51
|
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
|
52
|
In vitro aged, hiPSC-origin engineered heart tissue models with age-dependent functional deterioration to study myocardial infarction. Acta Biomater 2019; 94:372-391. [PMID: 31146032 DOI: 10.1016/j.actbio.2019.05.064] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2019] [Revised: 05/20/2019] [Accepted: 05/24/2019] [Indexed: 01/29/2023]
Abstract
Deaths attributed to ischemic heart disease increased by 41.7% from 1990 to 2013. This is primarily due to an increase in the aged population, however, research on cardiovascular disease (CVD) has been overlooking aging, a well-documented contributor to CVD. The use of young animals is heavily preferred due to lower costs and ready availability, despite the prominent differences between young and aged heart structure and function. Here we present the first human induced pluripotent stem cell (hiPSC)-derived cardiomyocyte (iCM)-based, in vitro aged myocardial tissue model as an alternative research platform. Within 4 months, iCMs go through accelerated senescence and show cellular characteristics of aging. Furthermore, the model tissues fabricated using aged iCMs, with stiffness resembling that of aged human heart, show functional and pharmacological deterioration specific to aged myocardium. Our novel tissue model with age-appropriate physiology and pathology presents a promising new platform for investigating CVD or other age-related diseases. STATEMENT OF SIGNIFICANCE: In vitro and in vivo models of cardiovascular disease are aimed to provide crucial insight on the pathology and treatment of these diseases. However, the contribution of age-dependent cardiovascular changes is greatly underestimated through the use of young animals and premature cardiomyocytes. Here, we developed in vitro aged cardiac tissue models that mimic the aged heart tissue microenvironment and cellular phenotype and present the first evidence that age-appropriate in vitro disease models can be developed to gain more physiologically-relevant insight on development, progression, and amelioration of cardiovascular diseases.
Collapse
|
53
|
Abushouk AI, Salem AMA, Saad A, Afifi AM, Afify AY, Afify H, Salem HSE, 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: 24] [Impact Index Per Article: 4.8] [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
| | - Ahmed M Afifi
- 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
|
54
|
Morin D, Long R, Panel M, Laure L, Taranu A, Gueguen C, Pons S, Leoni V, Caccia C, Vatner SF, Vatner DE, Qiu H, Depre C, Berdeaux A, Ghaleh B. Hsp22 overexpression induces myocardial hypertrophy, senescence and reduced life span through enhanced oxidative stress. Free Radic Biol Med 2019; 137:194-200. [PMID: 31047988 DOI: 10.1016/j.freeradbiomed.2019.04.035] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/05/2019] [Revised: 04/12/2019] [Accepted: 04/26/2019] [Indexed: 10/26/2022]
Abstract
H11 kinase/Hsp22 (Hsp22) is a small heat shock protein, which, when overexpressed cardiac specifically in transgenic (TG) mice, induces stable left ventricular (LV) hypertrophy. Hsp22 also increases oxidative phosphorylation and mitochondrial reactive oxygen species (ROS) production, mechanisms mediating LV hypertrophy, senescence and reduced lifespan. Therefore, we investigated whether ROS production mediates LV hypertrophy, senescence and reduced life span in Hsp22 TG mice. Survival curves revealed that TG mice had a 48% reduction in their mean life span compared to wild type (WT) mice. This was associated with a significant increase in senescence markers, such as p16, p19 mRNA levels as well as the percentage of β-galactosidase positive cells and telomerase activity. Oxidized (GSSG)/reduced (GSH) glutathione ratio, an indicator of oxidative stress, and ROS production from 3 major cellular sources was measured in cardiac tissue. Hearts from TG mice exhibited a decrease in GSH/GSSG ratio together with increased ROS production from all sources. To study the role of ROS, mice were treated with the antioxidant Tempol from weaning to their sacrifice. Chronic Tempol treatment abolished oxidative stress and overproduction of ROS, and reduced myocardial hypertrophy and Akt phosphorylation in TG mice. Tempol also significantly extended life span and prevented aging markers in TG mice. Taken together these results show that overexpression of Hsp22 increases oxidative stress responsible for the induction of hypertrophy and senescence and ultimately reduction in life span.
Collapse
Affiliation(s)
- Didier Morin
- U955-IMRB, Equipe 03, Inserm, UPEC, Ecole Nationale Vétérinaire d'Alfort, Créteil, France.
| | - Romain Long
- U955-IMRB, Equipe 03, Inserm, UPEC, Ecole Nationale Vétérinaire d'Alfort, Créteil, France
| | - Mathieu Panel
- U955-IMRB, Equipe 03, Inserm, UPEC, Ecole Nationale Vétérinaire d'Alfort, Créteil, France
| | - Lydie Laure
- U955-IMRB, Equipe 03, Inserm, UPEC, Ecole Nationale Vétérinaire d'Alfort, Créteil, France
| | - Adela Taranu
- U955-IMRB, Equipe 03, Inserm, UPEC, Ecole Nationale Vétérinaire d'Alfort, Créteil, France
| | - Cindy Gueguen
- U955-IMRB, Equipe 03, Inserm, UPEC, Ecole Nationale Vétérinaire d'Alfort, Créteil, France
| | - Sandrine Pons
- U955-IMRB, Equipe 03, Inserm, UPEC, Ecole Nationale Vétérinaire d'Alfort, Créteil, France
| | - Valerio Leoni
- Laboratory Medicine, Desio Hospital, University of Milano Bicocca, Milan, Italy
| | - Claudio Caccia
- Laboratory of Clinical Pathology and Medical Genetics, Institute Neurologico IRCCS Carlo Besta, Milano, Italy
| | - Stephen F Vatner
- Department of Cell Biology and Molecular Medicine, New Jersey Medical School, Rutgers, The State University of New Jersey, USA
| | - Dorothy E Vatner
- Department of Cell Biology and Molecular Medicine, New Jersey Medical School, Rutgers, The State University of New Jersey, USA
| | - Hongyu Qiu
- Department of Cell Biology and Molecular Medicine, New Jersey Medical School, Rutgers, The State University of New Jersey, USA
| | - Christophe Depre
- Department of Cell Biology and Molecular Medicine, New Jersey Medical School, Rutgers, The State University of New Jersey, USA
| | - Alain Berdeaux
- U955-IMRB, Equipe 03, Inserm, UPEC, Ecole Nationale Vétérinaire d'Alfort, Créteil, France
| | - Bijan Ghaleh
- U955-IMRB, Equipe 03, Inserm, UPEC, Ecole Nationale Vétérinaire d'Alfort, Créteil, France
| |
Collapse
|
55
|
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: 194] [Impact Index Per Article: 38.8] [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
|
56
|
Adult Cardiac Stem Cell Aging: A Reversible Stochastic Phenomenon? OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2019; 2019:5813147. [PMID: 30881594 PMCID: PMC6383393 DOI: 10.1155/2019/5813147] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/09/2018] [Accepted: 11/08/2018] [Indexed: 12/17/2022]
Abstract
Aging is by far the dominant risk factor for the development of cardiovascular diseases, whose prevalence dramatically increases with increasing age reaching epidemic proportions. In the elderly, pathologic cellular and molecular changes in cardiac tissue homeostasis and response to injury result in progressive deteriorations in the structure and function of the heart. Although the phenotypes of cardiac aging have been the subject of intense study, the recent discovery that cardiac homeostasis during mammalian lifespan is maintained and regulated by regenerative events associated with endogenous cardiac stem cell (CSC) activation has produced a crucial reconsideration of the biology of the adult and aged mammalian myocardium. The classical notion of the adult heart as a static organ, in terms of cell turnover and renewal, has now been replaced by a dynamic model in which cardiac cells continuously die and are then replaced by CSC progeny differentiation. However, CSCs are not immortal. They undergo cellular senescence characterized by increased ROS production and oxidative stress and loss of telomere/telomerase integrity in response to a variety of physiological and pathological demands with aging. Nevertheless, the old myocardium preserves an endogenous functionally competent CSC cohort which appears to be resistant to the senescent phenotype occurring with aging. The latter envisions the phenomenon of CSC ageing as a result of a stochastic and therefore reversible cell autonomous process. However, CSC aging could be a programmed cell cycle-dependent process, which affects all or most of the endogenous CSC population. The latter would infer that the loss of CSC regenerative capacity with aging is an inevitable phenomenon that cannot be rescued by stimulating their growth, which would only speed their progressive exhaustion. The resolution of these two biological views will be crucial to design and develop effective CSC-based interventions to counteract cardiac aging not only improving health span of the elderly but also extending lifespan by delaying cardiovascular disease-related deaths.
Collapse
|
57
|
Sato M, Miyata K, Tian Z, Kadomatsu T, Ujihara Y, Morinaga J, Horiguchi H, Endo M, Zhao J, Zhu S, Sugizaki T, Igata K, Muramatsu M, Minami T, Ito T, Bianchi ME, Mohri S, Araki K, Node K, Oike Y. Loss of Endogenous HMGB2 Promotes Cardiac Dysfunction and Pressure Overload-Induced Heart Failure in Mice. Circ J 2019; 83:368-378. [DOI: 10.1253/circj.cj-18-0925] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Affiliation(s)
- Michio Sato
- Department of Molecular Genetics, Graduate School of Medical Sciences, Kumamoto University
- Department of Cardiovascular Medicine, Saga University
| | - Keishi Miyata
- Department of Molecular Genetics, Graduate School of Medical Sciences, Kumamoto University
- Department of Immunology, Allergy and Vascular Medicine, Graduate School of Medical Sciences, Kumamoto University
| | - Zhe Tian
- Department of Molecular Genetics, Graduate School of Medical Sciences, Kumamoto University
| | - Tsuyoshi Kadomatsu
- Department of Molecular Genetics, Graduate School of Medical Sciences, Kumamoto University
| | | | - Jun Morinaga
- Department of Molecular Genetics, Graduate School of Medical Sciences, Kumamoto University
| | - Haruki Horiguchi
- Department of Molecular Genetics, Graduate School of Medical Sciences, Kumamoto University
| | - Motoyoshi Endo
- Department of Molecular Genetics, Graduate School of Medical Sciences, Kumamoto University
| | - Jiabin Zhao
- Department of Molecular Genetics, Graduate School of Medical Sciences, Kumamoto University
| | - Shunshun Zhu
- Department of Molecular Genetics, Graduate School of Medical Sciences, Kumamoto University
| | - Taichi Sugizaki
- Department of Molecular Genetics, Graduate School of Medical Sciences, Kumamoto University
| | - Kimihiro Igata
- Department of Molecular Genetics, Graduate School of Medical Sciences, Kumamoto University
| | - Masashi Muramatsu
- Division of Molecular and Vascular Biology, Institute of Resource Development and Analysis, Kumamoto University
| | - Takashi Minami
- Division of Molecular and Vascular Biology, Institute of Resource Development and Analysis, Kumamoto University
| | - Takashi Ito
- Department of Systems Biology in Thromboregulation, Kagoshima University Graduate School of Medical and Dental Science
| | - Marco E Bianchi
- Chromatin Dynamics Unit, San Raffaele University and Scientific Institute
| | - Satoshi Mohri
- First Department of Physiology, Kawasaki Medical School
| | - Kimi Araki
- Center for Metabolic Regulation of Healthy Aging, Graduate School of Medical Sciences, Kumamoto University
- Division of Developmental Genetics, Institute of Resource Development and Analysis, Kumamoto University
| | - Koichi Node
- Department of Cardiovascular Medicine, Saga University
| | - Yuichi Oike
- Department of Molecular Genetics, Graduate School of Medical Sciences, Kumamoto University
- Center for Metabolic Regulation of Healthy Aging, Graduate School of Medical Sciences, Kumamoto University
| |
Collapse
|
58
|
Marino F, Scalise M, Cianflone E, Mancuso T, Aquila I, Agosti V, Torella M, Paolino D, Mollace V, Nadal-Ginard B, Torella D. Role of c-Kit in Myocardial Regeneration and Aging. Front Endocrinol (Lausanne) 2019; 10:371. [PMID: 31275242 PMCID: PMC6593054 DOI: 10.3389/fendo.2019.00371] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/22/2019] [Accepted: 05/24/2019] [Indexed: 12/15/2022] Open
Abstract
c-Kit, a type III receptor tyrosine kinase (RTK), is involved in multiple intracellular signaling whereby it is mainly considered a stem cell factor receptor, which participates in vital functions of the mammalian body, including the human. Furthermore, c-kit is a necessary yet not sufficient marker to detect and isolate several types of tissue-specific adult stem cells. Accordingly, c-kit was initially used as a marker to identify and enrich for adult cardiac stem/progenitor cells (CSCs) that were proven to be clonogenic, self-renewing and multipotent, being able to differentiate into cardiomyocytes, endothelial cells and smooth muscle cells in vitro as well as in vivo after myocardial injury. Afterwards it was demonstrated that c-kit expression labels a heterogenous cardiac cell population, which is mainly composed by endothelial cells while only a very small fraction represents CSCs. Furthermore, c-kit as a signaling molecule is expressed at different levels in this heterogenous c-kit labeled cardiac cell pool, whereby c-kit low expressers are enriched for CSCs while c-kit high expressers are endothelial and mast cells. This heterogeneity in cell composition and expression levels has been neglected in recent genetic fate map studies focusing on c-kit, which have claimed that c-kit identifies cells with robust endothelial differentiation potential but with minimal if not negligible myogenic commitment potential. However, modification of c-kit gene for Cre Recombinase expression in these Cre/Lox genetic fate map mouse models produced a detrimental c-kit haploinsufficiency that prevents efficient labeling of true CSCs on one hand while affecting the regenerative potential of these cells on the other. Interestingly, c-kit haploinsufficiency in c-kit-deficient mice causes a worsening myocardial repair after injury and accelerates cardiac aging. Therefore, these studies have further demonstrated that adult c-kit-labeled CSCs are robustly myogenic and that the adult myocardium relies on c-kit expression to regenerate after injury and to counteract aging effects on cardiac structure and function.
