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.
J Mol Cell Cardiol ; 182: 86-91, 2023 09.
Artículo en Inglés | MEDLINE | ID: mdl-37517369

RESUMEN

Although the myocardial renewal rate in the adult mammalian heart is quite low, recent studies have identified genetic variants which can impact the degree of cardiomyocyte cell cycle reentry. Here we use the compound interest law to model the level of regenerative growth over time in mice exhibiting different rates of cardiomyocyte cell cycle reentry following myocardial injury. The modeling suggests that the limited ability of S-phase adult cardiomyocytes to progress through cytokinesis, rather than the ability to reenter the cell cycle per se, is a major contributor to the low levels of intrinsic regenerative growth in the adult myocardium.


Asunto(s)
Lesiones Cardíacas , Miocitos Cardíacos , Ratones , Animales , Miocitos Cardíacos/metabolismo , Miocardio/metabolismo , Corazón , Ciclo Celular , Lesiones Cardíacas/metabolismo , Citocinesis , Proliferación Celular , Mamíferos
2.
Circulation ; 147(2): 142-153, 2023 01 10.
Artículo en Inglés | MEDLINE | ID: mdl-36382596

RESUMEN

BACKGROUND: Identifying genetic variants that affect the level of cell cycle reentry and establishing the degree of cell cycle progression in those variants could help guide development of therapeutic interventions aimed at effecting cardiac regeneration. We observed that C57Bl6/NCR (B6N) mice have a marked increase in cardiomyocyte S-phase activity after permanent coronary artery ligation compared with infarcted DBA/2J (D2J) mice. METHODS: Cardiomyocyte cell cycle activity after infarction was monitored in D2J, (D2J×B6N)-F1, and (D2J×B6N)-F1×D2J backcross mice by means of bromodeoxyuridine or 5-ethynyl-2'-deoxyuridine incorporation using a nuclear-localized transgenic reporter to identify cardiomyocyte nuclei. Genome-wide quantitative trait locus analysis, fine scale genetic mapping, whole exome sequencing, and RNA sequencing analyses of the backcross mice were performed to identify the gene responsible for the elevated cardiomyocyte S-phase phenotype. RESULTS: (D2J×B6N)-F1 mice exhibited a 14-fold increase in cardiomyocyte S-phase activity in ventricular regions remote from infarct scar compared with D2J mice (0.798±0.09% versus 0.056±0.004%; P<0.001). Quantitative trait locus analysis of (D2J×B6N)-F1×D2J backcross mice revealed that the gene responsible for differential S-phase activity was located on the distal arm of chromosome 3 (logarithm of the odds score=6.38; P<0.001). Additional genetic and molecular analyses identified 3 potential candidates. Of these, Tnni3k (troponin I-interacting kinase) is expressed in B6N hearts but not in D2J hearts. Transgenic expression of TNNI3K in a D2J genetic background results in elevated cardiomyocyte S-phase activity after injury. Cardiomyocyte S-phase activity in both Tnni3k-expressing and Tnni3k-nonexpressing mice results in the formation of polyploid nuclei. CONCLUSIONS: These data indicate that Tnni3k expression increases the level of cardiomyocyte S-phase activity after injury.


Asunto(s)
Miocitos Cardíacos , Troponina I , Ratones , Animales , Troponina I/metabolismo , Ratones Endogámicos DBA , Miocitos Cardíacos/metabolismo , Ciclo Celular , Proliferación Celular , Proteínas Serina-Treonina Quinasas/genética , Proteínas Serina-Treonina Quinasas/metabolismo
4.
Dev Cell ; 44(4): 433-446.e7, 2018 02 26.
Artículo en Inglés | MEDLINE | ID: mdl-29486195

RESUMEN

Correlative evidence suggests that polyploidization of heart muscle, which occurs naturally in post-natal mammals, creates a barrier to heart regeneration. Here, we move beyond a correlation by demonstrating that experimental polyploidization of zebrafish cardiomyocytes is sufficient to suppress their proliferative potential during regeneration. Initially, we determined that zebrafish myocardium becomes susceptible to polyploidization upon transient cytokinesis inhibition mediated by dominant-negative Ect2. Using a transgenic strategy, we generated adult animals containing mosaic hearts composed of differentially labeled diploid and polyploid-enriched cardiomyocyte populations. Diploid cardiomyocytes outcompeted their polyploid neighbors in producing regenerated heart muscle. Moreover, hearts composed of equivalent proportions of diploid and polyploid cardiomyocytes failed to regenerate altogether, demonstrating that a critical percentage of diploid cardiomyocytes is required to achieve heart regeneration. Our data identify cardiomyocyte polyploidization as a barrier to heart regeneration and suggest that mobilizing rare diploid cardiomyocytes in the human heart will improve its regenerative capacity.


