Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 24
Filtrar
Más filtros











Base de datos
Intervalo de año de publicación
1.
bioRxiv ; 2024 Jun 25.
Artículo en Inglés | MEDLINE | ID: mdl-38948744

RESUMEN

Cyclin A2 (CCNA2) is a master regulatory gene of the cell cycle that is normally silenced in postnatal mammalian cardiomyocytes. We have previously demonstrated that it can induce significant cardiac repair in both small and large animals when delivered to the heart via a viral vector. To date, whether CCNA2 gene delivery can induce cytokinesis in isolated cardiomyocytes from adult human hearts has not been investigated. Therefore, we designed a human gene therapy vector featuring a replication-deficient, E1/E3-deleted human adenovirus five encoding human CCNA2 driven by the cardiac Troponin T promoter to enable the expression of CCNA2 in freshly isolated human cardiomyocytes. Utilizing time-lapse microscopy live imaging of cultured adult human cardiomyocytes isolated from a 21-year-old male, 41-year-old female, and 55-year-old male, we now report that human adult cardiomyocytes can be induced to undergo complete cytokinesis in response to CCNA2 gene delivery with preservation of sarcomere integrity in the resulting daughter cells. To elucidate the mechanistic underpinnings of CCNA2-dependent gene regulation in governing cardiomyocyte cytokinesis, we conducted single nucleus transcriptomics (snRNA-seq, 10X Genomics) analysis in hearts isolated from adult transgenic mice that constitutively express CCNA2 in cardiomyocytes (CCNA2-Tg) and non-transgenic mice (nTg). Remarkably, we identified a subpopulation of cardiomyocytes enriched with cytokinesis, proliferative, and reprogramming genes in hearts obtained from CCNA2-Tg mice as compared to hearts obtained from nTg mice. We also performed bulk RNA sequencing of human adult and fetal hearts, and we identified key reprogramming genes that are involved in CCNA2-induced cytokinesis. These results provide a compelling path forward for the clinical development of cardiac regenerative therapy based on strategic manipulation of the cardiomyocyte cell cycle.

2.
Methods Mol Biol ; 2803: 137-144, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38676890

RESUMEN

Myocardial infarction (MI) in mice is a widely used surgical model in preclinical cardiac repair studies to recapitulate human cardiovascular ischemic disease. Induction of reproducible infarct size is crucial for quantitative and analytical purpose. Here we describe a quick and reliable method to induce consistent infarct size in mice in less than a minute.


Asunto(s)
Modelos Animales de Enfermedad , Infarto del Miocardio , Animales , Infarto del Miocardio/patología , Ratones , Humanos , Miocardio/patología , Miocardio/metabolismo
3.
bioRxiv ; 2024 Jan 13.
Artículo en Inglés | MEDLINE | ID: mdl-37577721

RESUMEN

We report a unique population of multipotent cells isolated from the term human placenta, for the first time, that can differentiate into cardiomyocytes and vascular cells with clonal proliferative ability, migratory ability, and trancriptomic evidence of immune privilege. Caudal-type homeobox-2 (CDX2) is a conserved factor that regulates trophectoderm formation and placentation during early embryonic development but has not previously been implicated in developmentally conserved regenerative mechanisms. We had earlier reported that Cdx2 lineage cells in the mouse placenta are capable of restoring cardiac function after intravenous delivery in male mice with experimental cardiac injury (myocardial infarction). Here we demonstrate that CDX2-expressing cells are prevalent in the human chorion and are poised for cardiovascular differentiation. We examined the term placentas from 106 healthy patients and showed that isolated CDX2 cells can spontaneously differentiate into cardiomyocytes, functional vascular cells, and retain homing ability in vitro. Functional annotation from transcriptomics analysis supports enhanced cardiogenesis, vasculogenesis, immune modulation, and chemotaxis gene signatures in CDX2 cells. CDX2 cells can be clonally propagated in culture with retention of cardiovascular differentiation. Our data supports further use of this accessible and ethically feasible cell source in the design of therapeutic strategies for cardiovascular disease.