Collapse
Affiliation(s)
- Fabiola Marino
- Molecular and Cellular Cardiology, Department of Experimental and Clinical Medicine, University Magna Graecia, Catanzaro, Italy
- Department of Health Sciences, Interregional Research Center on Food Safety and Health (IRC-FSH), University Magna Graecia of Catanzaro, Catanzaro, Italy
| | - Mariangela Scalise
- Molecular and Cellular Cardiology, Department of Experimental and Clinical Medicine, University Magna Graecia, Catanzaro, Italy
| | - Eleonora Cianflone
- Molecular and Cellular Cardiology, Department of Experimental and Clinical Medicine, University Magna Graecia, Catanzaro, Italy
| | - Teresa Mancuso
- Molecular and Cellular Cardiology, Department of Experimental and Clinical Medicine, University Magna Graecia, Catanzaro, Italy
| | - Iolanda Aquila
- Molecular and Cellular Cardiology, Department of Experimental and Clinical Medicine, University Magna Graecia, Catanzaro, Italy
| | - Valter Agosti
- Interdepartmental Center of Services (CIS) of Genomics, Department of Experimental and Clinical Medicine, University Magna Graecia, Catanzaro, Italy
| | - Michele Torella
- Department of Cardiothoracic Sciences, University of Campania L. Vanvitelli, Naples, Italy
| | - Donatella Paolino
- Department of Experimental and Clinical Medicine, University Magna Graecia, Catanzaro, Italy
| | - Vincenzo Mollace
- Department of Health Sciences, Interregional Research Center on Food Safety and Health (IRC-FSH), University Magna Graecia of Catanzaro, Catanzaro, Italy
| | - Bernardo Nadal-Ginard
- Molecular and Cellular Cardiology, Department of Experimental and Clinical Medicine, University Magna Graecia, Catanzaro, Italy
- StemCell OpCo, Madrid, Spain
| | - Daniele Torella
- Molecular and Cellular Cardiology, Department of Experimental and Clinical Medicine, University Magna Graecia, Catanzaro, Italy
- *Correspondence: Daniele Torella
| |
Collapse
|
59
|
Dou X, Chen L, Lei M, Zellmer L, Jia Q, Ling P, He Y, Yang W, Liao DJ. Evaluating the Remote Control of Programmed Cell Death, with or without a Compensatory Cell Proliferation. Int J Biol Sci 2018; 14:1800-1812. [PMID: 30443184 PMCID: PMC6231223 DOI: 10.7150/ijbs.26962] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2018] [Accepted: 08/24/2018] [Indexed: 12/23/2022] Open
Abstract
Organisms and their different component levels, whether organelle, cellular or other, come by birth and go by death, and the deaths are often balanced by new births. Evolution on the one hand has built demise program(s) in cells of organisms but on the other hand has established external controls on the program(s). For instance, evolution has established death program(s) in animal cells so that the cells can, when it is needed, commit apoptosis or senescent death (SD) in physiological situations and stress-induced cell death (SICD) in pathological situations. However, these programmed cell deaths are not predominantly regulated by the cells that do the dying but, instead, are controlled externally and remotely by the cells' superior(s), i.e. their host tissue or organ or even the animal's body. Currently, it is still unclear whether a cell has only one death program or has several programs respectively controlling SD, apoptosis and SICD. In animals, apoptosis exterminates, in a physiological manner, healthy but no-longer needed cells to avoid cell redundancy, whereas suicidal SD and SICD, like homicidal necrosis, terminate ill but useful cells, which may be followed by regeneration of the live cells and by scar formation to heal the damaged organ or tissue. Therefore, “who dies” clearly differentiates apoptosis from SD, SICD and necrosis. In animals, apoptosis can occur only in those cell types that retain a lifelong ability of proliferation and never occurs in those cell types that can no longer replicate in adulthood. In cancer cells, SICD is strengthened, apoptosis is dramatically weakened while SD has been lost. Most published studies professed to be about apoptosis are actually about SICD, which has four basic and well-articulated pathways involving caspases or involving pathological alterations in the mitochondria, endoplasmic reticula, or lysosomes.
Collapse
Affiliation(s)
- Xixi Dou
- Key Laboratory of Biopharmaceuticals, Shandong Academy of Pharmaceutical Sciences, Jinan 250101, Shandong Province, P.R. China.,Technology Center, Shandong Freda Pharmaceutical Group, Jinan 250101, Shandong Province, P.R. China
| | - Lichan Chen
- College of Chemical Engineering, Huaqiao University, Xiamen 361021, Fujian Province, P.R. China
| | - Mingjuan Lei
- Hormel Institute, University of Minnesota, Austin, MN 55912, USA
| | - Lucas Zellmer
- Masonic Cancer Center, University of Minnesota, 435 E. River Road, Minneapolis, MN 55455, USA
| | - Qingwen Jia
- Key Laboratory of Biopharmaceuticals, Shandong Academy of Pharmaceutical Sciences, Jinan 250101, Shandong Province, P.R. China
| | - Peixue Ling
- Key Laboratory of Biopharmaceuticals, Shandong Academy of Pharmaceutical Sciences, Jinan 250101, Shandong Province, P.R. China.,Technology Center, Shandong Freda Pharmaceutical Group, Jinan 250101, Shandong Province, P.R. China
| | - Yan He
- Key Lab of Endemic and Ethnic Diseases of the Ministry of Education of China in Guizhou Medical University, Guiyang 550004, Guizhou Province, P.R. China
| | - Wenxiu Yang
- Department of Pathology, Guizhou Medical University Hospital, Guiyang 550004, Guizhou province, P.R. China
| | - Dezhong Joshua Liao
- Key Lab of Endemic and Ethnic Diseases of the Ministry of Education of China in Guizhou Medical University, Guiyang 550004, Guizhou Province, P.R. China.,Department of Pathology, Guizhou Medical University Hospital, Guiyang 550004, Guizhou province, P.R. China
| |
Collapse
|
60
|
Reyes ME, Ma J, Grove ML, Ater JL, Morrison AC, Hildebrandt MAT. RNA sequence analysis of inducible pluripotent stem cell-derived cardiomyocytes reveals altered expression of DNA damage and cell cycle genes in response to doxorubicin. Toxicol Appl Pharmacol 2018; 356:44-53. [PMID: 30031762 DOI: 10.1016/j.taap.2018.07.020] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2018] [Revised: 06/29/2018] [Accepted: 07/18/2018] [Indexed: 02/03/2023]
Abstract
Anthracyclines, such as doxorubicin, are highly effective chemotherapeutic agents, yet are associated with increased risk of cardiotoxicity. The genes and pathways involved in the development of heart damage following doxorubicin exposure in humans remain elusive. Our objective was to explore time- and dose-dependent changes in gene expression via RNA sequence (RNAseq) that mediate doxorubicin response in human iPSC-cardiomyocytes following 50, 150, or 450 nM exposure for 2, 7, or 12 days. Clustering and differential expression analyses were conducted to identify genes with altered expression. Samples clustered in dose and time-dependent manners, and MCM5, PRC1, NUSAP1, CENPF, CCNB1, MELK, AURKB, and RACGAP1 were consistently significantly differentially expressed between untreated and treated conditions. These genes were also significantly downregulated in pairwise analyses, which was validated by reverse transcription polymerase chain reaction (RT-PCR). Pathway analysis identified the top canonical pathways involved in response, implicating DNA damage repair response and the cell cycle as having roles in the development of doxorubicin-induced cardiotoxicity in the human cardiomyocyte.
Collapse
Affiliation(s)
- Monica E Reyes
- Department of Epidemiology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Jianzhong Ma
- Human Genetics Center, Department of Epidemiology, Human Genetics, and Environmental Sciences, School of Public Health, The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Megan L Grove
- Human Genetics Center, Department of Epidemiology, Human Genetics, and Environmental Sciences, School of Public Health, The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Joann L Ater
- Division of Pediatrics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Alanna C Morrison
- Human Genetics Center, Department of Epidemiology, Human Genetics, and Environmental Sciences, School of Public Health, The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Michelle A T Hildebrandt
- Department of Epidemiology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
| |
Collapse
|
61
|
Booth SA, Wadley GD, Marques FZ, Wlodek ME, Charchar FJ. Fetal growth restriction shortens cardiac telomere length, but this is attenuated by exercise in early life. Physiol Genomics 2018; 50:956-963. [PMID: 30192712 DOI: 10.1152/physiolgenomics.00042.2018] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
BACKGROUND AND AIMS Fetal and postnatal growth restriction cause a predisposition to cardiovascular disease (CVD) in adulthood. Telomeres are repetitive DNA-protein structures that protect chromosome ends, and the loss of these repeats (a reduction in telomere length) is associated with CVD. As exercise preserves telomere length and cardiovascular health, the aim of this study was to determine the effects of growth restriction and exercise training on cardiac telomere length and telomeric genes. METHODS AND RESULTS Pregnant Wistar Kyoto rats underwent bilateral uterine vessel ligation to induce uteroplacental insufficiency and fetal growth restriction ("Restricted"). Sham-operated rats had either intact litters ("Control") or their litters reduced to five pups with slowed postnatal growth ("Reduced"). Control, Restricted, and Reduced male rats were assigned to Sedentary, Early exercise (5-9 wk of age), or Late exercise (20-24 wk of age) groups. Hearts were excised at 24 wk of age for telomere length and gene expression measurements by quantitative PCR. Growth restriction shortened cardiac telomere length ( P < 0.001), but this was rescued by early exercise ( P < 0.001). Early and Late exercise increased cardiac weight index ( P < 0.001), but neither this nor telomere length was associated with expression of the telomeric genes Tert, Terc, Trf2, Pnuts, or Sirt1. DISCUSSION AND CONCLUSIONS Growth restriction shortens cardiac telomere length, reflecting the cardiac pathologies associated with low birth weight. Exercise in early life may offer long-term protective effects on cardiac telomere length, which could help prevent CVD in later life.
Collapse
Affiliation(s)
- S A Booth
- School of Health and Life Sciences, Federation University Australia , Victoria , Australia
| | - G D Wadley
- Institute for Physical Activity and Nutrition, School of Exercise and Nutrition Sciences, Deakin University , Geelong, Victoria , Australia
| | - F Z Marques
- School of Health and Life Sciences, Federation University Australia , Victoria , Australia.,Heart Failure Research Group, Baker Heart and Diabetes Institute , Melbourne , Australia.,Central Clinical School, Faculty of Medicine Nursing and Health Sciences, Monash University , Melbourne , Australia
| | - M E Wlodek
- Department of Physiology, The University of Melbourne , Parkville, Victoria , Australia
| | - F J Charchar
- School of Health and Life Sciences, Federation University Australia , Victoria , Australia.,Department of Physiology, The University of Melbourne , Parkville, Victoria , Australia.,Department of Cardiovascular Sciences, University of Leicester , Leicester , United Kingdom
| |
Collapse
|
62
|
Abstract
Most older individuals develop inflammageing, a condition characterized by elevated levels of blood inflammatory markers that carries high susceptibility to chronic morbidity, disability, frailty, and premature death. Potential mechanisms of inflammageing include genetic susceptibility, central obesity, increased gut permeability, changes to microbiota composition, cellular senescence, NLRP3 inflammasome activation, oxidative stress caused by dysfunctional mitochondria, immune cell dysregulation, and chronic infections. Inflammageing is a risk factor for cardiovascular diseases (CVDs), and clinical trials suggest that this association is causal. Inflammageing is also a risk factor for chronic kidney disease, diabetes mellitus, cancer, depression, dementia, and sarcopenia, but whether modulating inflammation beneficially affects the clinical course of non-CVD health problems is controversial. This uncertainty is an important issue to address because older patients with CVD are often affected by multimorbidity and frailty - which affect clinical manifestations, prognosis, and response to treatment - and are associated with inflammation by mechanisms similar to those in CVD. The hypothesis that inflammation affects CVD, multimorbidity, and frailty by inhibiting growth factors, increasing catabolism, and interfering with homeostatic signalling is supported by mechanistic studies but requires confirmation in humans. Whether early modulation of inflammageing prevents or delays the onset of cardiovascular frailty should be tested in clinical trials.