Asunto(s)
Animales Modificados Genéticamente/fisiología , Corazón/embriología , Infarto del Miocardio/patología , Miocardio/citología , Poliploidía , Regeneración/fisiología , Pez Cebra/fisiología , Animales , Animales Modificados Genéticamente/embriología , Proliferación Celular , Células Cultivadas , Corazón/fisiología , Infarto del Miocardio/metabolismo , Miocardio/metabolismo , Pez Cebra/embriología , Proteínas de Pez Cebra/genética , Proteínas de Pez Cebra/metabolismo
5.
Cardiovasc Res ; 114(3): 389-400, 2018 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-29016731

RESUMEN

AIMS: Recent studies have demonstrated electrotonic coupling between scar tissue and the surrounding myocardium in cryoinjured hearts. However, the electrical dynamics occurring at the myocyte-nonmyocyte interface in the fibrotic heart remain undefined. Here, we sought to develop an assay to interrogate the nonmyocyte cell type contributing to heterocellular coupling and to characterize, on a cellular scale, its voltage response in the infarct border zone of living hearts. METHODS AND RESULTS: We used two-photon laser scanning microscopy in conjunction with a voltage-sensitive dye to record transmembrane voltage changes simultaneously from cardiomyocytes and adjoined nonmyocytes in Langendorff-perfused mouse hearts with healing myocardial infarction. Transgenic mice with cardiomyocyte-restricted expression of a green fluorescent reporter protein underwent permanent coronary artery ligation and their hearts were subjected to voltage imaging 7-10 days later. Reporter-negative cells, i.e. nonmyocytes, in the infarct border zone exhibited depolarizing transients at a 1:1 coupling ratio with action potentials recorded simultaneously from adjacent, reporter-positive ventricular myocytes. The electrotonic responses in the nonmyocytes exhibited slower rates of de- and repolarization compared to the action potential waveform of juxtaposed myocytes. Voltage imaging in infarcted hearts expressing a fluorescent reporter specifically in myofibroblasts revealed that the latter were electrically coupled to border zone myocytes. Their voltage transient properties were indistinguishable from those of nonmyocytes in hearts with cardiomyocyte-restricted reporter expression. The density of connexin43 expression at myofibroblast-cardiomyocyte junctions was ∼5% of that in the intercalated disc regions of paired ventricular myocytes in the remote, uninjured myocardium, whereas the ratio of connexin45 to connexin43 expression levels at heterocellular contacts was ∼1%. CONCLUSION: Myofibroblasts contribute to the population of electrically coupled nonmyocytes in the infarct border zone. The slower kinetics of myofibroblast voltage responses may reflect low electrical conductivity across heterocellular junctions, in accordance with the paucity of connexin expression at myofibroblast-cardiomyocyte contacts.


Asunto(s)
Potenciales de Acción , Comunicación Celular , Infarto del Miocardio/metabolismo , Miocitos Cardíacos/metabolismo , Miofibroblastos/metabolismo , Animales , Conexina 43/metabolismo , Conexinas/metabolismo , Modelos Animales de Enfermedad , Conductividad Eléctrica , Genes Reporteros , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Preparación de Corazón Aislado , Cinética , Ratones Transgénicos , Microscopía Confocal , Microscopía de Fluorescencia por Excitación Multifotónica , Infarto del Miocardio/patología , Miocitos Cardíacos/patología , Miofibroblastos/patología
7.
PLoS One ; 10(6): e0131511, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26110646

RESUMEN

Parthenogenetic stem cells (PSCs) are a promising candidate donor for cell therapy applications. Similar to embryonic stem cells (ESCs) and induced pluripotent stem cells (iPSCs), PSCs exhibit self-renewing capacity and clonogenic proliferation in vitro. PSCs exhibit largely haploidentical genotype, and as such may constitute an attractive population for allogenic applications. In this study, PSCs isolated from transgenic mice carrying a cardiomyocyte-restricted reporter transgene to permit tracking of donor cells were genetically modified to carry a cardiomyocyte-restricted aminoglycoside phosphotransferase expression cassette (MHC-neor/pGK-hygror) to permit the generation of highly enriched cardiomyocyte cultures from spontaneously differentiating PSCs by simple selection with the neomycin analogue G148. Following engraftment into isogenic recipient hearts, the selected cardiomyocytes formed a functional syncytium with the host myocardium as evidenced by the presence of entrained intracellular calcium transients. These cells thus constitute a potential source of therapeutic donor cells.