4.
Heart Rhythm ; 21(4): 389-396, 2024 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-38159790

RESUMEN

BACKGROUND: Ultrastructural findings immediately after pulsed field ablation (PFA) of the myocardium have not been described. OBJECTIVES: The purpose of this study was to elucidate ultrastructural characteristics and differences between microsecond PFA at the 1- and 4-hour timepoints after pulse delivery and to compare them to irrigated radiofrequency ablation (RFA) lesions. METHODS: Healthy swine underwent endocardial PFA or RFA followed by necropsy. Discrete microsecond PFA and irrigated RFA lesions were created in the ventricles with a lattice tip ablation catheter. Lesions were delivered in a manner so as to allow sampling to occur 1 and 4 hours after ablation. All lesions were located at necropsy, and samples were carefully obtained from within the lesion core, lesion periphery, and adjacent healthy myocardium. Transmission electron microscopic assessment was performed after fixation using paraformaldehyde and glutaraldehyde. RESULTS: One hour after microsecond PFA delivery, myocytes were noted to be significantly and uniformly disrupted. Clustered, misaligned, swollen mitochondria coupled with degenerating nuclei and condensed chromatin were visualized. These findings progressed over the subsequent few hours with worsening edema. Similar changes were seen with RFA but reduced in severity. However, there was prominent extravasation of red blood cells with occlusion of capillaries that was not seen in PFA. At the lesion periphery, an abrupt change in the degree of myocyte damage was observed with PFA but not RFA. CONCLUSION: Transmission electron microscopy demonstrates evidence of widespread destruction of myocytes as early as an hour after PFA and corroborates known histologic features such as sparing of vessels and sharp lesion margins.


Asunto(s)
Fibrilación Atrial , Ablación por Catéter , Ablación por Radiofrecuencia , Porcinos , Animales , Ablación por Catéter/efectos adversos , Miocardio , Ventrículos Cardíacos/cirugía , Frecuencia Cardíaca , Fibrilación Atrial/cirugía
5.
Elife ; 122023 11 02.
Artículo en Inglés | MEDLINE | ID: mdl-37917118

RESUMEN

It may be possible to treat cardiac hypertrophy and injury by using drugs that inhibit a protein called SIRT2.


Asunto(s)
Corazón
6.
Curr Cardiol Rep ; 25(7): 615-620, 2023 07.
Artículo en Inglés | MEDLINE | ID: mdl-37266821

RESUMEN

PURPOSE OF REVIEW: This review explores efforts made over the previous three decades to determine mechanisms of cardiomyocyte cell division. Many investigators have explored cell therapy strategies in animal models and clinical trials over the past 2 decades with marginal results thus far in clinical testing. Hence, there is a greater focus now on strategies to induce cardiomyocyte proliferation. RECENT FINDINGS: Reports to induce reactivation of the cardiomyocyte cell cycle predated the focus on cell therapy, and we summarize the literature on this topic, which began with the very first transgenic mouse studies in cardiovascular science. These earlier studies form the foundation for the use of cell cycle manipulation in cardiac repair and should inform current and future investigations with respect to rigor of assessment in the degree of cardiomyocyte cell division and gold standard measures of cardiac functional improvement.


Asunto(s)
Corazón , Miocitos Cardíacos , Animales , Ratones , Humanos , Miocitos Cardíacos/fisiología , Proliferación Celular , Corazón/fisiología , Ciclo Celular/fisiología , Ratones Transgénicos , Regeneración
7.
Circ Arrhythm Electrophysiol ; 16(1): e011369, 2023 01.
Artículo en Inglés | MEDLINE | ID: mdl-36595634

RESUMEN

BACKGROUND: Pulsed field ablation (PFA) has recently been shown to penetrate ischemic scar, but details on its efficacy, risk of arrhythmias, and imaging insights are lacking. In a porcine model of myocardial scar, we studied the ability of ventricular PFA to penetrate scarred tissue, induce ventricular arrhythmias, and assess the influence of QRS gating during pulse delivery. METHODS: Of a total of 6 swine, 5 underwent coronary occlusion and 1 underwent radiofrequency ablation to create infarct scar and iatrogenic scar models, respectively. Two additional swine served as healthy controls. An 8 Fr focal PFA catheter was used to deliver bipolar, biphasic PFA (2.0 kV) lesions guided by electroanatomical mapping, fluoroscopy, and intracardiac echocardiography over both scarred and healthy myocardium. Swine underwent magnetic resonance imaging 2-7 days post-PFA. RESULTS: PFA successfully penetrated scar without significant difference in lesion depth between lesion at the infarct border (5.9±1.0 mm, n=41) and healthy myocardium (5.7±1.3 mm, n=26; P=0.53). PFA penetration of both infarct and iatrogenic radiofrequency abalation scar was observed in all examined sections. Sustained ventricular arrhythmias requiring defibrillation occurred in 4 of 187 (2.1%) ungated applications, whereas no ventricular arrhythmias occurred during gated PFA applications (0 of 64 [0%]). Dark-blood late-gadolinium-enhanced sequences allowed for improved endocardial border detection as well as lesion boundaries compared with conventional bright-blood late-gadolinium-enhanced sequences. CONCLUSIONS: PFA penetrates infarct and iatrogenic scar successfully to create deep lesions. Gated delivery eliminates the occurrence of ventricular arrhythmias observed with ungated porcine PFA. Optimized magnetic resonance imaging sequences can be helpful in detecting lesion boundaries.


Asunto(s)
Ablación por Catéter , Infarto del Miocardio , Taquicardia Ventricular , Animales , Porcinos , Cicatriz , Gadolinio , Arritmias Cardíacas/diagnóstico por imagen , Arritmias Cardíacas/cirugía , Electrofisiología Cardíaca , Enfermedad Iatrogénica , Ablación por Catéter/efectos adversos , Ablación por Catéter/métodos , Taquicardia Ventricular/diagnóstico por imagen , Taquicardia Ventricular/cirugía
8.
Circ Arrhythm Electrophysiol ; 15(9): e011120, 2022 09.
Artículo en Inglés | MEDLINE | ID: mdl-36074657

RESUMEN

BACKGROUND: Our understanding of catheter-based pulsed field ablation (PFA) of the ventricular myocardium is limited. We conducted a series of exploratory evaluations of ventricular PFA in swine ventricles. METHODS: A focal lattice-tip catheter was used to deliver proprietary biphasic monopolar PFA applications to swine ventricles under general anesthesia, with guidance from electroanatomical mapping, fluoroscopy, and intracardiac echocardiography. We conducted experiments to assess the impact of (1) delivery repetition (2×, 3×, or 4×) at each location, (2) epicardial PFA delivery, and (3) confluent areas of shallow healed endocardial scar created by prior PFA (4 weeks earlier) on subsequent endocardial PFA. Additional assessments included PFA optimized for the ventricle, lesion visualization by intracardiac echocardiography imaging, and immunohistochemical insights. RESULTS: Experiment no. 1: lesions (n=49) were larger with delivery repetition of either 4× or 3× versus 2×: length 17.6±3.9 or 14.2±2.0 versus 12.7±2.0 mm (P<0.01, P=0.22), width 13.4±1.8 or 10.6±1.3 versus 10.5±1.1 mm (P<0.01, P=1.00), and depth 6.1±2.1 or 5.1±1.3 versus 4.2±1.0 mm (P<0.01, P=0.21). Experiment no. 2: epicardial lesions (n=18) were reliably created and comparable to endocardial lesions: length 24.6±9.7 mm (n=5), width 15.6±4.6 mm, and depth 4.5±3.7 mm. Experiment no. 3: PFA (n=16) was able to penetrate to a depth of 4.8 (interquartile range, 4.5-5.4) mm in healthy myocardium versus 5.6 (interquartile range, 3.6-6.6) mm in adjacent healed endocardial scar (P=0.79), suggesting that superficial scar does not significantly impair PFA. Finally, we demonstrate, PFA optimized for the ventricle yielded adequate lesion dimensions, can result in myocardial activation, can be visualized by intracardiac echocardiography, and have unique immunohistochemical characteristics. CONCLUSIONS: This in vivo evaluation offers insights into the behavior of endocardial or epicardial PFA delivered using the lattice-tip catheter to normal or scarred porcine ventricular myocardium, thereby setting the stage for future clinical studies.


Asunto(s)
Ablación por Catéter , Cicatriz , Animales , Ablación por Catéter/métodos , Catéteres , Cicatriz/patología , Endocardio/diagnóstico por imagen , Endocardio/patología , Endocardio/cirugía , Ventrículos Cardíacos/diagnóstico por imagen , Ventrículos Cardíacos/patología , Ventrículos Cardíacos/cirugía , Porcinos
10.
Ann N Y Acad Sci ; 1487(1): 12-20, 2021 03.
Artículo en Inglés | MEDLINE | ID: mdl-32991755

RESUMEN

Organ and tissue repair are complex processes involving signaling molecules, growth factors, and cell cycle regulators that act in concert to promote cell division and differentiation at sites of injury. In embryonic development, progenitor fetal cells are actively involved in reparative mechanisms and display a biphasic interaction with the mother; and there is constant trafficking of fetal cells into maternal circulation and vice versa. This phenomenon of fetal microchimerism may have significant impact considering the primitive, multilineage nature of these cells. In published work, we have reported that fetal-derived placental cells expressing the homeodomain protein CDX2 retain all "stem" functional proteins of embryonic stem cells yet are endowed with additional functions in areas of growth, survival, homing, and immune modulation. These cells exhibit multipotency in vitro and in vivo, giving rise to spontaneously beating cardiomyocytes and vascular cells. In mouse models, CDX2 cells from female placentas can be administered intravenously to male mice subjected to myocardial infarction with subsequent homing of the CDX2 cells to infarcted areas and evidence of cellular regeneration with enhanced cardiac function. Elucidating the role of microchimeric fetal-derived placental cells may have broader scientific potential, as one can envision allogeneic cell therapy strategies targeted at tissue regeneration for a variety of organ systems.


Asunto(s)
Quimerismo/embriología , Regeneración/fisiología , Cicatrización de Heridas/fisiología , Animales , Femenino , Feto/inmunología , Feto/metabolismo , Humanos , Masculino , Intercambio Materno-Fetal/fisiología , Ratones , Trasplante de Órganos/métodos , Trasplante de Órganos/tendencias , Embarazo , Regeneración/genética , Regeneración/inmunología , Medicina Regenerativa/métodos , Medicina Regenerativa/tendencias , Transducción de Señal/fisiología , Cicatrización de Heridas/genética , Cicatrización de Heridas/inmunología
11.
Methods Mol Biol ; 2029: 235-246, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31273746

RESUMEN

Flow cytometry is a widely used laser-based technology for rapid analysis of the expression of cell surface antigens and intracellular molecules in various cell types including hematopoietic stem/progenitor cells (HSPCs). Multiparametric analysis of individual cells within a short time frame makes this tool attractive and indispensable in the field of stem cell research. This is accomplished by harnessing the specific light scattering ability of the cell type, which determines its size and internal complexity. In addition, use of fluorescently conjugated antibodies allows the detection of a specific surface or intracellular antigen present at that particular stage. Fluorescent Activated Cell Sorting (FACS) is used to separate a subset of cells from a heterogeneous cell population based on fluorescent labeling. Here we describe the general principles of flow cytometry and detailed methods for the isolation of HSPCs using flow cytometry as a tool.


Asunto(s)
Separación Celular/métodos , Citometría de Flujo/métodos , Células Madre Hematopoyéticas/citología , Células Madre/citología , Animales , Antígenos de Superficie/metabolismo , Colorantes Fluorescentes/metabolismo , Células Madre Hematopoyéticas/metabolismo , Humanos , Ratones , Células Madre/metabolismo
12.
Proc Natl Acad Sci U S A ; 116(24): 11786-11795, 2019 06 11.
Artículo en Inglés | MEDLINE | ID: mdl-31109997

RESUMEN

The extremely limited regenerative potential of adult mammalian hearts has prompted the need for novel cell-based therapies that can restore contractile function in heart disease. We have previously shown the regenerative potential of mixed fetal cells that were naturally found migrating to the injured maternal heart. Exploiting this intrinsic mechanism led to the current hypothesis that Caudal-type homeobox-2 (Cdx2) cells in placenta may represent a novel cell type for cardiac regeneration. Using a lineage-tracing strategy, we specifically labeled fetal-derived Cdx2 cells with enhanced green fluorescent protein (eGFP). Cdx2-eGFP cells from end-gestation placenta were assayed for cardiac differentiation in vitro and in vivo using a mouse model of myocardial infarction. We observed that these cells differentiated into spontaneously beating cardiomyocytes (CMs) and vascular cells in vitro, indicating multipotentiality. When administered via tail vein to infarcted wild-type male mice, they selectively and robustly homed to the heart and differentiated to CMs and blood vessels, resulting in significant improvement in contractility as noted by MRI. Proteomics and immune transcriptomics studies of Cdx2-eGFP cells compared with embryonic stem (ES) cells reveal that they appear to retain "stem"-related functions of ES cells but exhibit unique signatures supporting roles in homing and survival, with an ability to evade immune surveillance, which is critical for cell-based therapy. Cdx2-eGFP cells may potentially represent a therapeutic advance in allogeneic cell therapy for cardiac repair.


Asunto(s)
Factor de Transcripción CDX2/metabolismo , Feto/citología , Corazón/fisiología , Células Madre Multipotentes/citología , Miocitos Cardíacos/citología , Placenta/citología , Regeneración/fisiología , Animales , Diferenciación Celular/fisiología , Células Cultivadas , Células Madre Embrionarias/citología , Células Madre Embrionarias/metabolismo , Femenino , Feto/metabolismo , Proteínas Fluorescentes Verdes/metabolismo , Cardiopatías/metabolismo , Cardiopatías/terapia , Masculino , Ratones , Ratones Endogámicos C57BL , Células Madre Multipotentes/metabolismo , Infarto del Miocardio/patología , Miocitos Cardíacos/metabolismo , Placenta/metabolismo , Embarazo
14.
Sci Transl Med ; 6(224): 224ra27, 2014 Feb 19.
Artículo en Inglés | MEDLINE | ID: mdl-24553388

RESUMEN

Cyclin A2 (Ccna2), normally silenced after birth in the mammalian heart, can induce cardiac repair in small-animal models of myocardial infarction. We report that delivery of the Ccna2 gene to infarcted porcine hearts invokes a regenerative response. We used a catheter-based approach to occlude the left anterior descending artery in swine, which resulted in substantial myocardial infarction. A week later, we performed left lateral thoracotomy and injected adenovirus carrying complementary DNA encoding CCNA2 or null adenovirus into peri-infarct myocardium. Six weeks after treatment, we assessed cardiac contractile function using multimodality imaging including magnetic resonance imaging, which demonstrated ~18% increase in ejection fraction of Ccna2-treated pigs and ~4% decrease in control pigs. Histologic studies demonstrate in vivo evidence of increased cardiomyocyte mitoses, increased cardiomyocyte number, and decreased fibrosis in the experimental pigs. Using time-lapse microscopic imaging of cultured adult porcine cardiomyocytes, we also show that Ccna2 elicits cytokinesis of adult porcine cardiomyocytes with preservation of sarcomeric structure. These data provide a compelling framework for the design and development of cardiac regenerative therapies based on cardiomyocyte cell cycle regulation.


Asunto(s)
Ciclina A2/fisiología , Citocinesis , Infarto del Miocardio/terapia , Miocitos Cardíacos/citología , Regeneración , Animales , Infarto del Miocardio/fisiopatología , Porcinos
16.
Stem Cell Rev Rep ; 9(3): 303-12, 2013 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-22628110

RESUMEN

Epicardial resident stem cells are known to differentiate into cardiomyocytes during cardiac development, amongst other cell types. Whether epicardium-derived progenitor cells (EPDCs) retain this plasticity in the adult heart has been the topic of heated scientific debate. Priming with thymosin beta 4, a peptide which has been suggested to be critical for cardiac development and to have cardio-protective properties, was recently shown to induce differentiation of EPDCs into cardiomyocytes in a small animal model of myocardial infarction. This finding is in stark contrast to another recent study in which thymosin beta 4 treatment following myocardial infarction did not induce cardiomyocyte differentiation of EPDCs. While EPDCs seem to exhibit overall cardio-protective effects on the heart following myocardial infarction, they have not been shown to differentiate into cardiomyocytes in a clinically relevant setting. It will be important to understand why the ability of one therapeutic agent to induce cardiomyocyte differentiation of EPDCs seemingly depends on a single variable, i.e. the time of administration. Furthermore, in light of a recent report, it appears that thymosin beta 4 may be dispensable for cardiac development.


Asunto(s)
Diferenciación Celular/efectos de los fármacos , Infarto del Miocardio/terapia , Regeneración/efectos de los fármacos , Timosina/farmacología , Corazón , Humanos , Miocardio/citología , Miocardio/metabolismo , Miocitos Cardíacos/metabolismo , Trasplante de Células Madre , Células Madre/efectos de los fármacos , Células Madre/metabolismo
17.
Clin Transl Sci ; 5(4): 321-8, 2012 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-22883609

RESUMEN

Fetal cells enter the maternal circulation during pregnancies and can persist in blood and tissues for decades, creating a state of physiologic microchimerism. Microchimerism refers to acquisition of cells from another individual and can be due to bidirectional cell traffic between mother and fetus during pregnancy. Peripartum cardiomyopathy, a rare cardiac disorder associated with high mortality rates has the highest recovery rate amongst all etiologies of heart failure although the reason is unknown. Collectively, these observations led us to hypothesize that fetal cells enter the maternal circulation and may be recruited to the sites of myocardial disease or injury. The ability to genetically modify mice makes them an ideal system for studying the phenomenon of microchimerism in cardiac disease. Described here is a mouse model for ischemic cardiac injury during pregnancy designed to study microchimerism. Wild-type virgin female mice mated with eGFP male mice underwent ligation of the left anterior descending artery to induce a myocardial infarction at gestation day 12. We demonstrate the selective homing of eGFP cells to the site of cardiac injury without such homing to noninjured tissues suggesting the presence of precise signals sensed by fetal cells enabling them to target diseased myocardium specifically.


Asunto(s)
Modelos Animales de Enfermedad , Células Madre Fetales/citología , Intercambio Materno-Fetal , Isquemia Miocárdica/terapia , Complicaciones Cardiovasculares del Embarazo/terapia , Trasplante de Células Madre , Animales , Femenino , Citometría de Flujo , Técnica del Anticuerpo Fluorescente , Proteínas Fluorescentes Verdes/sangre , Masculino , Ratones , Ratones Endogámicos C57BL , Embarazo
18.
Circ Res ; 110(1): 82-93, 2012 Jan 06.
Artículo en Inglés | MEDLINE | ID: mdl-22082491

RESUMEN

RATIONALE: Fetal cells enter the maternal circulation during pregnancy and may persist in maternal tissue for decades as microchimeras. OBJECTIVE: Based on clinical observations of peripartum cardiomyopathy patients and the high rate of recovery they experience from heart failure, our objective was to determine whether fetal cells can migrate to the maternal heart and differentiate to cardiac cells. METHODS AND RESULTS: We report that fetal cells selectively home to injured maternal hearts and undergo differentiation into diverse cardiac lineages. Using enhanced green fluorescent protein (eGFP)-tagged fetuses, we demonstrate engraftment of multipotent fetal cells in injury zones of maternal hearts. In vivo, eGFP+ fetal cells form endothelial cells, smooth muscle cells, and cardiomyocytes. In vitro, fetal cells isolated from maternal hearts recapitulate these differentiation pathways, additionally forming vascular tubes and beating cardiomyocytes in a fusion-independent manner; ≈40% of fetal cells in the maternal heart express Caudal-related homeobox2 (Cdx2), previously associated with trophoblast stem cells, thought to solely form placenta. CONCLUSIONS: Fetal maternal stem cell transfer appears to be a critical mechanism in the maternal response to cardiac injury. Furthermore, we have identified Cdx2 cells as a novel cell type for potential use in cardiovascular regenerative therapy.


