Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 31
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Subcell Biochem ; 103: 45-78, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37120464

RESUMEN

During ageing molecular damage leads to the accumulation of several hallmarks of ageing including mitochondrial dysfunction, cellular senescence, genetic instability and chronic inflammation, which contribute to the development and progression of ageing-associated diseases including cardiovascular disease. Consequently, understanding how these hallmarks of biological ageing interact with the cardiovascular system and each other is fundamental to the pursuit of improving cardiovascular health globally. This review provides an overview of our current understanding of how candidate hallmarks contribute to cardiovascular diseases such as atherosclerosis, coronary artery disease and subsequent myocardial infarction, and age-related heart failure. Further, we consider the evidence that, even in the absence of chronological age, acute cellular stress leading to accelerated biological ageing expedites cardiovascular dysfunction and impacts on cardiovascular health. Finally, we consider the opportunities that modulating hallmarks of ageing offer for the development of novel cardiovascular therapeutics.


Asunto(s)
Enfermedades Cardiovasculares , Cardiopatías , Telomerasa , Humanos , Enfermedades Cardiovasculares/genética , Telomerasa/genética , Envejecimiento/genética , Senescencia Celular , Mitocondrias/genética
2.
EMBO J ; 38(5)2019 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-30737259

RESUMEN

Ageing is the biggest risk factor for cardiovascular disease. Cellular senescence, a process driven in part by telomere shortening, has been implicated in age-related tissue dysfunction. Here, we address the question of how senescence is induced in rarely dividing/post-mitotic cardiomyocytes and investigate whether clearance of senescent cells attenuates age-related cardiac dysfunction. During ageing, human and murine cardiomyocytes acquire a senescent-like phenotype characterised by persistent DNA damage at telomere regions that can be driven by mitochondrial dysfunction and crucially can occur independently of cell division and telomere length. Length-independent telomere damage in cardiomyocytes activates the classical senescence-inducing pathways, p21CIP and p16INK4a, and results in a non-canonical senescence-associated secretory phenotype, which is pro-fibrotic and pro-hypertrophic. Pharmacological or genetic clearance of senescent cells in mice alleviates detrimental features of cardiac ageing, including myocardial hypertrophy and fibrosis. Our data describe a mechanism by which senescence can occur and contribute to age-related myocardial dysfunction and in the wider setting to ageing in post-mitotic tissues.


Asunto(s)
Cardiomegalia/patología , Senescencia Celular , Daño del ADN , Fibrosis/patología , Mitosis , Miocitos Cardíacos/patología , Acortamiento del Telómero , Envejecimiento , Animales , Cardiomegalia/etiología , Femenino , Fibrosis/etiología , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Monoaminooxidasa/fisiología , Miocitos Cardíacos/metabolismo , Fenotipo , ARN/fisiología , Ratas Sprague-Dawley , Telomerasa/fisiología
3.
Age Ageing ; 52(7)2023 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-37466640

RESUMEN

Cellular senescence has emerged as a fundamental biological mechanism underpinning the ageing process and has been implicated in the pathogenesis of an increasing number of age-related conditions. Cellular senescence is a cell fate originally defined as an irreversible loss of replicative potential although it is now clear that it can be induced by a variety of mechanisms independent of replication and telomere attrition. The drivers include a persistent DNA damage response causing multiple alterations in cellular function. Senescent cells secrete a range of mediators that drive chronic inflammation and can convert other cells to the senescent state-the senescence-associated secretory phenotype. Much research to date has been conducted in animal models, but it is now clear that senescent cells accompany ageing in humans and their presence is an important driver of disease across systems. Proof-of-concept work suggests that preventing or reversing senescence may be a viable strategy to counteract human ageing and age-related disease. Possible interventions include exercise, nutrition and senolytics/senostatic drugs although there are a number of potential limitations to the use of senotherapeutics. These interventions are generally tested for single-organ conditions, but the real power of this approach is the potential to tackle multiple age-related conditions. The litmus test for this exciting new class of therapies, however, will be whether they can improve healthy life expectancy rather than merely extending lifespan. The outcomes measured in clinical studies need to reflect these aims if senotherapeutics are to gain the trust of clinicians, patients and the public.


