of Biomedical and Biotechnological Sciences, University of Catania, Catania, Italy
| | - Giuseppe Musumeci
- Department of Biomedical and Biotechnological Sciences, University of Catania, Catania, Italy
| | - Nunzio Vicario
- Department of Biomedical and Biotechnological Sciences, University of Catania, Catania, Italy
| | - Daniele Tibullo
- Department of Biomedical and Biotechnological Sciences, University of Catania, Catania, Italy
| | - Rosario Giuffrida
- Department of Biomedical and Biotechnological Sciences, University of Catania, Catania, Italy
| | - Rosalba Parenti
- Department of Biomedical and Biotechnological Sciences, University of Catania, Catania, Italy
| | - Debora Lo Furno
- Department of Biomedical and Biotechnological Sciences, University of Catania, Catania, Italy
| |
Collapse
|
21
|
Ivankovic I, Déan-Ben XL, Haas H, Kimm MA, Wildgruber M, Razansky D. Volumetric Optoacoustic Tomography Differentiates Myocardial Remodelling. Mol Imaging Biol 2021; 22:1235-1243. [PMID: 32394284 DOI: 10.1007/s11307-020-01498-5] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
PURPOSE Myocardial healing following myocardial infarction (MI) is a complex process that is yet to be fully understood. Clinical attempts in regeneration of the injured myocardium using cardiac stem cells faced major challenges, calling for a better understanding of the processes involved at a more basic level in order to foster translation. PROCEDURES We examined the feasibility of volumetric optoacoustic tomography (VOT) in studying healing of the myocardium in different models of MI, including permanent occlusion (PO) of the left coronary artery, temporary occlusion (ischemia-reperfusion-I/R) and infarcted c-kit mutants, a genetic mouse model with impaired cardiac healing. Murine hearts were imaged at 100 Hz frame rate using 800 nm excitation wavelength, corresponding to the peak absorption of indocyanine green (ICG) in plasma and the isosbestic point of haemoglobin. RESULTS The non-invasive real-time volumetric imaging capabilities of VOT have allowed the detection of significant variations in the pulmonary transit time (PTT), a parameter affected by MI, across different murine models. Upon intravenous injection of ICG, we were able to track alterations in cardiac perfusion in I/R models, which were absent in wild-type (wt) PO or kitW/kitW-v PO mice. The wt-PO and I/R models further exhibited irregularities in their cardiac cycles. CONCLUSIONS Clear differences in the PTT, ICG perfusion and cardiac cycle patterns were identified between the different models and days post MI. Overall, the results highlight the unique capacity of VOT for multi-parametric characterization of morphological and functional changes in murine models of MI.
Collapse
Affiliation(s)
- Ivana Ivankovic
- Faculty of Medicine and Institute of Pharmacology and Toxicology, University of Zurich, Zurich, Switzerland
- Institute for Biomedical Engineering and Department of Information Technology and Electrical Engineering, ETH Zurich, Zurich, Switzerland
| | - Xosé Luís Déan-Ben
- Faculty of Medicine and Institute of Pharmacology and Toxicology, University of Zurich, Zurich, Switzerland
- Institute for Biomedical Engineering and Department of Information Technology and Electrical Engineering, ETH Zurich, Zurich, Switzerland
| | - Helena Haas
- Department of Diagnostic and Interventional Radiology, Klinikum Rechts der Isar Technical University of Munich, Munich, Germany
| | - Melanie A Kimm
- Department of Diagnostic and Interventional Radiology, Klinikum Rechts der Isar Technical University of Munich, Munich, Germany
| | - Moritz Wildgruber
- Department of Diagnostic and Interventional Radiology, Klinikum Rechts der Isar Technical University of Munich, Munich, Germany
- Translational Research Imaging Center, Department of Clinical Radiology, Universitätsklinikum Münster, Munster, Germany
| | - Daniel Razansky
- Faculty of Medicine and Institute of Pharmacology and Toxicology, University of Zurich, Zurich, Switzerland.
- Institute for Biomedical Engineering and Department of Information Technology and Electrical Engineering, ETH Zurich, Zurich, Switzerland.
| |
Collapse
|
22
|
Marino F, Scalise M, Cianflone E, Salerno L, Cappetta D, Salerno N, De Angelis A, Torella D, Urbanek K. Physical Exercise and Cardiac Repair: The Potential Role of Nitric Oxide in Boosting Stem Cell Regenerative Biology. Antioxidants (Basel) 2021; 10:1002. [PMID: 34201562 PMCID: PMC8300666 DOI: 10.3390/antiox10071002] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2021] [Revised: 06/14/2021] [Accepted: 06/19/2021] [Indexed: 12/11/2022] Open
Abstract
Over the years strong evidence has been accumulated showing that aerobic physical exercise exerts beneficial effects on the prevention and reduction of cardiovascular risk. Exercise in healthy subjects fosters physiological remodeling of the adult heart. Concurrently, physical training can significantly slow-down or even reverse the maladaptive pathologic cardiac remodeling in cardiac diseases, improving heart function. The underlying cellular and molecular mechanisms of the beneficial effects of physical exercise on the heart are still a subject of intensive study. Aerobic activity increases cardiovascular nitric oxide (NO) released mainly through nitric oxidase synthase 3 activity, promoting endothelium-dependent vasodilation, reducing vascular resistance, and lowering blood pressure. On the reverse, an imbalance between increasing free radical production and decreased NO generation characterizes pathologic remodeling, which has been termed the "nitroso-redox imbalance". Besides these classical evidence on the role of NO in cardiac physiology and pathology, accumulating data show that NO regulate different aspects of stem cell biology, including survival, proliferation, migration, differentiation, and secretion of pro-regenerative factors. Concurrently, it has been shown that physical exercise generates physiological remodeling while antagonizes pathologic remodeling also by fostering cardiac regeneration, including new cardiomyocyte formation. This review is therefore focused on the possible link between physical exercise, NO, and stem cell biology in the cardiac regenerative/reparative response to physiological or pathological load. Cellular and molecular mechanisms that generate an exercise-induced cardioprotective phenotype are discussed in regards with myocardial repair and regeneration. Aerobic training can benefit cells implicated in cardiovascular homeostasis and response to damage by NO-mediated pathways that protect stem cells in the hostile environment, enhance their activation and differentiation and, in turn, translate to more efficient myocardial tissue regeneration. Moreover, stem cell preconditioning by and/or local potentiation of NO signaling can be envisioned as promising approaches to improve the post-transplantation stem cell survival and the efficacy of cardiac stem cell therapy.
Collapse
Affiliation(s)
- Fabiola Marino
- Department of Experimental and Clinical Medicine, Magna Graecia University, 88100 Catanzaro, Italy; (F.M.); (M.S.); (L.S.)
| | - Mariangela Scalise
- Department of Experimental and Clinical Medicine, Magna Graecia University, 88100 Catanzaro, Italy; (F.M.); (M.S.); (L.S.)
| | - Eleonora Cianflone
- Department of Medical and Surgical Sciences, Magna Graecia University, 88100 Catanzaro, Italy; (E.C.); (N.S.)
| | - Luca Salerno
- Department of Experimental and Clinical Medicine, Magna Graecia University, 88100 Catanzaro, Italy; (F.M.); (M.S.); (L.S.)
| | - Donato Cappetta
- Department of Experimental Medicine, University of Campania “L. Vanvitelli”, 80138 Naples, Italy; (D.C.); (A.D.A.)
| | - Nadia Salerno
- Department of Medical and Surgical Sciences, Magna Graecia University, 88100 Catanzaro, Italy; (E.C.); (N.S.)
| | - Antonella De Angelis
- Department of Experimental Medicine, University of Campania “L. Vanvitelli”, 80138 Naples, Italy; (D.C.); (A.D.A.)
| | - Daniele Torella
- Department of Experimental and Clinical Medicine, Magna Graecia University, 88100 Catanzaro, Italy; (F.M.); (M.S.); (L.S.)
| | - Konrad Urbanek
- Department of Experimental and Clinical Medicine, Magna Graecia University, 88100 Catanzaro, Italy; (F.M.); (M.S.); (L.S.)
| |
Collapse
|
23
|
Xu AA, Shapero KS, Geibig JA, Ma HWK, Jones AR, Hanna M, Pitts DR, Hillas E, Firpo MA, Peattie RA. Histologic evaluation of therapeutic responses in ischemic myocardium elicited by dual growth factor delivery from composite glycosaminoglycan hydrogels. Acta Histochem 2021; 123:151699. [PMID: 33662819 DOI: 10.1016/j.acthis.2021.151699] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2020] [Revised: 02/10/2021] [Accepted: 02/23/2021] [Indexed: 01/01/2023]
Abstract
In this project, the ability of dual growth factor-preloaded, silk-reinforced, composite hyaluronic acid-based hydrogels to elicit advantageous histologic responses when secured to ischemic myocardium was evaluated in vivo. Reinforced hydrogels containing both Vascular Endothelial Growth Factor (VEGF) and Platelet-derived Growth Factor (PDGF) were prepared by crosslinking chemically modified hyaluronic acid and heparin with poly(ethylene glycol)-diacrylate around a reinforcing silk mesh. Composite patches were sutured to the ventricular surface of ischemic myocardium in Sprague-Dawley rats, and the resulting angiogenic response was followed for 28 days. The gross appearance of treated hearts showed significantly reduced ischemic area and fibrous deposition compared to untreated control hearts. Histologic evaluation showed growth factor delivery to restore myofiber orientation to pre-surgical levels and to significantly increase elicited microvessel density and maturity by day 28 in infarcted myocardial tissue (p < 0.05). In addition, growth factor delivery reduced cell apoptosis and decreased the density of elicited mast cells and both CD68+ and anti-inflammatory CD163+ macrophages. These findings suggest that HA-based, dual growth factor-loaded hydrogels can successfully induce a series of beneficial responses in ischemic myocardium, and offer the potential for therapeutic improvement of ischemic myocardial remodeling.
Collapse
Affiliation(s)
- Alexander A Xu
- Department of Surgery, Tufts Medical Center, 800 Washington Street, Boston, MA, 02111, USA
| | - Kayle S Shapero
- Department of Surgery, Tufts Medical Center, 800 Washington Street, Boston, MA, 02111, USA
| | - Jared A Geibig
- Department of Surgery, Tufts Medical Center, 800 Washington Street, Boston, MA, 02111, USA
| | - Hsiang-Wei K Ma
- Department of Surgery, Tufts Medical Center, 800 Washington Street, Boston, MA, 02111, USA
| | - Alex R Jones
- Department of Surgery, Tufts Medical Center, 800 Washington Street, Boston, MA, 02111, USA
| | - Marina Hanna
- Department of Surgery, Tufts Medical Center, 800 Washington Street, Boston, MA, 02111, USA
| | - Daniel R Pitts
- Department of Surgery, Tufts Medical Center, 800 Washington Street, Boston, MA, 02111, USA
| | - Elaine Hillas
- Department of Surgery, School of Medicine, The University of Utah, 30 N., 1930 E., Salt Lake City, UT, 84132, USA
| | - Matthew A Firpo
- Department of Surgery, School of Medicine, The University of Utah, 30 N., 1930 E., Salt Lake City, UT, 84132, USA
| | - Robert A Peattie
- Department of Surgery, Tufts Medical Center, 800 Washington Street, Boston, MA, 02111, USA.
| |
Collapse
|
24
|
Zeng WR, Doran PM. Interactivity of biochemical and physical stimuli during epigenetic conditioning and cardiomyocytic differentiation of stem and progenitor cells derived from adult hearts. Integr Biol (Camb) 2021; 13:73-85. [PMID: 33704437 DOI: 10.1093/intbio/zyab003] [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: 08/25/2020] [Revised: 12/10/2020] [Accepted: 01/19/2021] [Indexed: 11/13/2022]
Abstract
Mixed populations of cardiosphere-derived stem and progenitor cells containing proliferative and cardiomyogenically committed cells were obtained from adult rat hearts. The cells were cultured in either static 2D monolayers or dynamic 3D scaffold systems with fluid flow. Cardiomyocyte lineage commitment in terms of GATA4 and Nkx2.5 expression was significantly enhanced in the dynamic 3D cultures compared with static 2D conditions. Treatment of the cells with 5-azacytidine (5-aza) produced different responses in the two culture systems, as activity of this chemical epigenetic conditioning agent depended on the cell attachment and hydrodynamic conditions provided during culture. Cell growth was unaffected by 5-aza in the static 2D cultures but was significantly reduced under dynamic 3D conditions relative to untreated controls. Myogenic differentiation measured as Mef2c expression was markedly upregulated by 5-aza in the dynamic 3D cultures but downregulated in the static 2D cultures. The ability of the physical environment to modulate the cellular cardiomyogenic response to 5-aza underscores the interactivity of biochemical and physical stimuli applied for cell differentiation. Accordingly, observations about the efficacy of 5-aza as a cardiomyocyte induction agent may not be applicable across different culture systems. Overall, use of dynamic 3D rather than static 2D culture was more beneficial for cardio-specific myogenesis than 5-aza treatment, which generated a more ambiguous differentiation response.
Collapse
Affiliation(s)
- Wendy R Zeng
- Faculty of Science, Engineering and Technology, Swinburne University of Technology, Melbourne, VIC, Australia
| | - Pauline M Doran
- Faculty of Science, Engineering and Technology, Swinburne University of Technology, Melbourne, VIC, Australia
| |
Collapse
|
25
|
Bhartiya D, Flora Y, Sharma D, Mohammad SA. Two Stem Cell Populations Including VSELs and CSCs Detected in the Pericardium of Adult Mouse Heart. Stem Cell Rev Rep 2021; 17:685-693. [PMID: 33492626 DOI: 10.1007/s12015-021-10119-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/05/2021] [Indexed: 10/22/2022]
Abstract
Adult mammalian heart is considered to be one of the least regenerative organs as it is not able to initiate endogenous regeneration in response to injury unlike in lower vertebrates and neonatal mammals. Evidence is now accumulating to suggest normal renewal and replacement of cardiomyocytes occurs even in middle-aged and old individuals. But underlying mechanisms leading to this are not yet clear. Do tissue-resident stem cells exist or somatic cells dedifferentiate leading to regeneration? Lot of attention is currently being focused on epicardium as it is involved in cardiac development, lodges multipotent progenitors and is a source of growth factors. Present study was undertaken to study the presence of stem cells in the pericardium. Intact adult mouse heart was subjected to partial enzymatic digestion to collect the pericardial cells dislodged from the surface. Pericardial cells suspension was processed to enrich the stem cells using our recently published protocol. Two populations of stem cells were successfully enriched from the pericardium of adult mouse heart along with distinct 'cardiospheres' with cytoplasmic continuity (formed by rapid proliferation and incomplete cytokinesis). These included very small embryonic-like stem cells (VSELs) and slightly bigger 'progenitors' cardiac stem cells (CSCs). Expression of pluripotent (Oct-4A, Sox-2, Nanog), primordial germ cells (Stella, Fragilis) and CSCs (Oct-4, Sca-1) specific transcripts was studied by RT-PCR. Stem cells expressed OCT-4, NANOG, SSEA-1, SCA-1 and c-KIT. c-KIT was expressed by cells of different sizes but only smaller CD45-c-KIT+ VSELs possess regenerative potential. Inadvertent loss of stem cells while processing for different experiments has led to misperceptions & controversies existing in the field of cardiac stem cells and requires urgent rectification. VSELs/CSCs have the potential to regenerate damaged cardiac tissue in the presence of paracrine support provided by the mesenchymal stromal cells.
Collapse
Affiliation(s)
- Deepa Bhartiya
- Stem Cell Biology Department, ICMR-National Institute for Research in Reproductive Health, Jehangir Merwanji Street, Parel, Mumbai, 400 012, India.
| | - Yash Flora
- Stem Cell Biology Department, ICMR-National Institute for Research in Reproductive Health, Jehangir Merwanji Street, Parel, Mumbai, 400 012, India
| | - Diksha Sharma
- Stem Cell Biology Department, ICMR-National Institute for Research in Reproductive Health, Jehangir Merwanji Street, Parel, Mumbai, 400 012, India
| | - Subhan Ali Mohammad
- Stem Cell Biology Department, ICMR-National Institute for Research in Reproductive Health, Jehangir Merwanji Street, Parel, Mumbai, 400 012, India
| |
Collapse
|
26
|
Carresi C, Scicchitano M, Scarano F, Macrì R, Bosco F, Nucera S, Ruga S, Zito MC, Mollace R, Guarnieri L, Coppoletta AR, Gliozzi M, Musolino V, Maiuolo J, Palma E, Mollace V. The Potential Properties of Natural Compounds in Cardiac Stem Cell Activation: Their Role in Myocardial Regeneration. Nutrients 2021; 13:275. [PMID: 33477916 PMCID: PMC7833367 DOI: 10.3390/nu13010275] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2020] [Revised: 01/07/2021] [Accepted: 01/12/2021] [Indexed: 12/12/2022] Open
Abstract
Cardiovascular diseases (CVDs), which include congenital heart disease, rhythm disorders, subclinical atherosclerosis, coronary heart disease, and many other cardiac disorders, cause about 30% of deaths globally; representing one of the main health problems worldwide. Among CVDs, ischemic heart diseases (IHDs) are one of the major causes of morbidity and mortality in the world. The onset of IHDs is essentially due to an unbalance between the metabolic demands of the myocardium and its supply of oxygen and nutrients, coupled with a low regenerative capacity of the heart, which leads to great cardiomyocyte (CM) loss; promoting heart failure (HF) and myocardial infarction (MI). To date, the first strategy recommended to avoid IHDs is prevention in order to reduce the underlying risk factors. In the management of IHDs, traditional therapeutic options are widely used to improve symptoms, attenuate adverse cardiac remodeling, and reduce early mortality rate. However, there are no available treatments that aim to improve cardiac performance by replacing the irreversible damaged cardiomyocytes (CMs). Currently, heart transplantation is the only treatment being carried out for irreversibly damaged CMs. Hence, the discovery of new therapeutic options seems to be necessary. Interestingly, recent experimental evidence suggests that regenerative stem cell medicine could be a useful therapeutic approach to counteract cardiac damage and promote tissue regeneration. To this end, researchers are tasked with answering one main question: how can myocardial regeneration be stimulated? In this regard, natural compounds from plant extracts seem to play a particularly promising role. The present review will summarize the recent advances in our knowledge of stem cell therapy in the management of CVDs; focusing on the main properties and potential mechanisms of natural compounds in stimulating and activating stem cells for myocardial regeneration.
Collapse
Affiliation(s)
- Cristina Carresi
- Institute of Research for Food Safety & Health IRC-FSH, University Magna Graecia, 88100 Catanzaro, Italy; (F.S.); (R.M.); (F.B.); (S.N.); (S.R.); (M.C.Z.); (R.M.); (L.G.); (A.R.C.); (M.G.); (V.M.); (J.M.); (E.P.); (V.M.)
| | - Miriam Scicchitano
- Institute of Research for Food Safety & Health IRC-FSH, University Magna Graecia, 88100 Catanzaro, Italy; (F.S.); (R.M.); (F.B.); (S.N.); (S.R.); (M.C.Z.); (R.M.); (L.G.); (A.R.C.); (M.G.); (V.M.); (J.M.); (E.P.); (V.M.)
| | - Federica Scarano
- Institute of Research for Food Safety & Health IRC-FSH, University Magna Graecia, 88100 Catanzaro, Italy; (F.S.); (R.M.); (F.B.); (S.N.); (S.R.); (M.C.Z.); (R.M.); (L.G.); (A.R.C.); (M.G.); (V.M.); (J.M.); (E.P.); (V.M.)
| | - Roberta Macrì
- Institute of Research for Food Safety & Health IRC-FSH, University Magna Graecia, 88100 Catanzaro, Italy; (F.S.); (R.M.); (F.B.); (S.N.); (S.R.); (M.C.Z.); (R.M.); (L.G.); (A.R.C.); (M.G.); (V.M.); (J.M.); (E.P.); (V.M.)
| | - Francesca Bosco
- Institute of Research for Food Safety & Health IRC-FSH, University Magna Graecia, 88100 Catanzaro, Italy; (F.S.); (R.M.); (F.B.); (S.N.); (S.R.); (M.C.Z.); (R.M.); (L.G.); (A.R.C.); (M.G.); (V.M.); (J.M.); (E.P.); (V.M.)
| | - Saverio Nucera
- Institute of Research for Food Safety & Health IRC-FSH, University Magna Graecia, 88100 Catanzaro, Italy; (F.S.); (R.M.); (F.B.); (S.N.); (S.R.); (M.C.Z.); (R.M.); (L.G.); (A.R.C.); (M.G.); (V.M.); (J.M.); (E.P.); (V.M.)
| | - Stefano Ruga
- Institute of Research for Food Safety & Health IRC-FSH, University Magna Graecia, 88100 Catanzaro, Italy; (F.S.); (R.M.); (F.B.); (S.N.); (S.R.); (M.C.Z.); (R.M.); (L.G.); (A.R.C.); (M.G.); (V.M.); (J.M.); (E.P.); (V.M.)
| | - Maria Caterina Zito
- Institute of Research for Food Safety & Health IRC-FSH, University Magna Graecia, 88100 Catanzaro, Italy; (F.S.); (R.M.); (F.B.); (S.N.); (S.R.); (M.C.Z.); (R.M.); (L.G.); (A.R.C.); (M.G.); (V.M.); (J.M.); (E.P.); (V.M.)
| | - Rocco Mollace
- Institute of Research for Food Safety & Health IRC-FSH, University Magna Graecia, 88100 Catanzaro, Italy; (F.S.); (R.M.); (F.B.); (S.N.); (S.R.); (M.C.Z.); (R.M.); (L.G.); (A.R.C.); (M.G.); (V.M.); (J.M.); (E.P.); (V.M.)
| | - Lorenza Guarnieri
- Institute of Research for Food Safety & Health IRC-FSH, University Magna Graecia, 88100 Catanzaro, Italy; (F.S.); (R.M.); (F.B.); (S.N.); (S.R.); (M.C.Z.); (R.M.); (L.G.); (A.R.C.); (M.G.); (V.M.); (J.M.); (E.P.); (V.M.)
| | - Anna Rita Coppoletta
- Institute of Research for Food Safety & Health IRC-FSH, University Magna Graecia, 88100 Catanzaro, Italy; (F.S.); (R.M.); (F.B.); (S.N.); (S.R.); (M.C.Z.); (R.M.); (L.G.); (A.R.C.); (M.G.); (V.M.); (J.M.); (E.P.); (V.M.)
| | - Micaela Gliozzi
- Institute of Research for Food Safety & Health IRC-FSH, University Magna Graecia, 88100 Catanzaro, Italy; (F.S.); (R.M.); (F.B.); (S.N.); (S.R.); (M.C.Z.); (R.M.); (L.G.); (A.R.C.); (M.G.); (V.M.); (J.M.); (E.P.); (V.M.)
| | - Vincenzo Musolino
- Institute of Research for Food Safety & Health IRC-FSH, University Magna Graecia, 88100 Catanzaro, Italy; (F.S.); (R.M.); (F.B.); (S.N.); (S.R.); (M.C.Z.); (R.M.); (L.G.); (A.R.C.); (M.G.); (V.M.); (J.M.); (E.P.); (V.M.)
| | - Jessica Maiuolo
- Institute of Research for Food Safety & Health IRC-FSH, University Magna Graecia, 88100 Catanzaro, Italy; (F.S.); (R.M.); (F.B.); (S.N.); (S.R.); (M.C.Z.); (R.M.); (L.G.); (A.R.C.); (M.G.); (V.M.); (J.M.); (E.P.); (V.M.)
| | - Ernesto Palma
- Institute of Research for Food Safety & Health IRC-FSH, University Magna Graecia, 88100 Catanzaro, Italy; (F.S.); (R.M.); (F.B.); (S.N.); (S.R.); (M.C.Z.); (R.M.); (L.G.); (A.R.C.); (M.G.); (V.M.); (J.M.); (E.P.); (V.M.)
- Nutramed S.c.a.r.l., Complesso Ninì Barbieri, Roccelletta di Borgia, 88100 Catanzaro, Italy
| | - Vincenzo Mollace
- Institute of Research for Food Safety & Health IRC-FSH, University Magna Graecia, 88100 Catanzaro, Italy; (F.S.); (R.M.); (F.B.); (S.N.); (S.R.); (M.C.Z.); (R.M.); (L.G.); (A.R.C.); (M.G.); (V.M.); (J.M.); (E.P.); (V.M.)
- Nutramed S.c.a.r.l., Complesso Ninì Barbieri, Roccelletta di Borgia, 88100 Catanzaro, Italy
| |
Collapse
|
27
|
Dobbin SJ, Petrie MC, Myles RC, Touyz RM, Lang NN. Cardiotoxic effects of angiogenesis inhibitors. Clin Sci (Lond) 2021; 135:71-100. [PMID: 33404052 PMCID: PMC7812690 DOI: 10.1042/cs20200305] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2020] [Revised: 12/07/2020] [Accepted: 12/10/2020] [Indexed: 02/06/2023]
Abstract
The development of new therapies for cancer has led to dramatic improvements in survivorship. Angiogenesis inhibitors represent one such advancement, revolutionising treatment for a wide range of malignancies. However, these drugs are associated with cardiovascular toxicities which can impact optimal cancer treatment in the short-term and may lead to increased morbidity and mortality in the longer term. Vascular endothelial growth factor inhibitors (VEGFIs) are associated with hypertension, left ventricular systolic dysfunction (LVSD) and heart failure as well as arterial and venous thromboembolism, QTc interval prolongation and arrhythmia. The mechanisms behind the development of VEGFI-associated LVSD and heart failure likely involve the combination of a number of myocardial insults. These include direct myocardial effects, as well as secondary toxicity via coronary or peripheral vascular damage. Cardiac toxicity may result from the 'on-target' effects of VEGF inhibition or 'off-target' effects resulting from inhibition of other tyrosine kinases. Similar mechanisms may be involved in the development of VEGFI-associated right ventricular (RV) dysfunction. Some VEGFIs can be associated with QTc interval prolongation and an increased risk of ventricular and atrial arrhythmia. Further pre-clinical and clinical studies and trials are needed to better understand the impact of VEGFI on the cardiovascular system. Once mechanisms are elucidated, therapies can be investigated in clinical trials and surveillance strategies for identifying VEGFI-associated cardiovascular complications can be developed.
Collapse
Affiliation(s)
- Stephen J.H. Dobbin
- BHF Glasgow Cardiovascular Research Centre, Institute of Cardiovascular and Medical Sciences, University of Glasgow, 126 University Place, Glasgow, United Kingdom, G12 8TA
| | - Mark C. Petrie
- BHF Glasgow Cardiovascular Research Centre, Institute of Cardiovascular and Medical Sciences, University of Glasgow, 126 University Place, Glasgow, United Kingdom, G12 8TA
| | - Rachel C. Myles
- BHF Glasgow Cardiovascular Research Centre, Institute of Cardiovascular and Medical Sciences, University of Glasgow, 126 University Place, Glasgow, United Kingdom, G12 8TA
| | - Rhian M. Touyz
- BHF Glasgow Cardiovascular Research Centre, Institute of Cardiovascular and Medical Sciences, University of Glasgow, 126 University Place, Glasgow, United Kingdom, G12 8TA
| | - Ninian N. Lang
- BHF Glasgow Cardiovascular Research Centre, Institute of Cardiovascular and Medical Sciences, University of Glasgow, 126 University Place, Glasgow, United Kingdom, G12 8TA
| |
Collapse
|
28
|
Cappetta D, De Angelis A, Flamini S, Cozzolino A, Bereshchenko O, Ronchetti S, Cianflone E, Gagliardi A, Ricci E, Rafaniello C, Rossi F, Riccardi C, Berrino L, Bruscoli S, Urbanek K. Deficit of glucocorticoid-induced leucine zipper amplifies angiotensin-induced cardiomyocyte hypertrophy and diastolic dysfunction. J Cell Mol Med 2021; 25:217-228. [PMID: 33247627 PMCID: PMC7810940 DOI: 10.1111/jcmm.15913] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2020] [Revised: 07/22/2020] [Accepted: 08/08/2020] [Indexed: 12/11/2022] Open
Abstract
Poor prognosis in heart failure and the lack of real breakthrough strategies validate targeting myocardial remodelling and the intracellular signalling involved in this process. So far, there are no effective strategies to counteract hypertrophy, an independent predictor of heart failure progression and death. Glucocorticoid-induced leucine zipper (GILZ) is involved in inflammatory signalling, but its role in cardiac biology is unknown. Using GILZ-knockout (KO) mice and an experimental model of hypertrophy and diastolic dysfunction, we addressed the role of GILZ in adverse myocardial remodelling. Infusion of angiotensin II (Ang II) resulted in myocardial dysfunction, inflammation, apoptosis, fibrosis, capillary rarefaction and hypertrophy. Interestingly, GILZ-KO showed more evident diastolic dysfunction and aggravated hypertrophic response compared with WT after Ang II administration. Both cardiomyocyte and left ventricular hypertrophy were more pronounced in GILZ-KO mice. On the other hand, Ang II-induced inflammatory and fibrotic phenomena, cell death and reduction in microvascular density, remained invariant between the WT and KO groups. The analysis of regulators of hypertrophic response, GATA4 and FoxP3, demonstrated an up-regulation in WT mice infused with Ang II; conversely, such an increase did not occur in GILZ-KO hearts. These data on myocardial response to Ang II in mice lacking GILZ indicate that this protein is a new element that can be mechanistically involved in cardiovascular pathology.
Collapse
Affiliation(s)
- Donato Cappetta
- Department of Experimental MedicineUniversity of Campania 'Luigi Vanvitelli'NaplesItaly
| | - Antonella De Angelis
- Department of Experimental MedicineUniversity of Campania 'Luigi Vanvitelli'NaplesItaly
| | - Sara Flamini
- Department of MedicineSection of PharmacologyUniversity of PerugiaPerugiaItaly
| | - Anna Cozzolino
- Department of Experimental MedicineUniversity of Campania 'Luigi Vanvitelli'NaplesItaly
| | - Oxana Bereshchenko
- Department of Philosophy, Social Sciences and EducationUniversity of PerugiaPerugiaItaly
| | - Simona Ronchetti
- Department of MedicineSection of PharmacologyUniversity of PerugiaPerugiaItaly
| | - Eleonora Cianflone
- Department of Medical and Surgical SciencesUniversity 'Magna Graecia' of CatanzaroCatanzaroItaly
| | - Andrea Gagliardi
- Department of MedicineSection of PharmacologyUniversity of PerugiaPerugiaItaly
| | - Erika Ricci
- Department of MedicineSection of PharmacologyUniversity of PerugiaPerugiaItaly
| | - Concetta Rafaniello
- Department of Experimental MedicineUniversity of Campania 'Luigi Vanvitelli'NaplesItaly
| | - Francesco Rossi
- Department of Experimental MedicineUniversity of Campania 'Luigi Vanvitelli'NaplesItaly
| | - Carlo Riccardi
- Department of MedicineSection of PharmacologyUniversity of PerugiaPerugiaItaly
| | - Liberato Berrino
- Department of Experimental MedicineUniversity of Campania 'Luigi Vanvitelli'NaplesItaly
| | - Stefano Bruscoli
- Department of MedicineSection of PharmacologyUniversity of PerugiaPerugiaItaly
| | - Konrad Urbanek
- Department of Experimental MedicineUniversity of Campania 'Luigi Vanvitelli'NaplesItaly
- Department of Experimental and Clinical MedicineUniversity 'Magna Graecia' of CatanzaroCatanzaroItaly
| |
Collapse
|
29
|
Cianflone E, Cappetta D, Mancuso T, Sabatino J, Marino F, Scalise M, Albanese M, Salatino A, Parrotta EI, Cuda G, De Angelis A, Berrino L, Rossi F, Nadal-Ginard B, Torella D, Urbanek K. Statins Stimulate New Myocyte Formation After Myocardial Infarction by Activating Growth and Differentiation of the Endogenous Cardiac Stem Cells. Int J Mol Sci 2020; 21:ijms21217927. [PMID: 33114544 PMCID: PMC7663580 DOI: 10.3390/ijms21217927] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2020] [Revised: 10/21/2020] [Accepted: 10/22/2020] [Indexed: 12/20/2022] Open
Abstract
The 3-hydroxy-3-methylglutaryl coenzyme A (HMG-CoA) reductase inhibitors (statins) exert pleiotropic effects on cardiac cell biology which are not yet fully understood. Here we tested whether statin treatment affects resident endogenous cardiac stem/progenitor cell (CSC) activation in vitro and in vivo after myocardial infarction (MI). Statins (Rosuvastatin, Simvastatin and Pravastatin) significantly increased CSC expansion in vitro as measured by both BrdU incorporation and cell growth curve. Additionally, statins increased CSC clonal expansion and cardiosphere formation. The effects of statins on CSC growth and differentiation depended on Akt phosphorylation. Twenty-eight days after myocardial infarction by permanent coronary ligation in rats, the number of endogenous CSCs in the infarct border zone was significantly increased by Rosuvastatin-treatment as compared to untreated controls. Additionally, commitment of the activated CSCs into the myogenic lineage (c-kitpos/Gata4pos CSCs) was increased by Rosuvastatin administration. Accordingly, Rosuvastatin fostered new cardiomyocyte formation after MI. Finally, Rosuvastatin treatment reversed the cardiomyogenic defects of CSCs in c-kit haploinsufficient mice, increasing new cardiomyocyte formation by endogenous CSCs in these mice after myocardial infarction. In summary, statins, by sustaining Akt activation, foster CSC growth and differentiation in vitro and in vivo. The activation and differentiation of the endogenous CSC pool and consequent new myocyte formation by statins improve myocardial remodeling after coronary occlusion in rodents. Similar effects might contribute to the beneficial effects of statins on human cardiovascular diseases.
Collapse
Affiliation(s)
- Eleonora Cianflone
- Department of Medical and Surgical Sciences, Magna Graecia University, 88100 Catanzaro, Italy; (E.C.); (J.S.); (M.A.); (E.I.P.); (B.N.-G.)
| | - Donato Cappetta
- Department of Experimental Medicine, University of Campania “L. Vanvitelli”, 80138 Naples, Italy; (D.C.); (A.D.A.); (L.B.); (F.R.)
| | - Teresa Mancuso
- Department of Experimental and Clinical Medicine, Magna Graecia University, 88100 Catanzaro, Italy; (T.M.); (F.M.); (M.S.); (A.S.); (G.C.)
| | - Jolanda Sabatino
- Department of Medical and Surgical Sciences, Magna Graecia University, 88100 Catanzaro, Italy; (E.C.); (J.S.); (M.A.); (E.I.P.); (B.N.-G.)
| | - Fabiola Marino
- Department of Experimental and Clinical Medicine, Magna Graecia University, 88100 Catanzaro, Italy; (T.M.); (F.M.); (M.S.); (A.S.); (G.C.)
| | - Mariangela Scalise
- Department of Experimental and Clinical Medicine, Magna Graecia University, 88100 Catanzaro, Italy; (T.M.); (F.M.); (M.S.); (A.S.); (G.C.)
| | - Michele Albanese
- Department of Medical and Surgical Sciences, Magna Graecia University, 88100 Catanzaro, Italy; (E.C.); (J.S.); (M.A.); (E.I.P.); (B.N.-G.)
| | - Alessandro Salatino
- Department of Experimental and Clinical Medicine, Magna Graecia University, 88100 Catanzaro, Italy; (T.M.); (F.M.); (M.S.); (A.S.); (G.C.)
| | - Elvira Immacolata Parrotta
- Department of Medical and Surgical Sciences, Magna Graecia University, 88100 Catanzaro, Italy; (E.C.); (J.S.); (M.A.); (E.I.P.); (B.N.-G.)
| | - Giovanni Cuda
- Department of Experimental and Clinical Medicine, Magna Graecia University, 88100 Catanzaro, Italy; (T.M.); (F.M.); (M.S.); (A.S.); (G.C.)
| | - Antonella De Angelis
- Department of Experimental Medicine, University of Campania “L. Vanvitelli”, 80138 Naples, Italy; (D.C.); (A.D.A.); (L.B.); (F.R.)
| | - Liberato Berrino
- Department of Experimental Medicine, University of Campania “L. Vanvitelli”, 80138 Naples, Italy; (D.C.); (A.D.A.); (L.B.); (F.R.)
| | - Francesco Rossi
- Department of Experimental Medicine, University of Campania “L. Vanvitelli”, 80138 Naples, Italy; (D.C.); (A.D.A.); (L.B.); (F.R.)
| | - Bernardo Nadal-Ginard
- Department of Medical and Surgical Sciences, Magna Graecia University, 88100 Catanzaro, Italy; (E.C.); (J.S.); (M.A.); (E.I.P.); (B.N.-G.)
| | - Daniele Torella
- Department of Experimental and Clinical Medicine, Magna Graecia University, 88100 Catanzaro, Italy; (T.M.); (F.M.); (M.S.); (A.S.); (G.C.)
- Correspondence: (D.T.); (K.U.)
| | - Konrad Urbanek
- Department of Experimental and Clinical Medicine, Magna Graecia University, 88100 Catanzaro, Italy; (T.M.); (F.M.); (M.S.); (A.S.); (G.C.)
- Correspondence: (D.T.); (K.U.)
| |
Collapse
|
30
|
Cardiac Stem Cell-Loaded Delivery Systems: A New Challenge for Myocardial Tissue Regeneration. Int J Mol Sci 2020; 21:ijms21207701. [PMID: 33080988 PMCID: PMC7589970 DOI: 10.3390/ijms21207701] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2020] [Revised: 10/13/2020] [Accepted: 10/16/2020] [Indexed: 02/06/2023] Open
Abstract
Cardiovascular disease (CVD) remains the leading cause of death in Western countries. Post-myocardial infarction heart failure can be considered a degenerative disease where myocyte loss outweighs any regenerative potential. In this scenario, regenerative biology and tissue engineering can provide effective solutions to repair the infarcted failing heart. The main strategies involve the use of stem and progenitor cells to regenerate/repair lost and dysfunctional tissue, administrated as a suspension or encapsulated in specific delivery systems. Several studies demonstrated that effectiveness of direct injection of cardiac stem cells (CSCs) is limited in humans by the hostile cardiac microenvironment and poor cell engraftment; therefore, the use of injectable hydrogel or pre-formed patches have been strongly advocated to obtain a better integration between delivered stem cells and host myocardial tissue. Several approaches were used to refine these types of constructs, trying to obtain an optimized functional scaffold. Despite the promising features of these stem cells’ delivery systems, few have reached the clinical practice. In this review, we summarize the advantages, and the novelty but also the current limitations of engineered patches and injectable hydrogels for tissue regenerative purposes, offering a perspective of how we believe tissue engineering should evolve to obtain the optimal delivery system applicable to the everyday clinical scenario.
Collapse
|
31
|
Gambini E, Martinelli I, Stadiotti I, Vinci MC, Scopece A, Eramo L, Sommariva E, Resta J, Benaouadi S, Cogliati E, Paolin A, Parini A, Pompilio G, Savagner F. Differences in Mitochondrial Membrane Potential Identify Distinct Populations of Human Cardiac Mesenchymal Progenitor Cells. Int J Mol Sci 2020; 21:ijms21207467. [PMID: 33050449 PMCID: PMC7590175 DOI: 10.3390/ijms21207467] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2020] [Revised: 10/05/2020] [Accepted: 10/07/2020] [Indexed: 02/07/2023] Open
Abstract
Adult human cardiac mesenchymal progenitor cells (hCmPC) are multipotent resident populations involved in cardiac homeostasis and heart repair. Even if the mechanisms have not yet been fully elucidated, the stem cell differentiation is guided by the mitochondrial metabolism; however, mitochondrial approaches to identify hCmPC with enhanced stemness and/or differentiation capability for cellular therapy are not established. Here we demonstrated that hCmPCs sorted for low and high mitochondrial membrane potential (using a lipophilic cationic dye tetramethylrhodamine methyl ester, TMRM), presented differences in energy metabolism from preferential glycolysis to oxidative rates. TMRM-high cells are highly efficient in terms of oxygen consumption rate, basal and maximal respiration, and spare respiratory capacity compared to TMRM-low cells. TMRM-high cells showed characteristics of pre-committed cells and were associated with higher in vitro differentiation capacity through endothelial, cardiac-like, and, to a lesser extent, adipogenic and chondro/osteogenic cell lineage, when compared with TMRM-low cells. Conversely, TMRM-low showed higher self-renewal potential. To conclude, we identified two hCmPC populations with different metabolic profile, stemness maturity, and differentiation potential. Our findings suggest that metabolic sorting can isolate cells with higher regenerative capacity and/or long-term survival. This metabolism-based strategy to select cells may be broadly applicable to therapies.
Collapse
Affiliation(s)
- Elisa Gambini
- Vascular Biology and Regenerative Medicine Unit, Centro Cardiologico Monzino-IRCCS, Via Carlo Parea 4, 20138 Milan, Italy; (I.S.); (M.C.V.); (A.S.); (L.E.); (E.S.); (J.R.); (G.P.)
- Correspondence:
| | - Ilenia Martinelli
- Institute of Metabolic and Cardiovascular Diseases (I2MC), Institut National de la Santé et de la Recherche Médicale (INSERM), Université de Toulouse, 31432 Toulouse, France; (I.M.); (S.B.); (A.P.); (F.S.)
| | - Ilaria Stadiotti
- Vascular Biology and Regenerative Medicine Unit, Centro Cardiologico Monzino-IRCCS, Via Carlo Parea 4, 20138 Milan, Italy; (I.S.); (M.C.V.); (A.S.); (L.E.); (E.S.); (J.R.); (G.P.)
| | - Maria Cristina Vinci
- Vascular Biology and Regenerative Medicine Unit, Centro Cardiologico Monzino-IRCCS, Via Carlo Parea 4, 20138 Milan, Italy; (I.S.); (M.C.V.); (A.S.); (L.E.); (E.S.); (J.R.); (G.P.)
| | - Alessandro Scopece
- Vascular Biology and Regenerative Medicine Unit, Centro Cardiologico Monzino-IRCCS, Via Carlo Parea 4, 20138 Milan, Italy; (I.S.); (M.C.V.); (A.S.); (L.E.); (E.S.); (J.R.); (G.P.)
| | - Luana Eramo
- Vascular Biology and Regenerative Medicine Unit, Centro Cardiologico Monzino-IRCCS, Via Carlo Parea 4, 20138 Milan, Italy; (I.S.); (M.C.V.); (A.S.); (L.E.); (E.S.); (J.R.); (G.P.)
| | - Elena Sommariva
- Vascular Biology and Regenerative Medicine Unit, Centro Cardiologico Monzino-IRCCS, Via Carlo Parea 4, 20138 Milan, Italy; (I.S.); (M.C.V.); (A.S.); (L.E.); (E.S.); (J.R.); (G.P.)
| | - Jessica Resta
- Vascular Biology and Regenerative Medicine Unit, Centro Cardiologico Monzino-IRCCS, Via Carlo Parea 4, 20138 Milan, Italy; (I.S.); (M.C.V.); (A.S.); (L.E.); (E.S.); (J.R.); (G.P.)
- Institute of Metabolic and Cardiovascular Diseases (I2MC), Institut National de la Santé et de la Recherche Médicale (INSERM), Université de Toulouse, 31432 Toulouse, France; (I.M.); (S.B.); (A.P.); (F.S.)
| | - Sabrina Benaouadi
- Institute of Metabolic and Cardiovascular Diseases (I2MC), Institut National de la Santé et de la Recherche Médicale (INSERM), Université de Toulouse, 31432 Toulouse, France; (I.M.); (S.B.); (A.P.); (F.S.)
| | - Elisa Cogliati
- Treviso Tissue Bank Foundation, Via Antonio Scarpa 9, 31100 Treviso, Italy; (E.C.); (A.P.)
| | - Adolfo Paolin
- Treviso Tissue Bank Foundation, Via Antonio Scarpa 9, 31100 Treviso, Italy; (E.C.); (A.P.)
| | - Angelo Parini
- Institute of Metabolic and Cardiovascular Diseases (I2MC), Institut National de la Santé et de la Recherche Médicale (INSERM), Université de Toulouse, 31432 Toulouse, France; (I.M.); (S.B.); (A.P.); (F.S.)
| | - Giulio Pompilio
- Vascular Biology and Regenerative Medicine Unit, Centro Cardiologico Monzino-IRCCS, Via Carlo Parea 4, 20138 Milan, Italy; (I.S.); (M.C.V.); (A.S.); (L.E.); (E.S.); (J.R.); (G.P.)
- Dipartimento di Scienze Cliniche e di Comunità, Università degli Studi di Milano, Via Festa del Perdono 7, 20122 Milan, Italy
| | - Frederique Savagner
- Institute of Metabolic and Cardiovascular Diseases (I2MC), Institut National de la Santé et de la Recherche Médicale (INSERM), Université de Toulouse, 31432 Toulouse, France; (I.M.); (S.B.); (A.P.); (F.S.)
| |
Collapse
|
32
|
Selviler-Sizer S, Kabak YB, Kabak M. Telocytes in the hearts of Saanen goats. Microsc Res Tech 2020; 84:548-554. [PMID: 33017500 DOI: 10.1002/jemt.23612] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2020] [Revised: 09/10/2020] [Accepted: 09/19/2020] [Indexed: 12/21/2022]
Abstract
Telocytes, new interstitial cells that have received significant attention in recent years, have been detected in many organs, including the heart. The distinction between telocytes and other interstitial cells can only be made based on their ultrastructural characterization and immunophenotypic features. In this study, we examined the interstitial cells in the healthy heart tissues of Saanen goats to determine whether they are telocytes or not, by using a scanning electron microscope (SEM) and immunohistochemical and immunofluorescence staining methods. The SEM revealed oval and round telocytes with two to four telopodes. Some telopodes also had podoms. The staining for immunohistochemical and immunofluorescence methods used for CD34, c-kit (CD117), and vimentin antibodies. Positive cells were detected in the heart muscle and heart valves by immunohistochemical staining. As these antigens can also be expressed by other non-telocyte cells, we used double immunofluorescence staining with CD34/c-kit and CD34/vimentin antibodies to identify true telocytes. Telocytes were determined in the right atrium and aortic valve. While telocytes were CD34+/c-kit+ and CD34+/vimentin+, fibroblasts were CD34-/vimentin+. These results confirm the presence of telocytes in the hearts of Saanen goats.
Collapse
Affiliation(s)
- Sedef Selviler-Sizer
- Department of Anatomy, Faculty of Veterinary Medicine, Ondokuz Mayis University, Samsun, Turkey
| | - Yonca Betil Kabak
- Department of Pathology, Faculty of Veterinary Medicine, Ondokuz Mayis University, Samsun, Turkey
| | - Murat Kabak
- Department of Anatomy, Faculty of Veterinary Medicine, Ondokuz Mayis University, Samsun, Turkey
| |
Collapse
|
33
|
Preconditioned and Genetically Modified Stem Cells for Myocardial Infarction Treatment. Int J Mol Sci 2020; 21:ijms21197301. [PMID: 33023264 PMCID: PMC7582407 DOI: 10.3390/ijms21197301] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2020] [Revised: 09/25/2020] [Accepted: 09/25/2020] [Indexed: 02/07/2023] Open
Abstract
Ischemic heart disease and myocardial infarction remain leading causes of mortality worldwide. Existing myocardial infarction treatments are incapable of fully repairing and regenerating the infarcted myocardium. Stem cell transplantation therapy has demonstrated promising results in improving heart function following myocardial infarction. However, poor cell survival and low engraftment at the harsh and hostile environment at the site of infarction limit the regeneration potential of stem cells. Preconditioning with various physical and chemical factors, as well as genetic modification and cellular reprogramming, are strategies that could potentially optimize stem cell transplantation therapy for clinical application. In this review, we discuss the most up-to-date findings related to utilizing preconditioned stem cells for myocardial infarction treatment, focusing mainly on preconditioning with hypoxia, growth factors, drugs, and biological agents. Furthermore, genetic manipulations on stem cells, such as the overexpression of specific proteins, regulation of microRNAs, and cellular reprogramming to improve their efficiency in myocardial infarction treatment, are discussed as well.
Collapse
|
34
|
Cardiac Fibroblast-Induced Pluripotent Stem Cell-Derived Exosomes as a Potential Therapeutic Mean for Heart Failure. Int J Mol Sci 2020; 21:ijms21197215. [PMID: 33003641 PMCID: PMC7582575 DOI: 10.3390/ijms21197215] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2020] [Revised: 09/23/2020] [Accepted: 09/24/2020] [Indexed: 12/20/2022] Open
Abstract
The limited regenerative capacity of the injured myocardium leads to remodeling and often heart failure. Novel therapeutic approaches are essential. Induced pluripotent stem cells (iPSC) differentiated into cardiomyocytes are a potential future therapeutics. We hypothesized that organ-specific reprogramed fibroblasts may serve an advantageous source for future cardiomyocytes. Moreover, exosomes secreted from those cells may have a beneficial effect on cardiac differentiation and/or function. We compared RNA from different sources of human iPSC using chip gene expression. Protein expression was evaluated as well as exosome micro-RNA levels and their impact on embryoid bodies (EBs) differentiation. Statistical analysis identified 51 genes that were altered (p ≤ 0.05), and confirmed in the protein level, cardiac fibroblasts-iPSCs (CF-iPSCs) vs. dermal fibroblasts-iPSCs (DF-iPSCs). Several miRs were altered especially miR22, a key regulator of cardiac hypertrophy and remodeling. Lower expression of miR22 in CF-iPSCs vs. DF-iPSCs was observed. EBs treated with these exosomes exhibited more beating EBs p = 0.05. vs. control. We identify CF-iPSC and its exosomes as a potential source for cardiac recovery induction. The decrease in miR22 level points out that our CF-iPSC-exosomes are naïve of congestive heart cell memory, making them a potential biological source for future therapy for the injured heart.
Collapse
|
35
|
Nicks AM, Kesteven SH, Li M, Wu J, Chan AY, Naqvi N, Husain A, Feneley MP, Smith NJ, Iismaa SE, Graham RM. Pressure overload by suprarenal aortic constriction in mice leads to left ventricular hypertrophy without c-Kit expression in cardiomyocytes. Sci Rep 2020; 10:15318. [PMID: 32948799 PMCID: PMC7501855 DOI: 10.1038/s41598-020-72273-3] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2020] [Accepted: 08/25/2020] [Indexed: 01/03/2023] Open
Abstract
Animal models of pressure overload are valuable for understanding hypertensive heart disease. We characterised a surgical model of pressure overload-induced hypertrophy in C57BL/6J mice produced by suprarenal aortic constriction (SAC). Compared to sham controls, at one week post-SAC systolic blood pressure was significantly elevated and left ventricular (LV) hypertrophy was evident by a 50% increase in the LV weight-to-tibia length ratio due to cardiomyocyte hypertrophy. As a result, LV end-diastolic wall thickness-to-chamber radius (h/R) ratio increased, consistent with the development of concentric hypertrophy. LV wall thickening was not sufficient to normalise LV wall stress, which also increased, resulting in LV systolic dysfunction with reductions in ejection fraction and fractional shortening, but no evidence of heart failure. Pathological LV remodelling was evident by the re-expression of fetal genes and coronary artery perivascular fibrosis, with ischaemia indicated by enhanced cardiomyocyte Hif1a expression. The expression of stem cell factor receptor, c-Kit, was low basally in cardiomyocytes and did not change following the development of robust hypertrophy, suggesting there is no role for cardiomyocyte c-Kit signalling in pathological LV remodelling following pressure overload.
Collapse
Affiliation(s)
- Amy M Nicks
- Division of Molecular Cardiology and Biophysics, Victor Chang Cardiac Research Institute, 405 Liverpool Street, Darlinghurst, Sydney, NSW, 2010, Australia
- St Vincent's Clinical School, University of New South Wales, Sydney, NSW, 2052, Australia
| | - Scott H Kesteven
- Division of Molecular Cardiology and Biophysics, Victor Chang Cardiac Research Institute, 405 Liverpool Street, Darlinghurst, Sydney, NSW, 2010, Australia
- St Vincent's Clinical School, University of New South Wales, Sydney, NSW, 2052, Australia
| | - Ming Li
- Division of Molecular Cardiology and Biophysics, Victor Chang Cardiac Research Institute, 405 Liverpool Street, Darlinghurst, Sydney, NSW, 2010, Australia
- Cardiac Regeneration Research Institute, Wenzhou Medical University, Wenzhou, 325035, China
| | - Jianxin Wu
- Division of Molecular Cardiology and Biophysics, Victor Chang Cardiac Research Institute, 405 Liverpool Street, Darlinghurst, Sydney, NSW, 2010, Australia
| | - Andrea Y Chan
- Division of Molecular Cardiology and Biophysics, Victor Chang Cardiac Research Institute, 405 Liverpool Street, Darlinghurst, Sydney, NSW, 2010, Australia
| | - Nawazish Naqvi
- Department of Medicine, Emory University, Atlanta, GA, 30322, USA
| | - Ahsan Husain
- Department of Medicine, Emory University, Atlanta, GA, 30322, USA
| | - Michael P Feneley
- Division of Molecular Cardiology and Biophysics, Victor Chang Cardiac Research Institute, 405 Liverpool Street, Darlinghurst, Sydney, NSW, 2010, Australia
- St Vincent's Clinical School, University of New South Wales, Sydney, NSW, 2052, Australia
| | - Nicola J Smith
- Division of Molecular Cardiology and Biophysics, Victor Chang Cardiac Research Institute, 405 Liverpool Street, Darlinghurst, Sydney, NSW, 2010, Australia
- St Vincent's Clinical School, University of New South Wales, Sydney, NSW, 2052, Australia
| | - Siiri E Iismaa
- Division of Molecular Cardiology and Biophysics, Victor Chang Cardiac Research Institute, 405 Liverpool Street, Darlinghurst, Sydney, NSW, 2010, Australia
- St Vincent's Clinical School, University of New South Wales, Sydney, NSW, 2052, Australia
| | - Robert M Graham
- Division of Molecular Cardiology and Biophysics, Victor Chang Cardiac Research Institute, 405 Liverpool Street, Darlinghurst, Sydney, NSW, 2010, Australia.
- St Vincent's Clinical School, University of New South Wales, Sydney, NSW, 2052, Australia.
| |
Collapse
|
36
|
PIM1 Promotes Survival of Cardiomyocytes by Upregulating c-Kit Protein Expression. Cells 2020; 9:cells9092001. [PMID: 32878131 PMCID: PMC7563506 DOI: 10.3390/cells9092001] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2020] [Revised: 08/21/2020] [Accepted: 08/27/2020] [Indexed: 12/17/2022] Open
Abstract
Enhancing cardiomyocyte survival is crucial to blunt deterioration of myocardial structure and function following pathological damage. PIM1 (Proviral Insertion site in Murine leukemia virus (PIM) kinase 1) is a cardioprotective serine threonine kinase that promotes cardiomyocyte survival and antagonizes senescence through multiple concurrent molecular signaling cascades. In hematopoietic stem cells, PIM1 interacts with the receptor tyrosine kinase c-Kit upstream of the ERK (Extracellular signal-Regulated Kinase) and Akt signaling pathways involved in cell proliferation and survival. The relationship between PIM1 and c-Kit activity has not been explored in the myocardial context. This study delineates the interaction between PIM1 and c-Kit leading to enhanced protection of cardiomyocytes from stress. Elevated c-Kit expression is induced in isolated cardiomyocytes from mice with cardiac-specific overexpression of PIM1. Co-immunoprecipitation and proximity ligation assay reveal protein–protein interaction between PIM1 and c-Kit. Following treatment with Stem Cell Factor, PIM1-overexpressing cardiomyocytes display elevated ERK activity consistent with c-Kit receptor activation. Functionally, elevated c-Kit expression confers enhanced protection against oxidative stress in vitro. This study identifies the mechanistic relationship between PIM1 and c-Kit in cardiomyocytes, demonstrating another facet of cardioprotection regulated by PIM1 kinase.
Collapse
|
37
|
Sayed RKA, Mokhtar DM, Fernández-Ortiz M, Fernández-Martínez J, Aranda-Martínez P, Escames G, Acuña-Castroviejo D. Lack of retinoid acid receptor-related orphan receptor alpha accelerates and melatonin supplementation prevents testicular aging. Aging (Albany NY) 2020; 12:12648-12668. [PMID: 32644943 PMCID: PMC7377884 DOI: 10.18632/aging.103654] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2020] [Accepted: 06/18/2020] [Indexed: 12/11/2022]
Abstract
The role of retinoid acid receptor-related orphan receptor alpha (RORα) on male reproductive functions during aging is unclear. Here, we analyze the morphological changes in the testis of both young and aged RORα-deficient mice, with and without melatonin supplementation. Young mutants showed vacuolation, degeneration and pyknosis of spermatogenic epithelium and Sertoli cells. Aged mutants showed atrophy of the seminiferous tubules and absence of mitotic spermatogenic cells. Absence of sperms in many tubules, loss of acrosomal cap, vacuolation and hypertrophy of Sertoli cells were detected in aged mice, with a significant reduction in the number of seminiferous tubules and a significant increase in the number of Leydig cells and telocytes. Repair in seminiferous tubules and interstitial tissues with enhancement of spermatogenesis was observed in melatonin-treated aged mice. Young mutants overexpressed VEGF that was weaker in aged animals and observed only in the spermatocytes, while melatonin increased VEGF expression in spermatocytes and spermatids. Caspase 3 increased in both young and aged mutant mice in all seminiferous tubules and interstitium; caspase 3 immunostaining in seminiferous tubules, however, showed a normal pattern of apoptosis with melatonin supplementation. The present study reports that age-dependent testicular changes in RORα mutant mice were recovered by melatonin treatment.
Collapse
Affiliation(s)
- Ramy K A Sayed
- Instituto de Biotecnología, Centro de Investigación Biomédica, Parque Tecnológico de Ciencias de la Salud, Universidad de Granada, Granada 18016, Spain.,Department of Anatomy and Embryology, Faculty of Veterinary Medicine, Sohag University, Sohag 82524, Egypt
| | - Doaa M Mokhtar
- Department of Anatomy and Histology, Faculty of Veterinary Medicine, Assiut University, Assiut 71526, Egypt
| | - Marisol Fernández-Ortiz
- Instituto de Biotecnología, Centro de Investigación Biomédica, Parque Tecnológico de Ciencias de la Salud, Universidad de Granada, Granada 18016, Spain.,Departamento de Fisiología, Facultad de Medicina, Universidad de Granada, Granada 18016, Spain
| | - José Fernández-Martínez
- Instituto de Biotecnología, Centro de Investigación Biomédica, Parque Tecnológico de Ciencias de la Salud, Universidad de Granada, Granada 18016, Spain.,Departamento de Fisiología, Facultad de Medicina, Universidad de Granada, Granada 18016, Spain
| | - Paula Aranda-Martínez
- Instituto de Biotecnología, Centro de Investigación Biomédica, Parque Tecnológico de Ciencias de la Salud, Universidad de Granada, Granada 18016, Spain.,Departamento de Fisiología, Facultad de Medicina, Universidad de Granada, Granada 18016, Spain
| | - Germaine Escames
- Instituto de Biotecnología, Centro de Investigación Biomédica, Parque Tecnológico de Ciencias de la Salud, Universidad de Granada, Granada 18016, Spain.,Departamento de Fisiología, Facultad de Medicina, Universidad de Granada, Granada 18016, Spain.,CIBER de Fragilidad y Envejecimiento, Ibs. Granada, Unidad de Gestión Clínica de Laboratorios Clínicos, Hospital Universitario San Cecilio, Granada 18016, Spain
| | - Darío Acuña-Castroviejo
- Instituto de Biotecnología, Centro de Investigación Biomédica, Parque Tecnológico de Ciencias de la Salud, Universidad de Granada, Granada 18016, Spain.,Departamento de Fisiología, Facultad de Medicina, Universidad de Granada, Granada 18016, Spain.,CIBER de Fragilidad y Envejecimiento, Ibs. Granada, Unidad de Gestión Clínica de Laboratorios Clínicos, Hospital Universitario San Cecilio, Granada 18016, Spain
| |
Collapse
|
38
|
Kotov G, Landzhov B, Stamenov N, Stanchev S, Iliev A. Changes in the number of mast cells, expression of fibroblast growth factor-2 and extent of interstitial fibrosis in established and advanced hypertensive heart disease. Ann Anat 2020; 232:151564. [PMID: 32603827 DOI: 10.1016/j.aanat.2020.151564] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2020] [Revised: 06/12/2020] [Accepted: 06/17/2020] [Indexed: 12/16/2022]
Abstract
INTRODUCTION An increasing number of studies have shed light on the role of cardiac mast cells in the pathogenesis of hypertension-induced myocardial remodeling. Mast cells promote fibroblast activation, myofibroblast differentiation and subsequent collagen accumulation through the action of tryptase, chymase, histamine and fibroblast growth factor-2. The aim of the present study was to report on the changes in the number of mast cells as evaluated through toluidine blue, tryptase and c-kit staining, to assess the extent of interstitial fibrosis and correlate it with the changes in the number of mast cells and to analyze the immunohistochemical expression of fibroblast growth factor-2 in two groups of spontaneously hypertensive rats indicative of established and advanced hypertensive heart disease. A novel aspect of our work was the analysis of all parameters in the right ventricle. MATERIAL AND METHODS For the present study, we used 6- and 12-month-old spontaneously hypertensive rats. A light microscopic study was conducted on sections stained with hematoxylin and eosin and toluidine blue. For the immunohistochemical study we used monoclonal antibodies against mast cell tryptase and fibroblast growth factor-2 and a polyclonal antibody against c-kit. The expression of fibroblast growth factor-2 was assessed semi-quantitatively through ImageJ. The number of mast cells was evaluated on toluidine blue-, tryptase- and c-kit-stained sections and a comparative statistical analysis with the Mann-Whitney test was conducted between the two age groups. A separate statistical analysis between results obtained through immunostaining for tryptase and for c-kit was conducted in each age group with the Wilcoxon signed-rank test. The extent of fibrosis was assessed quantitatively on slides stained with Mallory's trichrome stain as a percentage of the whole tissue and compared between the two age groups. Spearman's correlation was used to test whether a correlation exists between the number of mast cells and the percentage of interstitial fibrosis. RESULTS Mast cells with typical cytoplasmic granules were visualized in the interstitial tissue and in the perivascular zone in both age groups. In both ventricles, their number increased significantly in 12-month-old animals as evaluated through all three staining methods. Moreover, immunostaining for tryptase and for c-kit yielded comparable results. The immunoreactivity of fibroblast growth factor-2 increased in both ventricles in older animals. Expression of this protein was particularly intensive in the cytoplasm of connective tissue cells with the characteristic features of mast cells mainly found in the areas of fibrotic alterations in 12-month-old spontaneously hypertensive rats. In both ventricles, interstitial fibrosis was more extensive throughout the myocardium of older animals and was positively correlated with the changes in the number of mast cells in both age groups. CONCLUSION The present study reported for the first time that the increase in the number of mast cells, observed as hypertension-induced myocardial changes progress, is statistically significant and confirmed that this process takes place in both ventricles. This increase is accompanied by a higher expression of fibroblast growth factor-2 and is more strongly correlated with the more pronounced interstitial fibrosis in older animals, further supporting the role of mast cells in the structural changes taking place in the myocardium in response to systemic hypertension.
Collapse
Affiliation(s)
- Georgi Kotov
- Department of Anatomy, Histology and Embryology, Medical University of Sofia, Bulgaria.
| | - Boycho Landzhov
- Department of Anatomy, Histology and Embryology, Medical University of Sofia, Bulgaria
| | - Nikola Stamenov
- Department of Anatomy, Histology and Embryology, Medical University of Sofia, Bulgaria
| | - Stancho Stanchev
- Department of Anatomy, Histology and Embryology, Medical University of Sofia, Bulgaria
| | - Alexandar Iliev
- Department of Anatomy, Histology and Embryology, Medical University of Sofia, Bulgaria
| |
Collapse
|
39
|
Cianflone E, Torella M, Biamonte F, De Angelis A, Urbanek K, Costanzo FS, Rota M, Ellison-Hughes GM, Torella D. Targeting Cardiac Stem Cell Senescence to Treat Cardiac Aging and Disease. Cells 2020; 9:E1558. [PMID: 32604861 PMCID: PMC7349658 DOI: 10.3390/cells9061558] [Citation(s) in RCA: 79] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2020] [Revised: 06/19/2020] [Accepted: 06/25/2020] [Indexed: 12/13/2022] Open
Abstract
Adult stem/progenitor are a small population of cells that reside in tissue-specific niches and possess the potential to differentiate in all cell types of the organ in which they operate. Adult stem cells are implicated with the homeostasis, regeneration, and aging of all tissues. Tissue-specific adult stem cell senescence has emerged as an attractive theory for the decline in mammalian tissue and organ function during aging. Cardiac aging, in particular, manifests as functional tissue degeneration that leads to heart failure. Adult cardiac stem/progenitor cell (CSC) senescence has been accordingly associated with physiological and pathological processes encompassing both non-age and age-related decline in cardiac tissue repair and organ dysfunction and disease. Senescence is a highly active and dynamic cell process with a first classical hallmark represented by its replicative limit, which is the establishment of a stable growth arrest over time that is mainly secondary to DNA damage and reactive oxygen species (ROS) accumulation elicited by different intrinsic stimuli (like metabolism), as well as external stimuli and age. Replicative senescence is mainly executed by telomere shortening, the activation of the p53/p16INK4/Rb molecular pathways, and chromatin remodeling. In addition, senescent cells produce and secrete a complex mixture of molecules, commonly known as the senescence-associated secretory phenotype (SASP), that regulate most of their non-cell-autonomous effects. In this review, we discuss the molecular and cellular mechanisms regulating different characteristics of the senescence phenotype and their consequences for adult CSCs in particular. Because senescent cells contribute to the outcome of a variety of cardiac diseases, including age-related and unrelated cardiac diseases like diabetic cardiomyopathy and anthracycline cardiotoxicity, therapies that target senescent cell clearance are actively being explored. Moreover, the further understanding of the reversibility of the senescence phenotype will help to develop novel rational therapeutic strategies.
Collapse
Affiliation(s)
- Eleonora Cianflone
- Department of Medical and Surgical Sciences, Magna Graecia University, 88100 Catanzaro, Italy;
| | - Michele Torella
- Department of Translational Medical Sciences, AORN dei Colli/Monaldi Hospital, University of Campania “L. Vanvitelli”, Via Leonardo Bianchi, 80131 Naples, Italy;
| | - Flavia Biamonte
- Department of Experimental and Clinical Medicine and Interdepartmental Centre of Services (CIS), Magna Graecia University, 88100 Catanzaro, Italy; (F.B.); (F.S.C.)
| | - Antonella De Angelis
- Department of Experimental Medicine, Section of Pharmacology, University of Campania “L.Vanvitelli”, 80121 Naples, Italy;
| | - Konrad Urbanek
- Molecular and Cellular Cardiology, Department of Experimental and Clinical Medicine, Magna Graecia University, 88100 Catanzaro, Italy
| | - Francesco S. Costanzo
- Department of Experimental and Clinical Medicine and Interdepartmental Centre of Services (CIS), Magna Graecia University, 88100 Catanzaro, Italy; (F.B.); (F.S.C.)
| | - Marcello Rota
- Department of Physiology, New York Medical College, Valhalla, NY 10595, USA;
| | - 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 & Medicine, King’s College London, Guys Campus-Great Maze Pond rd, London SE1 1UL, UK;
| | - Daniele Torella
- Molecular and Cellular Cardiology, Department of Experimental and Clinical Medicine, Magna Graecia University, 88100 Catanzaro, Italy
| |
Collapse
|
40
|
Mancuso T, Barone A, Salatino A, Molinaro C, Marino F, Scalise M, Torella M, De Angelis A, Urbanek K, Torella D, Cianflone E. Unravelling the Biology of Adult Cardiac Stem Cell-Derived Exosomes to Foster Endogenous Cardiac Regeneration and Repair. Int J Mol Sci 2020; 21:E3725. [PMID: 32466282 PMCID: PMC7279257 DOI: 10.3390/ijms21103725] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2020] [Revised: 05/15/2020] [Accepted: 05/20/2020] [Indexed: 12/11/2022] Open
Abstract
Cardiac remuscularization has been the stated goal of the field of regenerative cardiology since its inception. Along with the refreshment of lost and dysfunctional cardiac muscle cells, the field of cell therapy has expanded in scope encompassing also the potential of the injected cells as cardioprotective and cardio-reparative agents for cardiovascular diseases. The latter has been the result of the findings that cell therapies so far tested in clinical trials exert their beneficial effects through paracrine mechanisms acting on the endogenous myocardial reparative/regenerative potential. The endogenous regenerative potential of the adult heart is still highly debated. While it has been widely accepted that adult cardiomyocytes (CMs) are renewed throughout life either in response to wear and tear and after injury, the rate and origin of this phenomenon are yet to be clarified. The adult heart harbors resident cardiac/stem progenitor cells (CSCs/CPCs), whose discovery and characterization were initially sufficient to explain CM renewal in response to physiological and pathological stresses, when also considering that adult CMs are terminally differentiated cells. The role of CSCs in CM formation in the adult heart has been however questioned by some recent genetic fate map studies, which have been proved to have serious limitations. Nevertheless, uncontested evidence shows that clonal CSCs are effective transplantable regenerative agents either for their direct myogenic differentiation and for their paracrine effects in the allogeneic setting. In particular, the paracrine potential of CSCs has been the focus of the recent investigation, whereby CSC-derived exosomes appear to harbor relevant regenerative and reparative signals underlying the beneficial effects of CSC transplantation. This review focuses on recent advances in our knowledge about the biological role of exosomes in heart tissue homeostasis and repair with the idea to use them as tools for new therapeutic biotechnologies for "cell-less" effective cardiac regeneration approaches.
Collapse
Affiliation(s)
- Teresa Mancuso
- Molecular and Cellular Cardiology, Department of Experimental and Clinical Medicine, Magna Graecia University, 88100 Catanzaro, Italy; (T.M.); (A.B.); (A.S.); (C.M.); (F.M.); (M.S.); (K.U.)
| | - Antonella Barone
- Molecular and Cellular Cardiology, Department of Experimental and Clinical Medicine, Magna Graecia University, 88100 Catanzaro, Italy; (T.M.); (A.B.); (A.S.); (C.M.); (F.M.); (M.S.); (K.U.)
| | - Alessandro Salatino
- Molecular and Cellular Cardiology, Department of Experimental and Clinical Medicine, Magna Graecia University, 88100 Catanzaro, Italy; (T.M.); (A.B.); (A.S.); (C.M.); (F.M.); (M.S.); (K.U.)
| | - Claudia Molinaro
- Molecular and Cellular Cardiology, Department of Experimental and Clinical Medicine, Magna Graecia University, 88100 Catanzaro, Italy; (T.M.); (A.B.); (A.S.); (C.M.); (F.M.); (M.S.); (K.U.)
| | - Fabiola Marino
- Molecular and Cellular Cardiology, Department of Experimental and Clinical Medicine, Magna Graecia University, 88100 Catanzaro, Italy; (T.M.); (A.B.); (A.S.); (C.M.); (F.M.); (M.S.); (K.U.)
| | - Mariangela Scalise
- Molecular and Cellular Cardiology, Department of Experimental and Clinical Medicine, Magna Graecia University, 88100 Catanzaro, Italy; (T.M.); (A.B.); (A.S.); (C.M.); (F.M.); (M.S.); (K.U.)
| | - Michele Torella
- Department of Translational Medical Sciences, AORN dei Colli/Monaldi Hospital, University of Campania “L. Vanvitelli”, Via Leonardo Bianchi, 80131 Naples, Italy;
| | - Antonella De Angelis
- Department of Experimental Medicine, Section of Pharmacology, University of Campania “L.Vanvitelli”, 80121 Naples, Italy;
| | - Konrad Urbanek
- Molecular and Cellular Cardiology, Department of Experimental and Clinical Medicine, Magna Graecia University, 88100 Catanzaro, Italy; (T.M.); (A.B.); (A.S.); (C.M.); (F.M.); (M.S.); (K.U.)
| | - Daniele Torella
- Molecular and Cellular Cardiology, Department of Experimental and Clinical Medicine, Magna Graecia University, 88100 Catanzaro, Italy; (T.M.); (A.B.); (A.S.); (C.M.); (F.M.); (M.S.); (K.U.)
| | - Eleonora Cianflone
- Molecular and Cellular Cardiology, Department of Medical and Surgical Sciences, Magna Graecia University, 88100 Catanzaro, Italy;
| |
Collapse
|
41
|
Cassani M, Fernandes S, Vrbsky J, Ergir E, Cavalieri F, Forte G. Combining Nanomaterials and Developmental Pathways to Design New Treatments for Cardiac Regeneration: The Pulsing Heart of Advanced Therapies. Front Bioeng Biotechnol 2020; 8:323. [PMID: 32391340 PMCID: PMC7193099 DOI: 10.3389/fbioe.2020.00323] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2019] [Accepted: 03/24/2020] [Indexed: 12/12/2022] Open
Abstract
The research for heart therapies is challenged by the limited intrinsic regenerative capacity of the adult heart. Moreover, it has been hampered by the poor results obtained by tissue engineering and regenerative medicine attempts at generating functional beating constructs able to integrate with the host tissue. For this reason, organ transplantation remains the elective treatment for end-stage heart failure, while novel strategies aiming to promote cardiac regeneration or repair lag behind. The recent discovery that adult cardiomyocytes can be ectopically induced to enter the cell cycle and proliferate by a combination of microRNAs and cardioprotective drugs, like anti-oxidant, anti-inflammatory, anti-coagulants and anti-platelets agents, fueled the quest for new strategies suited to foster cardiac repair. While proposing a revolutionary approach for heart regeneration, these studies raised serious issues regarding the efficient controlled delivery of the therapeutic cargo, as well as its timely removal or metabolic inactivation from the site of action. Especially, there is need for innovative treatment because of evidence of severe side effects caused by pleiotropic drugs. Biocompatible nanoparticles possess unique physico-chemical properties that have been extensively exploited for overcoming the limitations of standard medical therapies. Researchers have put great efforts into the optimization of the nanoparticles synthesis and functionalization, to control their interactions with the biological milieu and use as a viable alternative to traditional approaches. Nanoparticles can be used for diagnosis and deliver therapies in a personalized and targeted fashion. Regarding the treatment of cardiovascular diseases, nanoparticles-based strategies have provided very promising outcomes, in preclinical studies, during the last years. Efficient encapsulation of a large variety of cargos, specific release at the desired site and improvement of cardiac function are some of the main achievements reached so far by nanoparticle-based treatments in animal models. This work offers an overview on the recent nanomedical applications for cardiac regeneration and highlights how the versatility of nanomaterials can be combined with the newest molecular biology discoveries to advance cardiac regeneration therapies.
Collapse
Affiliation(s)
- Marco Cassani
- International Clinical Research Center, St Anne’s University Hospital, Brno, Czechia
| | - Soraia Fernandes
- International Clinical Research Center, St Anne’s University Hospital, Brno, Czechia
| | - Jan Vrbsky
- International Clinical Research Center, St Anne’s University Hospital, Brno, Czechia
| | - Ece Ergir
- International Clinical Research Center, St Anne’s University Hospital, Brno, Czechia
- Faculty of Technical Chemistry, Institute of Applied Synthetic Chemistry and Institute of Chemical Technologies and Analytics, Vienna University of Technology, Vienna, Austria
| | - Francesca Cavalieri
- School of Science, RMIT University, Melbourne, VIC, Australia
- Dipartimento di Scienze e Tecnologie Chimiche, Università di Roma “Tor Vergata”, Via Della Ricerca Scientifica, Rome, Italy
| | - Giancarlo Forte
- International Clinical Research Center, St Anne’s University Hospital, Brno, Czechia
| |
Collapse
|
42
|
Barreto S, Hamel L, Schiatti T, Yang Y, George V. Cardiac Progenitor Cells from Stem Cells: Learning from Genetics and Biomaterials. Cells 2019; 8:E1536. [PMID: 31795206 PMCID: PMC6952950 DOI: 10.3390/cells8121536] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2019] [Revised: 11/20/2019] [Accepted: 11/21/2019] [Indexed: 02/07/2023] Open
Abstract
Cardiac Progenitor Cells (CPCs) show great potential as a cell resource for restoring cardiac function in patients affected by heart disease or heart failure. CPCs are proliferative and committed to cardiac fate, capable of generating cells of all the cardiac lineages. These cells offer a significant shift in paradigm over the use of human induced pluripotent stem cell (iPSC)-derived cardiomyocytes owing to the latter's inability to recapitulate mature features of a native myocardium, limiting their translational applications. The iPSCs and direct reprogramming of somatic cells have been attempted to produce CPCs and, in this process, a variety of chemical and/or genetic factors have been evaluated for their ability to generate, expand, and maintain CPCs in vitro. However, the precise stoichiometry and spatiotemporal activity of these factors and the genetic interplay during embryonic CPC development remain challenging to reproduce in culture, in terms of efficiency, numbers, and translational potential. Recent advances in biomaterials to mimic the native cardiac microenvironment have shown promise to influence CPC regenerative functions, while being capable of integrating with host tissue. This review highlights recent developments and limitations in the generation and use of CPCs from stem cells, and the trends that influence the direction of research to promote better application of CPCs.
Collapse
Affiliation(s)
- Sara Barreto
- Guy Hilton Research Centre, School of Pharmacy & Bioengineering, Keele University, Staffordshire ST4 7QB, UK; (S.B.); (T.S.); (Y.Y.)
| | | | - Teresa Schiatti
- Guy Hilton Research Centre, School of Pharmacy & Bioengineering, Keele University, Staffordshire ST4 7QB, UK; (S.B.); (T.S.); (Y.Y.)
| | - Ying Yang
- Guy Hilton Research Centre, School of Pharmacy & Bioengineering, Keele University, Staffordshire ST4 7QB, UK; (S.B.); (T.S.); (Y.Y.)
| | - Vinoj George
- Guy Hilton Research Centre, School of Pharmacy & Bioengineering, Keele University, Staffordshire ST4 7QB, UK; (S.B.); (T.S.); (Y.Y.)
| |
Collapse
|
43
|
Gude NA, Sussman MA. Cardiac regenerative therapy: Many paths to repair. Trends Cardiovasc Med 2019; 30:338-343. [PMID: 31515053 DOI: 10.1016/j.tcm.2019.08.009] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/04/2019] [Revised: 08/14/2019] [Accepted: 08/29/2019] [Indexed: 12/17/2022]
Abstract
Cardiovascular disease remains the primary cause of death in the United States and in most nations worldwide, despite ongoing intensive efforts to promote cardiac health and treat heart failure. Replacing damaged myocardium represents perhaps the most promising treatment strategy, but also the most challenging given that the adult mammalian heart is notoriously resistant to endogenous repair. Cardiac regeneration following pathologic challenge would require proliferation of surviving tissue, expansion and differentiation of resident progenitors, or transdifferentiation of exogenously applied progenitor cells into functioning myocardium. Adult cardiomyocyte proliferation has been the focus of investigation for decades, recently enjoying a renaissance of interest as a therapeutic strategy for reversing cardiomyocyte loss due in large part to ongoing controversies and frustrations with myocardial cell therapy outcomes. The promise of cardiac cell therapy originated with reports of resident adult cardiac stem cells that could be isolated, expanded and reintroduced into damaged myocardium, producing beneficial effects in preclinical animal models. Despite modest functional improvements, Phase I clinical trials using autologous cardiac derived cells have proven safe and effective, setting the stage for an ongoing multi-center Phase II trial combining autologous cardiac stem cell types to enhance beneficial effects. This overview will examine the history of these two approaches for promoting cardiac repair and attempt to provide context for current and future directions in cardiac regenerative research.
Collapse
Affiliation(s)
- Natalie A Gude
- SDSU Heart Institute and Biology Department, San Diego State University, 5500 Campanile Drive, San Diego, CA 92182, USA
| | - Mark A Sussman
- SDSU Heart Institute and Biology Department, San Diego State University, 5500 Campanile Drive, San Diego, CA 92182, USA.
| |
Collapse
|