Asunto(s)
Diferenciación Celular/fisiología , Movimiento Celular/fisiología , Células Madre Fetales/citología , Intercambio Materno-Fetal/fisiología , Infarto del Miocardio/patología , Miocardio/citología , Complicaciones Cardiovasculares del Embarazo/patología , Animales , Factor de Transcripción CDX2 , Células Cultivadas , Endotelio Vascular/citología , Femenino , Células Madre Fetales/metabolismo , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Proteínas de Homeodominio/metabolismo , Técnicas In Vitro , Masculino , Ratones , Ratones Endogámicos , Ratones Transgénicos , Modelos Animales , Músculo Liso Vascular/citología , Miocitos Cardíacos/citología , Embarazo , Factores de Transcripción/metabolismo
19.
Ann N Y Acad Sci ; 1211: 113-26, 2010 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-21062300

RESUMEN

Heart disease is the leading cause of death in the industrialized world. This is partially attributed to the inability of cardiomyocytes to divide in a significant manner, and therefore the heart responds to injury through scar formation. One of the challenges of modern medicine is to develop novel therapeutic strategies to facilitate regeneration of cardiac muscle in the diseased heart. Numerous methods have been studied and a wide variety of cell types have been considered. To date, bone marrow stem cells, endogenous populations of cardiac stem cells, embryonic stem cells, and induced pluripotent stem cells have been investigated for their ability to regenerate infarcted myocardium, although stem cell transplantation has produced ambiguous results in human clinical trials. Several studies support another approach that seems very appealing: enhancing the limited endogenous regenerative capacity of the heart. The recent advances in stem cell and regenerative biology are giving rise to the view that cardiac regeneration, although not quite ready for clinical treatment, may translate into therapeutic reality in the not too distant future.


Asunto(s)
Células Madre Hematopoyéticas/fisiología , Miocitos Cardíacos/fisiología , Regeneración/fisiología , Animales , Corazón , Cardiopatías/fisiopatología , Cardiopatías/cirugía , Trasplante de Células Madre Hematopoyéticas/tendencias , Células Madre Hematopoyéticas/citología , Humanos , Miocitos Cardíacos/citología , Fenómenos Fisiológicos/fisiología
20.
Circ Res ; 100(12): 1741-8, 2007 Jun 22.
Artículo en Inglés | MEDLINE | ID: mdl-17495221

RESUMEN

Mammalian myocardial infarction is typically followed by scar formation with eventual ventricular dilation and heart failure. Here we present a novel model system in which mice constitutively expressing cyclin A2 in the myocardium elicit a regenerative response after infarction and exhibit significantly limited ventricular dilation with sustained and remarkably enhanced cardiac function. New cardiomyocyte formation was noted in the infarcted zones as well as cell cycle reentry of periinfarct myocardium with an increase in DNA synthesis and mitotic indices. The enhanced cardiac function was serially assessed over time by MRI. Furthermore, the constitutive expression of cyclin A2 appears to augment endogenous regenerative mechanisms via induction of side population cells with enhanced proliferative capacity. The ability of cultured transgenic cardiomyocytes to undergo cytokinesis provides mechanistic support for the regenerative capacity of cyclin A2.


Asunto(s)
Gasto Cardíaco Bajo/prevención & control , Ciclina A/fisiología , Corazón/fisiología , Infarto del Miocardio/fisiopatología , Miocitos Cardíacos/fisiología , Regeneración/fisiología , Animales , Gasto Cardíaco Bajo/fisiopatología , Ciclo Celular/fisiología , Proliferación Celular , Células Cultivadas , Ciclina A/genética , Ciclina A2 , Regulación de la Expresión Génica , Imagen por Resonancia Magnética , Ratones , Ratones Transgénicos , Índice Mitótico , Infarto del Miocardio/patología , Miocardio/patología , Miocitos Cardíacos/patología , Células Madre/fisiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA