Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 29
Filtrar
Más filtros

Tipo del documento
Intervalo de año de publicación
1.
Cardiology ; 146(2): 144-150, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33326980

RESUMEN

The radial artery is the preferred access site for cardiac catheterization because of patient comfort, early ambulation, and improved survival in acute coronary syndromes, when compared to the femoral artery route. However, it is associated with a high radial artery occlusion (RAO) rate, and patent haemostasis which can reduce this is extremely hard to implement in a busy clinical practice. Smaller sized sheaths are associated with less RAO but are uncommonly used as they could limit procedural prowess and complexity. Alternatively, the distal radial artery (dRA) approach appears to be safer with observed RAO rates of well under 1 percent without compromising benefits offered by the radial artery access. Default dRA can be accessed by palpation alone in most cases with some practice, and this can be improved further with ultrasound guidance. There is a subset of patients, especially in the elderly, where dRA access can be particularly challenging. To mitigate this, we propose a two-step cannulation strategy and illustrate this with a few cases with difficult dRA and radial artery anatomies.


Asunto(s)
Síndrome Coronario Agudo , Arteriopatías Oclusivas , Intervención Coronaria Percutánea , Anciano , Cateterismo Cardíaco , Angiografía Coronaria , Humanos , Arteria Radial/diagnóstico por imagen , Arteria Radial/cirugía , Resultado del Tratamiento , Ultrasonografía
2.
J Cardiovasc Electrophysiol ; 29(11): 1471-1479, 2018 11.
Artículo en Inglés | MEDLINE | ID: mdl-30230101

RESUMEN

OBJECTIVE: We tested whether ablation methodology and study design can explain the varying outcomes in terms of atrial fibrillation (AF)-free survival at 1 year. BACKGROUND: There have been numerous paroxysmal AF ablation trials, which are heterogeneous in their use of different ablation techniques and study design. A useful approach to understanding how these factors influence outcome is to dismantle the trials into individual arms and reconstitute them as a large meta-regression. METHODS: Data were collected from 66 studies (6941 patients). With freedom from AF as the dependent variable, we performed meta-regression using the individual study arm as the unit. RESULTS: Success rates did not change regardless of the technique used to produce pulmonary vein isolation (PVI). Neither was adjunctive lesion sets associated with any improvement in outcome. Studies that included more males and fewer hypertensive patients were found more likely to report better outcomes. The electrocardiography method selected to assess outcome also plays an important role. Outcomes were worse in studies that used regular telemonitoring (by 23%; P < 0.001) or in patients who had implantable loop recorders (by 21%; P = 0.006), rather than those with the less thorough periodic Holter monitoring. CONCLUSIONS: Outcomes of AF ablation studies involving PVI are not affected by the technologies used to produce PVI. Neither do adjunctive lesion sets change the outcome. Achieving high success rates in these studies appears to be dependent more on patient mix and on the thoroughness of AF detection protocols. These should be carefully considered when quoting the success rates of AF ablation procedures that are derived from such studies.


Asunto(s)
Técnicas de Ablación/métodos , Fibrilación Atrial/cirugía , Ablación por Catéter/métodos , Ensayos Clínicos Controlados Aleatorios como Asunto/métodos , Técnicas de Ablación/tendencias , Fibrilación Atrial/diagnóstico , Fibrilación Atrial/mortalidad , Ablación por Catéter/tendencias , Humanos , Selección de Paciente , Tasa de Supervivencia/tendencias , Resultado del Tratamiento
3.
Circulation ; 128(12): 1286-97, 2013 Sep 17.
Artículo en Inglés | MEDLINE | ID: mdl-23983250

RESUMEN

BACKGROUND: Little is known about the function of inositol 1,4,5-trisphosphate receptors (IP3Rs) in the adult heart experimentally. Moreover, whether these Ca(2+) release channels are present and play a critical role in human cardiomyocytes remains to be defined. IP3Rs may be activated after Gαq-protein-coupled receptor stimulation, affecting Ca(2+) cycling, enhancing myocyte performance, and potentially favoring an increase in the incidence of arrhythmias. METHODS AND RESULTS: IP3R function was determined in human left ventricular myocytes, and this analysis was integrated with assays in mouse myocytes to identify the mechanisms by which IP3Rs influence the electric and mechanical properties of the myocardium. We report that IP3Rs are expressed and operative in human left ventricular myocytes. After Gαq-protein-coupled receptor activation, Ca(2+) mobilized from the sarcoplasmic reticulum via IP3Rs contributes to the decrease in resting membrane potential, prolongation of the action potential, and occurrence of early afterdepolarizations. Ca(2+) transient amplitude and cell shortening are enhanced, and extrasystolic and dysregulated Ca(2+) elevations and contractions become apparent. These alterations in the electromechanical behavior of human cardiomyocytes are coupled with increased isometric twitch of the myocardium and arrhythmic events, suggesting that Gαq-protein-coupled receptor activation provides inotropic reserve, which is hampered by electric instability and contractile abnormalities. Additionally, our findings support the notion that increases in Ca(2+) load by IP3Rs promote Ca(2+) extrusion by forward-mode Na(+)/Ca(2+) exchange, an important mechanism of arrhythmic events. CONCLUSIONS: The Gαq-protein/coupled receptor/IP3R axis modulates the electromechanical properties of the human myocardium and its propensity to develop arrhythmias.