Asunto(s)
Senescencia Celular , Senoterapéuticos , Animales , Humanos , Senescencia Celular/fisiología , Envejecimiento/fisiología , Longevidad , Inflamación
4.
Cardiovasc Drugs Ther ; 36(1): 187-196, 2022 02.
Artículo en Inglés | MEDLINE | ID: mdl-32979174

RESUMEN

Ageing is the biggest risk factor for impaired cardiovascular health, with cardiovascular disease being the leading cause of death in 40% of individuals over 65 years old. Ageing is associated with both an increased prevalence of cardiovascular disease including heart failure, coronary artery disease, and myocardial infarction. Furthermore, ageing is associated with a poorer prognosis to these diseases. Genetic models allowing the elimination of senescent cells revealed that an accumulation of senescence contributes to the pathophysiology of cardiovascular ageing and promotes the progression of cardiovascular disease through the expression of a proinflammatory and profibrotic senescence-associated secretory phenotype. These studies have resulted in an effort to identify pharmacological therapeutics that enable the specific elimination of senescent cells through apoptosis induction. These senescent cell apoptosis-inducing compounds are termed senolytics and their potential to ameliorate age-associated cardiovascular disease is the focus of this review.


Asunto(s)
Enfermedades Cardiovasculares/tratamiento farmacológico , Senescencia Celular/efectos de los fármacos , Senoterapéuticos/farmacología , Anciano , Envejecimiento , Animales , Apoptosis/efectos de los fármacos , Enfermedades Cardiovasculares/fisiopatología , Progresión de la Enfermedad , Humanos , Pronóstico , Factores de Riesgo , Fenotipo Secretor Asociado a la Senescencia/fisiología
6.
Cells ; 13(4)2024 Feb 17.
Artículo en Inglés | MEDLINE | ID: mdl-38391966

RESUMEN

Cardiovascular diseases (CVDs), a group of disorders affecting the heart or blood vessels, are the primary cause of death worldwide, with an immense impact on patient quality of life and disability. According to the World Health Organization, CVD takes an estimated 17.9 million lives each year, where more than four out of five CVD deaths are due to heart attacks and strokes. In the decades to come, an increased prevalence of age-related CVD, such as atherosclerosis, coronary artery stenosis, myocardial infarction (MI), valvular heart disease, and heart failure (HF) will contribute to an even greater health and economic burden as the global average life expectancy increases and consequently the world's population continues to age. Considering this, it is important to focus our research efforts on understanding the fundamental mechanisms underlying CVD. In this review, we focus on cellular senescence and mitochondrial dysfunction, which have long been established to contribute to CVD. We also assess the recent advances in targeting mitochondrial dysfunction including energy starvation and oxidative stress, mitochondria dynamics imbalance, cell apoptosis, mitophagy, and senescence with a focus on therapies that influence both and therefore perhaps represent strategies with the most clinical potential, range, and utility.


Asunto(s)
Enfermedades Cardiovasculares , Insuficiencia Cardíaca , Enfermedades Mitocondriales , Infarto del Miocardio , Humanos , Calidad de Vida , Senescencia Celular
7.
Dev Cell ; 59(15): 1924-1939.e7, 2024 Aug 05.
Artículo en Inglés | MEDLINE | ID: mdl-38897197

RESUMEN

Selective degradation of damaged mitochondria by autophagy (mitophagy) is proposed to play an important role in cellular homeostasis. However, the molecular mechanisms and the requirement of mitochondrial quality control by mitophagy for cellular physiology are poorly understood. Here, we demonstrated that primary human cells maintain highly active basal mitophagy initiated by mitochondrial superoxide signaling. Mitophagy was found to be mediated by PINK1/Parkin-dependent pathway involving p62 as a selective autophagy receptor (SAR). Importantly, this pathway was suppressed upon the induction of cellular senescence and in naturally aged cells, leading to a robust shutdown of mitophagy. Inhibition of mitophagy in proliferating cells was sufficient to trigger the senescence program, while reactivation of mitophagy was necessary for the anti-senescence effects of NAD precursors or rapamycin. Furthermore, reactivation of mitophagy by a p62-targeting small molecule rescued markers of cellular aging, which establishes mitochondrial quality control as a promising target for anti-aging interventions.


Asunto(s)
Senescencia Celular , Mitocondrias , Mitofagia , Ubiquitina-Proteína Ligasas , Mitofagia/efectos de los fármacos , Humanos , Senescencia Celular/efectos de los fármacos , Mitocondrias/metabolismo , Mitocondrias/efectos de los fármacos , Ubiquitina-Proteína Ligasas/metabolismo , Ubiquitina-Proteína Ligasas/genética , Proteínas Quinasas/metabolismo , Fenotipo , Autofagia/efectos de los fármacos , Proteína Sequestosoma-1/metabolismo , Transducción de Señal/efectos de los fármacos , Sirolimus/farmacología , Superóxidos/metabolismo , Proteínas de Unión al ARN
8.
FASEB J ; 26(12): 4832-40, 2012 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-22919071

RESUMEN

While the mammalian heart has low, but functionally significant, levels of telomerase expression, the cellular population responsible remains incompletely characterized. This study aimed to identify the cell types responsible for cardiac telomerase activity in neonatal, adult, and cryoinjured adult hearts using transgenic mice expressing green fluorescent protein (GFP), driven by the promoter for murine telomerase reverse transcriptase (mTert), which is a necessary and rate-limiting component of telomerase. A rare population of mTert-GFP-expressing cells was identified that possessed all detectable cardiac telomerase RNA and telomerase activity. It was heterogeneous and included cells coexpressing markers of cardiomyocytic, endothelial, and mesenchymal lineages, putative cardiac stem cell markers, and, interestingly, cardiomyocytes with a differentiated phenotype. Quantification using both flow cytometry and immunofluorescence identified a significant decline in mTert-GFP cells in adult animals compared to neonates (∼9- and ∼20-fold, respectively). Cardiac injury resulted in a ∼6.45-fold expansion of this population (P<0.005) compared with sham-operated controls. This study identifies the cells responsible for cardiac telomerase activity, demonstrates a significant diminution with age but a marked response to injury, and, given the relationship between telomerase activity and stem cell populations, suggests that they represent a potential target for further investigation of cardiac regenerative potential.


Asunto(s)
Regulación del Desarrollo de la Expresión Génica , Regulación Enzimológica de la Expresión Génica , Miocardio/metabolismo , Telomerasa/genética , Factores de Edad , Animales , Animales Recién Nacidos , Antígenos Ly/genética , Antígenos Ly/metabolismo , Citometría de Flujo , Factor de Transcripción GATA4/genética , Factor de Transcripción GATA4/metabolismo , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Proteína Homeótica Nkx-2.5 , Proteínas de Homeodominio/genética , Proteínas de Homeodominio/metabolismo , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Ratones , Ratones Transgénicos , Microscopía Confocal , Miocardio/citología , Miocardio/enzimología , ARN Mensajero/genética , ARN Mensajero/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Telomerasa/metabolismo , Factores de Tiempo , Factores de Transcripción/genética , Factores de Transcripción/metabolismo
9.
NPJ Aging ; 9(1): 15, 2023 Jun 14.
Artículo en Inglés | MEDLINE | ID: mdl-37316516

RESUMEN

Myocardial infarction is a leading cause of morbidity and mortality. While reperfusion is now standard therapy, pathological remodelling leading to heart failure remains a clinical problem. Cellular senescence has been shown to contribute to disease pathophysiology and treatment with the senolytic navitoclax attenuates inflammation, reduces adverse myocardial remodelling and results in improved functional recovery. However, it remains unclear which senescent cell populations contribute to these processes. To identify whether senescent cardiomyocytes contribute to disease pathophysiology post-myocardial infarction, we established a transgenic model in which p16 (CDKN2A) expression was specifically knocked-out in the cardiomyocyte population. Following myocardial infarction, mice lacking cardiomyocyte p16 expression demonstrated no difference in cardiomyocyte hypertrophy but exhibited improved cardiac function and significantly reduced scar size in comparison to control animals. This data demonstrates that senescent cardiomyocytes participate in pathological myocardial remodelling. Importantly, inhibition of cardiomyocyte senescence led to reduced senescence-associated inflammation and decreased senescence-associated markers within other myocardial lineages, consistent with the hypothesis that cardiomyocytes promote pathological remodelling by spreading senescence to other cell-types. Collectively this study presents the demonstration that senescent cardiomyocytes are major contributors to myocardial remodelling and dysfunction following a myocardial infarction. Therefore, to maximise the potential for clinical translation, it is important to further understand the mechanisms underlying cardiomyocyte senescence and how to optimise senolytic strategies to target this cell lineage.

10.
Front Aging ; 3: 1058435, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36452034

RESUMEN

Cancer continues to place a heavy burden on healthcare systems around the world. Although cancer survivorship continues to improve, cardiotoxicity leading to cardiomyopathy and heart failure as a consequence of cancer therapy is rising, and yesterday's cancer survivors are fast becoming today's heart failure patients. Although the mechanisms driving cardiotoxicity are complex, cellular senescence is gaining attention as a major contributor to chemotherapy-induced cardiotoxicity and, therefore, may also represent a novel therapeutic target to prevent this disease. Cellular senescence is a well-recognized response to clinical doses of chemotherapies, including anthracyclines, and is defined by cell cycle exit, phenotypic alterations which include mitochondrial dysfunction, and the expression of the pro-senescent, pro-fibrotic, and pro-inflammatory senescence-associated phenotype. Senescence has an established involvement in promoting myocardial remodeling during aging, and studies have demonstrated that the elimination of senescence can attenuate the pathophysiology of several cardiovascular diseases. Most recently, pharmacology-mediated elimination of senescence, using a class of drugs termed senolytics, has been demonstrated to prevent myocardial dysfunction in preclinical models of chemotherapy-induced cardiotoxicity. In this review, we will discuss the evidence that anthracycline-induced senescence causes the long-term cardiotoxicity of anticancer chemotherapies, consider how the senescent phenotype may promote myocardial dysfunction, and examine the exciting possibility that targeting senescence may prove a therapeutic strategy to prevent or even reverse chemotherapy-induced cardiac dysfunction.

11.
Mech Ageing Dev ; 198: 111540, 2021 09.
Artículo en Inglés | MEDLINE | ID: mdl-34237321

RESUMEN

Ageing is the biggest risk factor for impaired cardiovascular health, with cardiovascular disease being the cause of death in 40 % of individuals over 65 years old. Ageing is associated with an increased prevalence of atherosclerosis, coronary artery stenosis and subsequent myocardial infarction, thoracic aortic aneurysm, valvular heart disease and heart failure. An accumulation of senescence and increased inflammation, caused by the senescence-associated secretory phenotype, have been implicated in the aetiology and progression of these age-associated diseases. Recently it has been demonstrated that compounds targeting components of anti-apoptotic pathways expressed by senescent cells can preferentially induce senescence cells to apoptosis and have been termed senolytics. In this review, we discuss the evidence demonstrating that senescence contributes to cardiovascular disease, with a particular focus on studies that indicate the promise of senotherapy. Based on these data we suggest novel indications for senolytics as a treatment of cardiovascular diseases which have yet to be studied in the context of senotherapy. Finally, while the potential benefits are encouraging, several complications may result from senolytic treatment. We, therefore, consider these challenges in the context of the cardiovascular system.


Asunto(s)
Envejecimiento , Proteínas Reguladoras de la Apoptosis/metabolismo , Enfermedades Cardiovasculares , Senescencia Celular , Senoterapéuticos/farmacología , Envejecimiento/inmunología , Envejecimiento/metabolismo , Enfermedades Cardiovasculares/etiología , Enfermedades Cardiovasculares/metabolismo , Enfermedades Cardiovasculares/terapia , Senescencia Celular/efectos de los fármacos , Senescencia Celular/fisiología , Humanos , Inflamación/metabolismo , Fenotipo Secretor Asociado a la Senescencia , Transducción de Señal/efectos de los fármacos
12.
Exp Dermatol ; 19(6): 546-8, 2010 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-20456497

RESUMEN

Human dermal papilla (DP) cells grown in two-dimensional (2D) culture have been studied extensively. However, key differences exist between DP cell activities in vivo and in vitro. Using a suspension method of cell culture to maintain DP cells, we created three-dimensional (3D) dermal spheres morphologically akin to intact (anagen) DPs. Analysis of these spheres using immunocytochemistry demonstrates that they have expression profiles different from papilla cells cultured in 2D but with many similarities to intact DPs. This method of DP cell culture may provide us with a tool to elucidate our understanding of signalling within the DP as it relates to induction, maintenance or even inhibition of hair growth.


Asunto(s)
Técnicas de Cultivo de Célula/métodos , Dermis/citología , Fibroblastos/citología , Folículo Piloso/citología , Modelos Biológicos , Esferoides Celulares/citología , Actinas/metabolismo , Proteína de la Poliposis Adenomatosa del Colon/genética , Fosfatasa Alcalina/metabolismo , Proteína Axina , Núcleo Celular/metabolismo , Conexina 43/metabolismo , Proteínas del Citoesqueleto/genética , Fibroblastos/metabolismo , Expresión Génica/genética , Glucógeno Sintasa Quinasa 3/genética , Glucógeno Sintasa Quinasa 3 beta , Proteoglicanos de Heparán Sulfato/metabolismo , Humanos , Factor de Unión 1 al Potenciador Linfoide/genética , Moléculas de Adhesión de Célula Nerviosa/genética , Antígeno Nuclear de Célula en Proliferación/metabolismo , Prostaglandina-Endoperóxido Sintasas/genética , Serina Endopeptidasas/genética , Esferoides Celulares/metabolismo , Versicanos/metabolismo , beta Catenina/metabolismo
13.
Aging Cell ; 19(10): e13249, 2020 10.
Artículo en Inglés | MEDLINE | ID: mdl-32996233

RESUMEN

A key component of cardiac ischemia-reperfusion injury (IRI) is the increased generation of reactive oxygen species, leading to enhanced inflammation and tissue dysfunction in patients following intervention for myocardial infarction. In this study, we hypothesized that oxidative stress, due to ischemia-reperfusion, induces senescence which contributes to the pathophysiology of cardiac IRI. We demonstrate that IRI induces cellular senescence in both cardiomyocytes and interstitial cell populations and treatment with the senolytic drug navitoclax after ischemia-reperfusion improves left ventricular function, increases myocardial vascularization, and decreases scar size. SWATH-MS-based proteomics revealed that biological processes associated with fibrosis and inflammation that were increased following ischemia-reperfusion were attenuated upon senescent cell clearance. Furthermore, navitoclax treatment reduced the expression of pro-inflammatory, profibrotic, and anti-angiogenic cytokines, including interferon gamma-induced protein-10, TGF-ß3, interleukin-11, interleukin-16, and fractalkine. Our study provides proof-of-concept evidence that cellular senescence contributes to impaired heart function and adverse remodeling following cardiac ischemia-reperfusion. We also establish that post-IRI the SASP plays a considerable role in the inflammatory response. Subsequently, senolytic treatment, at a clinically feasible time-point, attenuates multiple components of this response and improves clinically important parameters. Thus, cellular senescence represents a potential novel therapeutic avenue to improve patient outcomes following cardiac ischemia-reperfusion.


Asunto(s)
Senescencia Celular/fisiología , Daño por Reperfusión/metabolismo , Femenino , Humanos , Masculino
14.
Gene Expr Patterns ; 9(6): 454-60, 2009 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-19427408

RESUMEN

Syndecan-1 is a cell-surface heparan-sulphate proteoglycan that is involved in growth factor regulation, cell adhesion, proliferation, differentiation, blood coagulation, lipid metabolism, as well as tumour formation. In this study, investigation of discrete LCM captured dermal cells by semi-quantitative RT-PCR revealed Syndecan-1 mRNA transcripts were expressed only in the dermal condensation (DC) within this skin compartment during murine pelage hair follicle (HF) morphogenesis. Further immunofluorescence studies showed that, during early skin development, Syndecan-1 was expressed in the epidermis while being absent from the mesenchyme. As HF morphogenesis began ( approximately E14.5) Syndecan-1 expression was lost from the epithelial compartment of the HF and activated in HF mesenchymal cells. This Syndecan-1 expression profile was consistent between different hair follicle types including primary and secondary pelage, vibrissa, and tail hair follicles. Furthermore we show by using gene targeted mice lacking Syndecan-1 expression that Syndecan-1 is not required for follicle initiation and development.

15.
Exp Dermatol ; 18(9): 793-5, 2009 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-19695019

RESUMEN

Exogen is a distinct phase of the hair cycle describing the process by which the hair club fibre is shed from the follicle. This process is difficult to study in human skin and little is known about the mechanisms involved in the release of club fibres. We sought an alternative model system to study exogen in more detail, and therefore utilised the vibrissa system on the rodent mystacial pad. The time at which a vibrissa club hair will be lost can be predicted, based on the relative lengths of the new growing fibre and old club fibre. This timing phenomenon was exploited to investigate the club fibre within the follicle as it approaches final release, revealing key changes in the adhesive state of the club fibre within the epithelial sac as it approached release. We propose that exogen should be subdivided to represent variations in the club fibre status.


Asunto(s)
Folículo Piloso/fisiología , Vibrisas/crecimiento & desarrollo , Animales , Ratas
16.
Exp Eye Res ; 89(3): 435-8, 2009 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-19344714

RESUMEN

The corneal epithelium is continuously replaced by epithelial stem cells located in the basal layer of the limbus, located at the margin of the cornea. Studying how the stem cell niche is established at the limbus during development of the eye may lead to better understanding and treatments for diseases associated with limbal deficiencies. Using two highly specific commercially available antibodies, K10 was consistently detected suprabasally throughout the developing limbal epithelium of late gestation (20.5 dpc) and neonatal rat corneas, with interrupted expression in adult rat limbal epithelium. RT-PCR confirmed K10 expression at the transcript level in embryonic, neonatal and adult rat eyes. We have identified a time point where early stages of limbal development may be facilitated by the suprabasal expression of K10.


Asunto(s)
Queratina-10/metabolismo , Limbo de la Córnea/metabolismo , Envejecimiento/metabolismo , Animales , Proteínas del Ojo/metabolismo , Masculino , Conejos , Ratas
17.
Aging Cell ; 18(3): e12945, 2019 06.
Artículo en Inglés | MEDLINE | ID: mdl-30920115

RESUMEN

Cardiovascular disease is the leading cause of death in individuals over 60 years old. Aging is associated with an increased prevalence of coronary artery disease and a poorer prognosis following acute myocardial infarction (MI). With age, senescent cells accumulate in tissues, including the heart, and contribute to age-related pathologies. However, the role of senescence in recovery following MI has not been investigated. In this study, we demonstrate that treatment of aged mice with the senolytic drug, navitoclax, eliminates senescent cardiomyocytes and attenuates profibrotic protein expression in aged mice. Importantly, clearance of senescent cells improved myocardial remodelling and diastolic function as well as overall survival following MI. These data provide proof-of-concept evidence that senescent cells are major contributors to impaired function and increased mortality following MI and that senolytics are a potential new therapeutic avenue for MI.


Asunto(s)
Envejecimiento/efectos de los fármacos , Compuestos de Anilina/farmacología , Antineoplásicos/farmacología , Senescencia Celular/efectos de los fármacos , Infarto del Miocardio/tratamiento farmacológico , Sulfonamidas/farmacología , Enfermedad Aguda , Compuestos de Anilina/administración & dosificación , Animales , Antineoplásicos/administración & dosificación , Supervivencia Celular/efectos de los fármacos , Masculino , Ratones , Ratones Endogámicos C57BL , Infarto del Miocardio/patología , Sulfonamidas/administración & dosificación
19.
Exp Gerontol ; 109: 5-15, 2018 08.
Artículo en Inglés | MEDLINE | ID: mdl-29054534

RESUMEN

Cardiovascular disease (CVD) is the leading cause of death globally. One of the main risk factors for CVD is age, however the biological processes that occur in the heart during ageing are poorly understood. It is therefore important to understand the fundamental mechanisms driving heart ageing to enable the development of preventions and treatments targeting these processes. Cellular senescence is often described as the irreversible cell-cycle arrest which occurs in somatic cells. Emerging evidence suggests that cellular senescence plays a key role in heart ageing, however the cell-types involved and the underlying mechanisms are not yet elucidated. In this review we discuss the current understanding of how mechanisms known to contribute to senescence impact on heart ageing and CVD. Finally, we evaluate recent data suggesting that targeting senescent cells may be a viable therapy to counteract the ageing of the heart.


Asunto(s)
Envejecimiento/fisiología , Senescencia Celular/fisiología , Corazón/fisiología , Animales , Enfermedades Cardiovasculares/etiología , Daño del ADN , Reparación del ADN , Humanos , Mitocondrias/fisiología , Especies Reactivas de Oxígeno/metabolismo , Telómero
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA