Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
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
3.
Cardiovasc Res ; 115(5): 960-965, 2019 04 15.
Artículo en Inglés | MEDLINE | ID: mdl-30423020

RESUMEN

AIMS: Doxorubicin (DOX) is a widely used and effective anti-cancer therapeutic. DOX treatment is associated with both acute and late onset cardiotoxicity, limiting its overall efficacy. Here, the impact of cardiomyocyte cell cycle activation was examined in a juvenile model featuring aspects of acute and late onset DOX cardiotoxicity. METHODS AND RESULTS: Two-week old MHC-cycD2 transgenic mice (which express cyclin D2 in postnatal cardiomyocytes and exhibit sustained cardiomyocyte cell cycle activity; D2 mice) and their wild type (WT) littermates received weekly DOX injections for 5 weeks (25 mg/kg cumulative dose). One week after the last DOX treatment (acute stage), cardiac function was suppressed in both groups. Acute DOX cardiotoxicity in D2 and WT mice was associated with similar increases in the levels of cardiomyocyte apoptosis and Ku70/Ku80 expression (markers of DNA damage and oxidative stress), as well as similar reductions in hypertrophic cardiomyocyte growth. Cardiac dysfunction persisted in WT mice for 13 weeks following the last DOX treatment (late stage) and was accompanied by increased levels of cardiomyocyte apoptosis, Ku expression, and myocardial fibrosis. In contrast, D2 mice exhibited a progressive recovery in cardiac function, which was indistinguishable from saline-treated animals by 9 weeks following the last DOX treatment. Improved cardiac function was accompanied by reductions in the levels of late stage cardiomyocyte apoptosis, Ku expression, and myocardial fibrosis. CONCLUSION: These data suggest that cardiomyocyte cell cycle activity can promote recovery of cardiac function and preserve cardiac structure following DOX treatment.


Asunto(s)
Ciclina D2/metabolismo , Doxorrubicina , Insuficiencia Cardíaca/metabolismo , Miocitos Cardíacos/metabolismo , Animales , Apoptosis , Cardiotoxicidad , Ciclina D2/genética , Modelos Animales de Enfermedad , Fibrosis , Insuficiencia Cardíaca/inducido químicamente , Insuficiencia Cardíaca/genética , Insuficiencia Cardíaca/fisiopatología , Autoantígeno Ku/metabolismo , Ratones Endogámicos DBA , Ratones Transgénicos , Miocitos Cardíacos/patología , Recuperación de la Función , Regeneración , Transducción de Señal , Factores de Tiempo , Remodelación Ventricular
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
6.
J Clin Invest ; 127(12): 4285-4296, 2017 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-29083322

RESUMEN

Induction of the cell cycle is emerging as an intervention to treat heart failure. Here, we tested the hypothesis that enhanced cardiomyocyte renewal in transgenic mice expressing cyclin D2 would be beneficial during hemodynamic overload. We induced pressure overload by transthoracic aortic constriction (TAC) or volume overload by aortocaval shunt in cyclin D2-expressing and WT mice. Although cyclin D2 expression dramatically improved survival following TAC, it did not confer a survival advantage to mice following aortocaval shunt. Cardiac function decreased following TAC in WT mice, but was preserved in cyclin D2-expressing mice. On the other hand, cardiac structure and function were compromised in response to aortocaval shunt in both WT and cyclin D2-expressing mice. The preserved function and improved survival in cyclin D2-expressing mice after TAC was associated with an approximately 50% increase in cardiomyocyte number and exaggerated cardiac hypertrophy, as indicated by increased septum thickness. Aortocaval shunt did not further impact cardiomyocyte number in mice expressing cyclin D2. Following TAC, cyclin D2 expression attenuated cardiomyocyte hypertrophy, reduced cardiomyocyte apoptosis, fibrosis, calcium/calmodulin-dependent protein kinase IIδ phosphorylation, brain natriuretic peptide expression, and sustained capillarization. Thus, we show that cyclin D2-induced cardiomyocyte renewal reduced myocardial remodeling and dysfunction after pressure overload but not after volume overload.


Asunto(s)
Enfermedades de la Aorta/metabolismo , Cardiomegalia/metabolismo , Proliferación Celular , Ciclina D2/metabolismo , Insuficiencia Cardíaca/prevención & control , Miocitos Cardíacos/metabolismo , Animales , Enfermedades de la Aorta/genética , Enfermedades de la Aorta/patología , Cardiomegalia/genética , Cardiomegalia/patología , Constricción Patológica , Ciclina D2/genética , Insuficiencia Cardíaca/genética , Insuficiencia Cardíaca/metabolismo , Ratones , Ratones Transgénicos , Miocitos Cardíacos/patología
7.
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
8.
Anat Rec (Hoboken) ; 295(2): 234-48, 2012 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-22095914

RESUMEN

The paucity of mammalian adult cardiac myocytes (CM) proliferation following myocardial infarction (MI) and the remodeling of the necrotic tissue that ensues, result in non-regenerative repair. In contrast, zebrafish (ZF) can regenerate after an apical resection or cryoinjury of the heart. There is considerable interest in models where regeneration proceeds in the presence of necrotic tissue. We have developed and characterized a cautery injury model in the giant danio (GD), a species closely related to ZF, where necrotic tissue remains part of the ventricle, yet regeneration occurs. By light and transmission electron microscopy (TEM), we have documented four temporally overlapping processes: (1) a robust inflammatory response analogous to that observed in MI, (2) concomitant proliferation of epicardial cells leading to wound closure, (3) resorption of necrotic tissue and its replacement by granulation tissue, and (4) regeneration of the myocardial tissue driven by 5-EDU and [(3) H]thymidine incorporating CMs. In conclusion, our data suggest that the GD possesses robust repair mechanisms in the ventricle and can serve as an important model of cardiac inflammation, remodeling and regeneration.


Asunto(s)
Miocitos Cardíacos/patología , Regeneración/fisiología , Remodelación Ventricular/fisiología , Pez Cebra/fisiología , Animales , Proliferación Celular , Desoxiuridina/análogos & derivados , Desoxiuridina/metabolismo , Modelos Animales de Enfermedad , Tejido de Granulación/patología , Inflamación/patología , Miocitos Cardíacos/metabolismo , Miocitos Cardíacos/ultraestructura , Necrosis , Neovascularización Patológica/metabolismo , Neovascularización Patológica/patología , Pericardio/patología , Timidina/metabolismo , Cicatrización de Heridas/fisiología
9.
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
10.
Hum Mol Genet ; 20(23): 4582-96, 2011 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-21880664

RESUMEN

Arrhythmogenic right ventricular cardiomyopathy (ARVC) is an inheritable myocardial disorder associated with fibrofatty replacement of myocardium and ventricular arrhythmia. A subset of ARVC is categorized as Naxos disease, which is characterized by ARVC and a cutaneous disorder. A homozygous loss-of-function mutation of the Plakoglobin (Jup) gene, which encodes a major component of the desmosome and the adherens junction, had been identified in Naxos patients, although the underlying mechanism remained elusive. We generated Jup mutant mice by ablating Jup in cardiomyocytes. Jup mutant mice largely recapitulated the clinical manifestation of human ARVC: ventricular dilation and aneurysm, cardiac fibrosis, cardiac dysfunction and spontaneous ventricular arrhythmias. Ultra-structural analyses revealed that desmosomes were absent in Jup mutant myocardia, whereas adherens junctions and gap junctions were preserved. We found that ventricular arrhythmias were associated with progressive cardiomyopathy and fibrosis in Jup mutant hearts. Massive cell death contributed to the cardiomyocyte dropout in Jup mutant hearts. Despite the increase of ß-catenin at adherens junctions in Jup mutant cardiomyoicytes, the Wnt/ß-catenin-mediated signaling was not altered. Transforming growth factor-beta-mediated signaling was found significantly elevated in Jup mutant cardiomyocytes at the early stage of cardiomyopathy, suggesting an important pathogenic pathway for Jup-related ARVC. These findings have provided further insights for the pathogenesis of ARVC and potential therapeutic interventions.


Asunto(s)
Displasia Ventricular Derecha Arritmogénica/patología , Miocitos Cardíacos/metabolismo , Miocitos Cardíacos/patología , gamma Catenina/deficiencia , Animales , Arritmias Cardíacas/complicaciones , Arritmias Cardíacas/fisiopatología , Displasia Ventricular Derecha Arritmogénica/complicaciones , Displasia Ventricular Derecha Arritmogénica/fisiopatología , Muerte Celular , Desmosomas/metabolismo , Desmosomas/ultraestructura , Fibrosis , Eliminación de Gen , Sistema de Conducción Cardíaco/patología , Sistema de Conducción Cardíaco/fisiopatología , Humanos , Ratones , Ratones Mutantes , Miocardio/metabolismo , Miocardio/patología , Miocitos Cardíacos/ultraestructura , Especificidad de Órganos , Sarcómeros/metabolismo , Sarcómeros/ultraestructura , Vía de Señalización Wnt , gamma Catenina/metabolismo
11.
Circulation ; 122(10): 993-1003, 2010 Sep 07.
Artículo en Inglés | MEDLINE | ID: mdl-20733099

RESUMEN

BACKGROUND: Hemodynamic load regulates myocardial function and gene expression. We tested the hypothesis that afterload and preload, despite similar average load, result in different phenotypes. METHODS AND RESULTS: Afterload and preload were compared in mice with transverse aortic constriction (TAC) and aortocaval shunt (shunt). Compared with sham mice, 6 hours after surgery, systolic wall stress (afterload) was increased in TAC mice (+40%; P<0.05), diastolic wall stress (preload) was increased in shunt (+277%; P<0.05) and TAC mice (+74%; P<0.05), and mean total wall stress was similarly increased in TAC (69%) and shunt mice (67%) (P=NS, TAC versus shunt; each P<0.05 versus sham). At 1 week, left ventricular weight/tibia length was significantly increased by 22% in TAC and 29% in shunt mice (P=NS, TAC versus shunt). After 24 hours and 1 week, calcium/calmodulin-dependent protein kinase II signaling was increased in TAC. This resulted in altered calcium cycling, including increased L-type calcium current, calcium transients, fractional sarcoplasmic reticulum calcium release, and calcium spark frequency. In shunt mice, Akt phosphorylation was increased. TAC was associated with inflammation, fibrosis, and cardiomyocyte apoptosis. The latter was significantly reduced in calcium/calmodulin-dependent protein kinase IIdelta-knockout TAC mice. A total of 157 mRNAs and 13 microRNAs were differentially regulated in TAC versus shunt mice. After 8 weeks, fractional shortening was lower and mortality was higher in TAC versus shunt mice. CONCLUSIONS: Afterload results in maladaptive fibrotic hypertrophy with calcium/calmodulin-dependent protein kinase II-dependent altered calcium cycling and apoptosis. Preload is associated with Akt activation without fibrosis, little apoptosis, better function, and lower mortality. This indicates that different loads result in distinct phenotype differences that may require specific pharmacological interventions.


Asunto(s)
Insuficiencia Cardíaca/fisiopatología , Hemodinámica/fisiología , Hipertrofia Ventricular Izquierda/fisiopatología , Remodelación Ventricular/fisiología , Animales , Aorta/fisiopatología , Apoptosis/fisiología , Calcio/metabolismo , Proteína Quinasa Tipo 2 Dependiente de Calcio Calmodulina/genética , Proteína Quinasa Tipo 2 Dependiente de Calcio Calmodulina/metabolismo , Modelos Animales de Enfermedad , Femenino , Fibrosis , Estudio de Asociación del Genoma Completo , Insuficiencia Cardíaca/genética , Insuficiencia Cardíaca/mortalidad , Hipertrofia Ventricular Izquierda/genética , Hipertrofia Ventricular Izquierda/mortalidad , Ratones , Ratones Noqueados , MicroARNs/fisiología , Miocardio/patología , Miocitos Cardíacos/patología , Miocitos Cardíacos/fisiología , Transducción de Señal/fisiología
12.
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
13.
Circulation ; 121(18): 1992-2000, 2010 May 11.
Artículo en Inglés | MEDLINE | ID: mdl-20421520

RESUMEN

BACKGROUND: C-kit is a receptor tyrosine kinase family member expressed in hematopoietic stem cells. C-kit is also transiently expressed in cardiomyocyte precursors during development and in a rare cell population in the normal adult heart. In the present study, the cardiomyogenic potential of c-kit(+) cells isolated from normal neonatal, normal adult, and infarcted adult mouse hearts was evaluated. METHODS AND RESULTS: Magnetic activated cell sorting was used to prepare c-kit(+) cells from the hearts of ACT-EGFP/MHC-nLAC double transgenic mice. These animals exhibit widespread enhanced green fluorescent protein (EGFP) expression and cardiomyocyte-restricted nuclear beta-galactosidase activity, thus permitting simultaneous tracking of cell survival and differentiation. A subset of the c-kit(+) cells from double transgenic neonatal hearts acquired a cardiomyogenic phenotype when cocultured with fetal cardiomyocytes (2.4% of all EGFP(+) cells screened) but rarely when cultured alone or when cocultured with mouse fibroblasts (0.03% and 0.05% of the EGFP(+) cells screened, respectively). In contrast, c-kit(+) cells from normal adult double transgenic hearts failed to undergo cardiomyogenic differentiation when cocultured with nontransgenic fetal cardiomyocytes (>18 000 EGFP(+) cells screened) or when transplanted into normal or infarcted adult mouse hearts (14 EGFP(+) grafts examined). A single c-kit(+) cell from an infarcted double transgenic adult heart was observed to acquire a cardiomyogenic phenotype in coculture (>37 000 EGFP(+) cells screened). CONCLUSIONS: These data suggest that the ability of cardiac-resident c-kit(+) cells to acquire a cardiomyogenic phenotype is subject to temporal limitations or, alternatively, that the cardiomyogenic population is lost. Elucidation of the underlying molecular basis may permit robust cardiomyogenic induction in adult-derived cardiac c-kit(+) cells.


Asunto(s)
Células Madre Hematopoyéticas/citología , Infarto del Miocardio/patología , Infarto del Miocardio/terapia , Miocitos Cardíacos/citología , Proteínas Proto-Oncogénicas c-kit/genética , Factores de Edad , Animales , Animales Recién Nacidos , Diferenciación Celular/fisiología , Células Cultivadas , Técnicas de Cocultivo , Fibroblastos/citología , Fibroblastos/fisiología , Citometría de Flujo , Proteínas Fluorescentes Verdes/genética , Células Madre Hematopoyéticas/fisiología , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos DBA , Ratones Transgénicos , Infarto del Miocardio/fisiopatología , Miocitos Cardíacos/fisiología , Fenotipo
14.
J Mol Cell Cardiol ; 43(4): 504-16, 2007 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-17707399

RESUMEN

Human embryonic stem cell (hESC)-derived cardiomyocytes are a promising cell source for cardiac repair. Whether these cells can be transported long distance, survive, and mature in hearts subjected to ischemia/reperfusion with minimal infarction is unknown. Taking advantage of a constitutively GFP-expressing hESC line we investigated whether hESC-derived cardiomyocytes could be shipped and subsequently form grafts when transplanted into the left ventricular wall of athymic nude rats subjected to ischemia/reperfusion with minimal infarction. Co-localization of GFP-epifluorescence and cardiomyocyte-specific marker staining was utilized to analyze hESC-derived cardiomyocyte fate in a rat ischemia/reperfused myocardium. Differentiated, constitutively green fluorescent protein (GFP)-expressing hESCs (hES3-GFP; Envy) containing about 13% cardiomyocytes were differentiated in Singapore, and shipped in culture medium at 4 degrees C to Los Angeles (shipping time approximately 3 days). The cells were dissociated and a cell suspension (2 x 10(6) cells for each rat, n=10) or medium (n=10) was injected directly into the myocardium within the ischemic risk area 5 min after left coronary artery occlusion in athymic nude rats. After 15 min of ischemia, the coronary artery was reperfused. The hearts were harvested at various time points later and processed for histology, immunohistochemical staining, and fluorescence microscopy. In order to assess whether the hESC-derived cardiomyocytes might evade immune surveillance, 2 x 10(6) cells were injected into immune competent Sprague-Dawley rat hearts (n=2), and the hearts were harvested at 4 weeks after cell injection and examined as in the previous procedures. Even following 3 days of shipping, the hESC-derived cardiomyocytes within embryoid bodies (EBs) showed active and rhythmic contraction after incubation in the presence of 5% CO(2) at 37 degrees C. In the nude rats, following cell implantation, H&E, immunohistochemical staining and GFP epifluorescence demonstrated grafts in 9 out of 10 hearts. Cells that demonstrated GFP epifluorescence also stained positive (co-localized) for the muscle marker alpha-actinin and exhibited cross striations (sarcomeres). Furthermore, cells that stained positive for the antibody to GFP (immunohistochemistry) also stained positive for the muscle marker sarcomeric actin and demonstrated cross striations. At 4 weeks engrafted hESCs expressed connexin 43, suggesting the presence of nascent gap junctions between donor and host cells. No evidence of rejection was observed in nude rats as determined by inspection for lymphocytic infiltrate and/or giant cells. In contrast, hESC-derived cardiomyocytes injected into immune competent Sprague-Dawley rats resulted in an overt lymphocytic infiltrate. hESCs-derived cardiomyocytes can survive several days of shipping. Grafted cells survived up to 4 weeks after transplantation in hearts of nude rats subjected to ischemia/reperfusion with minimal infarction. They continued to express cardiac muscle markers and exhibit sarcomeric structure and they were well interspersed with the endogenous myocardium. However, hESC-derived cells did not escape immune surveillance in the xenograft setting in that they elicited a rejection phenomenon in immune competent rats.


Asunto(s)
Diferenciación Celular , Células Madre Embrionarias/citología , Células Madre Embrionarias/trasplante , Isquemia Miocárdica/terapia , Miocardio/citología , Miocitos Cardíacos/citología , Animales , Supervivencia Celular , Células Cultivadas , Conexina 43/metabolismo , Proteínas Fluorescentes Verdes/metabolismo , Humanos , Isquemia Miocárdica/patología , Miocardio/patología , Miocitos Cardíacos/metabolismo , Ratas , Ratas Desnudas , Trasplante de Células Madre , Trasplante Heterólogo
15.
Genomics ; 83(6): 1105-15, 2004 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-15177563

RESUMEN

msk, myocardial SNF1-like kinase, was originally isolated in a screen for kinases expressed during early cardiogenesis in the mouse. msk maps to the proximal end of mouse chromosome 17 in a region that is syntenic with human chromosome 21q22.3, where the gene for SNF1LK, a predicted protein that shares 80% identity at the amino acid level with Msk, is located. Accordingly, msk has been redesignated snf1lk. Interestingly, the region encompassing the SNF1LK locus has been implicated in congenital heart defects often observed in patients with Down syndrome. snf1lk is also expressed in skeletal muscle progenitor cells of the somite beginning at 9.5 dpc. These data suggest a more general role for snf1lk in the earliest stages of muscle growth and/or differentiation. Consistent with a role in cell cycling, we observe that Chinese hamster ovary cells that express a tetracycline-inducible SNF1LK kinase domain do not divide, but undergo additional rounds of replication to yield 8N and 16N cells. These data suggest a possible function for SNF1LK in G2/M regulation. We show data that indicate that SNF1LK does not share functional homology with other SNF1-related kinases, but represents a new subclass with novel molecular activities.


Asunto(s)
Ciclo Celular , Proteínas Serina-Treonina Quinasas/genética , Proteínas Serina-Treonina Quinasas/fisiología , Secuencia de Aminoácidos , Animales , Células CHO , Ciclo Celular/genética , Mapeo Cromosómico , Cricetinae , Cricetulus , Embrión de Mamíferos/metabolismo , Embrión de Mamíferos/ultraestructura , Fase G2/genética , Corazón/embriología , Humanos , Inmunoprecipitación , Ratones , Microtúbulos/metabolismo , Microtúbulos/ultraestructura , Datos de Secuencia Molecular , Desarrollo de Músculos/genética , Protamina Quinasa/genética , Proteínas Serina-Treonina Quinasas/metabolismo , Homología de Secuencia de Aminoácido
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...