Asunto(s)
Potenciales de Acción/fisiología , Señalización del Calcio/fisiología , Insuficiencia Cardíaca/fisiopatología , Receptores de Inositol 1,4,5-Trifosfato/fisiología , Miocitos Cardíacos/fisiología , Adulto , Animales , Arritmias Cardíacas/fisiopatología , Células Cultivadas , Femenino , Subunidades alfa de la Proteína de Unión al GTP Gq-G11/fisiología , Insuficiencia Cardíaca/genética , Ventrículos Cardíacos/citología , Humanos , Receptores de Inositol 1,4,5-Trifosfato/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Persona de Mediana Edad , Contracción Miocárdica/fisiología , Miocitos Cardíacos/metabolismo , Canal Liberador de Calcio Receptor de Rianodina/fisiología , Retículo Sarcoplasmático/fisiología , Transducción de Señal/fisiología
4.
N Engl J Med ; 364(19): 1795-806, 2011 May 12.
Artículo en Inglés | MEDLINE | ID: mdl-21561345

RESUMEN

BACKGROUND: Although progenitor cells have been described in distinct anatomical regions of the lung, description of resident stem cells has remained elusive. METHODS: Surgical lung-tissue specimens were studied in situ to identify and characterize human lung stem cells. We defined their phenotype and functional properties in vitro and in vivo. RESULTS: Human lungs contain undifferentiated human lung stem cells nested in niches in the distal airways. These cells are self-renewing, clonogenic, and multipotent in vitro. After injection into damaged mouse lung in vivo, human lung stem cells form human bronchioles, alveoli, and pulmonary vessels integrated structurally and functionally with the damaged organ. The formation of a chimeric lung was confirmed by detection of human transcripts for epithelial and vascular genes. In addition, the self-renewal and long-term proliferation of human lung stem cells was shown in serial-transplantation assays. CONCLUSIONS: Human lungs contain identifiable stem cells. In animal models, these cells participate in tissue homeostasis and regeneration. They have the undemonstrated potential to promote tissue restoration in patients with lung disease. (Funded by the National Institutes of Health.).


Asunto(s)
Pulmón/citología , Células Madre/fisiología , Adulto , Animales , Células Clonales , Femenino , Humanos , Pulmón/embriología , Pulmón/fisiología , Ratones , Ratones Endogámicos C57BL , Células Madre Pluripotentes , Proteínas Proto-Oncogénicas c-kit/análisis , Regeneración , Trasplante de Células Madre , Células Madre/química
5.
Circ Res ; 110(5): 701-15, 2012 Mar 02.
Artículo en Inglés | MEDLINE | ID: mdl-22275487

RESUMEN

RATIONALE: Embryonic and fetal myocardial growth is characterized by a dramatic increase in myocyte number, but whether the expansion of the myocyte compartment is dictated by activation and commitment of resident cardiac stem cells (CSCs), division of immature myocytes or both is currently unknown. OBJECTIVE: In this study, we tested whether prenatal cardiac development is controlled by activation and differentiation of CSCs and whether division of c-kit-positive CSCs in the mouse heart is triggered by spontaneous Ca(2+) oscillations. METHODS AND RESULTS: We report that embryonic-fetal c-kit-positive CSCs are self-renewing, clonogenic and multipotent in vitro and in vivo. The growth and commitment of c-kit-positive CSCs is responsible for the generation of the myocyte progeny of the developing heart. The close correspondence between values computed by mathematical modeling and direct measurements of myocyte number at E9, E14, E19 and 1 day after birth strongly suggests that the organogenesis of the embryonic heart is dependent on a hierarchical model of cell differentiation regulated by resident CSCs. The growth promoting effects of c-kit-positive CSCs are triggered by spontaneous oscillations in intracellular Ca(2+), mediated by IP3 receptor activation, which condition asymmetrical stem cell division and myocyte lineage specification. CONCLUSIONS: Myocyte formation derived from CSC differentiation is the major determinant of cardiac growth during development. Division of c-kit-positive CSCs in the mouse is promoted by spontaneous Ca(2+) spikes, which dictate the pattern of stem cell replication and the generation of a myocyte progeny at all phases of prenatal life and up to one day after birth.


Asunto(s)
Diferenciación Celular/fisiología , Células Madre Embrionarias/citología , Células Madre Embrionarias/metabolismo , Corazón/embriología , Miocitos Cardíacos/citología , Miocitos Cardíacos/fisiología , Proteínas Proto-Oncogénicas c-kit/metabolismo , Animales , Calcio/metabolismo , Señalización del Calcio/fisiología , Células Cultivadas , Técnicas de Cultivo de Embriones , Receptores de Inositol 1,4,5-Trifosfato/fisiología , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Modelos Animales , Modelos Teóricos , Organogénesis/fisiología , Proteínas Proto-Oncogénicas c-kit/genética
6.
Circ Res ; 111(7): 894-906, 2012 Sep 14.
Artículo en Inglés | MEDLINE | ID: mdl-22851539