Collapse
Affiliation(s)
- Luigi Ferrucci
- Translational Gerontology Branch, National Institute on Aging, NIH, Baltimore, MD, USA.
| | - Elisa Fabbri
- Department of Medical and Surgical Sciences, University of Bologna, Bologna, Italy
| |
Collapse
|
63
|
Nacarelli T, Azar A, Altinok O, Orynbayeva Z, Sell C. Rapamycin increases oxidative metabolism and enhances metabolic flexibility in human cardiac fibroblasts. GeroScience 2018; 40:10.1007/s11357-018-0030-2. [PMID: 29931650 PMCID: PMC6060207 DOI: 10.1007/s11357-018-0030-2] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2016] [Accepted: 06/14/2018] [Indexed: 01/07/2023] Open
Abstract
Inhibition of mTOR signaling using rapamycin has been shown to increase lifespan and healthspan in multiple model organisms; however, the precise mechanisms for the beneficial effects of rapamycin remain uncertain. We have previously reported that rapamycin delays senescence in human cells and that enhanced mitochondrial biogenesis and protection from mitochondrial stress is one component of the benefit provided by rapamycin treatment. Here, using two models of senescence, replicative senescence and senescence induced by the presence of the Hutchinson-Gilford progeria lamin A mutation, we report that senescence is accompanied by elevated glycolysis and increased oxidative phosphorylation, which are both reduced by rapamycin. Measurements of mitochondrial function indicate that direct mitochondria targets of rapamycin are succinate dehydrogenase and matrix alanine aminotransferase. Elevated activity of these enzymes could be part of complex mechanisms that enable mitochondria to resume their optimal oxidative phosphorylation and resist senescence. This interpretation is supported by the fact that rapamycin-treated cultures do not undergo a premature senescence in response to the replacement of glucose with galactose in the culture medium, which forces a greater reliance on oxidative phosphorylation. Additionally, long-term treatment with rapamycin increases expression of the mitochondrial carrier protein UCP2, which facilitates the movement of metabolic intermediates across the mitochondrial membrane. The results suggest that rapamycin impacts mitochondrial function both through direct interaction with the mitochondria and through altered gene expression of mitochondrial carrier proteins.
Collapse
Affiliation(s)
| | - Ashley Azar
- Department of Pathology and Laboratory Medicine, Drexel University College of Medicine, 245 North 15th Street, Philadelphia, PA, 19102, USA
| | - Oya Altinok
- Department of Surgery, Drexel University College of Medicine, 245 North 15th Street, Philadelphia, PA, 19102, USA
| | - Zulfiya Orynbayeva
- Department of Surgery, Drexel University College of Medicine, 245 North 15th Street, Philadelphia, PA, 19102, USA
| | - Christian Sell
- Department of Pathology and Laboratory Medicine, Drexel University College of Medicine, 245 North 15th Street, Philadelphia, PA, 19102, USA.
| |
Collapse
|
64
|
Teng CF, Jeng LB, Shyu WC. Role of Insulin-like Growth Factor 1 Receptor Signaling in Stem Cell Stemness and Therapeutic Efficacy. Cell Transplant 2018; 27:1313-1319. [PMID: 29882416 PMCID: PMC6168993 DOI: 10.1177/0963689718779777] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Evidence has emerged that stem cells represent a promising therapeutic tool for tissue engineering and regenerative medicine. Thus, identifying functional markers for selecting stem cells capable of superior self-renewal and pluripotency (or multipotency) and maintaining stem cell identity under appropriate culture conditions are critical for guiding the use of stem cells toward clinical applications. Many investigations have implicated the insulin-like growth factor 1 receptor (IGF1R) signaling in maintenance of stem cell characteristics and enhancement of stem cell therapy efficacy. IGF1R-expressing stem cells display robust pluripotent or multipotent properties. In this review, we summarize the essential roles of IGF1R signaling in self-renewal, pluripotency (or multipotency), and therapeutic efficacy of stem cells, including human embryonic stem cells, neural stem cells, cardiac stem cells, bone marrow mesenchymal stem cells, placental mesenchymal stem cells, and dental pulp mesenchymal stem cells. Modifying IGF1R signaling may thus provide potential strategies for maintaining stem cell properties and improving stem-cell-based therapeutic applications.
Collapse
Affiliation(s)
- Chiao-Fang Teng
- 1 Graduate Institute of Biomedical Sciences, China Medical University, Taichung, Taiwan.,2 Organ Transplantation Center, China Medical University Hospital, Taichung, Taiwan
| | - Long-Bin Jeng
- 2 Organ Transplantation Center, China Medical University Hospital, Taichung, Taiwan
| | - Woei-Cherng Shyu
- 1 Graduate Institute of Biomedical Sciences, China Medical University, Taichung, Taiwan.,3 Translational Medicine Research Center and Department of Neurology, China Medical University Hospital, Taichung, Taiwan.,4 Department of Occupational Therapy, Asia University, Taichung, Taiwan
| |
Collapse
|
65
|
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: 128] [Impact Index Per Article: 21.3] [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
|
66
|
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: 0.2] [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
|
67
|
Fuentes E, Fuentes M, Alarcón M, Palomo I. Immune System Dysfunction in the Elderly. AN ACAD BRAS CIENC 2018; 89:285-299. [PMID: 28423084 DOI: 10.1590/0001-3765201720160487] [Citation(s) in RCA: 128] [Impact Index Per Article: 21.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2016] [Accepted: 12/29/2016] [Indexed: 02/07/2023] Open
Abstract
Human aging is characterized by both physical and physiological frailty that profoundly affects the immune system. In this context aging is associated with declines in adaptive and innate immunity established as immunosenescence. Immunosenescence is a new concept that reflects the age-associated restructuring changes of innate and adaptive immune functions. Thus elderly individuals usually present chronic low-level inflammation, higher infection rates and chronic diseases. A study of alterations in the immune system during aging could provide a potentially useful biomarker for the evaluation of immune senescence treatment. The immune system is the result of the interplay between innate and adaptive immunity, yet the impact of aging on this function is unclear. In this article the function of the immune system during aging is explored.
Collapse
Affiliation(s)
- Eduardo Fuentes
- Platelet Research Laboratory, Department of Clinical Biochemistry and Immunohematology, Faculty of Health Sciences, Interdisciplinary Excellence Research Program on Healthy Aging/ PIEI-ES, Universidad de Talca, Postal Code 3460000, Casilla 747, Talca, Chile.,Núcleo Científico Multidisciplinario, Universidad de Talca, Postal Code 3460000, Casilla 747, Talca, Chile
| | - Manuel Fuentes
- Platelet Research Laboratory, Department of Clinical Biochemistry and Immunohematology, Faculty of Health Sciences, Interdisciplinary Excellence Research Program on Healthy Aging/ PIEI-ES, Universidad de Talca, Postal Code 3460000, Casilla 747, Talca, Chile
| | - Marcelo Alarcón
- Platelet Research Laboratory, Department of Clinical Biochemistry and Immunohematology, Faculty of Health Sciences, Interdisciplinary Excellence Research Program on Healthy Aging/ PIEI-ES, Universidad de Talca, Postal Code 3460000, Casilla 747, Talca, Chile
| | - Iván Palomo
- Platelet Research Laboratory, Department of Clinical Biochemistry and Immunohematology, Faculty of Health Sciences, Interdisciplinary Excellence Research Program on Healthy Aging/ PIEI-ES, Universidad de Talca, Postal Code 3460000, Casilla 747, Talca, Chile
| |
Collapse
|
68
|
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: 9.7] [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
|
69
|
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: 80] [Impact Index Per Article: 13.3] [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]
|
70
|
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: 2.0] [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]
|
71
|
Hekmatimoghaddam S, Dehghani Firoozabadi A, Zare-Khormizi MR, Pourrajab F. Sirt1 and Parp1 as epigenome safeguards and microRNAs as SASP-associated signals, in cellular senescence and aging. Ageing Res Rev 2017; 40:120-141. [PMID: 28993289 DOI: 10.1016/j.arr.2017.10.001] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2017] [Revised: 09/25/2017] [Accepted: 10/05/2017] [Indexed: 01/25/2023]
Abstract
Cellular senescence (CS) is underlying mechanism of organism aging and is closely interconnected with age-related diseases (ARDs). Thus, any attempt that influences CS, may be undertaken to reverse or inhibit senescence, whereby could prolong healthy life span. Until now, two main proposes are epigenetic and genetic modifications of cell fate. The first one concerns rejuvenation through effective reprogramming in cells undergoing senescence, or derived from very old or progeroid patients, by which is effective in vitro in induced pluripotent stem cells (iPSCs). The second approach concerns modification of senescence signaling pathways like as IGF-induced agents. However, senescence research has experienced an unprecedented advance over recent years, particularly with the discovery that the rate of senescence is controlled, at least to some extent, by epigenetic pathways and biochemical processes conserved in evolution. In this review we try to concentrate on very specific pathways (DNA damage response, DDR, and epigenetic modifiers) and very specific determinants (senescence-associated secretory phenotype, SASP-miRNAs) of human premature aging. A major challenge is to dissect the interconnectedness between the candidate elements and their relative contributions to aging, with the final goal of identifying new opportunities for design of novel anti-aging treatments or avoidance of age-associated manifestations. While knowing that aging is unavoidable and we cannot expect its elimination, but prolonging healthy life span is a goal worth serious consideration.
Collapse
Affiliation(s)
- Seyedhossein Hekmatimoghaddam
- Yazd Cardiovascular Research Center, Shahid Sadoughi University of Medical Sciences, Yazd, Iran; Department of Laboratory Sciences, School of Paramedicine, Shahid Sadoughi University of Medical Sciences, Yazd, Iran.
| | | | | | - Fatemeh Pourrajab
- Department of Biochemistry and Molecular Biology, School of Medicine, Shahid Sadoughi University of Medical Sciences, Yazd, Iran.
| |
Collapse
|
72
|
Cardiac Aging – Benefits of Exercise, Nrf2 Activation and Antioxidant Signaling. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2017; 999:231-255. [DOI: 10.1007/978-981-10-4307-9_13] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
|
73
|
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: 1.3] [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
|
74
|
Sharifi-Sanjani M, Oyster NM, Tichy ED, Bedi KC, Harel O, Margulies KB, Mourkioti F. Cardiomyocyte-Specific Telomere Shortening is a Distinct Signature of Heart Failure in Humans. J Am Heart Assoc 2017; 6:JAHA.116.005086. [PMID: 28882819 PMCID: PMC5634248 DOI: 10.1161/jaha.116.005086] [Citation(s) in RCA: 70] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Background Telomere defects are thought to play a role in cardiomyopathies, but the specific cell type affected by the disease in human hearts is not yet identified. The aim of this study was to systematically evaluate the cell type specificity of telomere shortening in patients with heart failure in relation to their cardiac disease, age, and sex. Methods and Results We studied cardiac tissues from patients with heart failure by utilizing telomere quantitative fluorescence in situ hybridization, a highly sensitive method with single‐cell resolution. In this study, total of 63 human left ventricular samples, including 37 diseased and 26 nonfailing donor hearts, were stained for telomeres in combination with cardiomyocyte‐ or α‐smooth muscle cell‐specific markers, cardiac troponin T, and smooth muscle actin, respectively, and assessed for telomere length. Patients with heart failure demonstrate shorter cardiomyocyte telomeres compared with nonfailing donors, which is specific only to cardiomyocytes within diseased human hearts and is associated with cardiomyocyte DNA damage. Our data further reveal that hypertrophic hearts with reduced ejection fraction exhibit the shortest telomeres. In contrast to other reported cell types, no difference in cardiomyocyte telomere length is evident with age. However, under the disease state, telomere attrition manifests in both young and older patients with cardiac hypertrophy. Finally, we demonstrate that cardiomyocyte‐telomere length is better sustained in women than men under diseased conditions. Conclusions This study provides the first evidence of cardiomyocyte‐specific telomere shortening in heart failure.
Collapse
Affiliation(s)
| | - Nicholas M Oyster
- Department of Orthopaedic Surgery, University of Connecticut, Storrs, CT
| | - Elisia D Tichy
- Department of Orthopaedic Surgery, University of Connecticut, Storrs, CT
| | - Kenneth C Bedi
- Cardiovascular Institute, University of Connecticut, Storrs, CT
| | - Ofer Harel
- College of Liberal Arts and Sciences, Department of Statistics, University of Connecticut, Storrs, CT
| | | | - Foteini Mourkioti
- Department of Orthopaedic Surgery, University of Connecticut, Storrs, CT .,Cardiovascular Institute, University of Connecticut, Storrs, CT.,Department of Cell and Developmental Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| |
Collapse
|
75
|
Bellio MA, Pinto MT, Florea V, Barrios PA, Taylor CN, Brown AB, Lamondin C, Hare JM, Schulman IH, Rodrigues CO. Hypoxic Stress Decreases c-Myc Protein Stability in Cardiac Progenitor Cells Inducing Quiescence and Compromising Their Proliferative and Vasculogenic Potential. Sci Rep 2017; 7:9702. [PMID: 28851980 PMCID: PMC5575078 DOI: 10.1038/s41598-017-09813-x] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2016] [Accepted: 07/31/2017] [Indexed: 12/27/2022] Open
Abstract
Cardiac progenitor cells (CPCs) have been shown to promote cardiac regeneration and improve heart function. However, evidence suggests that their regenerative capacity may be limited in conditions of severe hypoxia. Elucidating the mechanisms involved in CPC protection against hypoxic stress is essential to maximize their cardioprotective and therapeutic potential. We investigated the effects of hypoxic stress on CPCs and found significant reduction in proliferation and impairment of vasculogenesis, which were associated with induction of quiescence, as indicated by accumulation of cells in the G0-phase of the cell cycle and growth recovery when cells were returned to normoxia. Induction of quiescence was associated with a decrease in the expression of c-Myc through mechanisms involving protein degradation and upregulation of p21. Inhibition of c-Myc mimicked the effects of severe hypoxia on CPC proliferation, also triggering quiescence. Surprisingly, these effects did not involve changes in p21 expression, indicating that other hypoxia-activated factors may induce p21 in CPCs. Our results suggest that hypoxic stress compromises CPC function by inducing quiescence in part through downregulation of c-Myc. In addition, we found that c-Myc is required to preserve CPC growth, suggesting that modulation of pathways downstream of it may re-activate CPC regenerative potential under ischemic conditions.
Collapse
Affiliation(s)
- Michael A Bellio
- Interdisciplinary Stem Cell Institute, University of Miami Miller School of Medicine, Miami, Florida, United States of America
| | - Mariana T Pinto
- Interdisciplinary Stem Cell Institute, University of Miami Miller School of Medicine, Miami, Florida, United States of America
| | - Victoria Florea
- Interdisciplinary Stem Cell Institute, University of Miami Miller School of Medicine, Miami, Florida, United States of America
| | - Paola A Barrios
- Interdisciplinary Stem Cell Institute, University of Miami Miller School of Medicine, Miami, Florida, United States of America
| | - Christy N Taylor
- Interdisciplinary Stem Cell Institute, University of Miami Miller School of Medicine, Miami, Florida, United States of America
| | - Ariel B Brown
- Interdisciplinary Stem Cell Institute, University of Miami Miller School of Medicine, Miami, Florida, United States of America
| | - Courtney Lamondin
- Interdisciplinary Stem Cell Institute, University of Miami Miller School of Medicine, Miami, Florida, United States of America
| | - Joshua M Hare
- Interdisciplinary Stem Cell Institute, University of Miami Miller School of Medicine, Miami, Florida, United States of America
- Department of Medicine, Cardiovascular Division, University of Miami Miller School of Medicine, Miami, Florida, United States of America
| | - Ivonne H Schulman
- Interdisciplinary Stem Cell Institute, University of Miami Miller School of Medicine, Miami, Florida, United States of America
- Department of Medicine, Katz Family Division of Nephrology and Hypertension, University of Miami Miller School of Medicine, Miami, Florida, United States of America
| | - Claudia O Rodrigues
- Interdisciplinary Stem Cell Institute, University of Miami Miller School of Medicine, Miami, Florida, United States of America.
- Department of Molecular and Cellular Pharmacology, University of Miami Miller School of Medicine, Miami, Florida, United States of America.
| |
Collapse
|
76
|
Sack MN, Fyhrquist FY, Saijonmaa OJ, Fuster V, Kovacic JC. Basic Biology of Oxidative Stress and the Cardiovascular System: Part 1 of a 3-Part Series. J Am Coll Cardiol 2017; 70:196-211. [PMID: 28683968 DOI: 10.1016/j.jacc.2017.05.034] [Citation(s) in RCA: 149] [Impact Index Per Article: 21.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/17/2017] [Revised: 04/24/2017] [Accepted: 05/10/2017] [Indexed: 01/18/2023]
Abstract
The generation of reactive oxygen species (ROS) is a fundamental aspect of normal human biology. However, when ROS generation exceeds endogenous antioxidant capacity, oxidative stress arises. If unchecked, ROS production and oxidative stress mediate tissue and cell damage that can spiral in a cycle of inflammation and more oxidative stress. This article is part 1 of a 3-part series covering the role of oxidative stress in cardiovascular disease. The broad theme of this first paper is the mechanisms and biology of oxidative stress. Specifically, the authors review the basic biology of oxidative stress, relevant aspects of mitochondrial function, and stress-related cell death pathways (apoptosis and necrosis) as they relate to the heart and cardiovascular system. They then explore telomere biology and cell senescence. As important regulators and sensors of oxidative stress, telomeres are segments of repetitive nucleotide sequence at each end of a chromosome that protect the chromosome ends from deterioration.
Collapse
Affiliation(s)
- Michael N Sack
- Cardiovascular and Pulmonary Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland.
| | | | | | - Valentin Fuster
- Zena and Michael A. Wiener Cardiovascular Institute, Icahn School of Medicine at Mount Sinai, New York, New York; Marie-Josée and Henry R. Kravis Cardiovascular Health Center, Icahn School of Medicine at Mount Sinai, New York, New York; Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
| | - Jason C Kovacic
- Zena and Michael A. Wiener Cardiovascular Institute, Icahn School of Medicine at Mount Sinai, New York, New York.
| |
Collapse
|
77
|
Gevaert AB, Shakeri H, Leloup AJ, Van Hove CE, De Meyer GRY, Vrints CJ, Lemmens K, Van Craenenbroeck EM. Endothelial Senescence Contributes to Heart Failure With Preserved Ejection Fraction in an Aging Mouse Model. Circ Heart Fail 2017; 10:CIRCHEARTFAILURE.116.003806. [PMID: 28611124 DOI: 10.1161/circheartfailure.116.003806] [Citation(s) in RCA: 114] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/20/2016] [Accepted: 05/01/2017] [Indexed: 12/17/2022]
Abstract
BACKGROUND Because of global aging, the prevalence of heart failure with preserved ejection fraction (HFpEF) continues to rise. Although HFpEF pathophysiology remains incompletely understood, endothelial inflammation is stated to play a central role. Cellular senescence is a process of cellular growth arrest linked with aging and inflammation. We used mice with accelerated aging to investigate the role of cellular senescence in HFpEF development. METHODS AND RESULTS Senescence-accelerated mice (SAM, n=18) and control mice with normal senescence (n=15) were fed normal chow or a high-fat, high-salt diet (WD). Vascular and cardiac function was assessed at 8, 16, and 24 weeks of age. At 24 weeks, both SAM on WD (SAM-WD) and SAM on regular diet displayed endothelial dysfunction, as evidenced by impaired acetylcholine-induced relaxation of aortic segments and reduced basal nitric oxide. At week 24, SAM-WD had developed HFpEF, characterized by diastolic dysfunction, left ventricular hypertrophy, left atrial dilatation, and interstitial fibrosis. Also, exercise capacity was reduced and lung weight increased. Cardiovascular inflammation and senescence were assessed by immunohistochemical and immunofluorescence staining of hearts and aortas. SAM-WD showed increased endothelial inflammation (intercellular adhesion molecule 1 expression) and increased endothelial senescence (acetyl-p53/CD31 costaining). The latter correlated with diastolic function and intercellular adhesion molecule 1 expression. CONCLUSIONS SAM develop endothelial dysfunction. Adding a high-salt, high-fat diet accelerates endothelial senescence and instigates endothelial inflammation. This coincides with hemodynamic and structural changes typical of HFpEF. Targeting endothelial senescence could be a new therapeutic avenue in HFpEF.
Collapse
Affiliation(s)
- Andreas B Gevaert
- From the Research Group Physiopharmacology (A.B.G., H.S., A.J.L., C.E.V.H., G.R.Y.D.M., K.L.), and Research Group Cardiovascular Diseases, Translational Pathophysiological Research (A.B.G., C.E.V.H., C.J.V., K.L., E.M.V.C.), University of Antwerp, Belgium; and Laboratory for Cellular and Molecular Cardiology and Department of Cardiology, Antwerp University Hospital (UZA), Edegem, Belgium (A.B.G., C.J.V., E.M.V.C.).
| | - Hadis Shakeri
- From the Research Group Physiopharmacology (A.B.G., H.S., A.J.L., C.E.V.H., G.R.Y.D.M., K.L.), and Research Group Cardiovascular Diseases, Translational Pathophysiological Research (A.B.G., C.E.V.H., C.J.V., K.L., E.M.V.C.), University of Antwerp, Belgium; and Laboratory for Cellular and Molecular Cardiology and Department of Cardiology, Antwerp University Hospital (UZA), Edegem, Belgium (A.B.G., C.J.V., E.M.V.C.)
| | - Arthur J Leloup
- From the Research Group Physiopharmacology (A.B.G., H.S., A.J.L., C.E.V.H., G.R.Y.D.M., K.L.), and Research Group Cardiovascular Diseases, Translational Pathophysiological Research (A.B.G., C.E.V.H., C.J.V., K.L., E.M.V.C.), University of Antwerp, Belgium; and Laboratory for Cellular and Molecular Cardiology and Department of Cardiology, Antwerp University Hospital (UZA), Edegem, Belgium (A.B.G., C.J.V., E.M.V.C.)
| | - Cor E Van Hove
- From the Research Group Physiopharmacology (A.B.G., H.S., A.J.L., C.E.V.H., G.R.Y.D.M., K.L.), and Research Group Cardiovascular Diseases, Translational Pathophysiological Research (A.B.G., C.E.V.H., C.J.V., K.L., E.M.V.C.), University of Antwerp, Belgium; and Laboratory for Cellular and Molecular Cardiology and Department of Cardiology, Antwerp University Hospital (UZA), Edegem, Belgium (A.B.G., C.J.V., E.M.V.C.)
| | - Guido R Y De Meyer
- From the Research Group Physiopharmacology (A.B.G., H.S., A.J.L., C.E.V.H., G.R.Y.D.M., K.L.), and Research Group Cardiovascular Diseases, Translational Pathophysiological Research (A.B.G., C.E.V.H., C.J.V., K.L., E.M.V.C.), University of Antwerp, Belgium; and Laboratory for Cellular and Molecular Cardiology and Department of Cardiology, Antwerp University Hospital (UZA), Edegem, Belgium (A.B.G., C.J.V., E.M.V.C.)
| | - Christiaan J Vrints
- From the Research Group Physiopharmacology (A.B.G., H.S., A.J.L., C.E.V.H., G.R.Y.D.M., K.L.), and Research Group Cardiovascular Diseases, Translational Pathophysiological Research (A.B.G., C.E.V.H., C.J.V., K.L., E.M.V.C.), University of Antwerp, Belgium; and Laboratory for Cellular and Molecular Cardiology and Department of Cardiology, Antwerp University Hospital (UZA), Edegem, Belgium (A.B.G., C.J.V., E.M.V.C.)
| | - Katrien Lemmens
- From the Research Group Physiopharmacology (A.B.G., H.S., A.J.L., C.E.V.H., G.R.Y.D.M., K.L.), and Research Group Cardiovascular Diseases, Translational Pathophysiological Research (A.B.G., C.E.V.H., C.J.V., K.L., E.M.V.C.), University of Antwerp, Belgium; and Laboratory for Cellular and Molecular Cardiology and Department of Cardiology, Antwerp University Hospital (UZA), Edegem, Belgium (A.B.G., C.J.V., E.M.V.C.)
| | - Emeline M Van Craenenbroeck
- From the Research Group Physiopharmacology (A.B.G., H.S., A.J.L., C.E.V.H., G.R.Y.D.M., K.L.), and Research Group Cardiovascular Diseases, Translational Pathophysiological Research (A.B.G., C.E.V.H., C.J.V., K.L., E.M.V.C.), University of Antwerp, Belgium; and Laboratory for Cellular and Molecular Cardiology and Department of Cardiology, Antwerp University Hospital (UZA), Edegem, Belgium (A.B.G., C.J.V., E.M.V.C.)
| |
Collapse
|
78
|
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: 54] [Impact Index Per Article: 7.7] [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
|
79
|
Booth SA, Charchar FJ. Cardiac telomere length in heart development, function, and disease. Physiol Genomics 2017; 49:368-384. [DOI: 10.1152/physiolgenomics.00024.2017] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Telomeres are repetitive nucleoprotein structures at chromosome ends, and a decrease in the number of these repeats, known as a reduction in telomere length (TL), triggers cellular senescence and apoptosis. Heart disease, the worldwide leading cause of death, often results from the loss of cardiac cells, which could be explained by decreases in TL. Due to the cell-specific regulation of TL, this review focuses on studies that have measured telomeres in heart cells and critically assesses the relationship between cardiac TL and heart function. There are several lines of evidence that have identified rapid changes in cardiac TL during the onset and progression of heart disease as well as at critical stages of development. There are also many factors, such as the loss of telomeric proteins, oxidative stress, and hypoxia, that decrease cardiac TL and heart function. In contrast, antioxidants, calorie restriction, and exercise can prevent both cardiac telomere attrition and the progression of heart disease. TL in the heart is also indicative of proliferative potential and could facilitate the identification of cells suitable for cardiac rejuvenation. Although these findings highlight the involvement of TL in heart function, there are important questions regarding the validity of animal models, as well as several confounding factors, that need to be considered when interpreting results and planning future research. With these in mind, elucidating the telomeric mechanisms involved in heart development and the transition to disease holds promise to prevent cardiac dysfunction and potentiate regeneration after injury.
Collapse
Affiliation(s)
- S. A. Booth
- Faculty of Science and Technology, School of Applied and Biomedical Sciences, Federation University Australia, Balllarat, Australia
| | - F. J. Charchar
- Faculty of Science and Technology, School of Applied and Biomedical Sciences, Federation University Australia, Balllarat, Australia
- Department of Physiology, The University of Melbourne, Melbourne, Australia; and
- Department of Cardiovascular Sciences, University of Leicester, Leicester, United Kingdom
| |
Collapse
|
80
|
Frustaci A, Chimenti C, Doheny D, Desnick RJ. Evolution of cardiac pathology in classic Fabry disease: Progressive cardiomyocyte enlargement leads to increased cell death and fibrosis, and correlates with severity of ventricular hypertrophy. Int J Cardiol 2017; 248:257-262. [PMID: 28688718 DOI: 10.1016/j.ijcard.2017.06.079] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/16/2017] [Revised: 06/15/2017] [Accepted: 06/20/2017] [Indexed: 11/28/2022]
Abstract
BACKGROUND Fabry disease, an X-linked lysosomal storage disease, results from deficient α-galactosidase A (α-GalA) activity and the systemic accumulation of α-galactosyl-terminated glycosphingolipids. Two major phenotypes, "Classic" and "Later-Onset", lead to renal failure, and/or cardiac disease, and early demise. To date, the evolution and progression of the cardiac pathology and resultant clinical manifestations in family members of phenotype have not been well characterized. METHODS AND RESULTS In a Classic family with nine affected members (GLA mutation c.983delG), cardiac imaging, angiography, and cardiac biopsies were performed in four males and two heterozygous females. Tissues were examined histologically, ultrastructurally, and myocardial necrosis and apoptosis were evaluated by in situ ligation with hairpin probes. Increasing cardiac pathology correlated with ECG and cardiac magnetic resonance findings. Young affected males with "pre-hypertrophy" had 18-20μm cardiomyocyte diameters, <30% vacuolar areas in myocytes, and normal levels of necrosis and apoptosis. Patients with "moderate hypertrophy" (maximal wall thickness (MWT) ≤16mm) had 30-35μm cardiomyocyte diameters, ~45% vacuolar areas, and moderate levels of necrosis and apoptosis. In contrast, the oldest male with severe hypertrophy (MWT=21mm) had 38-40μm cell diameters, >60% vacuolar areas, and marked necrosis and apoptosis. CONCLUSION Progressive gender-specific cardiac pathology and clinical manifestations were documented in affected Classic family members. Increasing cardiomyocyte diameter was correlated with disease severity, age, and gender. Fibrosis was presumably caused by cell death of enlarged, substrate-engorged cardiomyocytes. These results support early enzyme therapy in Classic males to prevent/minimize irreversible cardiac damage.
Collapse
Affiliation(s)
- Andrea Frustaci
- Department of Cardiovascular, Respiratory, Nephrologic, Geriatric and Anesthesiologic Sciences, La Sapienza University, Rome, Italy
| | - Cristina Chimenti
- Department of Cardiovascular, Respiratory, Nephrologic, Geriatric and Anesthesiologic Sciences, La Sapienza University, Rome, Italy
| | - Dana Doheny
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Robert J Desnick
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, United States.
| |
Collapse
|
81
|
Cesselli D, Aleksova A, Sponga S, Cervellin C, Di Loreto C, Tell G, Beltrami AP. Cardiac Cell Senescence and Redox Signaling. Front Cardiovasc Med 2017; 4:38. [PMID: 28612009 PMCID: PMC5447053 DOI: 10.3389/fcvm.2017.00038] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2017] [Accepted: 05/10/2017] [Indexed: 12/12/2022] Open
Abstract
Aging is characterized by a progressive loss of the ability of the organism to cope with stressors and to repair tissue damage. As a result, chronic diseases, including cardiovascular disease, increase their prevalence with aging, underlining the existence of common mechanisms that lead to frailty and age-related diseases. In this frame, the progressive decline of the homeostatic and reparative function of primitive cells has been hypothesized to play a major role in the evolution of cardiac pathology to heart failure. Although initially it was believed that reactive oxygen species (ROS) were produced in an unregulated manner as a byproduct of cellular metabolism, causing macromolecular damage and aging, accumulating evidence indicate the major role played by redox signaling in physiology. Aim of this review is to critically revise evidence linking ROS to cell senescence and aging and to provide evidence of the primary role played by redox signaling, with a particular emphasis on the multifunctional protein APE1/Ref in stem cell biology. Finally, we will discuss evidence supporting the role of redox signaling in cardiovascular cells.
Collapse
Affiliation(s)
| | - Aneta Aleksova
- Cardiovascular Department, Azienda Sanitaria Universitaria Integrata di Trieste, University of Trieste, Trieste, Italy
| | - Sandro Sponga
- Cardiothoracic Surgery, Azienda Sanitaria Universitaria Integrata di Udine, Udine, Italy
| | | | | | - Gianluca Tell
- Department of Medicine, University of Udine, Udine, Italy
| | | |
Collapse
|
82
|
Monoamine Oxidases, Oxidative Stress, and Altered Mitochondrial Dynamics in Cardiac Ageing. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2017; 2017:3017947. [PMID: 28546851 PMCID: PMC5435992 DOI: 10.1155/2017/3017947] [Citation(s) in RCA: 70] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/06/2017] [Revised: 02/22/2017] [Accepted: 03/22/2017] [Indexed: 01/06/2023]
Abstract
The advances in healthcare over the past several decades have resulted in populations now living longer. With this increase in longevity, a wider prevalence of cardiovascular diseases is more common and known to be a major factor in rising healthcare costs. A wealth of scientific evidence has implicated cell senescence as an important component in the etiology of these age-dependent pathologies. A number of studies indicate that an excess of reactive oxygen species (ROS) contributes to trigger and accelerate the cardiac senescence processes, and a new role of monoamine oxidases, MAO-A and MAO-B, is emerging in this context. These mitochondrial enzymes regulate the level of catecholamines and serotonin by catalyzing their oxidative deamination in the heart. MAOs' expression substantially increases with ageing (6-fold MAO-A in the heart and 4-fold MAO-B in neuronal tissue), and their involvement in cardiac diseases is supposedly related to the formation of ROS, via the hydrogen peroxide produced during the substrate degradation. Here, we will review the most recent advances in this field and describe why MAOs could be effective targets in order to prevent age-associated cardiovascular disease.
Collapse
|
83
|
Chimenti C, Verardo R, Scopelliti F, Grande C, Petrosillo N, Piselli P, De Paulis R, Frustaci A. Myocardial expression of Toll-like receptor 4 predicts the response to immunosuppressive therapy in patients with virus-negative chronic inflammatory cardiomyopathy. Eur J Heart Fail 2017; 19:915-925. [DOI: 10.1002/ejhf.796] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/22/2016] [Revised: 01/22/2017] [Accepted: 01/30/2017] [Indexed: 12/21/2022] Open
Affiliation(s)
- Cristina Chimenti
- Cardiovascular, Respiratory, Nephrologic, Anesthesiologic and Geriatric Sciences; La Sapienza University; Rome Italy
- Cellular and Molecular Cardiology Laboratory, IRCCS INMI L. Spallanzani; Rome Italy
| | - Romina Verardo
- Cellular and Molecular Cardiology Laboratory, IRCCS INMI L. Spallanzani; Rome Italy
| | | | - Claudia Grande
- Cardiovascular, Respiratory, Nephrologic, Anesthesiologic and Geriatric Sciences; La Sapienza University; Rome Italy
| | - Nicola Petrosillo
- Clinical and Research Department, IRCCS INMI L. Spallanzani; Rome Italy
| | - Pierluca Piselli
- Unit of Clinical Epidemiology, IRCCS INMI L. Spallanzani; Rome Italy
| | | | - Andrea Frustaci
- Cardiovascular, Respiratory, Nephrologic, Anesthesiologic and Geriatric Sciences; La Sapienza University; Rome Italy
- Cellular and Molecular Cardiology Laboratory, IRCCS INMI L. Spallanzani; Rome Italy
| |
Collapse
|
84
|
D'Amario D, Leone AM, Narducci ML, Smaldone C, Lecis D, Inzani F, Luciani M, Siracusano A, La Neve F, Manchi M, Pelargonio G, Perna F, Bruno P, Massetti M, Pitocco D, Cappetta D, Esposito G, Urbanek K, De Angelis A, Rossi F, Piacentini R, Angelini G, Li Puma DD, Grassi C, De Paolis E, Capoluongo E, Silvestri V, Merlino B, Marano R, Crea F. Human cardiac progenitor cells with regenerative potential can be isolated and characterized from 3D-electro-anatomic guided endomyocardial biopsies. Int J Cardiol 2017; 241:330-343. [PMID: 28343765 DOI: 10.1016/j.ijcard.2017.02.106] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/06/2016] [Revised: 02/01/2017] [Accepted: 02/21/2017] [Indexed: 01/26/2023]
Abstract
AIMS In the present study, we aimed to develop a percutaneous approach and a reproducible methodology for the isolation and expansion of Cardiac Progenitor Cells (CPCs) from EndoMyocardial Biopsies (EMB) in vivo. Moreover, in an animal model of non-ischemic heart failure (HF), we would like to test whether CPCs obtained by this methodology may engraft the myocardium and differentiate. METHODS AND RESULTS EMB were obtained using a preformed sheath and a disposable bioptome, advanced via right femoral vein in 12 healthy mini pigs, to the right ventricle. EMB were enzymatically dissociated, cells were expanded and sorted for c-kit. We used 3D-Electro-Anatomic Mapping (3D-EAM) to obtain CPCs from 32 patients affected by non-ischemic cardiomyopathy. The in vivo regenerative potential of CPCs was tested in a rodent model of drug-induced non-ischemic cardiomyopathy. c-kit positive CPCs replicative capacity was assessed in 30 patients. Telomere length averaged 7.4±0.4kbp and telomerase activity was present in all preparations (1.7×105 copies). The in situ hybridization experiments showed that injected human CPCs may acquire a neonatal myocyte phenotype given the expression of the alpha-sarcomeric actin together with the presence of the Alu probe, suggesting a beneficial impact on LV performance. CONCLUSIONS The success in obtaining CPCs characterized by high regenerative potential, in vitro and in vivo, from EMB indicates that harvesting without thoracotomy in patients affected by either ischemic or non-ischemic cardiomyopathy is feasible. These initial results may potentially expand the future application of CPCs to all patients affected by HF not undergoing surgical procedures.
Collapse
Affiliation(s)
- Domenico D'Amario
- Department of Cardiovascular Medicine, Catholic University of the Sacred Heart, Rome, Italy.
| | - Antonio Maria Leone
- Department of Cardiovascular Medicine, Catholic University of the Sacred Heart, Rome, Italy
| | - Maria Lucia Narducci
- Department of Cardiovascular Medicine, Catholic University of the Sacred Heart, Rome, Italy
| | - Costantino Smaldone
- Department of Cardiovascular Medicine, Catholic University of the Sacred Heart, Rome, Italy
| | - Dalgisio Lecis
- Department of Cardiovascular Medicine, Catholic University of the Sacred Heart, Rome, Italy
| | - Frediano Inzani
- Department of Pathology, Catholic University of the Sacred Heart, Rome, Italy
| | - Marco Luciani
- Department of Cardiovascular Medicine, Catholic University of the Sacred Heart, Rome, Italy
| | - Andrea Siracusano
- Department of Cardiovascular Medicine, Catholic University of the Sacred Heart, Rome, Italy
| | - Federica La Neve
- Department of Cardiovascular Medicine, Catholic University of the Sacred Heart, Rome, Italy
| | - Melissa Manchi
- Department of Cardiovascular Medicine, Catholic University of the Sacred Heart, Rome, Italy
| | - Gemma Pelargonio
- Department of Cardiovascular Medicine, Catholic University of the Sacred Heart, Rome, Italy
| | - Francesco Perna
- Department of Cardiovascular Medicine, Catholic University of the Sacred Heart, Rome, Italy
| | - Piergiorgio Bruno
- Department of Cardiovascular Medicine, Catholic University of the Sacred Heart, Rome, Italy
| | - Massimo Massetti
- Department of Cardiovascular Medicine, Catholic University of the Sacred Heart, Rome, Italy
| | - Dario Pitocco
- Institute of Internal Medicine and Diabetes Care Unit, Catholic University of the Sacred Heart, Rome, Italy
| | - Donato Cappetta
- Department of Experimental Medicine, Section of Pharmacology, Università della Campania "Luigi Vanvitelli", Naples, Italy
| | - Grazia Esposito
- Department of Experimental Medicine, Section of Pharmacology, Università della Campania "Luigi Vanvitelli", Naples, Italy
| | - Konrad Urbanek
- Department of Experimental Medicine, Section of Pharmacology, Università della Campania "Luigi Vanvitelli", Naples, Italy
| | - Antonella De Angelis
- Department of Experimental Medicine, Section of Pharmacology, Università della Campania "Luigi Vanvitelli", Naples, Italy
| | - Francesco Rossi
- Department of Experimental Medicine, Section of Pharmacology, Università della Campania "Luigi Vanvitelli", Naples, Italy
| | - Roberto Piacentini
- Institute of Human Physiology, Catholic University of the Sacred Heart, Rome, Italy
| | - Giulia Angelini
- Department of Cardiovascular Medicine, Catholic University of the Sacred Heart, Rome, Italy
| | | | - Claudio Grassi
- Institute of Human Physiology, Catholic University of the Sacred Heart, Rome, Italy; San Raffaele Pisana Scientific Institute for Research, Hospitalization and Health Care, Rome, Italy
| | - Elisa De Paolis
- Laboratory of Molecular Biology, Institute of Biochemistry and Clinical Biochemistry, Catholic University of the Sacred Heart, Rome, Italy
| | - Ettore Capoluongo
- Laboratory of Molecular Biology, Institute of Biochemistry and Clinical Biochemistry, Catholic University of the Sacred Heart, Rome, Italy
| | - Valentina Silvestri
- Department of Radiological Sciences, Institute of Radiology, Catholic University of the Sacred Heart, Rome, Italy
| | - Biagio Merlino
- Department of Radiological Sciences, Institute of Radiology, Catholic University of the Sacred Heart, Rome, Italy
| | - Riccardo Marano
- Department of Radiological Sciences, Institute of Radiology, Catholic University of the Sacred Heart, Rome, Italy
| | - Filippo Crea
- Department of Cardiovascular Medicine, Catholic University of the Sacred Heart, Rome, Italy.
| |
Collapse
|
85
|
Kannappan R, Matsuda A, Ferreira-Martins J, Zhang E, Palano G, Czarna A, Cabral-Da-Silva MC, Bastos-Carvalho A, Sanada F, Ide N, Rota M, Blasco MA, Serrano M, Anversa P, Leri A. p53 Modulates the Fate of Cardiac Progenitor Cells Ex Vivo and in the Diabetic Heart In Vivo. EBioMedicine 2017; 16:224-237. [PMID: 28163043 PMCID: PMC5474510 DOI: 10.1016/j.ebiom.2017.01.028] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2016] [Revised: 01/20/2017] [Accepted: 01/20/2017] [Indexed: 12/01/2022] Open
Abstract
p53 is an important modulator of stem cell fate, but its role in cardiac progenitor cells (CPCs) is unknown. Here, we tested the effects of a single extra-copy of p53 on the function of CPCs in the presence of oxidative stress mediated by doxorubicin in vitro and type-1 diabetes in vivo. CPCs were obtained from super-p53 transgenic mice (p53-tg), in which the additional allele is regulated in a manner similar to the endogenous protein. Old CPCs with increased p53 dosage showed a superior ability to sustain oxidative stress, repair DNA damage and restore cell division. With doxorubicin, a larger fraction of CPCs carrying an extra-copy of the p53 allele recruited γH2A.X reestablishing DNA integrity. Enhanced p53 expression resulted in a superior tolerance to oxidative stress in vivo by providing CPCs with defense mechanisms necessary to survive in the milieu of the diabetic heart; they engrafted in regions of tissue injury and in three days acquired the cardiomyocyte phenotype. The biological advantage provided by the increased dosage of p53 in CPCs suggests that this genetic strategy may be translated to humans to increase cellular engraftment and growth, critical determinants of successful cell therapy for the failing heart. p53 improves the ability of CPCs to sustain oxidative stress. p53 promotes the restoration of DNA integrity and cell division. p53 enhances the engraftment of CPCs in the diabetic heart.
Ongoing clinical trials with autologous cardiac stem cells (CSCs) are faced with a critical limitation which is related to the modest amount of retained cells within the damaged myocardium. We have developed a strategy that overcomes in part this problem enhancing the number of CSCs able to engraft within the pathologic organ. Additionally, these genetically modified CSCs can be generated in large number, raising the possibility that multiple temporally distinct deliveries of cells can be introduced to restore the structural and functional integrity of the decompensated heart.
Collapse
Affiliation(s)
- Ramaswamy Kannappan
- Departments of Anesthesia and Medicine, and Division of Cardiovascular Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Alex Matsuda
- Departments of Anesthesia and Medicine, and Division of Cardiovascular Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA; Cardiocentro Ticino Foundation, Swiss Institute for Regenerative Medicine (SIRM), Via Tesserete 48, 6900 Lugano, Switzerland
| | - João Ferreira-Martins
- Departments of Anesthesia and Medicine, and Division of Cardiovascular Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Eric Zhang
- Departments of Anesthesia and Medicine, and Division of Cardiovascular Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Giorgia Palano
- Departments of Anesthesia and Medicine, and Division of Cardiovascular Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Anna Czarna
- Departments of Anesthesia and Medicine, and Division of Cardiovascular Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA; Cardiocentro Ticino Foundation, Swiss Institute for Regenerative Medicine (SIRM), Via Tesserete 48, 6900 Lugano, Switzerland
| | - Mauricio Castro Cabral-Da-Silva
- Departments of Anesthesia and Medicine, and Division of Cardiovascular Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Adriana Bastos-Carvalho
- Departments of Anesthesia and Medicine, and Division of Cardiovascular Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Fumihiro Sanada
- Departments of Anesthesia and Medicine, and Division of Cardiovascular Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Noriko Ide
- Departments of Anesthesia and Medicine, and Division of Cardiovascular Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Marcello Rota
- Departments of Anesthesia and Medicine, and Division of Cardiovascular Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Maria A Blasco
- Spanish National Cancer Research Centre (CNIO), Madrid E-28029, Spain
| | - Manuel Serrano
- Spanish National Cancer Research Centre (CNIO), Madrid E-28029, Spain
| | - Piero Anversa
- Departments of Anesthesia and Medicine, and Division of Cardiovascular Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA; Cardiocentro Ticino Foundation, Swiss Institute for Regenerative Medicine (SIRM), Via Tesserete 48, 6900 Lugano, Switzerland
| | - Annarosa Leri
- Departments of Anesthesia and Medicine, and Division of Cardiovascular Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA; Cardiocentro Ticino Foundation, Swiss Institute for Regenerative Medicine (SIRM), Via Tesserete 48, 6900 Lugano, Switzerland.
| |
Collapse
|
86
|
Der Sarkissian S, Lévesque T, Noiseux N. Optimizing stem cells for cardiac repair: Current status and new frontiers in regenerative cardiology. World J Stem Cells 2017; 9:9-25. [PMID: 28154736 PMCID: PMC5253186 DOI: 10.4252/wjsc.v9.i1.9] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/01/2016] [Revised: 09/20/2016] [Accepted: 10/24/2016] [Indexed: 02/06/2023] Open
Abstract
Cell therapy has the potential to improve healing of ischemic heart, repopulate injured myocardium and restore cardiac function. The tremendous hope and potential of stem cell therapy is well understood, yet recent trials involving cell therapy for cardiovascular diseases have yielded mixed results with inconsistent data thereby readdressing controversies and unresolved questions regarding stem cell efficacy for ischemic cardiac disease treatment. These controversies are believed to arise by the lack of uniformity of the clinical trial methodologies, uncertainty regarding the underlying reparative mechanisms of stem cells, questions concerning the most appropriate cell population to use, the proper delivery method and timing in relation to the moment of infarction, as well as the poor stem cell survival and engraftment especially in a diseased microenvironment which is collectively acknowledged as a major hindrance to any form of cell therapy. Indeed, the microenvironment of the failing heart exhibits pathological hypoxic, oxidative and inflammatory stressors impairing the survival of transplanted cells. Therefore, in order to observe any significant therapeutic benefit there is a need to increase resilience of stem cells to death in the transplant microenvironment while preserving or better yet improving their reparative functionality. Although stem cell differentiation into cardiomyocytes has been observed in some instance, the prevailing reparative benefits are afforded through paracrine mechanisms that promote angiogenesis, cell survival, transdifferentiate host cells and modulate immune responses. Therefore, to maximize their reparative functionality, ex vivo manipulation of stem cells through physical, genetic and pharmacological means have shown promise to enable cells to thrive in the post-ischemic transplant microenvironment. In the present work, we will overview the current status of stem cell therapy for ischemic heart disease, discuss the most recurring cell populations employed, the mechanisms by which stem cells deliver a therapeutic benefit and strategies that have been used to optimize and increase survival and functionality of stem cells including ex vivo preconditioning with drugs and a novel “pharmaco-optimizer” as well as genetic modifications.
Collapse
|
87
|
Shen L, Wang H, Bei Y, Cretoiu D, Cretoiu SM, Xiao J. Formation of New Cardiomyocytes in Exercise. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2017; 999:91-102. [DOI: 10.1007/978-981-10-4307-9_6] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
|
88
|
Abstract
Telomeres are tandem repeat DNA sequences present at the ends of each eukaryotic chromosome to stabilize the genome structure integrity. Telomere lengths progressively shorten with each cell division. Inflammation and oxidative stress, which are implicated as major mechanisms underlying cardiovascular diseases, increase the rate of telomere shortening and lead to cellular senescence. In clinical studies, cardiovascular risk factors such as smoking, obesity, sedentary lifestyle, and hypertension have been associated with short leukocyte telomere length. In addition, low telomerase activity and short leukocyte telomere length have been observed in atherosclerotic plaque and associated with plaque instability, thus stroke or acute myocardial infarction. The aging myocardium with telomere shortening and accumulation of senescent cells limits the tissue regenerative capacity, contributing to systolic or diastolic heart failure. In addition, patients with ion-channel defects might have genetic imbalance caused by oxidative stress-related accelerated telomere shortening, which may subsequently cause sudden cardiac death. Telomere length can serve as a marker for the biological status of previous cell divisions and DNA damage with inflammation and oxidative stress. It can be integrated into current risk prediction and stratification models for cardiovascular diseases and can be used in precise personalized treatments. In this review, we summarize the current understanding of telomeres and telomerase in the aging process and their association with cardiovascular diseases. In addition, we discuss therapeutic interventions targeting the telomere system in cardiovascular disease treatments.
Collapse
|
89
|
Yeh JK, Wang CY. Telomeres and Telomerase in Cardiovascular Diseases. Genes (Basel) 2016; 7:genes7090058. [PMID: 27598203 PMCID: PMC5042389 DOI: 10.3390/genes7090058] [Citation(s) in RCA: 126] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2016] [Revised: 08/25/2016] [Accepted: 08/29/2016] [Indexed: 12/20/2022] Open
Abstract
Telomeres are tandem repeat DNA sequences present at the ends of each eukaryotic chromosome to stabilize the genome structure integrity. Telomere lengths progressively shorten with each cell division. Inflammation and oxidative stress, which are implicated as major mechanisms underlying cardiovascular diseases, increase the rate of telomere shortening and lead to cellular senescence. In clinical studies, cardiovascular risk factors such as smoking, obesity, sedentary lifestyle, and hypertension have been associated with short leukocyte telomere length. In addition, low telomerase activity and short leukocyte telomere length have been observed in atherosclerotic plaque and associated with plaque instability, thus stroke or acute myocardial infarction. The aging myocardium with telomere shortening and accumulation of senescent cells limits the tissue regenerative capacity, contributing to systolic or diastolic heart failure. In addition, patients with ion-channel defects might have genetic imbalance caused by oxidative stress-related accelerated telomere shortening, which may subsequently cause sudden cardiac death. Telomere length can serve as a marker for the biological status of previous cell divisions and DNA damage with inflammation and oxidative stress. It can be integrated into current risk prediction and stratification models for cardiovascular diseases and can be used in precise personalized treatments. In this review, we summarize the current understanding of telomeres and telomerase in the aging process and their association with cardiovascular diseases. In addition, we discuss therapeutic interventions targeting the telomere system in cardiovascular disease treatments.
Collapse
Affiliation(s)
- Jih-Kai Yeh
- Department of Cardiology, Chang Gung Memorial Hospital, 33305 Taoyuan, Taiwan.
| | - Chao-Yung Wang
- Department of Cardiology, Chang Gung Memorial Hospital, 33305 Taoyuan, Taiwan.
- Chang Gung University College of Medicine, 33302 Taoyuan, Taiwan.
| |
Collapse
|
90
|
Gianfranceschi G, Caragnano A, Piazza S, Manini I, Ciani Y, Verardo R, Toffoletto B, Finato N, Livi U, Beltrami CA, Scoles G, Sinagra G, Aleksova A, Cesselli D, Beltrami AP. Critical role of lysosomes in the dysfunction of human Cardiac Stem Cells obtained from failing hearts. Int J Cardiol 2016; 216:140-50. [DOI: 10.1016/j.ijcard.2016.04.155] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/06/2015] [Revised: 04/10/2016] [Accepted: 04/20/2016] [Indexed: 12/01/2022]
|
91
|
Nakayama H, Nishida K, Otsu K. Macromolecular Degradation Systems and Cardiovascular Aging. Circ Res 2016; 118:1577-92. [DOI: 10.1161/circresaha.115.307495] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/28/2015] [Accepted: 02/29/2016] [Indexed: 11/16/2022]
Abstract
Aging-related cardiovascular diseases are a rapidly increasing problem worldwide. Cardiac aging demonstrates progressive decline of diastolic dysfunction of ventricle and increase in ventricular and arterial stiffness accompanied by increased fibrosis stimulated by angiotensin II and proinflammatory cytokines. Reactive oxygen species and multiple signaling pathways on cellular senescence play major roles in the process. Aging is also associated with an alteration in steady state of macromolecular dynamics including a dysfunction of protein synthesis and degradation. Currently, impaired macromolecular degradation is considered to be closely related to enhanced inflammation and be involved in the process and mechanism of cardiac aging. Herein, we review the role and mechanisms of the degradation system of intracellular macromolecules in the process and pathophysiology of cardiovascular aging.
Collapse
Affiliation(s)
- Hiroyuki Nakayama
- From the Laboratory of Clinical Science and Biomedicine, Graduate School of Pharmaceutical Sciences, Osaka University, Suita, Osaka, Japan (H.N.); and Cardiovascular Division, King’s College London British Heart Foundation Centre of Research Excellence, London, United Kingdom (K.N., K.O.)
| | - Kazuhiko Nishida
- From the Laboratory of Clinical Science and Biomedicine, Graduate School of Pharmaceutical Sciences, Osaka University, Suita, Osaka, Japan (H.N.); and Cardiovascular Division, King’s College London British Heart Foundation Centre of Research Excellence, London, United Kingdom (K.N., K.O.)
| | - Kinya Otsu
- From the Laboratory of Clinical Science and Biomedicine, Graduate School of Pharmaceutical Sciences, Osaka University, Suita, Osaka, Japan (H.N.); and Cardiovascular Division, King’s College London British Heart Foundation Centre of Research Excellence, London, United Kingdom (K.N., K.O.)
| |
Collapse
|
92
|
Raymond AR, Becker J, Woodiwiss AJ, Booysen HL, Norton GR, Brooksbank RL. Ethanol-Associated Cardiomyocyte Apoptosis and Left Ventricular Dilation Are Unrelated to Changes in Myocardial Telomere Length in Rats. J Card Fail 2016; 22:294-302. [DOI: 10.1016/j.cardfail.2015.06.013] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2015] [Revised: 04/07/2015] [Accepted: 06/15/2015] [Indexed: 12/26/2022]
|
93
|
Cui J, Zhang F, Wang Y, Liu J, Ming X, Hou J, Lv B, Fang S, Yu B. Macrophage migration inhibitory factor promotes cardiac stem cell proliferation and endothelial differentiation through the activation of the PI3K/Akt/mTOR and AMPK pathways. Int J Mol Med 2016; 37:1299-309. [PMID: 27035848 PMCID: PMC4829139 DOI: 10.3892/ijmm.2016.2542] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2015] [Accepted: 03/16/2016] [Indexed: 01/05/2023] Open
Abstract
Macrophage migration inhibitory factor (MIF) has pleiotropic immune functions in a number of inflammatory diseases. Recent evidence from expression and functional studies has indicated that MIF is involved in various aspects of cardiovascular disease. In this study, we aimed to determine whether MIF supports in vitro c-kit+CD45− cardiac stem cell (CSC) survival, proliferation and differentiation into endothelial cells, as well as the possible mechanisms involved. We observed MIF receptor (CD74) expression in mouse CSCs (mCSCs) using PCR and immunofluorescence staining, and MIF secretion by mCSCs using PCR and ELISA in vitro. Increasing amounts of exogenous MIF did not affect CD74 expression, but promoted mCSC survival, proliferation and endothelial differentiation. By contrast, treatment with an MIF inhibitor (ISO-1) or siRNA targeting CD74 (CD74-siRNA) suppressed the biological changes induced by MIF in the mCSCs. Increasing amounts of MIF increased the phosphorylation of Akt and mammalian target of rapamycin (mTOR), which are known to support cell survival, proliferation and differentiation. These effects of MIF on the mCSCs were abolished by LY294002 [a phosphoinositide 3-kinase (PI3K) inhibitor] and MK-2206 (an Akt inhibitor). Moreover, adenosine monophosphate-activated protein kinase (AMPK) phosphorylation increased following treatment with MIF. The AMPK inhibitor, compound C, partly blocked the pro-proliferative effects of MIF on the mCSCs. In conclusion, our results suggest that MIF promotes mCSC survival, proliferation and endothelial differentiation through the activation of the PI3K/Akt/mTOR and AMPK signaling pathways. Thus, MIF may prove to be a potential therapeutic factor in the treatment of heart failure and myocardial infarction by activating CSCs.
Collapse
Affiliation(s)
- Jinjin Cui
- Department of Cardiology, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150081, P.R. China
| | - Fengyun Zhang
- Department of Cardiology, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150081, P.R. China
| | - Yongshun Wang
- Department of Cardiology, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150081, P.R. China
| | - Jingjin Liu
- Department of Cardiology, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150081, P.R. China
| | - Xing Ming
- Department of Cardiology, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150081, P.R. China
| | - Jingbo Hou
- Department of Cardiology, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150081, P.R. China
| | - Bo Lv
- Department of Cardiology, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150081, P.R. China
| | - Shaohong Fang
- The Key Laboratory of Myocardial Ischemia, Chinese Ministry of Education, Harbin, Heilongjiang 150081, P.R. China
| | - Bo Yu
- Department of Cardiology, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150081, P.R. China
| |
Collapse
|
94
|
Abstract
Following the recognition that hematopoietic stem cells improve the outcome of myocardial infarction in animal models, bone marrow mononuclear cells, CD34-positive cells, and mesenchymal stromal cells have been introduced clinically. The intracoronary or intramyocardial injection of these cell classes has been shown to be safe and to produce a modest but significant enhancement in systolic function. However, the identification of resident cardiac stem cells in the human heart (hCSCs) has created great expectation concerning the potential implementation of this category of autologous cells for the management of the human disease. Although phase 1 clinical trials have been conducted with encouraging results, the search for the most powerful hCSC for myocardial regeneration is in its infancy. This manuscript discusses the efforts performed in our laboratory to characterize the critical biological variables that define the growth reserve of hCSCs. Based on the theory of the immortal DNA template, we propose that stem cells retaining the old DNA represent 1 of the most powerful cells for myocardial regeneration. Similarly, the expression of insulin-like growth factor-1 receptors in hCSCs recognizes a cell phenotype with superior replicating reserve. However, the impressive recovery in ventricular hemodynamics and anatomy mediated by clonal hCSCs carrying the "mother" DNA underscores the clinical relevance of this hCSC class for the treatment of human heart failure.
Collapse
|
95
|
Abstract
Much has changed since our survey of the landscape for myocardial regeneration powered by adult stem cells 4 years ago.(1) The intervening years since that first review has witnessed an explosive expansion of studies that advance both understanding and implementation of adult stem cells in promoting myocardial repair. Painstaking research from innumerable laboratories throughout the world is prying open doors that may lead to restoration of myocardial structure and function in the wake of pathological injury. This global effort has produced deeper mechanistic comprehension coupled with an evolving appreciation for the complexity of myocardial regeneration in the adult context. Undaunted by both known and (as yet) unknown challenges, pursuit of myocardial regenerative medicine mediated by adult stem cell therapy has gathered momentum fueled by tantalizing clues and visionary goals. This concise review takes a somewhat different perspective than our initial treatise, taking stock of the business sector that has become an integral part of the field while concurrently updating state of affairs in cutting edge research. Looking retrospectively at advancement over the years as all reviews eventually must, the fundamental lesson to be learned is best explained by Jonatan Mårtensson: "Success will never be a big step in the future. Success is a small step taken just now."
Collapse
Affiliation(s)
- Kathleen M Broughton
- From the San Diego State University Heart Institute and the Integrated Regenerative Research Institute, San Diego, CA
| | - Mark A Sussman
- From the San Diego State University Heart Institute and the Integrated Regenerative Research Institute, San Diego, CA.
| |
Collapse
|
96
|
De Angelis A, Urbanek K, Cappetta D, Piegari E, Ciuffreda LP, Rivellino A, Russo R, Esposito G, Rossi F, Berrino L. Doxorubicin cardiotoxicity and target cells: a broader perspective. CARDIO-ONCOLOGY (LONDON, ENGLAND) 2016; 2:2. [PMID: 33530140 PMCID: PMC7837148 DOI: 10.1186/s40959-016-0012-4] [Citation(s) in RCA: 44] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 02/08/2016] [Accepted: 02/18/2016] [Indexed: 12/12/2022]
Abstract
The cardiotoxicity of doxorubicin is becoming an interdisciplinary point of interest given a growing population of cancer survivors. The complex and not completely understood pathogenesis of this complication makes difficult to design successful preventive or curative measures. Although cardiomyocyte has been considered a classical cellular target, other cells including various types of undifferentiated cells are involved in myocardial homeostasis. Such perspective may shed light on previously unrecognized aspects of cardiotoxicity and promote new experimental and clinical cardioprotective strategies. In this review, different cellular targets of doxorubicin are discussed with the focus on cardiac progenitor cells, oxidative stress, DNA damage, senescence and apoptosis all of which contribute to their compromised functional properties.
Collapse
Affiliation(s)
- Antonella De Angelis
- Department of Experimental Medicine, Section of Pharmacology, Second University of Naples, via Costantinopoli 16, 80138 Naples, Italy
| | - Konrad Urbanek
- Department of Experimental Medicine, Section of Pharmacology, Second University of Naples, via Costantinopoli 16, 80138 Naples, Italy
| | - Donato Cappetta
- Department of Experimental Medicine, Section of Pharmacology, Second University of Naples, via Costantinopoli 16, 80138 Naples, Italy
| | - Elena Piegari
- Department of Experimental Medicine, Section of Pharmacology, Second University of Naples, via Costantinopoli 16, 80138 Naples, Italy
| | - Loreta Pia Ciuffreda
- Department of Experimental Medicine, Section of Pharmacology, Second University of Naples, via Costantinopoli 16, 80138 Naples, Italy
| | - Alessia Rivellino
- Department of Experimental Medicine, Section of Pharmacology, Second University of Naples, via Costantinopoli 16, 80138 Naples, Italy
| | - Rosa Russo
- Department of Experimental Medicine, Section of Pharmacology, Second University of Naples, via Costantinopoli 16, 80138 Naples, Italy
| | - Grazia Esposito
- Department of Experimental Medicine, Section of Pharmacology, Second University of Naples, via Costantinopoli 16, 80138 Naples, Italy
| | - Francesco Rossi
- Department of Experimental Medicine, Section of Pharmacology, Second University of Naples, via Costantinopoli 16, 80138 Naples, Italy
| | - Liberato Berrino
- Department of Experimental Medicine, Section of Pharmacology, Second University of Naples, via Costantinopoli 16, 80138 Naples, Italy
| |
Collapse
|
97
|
Correia-Melo C, Marques FDM, Anderson R, Hewitt G, Hewitt R, Cole J, Carroll BM, Miwa S, Birch J, Merz A, Rushton MD, Charles M, Jurk D, Tait SWG, Czapiewski R, Greaves L, Nelson G, Bohlooly-Y M, Rodriguez-Cuenca S, Vidal-Puig A, Mann D, Saretzki G, Quarato G, Green DR, Adams PD, von Zglinicki T, Korolchuk VI, Passos JF. Mitochondria are required for pro-ageing features of the senescent phenotype. EMBO J 2016; 35:724-42. [PMID: 26848154 PMCID: PMC4818766 DOI: 10.15252/embj.201592862] [Citation(s) in RCA: 484] [Impact Index Per Article: 60.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2015] [Accepted: 01/12/2016] [Indexed: 01/07/2023] Open
Abstract
Cell senescence is an important tumour suppressor mechanism and driver of ageing. Both functions are dependent on the development of the senescent phenotype, which involves an overproduction of pro‐inflammatory and pro‐oxidant signals. However, the exact mechanisms regulating these phenotypes remain poorly understood. Here, we show the critical role of mitochondria in cellular senescence. In multiple models of senescence, absence of mitochondria reduced a spectrum of senescence effectors and phenotypes while preserving ATP production via enhanced glycolysis. Global transcriptomic analysis by RNA sequencing revealed that a vast number of senescent‐associated changes are dependent on mitochondria, particularly the pro‐inflammatory phenotype. Mechanistically, we show that the ATM, Akt and mTORC1 phosphorylation cascade integrates signals from the DNA damage response (DDR) towards PGC‐1β‐dependent mitochondrial biogenesis, contributing to a ROS‐mediated activation of the DDR and cell cycle arrest. Finally, we demonstrate that the reduction in mitochondrial content in vivo, by either mTORC1 inhibition or PGC‐1β deletion, prevents senescence in the ageing mouse liver. Our results suggest that mitochondria are a candidate target for interventions to reduce the deleterious impact of senescence in ageing tissues.
Collapse
Affiliation(s)
- Clara Correia-Melo
- Institute for Cell and Molecular Biosciences, Campus for Ageing and Vitality, Newcastle University Institute for Ageing, Newcastle University, Newcastle upon Tyne, UK GABBA Program, Abel Salazar Biomedical Sciences Institute University of Porto, Porto, Portugal
| | - Francisco D M Marques
- Institute for Cell and Molecular Biosciences, Campus for Ageing and Vitality, Newcastle University Institute for Ageing, Newcastle University, Newcastle upon Tyne, UK
| | - Rhys Anderson
- Institute for Cell and Molecular Biosciences, Campus for Ageing and Vitality, Newcastle University Institute for Ageing, Newcastle University, Newcastle upon Tyne, UK
| | - Graeme Hewitt
- Institute for Cell and Molecular Biosciences, Campus for Ageing and Vitality, Newcastle University Institute for Ageing, Newcastle University, Newcastle upon Tyne, UK
| | - Rachael Hewitt
- Institute of Cancer Sciences, CR-UK Beatson Institute, University of Glasgow, Glasgow, UK
| | - John Cole
- Institute of Cancer Sciences, CR-UK Beatson Institute, University of Glasgow, Glasgow, UK
| | - Bernadette M Carroll
- Institute for Cell and Molecular Biosciences, Campus for Ageing and Vitality, Newcastle University Institute for Ageing, Newcastle University, Newcastle upon Tyne, UK
| | - Satomi Miwa
- Institute for Cell and Molecular Biosciences, Campus for Ageing and Vitality, Newcastle University Institute for Ageing, Newcastle University, Newcastle upon Tyne, UK
| | - Jodie Birch
- Institute for Cell and Molecular Biosciences, Campus for Ageing and Vitality, Newcastle University Institute for Ageing, Newcastle University, Newcastle upon Tyne, UK
| | - Alina Merz
- Institute for Cell and Molecular Biosciences, Campus for Ageing and Vitality, Newcastle University Institute for Ageing, Newcastle University, Newcastle upon Tyne, UK
| | - Michael D Rushton
- Institute for Cell and Molecular Biosciences, Campus for Ageing and Vitality, Newcastle University Institute for Ageing, Newcastle University, Newcastle upon Tyne, UK
| | - Michelle Charles
- Institute for Cell and Molecular Biosciences, Campus for Ageing and Vitality, Newcastle University Institute for Ageing, Newcastle University, Newcastle upon Tyne, UK
| | - Diana Jurk
- Institute for Cell and Molecular Biosciences, Campus for Ageing and Vitality, Newcastle University Institute for Ageing, Newcastle University, Newcastle upon Tyne, UK
| | - Stephen W G Tait
- Institute of Cancer Sciences, CR-UK Beatson Institute, University of Glasgow, Glasgow, UK
| | - Rafal Czapiewski
- Institute for Cell and Molecular Biosciences, Campus for Ageing and Vitality, Newcastle University Institute for Ageing, Newcastle University, Newcastle upon Tyne, UK
| | - Laura Greaves
- Wellcome Trust Centre for Mitochondrial Research, Newcastle University Centre for Brain Ageing and Vitality, Newcastle University, Newcastle upon Tyne, UK
| | - Glyn Nelson
- Institute for Cell and Molecular Biosciences, Campus for Ageing and Vitality, Newcastle University Institute for Ageing, Newcastle University, Newcastle upon Tyne, UK
| | | | - Sergio Rodriguez-Cuenca
- Metabolic Research Laboratories, Wellcome Trust-MRC Institute of Metabolic Science, Addenbrooke's Hospital, University of Cambridge, Cambridge, UK
| | - Antonio Vidal-Puig
- Metabolic Research Laboratories, Wellcome Trust-MRC Institute of Metabolic Science, Addenbrooke's Hospital, University of Cambridge, Cambridge, UK
| | - Derek Mann
- Faculty of Medical Sciences, Institute of Cellular Medicine, Newcastle University, Newcastle upon Tyne, UK
| | - Gabriele Saretzki
- Institute for Cell and Molecular Biosciences, Campus for Ageing and Vitality, Newcastle University Institute for Ageing, Newcastle University, Newcastle upon Tyne, UK
| | - Giovanni Quarato
- Department of Immunology, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Douglas R Green
- Department of Immunology, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Peter D Adams
- Institute of Cancer Sciences, CR-UK Beatson Institute, University of Glasgow, Glasgow, UK
| | - Thomas von Zglinicki
- Institute for Cell and Molecular Biosciences, Campus for Ageing and Vitality, Newcastle University Institute for Ageing, Newcastle University, Newcastle upon Tyne, UK
| | - Viktor I Korolchuk
- Institute for Cell and Molecular Biosciences, Campus for Ageing and Vitality, Newcastle University Institute for Ageing, Newcastle University, Newcastle upon Tyne, UK
| | - João F Passos
- Institute for Cell and Molecular Biosciences, Campus for Ageing and Vitality, Newcastle University Institute for Ageing, Newcastle University, Newcastle upon Tyne, UK
| |
Collapse
|
98
|
Sridharan L, Maurer MS. Paradoxical Low Flow/Low Gradient Severe Aortic Stenosis: an Emerging Phenotype in Older Adults with Valve Disease. CURRENT GERIATRICS REPORTS 2015. [DOI: 10.1007/s13670-015-0139-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
|
99
|
Hohensinner PJ, Kaun C, Buchberger E, Ebenbauer B, Demyanets S, Huk I, Eppel W, Maurer G, Huber K, Wojta J. Age intrinsic loss of telomere protection via TRF1 reduction in endothelial cells. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2015; 1863:360-7. [PMID: 26658719 DOI: 10.1016/j.bbamcr.2015.11.034] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/17/2015] [Revised: 11/26/2015] [Accepted: 11/28/2015] [Indexed: 12/30/2022]
Abstract
Aging is a major factor predisposing for multiple diseases. Telomeres at the ends of chromosomes protect the integrity of chromosomal DNA. A specialized six-protein complex termed shelterin protects the telomere from unwanted interaction with DNA damage pathways. The aim of our study was to evaluate the integrity of telomeres and the stability of telomere protection during aging in endothelial cells (EC). We describe that aging EC can be characterized by an increased cell size (40%, p=0.02) and increased expression of PAI 1 (4 fold, p=0.02), MCP1 (10 fold, p=0.001) and GMCSF (15 fold, p=0.004). Telomeric state in aging cells is defined by an increased telomere oxidation (27%, p=0.01), reduced telomere length (62%, p=0.02), and increased DNA damage foci formation (5% in young EC versus 16% in aged EC, p=0.003). This telomeric dysfunction is accompanied by a reduction in the shelterin component TRF1 (33% mRNA, p=0.001; 24% protein, p=0.007). Overexpression of TRF1 in aging EC reduced telomere-associated DNA damage foci to 5% (p=0.02) and reduced expression levels of MCP1 (18% reduction, p=0.008). Aged EC have increased telomere damage and an intrinsic loss of telomere protection. Reestablishing telomere integrity could therefore be a target for rejuvenating endothelial cell function.
Collapse
Affiliation(s)
- P J Hohensinner
- Department of Internal Medicine II, Cardiology, Medical University of Vienna, Vienna, Austria.
| | - C Kaun
- Department of Internal Medicine II, Cardiology, Medical University of Vienna, Vienna, Austria
| | - E Buchberger
- Department of Surgery, Medical University of Vienna, Vienna, Austria
| | - B Ebenbauer
- Department of Internal Medicine II, Cardiology, Medical University of Vienna, Vienna, Austria; Ludwig Boltzmann Cluster for Cardiovascular Research, Vienna, Austria
| | - S Demyanets
- Department of Internal Medicine II, Cardiology, Medical University of Vienna, Vienna, Austria; Department of Laboratory Medicine, Medical University of Vienna, Vienna, Austria
| | - I Huk
- Department of Surgery, Division of Vascular Surgery, Medical University of Vienna, Vienna, Austria
| | - W Eppel
- Department of Gynecology, Medical University of Vienna, Vienna, Austria
| | - G Maurer
- Department of Internal Medicine II, Cardiology, Medical University of Vienna, Vienna, Austria
| | - K Huber
- Ludwig Boltzmann Cluster for Cardiovascular Research, Vienna, Austria; 3rd Medical Department, Wilhelminenhospital, Vienna, Austria
| | - J Wojta
- Department of Internal Medicine II, Cardiology, Medical University of Vienna, Vienna, Austria; Ludwig Boltzmann Cluster for Cardiovascular Research, Vienna, Austria; Core Facilities, Medical University of Vienna, Vienna, Austria
| |
Collapse
|
100
|
Mitochondria: Are they causal players in cellular senescence? BIOCHIMICA ET BIOPHYSICA ACTA-BIOENERGETICS 2015; 1847:1373-9. [DOI: 10.1016/j.bbabio.2015.05.017] [Citation(s) in RCA: 106] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/10/2015] [Revised: 05/20/2015] [Accepted: 05/21/2015] [Indexed: 12/25/2022]
|