Asunto(s)
Miocitos Cardíacos/citología , Células Madre/citología , Animales , Diferenciación Celular , Proliferación Celular , Trasplante de Células/métodos , Electroporación , Femenino , Genotipo , Proteínas Fluorescentes Verdes/metabolismo , Kanamicina Quinasa/genética , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos DBA , Ratones Transgénicos , Miocardio/metabolismo , Partenogénesis , Reacción en Cadena de la Polimerasa , Transfección , Transgenes
8.
PLoS One ; 9(12): e115871, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25545368

RESUMEN

OBJECTIVES: Neuregulin 1 signaling plays an important role in cardiac trabecular development, and in sustaining functional integrity in adult hearts. Treatment with neuregulin 1 enhances adult cardiomyocyte differentiation, survival and/or function in vitro and in vivo. It has also been suggested that recombinant neuregulin 1ß1 (NRG1ß1) induces cardiomyocyte proliferation in normal and injured adult hearts. Here we further explore the impact of neuregulin 1 signaling on adult cardiomyocyte cell cycle activity. METHODS AND RESULTS: Adult mice were subjected to 9 consecutive daily injections of recombinant NRG1ß1 or vehicle, and cardiomyocyte DNA synthesis was quantitated via bromodeoxyuridine (BrdU) incorporation, which was delivered using mini-osmotic pumps over the entire duration of NRG1ß1 treatment. NRG1ß1 treatment inhibited baseline rates of cardiomyocyte DNA synthesis in normal mice (cardiomyocyte labelling index: 0.019±0.005% vs. 0.003±0.001%, saline vs. NRG1ß1, P<0.05). Acute NRG1ß1 treatment did result in activation of Erk1/2 and cardiac myosin regulatory light chain (down-stream mediators of neuregulin signalling), as well as activation of DNA synthesis in non-cardiomyocytes, validating the biological activity of the recombinant protein. In other studies, mice were subjected to permanent coronary artery occlusion, and cardiomyocyte DNA synthesis was monitored via tritiated thymidine incorporation which was delivered as a single injection 7 days post-infarction. Daily NRG1ß1 treatment had no impact on cardiomyocyte DNA synthesis in the infarcted myocardium (cardiomyocyte labelling index: 0.039±0.011% vs. 0.027±0.021%, saline vs. NRG1ß1, P>0.05). SUMMARY: These data indicate that NRG1ß1 treatment does not increase cardiomyocyte DNA synthesis (and consequently does not increase the rate of cardiomyocyte renewal) in normal or infarcted adult mouse hearts. Thus, any improvement in cardiac structure and function observed following neuregulin treatment of injured hearts likely occurs independently of overt myocardial regeneration.


Asunto(s)
Replicación del ADN/efectos de los fármacos , Infarto del Miocardio/tratamiento farmacológico , Neurregulina-1/farmacología , Animales , ADN/biosíntesis , Ratones Endogámicos DBA , Ratones Transgénicos , Infarto del Miocardio/metabolismo , Miocitos Cardíacos/efectos de los fármacos , Neurregulina-1/uso terapéutico , Proteínas Recombinantes/farmacología , Proteínas Recombinantes/uso terapéutico
9.
J Neurosci Methods ; 221: 48-61, 2014 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-24056230

RESUMEN

BACKGROUND: Sympathetic nerve wiring in the mammalian heart has remained largely unexplored. Resolving the wiring diagram of the cardiac sympathetic network would help establish the structural underpinnings of neurocardiac coupling. NEW METHOD: We used two-photon excitation fluorescence microscopy, combined with a computer-assisted 3-D tracking algorithm, to map the local sympathetic circuits in living hearts from adult transgenic mice expressing enhanced green fluorescent protein (EGFP) in peripheral adrenergic neurons. RESULTS: Quantitative co-localization analyses confirmed that the intramyocardial EGFP distribution recapitulated the anatomy of the sympathetic arbor. In the left ventricular subepicardium of the uninjured heart, the sympathetic network was composed of multiple subarbors, exhibiting variable branching and looping topology. Axonal branches did not overlap with each other within their respective parental subarbor nor with neurites of annexed subarbors. The sympathetic network in the border zone of a 2-week-old myocardial infarction was characterized by substantive rewiring, which included spatially heterogeneous loss and gain of sympathetic fibers and formation of multiple, predominately nested, axon loops of widely variable circumference and geometry. COMPARISON WITH EXISTING METHODS: In contrast to mechanical tissue sectioning methods that may involve deformation of tissue and uncertainty in registration across sections, our approach preserves continuity of structure, which allows tracing of neurites over distances, and thus enables derivation of the three-dimensional and topological morphology of cardiac sympathetic nerves. CONCLUSIONS: Our assay should be of general utility to unravel the mechanisms governing sympathetic axon spacing during development and disease.


Asunto(s)
Algoritmos , Corazón/inervación , Imagenología Tridimensional/métodos , Sistema Nervioso Simpático/anatomía & histología , Animales , Proteínas Fluorescentes Verdes/genética , Humanos , Ratones , Ratones Transgénicos , Microscopía Fluorescente
10.
J Clin Invest ; 123(3): 1285-98, 2013 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-23434590

RESUMEN

Uniparental parthenotes are considered an unwanted byproduct of in vitro fertilization. In utero parthenote development is severely compromised by defective organogenesis and in particular by defective cardiogenesis. Although developmentally compromised, apparently pluripotent stem cells can be derived from parthenogenetic blastocysts. Here we hypothesized that nonembryonic parthenogenetic stem cells (PSCs) can be directed toward the cardiac lineage and applied to tissue-engineered heart repair. We first confirmed similar fundamental properties in murine PSCs and embryonic stem cells (ESCs), despite notable differences in genetic (allelic variability) and epigenetic (differential imprinting) characteristics. Haploidentity of major histocompatibility complexes (MHCs) in PSCs is particularly attractive for allogeneic cell-based therapies. Accordingly, we confirmed acceptance of PSCs in MHC-matched allotransplantation. Cardiomyocyte derivation from PSCs and ESCs was equally effective. The use of cardiomyocyte-restricted GFP enabled cell sorting and documentation of advanced structural and functional maturation in vitro and in vivo. This included seamless electrical integration of PSC-derived cardiomyocytes into recipient myocardium. Finally, we enriched cardiomyocytes to facilitate engineering of force-generating myocardium and demonstrated the utility of this technique in enhancing regional myocardial function after myocardial infarction. Collectively, our data demonstrate pluripotency, with unrestricted cardiogenicity in PSCs, and introduce this unique cell type as an attractive source for tissue-engineered heart repair.


Asunto(s)
Células Madre Embrionarias/fisiología , Corazón/fisiología , Ingeniería de Tejidos , Potenciales de Acción , Animales , Antígenos de Diferenciación/genética , Antígenos de Diferenciación/metabolismo , Señalización del Calcio , Diferenciación Celular , Forma de la Célula , Células Cultivadas , Células Madre Embrionarias/metabolismo , Epigénesis Genética , Genotipo , Histocompatibilidad/genética , Antígenos de Histocompatibilidad Clase II/genética , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Endogámicos DBA , Ratones SCID , Contracción Miocárdica , Miocitos Cardíacos/metabolismo , Miocitos Cardíacos/fisiología , Técnicas de Cultivo de Órganos , Organoides/trasplante , Partenogénesis , Fenotipo , Trasplante de Células Madre , Trasplante Homólogo
11.
Biochim Biophys Acta ; 1833(4): 799-803, 2013 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-23142641

RESUMEN

Interventions to effect therapeutic cardiomyocyte renewal have received considerable interest of late. Such interventions, if successful, could give rise to myocardial regeneration in diseased hearts. Regenerative interventions fall into two broad categories, namely approaches based on promoting renewal of pre-existing cardiomyocytes and approaches based on cardiomyogenic stem cell activity. The latter category can be further subdivided into approaches promoting differentiation of endogenous cardiomyogenic stem cells, approaches wherein cardiomyogenic stem cells are harvested, amplified or enriched ex vivo, and subsequently engrafted into the heart, and approaches wherein an exogenous stem cell is induced to differentiate in vitro, and the resulting cardiomyocytes are engrafted into the heart. There is disagreement in the literature regarding the degree to which cardiomyocyte renewal occurs in the normal and injured heart, the mechanism(s) by which this occurs, and the degree to which therapeutic interventions can enhance regenerative growth. This review discusses several caveats which are encountered when attempting to measure cardiomyocyte renewal in vivo which likely contribute, at least in part, to the disagreement regarding the levels at which this occurs in normal, injured and treated hearts. This article is part of a Special Issue entitled: Cardiomyocyte biology: Cardiac pathways of differentiation, metabolism and contraction.


Asunto(s)
Miocardio/citología , Miocitos Cardíacos/citología , Células Madre/citología , Animales , Bromodesoxiuridina , Diferenciación Celular , Núcleo Celular/metabolismo , Núcleo Celular/ultraestructura , Proliferación Celular , Rastreo Celular , Genes Reporteros , Integrasas , Ratones , Ratones Transgénicos , Miocardio/metabolismo , Miocitos Cardíacos/metabolismo , Regeneración , Células Madre/metabolismo , beta-Galactosidasa
12.
Pediatr Cardiol ; 33(6): 929-37, 2012 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-22481568

RESUMEN

Although the adult mammalian myocardium exhibits a limited ability to undergo regenerative growth, its intrinsic renewal rate is insufficient to compensate for myocyte loss during cardiac disease. Transplantation of donor cardiomyocytes or cardiomyogenic stem cells is considered a promising strategy for reconstitution of cardiac mass, provided the engrafted cells functionally integrate with host myocardium and actively contribute to its contractile force. The authors previously developed a two-photon fluorescence microscopy-based assay that allows in situ screening of donor cell function after intracardiac delivery of the cells. This report reviews the techniques of two-photon fluorescence microscopy and summarizes its application for quantifying the extent to which a variety of donor cell types stably and functionally couple with the recipient myocardium.


Asunto(s)
Calcio/metabolismo , Microscopía Fluorescente/métodos , Miocardio/citología , Miocitos Cardíacos/trasplante , Animales , Humanos , Ratones , Miocardio/metabolismo , Miocitos Cardíacos/metabolismo , Trasplantes
13.
Eur Heart J ; 33(1): 129-37, 2012 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-21849352

RESUMEN

AIMS: Although pharmacological interventions that mobilize stem cells and enhance their homing to damaged tissue can limit adverse post-myocardial infarction (MI) remodelling, cardiomyocyte renewal with this approach is limited. While experimental cell cycle induction can promote cardiomyocyte renewal following MI, this process must compete with the more rapid processes of scar formation and adverse remodelling. The current study tested the hypothesis that the combination of enhanced stem cell mobilization/homing and cardiomyocyte cell cycle induction would result in increased myocardial renewal in injured hearts. METHODS AND RESULTS: Myocardial infarction was induced by coronary artery ligation in adult MHC-cycD2 transgenic mice (which exhibit constitutive cardiomyocyte cell cycle activity) and their non-transgenic littermates. Mice were then treated with saline or with granulocyte colony-stimulating factor (G-CSF) plus the dipeptidylpeptidase-IV (DPP-IV) inhibitor Diprotin A (DipA) for 7 days. Infarct thickness and cardiomyocyte number/infarct/section were significantly improved in MHC-cycD2 mice with G-CSF plus DipA treatment when compared with MHC-cycD2 transgene expression or G-CSF plus DipA treatment alone. Echocardiographic analyses revealed that stem cell mobilization/homing and cardiomyocyte cell cycle activation had an additive effect on functional recovery. CONCLUSION: These data strongly suggest that G-CSF plus DPP-IV inhibition, combined with cardiomyocyte cell cycle activation, leads to enhanced myocardial regeneration following MI. The data are also consistent with the notion that altering adverse post-injury remodelling renders the myocardium more permissive for cardiomyocyte repopulation.


Asunto(s)
Inhibidores de la Dipeptidil-Peptidasa IV/farmacología , Factor Estimulante de Colonias de Granulocitos/farmacología , Corazón/fisiología , Infarto del Miocardio/fisiopatología , Regeneración/fisiología , Animales , Ciclina D2/metabolismo , Combinación de Medicamentos , Movilización de Célula Madre Hematopoyética/métodos , Ratones , Ratones Transgénicos , Miocitos Cardíacos/metabolismo , Distribución Aleatoria , Volumen Sistólico/fisiología
14.
Am J Physiol Cell Physiol ; 298(6): C1603-9, 2010 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-20457832

RESUMEN

Accurate nuclear identification is crucial for distinguishing the role of cardiac myocytes in intrinsic and experimentally induced regenerative growth of the myocardium. Conventional histologic analysis of myocyte nuclei relies on the optical sectioning capabilities of confocal microscopy in conjunction with immunofluorescent labeling of cytoplasmic proteins such as troponin T, and dyes that bind to double-strand DNA to identify nuclei. Using heart sections from transgenic mice in which the cardiomyocyte-restricted alpha-cardiac myosin heavy chain promoter targeted the expression of nuclear localized beta-galactosidase reporter in >99% of myocytes, we systematically compared the fidelity of conventional myocyte nuclear identification using confocal microscopy, with and without the aid of a membrane marker. The values obtained with these assays were then compared with those obtained with anti-beta-galactosidase immune reactivity in the same samples. In addition, we also studied the accuracy of anti-GATA4 immunoreactivity for myocyte nuclear identification. Our results demonstrate that, although these strategies are capable of identifying myocyte nuclei, the level of interobserver agreement and margin of error can compromise accurate identification of rare events, such as cardiomyocyte apoptosis and proliferation. Thus these data indicate that morphometric approaches based on segmentation are justified only if the margin of error for measuring the event in question has been predetermined and deemed to be small and uniform. We also illustrate the value of a transgene-based approach to overcome these intrinsic limitations of identifying myocyte nuclei. This latter approach should prove quite useful when measuring rare events.


Asunto(s)
Núcleo Celular/metabolismo , Inmunohistoquímica , Microscopía Confocal , Miocitos Cardíacos/metabolismo , Coloración y Etiquetado/métodos , Animales , Biomarcadores/metabolismo , Miosinas Cardíacas/genética , Proliferación Celular , Factor de Transcripción GATA4/metabolismo , Genes Reporteros , Ratones , Ratones Endogámicos DBA , Ratones Transgénicos , Cadenas Pesadas de Miosina/genética , Variaciones Dependientes del Observador , Regiones Promotoras Genéticas , Reproducibilidad de los Resultados , Troponina T/metabolismo , Aglutininas del Germen de Trigo , beta-Galactosidasa/biosíntesis , beta-Galactosidasa/genética
15.
Circulation ; 119(1): 99-106, 2009 Jan 06.
Artículo en Inglés | MEDLINE | ID: mdl-19103993

RESUMEN

BACKGROUND: Doxorubicin is used to treat childhood and adult cancer. Doxorubicin treatment is associated with both acute and chronic cardiotoxicity. The cardiotoxic effects of doxorubicin are cumulative, which limits its chemotherapeutic dose. Free radical generation and p53-dependent apoptosis are thought to contribute to doxorubicin-induced cardiotoxicity. METHODS AND RESULTS: Adult transgenic (MHC-CB7) mice expressing cardiomyocyte-restricted dominant-interfering p53 and their nontransgenic littermates were treated with doxorubicin (20 mg/kg cumulative dose). Nontransgenic mice exhibited reduced left ventricular systolic function (predoxorubicin fractional shortening [FS] 61+/-2%, postdoxorubicin FS 45+/-2%, mean+/-SEM, P<0.008), reduced cardiac mass, and high levels of cardiomyocyte apoptosis 7 days after the initiation of doxorubicin treatment. In contrast, doxorubicin-treated MHC-CB7 mice exhibited normal left ventricular systolic function (predoxorubicin FS 63+/-2%, postdoxorubicin FS 60+/-2%, P>0.008), normal cardiac mass, and low levels of cardiomyocyte apoptosis. Western blot analyses indicated that mTOR (mammalian target of rapamycin) signaling was inhibited in doxorubicin-treated nontransgenic mice but not in doxorubicin-treated MHC-CB7 mice. Accordingly, transgenic mice with cardiomyocyte-restricted, constitutively active mTOR expression (MHC-mTORca) were studied. Left ventricular systolic function (predoxorubicin FS 64+/-2%, postdoxorubicin FS 60+/-3%, P>0.008) and cardiac mass were normal in doxorubicin-treated MHC-mTORca mice, despite levels of cardiomyocyte apoptosis similar to those seen in doxorubicin-treated nontransgenic mice. CONCLUSIONS: These data suggest that doxorubicin treatment induces acute cardiac dysfunction and reduces cardiac mass via p53-dependent inhibition of mTOR signaling and that loss of myocardial mass, and not cardiomyocyte apoptosis, is the major contributor to acute doxorubicin cardiotoxicity.


Asunto(s)
Antibióticos Antineoplásicos/toxicidad , Proteínas Portadoras/metabolismo , Doxorrubicina/toxicidad , Cardiopatías/inducido químicamente , Cardiopatías/metabolismo , Fosfotransferasas (Aceptor de Grupo Alcohol)/metabolismo , Proteína p53 Supresora de Tumor/metabolismo , Enfermedad Aguda , Animales , Apoptosis , Proteínas Portadoras/genética , Cardiopatías/patología , Ratones , Ratones Endogámicos DBA , Ratones Transgénicos , Miocitos Cardíacos/efectos de los fármacos , Miocitos Cardíacos/metabolismo , Miocitos Cardíacos/patología , Fosfotransferasas (Aceptor de Grupo Alcohol)/genética , Transducción de Señal/efectos de los fármacos , Transducción de Señal/fisiología , Serina-Treonina Quinasas TOR , Proteína p53 Supresora de Tumor/genética
16.
PLoS One ; 3(4): e1929, 2008 Apr 16.
Artículo en Inglés | MEDLINE | ID: mdl-18414652

RESUMEN

AIMS: Cultured cardiac explants produce a heterogeneous population of cells including a distinctive population of refractile cells described here as small round cardiac explant derived cells (EDCs). The aim of this study was to explore the source, morphology and cardiogenic potential of EDCs. METHODS: Transgenic MLC2v-Cre/ZEG, and actin-eGFP mice were used for lineage-tracing of EDCs in vitro and in vivo. C57B16 mice were used as cell transplant recipients of EDCs from transgenic hearts, as well as for the general characterisation of EDCs. The activation of cardiac-specific markers were analysed by: immunohistochemistry with bright field and immunofluorescent microscopy, electron microscopy, PCR and RT-PCR. Functional engraftment of transplanted cells was further investigated with calcium transient studies. RESULTS: Production of EDCs was highly dependent on the retention of blood-derived cells or factors in the cultured explants. These cells shared some characteristics of cardiac myocytes in vitro and survived engraftment in the adult heart in vivo. However, EDCs failed to differentiate into functional cardiac myocytes in vivo as demonstrated by the absence of stimulation-evoked intracellular calcium transients following transplantation into the peri-infarct zone. CONCLUSIONS: This study highlights that positive identification based upon one parameter alone such as morphology or immunofluorescene is not adequate to identify the source, fate and function of adult cardiac explant derived cells.


Asunto(s)
Corazón/anatomía & histología , Miocardio/citología , Miocitos Cardíacos/citología , Animales , Calcio/metabolismo , Linaje de la Célula , Células Cultivadas , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Microscopía Electrónica , Microscopía Fluorescente , Perfusión , Reacción en Cadena de la Polimerasa , Transgenes
17.
Nature ; 453(7194): 524-8, 2008 May 22.
Artículo en Inglés | MEDLINE | ID: mdl-18432194

RESUMEN

The functional heart is comprised of distinct mesoderm-derived lineages including cardiomyocytes, endothelial cells and vascular smooth muscle cells. Studies in the mouse embryo and the mouse embryonic stem cell differentiation model have provided evidence indicating that these three lineages develop from a common Flk-1(+) (kinase insert domain protein receptor, also known as Kdr) cardiovascular progenitor that represents one of the earliest stages in mesoderm specification to the cardiovascular lineages. To determine whether a comparable progenitor is present during human cardiogenesis, we analysed the development of the cardiovascular lineages in human embryonic stem cell differentiation cultures. Here we show that after induction with combinations of activin A, bone morphogenetic protein 4 (BMP4), basic fibroblast growth factor (bFGF, also known as FGF2), vascular endothelial growth factor (VEGF, also known as VEGFA) and dickkopf homolog 1 (DKK1) in serum-free media, human embryonic-stem-cell-derived embryoid bodies generate a KDR(low)/C-KIT(CD117)(neg) population that displays cardiac, endothelial and vascular smooth muscle potential in vitro and, after transplantation, in vivo. When plated in monolayer cultures, these KDR(low)/C-KIT(neg) cells differentiate to generate populations consisting of greater than 50% contracting cardiomyocytes. Populations derived from the KDR(low)/C-KIT(neg) fraction give rise to colonies that contain all three lineages when plated in methylcellulose cultures. Results from limiting dilution studies and cell-mixing experiments support the interpretation that these colonies are clones, indicating that they develop from a cardiovascular colony-forming cell. Together, these findings identify a human cardiovascular progenitor that defines one of the earliest stages of human cardiac development.


Asunto(s)
Células Madre Embrionarias/citología , Células Madre Embrionarias/metabolismo , Miocitos Cardíacos/citología , Receptor 2 de Factores de Crecimiento Endotelial Vascular/metabolismo , Activinas/farmacología , Proteína Morfogenética Ósea 4 , Proteínas Morfogenéticas Óseas/farmacología , Diferenciación Celular/efectos de los fármacos , Línea Celular , Linaje de la Célula/efectos de los fármacos , Células Madre Embrionarias/efectos de los fármacos , Células Madre Embrionarias/trasplante , Factor 2 de Crecimiento de Fibroblastos/farmacología , Humanos , Péptidos y Proteínas de Señalización Intercelular/farmacología , Miocitos Cardíacos/efectos de los fármacos , Miocitos Cardíacos/metabolismo , Técnicas de Placa-Clamp , Proteínas Proto-Oncogénicas c-kit/genética , Factor A de Crecimiento Endotelial Vascular/farmacología , Receptor 2 de Factores de Crecimiento Endotelial Vascular/deficiencia , Receptor 2 de Factores de Crecimiento Endotelial Vascular/genética
18.
Mol Ther ; 16(6): 1129-37, 2008 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-18431364

RESUMEN

The cardiomyogenic potential of adult bone marrow (BM) cells after being directly transplanted into the ischemically injured heart remains a controversial issue. In this study, we investigated the ability of transplanted BM cells to develop intracellular calcium ([Ca(2+)](i)) transients in response to membrane depolarization in situ. Low-density mononuclear (LDM) BM cells, c-kit-enriched (c-kit(enr)) BM cells, and highly enriched lin(-) c-kit(+) BM cells were obtained from adult transgenic mice ubiquitously expressing enhanced green fluorescent protein (EGFP), and injected into peri-infarct myocardiums of nontransgenic mice. After 9-10 days the mice were killed, and the hearts were removed, perfused in Langendorff mode, loaded with the calcium-sensitive fluorophore rhod-2, and subjected to two-photon laser scanning fluorescence microscopy (TPLSM) to monitor action potential-induced [Ca(2+)](i) transients in EGFP-expressing donor-derived cells and non-expressing host cardiomyocytes. Whereas spontaneous and electrically evoked [Ca(2+)](i) transients were found to occur synchronously in host cardiomyocytes along the graft-host border and in areas remote from the infarct, they were absent in all of the >3,000 imaged BM-derived cells that were located in clusters throughout the infarct scar or peri-infarct zone. We conclude that engrafted BM-derived cells lack attributes of functioning cardiomyocytes, calling into question the concept that adult BM cells can give rise to substantive cardiomyocyte regeneration within the infarcted heart.


Asunto(s)
Células de la Médula Ósea/citología , Infarto del Miocardio/terapia , Miocardio/metabolismo , Miocitos Cardíacos/citología , Animales , Antígenos CD34/biosíntesis , Calcio/metabolismo , Linaje de la Célula , Trasplante de Células , Pollos , Proteínas Fluorescentes Verdes/metabolismo , Ratones , Ratones Endogámicos C57BL , Microscopía Fluorescente , Modelos Biológicos
19.
Cardiovasc Res ; 78(1): 18-25, 2008 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-18079102

RESUMEN

AIMS: Cardiomyocyte loss is a major contributor to the decreased cardiac function observed in diseased hearts. Previous studies have shown that cardiomyocyte-restricted cyclin D2 expression resulted in sustained cell cycle activity following myocardial injury in transgenic (MHC-cycD2) mice. Here, we investigated the effects of this cell cycle activation on cardiac function following myocardial infarction (MI). METHODS AND RESULTS: MI was induced in transgenic and non-transgenic mice by left coronary artery occlusion. At 7, 60, and 180 days after MI, left ventricular pressure-volume measurements were recorded and histological analysis was performed. MI had a similar adverse effect on cardiac function in transgenic and non-transgenic mice at 7 days post-injury. No improvement in cardiac function was observed in non-transgenic mice at 60 and 180 days post-MI. In contrast, the transgenic animals exhibited a progressive and marked increase in cardiac function at subsequent time points. Improved cardiac function in the transgenic mice at 60 and 180 days post-MI correlated positively with the presence of newly formed myocardial tissue which was not apparent at 7 days post-MI. Intracellular calcium transient imaging indicated that cardiomyocytes present in the newly formed myocardium participated in a functional syncytium with the remote myocardium. CONCLUSION: These findings indicate that cardiomyocyte cell cycle activation leads to improvement of cardiac function and morphology following MI and may represent an important clinical strategy to promote myocardial regeneration.


Asunto(s)
Ciclo Celular , Proliferación Celular , Ciclinas/metabolismo , Infarto del Miocardio/metabolismo , Miocitos Cardíacos/metabolismo , Función Ventricular Izquierda , Animales , Señalización del Calcio , Ciclina D2 , Ciclinas/genética , Modelos Animales de Enfermedad , Masculino , Ratones , Ratones Endogámicos DBA , Ratones Transgénicos , Infarto del Miocardio/patología , Infarto del Miocardio/fisiopatología , Miocitos Cardíacos/patología , Cadenas Pesadas de Miosina/genética , Regiones Promotoras Genéticas , Regeneración , Factores de Tiempo , Miosinas Ventriculares/genética , Presión Ventricular
20.
Circ Res ; 96(1): 110-8, 2005 Jan 07.
Artículo en Inglés | MEDLINE | ID: mdl-15576649

RESUMEN

Restriction point transit and commitment to a new round of cell division is regulated by the activity of cyclin-dependent kinase 4 and its obligate activating partners, the D-type cyclins. In this study, we examined the ability of D-type cyclins to promote cardiomyocyte cell cycle activity. Adult transgenic mice expressing cyclin D1, D2, or D3 under the regulation of the alpha cardiac myosin heavy chain promoter exhibited high rates of cardiomyocyte DNA synthesis under baseline conditions. Cardiac injury in mice expressing cyclin D1 or D3 resulted in cytoplasmic cyclin D accumulation, with a concomitant reduction in the level of cardiomyocyte DNA synthesis. In contrast, cardiac injury in mice expressing cyclin D2 did not alter subcellular cyclin localization. Consequently, cardiomyocyte cell cycle activity persisted in injured hearts expressing cyclin D2, ultimately resulting in infarct regression. These data suggested that modulation of D-type cyclins could be exploited to promote regenerative growth in injured hearts.


Asunto(s)
Ciclinas/fisiología , Replicación del ADN , Terapia Genética , Infarto del Miocardio/terapia , Miocitos Cardíacos/metabolismo , Factores de Edad , Animales , Animales Recién Nacidos , Cardiomegalia/inducido químicamente , Enfermedad Coronaria/complicaciones , Enfermedad Coronaria/metabolismo , Ciclina D1/genética , Ciclina D1/fisiología , Ciclina D2 , Ciclina D3 , Quinasa 4 Dependiente de la Ciclina , Quinasas Ciclina-Dependientes/metabolismo , Ciclinas/biosíntesis , Ciclinas/genética , Electrocoagulación/efectos adversos , Fibroblastos/metabolismo , Regulación de la Expresión Génica , Lesiones Cardíacas/genética , Lesiones Cardíacas/metabolismo , Lesiones Cardíacas/patología , Lesiones Cardíacas/terapia , Isoproterenol/toxicidad , Ligadura , Ratones , Ratones Endogámicos DBA , Ratones Transgénicos , Infarto del Miocardio/genética , Infarto del Miocardio/metabolismo , Infarto del Miocardio/patología , Miocitos Cardíacos/patología , Cadenas Pesadas de Miosina/genética , Regiones Promotoras Genéticas , Proteínas Proto-Oncogénicas/metabolismo , Proteínas Recombinantes de Fusión/fisiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA