1
|
Wood C, Saltera Z, Garcia I, Nguyen M, Rios A, Oropeza J, Ugwa D, Mukherjee U, Sehar U, Reddy PH. Age-associated changes in the heart: implications for COVID-19 therapies. Aging (Albany NY) 2025; 17:206251. [PMID: 40372276 DOI: 10.18632/aging.206251] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2025] [Accepted: 04/22/2025] [Indexed: 05/16/2025]
Abstract
Cardiac aging involves progressive structural, functional, cellular, and molecular changes that impair heart function. This review explores key mechanisms, including oxidative stress, mitochondrial dysfunction, impaired autophagy, and chronic low-grade inflammation. Excess reactive oxygen species (ROS) damage heart muscle cells, contributing to fibrosis and cellular aging. Mitochondrial dysfunction reduces energy production and increases oxidative stress, accelerating cardiac decline. Impaired autophagy limits the removal of damaged proteins and organelles, while inflammation activates signaling molecules that drive tissue remodeling. Gender differences reveal estrogen's protective role in premenopausal women, with men showing greater susceptibility to heart muscle dysfunction and injury. After menopause, women lose this hormonal protection, increasing their risk of cardiovascular conditions. Ethnic disparities, particularly among underserved minority populations, emphasize how social factors such as access to care, environment, and chronic stress contribute to worsening cardiovascular outcomes. The coronavirus disease pandemic has introduced further challenges by increasing the incidence of heart damage through inflammation, blood clots, and long-term heart failure, especially in older adults with existing metabolic conditions like diabetes and high blood pressure. The virus's interaction with receptors on heart and blood vessel cells, along with a weakened immune response in older adults, intensifies cardiac aging. Emerging therapies include delivery of therapeutic extracellular vesicles, immune cell modulation, and treatments targeting mitochondria. In addition, lifestyle strategies such as regular physical activity, nutritional improvements, and stress reduction remain vital to maintaining cardiac health. Understanding how these biological and social factors intersect is critical to developing targeted strategies that promote healthy aging of the heart.
Collapse
Affiliation(s)
- Colby Wood
- Department of Internal Medicine, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA
| | - Zach Saltera
- Department of Internal Medicine, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA
| | - Isaiah Garcia
- Department of Internal Medicine, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA
| | - Michelle Nguyen
- Department of Internal Medicine, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA
| | - Andres Rios
- Department of Internal Medicine, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA
| | - Jacqui Oropeza
- Department of Internal Medicine, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA
| | - Destiny Ugwa
- Department of Internal Medicine, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA
| | - Upasana Mukherjee
- Department of Internal Medicine, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA
| | - Ujala Sehar
- Department of Internal Medicine, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA
| | - P Hemachandra Reddy
- Department of Internal Medicine, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA
- Nutritional Sciences Department, College Human Sciences, Texas Tech University, Lubbock, TX 79409, USA
- Department of Pharmacology and Neuroscience, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA
- Department of Neurology, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA
- Department of Public Health, Graduate School of Biomedical Sciences, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA
- Department of Speech, Language, and Hearing Sciences, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA
| |
Collapse
|
2
|
Li D, Li Y, Ding H, Wang Y, Xie Y, Zhang X. Cellular Senescence in Cardiovascular Diseases: From Pathogenesis to Therapeutic Challenges. J Cardiovasc Dev Dis 2023; 10:439. [PMID: 37887886 PMCID: PMC10607269 DOI: 10.3390/jcdd10100439] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2023] [Revised: 10/17/2023] [Accepted: 10/19/2023] [Indexed: 10/28/2023] Open
Abstract
Cellular senescence (CS), classically considered a stable cell cycle withdrawal, is hallmarked by a progressive decrease in cell growth, differentiation, and biological activities. Senescent cells (SNCs) display a complicated senescence-associated secretory phenotype (SASP), encompassing a variety of pro-inflammatory factors that exert influence on the biology of both the cell and surrounding tissue. Among global mortality causes, cardiovascular diseases (CVDs) stand out, significantly impacting the living quality and functional abilities of patients. Recent data suggest the accumulation of SNCs in aged or diseased cardiovascular systems, suggesting their potential role in impairing cardiovascular function. CS operates as a double-edged sword: while it can stimulate the restoration of organs under physiological conditions, it can also participate in organ and tissue dysfunction and pave the way for multiple chronic diseases under pathological states. This review explores the mechanisms that underlie CS and delves into the distinctive features that characterize SNCs. Furthermore, we describe the involvement of SNCs in the progression of CVDs. Finally, the study provides a summary of emerging interventions that either promote or suppress senescence and discusses their therapeutic potential in CVDs.
Collapse
Affiliation(s)
- Dan Li
- Department of Cardiovascular Medicine, Lanzhou University Second Hospital, Lanzhou 730030, China; (D.L.); (H.D.); (Y.W.); (Y.X.)
| | - Yongnan Li
- Department of Cardiac Surgery, Lanzhou University Second Hospital, Lanzhou 730030, China;
| | - Hong Ding
- Department of Cardiovascular Medicine, Lanzhou University Second Hospital, Lanzhou 730030, China; (D.L.); (H.D.); (Y.W.); (Y.X.)
| | - Yuqin Wang
- Department of Cardiovascular Medicine, Lanzhou University Second Hospital, Lanzhou 730030, China; (D.L.); (H.D.); (Y.W.); (Y.X.)
| | - Yafei Xie
- Department of Cardiovascular Medicine, Lanzhou University Second Hospital, Lanzhou 730030, China; (D.L.); (H.D.); (Y.W.); (Y.X.)
| | - Xiaowei Zhang
- Department of Cardiovascular Medicine, Lanzhou University Second Hospital, Lanzhou 730030, China; (D.L.); (H.D.); (Y.W.); (Y.X.)
| |
Collapse
|
3
|
Suda M, Paul KH, Minamino T, Miller JD, Lerman A, Ellison-Hughes GM, Tchkonia T, Kirkland JL. Senescent Cells: A Therapeutic Target in Cardiovascular Diseases. Cells 2023; 12:1296. [PMID: 37174697 PMCID: PMC10177324 DOI: 10.3390/cells12091296] [Citation(s) in RCA: 46] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2023] [Revised: 04/27/2023] [Accepted: 04/29/2023] [Indexed: 05/15/2023] Open
Abstract
Senescent cell accumulation has been observed in age-associated diseases including cardiovascular diseases. Senescent cells lack proliferative capacity and secrete senescence-associated secretory phenotype (SASP) factors that may cause or worsen many cardiovascular diseases. Therapies targeting senescent cells, especially senolytic drugs that selectively induce senescent cell removal, have been shown to delay, prevent, alleviate, or treat multiple age-associated diseases in preclinical models. Some senolytic clinical trials have already been completed or are underway for a number of diseases and geriatric syndromes. Understanding how cellular senescence affects the various cell types in the cardiovascular system, such as endothelial cells, vascular smooth muscle cells, fibroblasts, immune cells, progenitor cells, and cardiomyocytes, is important to facilitate translation of senotherapeutics into clinical interventions. This review highlights: (1) the characteristics of senescent cells and their involvement in cardiovascular diseases, focusing on the aforementioned cardiovascular cell types, (2) evidence about senolytic drugs and other senotherapeutics, and (3) the future path and clinical potential of senotherapeutics for cardiovascular diseases.
Collapse
Affiliation(s)
- Masayoshi Suda
- Department of Physiology and Biomedical Engineering, Mayo Clinic, 200 First St., S.W., Rochester, MN 55905, USA
- Department of Cardiovascular Biology and Medicine, Juntendo University Graduate School of Medicine, 3-1-3 Hongo, Bunkyo-ku, Tokyo 113-8421, Japan
| | - Karl H. Paul
- Department of Physiology and Biomedical Engineering, Mayo Clinic, 200 First St., S.W., Rochester, MN 55905, USA
- Department of Physiology and Pharmacology, Karolinska Institutet, Solnavägen 9, 171 65 Solna, Sweden
| | - Tohru Minamino
- Department of Cardiovascular Biology and Medicine, Juntendo University Graduate School of Medicine, 3-1-3 Hongo, Bunkyo-ku, Tokyo 113-8421, Japan
- Japan Agency for Medical Research and Development-Core Research for Evolutionary Medical Science and Technology (AMED-CREST), Japan Agency for Medical Research and Development, Tokyo 100-0004, Japan
| | - Jordan D. Miller
- Division of Cardiovascular Surgery, Mayo Clinic College of Medicine, 200 First St., S.W., Rochester, MN 55905, USA
| | - Amir Lerman
- Department of Cardiovascular Medicine, Mayo Clinic, 200 First St., S.W., Rochester, MN 55905, USA
| | - Georgina M. Ellison-Hughes
- Centre for Human and Applied Physiological Sciences, School of Basic and Medical Biosciences, Faculty of Life Sciences & Medicine, Guy’s Campus, King’s College London, London SE1 1UL, UK
- Centre for Stem Cells and Regenerative Medicine, School of Basic and Medical Biosciences, Faculty of Life Sciences & Medicine, Guy’s Campus, King’s College London, London SE1 1UL, UK
| | - Tamar Tchkonia
- Department of Physiology and Biomedical Engineering, Mayo Clinic, 200 First St., S.W., Rochester, MN 55905, USA
| | - James L. Kirkland
- Department of Physiology and Biomedical Engineering, Mayo Clinic, 200 First St., S.W., Rochester, MN 55905, USA
- Division of General Internal Medicine, Department of Medicine, Mayo Clinic, 200 First St., S.W., Rochester, MN 55905, USA
| |
Collapse
|
4
|
Kaur G, Wang X, Li X, Ong H, He X, Cai C. Overexpression of GREM1 Improves the Survival Capacity of Aged Cardiac Mesenchymal Progenitor Cells via Upregulation of the ERK/NRF2-Associated Antioxidant Signal Pathway. Cells 2023; 12:1203. [PMID: 37190112 PMCID: PMC10136744 DOI: 10.3390/cells12081203] [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: 03/13/2023] [Revised: 04/11/2023] [Accepted: 04/17/2023] [Indexed: 05/17/2023] Open
Abstract
Ischemic heart disease is the leading cause of mortality in the United States. Progenitor cell therapy can restore myocardial structure and function. However, its efficacy is severely limited by cell aging and senescence. Gremlin-1 (GREM1), a member of the bone morphogenetic protein antagonist family, has been implicated in cell proliferation and survival. However, GREM1's role in cell aging and senescence has never been investigated in human cardiac mesenchymal progenitor cells (hMPCs). Therefore, this study assessed the hypothesis that overexpression of GREM1 rejuvenates the cardiac regenerative potential of aging hMPCs to a youthful stage and therefore allows better capacity for myocardial repair. We recently reported that a subpopulation of hMPCs with low mitochondrial membrane potential can be sorted from right atrial appendage-derived cells in patients with cardiomyopathy and exhibit cardiac reparative capacity in a mouse model of myocardial infarction. In this study, lentiviral particles were used to overexpress GREM1 in these hMPCs. Protein and mRNA expression were assessed through Western blot and RT-qPCR. FACS analysis for Annexin V/PI staining and lactate dehydrogenase assay were used to assess cell survival. It was observed that cell aging and cell senescence led to a decrease in GREM1 expression. In addition, overexpression of GREM1 led to a decrease in expression of senescence genes. Overexpression of GREM1 led to no significant change in cell proliferation. However, GREM1 appeared to have an anti-apoptotic effect, with an increase in survival and decrease in cytotoxicity evident in GREM1-overexpressing hMPCs. Overexpressing GREM1 also induced cytoprotective properties by decreasing reactive oxidative species and mitochondrial membrane potential. This result was associated with increased expression of antioxidant proteins, such as SOD1 and catalase, and activation of the ERK/NRF2 survival signal pathway. Inhibition of ERK led to a decrease in GREM1-mediated rejuvenation in terms of cell survival, which suggests that an ERK-dependent pathway may be involved. Taken altogether, these results indicate that overexpression of GREM1 can allow aging hMPCs to adopt a more robust phenotype with improved survival capacity, which is associated with an activated ERK/NRF2 antioxidant signal pathway.
Collapse
Affiliation(s)
- Gurleen Kaur
- Department of Molecular and Cellular Physiology, Department of Medicine, Albany Medical College, Albany, NY 12208, USA; (G.K.); (X.W.); (X.L.)
- Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Xiaoliang Wang
- Department of Molecular and Cellular Physiology, Department of Medicine, Albany Medical College, Albany, NY 12208, USA; (G.K.); (X.W.); (X.L.)
- Division of Surgical Sciences, Department of Surgery, University of Virginia, Charlottesville, VA 22903, USA
- Department of Surgery, Davis Heart and Lung Research Institute, The Ohio State University Wexner Medical Center, Columbus, OH 43210, USA; (H.O.); (X.H.)
| | - Xiuchun Li
- Department of Molecular and Cellular Physiology, Department of Medicine, Albany Medical College, Albany, NY 12208, USA; (G.K.); (X.W.); (X.L.)
- Department of Surgery, Davis Heart and Lung Research Institute, The Ohio State University Wexner Medical Center, Columbus, OH 43210, USA; (H.O.); (X.H.)
| | - Hannah Ong
- Department of Surgery, Davis Heart and Lung Research Institute, The Ohio State University Wexner Medical Center, Columbus, OH 43210, USA; (H.O.); (X.H.)
| | - Xiangfei He
- Department of Surgery, Davis Heart and Lung Research Institute, The Ohio State University Wexner Medical Center, Columbus, OH 43210, USA; (H.O.); (X.H.)
| | - Chuanxi Cai
- Department of Molecular and Cellular Physiology, Department of Medicine, Albany Medical College, Albany, NY 12208, USA; (G.K.); (X.W.); (X.L.)
- Division of Surgical Sciences, Department of Surgery, University of Virginia, Charlottesville, VA 22903, USA
- Department of Surgery, Davis Heart and Lung Research Institute, The Ohio State University Wexner Medical Center, Columbus, OH 43210, USA; (H.O.); (X.H.)
| |
Collapse
|
5
|
Meenakshi-Siddharthan DV, Livia C, Peterson TE, Stalboerger P, Attia ZI, Clavell AL, Friedman PA, Kapa S, Noseworthy PA, Schafer MJ, Stulak JM, Behfar A, Boilson BA. Artificial Intelligence-Derived Electrocardiogram Assessment of Cardiac Age and Molecular Markers of Senescence in Heart Failure. Mayo Clin Proc 2023; 98:372-385. [PMID: 36868745 DOI: 10.1016/j.mayocp.2022.10.026] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/06/2022] [Revised: 05/27/2022] [Accepted: 10/12/2022] [Indexed: 03/05/2023]
Abstract
OBJECTIVE To ascertain whether heart failure (HF) itself is a senescent phenomenon independent of age, and how this is reflected at a molecular level in the circulating progenitor cell niche, and at a substrate level using a novel electrocardiogram (ECG)-based artificial intelligence platform. PATIENTS AND METHODS Between October 14, 2016, and October 29, 2020, CD34+ progenitor cells were analyzed by flow cytometry and isolated by magnetic-activated cell sorting from patients of similar age with New York Heart Association functional classes IV (n = 17) and I-II (n = 10) heart failure with reduced ejection fraction and healthy controls (n = 10). CD34+ cellular senescence was quantitated by human telomerase reverse transcriptase expression and telomerase expression by quantitative polymerase chain reaction, and senescence-associated secretory phenotype (SASP) protein expression assayed in plasma. An ECG-based artificial intelligence (AI) algorithm was used to determine cardiac age and difference from chronological age (AI ECG age gap). RESULTS CD34+ counts and telomerase expression were significantly reduced and AI ECG age gap and SASP expression increased in all HF groups compared with healthy controls. Expression of SASP protein was closely associated with telomerase activity and severity of HF phenotype and inflammation. Telomerase activity was more closely associated with CD34+ cell counts and AI ECG age gap. CONCLUSION We conclude from this pilot study that HF may promote a senescent phenotype independent of chronological age. We show for the first time that the AI ECG in HF shows a phenotype of cardiac aging beyond chronological age, and appears to be associated with cellular and molecular evidence of senescence.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | - Marissa J Schafer
- Department of Physiology and Biomedical Engineering; Robert and Arlene Kogod Center on Aging
| | | | - Atta Behfar
- Van Cleve Cardiac Regeneration Program; Department of Cardiovascular Diseases
| | | |
Collapse
|
6
|
Desmin deficiency affects the microenvironment of the cardiac side population and Sca1+ stem cell population of the adult heart and impairs their cardiomyogenic commitment. Cell Tissue Res 2022; 389:309-326. [DOI: 10.1007/s00441-022-03643-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2022] [Accepted: 05/20/2022] [Indexed: 11/02/2022]
|
7
|
Romano V, Belviso I, Sacco AM, Cozzolino D, Nurzynska D, Amarelli C, Maiello C, Sirico F, Di Meglio F, Castaldo C. Human Cardiac Progenitor Cell-Derived Extracellular Vesicles Exhibit Promising Potential for Supporting Cardiac Repair in Vitro. Front Physiol 2022; 13:879046. [PMID: 35669580 PMCID: PMC9163838 DOI: 10.3389/fphys.2022.879046] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2022] [Accepted: 05/02/2022] [Indexed: 11/13/2022] Open
Abstract
Although human Cardiac Progenitor Cells (hCPCs) are not retained by host myocardium they still improve cardiac function when injected into ischemic heart. Emerging evidence supports the hypothesis that hCPC beneficial effects are induced by paracrine action on resident cells. Extracellular vesicles (EVs) are an intriguing mechanism of cell communication based on the transport and transfer of peptides, lipids, and nucleic acids that have the potential to modulate signaling pathways, cell growth, migration, and proliferation of recipient cells. We hypothesize that EVs are involved in the paracrine effects elicited by hCPCs and held accountable for the response of the infarcted myocardium to hCPC-based cell therapy. To test this theory, we collected EVs released by hCPCs isolated from healthy myocardium and evaluated the effects they elicited when administered to resident hCPC and cardiac fibroblasts (CFs) isolated from patients with post-ischemic end-stage heart failure. Evidence emerging from our study indicated that hCPC-derived EVs impacted upon proliferation and survival of hCPCs residing in the ischemic heart and regulated the synthesis and deposition of extracellular-matrix by CFs. These findings suggest that beneficial effects exerted by hCPC injection are, at least to some extent, ascribable to the delivery of signals conveyed by EVs.
Collapse
Affiliation(s)
- Veronica Romano
- Department of Public Health, University of Naples Federico II, Naples, Italy
| | - Immacolata Belviso
- Department of Public Health, University of Naples Federico II, Naples, Italy
| | - Anna Maria Sacco
- Department of Public Health, University of Naples Federico II, Naples, Italy
| | - Domenico Cozzolino
- Department of Public Health, University of Naples Federico II, Naples, Italy
| | - Daria Nurzynska
- Department of Medicine, Surgery and Dentistry “Scuola Medica Salernitana”/DIPMED, University of Salerno, Baronissi, Italy
| | - Cristiano Amarelli
- Department of Cardiovascular Surgery and Transplant, Monaldi Hospital, Naples, Italy
| | - Ciro Maiello
- Department of Cardiovascular Surgery and Transplant, Monaldi Hospital, Naples, Italy
| | - Felice Sirico
- Department of Public Health, University of Naples Federico II, Naples, Italy
| | - Franca Di Meglio
- Department of Public Health, University of Naples Federico II, Naples, Italy
| | - Clotilde Castaldo
- Department of Public Health, University of Naples Federico II, Naples, Italy
| |
Collapse
|
8
|
Budhathoki S, Graham C, Sethu P, Kannappan R. Engineered Aging Cardiac Tissue Chip Model for Studying Cardiovascular Disease. Cells Tissues Organs 2022; 211:348-359. [PMID: 34365455 PMCID: PMC8818062 DOI: 10.1159/000516954] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2021] [Accepted: 04/29/2021] [Indexed: 01/03/2023] Open
Abstract
Due to the rapidly growing number of older people worldwide and the concomitant increase in cardiovascular complications, there is an urgent need for age-related cardiac disease modeling and drug screening platforms. In the present study, we developed a cardiac tissue chip model that incorporates hemodynamic loading and mimics essential aspects of the infarcted aging heart. We induced cellular senescence in H9c2 myoblasts using low-dose doxorubicin treatment. These senescent cells were then used to engineer cardiac tissue fibers, which were subjected to hemodynamic stresses associated with pressure-volume changes in the heart. Myocardial ischemia was modeled in the engineered cardiac tissue via hypoxic treatment. Our results clearly show that acute low-dose doxorubicin treatment-induced senescence, as evidenced by morphological and molecular markers, including enlarged and flattened nuclei, DNA damage response foci, and increased expression of cell cycle inhibitor p16INK4a, p53, and ROS. Under normal hemodynamic load, the engineered cardiac tissues demonstrated cell alignment and retained cardiac cell characteristics. Our senescent cardiac tissue model of hypoxia-induced myocardial infarction recapitulated the pathological disease hallmarks such as increased cell death and upregulated expression of ANP and BNP. In conclusion, the described methodology provides a novel approach to generate stress-induced aging cardiac cell phenotypes and engineer cardiac tissue chip models to study the cardiovascular disease pathologies associated with aging.
Collapse
Affiliation(s)
- Sachin Budhathoki
- Division of Cardiovascular Disease, Departments of Medicine, The University of Alabama at Birmingham, Birmingham, AL, USA
| | - Caleb Graham
- Division of Cardiovascular Disease, Departments of Medicine, The University of Alabama at Birmingham, Birmingham, AL, USA
| | - Palaniappan Sethu
- Division of Cardiovascular Disease, Departments of Medicine, The University of Alabama at Birmingham, Birmingham, AL, USA
| | - Ramaswamy Kannappan
- Division of Cardiovascular Disease, Departments of Medicine, The University of Alabama at Birmingham, Birmingham, AL, USA
| |
Collapse
|
9
|
Opportunities and Challenges in Stem Cell Aging. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2021; 1341:143-175. [PMID: 33748933 DOI: 10.1007/5584_2021_624] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Studying aging, as a physiological process that can cause various pathological phenotypes, has attracted lots of attention due to its increasing burden and prevalence. Therefore, understanding its mechanism to find novel therapeutic alternatives for age-related disorders such as neurodegenerative and cardiovascular diseases is essential. Stem cell senescence plays an important role in aging. In the context of the underlying pathways, mitochondrial dysfunction, epigenetic and genetic alterations, and other mechanisms have been studied and as a consequence, several rejuvenation strategies targeting these mechanisms like pharmaceutical interventions, genetic modification, and cellular reprogramming have been proposed. On the other hand, since stem cells have great potential for disease modeling, they have been useful for representing aging and its associated disorders. Accordingly, the main mechanisms of senescence in stem cells and promising ways of rejuvenation, along with some examples of stem cell models for aging are introduced and discussed. This review aims to prepare a comprehensive summary of the findings by focusing on the most recent ones to shine a light on this area of research.
Collapse
|
10
|
Villarreal-Leal RA, Cooke JP, Corradetti B. Biomimetic and immunomodulatory therapeutics as an alternative to natural exosomes for vascular and cardiac applications. NANOMEDICINE : NANOTECHNOLOGY, BIOLOGY, AND MEDICINE 2021; 35:102385. [PMID: 33774130 PMCID: PMC8238887 DOI: 10.1016/j.nano.2021.102385] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/04/2020] [Revised: 02/21/2021] [Accepted: 03/03/2021] [Indexed: 02/07/2023]
Abstract
Inflammation is a central mechanism in cardiovascular diseases (CVD), where sustained oxidative stress and immune responses contribute to cardiac remodeling and impairment. Exosomes are extracellular vesicles released by cells to communicate with their surroundings and to modulate the tissue microenvironment. Recent evidence indicates their potential as cell-free immunomodulatory therapeutics for CVD, preventing cell death and fibrosis while inducing wound healing and angiogenesis. Biomimetic exosomes are semi-synthetic particles engineered using essential moieties present in natural exosomes (lipids, RNA, proteins) to reproduce their therapeutic effects while improving on scalability and standardization due to the ample range of moieties available to produce them. In this review, we provide an up-to-date description of the use of exosomes for CVD and offer our vision on the areas of opportunity for the development of biomimetic strategies. We also discuss the current limitations to overcome in the process towards their translation into clinic.
Collapse
Affiliation(s)
- Ramiro A Villarreal-Leal
- Tecnologico de Monterrey, Escuela de Medicina y Ciencias de la Salud, Monterrey, Nuevo León, Mexico
| | - John P Cooke
- RNA Therapeutics Program, Department of Cardiovascular Sciences (R.S., J.P.C.), Houston Methodist Research Institute, TX, USA; Houston Methodist DeBakey Heart and Vascular Center (J.P.C.), Houston Methodist Hospital, TX, USA
| | - Bruna Corradetti
- Department of Nanomedicine, Houston Methodist Research Institute, Houston, TX, USA; Center of NanoHealth, Swansea University Medical School, Swansea, UK.
| |
Collapse
|
11
|
Hasan AS, Luo L, Baba S, Li TS. Estrogen is required for maintaining the quality of cardiac stem cells. PLoS One 2021; 16:e0245166. [PMID: 33481861 PMCID: PMC7822545 DOI: 10.1371/journal.pone.0245166] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2020] [Accepted: 12/22/2020] [Indexed: 12/24/2022] Open
Abstract
Compared to the age-matched men, the incidence of cardiovascular diseases is lower in premenopausal but higher in postmenopausal women, suggesting the cardio-protective role of estrogen in females. Although cardiac stem cells (CSCs) express estrogen receptors, yet the effects of estrogen on CSCs remain unclear. In this study, we investigated the potential role of estrogen in maintaining the quality of CSCs by in vivo and in vitro experiments. For the in vivo study, estrogen deficiency was induced by ovariectomy in 6-weeks-old C57BL/6 female mice, and then randomly given 17β-estradiol (E2) replacements at a low dose (0.01 mg/60 days) and high dose (0.18 mg/60 days), or vehicle treatment. All mice were killed 2 months after treatments, and heart tissues were collected for ex vivo expansion of CSCs. Compared to age-matched healthy controls, estrogen deficiency slightly decreased the yield of CSCs with significantly lower telomerase activity and more DNA damage. Interestingly, E2 replacements at low and high doses significantly increased the yield of CSCs and reversed the quality impairment of CSCs following estrogen deficiency. For the in vitro study, twice-passaged CSCs from the hearts of adult healthy female mice were cultured with the supplement of 0.01, 0.1, and 1 μM E2 in the medium for 3 days. We found that E2 supplement increased c-kit expression, increased proliferative activity, improved telomerase activity, and reduced DNA damage of CSCs in a dose-dependent manner. Our data suggested the potential role of estrogen in maintaining the quality of CSCs, providing new insight into the cardio-protective effects of estrogen.
Collapse
Affiliation(s)
- Al Shaimaa Hasan
- Department of Stem Cell Biology, Atomic Bomb Disease Institute, Nagasaki University, Nagasaki, Japan
- Department of Medical Pharmacology, Qena Faculty of Medicine, South Valley University, Qena, Egypt
| | - Lan Luo
- Department of Stem Cell Biology, Atomic Bomb Disease Institute, Nagasaki University, Nagasaki, Japan
- School of Medical Technology, Xuzhou Medical University, Xuzhou City, Jiangsu Province, China
| | - Satoko Baba
- Department of Stem Cell Biology, Atomic Bomb Disease Institute, Nagasaki University, Nagasaki, Japan
| | - Tao-Sheng Li
- Department of Stem Cell Biology, Atomic Bomb Disease Institute, Nagasaki University, Nagasaki, Japan
- * E-mail:
| |
Collapse
|
12
|
Caponnetto F, Manini I, Bulfoni M, Zingaretti N, Miotti G, Di Loreto C, Cesselli D, Mariuzzi L, Parodi PC. Human Adipose-Derived Stem Cells in Madelung's Disease: Morphological and Functional Characterization. Cells 2020; 10:cells10010044. [PMID: 33396896 PMCID: PMC7824042 DOI: 10.3390/cells10010044] [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: 12/01/2020] [Revised: 12/21/2020] [Accepted: 12/28/2020] [Indexed: 12/17/2022] Open
Abstract
Madelung Disease (MD) is a syndrome characterized by the accumulation of aberrant symmetric adipose tissue deposits. The etiology of this disease is yet to be elucidated, even though the presence of comorbidities, either genetic or environmental, has been reported. For this reason, establishing an in vitro model for MD is considered crucial to get insights into its physiopathology. We previously established a protocol for isolation and culture of stem cells from diseased tissues. Therefore, we isolated human adipose-derived stem cells (ASC) from MD patients and compared these cells with those isolated from healthy subjects in terms of surface phenotype, growth kinetic, adipogenic differentiation potential, and molecular alterations. Moreover, we evaluated the ability of the MD-ASC secretome to affect healthy ASC. The results reported a difference in the growth kinetic and surface markers of MD-ASC compared to healthy ASC but not in adipogenic differentiation. The most commonly described mitochondrial mutations were not observed. Still, MD-ASC secretome was able to shift the healthy ASC phenotype to an MD phenotype. This work provides evidence of the possibility of exploiting a patient-based in vitro model for better understanding MD pathophysiology, possibly favoring the development of novel target therapies.
Collapse
Affiliation(s)
- Federica Caponnetto
- Department of Medicine, University of Udine, 33100 Udine, Italy; (M.B.); (C.D.L.); (D.C.); (L.M.); (P.C.P.)
- Correspondence: ; Tel.: +39-04-3255-9412
| | - Ivana Manini
- Institute of Pathology, University Hospital of Udine, 33100 Udine, Italy;
| | - Michela Bulfoni
- Department of Medicine, University of Udine, 33100 Udine, Italy; (M.B.); (C.D.L.); (D.C.); (L.M.); (P.C.P.)
| | - Nicola Zingaretti
- Department of Medical Area (DAME), Clinic of Plastic and Reconstructive Surgery, Academic Hospital of Udine, University of Udine, 33100 Udine, Italy; (N.Z.); (G.M.)
| | - Giovanni Miotti
- Department of Medical Area (DAME), Clinic of Plastic and Reconstructive Surgery, Academic Hospital of Udine, University of Udine, 33100 Udine, Italy; (N.Z.); (G.M.)
| | - Carla Di Loreto
- Department of Medicine, University of Udine, 33100 Udine, Italy; (M.B.); (C.D.L.); (D.C.); (L.M.); (P.C.P.)
- Institute of Pathology, University Hospital of Udine, 33100 Udine, Italy;
| | - Daniela Cesselli
- Department of Medicine, University of Udine, 33100 Udine, Italy; (M.B.); (C.D.L.); (D.C.); (L.M.); (P.C.P.)
- Institute of Pathology, University Hospital of Udine, 33100 Udine, Italy;
| | - Laura Mariuzzi
- Department of Medicine, University of Udine, 33100 Udine, Italy; (M.B.); (C.D.L.); (D.C.); (L.M.); (P.C.P.)
- Institute of Pathology, University Hospital of Udine, 33100 Udine, Italy;
| | - Pier Camillo Parodi
- Department of Medicine, University of Udine, 33100 Udine, Italy; (M.B.); (C.D.L.); (D.C.); (L.M.); (P.C.P.)
- Department of Medical Area (DAME), Clinic of Plastic and Reconstructive Surgery, Academic Hospital of Udine, University of Udine, 33100 Udine, Italy; (N.Z.); (G.M.)
| |
Collapse
|
13
|
Saheera S, Potnuri AG, Krishnamurthy P. Nano-Vesicle (Mis)Communication in Senescence-Related Pathologies. Cells 2020; 9:E1974. [PMID: 32859053 PMCID: PMC7564330 DOI: 10.3390/cells9091974] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2020] [Revised: 08/23/2020] [Accepted: 08/25/2020] [Indexed: 12/11/2022] Open
Abstract
Extracellular vesicles are a heterogeneous group of cell-derived membranous structures comprising of exosomes, apoptotic bodies, and microvesicles. Of the extracellular vesicles, exosomes are the most widely sorted and extensively explored for their contents and function. The size of the nanovesicular structures (exosomes) range from 30 to 140 nm and are present in various biological fluids such as saliva, plasma, urine etc. These cargo-laden extracellular vesicles arise from endosome-derived multivesicular bodies and are known to carry proteins and nucleic acids. Exosomes are involved in multiple physiological and pathological processes, including cellular senescence. Exosomes mediate signaling crosstalk and play a critical role in cell-cell communications. Exosomes have evolved as potential biomarkers for aging-related diseases. Aging, a physiological process, involves a progressive decline of function of organs with a loss of homeostasis and increasing probability of illness and death. The review focuses on the classic view of exosome biogenesis, biology, and age-associated changes. Owing to their ability to transport biological information among cells, the review also discusses the interplay of senescent cell-derived exosomes with the aging process, including the susceptibility of the aging population to COVID-19 infections.
Collapse
Affiliation(s)
- Sherin Saheera
- Department of Cardiovascular Medicine, University of Massachusetts Medical School, Worcester, MA 01605, USA;
| | - Ajay Godwin Potnuri
- Department of Animal Physiology, Indian Council for Medical Research—National Animal Resource Facility for Biomedical Research, Genome Valley, Shamirpet, Hyderabad, Telangana 500078, India;
| | - Prasanna Krishnamurthy
- Department of Biomedical Engineering, School of Medicine and School of Engineering, The University of Alabama at Birmingham, 1675 University Blvd, Volker Hall G094, Birmingham, AL 35294, USA
| |
Collapse
|
14
|
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: 80] [Impact Index Per Article: 16.0] [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
|
15
|
Sameri S, Samadi P, Dehghan R, Salem E, Fayazi N, Amini R. Stem Cell Aging in Lifespan and Disease: A State-of-the-Art Review. Curr Stem Cell Res Ther 2020; 15:362-378. [DOI: 10.2174/1574888x15666200213105155] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2019] [Revised: 11/09/2019] [Accepted: 12/31/2019] [Indexed: 12/11/2022]
Abstract
Aging is considered as inevitable changes at different levels of genome, cell, and organism.
From the accumulation of DNA damages to imperfect protein homeostasis, altered cellular communication
and exhaustion of stem cells, aging is a major risk factor for many prevalent diseases, such as
cancer, cardiovascular disease, pulmonary disease, diabetes, and neurological disorders. The cells are
dynamic systems, which, through a cycle of processes such as replication, growth, and death, could
replenish the bodies’ organs and tissues, keeping an entire organism in optimal working order. In many
different tissues, adult stem cells are behind these processes, replenishing dying cells to maintain normal
tissue function and regenerating injured tissues. Therefore, adult stem cells play a vital role in preventing
the aging of organs and tissues, and can delay aging. However, during aging, these cells also
undergo some detrimental changes such as alterations in the microenvironment, a decline in the regenerative
capacity, and loss of function. This review aimed to discuss age-related changes of stem cells in
different tissues and cells, including skin, muscles, brain, heart, hair follicles, liver, and lung.
Collapse
Affiliation(s)
- Saba Sameri
- Research Center for Molecular Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Pouria Samadi
- Research Center for Molecular Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Razieh Dehghan
- Research Center for Molecular Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Elham Salem
- Research Center for Molecular Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Nashmin Fayazi
- Research Center for Molecular Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Razieh Amini
- Research Center for Molecular Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
| |
Collapse
|
16
|
Saheera S, Krishnamurthy P. Cardiovascular Changes Associated with Hypertensive Heart Disease and Aging. Cell Transplant 2020; 29:963689720920830. [PMID: 32393064 PMCID: PMC7586256 DOI: 10.1177/0963689720920830] [Citation(s) in RCA: 75] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Cardiovascular diseases are the leading cause of mortality and morbidity worldwide and account for more than 17.9 million deaths (World Health Organization report). Hypertension and aging are two major risk factors for the development of cardiac structural and functional abnormalities. Hypertension, or elevated blood pressure, if left untreated can result in myocardial hypertrophy leading to heart failure (HF). Left ventricular hypertrophy consequent to pressure overload is recognized as the most important predictor of congestive HF and sudden death. The pathological changes occurring during hypertensive heart disease are very complex and involve many cellular and molecular alterations. In contrast, the cardiac changes that occur with aging are a slow but life-long process and involve all of the structural components in the heart and vasculature. However, these structural changes in the cardiovascular system lead to alterations in overall cardiac physiology and function. The pace at which these pathophysiological changes occur varies between individuals owing to many genetic and environmental risk factors. This review highlights the molecular mechanisms of cardiac structural and functional alterations associated with hypertension and aging.
Collapse
Affiliation(s)
- Sherin Saheera
- Department of Cardiovascular Medicine, University of Massachusetts Medical School, Worcester, USA
| | - Prasanna Krishnamurthy
- Department of Biomedical Engineering, School of Medicine and School of Engineering, The University of Alabama at Birmingham, USA
| |
Collapse
|
17
|
Atkinson SP. A Preview of Selected Articles. Stem Cells Transl Med 2020; 9:543-546. [PMID: 32329241 PMCID: PMC7180293 DOI: 10.1002/sctm.20-0135] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2020] [Accepted: 03/27/2020] [Indexed: 11/11/2022] Open
|
18
|
Park JH, Lee NK, Lim HJ, Ji ST, Kim YJ, Jang WB, Kim DY, Kang S, Yun J, Ha JS, Kim H, Lee D, Baek SH, Kwon SM. Pharmacological inhibition of mTOR attenuates replicative cell senescence and improves cellular function via regulating the STAT3-PIM1 axis in human cardiac progenitor cells. Exp Mol Med 2020; 52:615-628. [PMID: 32273566 PMCID: PMC7210934 DOI: 10.1038/s12276-020-0374-4] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2019] [Revised: 09/08/2019] [Accepted: 10/29/2019] [Indexed: 12/19/2022] Open
Abstract
The mammalian target of rapamycin (mTOR) signaling pathway efficiently regulates the energy state of cells and maintains tissue homeostasis. Dysregulation of the mTOR pathway has been implicated in several human diseases. Rapamycin is a specific inhibitor of mTOR and pharmacological inhibition of mTOR with rapamycin promote cardiac cell generation from the differentiation of mouse and human embryonic stem cells. These studies strongly implicate a role of sustained mTOR activity in the differentiating functions of embryonic stem cells; however, they do not directly address the required effect for sustained mTOR activity in human cardiac progenitor cells. In the present study, we evaluated the effect of mTOR inhibition by rapamycin on the cellular function of human cardiac progenitor cells and discovered that treatment with rapamycin markedly attenuated replicative cell senescence in human cardiac progenitor cells (hCPCs) and promoted their cellular functions. Furthermore, rapamycin not only inhibited mTOR signaling but also influenced signaling pathways, including STAT3 and PIM1, in hCPCs. Therefore, these data reveal a crucial function for rapamycin in senescent hCPCs and provide clinical strategies based on chronic mTOR activity.
Collapse
Affiliation(s)
- Ji Hye Park
- Laboratory of Regenerative Medicine and Stem Cell Biology, Department of Physiology, Medical Research Institute, School of Medicine, Pusan National University, Yangsan, 50612, Republic of Korea
- Research Institute of Convergence Biomedical Science and Technology, Pusan National University School of Medicine, Yangsan, 50612, Republic of Korea
- R&D Center for Advanced Pharmaceuticals & Evaluation, Korea Institute of Toxicology, Korea Research Institute of Chemical Technology, 141 Gajeong-ro, Yuseong-gu, Daejeon, 34114, South Korea
| | - Na Kyoung Lee
- Laboratory of Regenerative Medicine and Stem Cell Biology, Department of Physiology, Medical Research Institute, School of Medicine, Pusan National University, Yangsan, 50612, Republic of Korea
- Research Institute of Convergence Biomedical Science and Technology, Pusan National University School of Medicine, Yangsan, 50612, Republic of Korea
| | - Hye Ji Lim
- Laboratory of Regenerative Medicine and Stem Cell Biology, Department of Physiology, Medical Research Institute, School of Medicine, Pusan National University, Yangsan, 50612, Republic of Korea
- Research Institute of Convergence Biomedical Science and Technology, Pusan National University School of Medicine, Yangsan, 50612, Republic of Korea
| | - Seung Taek Ji
- Laboratory of Regenerative Medicine and Stem Cell Biology, Department of Physiology, Medical Research Institute, School of Medicine, Pusan National University, Yangsan, 50612, Republic of Korea
- Research Institute of Convergence Biomedical Science and Technology, Pusan National University School of Medicine, Yangsan, 50612, Republic of Korea
| | - Yeon-Ju Kim
- Laboratory of Regenerative Medicine and Stem Cell Biology, Department of Physiology, Medical Research Institute, School of Medicine, Pusan National University, Yangsan, 50612, Republic of Korea
- Research Institute of Convergence Biomedical Science and Technology, Pusan National University School of Medicine, Yangsan, 50612, Republic of Korea
| | - Woong Bi Jang
- Laboratory of Regenerative Medicine and Stem Cell Biology, Department of Physiology, Medical Research Institute, School of Medicine, Pusan National University, Yangsan, 50612, Republic of Korea
- Research Institute of Convergence Biomedical Science and Technology, Pusan National University School of Medicine, Yangsan, 50612, Republic of Korea
| | - Da Yeon Kim
- Laboratory of Regenerative Medicine and Stem Cell Biology, Department of Physiology, Medical Research Institute, School of Medicine, Pusan National University, Yangsan, 50612, Republic of Korea
- Research Institute of Convergence Biomedical Science and Technology, Pusan National University School of Medicine, Yangsan, 50612, Republic of Korea
| | - Songhwa Kang
- Laboratory of Regenerative Medicine and Stem Cell Biology, Department of Physiology, Medical Research Institute, School of Medicine, Pusan National University, Yangsan, 50612, Republic of Korea
- Research Institute of Convergence Biomedical Science and Technology, Pusan National University School of Medicine, Yangsan, 50612, Republic of Korea
| | - Jisoo Yun
- Laboratory of Regenerative Medicine and Stem Cell Biology, Department of Physiology, Medical Research Institute, School of Medicine, Pusan National University, Yangsan, 50612, Republic of Korea
- Research Institute of Convergence Biomedical Science and Technology, Pusan National University School of Medicine, Yangsan, 50612, Republic of Korea
| | - Jong Seong Ha
- Laboratory of Regenerative Medicine and Stem Cell Biology, Department of Physiology, Medical Research Institute, School of Medicine, Pusan National University, Yangsan, 50612, Republic of Korea
- Research Institute of Convergence Biomedical Science and Technology, Pusan National University School of Medicine, Yangsan, 50612, Republic of Korea
| | - Hyungtae Kim
- Department of Thoracic and Cardiovascular Surgery, Pusan National University Yangsan Hospital, School of Medicine, Pusan National University, Yangsan, 50612, Republic of Korea
| | - Dongjun Lee
- Department of Convergence Medical Science, Pusan National University School of Medicine, Yangsan, 50612, Republic of Korea.
| | - Sang Hong Baek
- Laboratory of Cardiovascular Disease, Division of Cardiology, School of Medicine, The Catholic University of Korea, Seoul, 137-040, Republic of Korea.
| | - Sang-Mo Kwon
- Laboratory of Regenerative Medicine and Stem Cell Biology, Department of Physiology, Medical Research Institute, School of Medicine, Pusan National University, Yangsan, 50612, Republic of Korea.
- Department of Thoracic and Cardiovascular Surgery, Pusan National University Yangsan Hospital, School of Medicine, Pusan National University, Yangsan, 50612, Republic of Korea.
| |
Collapse
|
19
|
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
|
20
|
Ng WH, Yong YK, Ramasamy R, Ngalim SH, Lim V, Shaharuddin B, Tan JJ. Human Wharton's Jelly-Derived Mesenchymal Stem Cells Minimally Improve the Growth Kinetics and Cardiomyocyte Differentiation of Aged Murine Cardiac c-kit Cells in In Vitro without Rejuvenating Effect. Int J Mol Sci 2019; 20:ijms20225519. [PMID: 31698679 PMCID: PMC6887783 DOI: 10.3390/ijms20225519] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2019] [Revised: 09/30/2019] [Accepted: 09/30/2019] [Indexed: 01/09/2023] Open
Abstract
Cardiac c-kit cells show promise in regenerating an injured heart. While heart disease commonly affects elderly patients, it is unclear if autologous cardiac c-kit cells are functionally competent and applicable to these patients. This study characterised cardiac c-kit cells (CCs) from aged mice and studied the effects of human Wharton’s Jelly-derived mesenchymal stem cells (MSCs) on the growth kinetics and cardiac differentiation of aged CCs in vitro. CCs were isolated from 4-week- and 18-month-old C57/BL6N mice and were directly co-cultured with MSCs or separated by transwell insert. Clonogenically expanded aged CCs showed comparable telomere length to young CCs. However, these cells showed lower Gata4, Nkx2.5, and Sox2 gene expressions, with changes of 2.4, 3767.0, and 4.9 folds, respectively. Direct co-culture of both cells increased aged CC migration, which repopulated 54.6 ± 4.4% of the gap area as compared to aged CCs with MSCs in transwell (42.9 ± 2.6%) and CCs without MSCs (44.7 ± 2.5%). Both direct and transwell co-culture improved proliferation in aged CCs by 15.0% and 16.4%, respectively, as traced using carboxyfluorescein succinimidyl ester (CFSE) for three days. These data suggest that MSCs can improve the growth kinetics of aged CCs. CCs retaining intact telomere are present in old hearts and could be obtained based on their self-renewing capability. Although these aged CCs with reduced growth kinetics are improved by MSCs via cell–cell contact, the effect is minimal.
Collapse
Affiliation(s)
- Wai Hoe Ng
- Advanced Medical and Dental Institute, Universiti Sains Malaysia, Bertam 13200, Kepala Batas, Penang, Malaysia; (W.H.N.); (S.H.N.); (V.L.); (B.S.)
| | - Yoke Keong Yong
- Department of Human Anatomy, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, Serdang 43400, Selangor Darul Ehsan, Malaysia;
| | - Rajesh Ramasamy
- Department of Pathology, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, Serdang 43400, Selangor Darul Ehsan, Malaysia;
| | - Siti Hawa Ngalim
- Advanced Medical and Dental Institute, Universiti Sains Malaysia, Bertam 13200, Kepala Batas, Penang, Malaysia; (W.H.N.); (S.H.N.); (V.L.); (B.S.)
| | - Vuanghao Lim
- Advanced Medical and Dental Institute, Universiti Sains Malaysia, Bertam 13200, Kepala Batas, Penang, Malaysia; (W.H.N.); (S.H.N.); (V.L.); (B.S.)
| | - Bakiah Shaharuddin
- Advanced Medical and Dental Institute, Universiti Sains Malaysia, Bertam 13200, Kepala Batas, Penang, Malaysia; (W.H.N.); (S.H.N.); (V.L.); (B.S.)
| | - Jun Jie Tan
- Advanced Medical and Dental Institute, Universiti Sains Malaysia, Bertam 13200, Kepala Batas, Penang, Malaysia; (W.H.N.); (S.H.N.); (V.L.); (B.S.)
- Correspondence: ; Tel.: +045622422
| |
Collapse
|
21
|
Abstract
Telomeres, the protective ends of linear chromosomes, shorten throughout an individual's lifetime. Accumulation of critically short telomeres is proposed to be a primary molecular cause of aging and age-associated diseases. Mutations in telomere maintenance genes are associated with pathologies referred to as or telomeropathies. The rate of telomere shortening throughout life is determined by endogenous (genetic) and external (nongenetic) factors. Therapeutic strategies based on telomerase activation are being developed to treat and prevent telomere-associated diseases, namely aging-related diseases and telomeropathies. Here, we review the molecular mechanisms underlying telomere driven diseases with particular emphasis on cardiovascular diseases.
Collapse
Affiliation(s)
- Paula Martínez
- From the Telomeres and Telomerase Group, Molecular Oncology Program, Spanish National Cancer Centre (CNIO), Madrid, Spain
| | - Maria A Blasco
- From the Telomeres and Telomerase Group, Molecular Oncology Program, Spanish National Cancer Centre (CNIO), Madrid, Spain
| |
Collapse
|
22
|
Abstract
Cardiac ageing manifests as a decline in function leading to heart failure. At the cellular level, ageing entails decreased replicative capacity and dysregulation of cellular processes in myocardial and nonmyocyte cells. Various extrinsic parameters, such as lifestyle and environment, integrate important signalling pathways, such as those involving inflammation and oxidative stress, with intrinsic molecular mechanisms underlying resistance versus progression to cellular senescence. Mitigation of cardiac functional decline in an ageing organism requires the activation of enhanced maintenance and reparative capacity, thereby overcoming inherent endogenous limitations to retaining a youthful phenotype. Deciphering the molecular mechanisms underlying dysregulation of cellular function and renewal reveals potential interventional targets to attenuate degenerative processes at the cellular and systemic levels to improve quality of life for our ageing population. In this Review, we discuss the roles of extrinsic and intrinsic factors in cardiac ageing. Animal models of cardiac ageing are summarized, followed by an overview of the current and possible future treatments to mitigate the deleterious effects of cardiac ageing.
Collapse
|
23
|
Enhanced cardiac repair by telomerase reverse transcriptase over-expression in human cardiac mesenchymal stromal cells. Sci Rep 2019; 9:10579. [PMID: 31332256 PMCID: PMC6646304 DOI: 10.1038/s41598-019-47022-w] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2019] [Accepted: 07/08/2019] [Indexed: 12/11/2022] Open
Abstract
We have previously reported a subpopulation of mesenchymal stromal cells (MSCs) within the platelet-derived growth factor receptor-alpha (PDGFRα)/CD90 co-expressing cardiac interstitial and adventitial cell fraction. Here we further characterise PDGFRα/CD90-expressing cardiac MSCs (PDGFRα + cMSCs) and use human telomerase reverse transcriptase (hTERT) over-expression to increase cMSCs ability to repair the heart after induced myocardial infarction. hTERT over-expression in PDGFRα + cardiac MSCs (hTERT + PDGFRα + cMSCs) modulates cell differentiation, proliferation, survival and angiogenesis related genes. In vivo, transplantation of hTERT + PDGFRα + cMSCs in athymic rats significantly increased left ventricular function, reduced scar size, increased angiogenesis and proliferation of both cardiomyocyte and non-myocyte cell fractions four weeks after myocardial infarction. In contrast, transplantation of mutant hTERT + PDGFRα + cMSCs (which generate catalytically-inactive telomerase) failed to replicate this cardiac functional improvement, indicating a telomerase-dependent mechanism. There was no hTERT + PDGFRα + cMSCs engraftment 14 days after transplantation indicating functional improvement occurred by paracrine mechanisms. Mass spectrometry on hTERT + PDGFRα + cMSCs conditioned media showed increased proteins associated with matrix modulation, angiogenesis, cell proliferation/survival/adhesion and innate immunity function. Our study shows that hTERT can activate pro-regenerative signalling within PDGFRα + cMSCs and enhance cardiac repair after myocardial infarction. An increased understanding of hTERT’s role in mesenchymal stromal cells from various organs will favourably impact clinical regenerative and anti-cancer therapies.
Collapse
|
24
|
Aquila G, Kostina A, Vieceli Dalla Sega F, Shlyakhto E, Kostareva A, Marracino L, Ferrari R, Rizzo P, Malaschicheva A. The Notch pathway: a novel therapeutic target for cardiovascular diseases? Expert Opin Ther Targets 2019; 23:695-710. [PMID: 31304807 DOI: 10.1080/14728222.2019.1641198] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Introduction: The Notch pathway is involved in determining cell fate during development and postnatally in continuously renewing tissues, such as the endothelium, the epithelium, and in the stem cells pool. The dysregulation of the Notch pathway is one of the causes of limited response, or resistance, to available cancer treatments and novel therapeutic strategies based on Notch inhibition are being investigated in preclinical and clinical studies in oncology. A large body of evidence now shows that the dysregulation of the Notch pathway is also involved in the pathophysiology of cardiovascular diseases (CVDs). Areas covered: This review discusses the molecular mechanisms involving Notch which underlie heart failure, aortic valve calcification, and aortic aneurysm. Expert opinion: Despite the existence of preventive, pharmacological and surgical interventions approaches, CVDs are the first causes of mortality worldwide. The Notch pathway is becoming increasingly recognized as being involved in heart failure, aortic aneurysm and aortic valve calcification, which are among the most common global causes of mortality due to CVDs. As already shown in cancer, the dissection of the biological processes and molecular mechanisms involving Notch should pave the way for new strategies to prevent and cure these diseases.
Collapse
Affiliation(s)
- Giorgio Aquila
- Department of Medical Sciences, University of Ferrara , Ferrara , Italy
| | - Aleksandra Kostina
- Laboratory of Molecular Cardiology, Almazov National Medical Research Centre , St-Petersburg , Russia.,Laboratory of Regenerative Biomedicine, Institute of Cytology, Russian Academy of Sciences , St-Petersburg , Russia
| | | | - Eugeniy Shlyakhto
- Laboratory of Molecular Cardiology, Almazov National Medical Research Centre , St-Petersburg , Russia
| | - Anna Kostareva
- Laboratory of Molecular Cardiology, Almazov National Medical Research Centre , St-Petersburg , Russia
| | - Luisa Marracino
- Department of Morphology, Surgery and Experimental Medicine and Laboratory for Technologies of Advanced Therapies (LTTA), University of Ferrara , Ferrara , Italy
| | - Roberto Ferrari
- Department of Medical Sciences, University of Ferrara , Ferrara , Italy.,Maria Cecilia Hospital, GVM Care & Research , Cotignola , Italy
| | - Paola Rizzo
- Maria Cecilia Hospital, GVM Care & Research , Cotignola , Italy.,Department of Morphology, Surgery and Experimental Medicine and Laboratory for Technologies of Advanced Therapies (LTTA), University of Ferrara , Ferrara , Italy
| | - Anna Malaschicheva
- Laboratory of Molecular Cardiology, Almazov National Medical Research Centre , St-Petersburg , Russia.,Laboratory of Regenerative Biomedicine, Institute of Cytology, Russian Academy of Sciences , St-Petersburg , Russia.,Department of Embryology, Faculty of Biology, Saint-Petersburg State University , St. Petersburg , Russia
| |
Collapse
|
25
|
Safety profiling of genetically engineered Pim-1 kinase overexpression for oncogenicity risk in human c-kit+ cardiac interstitial cells. Gene Ther 2019; 26:324-337. [PMID: 31239537 DOI: 10.1038/s41434-019-0084-5] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2019] [Revised: 03/19/2019] [Accepted: 05/14/2019] [Indexed: 12/11/2022]
Abstract
Advancement of stem cell-based treatment will involve next-generation approaches to enhance therapeutic efficacy which is often modest, particularly in the context of myocardial regenerative therapy. Our group has previously demonstrated the beneficial effect of genetic modification of cardiac stem cells with Pim-1 kinase overexpression to rejuvenate aged cells as well as potentiate myocardial repair. Despite these encouraging findings, concerns were raised regarding potential for oncogenic risk associated with Pim-1 kinase overexpression. Testing of Pim-1 engineered c-kit+ cardiac interstitial cells (cCIC) derived from heart failure patient samples for indices of oncogenic risk was undertaken using multiple assessments including soft agar colony formation, micronucleation, gamma-Histone 2AX foci, and transcriptome profiling. Collectively, findings demonstrate comparable phenotypic and biological properties of cCIC following Pim-1 overexpression compared with using baseline control cells with no evidence for oncogenic phenotype. Using a highly selective and continuous sensor for quantitative assessment of PIM1 kinase activity revealed a sevenfold increase in Pim-1 engineered vs. control cells. Kinase activity profiling using a panel of sensors for other kinases demonstrates elevation of IKKs), AKT/SGK, CDK1-3, p38, and ERK1/2 in addition to Pim-1 consistent with heightened kinase activity correlating with Pim-1 overexpression that may contribute to Pim-1-mediated effects. Enhancement of cellular survival, proliferation, and other beneficial properties to augment stem cell-mediated repair without oncogenic risk is a feasible, logical, and safe approach to improve efficacy and overcome current limitations inherent to cellular adoptive transfer therapeutic interventions.
Collapse
|
26
|
Abushouk AI, Salem AMA, Saad A, Afifi AM, Afify AY, Afify H, Salem HS, Ghanem E, Abdel-Daim MM. Mesenchymal Stem Cell Therapy for Doxorubicin-Induced Cardiomyopathy: Potential Mechanisms, Governing Factors, and Implications of the Heart Stem Cell Debate. Front Pharmacol 2019; 10:635. [PMID: 31258475 PMCID: PMC6586740 DOI: 10.3389/fphar.2019.00635] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2018] [Accepted: 05/17/2019] [Indexed: 12/13/2022] Open
Abstract
Over the past decades, researchers have reported several mechanisms for doxorubicin (DOX)-induced cardiomyopathy, including oxidative stress, inflammation, and apoptosis. Another mechanism that has been suggested is that DOX interferes with the cell cycle and induces oxidative stress in C-kit+ cells (commonly known as cardiac progenitor cells), reducing their regenerative capacity. Cardiac regeneration through enhancing the regenerative capacity of these cells or administration of other stem cells types has been the axis of several studies over the past 20 years. Several experiments revealed that local or systemic injections with mesenchymal stem cells (MSCs) were associated with significantly improved cardiac function, ameliorated inflammatory response, and reduced myocardial fibrosis. They also showed that several factors can affect the outcome of MSC treatment for DOX cardiomyopathy, including the MSC type, dose, route, and timing of administration. However, there is growing evidence that the C-kit+ cells do not have a cardiac regenerative potential in the adult mammalian heart. Similarly, the protective mechanisms of MSCs against DOX-induced cardiomyopathy are not likely to include direct differentiation into cardiomyocytes and probably occur through paracrine secretion, antioxidant and anti-inflammatory effects. Better understanding of the involved mechanisms and the factors governing the outcomes of MSCs therapy are essential before moving to clinical application in patients with DOX-induced cardiomyopathy.
Collapse
Affiliation(s)
| | | | - Anas Saad
- Faculty of Medicine, Ain Shams University, Cairo, Egypt
| | | | | | - Hesham Afify
- Wake Forest University, Winston-Salem, NC, United States
| | | | - Esraa Ghanem
- Faculty of Medicine, Al-Azhar University, Cairo, Egypt
| | - Mohamed M. Abdel-Daim
- Department of Pharmacology, Faculty of Veterinary Medicine, Suez Canal University, Ismailia, Egypt
| |
Collapse
|
27
|
mTOR and Aging: An Old Fashioned Dress. Int J Mol Sci 2019; 20:ijms20112774. [PMID: 31174250 PMCID: PMC6600378 DOI: 10.3390/ijms20112774] [Citation(s) in RCA: 65] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2019] [Revised: 06/03/2019] [Accepted: 06/04/2019] [Indexed: 12/12/2022] Open
Abstract
Aging is a physiologic/pathologic process characterized by a progressive impairment of cellular functions, supported by the alterations of several molecular pathways, leading to an increased cell susceptibility to injury. This deterioration is the primary risk factor for several major human pathologies. Numerous cellular processes, including genomic instability, telomere erosion, epigenetic alterations, loss of proteostasis, deregulated nutrient-sensing, mitochondrial dysfunction, stem cell exhaustion, and altered intercellular signal transduction represent common denominators of aging in different organisms. Mammalian target of rapamycin (mTOR) is an evolutionarily conserved nutrient sensing protein kinase that regulates growth and metabolism in all eukaryotic cells. Studies in flies, worms, yeast, and mice support the hypothesis that the mTOR signalling network plays a pivotal role in modulating aging. mTOR is emerging as the most robust mediator of the protective effects of various forms of dietary restriction, which has been shown to extend lifespan and slow the onset of age-related diseases across species. Herein we discuss the role of mTor signalling network in the development of classic age-related diseases, focused on cardiovascular system, immune response, and cancer.
Collapse
|
28
|
Lewis‐McDougall FC, Ruchaya PJ, Domenjo‐Vila E, Shin Teoh T, Prata L, Cottle BJ, Clark JE, Punjabi PP, Awad W, Torella D, Tchkonia T, Kirkland JL, Ellison‐Hughes GM. Aged-senescent cells contribute to impaired heart regeneration. Aging Cell 2019; 18:e12931. [PMID: 30854802 PMCID: PMC6516154 DOI: 10.1111/acel.12931] [Citation(s) in RCA: 216] [Impact Index Per Article: 36.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2018] [Revised: 01/09/2019] [Accepted: 01/31/2019] [Indexed: 12/19/2022] Open
Abstract
Aging leads to increased cellular senescence and is associated with decreased potency of tissue-specific stem/progenitor cells. Here, we have done an extensive analysis of cardiac progenitor cells (CPCs) isolated from human subjects with cardiovascular disease, aged 32-86 years. In aged subjects (>70 years old), over half of CPCs are senescent (p16INK4A , SA-β-gal, DNA damage γH2AX, telomere length, senescence-associated secretory phenotype [SASP]), unable to replicate, differentiate, regenerate or restore cardiac function following transplantation into the infarcted heart. SASP factors secreted by senescent CPCs renders otherwise healthy CPCs to senescence. Elimination of senescent CPCs using senolytics abrogates the SASP and its debilitative effect in vitro. Global elimination of senescent cells in aged mice (INK-ATTAC or wild-type mice treated with D + Q senolytics) in vivo activates resident CPCs and increased the number of small Ki67-, EdU-positive cardiomyocytes. Therapeutic approaches that eliminate senescent cells may alleviate cardiac deterioration with aging and restore the regenerative capacity of the heart.
Collapse
Affiliation(s)
- Fiona C. Lewis‐McDougall
- School of Basic and Medical Biosciences, Faculty of Life Sciences & MedicineKings College LondonLondonUK
| | - Prashant J. Ruchaya
- School of Basic and Medical Biosciences, Faculty of Life Sciences & MedicineKings College LondonLondonUK
| | - Eva Domenjo‐Vila
- School of Basic and Medical Biosciences, Faculty of Life Sciences & MedicineKings College LondonLondonUK
| | - Tze Shin Teoh
- School of Basic and Medical Biosciences, Faculty of Life Sciences & MedicineKings College LondonLondonUK
| | - Larissa Prata
- Robert and Arlene Kogod Center on AgingMayo Clinic College of MedicineRochesterMinnesota
| | - Beverley J. Cottle
- School of Basic and Medical Biosciences, Faculty of Life Sciences & MedicineKings College LondonLondonUK
| | - James E. Clark
- School of Cardiovascular Medicine & Sciences, Faculty of Life Sciences & MedicineKings College LondonLondonUK
| | | | | | - Daniele Torella
- Molecular and Cellular Cardiology, Department of Medical and Surgical SciencesMagna Graecia UniversityCatanzaroItaly
| | - Tamara Tchkonia
- Robert and Arlene Kogod Center on AgingMayo Clinic College of MedicineRochesterMinnesota
| | - James L. Kirkland
- Robert and Arlene Kogod Center on AgingMayo Clinic College of MedicineRochesterMinnesota
| | - Georgina M. Ellison‐Hughes
- School of Basic and Medical Biosciences, Faculty of Life Sciences & MedicineKings College LondonLondonUK
| |
Collapse
|
29
|
Parizadeh SM, Jafarzadeh‐Esfehani R, Ghandehari M, Parizadeh MR, Ferns GA, Avan A, Hassanian SM. Stem cell therapy: A novel approach for myocardial infarction. J Cell Physiol 2019; 234:16904-16912. [DOI: 10.1002/jcp.28381] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2018] [Revised: 01/21/2019] [Accepted: 01/24/2019] [Indexed: 12/12/2022]
Affiliation(s)
| | - Reza Jafarzadeh‐Esfehani
- Department of Medical Genetics Faculty of Medicine, Mashhad University of Medical Sciences Mashhad Iran
| | - Maryam Ghandehari
- Metabolic Syndrome Research Center Mashhad University of Medical Sciences Mashhad Iran
- Student Research Committee, Faculty of Medicine Islamic Azad University, Mashhad Branch Mashhad Iran
| | - Mohammad Reza Parizadeh
- Metabolic Syndrome Research Center Mashhad University of Medical Sciences Mashhad Iran
- Department of Clinical Biochemistry Faculty of Medicine, Mashhad University of Medical Sciences Mashhad Iran
| | - Gordon A. Ferns
- Brighton & Sussex Medical School Division of Medical Education Brighton UK
| | - Amir Avan
- Metabolic Syndrome Research Center Mashhad University of Medical Sciences Mashhad Iran
- Department of Modern Sciences and Technologies, Faculty of Medicine Mashhad University of Medical Sciences Mashhad Iran
| | - Seyed Mahdi Hassanian
- Metabolic Syndrome Research Center Mashhad University of Medical Sciences Mashhad Iran
- Department of Clinical Biochemistry Faculty of Medicine, Mashhad University of Medical Sciences Mashhad Iran
| |
Collapse
|
30
|
Korski KI, Kubli DA, Wang BJ, Khalafalla FG, Monsanto MM, Firouzi F, Echeagaray OH, Kim T, Adamson RM, Dembitsky WP, Gustafsson ÅB, Sussman MA. Hypoxia Prevents Mitochondrial Dysfunction and Senescence in Human c-Kit + Cardiac Progenitor Cells. Stem Cells 2019; 37:555-567. [PMID: 30629785 DOI: 10.1002/stem.2970] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2018] [Revised: 12/10/2018] [Accepted: 12/17/2018] [Indexed: 12/21/2022]
Abstract
Senescence-associated dysfunction deleteriously affects biological activities of human c-Kit+ cardiac progenitor cells (hCPCs), particularly under conditions of in vitro culture. In comparison, preservation of self-renewal and decreases in mitochondrial reactive oxygen species (ROS) are characteristics of murine CPCs in vivo that reside within hypoxic niches. Recapitulating hypoxic niche oxygen tension conditions of ∼1% O2 in vitro for expansion of hCPCs rather than typical normoxic cell culture conditions (21% O2 ) could provide significant improvement of functional and biological activities of hCPCs. hCPCs were isolated and expanded under permanent hypoxic (hCPC-1%) or normoxic (hCPC-21%) conditions from left ventricular tissue explants collected during left ventricular assist device implantation. hCPC-1% exhibit increased self-renewal and suppression of senescence characteristics relative to hCPC-21%. Oxidative stress contributed to higher susceptibility to apoptosis, as well as decreased mitochondrial function in hCPC-21%. Hypoxia prevented accumulation of dysfunctional mitochondria, supporting higher oxygen consumption rates and mitochondrial membrane potential. Mitochondrial ROS was an upstream mediator of senescence since treatment of hCPC-1% with mitochondrial inhibitor antimycin A recapitulated mitochondrial dysfunction and senescence observed in hCPC-21%. NAD+ /NADH ratio and autophagic flux, which are key factors for mitochondrial function, were higher in hCPC-1%, but hCPC-21% were highly dependent on BNIP3/NIX-mediated mitophagy to maintain mitochondrial function. Overall, results demonstrate that supraphysiological oxygen tension during in vitro expansion initiates a downward spiral of oxidative stress, mitochondrial dysfunction, and cellular energy imbalance culminating in early proliferation arrest of hCPCs. Senescence is inhibited by preventing ROS through hypoxic culture of hCPCs. Stem Cells 2019;37:555-567.
Collapse
Affiliation(s)
- Kelli I Korski
- Department of Biology and Integrated Regenerative Research Institute, San Diego State University, San Diego, California, USA
| | - Dieter A Kubli
- Department of Biology and Integrated Regenerative Research Institute, San Diego State University, San Diego, California, USA
| | - Bingyan J Wang
- Department of Biology and Integrated Regenerative Research Institute, San Diego State University, San Diego, California, USA
| | - Farid G Khalafalla
- Department of Biology and Integrated Regenerative Research Institute, San Diego State University, San Diego, California, USA
| | - Megan M Monsanto
- Department of Biology and Integrated Regenerative Research Institute, San Diego State University, San Diego, California, USA
| | - Fareheh Firouzi
- Department of Biology and Integrated Regenerative Research Institute, San Diego State University, San Diego, California, USA
| | - Oscar H Echeagaray
- Department of Biology and Integrated Regenerative Research Institute, San Diego State University, San Diego, California, USA
| | - Taeyong Kim
- Department of Biology and Integrated Regenerative Research Institute, San Diego State University, San Diego, California, USA
| | - Robert M Adamson
- Division of Cardiology, Sharp Hospital, San Diego, California, USA
| | | | - Åsa B Gustafsson
- The Skaggs School of Pharmacy and Pharmaceutical Sciences, Department of Pharmacology, University of California San Diego, La Jolla, California, USA
| | - Mark A Sussman
- Department of Biology and Integrated Regenerative Research Institute, San Diego State University, San Diego, California, USA
| |
Collapse
|
31
|
Khatiwala RV, Zhang S, Li X, Devejian N, Bennett E, Cai C. Inhibition of p16 INK4A to Rejuvenate Aging Human Cardiac Progenitor Cells via the Upregulation of Anti-oxidant and NFκB Signal Pathways. Stem Cell Rev Rep 2018; 14:612-625. [PMID: 29675777 DOI: 10.1007/s12015-018-9815-z] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Autologous human cardiac stem/progenitor cell (hCPC) therapy is a promising treatment that has come into use in recent years for patients with cardiomyopathy. Though innovative in theory, a major hindrance to the practical application of this treatment is that the hCPCs of elderly patients, who are most susceptible to myocardial disease, are senescent and prone to cell death. Rejuvenating hCPCs from elderly patients may help overcome this obstacle, and can be accomplished by reversing entry into the cellular stage of senescence. p16INK4A, a cyclin dependent kinase inhibitor, is an important player in the regulation of cell senescence. In this study, we investigated whether knockdown of p16INK4A will rejuvenate aging hCPCs to a youthful phenotype. Our data indicated that upregulation of p16INK4A is associated with hCPC senescence. Both cell proliferation and survival capacity were significantly increased in hCPCs infected with lentivirus expressing p16INK4A shRNA when compared to control hCPCs. The knockdown of p16INK4A also induced antioxidant properties as indicated by a 50% decrease in ROS generation at basal cell metabolism, and a 25% decrease in ROS generation after exposure to oxidative stress. Genes associated with cell senescence (p21CIP1), anti-apoptosis (BCL2 and MCL1), anti-oxidant (CYGB, PRDX1 and SRXN1), and NFκB signal pathway (p65, IKBKB, HMOX1, etc.), were significantly upregulated after the p16INK4A knockdown. Knocking down the NFĸB-p65 expression also significantly diminished the cytoprotective effect caused by the p16INK4A knockdown. Our results suggest that genetic knockdown of p16INK4A may play a significant role in inducing antioxidant effects and extending lifespan of aging hCPCs. This genetic modification may enhance the effectiveness of autologous hCPC therapy for repair of infarcted myocardium.
Collapse
Affiliation(s)
- Roshni V Khatiwala
- Department of Molecular and Cellular Physiology, Center for Cardiovascular Sciences, & Department of Medicine, Albany Medical College, Albany, NY, 12208, USA
| | - Shuning Zhang
- Department of Molecular and Cellular Physiology, Center for Cardiovascular Sciences, & Department of Medicine, Albany Medical College, Albany, NY, 12208, USA
| | - Xiuchun Li
- Department of Molecular and Cellular Physiology, Center for Cardiovascular Sciences, & Department of Medicine, Albany Medical College, Albany, NY, 12208, USA
| | - Neil Devejian
- Division of Pediatric Cardiothoracic Surgery, Albany Medical Center, Albany, NY, 12208, USA
| | - Edward Bennett
- Division of Cardiothoracic Surgery, Albany Medical Center, Albany, NY, 12208, USA
| | - Chuanxi Cai
- Department of Molecular and Cellular Physiology, Center for Cardiovascular Sciences, & Department of Medicine, Albany Medical College, Albany, NY, 12208, USA.
| |
Collapse
|
32
|
Ren J, Sowers JR, Zhang Y. Metabolic Stress, Autophagy, and Cardiovascular Aging: from Pathophysiology to Therapeutics. Trends Endocrinol Metab 2018; 29:699-711. [PMID: 30145108 PMCID: PMC6151141 DOI: 10.1016/j.tem.2018.08.001] [Citation(s) in RCA: 82] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/14/2018] [Revised: 07/30/2018] [Accepted: 08/02/2018] [Indexed: 12/11/2022]
Abstract
Recent advances in health care have improved the management of cardiometabolic disorders, and prolonged lifespan. However, the ever-rising prevalence of metabolic stress related to obesity (insulin resistance, diabetes, hypertension, and dyslipidemia) has greatly challenged geriatric care. The ubiquitin-proteasome system and autophagy-lysosomal pathways represent two major, yet distinct cellular machineries, for degradation and removal of damaged or long-lived proteins and organelles; the function of which declines with aging. To seek new strategies for cardiovascular aging under various metabolic diseases, it is imperative to understand the precise role for metabolic stress and protein quality control, in particular autophagy, in premature cardiovascular aging. Targeting metabolic stress and autophagy may offer exciting new avenues for the management of cardiovascular aging.
Collapse
Affiliation(s)
- Jun Ren
- Department of Cardiology, Fudan University Zhongshan Hospital, Shanghai, 200032, China; Center for Cardiovascular Research and Alternative Medicine, University of Wyoming College of Health Sciences, Laramie, WY 82071, USA.
| | - James R Sowers
- Diabetes and Dalton Cardiovascular Center and Harry S. Truman Memorial VA Research, University of Missouri-Columbia School of Medicine, Columbia, MO 65212, USA
| | - Yingmei Zhang
- Department of Cardiology, Fudan University Zhongshan Hospital, Shanghai, 200032, China; Center for Cardiovascular Research and Alternative Medicine, University of Wyoming College of Health Sciences, Laramie, WY 82071, USA.
| |
Collapse
|
33
|
Cianflone E, Aquila I, Scalise M, Marotta P, Torella M, Nadal-Ginard B, Torella D. Molecular basis of functional myogenic specification of Bona Fide multipotent adult cardiac stem cells. Cell Cycle 2018; 17:927-946. [PMID: 29862928 PMCID: PMC6103696 DOI: 10.1080/15384101.2018.1464852] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2018] [Revised: 06/01/2018] [Accepted: 04/08/2018] [Indexed: 01/14/2023] Open
Abstract
Ischemic Heart Disease (IHD) remains the developed world's number one killer. The improved survival from Acute Myocardial Infarction (AMI) and the progressive aging of western population brought to an increased incidence of chronic Heart Failure (HF), which assumed epidemic proportions nowadays. Except for heart transplantation, all treatments for HF should be considered palliative because none of the current therapies can reverse myocardial degeneration responsible for HF syndrome. To stop the HF epidemic will ultimately require protocols to reduce the progressive cardiomyocyte (CM) loss and to foster their regeneration. It is now generally accepted that mammalian CMs renew throughout life. However, this endogenous regenerative reservoir is insufficient to repair the extensive damage produced by AMI/IHD while the source and degree of CM turnover remains strongly disputed. Independent groups have convincingly shown that the adult myocardium harbors bona-fide tissue specific cardiac stem cells (CSCs). Unfortunately, recent reports have challenged the identity and the endogenous myogenic capacity of the c-kit expressing CSCs. This has hampered progress and unless this conflict is settled, clinical tests of repair/regenerative protocols are unlikely to provide convincing answers about their clinical potential. Here we review recent data that have eventually clarified the specific phenotypic identity of true multipotent CSCs. These cells when coaxed by embryonic cardiac morphogens undergo a precisely orchestrated myogenic commitment process robustly generating bona-fide functional cardiomyocytes. These data should set the path for the revival of further investigation untangling the regenerative biology of adult CSCs to harness their potential for HF prevention and treatment.
Collapse
Affiliation(s)
- Eleonora Cianflone
- Molecular and Cellular Cardiology, Department of Medical and Surgical Sciences, Magna Graecia University, Catanzaro, Italy
| | - Iolanda Aquila
- Molecular and Cellular Cardiology, Department of Medical and Surgical Sciences, Magna Graecia University, Catanzaro, Italy
| | - Mariangela Scalise
- Molecular and Cellular Cardiology, Department of Medical and Surgical Sciences, Magna Graecia University, Catanzaro, Italy
| | - Pina Marotta
- Molecular and Cellular Cardiology, Department of Medical and Surgical Sciences, Magna Graecia University, Catanzaro, Italy
| | - Michele Torella
- Department of Cardiothoracic Sciences, University of Campania Campus “Salvatore Venuta” Viale Europa- Loc. Germaneto “L. Vanvitelli”, Naples, Italy
| | - Bernardo Nadal-Ginard
- Molecular and Cellular Cardiology, Department of Medical and Surgical Sciences, Magna Graecia University, Catanzaro, Italy
| | - Daniele Torella
- Molecular and Cellular Cardiology, Department of Medical and Surgical Sciences, Magna Graecia University, Catanzaro, Italy
| |
Collapse
|
34
|
Shakeri H, Lemmens K, Gevaert AB, De Meyer GRY, Segers VFM. Cellular senescence links aging and diabetes in cardiovascular disease. Am J Physiol Heart Circ Physiol 2018; 315:H448-H462. [PMID: 29750567 DOI: 10.1152/ajpheart.00287.2018] [Citation(s) in RCA: 89] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Aging is a powerful independent risk factor for cardiovascular diseases such as atherosclerosis and heart failure. Concomitant diabetes mellitus strongly reinforces this effect of aging on cardiovascular disease. Cellular senescence is a fundamental mechanism of aging and appears to play a crucial role in the onset and prognosis of cardiovascular disease in the context of both aging and diabetes. Senescent cells are in a state of cell cycle arrest but remain metabolically active by secreting inflammatory factors. This senescence-associated secretory phenotype is a trigger of chronic inflammation, oxidative stress, and decreased nitric oxide bioavailability. A complex interplay between these three mechanisms results in age- and diabetes-associated cardiovascular damage. In this review, we summarize current knowledge on cellular senescence and its secretory phenotype, which might be the missing link between aging and diabetes contributing to cardiovascular disease.
Collapse
Affiliation(s)
- Hadis Shakeri
- Laboratory of Physiopharmacology, University of Antwerp , Antwerp , Belgium
| | - Katrien Lemmens
- Laboratory of Physiopharmacology, University of Antwerp , Antwerp , Belgium
| | - Andreas B Gevaert
- Laboratory of Physiopharmacology, University of Antwerp , Antwerp , Belgium.,Laboratory for Cellular and Molecular Cardiology, Department of Cardiology, University Hospital Antwerp, Edegem, Belgium
| | - Guido R Y De Meyer
- Laboratory of Physiopharmacology, University of Antwerp , Antwerp , Belgium
| | - Vincent F M Segers
- Laboratory of Physiopharmacology, University of Antwerp , Antwerp , Belgium.,Department of Cardiology, University Hospital Antwerp, Edegem, Belgium
| |
Collapse
|
35
|
Childs BG, Li H, van Deursen JM. Senescent cells: a therapeutic target for cardiovascular disease. J Clin Invest 2018; 128:1217-1228. [PMID: 29608141 DOI: 10.1172/jci95146] [Citation(s) in RCA: 131] [Impact Index Per Article: 18.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Cellular senescence, a major tumor-suppressive cell fate, has emerged from humble beginnings as an in vitro phenomenon into recognition as a fundamental mechanism of aging. In the process, senescent cells have attracted attention as a therapeutic target for age-related diseases, including cardiovascular disease (CVD), the leading cause of morbidity and mortality in the elderly. Given the aging global population and the inadequacy of current medical management, attenuating the health care burden of CVD would be transformative to clinical practice. Here, we review the evidence that cellular senescence drives CVD in a bimodal fashion by both priming the aged cardiovascular system for disease and driving established disease forward. Hence, the growing field of senotherapy (neutralizing senescent cells for therapeutic benefit) is poised to contribute to both prevention and treatment of CVD.
Collapse
Affiliation(s)
| | - Hu Li
- Department of Molecular Pharmacology and Experimental Therapeutics, and
| | - Jan M van Deursen
- Department of Biochemistry and Molecular Biology.,Department of Pediatric and Adolescent Medicine, Mayo Clinic, Rochester, Minnesota, USA
| |
Collapse
|
36
|
Gerisch M, Smettan J, Ebert S, Athelogou M, Brand-Saberi B, Spindler N, Mueller WC, Giri S, Bader A. Qualitative and Quantitative Analysis of Cardiac Progenitor Cells in Cases of Myocarditis and Cardiomyopathy. Front Genet 2018; 9:72. [PMID: 29559994 PMCID: PMC5845648 DOI: 10.3389/fgene.2018.00072] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2017] [Accepted: 02/16/2018] [Indexed: 11/24/2022] Open
Abstract
We aimed to identify and quantify CD117+ and CD90+ endogenous cardiac progenitor cells (CPC) in human healthy and diseased hearts. We hypothesize that these cells perform a locally acting, contributing function in overcoming medical conditions of the heart by endogenous means. Human myocardium biopsies were obtained from 23 patients with the following diagnoses: Dilatative cardiomyopathy (DCM), ischemic cardiomyopathy (ICM), myocarditis, and controls from healthy cardiac patients. High-resolution scanning microscopy of the whole slide enabled a computer-based immunohistochemical quantification of CD117 and CD90. Those signals were evaluated by Definiens Tissue Phenomics® Technology. Co-localization of CD117 and CD90 was determined by analyzing comparable serial sections. CD117+/CD90+ cardiac cells were detected in all biopsies. The highest expression of CD90 was revealed in the myocarditis group. CD117 was significantly higher in all patient groups, compared to healthy specimens (*p < 0.05). The highest co-expression was found in the myocarditis group (6.75 ± 3.25 CD90+CD117+ cells/mm2) followed by ICM (4 ± 1.89 cells/mm2), DCM (1.67 ± 0.58 cells/mm2), and healthy specimens (1 ± 0.43 cells/mm2). We conclude that the human heart comprises a fraction of local CD117+ and CD90+ cells. We hypothesize that these cells are part of local endogenous progenitor cells due to the co-expression of CD90 and CD117. With novel digital image analysis technologies, a quantification of the CD117 and CD90 signals is available. Our experiments reveal an increase of CD117 and CD90 in patients with myocarditis.
Collapse
Affiliation(s)
- Marie Gerisch
- Applied Stem Cell Biology and Cell Technology, Biomedical and Biotechnological Center, University of Leipzig, Leipzig, Germany
| | - Jan Smettan
- Division of Cardiology and Angiology, Department of Internal Medicine, Neurology and Dermatology, University Hospital Leipzig, Leipzig, Germany
| | - Sabine Ebert
- Applied Stem Cell Biology and Cell Technology, Biomedical and Biotechnological Center, University of Leipzig, Leipzig, Germany
| | | | - Beate Brand-Saberi
- Department of Anatomy and Molecular Embryology, Institute of Anatomy, Faculty of Medicine, Ruhr-University Bochum, Bochum, Germany
| | - Nick Spindler
- Department of Orthopedics, Trauma and Plastic Surgery, University Hospital Leipzig, Leipzig, Germany
| | - Wolf C Mueller
- Department of Neuropathology, University Hospital Leipzig, Leipzig, Germany
| | - Shibashish Giri
- Applied Stem Cell Biology and Cell Technology, Biomedical and Biotechnological Center, University of Leipzig, Leipzig, Germany.,Department of Plastic and Hand Surgery, University Hospital Rechts der Isar, Munich Technical University, Munich, Germany
| | - Augustinus Bader
- Applied Stem Cell Biology and Cell Technology, Biomedical and Biotechnological Center, University of Leipzig, Leipzig, Germany
| |
Collapse
|
37
|
Wu Q, Zhan J, Pu S, Qin L, Li Y, Zhou Z. Influence of aging on the activity of mice Sca-1+CD31- cardiac stem cells. Oncotarget 2018; 8:29-41. [PMID: 27980224 PMCID: PMC5352119 DOI: 10.18632/oncotarget.13930] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2016] [Accepted: 11/24/2016] [Indexed: 12/17/2022] Open
Abstract
Therapeutic application of cardiac resident stem/progenitor cells (CSC/CPCs) is limited due to decline of their regenerative potential with donor age. A variety of studies have shown that the cardiac aging was the problem of the stem cells, but little is known about the impact of age on the subgroups CSC/CPCs, the relationship between subgroups CSC/CPCs ageing and age-related dysfunction. Here, we studied Sca-1+CD31− subgroups of CSCs from younger(2~3months) and older(22~24months) age mice, biological differentiation was realized using specific mediums for 14 days to induce cardiomyocyte, smooth muscle cells or endothelial cells and immunostain analysis of differentiated cell resulting were done. Proliferation and cell cycle were measured by flow cytometry assay, then used microarray to dissect variability from younger and older mice. Although the number of CSCs was higher in older mice, the advanced age significantly reduced the differentiation ability into cardiac cell lineages and the proliferation ability. Transcriptional changes in Sca-1+CD31− subgroups of CSCs during aging are related to Vitamin B6 metabolism, circadian rhythm, Tyrosine metabolism, Complement and coagulation cascades. Taking together these results indicate that Cardiac resident stem/progenitor cells have significant differences in their proliferative, pluripotency and gene profiles and those differences are age depending.
Collapse
Affiliation(s)
- Qiong Wu
- School of Life Sciences, Guangxi Normal University, Guilin, China.,Guangxi Universities Key Laboratory of Stem cell and Biopharmaceutical Technology, Guangxi Normal University, Guilin, China.,Research Center for Biomedical Sciences, Guangxi Normal University, Guilin, China
| | - Jinxi Zhan
- School of Life Sciences, Guangxi Normal University, Guilin, China.,Guangxi Universities Key Laboratory of Stem cell and Biopharmaceutical Technology, Guangxi Normal University, Guilin, China.,Research Center for Biomedical Sciences, Guangxi Normal University, Guilin, China
| | - Shiming Pu
- School of Life Sciences, Guangxi Normal University, Guilin, China.,Guangxi Universities Key Laboratory of Stem cell and Biopharmaceutical Technology, Guangxi Normal University, Guilin, China.,Research Center for Biomedical Sciences, Guangxi Normal University, Guilin, China
| | - Liu Qin
- School of Life Sciences, Guangxi Normal University, Guilin, China.,Guangxi Universities Key Laboratory of Stem cell and Biopharmaceutical Technology, Guangxi Normal University, Guilin, China.,Research Center for Biomedical Sciences, Guangxi Normal University, Guilin, China
| | - Yun Li
- School of Life Sciences, Guangxi Normal University, Guilin, China.,Guangxi Universities Key Laboratory of Stem cell and Biopharmaceutical Technology, Guangxi Normal University, Guilin, China.,Research Center for Biomedical Sciences, Guangxi Normal University, Guilin, China
| | - Zuping Zhou
- School of Life Sciences, Guangxi Normal University, Guilin, China.,Guangxi Universities Key Laboratory of Stem cell and Biopharmaceutical Technology, Guangxi Normal University, Guilin, China.,Research Center for Biomedical Sciences, Guangxi Normal University, Guilin, China
| |
Collapse
|
38
|
Matsumoto C, Jiang Y, Emathinger J, Quijada P, Nguyen N, De La Torre A, Moshref M, Nguyen J, Levinson AB, Shin M, Sussman MA, Hariharan N. Short Telomeres Induce p53 and Autophagy and Modulate Age-Associated Changes in Cardiac Progenitor Cell Fate. Stem Cells 2018; 36:868-880. [PMID: 29441645 DOI: 10.1002/stem.2793] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2017] [Revised: 01/07/2018] [Accepted: 01/24/2018] [Indexed: 12/12/2022]
Abstract
Aging severely limits myocardial repair and regeneration. Delineating the impact of age-associated factors such as short telomeres is critical to enhance the regenerative potential of cardiac progenitor cells (CPCs). We hypothesized that short telomeres activate p53 and induce autophagy to elicit the age-associated change in CPC fate. We isolated CPCs and compared mouse strains with different telomere lengths for phenotypic characteristics of aging. Wild mouse strain Mus musculus castaneus (CAST) possessing short telomeres exhibits early cardiac aging with cardiac dysfunction, hypertrophy, fibrosis, and senescence, as compared with common lab strains FVB and C57 bearing longer telomeres. CAST CPCs with short telomeres demonstrate altered cell fate as characterized by cell cycle arrest, senescence, basal commitment, and loss of quiescence. Elongation of telomeres using a modified mRNA for telomerase restores youthful properties to CAST CPCs. Short telomeres induce autophagy in CPCs, a catabolic protein degradation process, as evidenced by reduced p62 and increased accumulation of autophagic puncta. Pharmacological inhibition of autophagosome formation reverses the cell fate to a more youthful phenotype. Mechanistically, cell fate changes induced by short telomeres are partially p53 dependent, as p53 inhibition rescues senescence and commitment observed in CAST CPCs, coincident with attenuation of autophagy. In conclusion, short telomeres activate p53 and autophagy to tip the equilibrium away from quiescence and proliferation toward differentiation and senescence, leading to exhaustion of CPCs. This study provides the mechanistic basis underlying age-associated cell fate changes that will enable identification of molecular strategies to prevent senescence of CPCs. Stem Cells 2018;36:868-880.
Collapse
Affiliation(s)
- Collin Matsumoto
- Department of Pharmacology, University of California at Davis, Davis, California, USA
| | - Yan Jiang
- Department of Pharmacology, University of California at Davis, Davis, California, USA
| | | | - Pearl Quijada
- Department of Biology, San Diego State University, San Diego, California, USA
| | - Nathalie Nguyen
- Department of Biology, San Diego State University, San Diego, California, USA
| | - Andrea De La Torre
- Department of Biology, San Diego State University, San Diego, California, USA
| | - Maryam Moshref
- Department of Pharmacology, University of California at Davis, Davis, California, USA
| | - Jonathan Nguyen
- Department of Biology, San Diego State University, San Diego, California, USA
| | - Aimee B Levinson
- Department of Pharmacology, University of California at Davis, Davis, California, USA
| | - Minyoung Shin
- Department of Pharmacology, University of California at Davis, Davis, California, USA
| | - Mark A Sussman
- Department of Biology, San Diego State University, San Diego, California, USA
| | - Nirmala Hariharan
- Department of Pharmacology, University of California at Davis, Davis, California, USA.,Department of Biology, San Diego State University, San Diego, California, USA
| |
Collapse
|
39
|
Saheera S, Potnuri AG, Nair RR. Modulation of cardiac stem cell characteristics by metoprolol in hypertensive heart disease. Hypertens Res 2018; 41:253-262. [PMID: 29449707 DOI: 10.1038/s41440-018-0015-7] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2017] [Revised: 06/19/2017] [Accepted: 08/04/2017] [Indexed: 11/09/2022]
Abstract
Cardiac stem cells (CSCs) play a vital role in cardiac remodeling. Uncontrolled hypertension leads to cardiac hypertrophy, followed by cardiac failure. Pathological remodeling is associated with enhanced oxidative stress. Decreased cardiac stem cell efficiency is speculated in heart diseases. Maintaining a healthy stem cell population is essential for preventing progressive cardiac remodeling. Some anti-hypertensive drugs are cardioprotective. However, the effect of these drugs on CSCs has not been investigated. Metoprolol is a cardioprotective anti-hypertensive agent. To examine whether metoprolol can prevent the deterioration of CSC efficiency, spontaneously hypertensive rats (SHRs) were treated with this drug, and the effects on stem cell function were evaluated. Six-month-old male SHRs were treated with metoprolol (50 mg × kg-1per day) for 2 months. The effectiveness of the treatment at reducing blood pressure and reducing hypertrophy was ensured, and the animals were killed. Cardiac stem cells were isolated from the atrial tissue, and the effect of metoprolol on stem cell migration, proliferation, differentiation, and survival was evaluated by comparing the treated SHRs with untreated SHRs and normotensive Wistar rats. Compared to the Wistar rats, the SHR rats presented with a decrease in stem cell migration and proliferation and an increase in intracellular oxidative stress and senescence. Treating SHRs with metoprolol increased CSC migration and proliferation potential and stemness retention. Cellular senescence and oxidative stress were reduced. The attributes of stem cells from the metoprolol-treated SHRs were comparable to those of the Wistar rats. The restoration of stem cell efficiency is expected to prevent hypertension-induced progressive cardiac remodeling.
Collapse
Affiliation(s)
- Sherin Saheera
- Division of Cellular and Molecular Cardiology, Sree Chitra Tirunal Institute for Medical Sciences and Technology, Trivandrum, Thiruvananthapuram, Kerala, 695011, India
| | - Ajay Godwin Potnuri
- Division of Cellular and Molecular Cardiology, Sree Chitra Tirunal Institute for Medical Sciences and Technology, Trivandrum, Thiruvananthapuram, Kerala, 695011, India
| | - Renuka R Nair
- Division of Cellular and Molecular Cardiology, Sree Chitra Tirunal Institute for Medical Sciences and Technology, Trivandrum, Thiruvananthapuram, Kerala, 695011, India.
| |
Collapse
|
40
|
Piegari E, Russo R, Cappetta D, Esposito G, Urbanek K, Dell'Aversana C, Altucci L, Berrino L, Rossi F, De Angelis A. MicroRNA-34a regulates doxorubicin-induced cardiotoxicity in rat. Oncotarget 2018; 7:62312-62326. [PMID: 27694688 PMCID: PMC5308729 DOI: 10.18632/oncotarget.11468] [Citation(s) in RCA: 58] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2016] [Accepted: 07/26/2016] [Indexed: 12/22/2022] Open
Abstract
New strategies to prevent and early detect the cardiotoxic effects of the anticancer drug doxorubicin (DOXO) are required. MicroRNAs emerged as potential diagnostic, therapeutic and prognostic approaches in cardiovascular diseases. MiR-34a has a role in cardiac dysfunction and ageing and is involved in several cellular processes associated with DOXO cardiotoxicity. Our in vitro and in vivo results indicated that after DOXO exposure the levels of miR-34a are enhanced in cardiac cells, including Cardiac Progenitor Cells (CPCs). Since one of the determining event responsible for the initiation and evolution of the DOXO toxicity arises at the level of the CPC compartment, we evaluated if miR-34a pharmacological inhibition in these cells ameliorates the detrimental aftermath of the drug. AntimiR-34a has beneficial consequences on vitality, proliferation, apoptosis and senescence of DOXO-treated rat CPC. These effects are mediated by an increase of prosurvival miR-34a targets Bcl-2 and SIRT1, accompanied by a decrease of acetylated-p53 and p16INK4a. Importantly, miR-34a silencing also reduces the release of this miRNA from DOXO-exposed rCPCs, decreasing its negative paracrine effects on other rat cardiac cells. In conclusion, the silencing of miR-34a could represent a future therapeutic option for cardioprotection in DOXO toxicity and at the same time, it could be considered as a circulating biomarker for anthracycline-induced cardiac damage.
Collapse
Affiliation(s)
- Elena Piegari
- Department of Experimental Medicine, Section of Pharmacology, Second University of Naples, Naples, Italy
| | - Rosa Russo
- Department of Experimental Medicine, Section of Pharmacology, Second University of Naples, Naples, Italy
| | - Donato Cappetta
- Department of Experimental Medicine, Section of Pharmacology, Second University of Naples, Naples, Italy
| | - Grazia Esposito
- Department of Experimental Medicine, Section of Pharmacology, Second University of Naples, Naples, Italy
| | - Konrad Urbanek
- Department of Experimental Medicine, Section of Pharmacology, Second University of Naples, Naples, Italy
| | | | - Lucia Altucci
- Institute of Genetics and Biophysics, IGB 'Adriano Buzzati-Traverso', Naples, Italy.,Department of Biochemistry, Biophysics and General Pathology, Second University of Naples, Naples, Italy
| | - Liberato Berrino
- Department of Experimental Medicine, Section of Pharmacology, Second University of Naples, Naples, Italy
| | - Francesco Rossi
- Department of Experimental Medicine, Section of Pharmacology, Second University of Naples, Naples, Italy
| | - Antonella De Angelis
- Department of Experimental Medicine, Section of Pharmacology, Second University of Naples, Naples, Italy
| |
Collapse
|
41
|
Cappetta D, Rossi F, Piegari E, Quaini F, Berrino L, Urbanek K, De Angelis A. Doxorubicin targets multiple players: A new view of an old problem. Pharmacol Res 2018; 127:4-14. [DOI: 10.1016/j.phrs.2017.03.016] [Citation(s) in RCA: 122] [Impact Index Per Article: 17.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/07/2017] [Revised: 03/16/2017] [Accepted: 03/16/2017] [Indexed: 01/22/2023]
|
42
|
Cesselli D, Aleksova A, Mazzega E, Caragnano A, Beltrami AP. Cardiac stem cell aging and heart failure. Pharmacol Res 2018; 127:26-32. [DOI: 10.1016/j.phrs.2017.01.013] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/25/2016] [Revised: 01/07/2017] [Accepted: 01/11/2017] [Indexed: 12/11/2022]
|
43
|
Saheera S, Nair RR. Accelerated decline in cardiac stem cell efficiency in Spontaneously hypertensive rat compared to normotensive Wistar rat. PLoS One 2017; 12:e0189129. [PMID: 29232369 PMCID: PMC5726722 DOI: 10.1371/journal.pone.0189129] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2017] [Accepted: 11/07/2017] [Indexed: 12/29/2022] Open
Abstract
Cardiac hypertrophy is recognized as an independent risk factor for cardiac failure. Efficient management of hypertensive heart disease requires identification of factors that can possibly mediate the transition from hypertrophy to failure. Resident cardiac stem cells have a prominent role in the maintenance of cardiac tissue homeostasis. Decline in the proportion of healthy cardiac stem cells (CSCs) can affect tissue regeneration. In pathological conditions, apart from natural aging, an adverse microenvironment can lead to decrease in efficiency of CSCs. A systematic analysis of cardiac stem cell characteristics in pathological conditions has not been reported so far. Therefore, this study was designed with the objective of examining the age associated variation in stem cell attributes of Spontaneously hypertensive rat (SHR) in comparison with normotensive Wistar rat. Spontaneously hypertensive rat was used as the experimental model since the cardiac remodeling resembles the clinical course of hypertensive heart disease. CSCs were isolated from atrial explants. Stem cell attributes were assessed in 1-week, 6, 12 and 18-month-old male SHR, in comparison with age matched Wistar rats. In 1-week-old pups, stem cell attributes of SHR and Wistar were comparable. Migration potential, proliferative capacity, TERT expression, telomerase activity and the proportion of c-kit+ cells decreased with age, both in SHR and Wistar. DNA damage and the proportion of senescent CSCs increased with age both in SHR and Wistar rats. Age associated increase was observed in the oxidative stress of stem cells, possibly mediated by the enhanced oxidative stress in the microenvironment. The changes were more pronounced in SHR, and as early as six months of age, there was significant decrease in efficiency of CSCs of SHR compared to Wistar. The density of healthy CSCs determined as a fraction of the differentiated cells was remarkably low in 18-month-old SHR. Age associated decrease in functionally efficient CSCs was therefore accelerated in SHR. Considering the vital role of CSCs in the maintenance of a healthy myocardium, decrease in functionally efficient CSCs can be a precipitating factor in pathological cardiac remodeling. Elevated ROS levels in CSCs of SHR lends scope for speculation that decrease in efficiency of CSCs is mediated by oxidative stress; and that modulation of the microenvironment by therapeutic interventions can restore a healthy stem cell population and facilitate maintenance of cardiac homeostasis and prevent cardiac decompensation.
Collapse
Affiliation(s)
- Sherin Saheera
- Division of Cellular and Molecular Cardiology, Sree Chitra Tirunal Institute for Medical Sciences and Technology, Trivandrum, Thiruvananthapuram, Kerala, India
| | - Renuka R. Nair
- Division of Cellular and Molecular Cardiology, Sree Chitra Tirunal Institute for Medical Sciences and Technology, Trivandrum, Thiruvananthapuram, Kerala, India
- * E-mail:
| |
Collapse
|
44
|
A Combination of Allogeneic Stem Cells Promotes Cardiac Regeneration. J Am Coll Cardiol 2017; 70:2504-2515. [PMID: 29145950 DOI: 10.1016/j.jacc.2017.09.036] [Citation(s) in RCA: 60] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/15/2017] [Revised: 09/15/2017] [Accepted: 09/15/2017] [Indexed: 12/15/2022]
Abstract
BACKGROUND The combination of autologous mesenchymal stem cells (MSCs) and cardiac stem cells (CSCs) synergistically reduces scar size and improves cardiac function in ischemic cardiomyopathy. Whereas allogeneic (allo-)MSCs are immunoevasive, the capacity of CSCs to similarly elude the immune system remains controversial, potentially limiting the success of allogeneic cell combination therapy (ACCT). OBJECTIVES This study sought to test the hypothesis that ACCT synergistically promotes cardiac regeneration without provoking immunologic reactions. METHODS Göttingen swine with experimental ischemic cardiomyopathy were randomized to receive transendocardial injections of allo-MSCs + allo-CSCs (ACCT: 200 million MSCs/1 million CSCs, n = 7), 200 million allo-MSCs (n = 8), 1 million allo-CSCs (n = 4), or placebo (Plasma-Lyte A, n = 6). Swine were assessed by cardiac magnetic resonance imaging and pressure volume catheterization. Immune response was tested by histologic analyses. RESULTS Both ACCT and allo-MSCs reduced scar size by -11.1 ± 4.8% (p = 0.012) and -9.5 ± 4.8% (p = 0.047), respectively. Only ACCT, but not MSCs or CSCs, prevented ongoing negative remodeling by offsetting increases in chamber volumes. Importantly, ACCT exerted the greatest effect on systolic function, improving the end-systolic pressure-volume relation (+0.98 ± 0.41 mm Hg/ml; p = 0.016). The ACCT group had more phospho-histone H3+ (a marker of mitosis) cardiomyocytes (p = 0.04), and noncardiomyocytes (p = 0.0002) than did the placebo group in some regions of the heart. Inflammatory sites in ACCT and MSC-treated swine contained immunotolerant CD3+/CD25+/FoxP3+ regulatory T cells (p < 0.0001). Histologic analysis showed absent to low-grade inflammatory infiltrates without cardiomyocyte necrosis. CONCLUSIONS ACCT demonstrates synergistic effects to enhance cardiac regeneration and left ventricular functional recovery in a swine model of chronic ischemic cardiomyopathy without adverse immunologic reaction. Clinical translation to humans is warranted.
Collapse
|
45
|
Khalafalla FG, Kayani W, Kassab A, Ilves K, Monsanto MM, Alvarez R, Chavarria M, Norman B, Dembitsky WP, Sussman MA. Empowering human cardiac progenitor cells by P2Y 14 nucleotide receptor overexpression. J Physiol 2017; 595:7135-7148. [PMID: 28980705 DOI: 10.1113/jp274980] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2017] [Accepted: 09/27/2017] [Indexed: 01/10/2023] Open
Abstract
KEY POINTS Autologous cardiac progenitor cell (CPC) therapy is a promising approach for treatment of heart failure (HF). There is an unmet need to identify inherent deficits in aged/diseased human CPCs (hCPCs) derived from HF patients in the attempts to augment their regenerative capacity prior to use in the clinical setting. Here we report significant functional correlations between phenotypic properties of hCPCs isolated from cardiac biopsies of HF patients, clinical parameters of patients and expression of the P2Y14 purinergic receptor (P2Y14 R), a crucial detector for extracellular UDP-sugars released during injury/stress. P2Y14 R is downregulated in hCPCs derived from HF patients with lower ejection fraction or diagnosed with diabetes. Augmenting P2Y14 R expression levels in aged/diseased hCPCs antagonizes senescence and improves functional responses. This study introduces purinergic signalling modulation as a potential strategy to rejuvenate and improve phenotypic characteristics of aged/functionally compromised hCPCs prior to transplantation in HF patients. ABSTRACT Autologous cardiac progenitor cell therapy is a promising alternative approach to current inefficient therapies for heart failure (HF). However, ex vivo expansion and pharmacological/genetic modification of human cardiac progenitor cells (hCPCs) are necessary interventions to rejuvenate aged/diseased cells and improve their regenerative capacities. This study was designed to assess the potential of improving hCPC functional capacity by targeting the P2Y14 purinergic receptor (P2Y14 R), which has been previously reported to induce regenerative and anti-senescence responses in a variety of experimental models. c-Kit+ hCPCs were isolated from cardiac biopsies of multiple HF patients undergoing left ventricular assist device implantation surgery. Significant correlations existed between the expression of P2Y14 R in hCPCs and clinical parameters of HF patients. P2Y14 R was downregulated in hCPCs derived from patients with a relatively lower ejection fraction and patients diagnosed with diabetes. hCPC lines with lower P2Y14 R expression did not respond to P2Y14 R agonist UDP-glucose (UDP-Glu) while hCPCs with higher P2Y14 R expression showed enhanced proliferation in response to UDP-Glu stimulation. Mechanistically, UDP-Glu stimulation enhanced the activation of canonical growth signalling pathways ERK1/2 and AKT. Restoring P2Y14 R expression levels in functionally compromised hCPCs via lentiviral-mediated overexpression improved proliferation, migration and survival under stress stimuli. Additionally, P2Y14 R overexpression reversed senescence-associated morphology and reduced levels of molecular markers of senescence p16INK4a , p53, p21 and mitochondrial reactive oxygen species. Findings from this study unveil novel biological roles of the UDP-sugar receptor P2Y14 in hCPCs and suggest purinergic signalling modulation as a promising strategy to improve phenotypic properties of functionally impaired hCPCs.
Collapse
Affiliation(s)
- Farid G Khalafalla
- San Diego Heart Research Institute, San Diego State University, 5500 Campanile Drive, San Diego, CA, 92182, USA
| | - Waqas Kayani
- San Diego Heart Research Institute, San Diego State University, 5500 Campanile Drive, San Diego, CA, 92182, USA
| | - Arwa Kassab
- San Diego Heart Research Institute, San Diego State University, 5500 Campanile Drive, San Diego, CA, 92182, USA
| | - Kelli Ilves
- San Diego Heart Research Institute, San Diego State University, 5500 Campanile Drive, San Diego, CA, 92182, USA
| | - Megan M Monsanto
- San Diego Heart Research Institute, San Diego State University, 5500 Campanile Drive, San Diego, CA, 92182, USA
| | - Roberto Alvarez
- San Diego Heart Research Institute, San Diego State University, 5500 Campanile Drive, San Diego, CA, 92182, USA
| | - Monica Chavarria
- San Diego Heart Research Institute, San Diego State University, 5500 Campanile Drive, San Diego, CA, 92182, USA
| | - Benjamin Norman
- San Diego Heart Research Institute, San Diego State University, 5500 Campanile Drive, San Diego, CA, 92182, USA
| | | | - Mark A Sussman
- San Diego Heart Research Institute, San Diego State University, 5500 Campanile Drive, San Diego, CA, 92182, USA
| |
Collapse
|
46
|
Cardiac Nonmyocyte Cell Functions and Crosstalks in Response to Cardiotoxic Drugs. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2017; 2017:1089359. [PMID: 29201269 PMCID: PMC5671742 DOI: 10.1155/2017/1089359] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/29/2017] [Accepted: 09/24/2017] [Indexed: 01/06/2023]
Abstract
The discovery of the molecular mechanisms involved in the cardiac responses to anticancer drugs represents the current goal of cardio-oncology research. The oxidative stress has a pivotal role in cardiotoxic responses, affecting the function of all types of cardiac cells, and their functional crosstalks. Generally, cardiomyocytes are the main target of research studies on cardiotoxicity, but recently the contribution of the other nonmyocyte cardiac cells is becoming of growing interest. This review deals with the role of oxidative stress, induced by anticancer drugs, in cardiac nonmyocyte cells (fibroblasts, vascular cells, and immune cells). The alterations of functional interplays among these cardiac cells are discussed, as well. These interesting recent findings increase the knowledge about cardiotoxicity and suggest new molecular targets for both diagnosis and therapy.
Collapse
|
47
|
Strategies to Enhance the Effectiveness of Adult Stem Cell Therapy for Ischemic Heart Diseases Affecting the Elderly Patients. Stem Cell Rev Rep 2017; 12:214-23. [PMID: 26779896 DOI: 10.1007/s12015-016-9642-z] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
Myocardial infarctions and chronic ischemic heart disease both commonly and disproportionately affect elderly patients more than any other patient population. Despite available treatments, heart tissue is often permanently damaged as a result of cardiac injury. This review aims to summarize recent literature proposing the use of modified autologous adult stem cells to promote healing of post-infarct cardiac tissue. This novel cellular treatment involves isolation of adult stem cells from the patient, in vitro manipulation of these stem cells, and subsequent transplantation back into the patient's own heart to accelerate healing. One of the hindrances affecting this process is that cardiac issues are increasingly common in elderly patients, and stem cells recovered from their tissues tend to be pre-senescent or already in senescence. As a result, harsh in vitro manipulations can cause the aged stem cells to undergo massive in vivo apoptosis after transplantation. The consensus in literature is that inhibition or reversal of senescence onset in adult stem cells would be of utmost benefit. In fact, it is believed that this strategy may lower stem cell mortality and coerce aged stem cells into adopting more resilient phenotypes similar to that of their younger counterparts. This review will discuss a selection of the most efficient and most-recent strategies used experimentally to enhance the effectiveness of current stem cell therapies for ischemic heart diseases.
Collapse
|
48
|
Saheera S, Potnuri AG, Nair R. Histamine-2 receptor antagonist famotidine modulates cardiac stem cell characteristics in hypertensive heart disease. PeerJ 2017; 5:e3882. [PMID: 29038754 PMCID: PMC5637875 DOI: 10.7717/peerj.3882] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2017] [Accepted: 09/12/2017] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND Cardiac stem cells (CSCs) play a vital role in cardiac homeostasis. A decrease in the efficiency of cardiac stem cells is speculated in various cardiac abnormalities. The maintenance of a healthy stem cell population is essential for the prevention of adverse cardiac remodeling leading to cardiac failure. Famotidine, a histamine-2 receptor antagonist, is currently used to treat ulcers of the stomach and intestines. In repurposing the use of the drug, reduction of cardiac hypertrophy and improvement in cardiac function of spontaneously hypertensive rats (SHR) was reported by our group. Given that stem cells are affected in cardiac pathologies, the effect of histamine-2 receptor antagonism on CSC characteristics was investigated. METHODS To examine whether famotidine has a positive effect on CSCs, spontaneously hypertensive rats (SHR) treated with the drug were sacrificed; and CSCs isolated from atrial appendages was evaluated. Six-month-old male SHRs were treated with famotidine (30 mg/kg/day) for two months. The effect of famotidine treatment on migration, proliferation and survival of CSCs was compared with untreated SHRs and normotensive Wistar rats. RESULTS Functional efficiency of CSCs from SHR was compromised relative to that in Wistar rat. Famotidine increased the migration and proliferation potential, along with retention of stemness of CSCs in treated SHRs. Cellular senescence and oxidative stress were also reduced. The expression of H2R was unaffected by the treatment. DISCUSSION As anticipated, CSCs from SHRs were functionally impaired. Stem cell attributes of famotidine-treated SHRs was comparable to that of Wistar rats. Therefore, in addition to being cardioprotective, the histamine 2 receptor antagonist modulated cardiac stem cells characteristics. Restoration of stem cell efficiency by famotidine is possibly mediated by reduction of oxidative stress as the expression of H2R was unaffected by the treatment. Maintenance of healthy stem cell population is suggested as a possible mechanism underlying the cardioprotective effect of famotidine.
Collapse
Affiliation(s)
- Sherin Saheera
- Division of Cellular and Molecular Cardiology, Sree Chitra Tirunal Institute for Medical Sciences and Technology, Thiruvananthapuram, Kerala, India
| | - Ajay G Potnuri
- Division of Cellular and Molecular Cardiology, Sree Chitra Tirunal Institute for Medical Sciences and Technology, Thiruvananthapuram, Kerala, India
| | - Renuka Nair
- Division of Cellular and Molecular Cardiology, Sree Chitra Tirunal Institute for Medical Sciences and Technology, Thiruvananthapuram, Kerala, India
| |
Collapse
|
49
|
The Light and Shadow of Senescence and Inflammation in Cardiovascular Pathology and Regenerative Medicine. Mediators Inflamm 2017; 2017:7953486. [PMID: 29118467 PMCID: PMC5651105 DOI: 10.1155/2017/7953486] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2017] [Accepted: 09/12/2017] [Indexed: 02/06/2023] Open
Abstract
Recent epidemiologic studies evidence a dramatic increase of cardiovascular diseases, especially associated with the aging of the world population. During aging, the progressive impairment of the cardiovascular functions results from the compromised tissue abilities to protect the heart against stress. At the molecular level, in fact, a gradual weakening of the cellular processes regulating cardiovascular homeostasis occurs in aging cells. Atherosclerosis and heart failure are particularly correlated with aging-related cardiovascular senescence, that is, the inability of cells to progress in the mitotic program until completion of cytokinesis. In this review, we explore the intrinsic and extrinsic causes of cellular senescence and their role in the onset of these cardiovascular pathologies. Additionally, we dissect the effects of aging on the cardiac endogenous and exogenous reservoirs of stem cells. Finally, we offer an overview on the strategies of regenerative medicine that have been advanced in the quest for heart rejuvenation.
Collapse
|
50
|
Berberine-induced cardioprotection and Sirt3 modulation in doxorubicin-treated H9c2 cardiomyoblasts. Biochim Biophys Acta Mol Basis Dis 2017; 1863:2904-2923. [PMID: 28760703 DOI: 10.1016/j.bbadis.2017.07.030] [Citation(s) in RCA: 59] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2017] [Revised: 07/18/2017] [Accepted: 07/27/2017] [Indexed: 12/22/2022]
Abstract
Doxorubicin (DOX) is one of the most widely used anti-neoplastic agents. However, treatment with DOX is associated with cumulative cardiotoxicity inducing progressive cardiomyocyte death. Sirtuin 3 (Sirt3), a mitochondrial deacetylase, regulates the activity of proteins involved in apoptosis, autophagy and metabolism. Our hypothesis is that pharmacological modulation by berberine (BER) pre-conditioning of Sirt3 protein levels decreases DOX-induced cardiotoxicity. Our results showed that DOX induces cell death in all experimental groups. Increase in Sirt3 content by transfection-mediated overexpression decreased DOX cytotoxicity, mostly by maintaining mitochondrial network integrity and reducing oxidative stress. p53 was upregulated by DOX, and appeared to be a direct target of Sirt3, suggesting that Sirt3-mediated protection against cell death could be related to this protein. BER pre-treatment increased Sirt3 and Sirt1 protein levels in the presence of DOX and inhibited DOX-induced caspase 9 and 3-like activation. Moreover, BER modulated autophagy in DOX-treated H9c2 cardiomyoblasts. Interestingly, mitochondrial biogenesis markers were upregulated in in BER/DOX-treated cells. Sirt3 over-expression contributes to decrease DOX cytotoxicity on H9c2 cardiomyoblasts, while BER can be used as a modulator of Sirtuin function and cell quality control pathways to decrease DOX toxicity.
Collapse
|