RESUMEN

RATIONALE: According to the immortal DNA strand hypothesis, dividing stem cells selectively segregate chromosomes carrying the old template DNA, opposing accumulation of mutations resulting from nonrepaired replication errors and attenuating telomere shortening. OBJECTIVE: Based on the premise of the immortal DNA strand hypothesis, we propose that stem cells retaining the old DNA would represent the most powerful cells for myocardial regeneration. METHODS AND RESULTS: Division of human cardiac stem cells (hCSCs) by nonrandom and random segregation of chromatids was documented by clonal assay of bromodeoxyuridine-tagged hCSCs. Additionally, their growth properties were determined by a series of in vitro and in vivo studies. We report that a small class of hCSCs retain during replication the mother DNA and generate 2 daughter cells, which carry the old and new DNA, respectively. hCSCs with immortal DNA form a pool of nonsenescent cells with longer telomeres and higher proliferative capacity. The self-renewal and long-term repopulating ability of these cells was shown in serial-transplantation assays in the infarcted heart; these cells created a chimeric organ, composed of spared rat and regenerated human cardiomyocytes and coronary vessels, leading to a remarkable restoration of cardiac structure and function. The documentation that hCSCs divide by asymmetrical and symmetrical chromatid segregation supports the view that the human heart is a self-renewing organ regulated by a compartment of resident hCSCs. CONCLUSIONS: The impressive recovery in ventricular hemodynamics and anatomy mediated by clonal hCSCs carrying the "mother" DNA underscores the clinical relevance of this stem cell class for the management of heart failure in humans.


Asunto(s)
Cromátides/fisiología , Segregación Cromosómica/fisiología , Corazón/fisiología , Infarto del Miocardio/terapia , Miocardio/citología , Regeneración/fisiología , Trasplante de Células Madre , Células Madre/citología , Adolescente , Adulto , Anciano , Anciano de 80 o más Años , Animales , Bromodesoxiuridina , Proliferación Celular , Células Cultivadas , Niño , Preescolar , Cromátides/ultraestructura , ADN/fisiología , Femenino , Humanos , Técnicas In Vitro , Lactante , Masculino , Persona de Mediana Edad , Modelos Animales , Infarto del Miocardio/patología , Infarto del Miocardio/fisiopatología , Ratas , Ratas Endogámicas F344 , Células Madre/fisiología , Telómero/ultraestructura , Adulto Joven
7.
Circulation ; 126(15): 1869-81, 2012 Oct 09.
Artículo en Inglés | MEDLINE | ID: mdl-22955965

RESUMEN

BACKGROUND: Two opposite views of cardiac growth are currently held; one views the heart as a static organ characterized by a large number of cardiomyocytes that are present at birth and live as long as the organism, and the other views the heart a highly plastic organ in which the myocyte compartment is restored several times during the course of life. METHODS AND RESULTS: The average age of cardiomyocytes, vascular endothelial cells (ECs), and fibroblasts and their turnover rates were measured by retrospective (14)C birth dating of cells in 19 normal hearts 2 to 78 years of age and in 17 explanted failing hearts 22 to 70 years of age. We report that the human heart is characterized by a significant turnover of ventricular myocytes, ECs, and fibroblasts, physiologically and pathologically. Myocyte, EC, and fibroblast renewal is very high shortly after birth, decreases during postnatal maturation, remains relatively constant in the adult organ, and increases dramatically with age. From 20 to 78 years of age, the adult human heart entirely replaces its myocyte, EC, and fibroblast compartment ≈8, ≈6, and ≈8 times, respectively. Myocyte, EC, and fibroblast regeneration is further enhanced with chronic heart failure. CONCLUSIONS: The human heart is a highly dynamic organ that retains a remarkable degree of plasticity throughout life and in the presence of chronic heart failure. However, the ability to regenerate cardiomyocytes, vascular ECs, and fibroblasts cannot prevent the manifestations of myocardial aging or oppose the negative effects of ischemic and idiopathic dilated cardiomyopathy.


Asunto(s)
Insuficiencia Cardíaca/fisiopatología , Desarrollo de Músculos/fisiología , Miocitos Cardíacos/fisiología , Adolescente , Adulto , Anciano , Envejecimiento , Niño , Preescolar , Células Endoteliales/fisiología , Fibroblastos/fisiología , Corazón/fisiología , Humanos , Persona de Mediana Edad , Miocitos Cardíacos/citología , Regeneración , Donantes de Tejidos , Adulto Joven
8.
Circ Res ; 108(9): 1071-83, 2011 Apr 29.
Artículo en Inglés | MEDLINE | ID: mdl-21415392

RESUMEN

RATIONALE: Understanding the mechanisms that regulate trafficking of human cardiac stem cells (hCSCs) may lead to development of new therapeutic approaches for the failing heart. OBJECTIVE: We tested whether the motility of hCSCs in immunosuppressed infarcted animals is controlled by the guidance system that involves the interaction of Eph receptors with ephrin ligands. METHODS AND RESULTS: Within the cardiac niches, cardiomyocytes expressed preferentially the ephrin A1 ligand, whereas hCSCs possessed the EphA2 receptor. Treatment of hCSCs with ephrin A1 resulted in the rapid internalization of the ephrin A1-EphA2 complex, posttranslational modifications of Src kinases, and morphological changes consistent with the acquisition of a motile cell phenotype. Ephrin A1 enhanced the motility of hCSCs in vitro, and their migration in vivo following acute myocardial infarction. At 2 weeks after infarction, the volume of the regenerated myocardium was 2-fold larger in animals injected with ephrin A1-activated hCSCs than in animals receiving control hCSCs; this difference was dictated by a greater number of newly formed cardiomyocytes and coronary vessels. The increased recovery in myocardial mass with ephrin A1-treated hCSCs was characterized by further restoration of cardiac function and by a reduction in arrhythmic events. CONCLUSIONS: Ephrin A1 promotes the motility of EphA2-positive hCSCs, facilitates their migration to the area of damage, and enhances cardiac repair. Thus, in situ stimulation of resident hCSCs with ephrin A1 or their ex vivo activation before myocardial delivery improves cell targeting to sites of injury, possibly providing a novel strategy for the management of the diseased heart.


Asunto(s)
Efrina-A1/genética , Efrina-A2/genética , Células Madre Hematopoyéticas/citología , Infarto del Miocardio/fisiopatología , Miocitos Cardíacos/citología , Animales , Adhesión Celular/fisiología , Membrana Celular/metabolismo , Movimiento Celular/fisiología , Citoplasma/metabolismo , Efrina-A1/metabolismo , Efrina-A2/metabolismo , Expresión Génica/fisiología , Proteínas Fluorescentes Verdes/genética , Humanos , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Infarto del Miocardio/patología , Infarto del Miocardio/terapia , Ratas , Ratas Endogámicas F344 , Taquicardia Ventricular/patología , Taquicardia Ventricular/fisiopatología , Taquicardia Ventricular/terapia
9.
Circ Res ; 108(12): 1467-81, 2011 Jun 10.
Artículo en Inglés | MEDLINE | ID: mdl-21546606

RESUMEN

RATIONALE: Age and coronary artery disease may negatively affect the function of human cardiac stem cells (hCSCs) and their potential therapeutic efficacy for autologous cell transplantation in the failing heart. OBJECTIVE: Insulin-like growth factor (IGF)-1, IGF-2, and angiotensin II (Ang II), as well as their receptors, IGF-1R, IGF-2R, and AT1R, were characterized in c-kit(+) hCSCs to establish whether these systems would allow us to separate hCSC classes with different growth reserve in the aging and diseased myocardium. METHODS AND RESULTS: C-kit(+) hCSCs were collected from myocardial samples obtained from 24 patients, 48 to 86 years of age, undergoing elective cardiac surgery for coronary artery disease. The expression of IGF-1R in hCSCs recognized a young cell phenotype defined by long telomeres, high telomerase activity, enhanced cell proliferation, and attenuated apoptosis. In addition to IGF-1, IGF-1R(+) hCSCs secreted IGF-2 that promoted myocyte differentiation. Conversely, the presence of IGF-2R and AT1R, in the absence of IGF-1R, identified senescent hCSCs with impaired growth reserve and increased susceptibility to apoptosis. The ability of IGF-1R(+) hCSCs to regenerate infarcted myocardium was then compared with that of unselected c-kit(+) hCSCs. IGF-1R(+) hCSCs improved cardiomyogenesis and vasculogenesis. Pretreatment of IGF-1R(+) hCSCs with IGF-2 resulted in the formation of more mature myocytes and superior recovery of ventricular structure. CONCLUSIONS: hCSCs expressing only IGF-1R synthesize both IGF-1 and IGF-2, which are potent modulators of stem cell replication, commitment to the myocyte lineage, and myocyte differentiation, which points to this hCSC subset as the ideal candidate cell for the management of human heart failure.


Asunto(s)
Enfermedad de la Arteria Coronaria/metabolismo , Infarto del Miocardio/metabolismo , Miocardio/metabolismo , Miocitos Cardíacos/metabolismo , Receptor IGF Tipo 1/metabolismo , Regeneración , Células Madre/metabolismo , Angiotensina II/metabolismo , Diferenciación Celular , Enfermedad de la Arteria Coronaria/patología , Enfermedad de la Arteria Coronaria/terapia , Femenino , Humanos , Factor I del Crecimiento Similar a la Insulina/biosíntesis , Factor II del Crecimiento Similar a la Insulina/metabolismo , Masculino , Infarto del Miocardio/patología , Infarto del Miocardio/terapia , Miocardio/patología , Miocitos Cardíacos/patología , Receptor IGF Tipo 2/metabolismo , Trasplante de Células Madre , Células Madre/patología , Trasplante Autólogo
10.
Circulation ; 123(12): 1287-96, 2011 Mar 29.
Artículo en Inglés | MEDLINE | ID: mdl-21403094

RESUMEN

BACKGROUND: Cardiac stem cells (CSCs) delivered to the infarcted heart generate a large number of small fetal-neonatal cardiomyocytes that fail to acquire the differentiated phenotype. However, the interaction of CSCs with postmitotic myocytes results in the formation of cells with adult characteristics. METHODS AND RESULTS: On the basis of results of in vitro and in vivo assays, we report that the commitment of human CSCs (hCSCs) to the myocyte lineage and the generation of mature working cardiomyocytes are influenced by microRNA-499 (miR-499), which is barely detectable in hCSCs but is highly expressed in postmitotic human cardiomyocytes. miR-499 traverses gap junction channels and translocates to structurally coupled hCSCs favoring their differentiation into functionally competent cells. Expression of miR-499 in hCSCs represses the miR-499 target genes Sox6 and Rod1, enhancing cardiomyogenesis in vitro and after infarction in vivo. Although cardiac repair was detected in all cell-treated infarcted hearts, the aggregate volume of the regenerated myocyte mass and myocyte cell volume were greater in animals injected with hCSCs overexpressing miR-499. Treatment with hCSCs resulted in an improvement in ventricular function, consisting of a better preservation of developed pressure and positive and negative dP/dt after infarction. An additional positive effect on cardiac performance occurred with miR-499, pointing to enhanced myocyte differentiation/hypertrophy as the mechanism by which miR-499 potentiated the restoration of myocardial mass and function in the infarcted heart. CONCLUSIONS: The recognition that miR-499 promotes the differentiation of hCSCs into mechanically integrated cardiomyocytes has important clinical implications for the treatment of human heart failure.


Asunto(s)
Células Madre Adultas/citología , MicroARNs/fisiología , Infarto del Miocardio/terapia , Miocitos Cardíacos/citología , Trasplante de Células Madre , Células Madre Adultas/fisiología , Animales , Diferenciación Celular/fisiología , División Celular/fisiología , Células Cultivadas , Técnicas de Cocultivo , Modelos Animales de Enfermedad , Uniones Comunicantes/fisiología , Expresión Génica/fisiología , Humanos , Infarto del Miocardio/patología , Miocitos Cardíacos/fisiología , Proteína de Unión al Tracto de Polipirimidina , Proteínas de Unión al ARN/genética , Ratas , Regeneración/fisiología , Factores de Transcripción SOXD/genética
11.
Circ Res ; 107(2): 305-15, 2010 Jul 23.
Artículo en Inglés | MEDLINE | ID: mdl-20522802

RESUMEN

RATIONALE: The ability of the human heart to regenerate large quantities of myocytes remains controversial, and the extent of myocyte renewal claimed by different laboratories varies from none to nearly 20% per year. OBJECTIVE: To address this issue, we examined the percentage of myocytes, endothelial cells, and fibroblasts labeled by iododeoxyuridine in postmortem samples obtained from cancer patients who received the thymidine analog for therapeutic purposes. Additionally, the potential contribution of DNA repair, polyploidy, and cell fusion to the measurement of myocyte regeneration was determined. METHODS AND RESULTS: The fraction of myocytes labeled by iododeoxyuridine ranged from 2.5% to 46%, and similar values were found in fibroblasts and endothelial cells. An average 22%, 20%, and 13% new myocytes, fibroblasts, and endothelial cells were generated per year, suggesting that the lifespan of these cells was approximately 4.5, 5, and 8 years, respectively. The newly formed cardiac cells showed a fully differentiated adult phenotype and did not express the senescence-associated protein p16(INK4a). Moreover, measurements by confocal microscopy and flow cytometry documented that the human heart is composed predominantly of myocytes with 2n diploid DNA content and that tetraploid and octaploid nuclei constitute only a small fraction of the parenchymal cell pool. Importantly, DNA repair, ploidy formation, and cell fusion were not implicated in the assessment of myocyte regeneration. CONCLUSIONS: Our findings indicate that the human heart has a significant growth reserve and replaces its myocyte and nonmyocyte compartment several times during the course of life.


Asunto(s)
Proliferación Celular , Células Endoteliales/patología , Fibroblastos/patología , Desarrollo de Músculos , Miocardio/patología , Miocitos Cardíacos/patología , Neoplasias/patología , Adulto , Factores de Edad , Anciano , Animales , Autopsia , Muerte Celular , Fusión Celular , Núcleo Celular/patología , Proliferación Celular/efectos de los fármacos , Reparación del ADN , Células Endoteliales/efectos de los fármacos , Femenino , Fibroblastos/efectos de los fármacos , Citometría de Flujo , Humanos , Idoxuridina/uso terapéutico , Inmunohistoquímica , Masculino , Microscopía Confocal , Persona de Mediana Edad , Desarrollo de Músculos/efectos de los fármacos , Miocitos Cardíacos/efectos de los fármacos , Neoplasias/tratamiento farmacológico , Fenotipo , Poliploidía , Fármacos Sensibilizantes a Radiaciones/uso terapéutico , Ratas , Ratas Endogámicas F344 , Regeneración , Factores de Tiempo , Adulto Joven
12.
Circ Res ; 107(3): 429-41, 2010 Aug 06.
Artículo en Inglés | MEDLINE | ID: mdl-20558824

RESUMEN

RATIONALE: Physiological hypertrophy in the developing heart has been considered the product of an increase in volume of preexisting fetal cardiomyocytes in the absence of myocyte formation. OBJECTIVE: In this study, we tested whether the mouse heart at birth has a pool of cardiac stem cells (CSCs) that differentiate into myocytes contributing to the postnatal expansion of the parenchymal cell compartment. METHODS AND RESULTS: We have found that the newborn heart contains a population of c-kit-positive CSCs that are lineage negative, self-renewing, and multipotent. CSCs express the Notch1 receptor and show the nuclear localization of its active fragment, N1ICD. In 60% of cases, N1ICD was coupled with the presence of Nkx2.5, indicating that the commitment of CSCs to the myocyte lineage is regulated by Notch1. Importantly, overexpression of N1ICD in neonatal CSCs significantly expanded the proportion of transit-amplifying myocytes. To establish whether these in vitro findings had a functional counterpart in vivo, the Notch pathway was blocked in newborn mice by administration of a gamma-secretase inhibitor. This intervention resulted in the development of a dilated myopathy and high mortality rates. Ventricular decompensation was characterized by a 62% reduction in amplifying myocytes, which resulted in a 54% decrease in myocyte number. After cessation of Notch blockade and recovery of myocyte regeneration, cardiac anatomy and function were largely restored. CONCLUSIONS: Notch1 signaling is a critical determinant of CSC growth and differentiation; when this cascade of events is altered, cardiomyogenesis is impaired, physiological cardiac hypertrophy is prevented, and a life-threatening myopathy supervenes.


Asunto(s)
Cardiomiopatía Dilatada/etiología , Miocitos Cardíacos/citología , Miocitos Cardíacos/fisiología , Receptor Notch1/antagonistas & inhibidores , Actinina/metabolismo , Actinas/metabolismo , Animales , Animales Recién Nacidos , Capilares/citología , Capilares/fisiología , Cardiomiopatía Dilatada/fisiopatología , Diferenciación Celular , División Celular , Corazón/crecimiento & desarrollo , Humanos , Recién Nacido , Ratones , Receptor Notch1/fisiología , Receptores Notch/antagonistas & inhibidores , Receptores Notch/fisiología , Células Madre/citología , Células Madre/fisiología , Factores de Transcripción/metabolismo
13.
Proc Natl Acad Sci U S A ; 106(40): 17169-74, 2009 Oct 06.
Artículo en Inglés | MEDLINE | ID: mdl-19805158

RESUMEN

An analysis of the clonality of cardiac progenitor cells (CPCs) and myocyte turnover in vivo requires genetic tagging of the undifferentiated cells so that the clonal marker of individual mother cells is traced in the specialized progeny. CPC niches in the atria and apex of the mouse heart were infected with a lentivirus carrying EGFP, and the destiny of the tagged cells was determined 1-5 months later. A common integration site was identified in isolated CPCs, cardiomyocytes, endothelial cells (ECs), and fibroblasts, documenting CPC self-renewal and multipotentiality and the clonal origin of the differentiated cell populations. Subsequently, the degree of EGFP-lentiviral infection of CPCs was evaluated 2-4 days after injection, and the number of myocytes expressing the reporter gene was measured 6 months later. A BrdU pulse-chasing protocol was also introduced as an additional assay for the analysis of myocyte turnover. Over a period of 6 months, each EGFP-positive CPC divided approximately eight times generating 230 cardiomyocytes; this value was consistent with the number of newly formed cells labeled by BrdU. To determine whether, human CPCs (hCPCs) are self-renewing and multipotent, these cells were transduced with the EGFP-lentivirus and injected after acute myocardial infarction in immunosuppressed rats. hCPCs, myocytes, ECs, and fibroblasts collected from the regenerated myocardium showed common viral integration sites in the human genome. Thus, our results indicate that the adult heart contains a pool of resident stem cells that regulate cardiac homeostasis and repair.


Asunto(s)
Diferenciación Celular , Proliferación Celular , Miocitos Cardíacos/citología , Miocitos Cardíacos/metabolismo , Células 3T3 , Animales , Secuencia de Bases , Linaje de la Célula , Células Clonales/citología , Células Clonales/metabolismo , Células Endoteliales/citología , Células Endoteliales/metabolismo , Femenino , Fibroblastos/citología , Fibroblastos/metabolismo , Vectores Genéticos/genética , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Humanos , Inmunohistoquímica , Lentivirus/genética , Ratones , Datos de Secuencia Molecular , Miocardio/citología , Miocardio/metabolismo , Cadenas Pesadas de Miosina/genética , Cadenas Pesadas de Miosina/metabolismo , Ratas , Ratas Endogámicas F344 , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Factores de Tiempo
14.
Circulation ; 121(2): 276-92, 2010 Jan 19.
Artículo en Inglés | MEDLINE | ID: mdl-20038740

RESUMEN

BACKGROUND: Anthracyclines are the most effective drugs available in the treatment of neoplastic diseases; however, they have profound consequences on the structure and function of the heart, which over time cause a cardiomyopathy that leads to congestive heart failure. METHODS AND RESULTS: Administration of doxorubicin in rats led to a dilated myopathy, heart failure, and death. To test whether the effects of doxorubicin on cardiac anatomy and function were mediated by alterations in cardiac progenitor cells (CPCs), these cells were exposed to the anthracycline, which increased the formation of reactive oxygen species and caused increases in DNA damage, expression of p53, telomere attrition, and apoptosis. Additionally, doxorubicin resulted in cell-cycle arrest at the G2/M transition, which led to a significant decrease in CPC growth. Doxorubicin elicited multiple molecular adaptations; the massive apoptotic death that occurred in CPCs in the presence of anthracycline imposed on the surviving CPC pool the activation of several pathways aimed at preservation of the primitive state, cell division, lineage differentiation, and repair of damaged DNA. To establish whether delivery of syngeneic progenitor cells opposed the progression of doxorubicin cardiotoxicity, enhanced green fluorescent protein-labeled CPCs were injected in the failing myocardium; this treatment promoted regeneration of cardiomyocytes and vascular structures, which improved ventricular performance and rate of animal survival. CONCLUSIONS: Our results raise the possibility that autologous CPCs can be obtained before antineoplastic drugs are given to cancer patients and subsequently administered to individuals who are particularly sensitive to the cardiotoxicity of these agents for prevention or management of heart failure.


Asunto(s)
Antraciclinas/efectos adversos , Cardiomiopatías/inducido químicamente , Cardiomiopatías/terapia , Regeneración , Trasplante de Células Madre , Células Madre/efectos de los fármacos , Animales , Cardiomiopatías/patología , Cardiomiopatía Dilatada/inducido químicamente , Cardiomiopatía Dilatada/patología , Cardiomiopatía Dilatada/terapia , Recuento de Células , Doxorrubicina/efectos adversos , Insuficiencia Cardíaca/inducido químicamente , Insuficiencia Cardíaca/patología , Insuficiencia Cardíaca/terapia , Humanos , Miocitos Cardíacos , Neoplasias/complicaciones , Neoplasias/tratamiento farmacológico , Ratas , Células Madre/fisiología
15.
Circ Res ; 105(8): 764-74, 2009 Oct 09.
Artículo en Inglés | MEDLINE | ID: mdl-19745162

RESUMEN

RATIONALE: The adult heart possesses a pool of progenitor cells stored in myocardial niches, but the mechanisms involved in the activation of this cell compartment are currently unknown. OBJECTIVE: Ca2+ promotes cell growth raising the possibility that changes in intracellular Ca2+ initiate division of c-kit-positive human cardiac progenitor cells (hCPCs) and determine their fate. METHODS AND RESULTS: Ca2+ oscillations were identified in hCPCs and these events occurred independently from coupling with cardiomyocytes or the presence of extracellular Ca2+. These findings were confirmed in the heart of transgenic mice in which enhanced green fluorescent protein was under the control of the c-kit promoter. Ca2+ oscillations in hCPCs were regulated by the release of Ca2+ from the endoplasmic reticulum through activation of inositol 1,4,5-triphosphate receptors (IP3Rs) and the reuptake of Ca2+ by the sarco-/endoplasmic reticulum Ca2+ pump (SERCA). IP3Rs and SERCA were highly expressed in hCPCs, whereas ryanodine receptors were not detected. Although Na+-Ca2+ exchanger, store-operated Ca2+ channels and plasma membrane Ca2+ pump were present and functional in hCPCs, they had no direct effects on Ca2+ oscillations. Conversely, Ca2+ oscillations and their frequency markedly increased with ATP and histamine which activated purinoceptors and histamine-1 receptors highly expressed in hCPCs. Importantly, Ca2+ oscillations in hCPCs were coupled with the entry of cells into the cell cycle and 5-bromodeoxyuridine incorporation. Induction of Ca2+ oscillations in hCPCs before their intramyocardial delivery to infarcted hearts was associated with enhanced engraftment and expansion of these cells promoting the generation of a large myocyte progeny. CONCLUSION: IP3R-mediated Ca2+ mobilization control hCPC growth and their regenerative potential.


Asunto(s)
Relojes Biológicos/fisiología , Calcio/metabolismo , Miocardio/metabolismo , Miocitos Cardíacos/metabolismo , Células Madre/metabolismo , Adenosina Trifosfato/farmacología , Adulto , Animales , Retículo Endoplásmico/metabolismo , Regulación de la Expresión Génica/efectos de los fármacos , Regulación de la Expresión Génica/fisiología , Histamina/farmacología , Humanos , Receptores de Inositol 1,4,5-Trifosfato/metabolismo , Ratones , Ratones Transgénicos , Miocardio/citología , Miocitos Cardíacos/citología , Proteínas Proto-Oncogénicas c-kit/metabolismo , Receptores Histamínicos/metabolismo , Receptores Purinérgicos/metabolismo , ATPasas Transportadoras de Calcio del Retículo Sarcoplásmico/metabolismo , Células Madre/citología
17.
J Biomed Biotechnol ; 2010: 807084, 2010.
Artículo en Inglés | MEDLINE | ID: mdl-20625419

RESUMEN

Although systole was for long considered the core of cardiac function, hemodynamic performance is evenly dependent on appropriate systolic and diastolic functions. The recognition that isolated diastolic dysfunction is the major culprit for approximately fifty percent of all heart failure cases imposes a deeper understanding of its underlying mechanisms so that better diagnostic and therapeutic strategies can be designed. Risk factors leading to diastolic dysfunction affect myocardial relaxation and/or its material properties by disrupting the homeostasis of cardiomyocytes as well as their relation with surrounding matrix and vascular structures. As a consequence, slower ventricular relaxation and higher myocardial stiffness may result in higher ventricular filling pressures and in the risk of hemodynamic decompensation. Thus, determining the mechanisms of diastolic function and their implications in the pathophysiology of heart failure with normal ejection fraction has become a prominent field in basic and clinical research.


Asunto(s)
Diástole/fisiología , Miocardio/metabolismo , Animales , Matriz Extracelular/metabolismo , Insuficiencia Cardíaca/fisiopatología , Humanos , Miocitos Cardíacos/metabolismo , Factores de Riesgo , Volumen Sistólico/fisiología
18.
Circ Res ; 103(1): 107-16, 2008 Jul 03.
Artículo en Inglés | MEDLINE | ID: mdl-18556576

RESUMEN

Ischemic heart disease is characterized chronically by a healed infarct, foci of myocardial scarring, cavitary dilation, and impaired ventricular performance. These alterations can only be reversed by replacement of scarred tissue with functionally competent myocardium. We tested whether cardiac progenitor cells (CPCs) implanted in proximity of healed infarcts or resident CPCs stimulated locally by hepatocyte growth factor and insulin-like growth factor-1 invade the scarred myocardium and generate myocytes and coronary vessels improving the hemodynamics of the infarcted heart. Hepatocyte growth factor is a powerful chemoattractant of CPCs, and insulin-like growth factor-1 promotes their proliferation and survival. Injection of CPCs or growth factors led to the replacement of approximately 42% of the scar with newly formed myocardium, attenuated ventricular dilation and prevented the chronic decline in function of the infarcted heart. Cardiac repair was mediated by the ability of CPCs to synthesize matrix metalloproteinases that degraded collagen proteins, forming tunnels within the fibrotic tissue during their migration across the scarred myocardium. New myocytes had a 2n karyotype and possessed 2 sex chromosomes, excluding cell fusion. Clinically, CPCs represent an ideal candidate cell for cardiac repair in patients with chronic heart failure. CPCs may be isolated from myocardial biopsies and, following their expansion in vitro, administered back to the same patients avoiding the adverse effects associated with the use of nonautologous cells. Alternatively, growth factors may be delivered locally to stimulate resident CPCs and promote myocardial regeneration. These forms of treatments could be repeated over time to reduce progressively tissue scarring and expand the working myocardium.


Asunto(s)
Cicatriz/terapia , Insuficiencia Cardíaca/terapia , Infarto del Miocardio/terapia , Miocardio , Trasplante de Células Madre , Células Madre , Animales , Movimiento Celular/efectos de los fármacos , Enfermedad Crónica , Cicatriz/etiología , Cicatriz/metabolismo , Cicatriz/patología , Colágeno/metabolismo , Colagenasas/biosíntesis , Diploidia , Insuficiencia Cardíaca/metabolismo , Insuficiencia Cardíaca/patología , Hemodinámica , Factor de Crecimiento de Hepatocito/metabolismo , Factor de Crecimiento de Hepatocito/farmacología , Humanos , Factor I del Crecimiento Similar a la Insulina/metabolismo , Factor I del Crecimiento Similar a la Insulina/farmacología , Infarto del Miocardio/complicaciones , Infarto del Miocardio/metabolismo , Infarto del Miocardio/patología , Miocardio/metabolismo , Miocardio/patología , Miocitos Cardíacos/metabolismo , Miocitos Cardíacos/patología , Ratas , Regeneración/efectos de los fármacos , Trasplante de Células Madre/métodos , Células Madre/metabolismo , Células Madre/patología , Trasplante Homólogo , Disfunción Ventricular/etiología , Disfunción Ventricular/metabolismo , Disfunción Ventricular/patología , Disfunción Ventricular/terapia
19.
Peptides ; 30(2): 419-25, 2009 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-19028535

RESUMEN

Diastole plays a central role in cardiovascular homeostasis. Its two main determinants, myocardial relaxation and passive properties of the ventricular wall, are nowadays regarded as physiological mechanisms susceptible of active modulation. Furthermore, diastolic dysfunction and heart failure with normal ejection fraction (previously called diastolic heart failure) are two subjects of major clinical relevance and an intense area of research. The role of several neurohumoral mediators like angiotensin-II and endothelin-1 on the modulation of diastolic function was systematically described as having only chronic deleterious effects such as cardiac hypertrophy and fibrosis. However, over the last years a growing body of evidence described a new role for several peptides on the acute modulation of diastolic function. In the acute setting, some of these mediators may have the potential to induce an adaptive cardiac response. In this review, we describe the role of angiotensin-II, endothelin-1, nitric oxide, urotensin-II and ghrelin on the acute modulation of diastolic function, emphasizing its pathophysiological relevance. Only a thorough understanding of diastolic physiology as well as its active modulation, both in the acute and chronic settings, will improve our knowledge on diastolic dysfunction and allow us to solve the enigmas of heart failure with normal ejection fraction.


Asunto(s)
Diástole/fisiología , Insuficiencia Cardíaca Diastólica/metabolismo , Neurotransmisores/metabolismo , Angiotensina II/metabolismo , Animales , Endotelina-1/metabolismo , Ghrelina/metabolismo , Humanos , Contracción Miocárdica/fisiología , Óxido Nítrico/metabolismo , Urotensinas/metabolismo
20.
JACC Clin Electrophysiol ; 5(8): 968-976, 2019 08.
Artículo en Inglés | MEDLINE | ID: mdl-31439299

RESUMEN

OBJECTIVES: This meta-analysis examined the ability of pulmonary vein isolation (PVI) to prevent atrial fibrillation in randomized controlled trials (RCTs) in which the patients not receiving PVI nevertheless underwent a procedure. BACKGROUND: PVI is a commonly used procedure for the treatment of atrial fibrillation (AF), and its efficacy has usually been judged against therapy with anti-arrhythmic drugs in open-label trials. There have been several RCTs of AF ablation in which both arms received an ablation, but the difference between the treatment arms was inclusion or omission of PVI. These trials of an ablation strategy with PVI versus an ablation strategy without PVI may provide a more rigorous method for evaluating the efficacy of PVI. METHODS: Medline and Cochrane databases were searched for RCTs comparing ablation including PVI with ablation excluding PVI. The primary efficacy endpoint was freedom from atrial fibrillation (AF) and atrial tachycardia at 12 months. A random-effects meta-analysis was performed using the restricted maximum likelihood estimator. RESULTS: Overall, 6 studies (n = 610) met inclusion criteria. AF recurrence was significantly lower with an ablation including PVI than an ablation without PVI (RR: 0.54; 95% confidence interval [CI]: 0.33 to 0.89; p = 0.0147; I2 = 79.7%). Neither the type of AF (p = 0.48) nor the type of non-PVI ablation (p = 0.21) was a significant moderator of the effect size. In 3 trials the non-PVI ablation procedure was performed in both arms, whereas PVI was performed in only 1 arm. In these studies, AF recurrence was significantly lower when PVI was included (RR: 0.32; 95% CI: 0.14 to 0.73; p = 0.007, I2 78%). CONCLUSIONS: In RCTs where both arms received an ablation, and therefore an expectation amongst patients and doctors of benefit, being randomized to PVI had a striking effect, reducing AF recurrence by a half.


Asunto(s)
Fibrilación Atrial/cirugía , Ablación por Catéter , Venas Pulmonares/cirugía , Ablación por Catéter/efectos adversos , Ablación por Catéter/métodos , Ablación por Catéter/estadística & datos numéricos , Femenino , Humanos , Masculino , Persona de Mediana Edad , Ensayos Clínicos Controlados Aleatorios como Asunto , Recurrencia
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA