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

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Hum Mol Genet ; 22(7): 1395-403, 2013 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-23300193

RESUMEN

In this paper, we report a novel heterozygous mutation of A285V codon conversion on exon 4 of the desmin (DES), using whole exome sequencing (WES) in an isolated proband with documented dilated cardiomyopathy (DCM). This mutation is predicted to cause three-dimensional structure changes of DES. Immunohistological and electron microscopy studies demonstrated diffuse abnormal DES aggregations in DCM-induced-pluripotent stem cell (iPSC)-derived cardiomyocytes, and control-iPSC-derived cardiomyocytes transduced with A285V-DES. DCM-iPSC-derived cardiomyocytes also exhibited functional abnormalities in vitro. This is the first demonstration that patient-specific iPSC-derived cardiomyocytes can be used to provide histological and functional confirmation of a suspected genetic basis for DCM identified by WES.


Asunto(s)
Cardiomiopatía Dilatada/genética , Desmina/genética , Células Madre Pluripotentes Inducidas/fisiología , Miocitos Cardíacos/metabolismo , Adulto , Secuencia de Aminoácidos , Secuencia de Bases , Cardiomiopatía Dilatada/diagnóstico por imagen , Cardiomiopatía Dilatada/fisiopatología , Diferenciación Celular , Desmina/química , Desmina/metabolismo , Exoma , Exones , Células HEK293 , Secuenciación de Nucleótidos de Alto Rendimiento , Humanos , Masculino , Datos de Secuencia Molecular , Mutación Missense , Linaje , Fenotipo , Análisis de Secuencia de ADN , Volumen Sistólico/genética , Ultrasonografía , Disfunción Ventricular Izquierda/diagnóstico por imagen , Disfunción Ventricular Izquierda/genética , Disfunción Ventricular Izquierda/fisiopatología
2.
Hum Mol Genet ; 21(1): 32-45, 2012 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-21949351

RESUMEN

Many human diseases share a developmental origin that manifests during childhood or maturity. Aneuploid syndromes are caused by supernumerary or reduced number of chromosomes and represent an extreme example of developmental disease, as they have devastating consequences before and after birth. Investigating how alterations in gene dosage drive these conditions is relevant because it might help treat some clinical aspects. It may also provide explanations as to how quantitative differences in gene expression determine phenotypic diversity and disease susceptibility among natural populations. Here, we aimed to produce induced pluripotent stem cell (iPSC) lines that can be used to improve our understanding of aneuploid syndromes. We have generated iPSCs from monosomy X [Turner syndrome (TS)], trisomy 8 (Warkany syndrome 2), trisomy 13 (Patau syndrome) and partial trisomy 11;22 (Emanuel syndrome), using either skin fibroblasts from affected individuals or amniocytes from antenatal diagnostic tests. These cell lines stably maintain the karyotype of the donors and behave like embryonic stem cells in all tested assays. TS iPSCs were used for further studies including global gene expression analysis and tissue-specific directed differentiation. Multiple clones displayed lower levels of the pseudoautosomal genes ASMTL and PPP2R3B than the controls. Moreover, they could be transformed into neural-like, hepatocyte-like and heart-like cells, but displayed insufficient up-regulation of the pseudoautosomal placental gene CSF2RA during embryoid body formation. These data support that abnormal organogenesis and early lethality in TS are not caused by a tissue-specific differentiation blockade, but rather involves other abnormalities including impaired placentation.


Asunto(s)
Aneuploidia , Trastornos de los Cromosomas/genética , Células Madre Pluripotentes Inducidas/citología , Diferenciación Celular , Células Cultivadas , Trastornos de los Cromosomas/metabolismo , Trastornos de los Cromosomas/fisiopatología , Femenino , Expresión Génica , Humanos , Células Madre Pluripotentes Inducidas/metabolismo , Lactante , Masculino , Modelos Genéticos
3.
Am J Physiol Cell Physiol ; 303(2): C115-25, 2012 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-22357737

RESUMEN

The successful generation of a high yield of mesenchymal stem cells (MSCs) from human induced pluripotent stem cells (iPSCs) may represent an unlimited cell source with superior therapeutic benefits for tissue regeneration to bone marrow (BM)-derived MSCs. We investigated whether the differential expression of ion channels in iPSC-MSCs was responsible for their higher proliferation capacity than BM-MSCs. The expression of ion channels for K(+), Na(+), Ca(2+), and Cl(-) was examined by RT-PCR. The electrophysiological properties of iPSC-MSCs and BM-MSCs were then compared by patch-clamp experiments to verify their functional roles. Significant mRNA expression of ion channel genes including KCa1.1, KCa3.1, KCNH1, Kir2.1, SCN9A, CACNA1C, and Clcn3 was observed in both human iPSC-MSCs and BM-MSCs, whereas Kir2.2 and Kir2.3 were only detected in human iPSC-MSCs. Five types of currents [big-conductance Ca(2+)-activated K(+) current (BK(Ca)), delayed rectifier K(+) current (IK(DR)), inwardly rectifying K(+) current (I(Kir)), Ca(2+)-activated K(+) current (IK(Ca)), and chloride current (I(Cl))] were found in iPSC-MSCs (83%, 47%, 11%, 5%, and 4%, respectively) but only four of them (BK(Ca), IK(DR), I(Kir), and IK(Ca)) were identified in BM-MSCs (76%, 25%, 22%, and 11%, respectively). Cell proliferation was examined with MTT or bromodeoxyuridine assay, and doubling times were 2.66 and 3.72 days for iPSC-MSCs and BM-MSCs, respectively, showing a 1.4-fold discrepancy. Blockade of IK(DR) with short hairpin RNA or human ether-à-go-go 1 (hEAG1) channel blockers, 4-AP and astemizole, significantly reduced the rate of proliferation of human iPSC-MSCs. These treatments also decreased the rate of proliferation of human BM-MSCs albeit to a lesser extent. These findings demonstrate that the hEAG1 channel plays a crucial role in controlling the proliferation rate of human iPSC-MSCs and to a lesser extent in BM-MSCs.


Asunto(s)
Proliferación Celular , Canales de Potasio Éter-A-Go-Go/fisiología , Células Madre Pluripotentes Inducidas/fisiología , Células Madre Mesenquimatosas/fisiología , Células Cultivadas , Colorantes , Humanos , Potenciales de la Membrana , Sales de Tetrazolio , Tiazoles
4.
Pharmacol Res ; 65(2): 182-8, 2012 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-22005391

RESUMEN

Although vascular effects of selective estrogen receptor modulators (SERMs) have been extensively examined in conduit arteries, whether SERMs could favorably modulate myogenic response in resistance arteries is unknown. The impact of raloxifene therapy and cholesterol diet on myogenic constriction during estrogen deficiency is unresolved. This study investigated changes in vascular reactivity and myogenic responses in female ovariectomized (Ovx) hamsters fed high-cholesterol diet (HCD) with and without chronic treatment of raloxifene. Functional studies were performed on hamster septal coronary arteries cannulated in a pressure myograph. Acetylcholine (ACh)-induced dilatation was reduced in arteries from cholesterol-fed Ovx hamsters, but not in those from cholesterol-fed hamsters, while pressure-induced myogenic constriction was unaffected. Chronic treatment with raloxifene restored ACh-induced dilatation in cholesterol-fed Ovx hamsters. U46619-induced constriction was increased in arteries from cholesterol-fed Ovx hamsters but not from cholesterol-fed control hamsters, which was normalized by chronic raloxifene treatment. The pressure-diameter relationship is presented as normalized diameter versus intraluminal pressure, while the effect of ACh or U46619 is expressed as percentage of tone at 80 mm Hg. Two-way analysis of variance (ANOVA) followed by Bonferroni post-tests were used for statistical evaluation among different treatment groups. P<0.05 was taken as statistically significant. The present results show that chronic treatment with raloxifene could benefit myogenically active coronary arteries by (i) restoring ACh-induced dilatation and (ii) reducing U46619-induced constriction without affecting pressure-induced myogenic responses in cholesterol-fed hamsters during estrogen deficiency. If such benefit can be observed in humans, raloxifene and other SERMs may be useful to preserve endothelial function and curtail vascular hypersensitivity in resistance coronary arteries in post-menopausal women with hypercholesterolemia or hyperlipidemia, a lipid condition implicated in the pathogenesis of myocardial infarction.


Asunto(s)
Colesterol en la Dieta/administración & dosificación , Vasos Coronarios/efectos de los fármacos , Endotelio Vascular/efectos de los fármacos , Clorhidrato de Raloxifeno/farmacología , Acetilcolina/farmacología , Animales , Colesterol en la Dieta/sangre , Colesterol en la Dieta/toxicidad , LDL-Colesterol/sangre , Cricetinae , Endotelio Vascular/fisiología , Femenino , Lipasa/sangre , Mesocricetus , Desarrollo de Músculos/efectos de los fármacos , Desarrollo de Músculos/fisiología , NG-Nitroarginina Metil Éster/farmacología , Ovariectomía/métodos , Vasoconstricción/efectos de los fármacos , Vasoconstricción/fisiología , Vasodilatación/efectos de los fármacos , Vasodilatación/fisiología
5.
J Am Soc Nephrol ; 22(7): 1221-8, 2011 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-21636641

RESUMEN

Forced expression of selected transcription factors can transform somatic cells into embryonic stem cell (ESC)-like cells, termed induced pluripotent stem cells (iPSCs). There is no consensus regarding the preferred tissue from which to harvest donor cells for reprogramming into iPSCs, and some donor cell types may be more prone than others to accumulation of epigenetic imprints and somatic cell mutations. Here, we present a simple, reproducible, noninvasive method for generating human iPSCs from renal tubular cells present in urine. This procedure eliminates many problems associated with other protocols, and the resulting iPSCs display an excellent ability to differentiate. These data suggest that urine may be a preferred source for generating iPSCs.


Asunto(s)
Células Madre Pluripotentes Inducidas , Túbulos Renales/citología , Orina/citología , Anciano , Femenino , Técnicas de Transferencia de Gen , Humanos , Masculino , Adulto Joven
6.
Arterioscler Thromb Vasc Biol ; 30(5): 992-9, 2010 May.
Artículo en Inglés | MEDLINE | ID: mdl-20185791

RESUMEN

OBJECTIVE: Selective estrogen receptor modulators (SERMs) inhibit constriction of mammalian conduit arteries. However, it is unknown whether SERMs at therapeutically achievable concentrations could reduce vascular tone in resistance arteries. The present study aimed to examine roles of Ca(2+) influx in endothelium and endothelial nitric oxide synthase (eNOS) activation in dilatations induced by raloxifene, a second-generation SERM in myogenically active arteries. METHODS AND RESULTS: Small mesenteric arteries from Sprague-Dawley rats were isolated and mounted in a pressure myograph for measurement of changes in vessel diameter. [Ca(2+)](i) images on native endothelial cells of intact arteries were determined by the fluorescence imaging technique, and phosphorylation of eNOS was assayed by Western blotting. Raloxifene (0.3 to 10 nmol/L) produced dilatations on established steady myogenic constriction. Female rat arteries dilated significantly more in response to raloxifene than male arteries. Raloxifene-induced dilatations of female arteries were blunted by N(G)-nitro-l-arginine methyl ester but unaffected by 1400W, charybdotoxin plus apamin, wortmannin, or LY294002. Raloxifene (3 nmol/L) triggered rises in endothelial cell [Ca(2+)](i) and increased eNOS phosphorylation at Ser1177. Both effects were greater in arteries from female rats than in arteries from male rats. Increases in endothelial cell [Ca(2+)](i) and in eNOS phosphorylation were prevented by removal of extracellular Ca(2+) ions. Finally, ICI 182,780 did not affect the raloxifene-stimulated rise in endothelial cell [Ca(2+)](i), eNOS phosphorylation, and vasodilatations. Chronic raloxifene treatment reduced myogenic constriction in arteries from female but not male rats. CONCLUSION: Raloxifene at therapeutically relevant concentrations inhibits myogenic constriction by an NO-dependent mechanism that causally involves the elevated [Ca(2+)](i) in endothelial cells and subsequent eNOS activation. Raloxifene dilates resistance arteries more effectively in female rats, indicating its significant gender-related action on endothelial cells in microcirculation.


Asunto(s)
Señalización del Calcio/efectos de los fármacos , Arterias Mesentéricas/efectos de los fármacos , Óxido Nítrico Sintasa de Tipo III/metabolismo , Clorhidrato de Raloxifeno/farmacología , Moduladores Selectivos de los Receptores de Estrógeno/farmacología , Vasodilatación/efectos de los fármacos , Vasodilatadores/farmacología , Androstadienos/farmacología , Animales , Apamina/farmacología , Presión Sanguínea/efectos de los fármacos , Caribdotoxina/farmacología , Cromonas/farmacología , Relación Dosis-Respuesta a Droga , Activación Enzimática , Inhibidores Enzimáticos/farmacología , Estradiol/análogos & derivados , Estradiol/farmacología , Antagonistas de Estrógenos/farmacología , Femenino , Fulvestrant , Técnicas In Vitro , Masculino , Arterias Mesentéricas/enzimología , Morfolinas/farmacología , NG-Nitroarginina Metil Éster/farmacología , Óxido Nítrico/metabolismo , Óxido Nítrico Sintasa de Tipo III/antagonistas & inhibidores , Ovariectomía , Fosforilación , Bloqueadores de los Canales de Potasio/farmacología , Inhibidores de Proteínas Quinasas/farmacología , Ratas , Ratas Sprague-Dawley , Factores Sexuales , Factores de Tiempo , Resistencia Vascular/efectos de los fármacos , Wortmanina
7.
J Mol Cell Cardiol ; 48(6): 1129-37, 2010 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-20116384

RESUMEN

Hypoxia plays an important role in the proliferation, differentiation and maintenance of the cardiovascular system during development. While low oxygen tension appears to direct the cultured embryonic stem cells (ESCs) to differentiate into cardiomyocytes, the underlying molecular mechanism remains unclear. At a molecular level, hypoxia inducible factor-1 (HIF-1) plays an important role in handling the hypoxia signal. In the present study, we demonstrated that expression of exogenous HIF-1 alpha cDNA into murine ESCs significantly promoted cardiogenesis as indicated by a higher percentage of beating embryoid body and troponin-T positive cell counts as well as increased expression of early and late cardiac markers, such as GATA-binding protein 4 and 6, NK2 transcription factor related locus 5, alpha-myosin heavy chain, beta-myosin heavy chain and myosin light chain 2 ventricular transcripts. In addition, the transduced cells exhibited increased mRNA levels of cardiotrophin-1 and vascular endothelial growth factor, along with phosphorylation of eNOS [p-eNOS (ser1171)]. Application of NOS inhibitors, diphenyleneiodonium chloride (DPI), N(omega)-Nitro-L-arginine methyl ester hydrochloride (L-NAME) or N(omega)-Nitro-L-arginine (L-NNA) abolished the HIF-1 alpha stimulated cardiac differentiation. With the clues of upregulated mRNA expression of calcium handling proteins, ryanodine receptor 2, sodium calcium exchanger and sarcoplasmic/endoplasmic reticulum calcium ATPase, in the transduced HIF-1 alpha ESCs, further study indicated that the maximum upstroke and decay velocity was significantly increased in both non-caffeine and caffeine-induced calcium transient in ESCs-derived cardiomyocytes. This suggests a well developed function of the sarcoplasmic reticulum in ESC-derived cardiomyocytes. Electrophysiological study also indicated that a portion of the HIF-1 alpha-transduced cells exhibited prominent phase-4 depolarization. These findings suggest that keen activation of the HIF-1 pathway enhances differentiation and maturation of cardiomyocytes derived from ESCs.


Asunto(s)
Células Madre Embrionarias/citología , Subunidad alfa del Factor 1 Inducible por Hipoxia/biosíntesis , Miocardio/metabolismo , Animales , Diferenciación Celular , Citocinas/biosíntesis , Corazón/fisiología , Hipoxia/metabolismo , Ratones , Modelos Biológicos , ARN Mensajero/metabolismo , Transducción de Señal , Troponina T/biosíntesis , Regulación hacia Arriba , Factor A de Crecimiento Endotelial Vascular/metabolismo
8.
Europace ; 12(8): 1178-87, 2010 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-20472688

RESUMEN

AIMS: A better understanding of the ionic mechanisms for cardiac automaticity can lead to better strategies for engineering bio-artificial pacemakers. Here, we attempted to better define the relative contribution of I(f) and I(K1) in the generation of spontaneous action potentials (SAPs) in cardiomyocytes (CMs). METHODS AND RESULTS: Monolayers of neonatal rat ventricular myocytes (NRVMs) were transduced with a recombinant adenovirus (Ad) to express a gating-engineered HCN1 construct (HCN1-DeltaDeltaDelta) for patch-clamp and multielectrode array (MEA) recordings. Single NRVMs exhibited a bi-phasic response in the generation of SAPs (62.6 +/- 17.4 b.p.m., Days 1-2; 194.3 +/- 12.3 b.p.m., Days 3-4; 73% quiescent, Days 9-10). Although automaticity time-dependently decreased and subsequently ceased, I(f) remained fairly stable (-5.2 +/- 1.1 pA/pF, Days 1-2; -5.1 +/- 1.4 pA/pF, Days 7-8; -4.3 +/- 1.3 pA/pF, Days 13-14). In contrast, I(K1) declined rapidly (from -16.9 +/- 2.7 pA/pF on Days 1-2 to -4.4 +/- 1.6 pA/pF on Days 5-6). Maximum diastolic potential/resting membrane potential (r = 0.89) and action potential duration at 50% (APD(50), r = 0.73) and 90% (APD(90), r = 0.75) but not the firing rate (r = -0.3) were positively correlated to the I(K1). Similarly, monolayer NRVMs ceased to spontaneously fire after long-term culture. Ad-HCN1-DeltaDeltaDelta transduction restored pacing in silenced individual and monolayer NRVMs but with reduced conduction velocity and field potential amplitude. CONCLUSION: We conclude that the combination of I(K1) and I(f) primes CMs for bio-artificial pacing by determining the threshold. However, I(f) functions as a membrane potential oscillator to determine the basal firing frequency. Future engineering of automaticity in the multicellular setting needs to have conduction taken into consideration.


Asunto(s)
Potenciales de Acción/fisiología , Relojes Biológicos/fisiología , Ventrículos Cardíacos/citología , Miocitos Cardíacos/fisiología , Marcapaso Artificial , Animales , Animales Recién Nacidos , Técnicas de Cultivo de Célula/métodos , Canales Catiónicos Regulados por Nucleótidos Cíclicos/genética , Canales Regulados por Nucleótidos Cíclicos Activados por Hiperpolarización , Contracción Miocárdica/fisiología , Miocitos Cardíacos/citología , Técnicas de Placa-Clamp , Canales de Potasio/genética , Ratas , Ratas Wistar , Transducción Genética
9.
Pflugers Arch ; 459(1): 25-38, 2009 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-19756722

RESUMEN

If (or Ih), encoded by the hyperpolarization-activated, cyclic nucleotide-gated (HCN1-4) channel gene family, contributes significantly to cardiac pacing. Bradycardic agents such as ZD7288 that target HCN channels have been developed, but the molecular configuration of their receptor is poorly defined. Here, we probed the drug receptor by systematically introducing alanine scanning substitutions into the selectivity filter (C347A, I348A, G349A, Y350A, G351A in the P-loop), outer (P355A, V356A, S357A, M358A in the P-S6 linker), and inner (M377A, F378A, V379A in S6) pore vestibules of HCN1 channels. When heterologously expressed in human embryonic kidney 293 cells for patch-clamp recordings, I348A, G349A, Y350A, G351A, P355A, and V356A did not produce measurable currents. The half-blocking concentration (IC50) of wild type (WT) for ZD7288 was 25.8+/-9.7microM. While the IC50 of M358A was identical to WT, those of C347A, S357A, F378A, and V379A markedly increased to 137.6+/-56.4, 113.3+/-34.1, 587.1+/-167.5, and 1726.3+/-673.4microM, respectively (p<0.05). Despite the proximity of the S6 residues studied, M377A was hypersensitive (IC50=5.1+/-0.7microM; p<0.05) implicating site specificity. To explore the energetic interactions among the S6 residues, double and triple substitutions (M377A/F378A, M377A/V379A, F378A/V379A, and M377A/F378A/V379A) were generated for thermodynamic cycle analysis. Specific interactions with coupling energies (deltadeltaG)>1kT for M377-F378 and F378-V379 but not M377-V379 were identified. Based on these new data and others, we proposed a refined drug-blocking model that may lead to improved antiarrhythmics and bioartificial pacemaker designs.


Asunto(s)
Cardiotónicos/farmacología , Canales Catiónicos Regulados por Nucleótidos Cíclicos/química , Canales de Potasio/química , Estructura Cuaternaria de Proteína , Pirimidinas/farmacología , Secuencia de Aminoácidos , Animales , Bradicardia/tratamiento farmacológico , Canales Catiónicos Regulados por Nucleótidos Cíclicos/metabolismo , Humanos , Canales Regulados por Nucleótidos Cíclicos Activados por Hiperpolarización , Ratones , Microscopía Fluorescente , Datos de Secuencia Molecular , Técnicas de Placa-Clamp , Canales de Potasio/metabolismo , Estructura Cuaternaria de Proteína/efectos de los fármacos
10.
Biochem Biophys Res Commun ; 379(4): 898-903, 2009 Feb 20.
Artículo en Inglés | MEDLINE | ID: mdl-19135029

RESUMEN

The therapeutic potential of transplantation of embryonic stem cells (ESCs) in animal model of myocardial infarction has been consistently demonstrated. The development of superparamagnetic iron oxide (SPIO) nanoparticles labeling and cardiac magnetic resonance imaging (MRI) have been increasingly used to track the migration of transplanted cells in vivo allowing cell fate determination. However, the impact of SPIO- labeling on cell phenotype and cardiac differentiation capacity of ESCs remains unclear. In this study, we demonstrated that ESCs labeled with SPIO compared to their unlabeled counterparts had similar cardiogenic capacity, and SPIO-labeling did not affect calcium-handling property of ESC-derived cardiomyocytes. Moreover, transplantation of SPIO-labeled ESCs via direct intra-myocardial injection to infarct myocardium resulted in significant improvement in heart function. These findings demonstrated the feasibility of in vivo ESC tracking using SPIO-labeling and cardiac MRI without affecting the cardiac differentiation potential and functional properties of ESCs.


Asunto(s)
Diferenciación Celular/efectos de los fármacos , Células Madre Embrionarias/efectos de los fármacos , Compuestos Férricos/efectos adversos , Miocitos Cardíacos/efectos de los fármacos , Nanopartículas/efectos adversos , Coloración y Etiquetado/métodos , Animales , Línea Celular , Movimiento Celular/efectos de los fármacos , Células Madre Embrionarias/citología , Células Madre Embrionarias/trasplante , Proteínas Fluorescentes Verdes/análisis , Proteínas Fluorescentes Verdes/biosíntesis , Imagen por Resonancia Magnética/métodos , Ratones , Miocitos Cardíacos/citología
11.
J Cardiovasc Electrophysiol ; 20(9): 1048-54, 2009 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-19460073

RESUMEN

INTRODUCTION: Normal heart rhythms originate in the sinoatrial node. HCN-encoded funny current (I(f)) and the Kir2-encoded inward rectifier (I(K1)) counteract each other by respectively oscillating and stabilizing the negative resting membrane potential, and controlling action potential firing. Therefore, I(K1) suppression and I(f) overexpression have been independently exploited to convert cardiomyocytes (CMs) into AP-firing bioartificial pacemakers. Although the 2 strategies have been largely assumed synergistic, their complementarity has not been investigated. METHODS AND RESULTS: We explored the interrelationships of automaticity, I(f) and I(K1) by transducing single left ventricular (LV) CMs isolated from guinea pig hearts with the recombinant adenoviruses Ad-CMV-GFP-IRES-HCN1-AAA and/or Ad-CGI-Kir2.1 to mediate their current densities via a whole-cell patch clamp technique at 37 degrees C. Results showed that Ad-CGI-HCN1-AAA but not Ad-CGI-Kir2.1 transduction induced automaticity (181.1 +/- 13.1 bpm). Interestingly, Ad-CGI-HCN1-AAA/Ad-CGI-Kir2.1 cotransduction significantly promoted the induced firing frequency (320.0 +/- 15.8 bpm; P < 0.05). Correlation analysis revealed that the firing frequency, phase-4 slope and APD(90) of AP-firing LV CMs were correlated with I(f) (R(2) > 0.7) only when -2 >I(K1) >-4 pA/pF but not with I(K1) over the entire I(f) ranges examined (0.02 < R(2) < 0.4). Unlike I(f), I(K1) displayed correlation with neither the phase-4 slope (R(2)= 0.02) nor phase-4 length (R(2)= 0.04) when -2 > I(f) > -4 pA/pF. As anticipated, however, APD(90) was correlated with I(K1) (R(2)= 0.4). CONCLUSION: We conclude that an optimal level of I(K1) maintains a voltage range for I(f) to operate most effectively during a dynamic cardiac cycle.


Asunto(s)
Potenciales de Acción/fisiología , Canales Catiónicos Regulados por Nucleótidos Cíclicos/genética , Ingeniería Genética/métodos , Sistema de Conducción Cardíaco/fisiología , Activación del Canal Iónico/fisiología , Potenciales de la Membrana/fisiología , Canales de Potasio/genética , Potasio/metabolismo , Animales , Femenino , Cobayas , Canales Regulados por Nucleótidos Cíclicos Activados por Hiperpolarización
12.
Biochem Biophys Res Commun ; 372(4): 553-8, 2008 Aug 08.
Artículo en Inglés | MEDLINE | ID: mdl-18503758

RESUMEN

Although both the H1 and HES2 human embryonic stem cell lines (NIH codes: WA01 and ES02, respectively) are capable of forming all three germ layers and their derivatives, various lines of evidence including the need to use different protocols to induce cardiac differentiation hint that they have distinct preferences to become chamber-specific heart cells. However, a direct systematic comparison has not been reported. Here we electrophysiologically demonstrated that the distributions of ventricular-, atrial- and pacemaker-like derivatives were indeed different (ratios=39:61:0 and 64:33:3 for H1 and HES2, respectively). Based on these results, we hypothesized the differences in their cardiogenic potentials are imprinted in the proteomes of undifferentiated H1 and HES2. Using multiplexing, high-resolution 2-D Differential In Gel Electrophoresis (DIGE) to minimize gel-to-gel variations that are common in conventional 2-D gels, a total of 2000 individual protein spots were separated. Of which, 55 were >2-fold differentially expressed in H1 and HES2 (p<0.05) and identified by mass spectrometery. Bioinformatic analysis of these protein differences further revealed candidate pathways that contribute to the H1 and HES2 phenotypes. We conclude that H1 and HES2 have predetermined preferences to become ventricular, atrial, and pacemaker cells due to discrete differences in their proteomes. These results improve our basic understanding of hESCs and may lead to mechanism-based methods for their directed cardiac differentiation into chamber-specific cardiomyocytes.


Asunto(s)
Células Madre Embrionarias/citología , Miocitos Cardíacos/citología , Células Madre Pluripotentes/citología , Proteoma , Potenciales de Acción , Línea Celular , Electroforesis en Gel Bidimensional , Células Madre Embrionarias/metabolismo , Impresión Genómica , Corazón/embriología , Humanos , Miocitos Cardíacos/metabolismo , Miocitos Cardíacos/fisiología , Organogénesis/genética , Células Madre Pluripotentes/metabolismo , Biosíntesis de Proteínas , Proteoma/genética
13.
Eur J Pharmacol ; 488(1-3): 169-72, 2004 Mar 19.
Artículo en Inglés | MEDLINE | ID: mdl-15044048

RESUMEN

Urocortin relaxes rat pulmonary arteries partly through a cyclic AMP-dependent but Ca(2+) channel-independent mechanism. However, other participating mechanisms are relatively unknown. The present study was designed to examine whether the forward mode of Na(+)-Ca(2+) exchangers play a role in the relaxant responses to urocortin in isolated rat small pulmonary arteries. Endothelium-denuded rings were mounted on small vessel myographs for measurement of changes in isometric tension. Urocortin inhibited 9,11-dideoxy-11alpha,9alpha-epoxy-methanoprostaglandin F(2alpha) (U46619)-induced contraction in a concentration-dependent manner and this inhibition was reversed by astressin, a corticotropin-releasing factor receptor antagonist. Micromolar concentrations of nickel (Ni(2+)) chloride, a putative inhibitor of the Na(+)-Ca(2+) exchanger, reduced the relaxant responses to urocortin. Urocortin-induced relaxation was abolished in a Na(+)-free solution, a condition that eliminates influence of the forward mode of Na(+)-Ca(2+) exchanger. In contrast, the relaxant responses to atrial natriuretic peptide or forskolin were unaffected by Ni(2+) or with removal of extracellular Na(+). The present results provide indirect evidence suggesting that stimulation of Na(+)-Ca(2+) exchangers may contribute to urocortin-induced endothelium-independent pulmonary artery relaxation.


Asunto(s)
Hormona Liberadora de Corticotropina/antagonistas & inhibidores , Hormona Liberadora de Corticotropina/farmacología , Músculo Liso Vascular/efectos de los fármacos , Níquel/farmacología , Arteria Pulmonar/efectos de los fármacos , Ácido 15-Hidroxi-11 alfa,9 alfa-(epoximetano)prosta-5,13-dienoico/farmacología , Animales , Factor Natriurético Atrial/farmacología , Colforsina/farmacología , Técnicas In Vitro , Masculino , Contracción Muscular/efectos de los fármacos , Relajación Muscular/efectos de los fármacos , Potasio/farmacología , Ratas , Ratas Sprague-Dawley , Intercambiador de Sodio-Calcio/antagonistas & inhibidores , Urocortinas , Vasoconstrictores/farmacología
14.
Physiol Rep ; 2(2): e00237, 2014 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-24744906

RESUMEN

Mesenchymal stem cells (MSCs) derived from human-induced pluripotent stem cells (iPSCs) show superior proliferative capacity and therapeutic potential than those derived from bone marrow (BM). Ectopic expression of myocardin further improved the therapeutic potential of BM-MSCs in a mouse model of myocardial infarction. The aim was of this study was to assess whether forced myocardin expression in iPSC-MSCs could further enhance their transdifferentiation to cardiomyocytes and improve their electrophysiological properties for cardiac regeneration. Myocardin was overexpressed in iPSC-MSCs using viral vectors (adenovirus or lentivirus). The expression of smooth muscle cell and cardiomyocyte markers, and ion channel genes was examined by reverse transcription-polymerase chain reaction (RT-PCR), immunofluorescence staining and patch clamp. The conduction velocity of the neonatal rat ventricular cardiomyocytes cocultured with iPSC-MSC monolayer was measured by multielectrode arrays recording plate. Myocardin induced the expression of α-MHC, GATA4, α-actinin, cardiac MHC, MYH11, calponin, and SM α-actin, but not cTnT, ß-MHC, and MLC2v in iPSC-MSCs. Overexpression of myocardin in iPSC-MSC enhanced the expression of SCN9A and CACNA1C, but reduced that of KCa3.1 and Kir2.2 in iPSC-MSCs. Moreover, BKCa, IKir, ICl, Ito and INa.TTX were detected in iPSC-MSC with myocardin overexpression; while only BKCa, IKir, ICl, IKDR, and IKCa were noted in iPSC-MSC transfected with green florescence protein. Furthermore, the conduction velocity of iPSC-MSC was significantly increased after myocardin overexpression. Overexpression of myocardin in iPSC-MSCs resulted in partial transdifferentiation into cardiomyocytes phenotype and improved the electrical conduction during integration with mature cardiomyocytes.

15.
World J Stem Cells ; 5(3): 86-97, 2013 Jul 26.
Artículo en Inglés | MEDLINE | ID: mdl-23904910

RESUMEN

AIM: To development of an improved p38 MAPK inhibitor-based serum-free medium for embryoid body cardiomyocyte differentiation of human pluripotent stem cells. METHODS: Human embryonic stem cells (hESC) differentiated to cardiomyocytes (CM) using a p38 MAPK inhibitor (SB203580) based serum-free medium (SB media). Nutrient supplements known to increase cell viability were added to SB medium. The ability of these supplements to improve cardiomyogenesis was evaluated by measurements of cell viability, total cell count, and the expression of cardiac markers via flow cytometry. An improved medium containing Soy hydrolysate (HySoy) and bovine serum albumin (BSA) (SupSB media) was developed and tested on 2 additional cell lines (H1 and Siu-hiPSC). Characterization of the cardiomyocytes was done by immunohistochemistry, electrophysiology and quantitative real-time reverse transcription-polymerase chain reaction. RESULTS: hESC cell line, HES-3, differentiating in SB medium for 16 d resulted in a cardiomyocyte yield of 0.07 ± 0.03 CM/hESC. A new medium (SupSB media) was developed with the addition of HySoy and BSA to SB medium. This medium resulted in 2.6 fold increase in cardiomyocyte yield (0.21 ± 0.08 CM/hESC). The robustness of SupSB medium was further demonstrated using two additional pluripotent cell lines (H1, hESC and Siu1, hiPSC), showing a 15 and 9 fold increase in cardiomyocyte yield respectively. The age (passage number) of the pluripotent cells did not affect the cardiomyocyte yields. Embryoid body (EB) cardiomyocytes formed in SupSB medium expressed canonical cardiac markers (sarcomeric α-actinin, myosin heavy chain and troponin-T) and demonstrated all three major phenotypes: nodal-, atrial- and ventricular-like. Electrophysiological characteristics (maximum diastolic potentials and action potential durations) of cardiomyocytes derived from SB and SupSB media were similar. CONCLUSION: The nutrient supplementation (HySoy and BSA) leads to increase in cell viability, cell yield and cardiac marker expression during cardiomyocyte differentiation, translating to an overall increase in cardiomyocyte yield.

16.
Heart Rhythm ; 10(2): 273-82, 2013 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-23041574

RESUMEN

BACKGROUND: Cellular replacement strategies using embryonic stem cell-derived cardiomyocytes (ESC-CMs) have been shown to improve left ventricular (LV) ejection fraction and prevent LV remodeling post-myocardial infarction (MI). Nonetheless, the immature electrical phenotypes of ESC-CMs may increase the risk of ventricular tachyarrhythmias (VTs) and sudden death. OBJECTIVE: To investigate whether the forced expression of Kir2.1-encoded inward rectifying K(+) channels that are otherwise absent in ESC-CMs would attenuate their proarrhythmic risk after transplantation post-MI. METHODS: Mouse ESC line stably transduced with a lentivirus (LentV)-based doxycycline (DOX)-inducible system coexpressing the transgenes Kir2.1 and a dsRed (LentV-THM-Kir2.1-GFP/LentV-TR-KRAB-dsRed) was differentiated into ESC-CMs with (DOX(+)) or without (DOX(-)) treatment with DOX. Detailed in vitro and in vivo assessments of LV function and cardiac electrophysiology were measured 4 weeks after transplantation. RESULTS: ESC-CM DOX(+) with atrial and ventricular phenotype exhibited more hyperpolarizing resting membrane potential than did ESC-CM DOX(-) (P< .05). Transplantations of ESC-CM DOX(-) and ESC-CM DOX(+) both significantly improved LV ejection fraction, LV end-systolic diameter, end-systolic pressure-volume relationship, and positive maximal and negative pressure derivative (P< .05) at 4 weeks compared with the MI group; however, the DOX(-) group (22 of 40, 55%) had a significantly higher early sudden death rate than the DOX(+) group (13 of 40, 32.5%; P = .036). Telemetry monitoring revealed that the DOX(-) group (6.09%±3.65%) had significantly more episodes of spontaneous VT compared with the DOX(+) group (0.92%±0.81%; P< .05). In vivo programmed electrical stimulation at 2 weeks resulted in a significantly higher incidence of inducible VT in the DOX(-) group (9 of 16, 56.25%) compared with the DOX(+) group (3 of 16, 18.75%; P = .031). CONCLUSIONS: Forced expression of Kir2.1 in ESC-CMs improves their electrical phenotypes and lowers the risk of inducible and spontaneous VT after post-MI transplantation.


Asunto(s)
Células Madre Embrionarias/trasplante , Trasplante de Corazón/efectos adversos , Infarto del Miocardio/terapia , Canales de Potasio de Rectificación Interna/genética , Taquicardia Ventricular/terapia , Remodelación Ventricular/fisiología , Animales , Células Cultivadas , Modelos Animales de Enfermedad , Doxiciclina/farmacología , Electrocardiografía/métodos , Regulación de la Expresión Génica , Trasplante de Corazón/métodos , Inmunohistoquímica , Masculino , Ratones , Ratones Endogámicos , Infarto del Miocardio/diagnóstico , Miocitos Cardíacos/metabolismo , Fenotipo , Complicaciones Posoperatorias/diagnóstico , Complicaciones Posoperatorias/terapia , Canales de Potasio de Rectificación Interna/metabolismo , Medición de Riesgo , Sensibilidad y Especificidad , Taquicardia Ventricular/etiología
17.
PLoS One ; 8(3): e57876, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23472116

RESUMEN

Functional endothelial-like cells (EC) have been successfully derived from different cell sources and potentially used for treatment of cardiovascular diseases; however, their relative therapeutic efficacy remains unclear. We differentiated functional EC from human bone marrow mononuclear cells (BM-EC), human embryonic stem cells (hESC-EC) and human induced pluripotent stem cells (hiPSC-EC), and compared their in-vitro tube formation, migration and cytokine expression profiles, and in-vivo capacity to attenuate hind-limb ischemia in mice. Successful differentiation of BM-EC was only achieved in 1/6 patient with severe coronary artery disease. Nevertheless, BM-EC, hESC-EC and hiPSC-EC exhibited typical cobblestone morphology, had the ability of uptaking DiI-labeled acetylated low-density-lipoprotein, and binding of Ulex europaeus lectin. In-vitro functional assay demonstrated that hiPSC-EC and hESC-EC had similar capacity for tube formation and migration as human umbilical cord endothelial cells (HUVEC) and BM-EC (P>0.05). While increased expression of major angiogenic factors including epidermal growth factor, hepatocyte growth factor, vascular endothelial growth factor, placental growth factor and stromal derived factor-1 were observed in all EC cultures during hypoxia compared with normoxia (P<0.05), the magnitudes of cytokine up-regulation upon hypoxic were more dramatic in hiPSC-EC and hESC-EC (P<0.05). Compared with medium, transplanting BM-EC (n = 6), HUVEC (n = 6), hESC-EC (n = 8) or hiPSC-EC (n = 8) significantly attenuated severe hind-limb ischemia in mice via enhancement of neovascularization. In conclusion, functional EC can be generated from hECS and hiPSC with similar therapeutic efficacy for attenuation of severe hind-limb ischemia. Differentiation of functional BM-EC was more difficult to achieve in patients with cardiovascular diseases, and hESC-EC or iPSC-EC are readily available as "off-the-shelf" format for the treatment of tissue ischemia.


Asunto(s)
Células Endoteliales/trasplante , Células Madre Pluripotentes Inducidas/citología , Isquemia/patología , Isquemia/terapia , Trasplante de Células Madre , Anciano , Proteínas Angiogénicas/metabolismo , Animales , Células de la Médula Ósea/citología , Diferenciación Celular , Hipoxia de la Célula , Medios de Cultivo Condicionados/farmacología , Citocinas/metabolismo , Células Endoteliales/citología , Miembro Posterior/patología , Humanos , Masculino , Ratones , Persona de Mediana Edad , Neovascularización Fisiológica , Perfusión , Factores de Tiempo
18.
J Cardiovasc Transl Res ; 6(6): 989-99, 2013 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-24081385

RESUMEN

While human embryonic stem cells (hESCs) can differentiate into functional cardiomyocytes, their immature phenotypes limit their therapeutic application for myocardial regeneration. We sought to determine whether electrical stimulation could enhance the differentiation and maturation of hESC-derived cardiomyocytes. Cardiac differentiation was induced in a HES3 hESC line via embryoid bodies formation treated with a p38 MAP kinase inhibitor. Detailed molecular and functional analysis were performed in those hESC-derived cardiomyocytes cultured for 4 days in the absence or presence of electrical field stimulation (6.6 V/cm, 1 Hz, and 2 ms pulses) using an eight-channel C-Pace stimulator (Ion-Optics Co., MA). Upon electrical stimulation, quantitative polymerase chain reaction demonstrated significant upregulation of cardiac-specific gene expression including HCN1, MLC2V, SCN5A, SERCA, Kv4.3, and GATA4; immunostaining and flow cytometry analysis revealed cellular elongation and an increased proportion of troponin-T positive cells (6.3 ± 1.2% vs. 15.8 ± 2.1%; n = 3, P < 0.01). Electrophysiological studies showed an increase in the proportion of ventricular-like hESC-derived cardiomyocytes (48 vs. 29%, P < 0.05) with lengthening of their action potential duration at 90% repolarization (387.7 ± 35.35; n = 11 vs. 291.8 ± 20.82; n = 10, P < 0.05) and 50% repolarization (313.9 ± 27.94; n = 11 vs. 234.0 ± 16.10; n = 10, P < 0.05) after electrical stimulation. Nonetheless, the membrane diastolic potentials and action potential upstrokes of different hESC-derived cardiomyocyte phenotypes, and the overall beating rate remained unchanged (all P > 0.05). Fluorescence confocal imaging revealed that electrical stimulation significantly increased both spontaneous and caffeine-induced calcium flux in the hESC-derived cardiomyocytes (approximately 1.6-fold for both cases; P < 0.01). In conclusion, electrical field stimulation increased the expression of cardiac-specific genes and the yield of differentiation, promoted ventricular-like phenotypes, and improved the calcium handling of hESC-derived cardiomyocytes.


Asunto(s)
Diferenciación Celular , Células Madre Embrionarias/fisiología , Miocitos Cardíacos/fisiología , Señalización del Calcio , Línea Celular , Linaje de la Célula , Estimulación Eléctrica , Células Madre Embrionarias/efectos de los fármacos , Células Madre Embrionarias/enzimología , Perfilación de la Expresión Génica , Regulación de la Expresión Génica , Humanos , Potenciales de la Membrana , Miocitos Cardíacos/efectos de los fármacos , Miocitos Cardíacos/enzimología , Fenotipo , Inhibidores de Proteínas Quinasas/farmacología , Factores de Tiempo , Proteínas Quinasas p38 Activadas por Mitógenos/antagonistas & inhibidores , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
19.
Aging (Albany NY) ; 4(11): 803-822, 2012 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-23362510

RESUMEN

AIMS: We identified an autosomal dominant non­sense mutation (R225X) in exon 4 of the lamin A/C (LMNA) gene in a Chinese family spanning 3 generations with familial dilated cardiomyopathy (DCM). In present study, we aim to generate induced pluripotent stem cells derived cardiomyocytes (iPSC­CMs) from an affected patient with R225X and another patient bearing LMNA frame­shift mutation for drug screening. METHODS AND RESULTS: Higher prevalence of nuclear bleb formation and micronucleation was present in LMNA(R225X/WT) and LMNA(Framshift/WT) iPSC­CMs. Under field electrical stimulation, percentage of LMNA­mutated iPSC­CMs exhibiting nuclear senescence and cellular apoptosis markedly increased. shRNA knockdown of LMNA replicated those phenotypes of the mutated LMNA field electrical stress. Pharmacological blockade of ERK1/2 pathway with MEK1/2 inhibitors, U0126 and selumetinib (AZD6244) significantly attenuated the pro­apoptotic effects of field electric stimulation on the mutated LMNA iPSC­CMs. CONCLUSION: LMNA­related DCM was modeled in­vitro using patient­specific iPSC­CMs. Our results demonstrated that haploinsufficiency due to R225X LMNA non­sense mutation was associated with accelerated nuclear senescence and apoptosis of iPSC­ CMs under electrical stimulation, which can be significantly attenuated by therapeutic blockade of stress­related ERK1/2 pathway.


Asunto(s)
Envejecimiento/fisiología , Cardiomiopatía Dilatada , Células Madre Pluripotentes Inducidas/citología , Lamina Tipo A/genética , Modelos Biológicos , Miocitos Cardíacos/citología , Western Blotting , Cardiomiopatía Dilatada/genética , Cardiomiopatía Dilatada/metabolismo , Cardiomiopatía Dilatada/patología , Diferenciación Celular/fisiología , Femenino , Fibroblastos/citología , Técnica del Anticuerpo Fluorescente , Humanos , Células Madre Pluripotentes Inducidas/metabolismo , Masculino , Persona de Mediana Edad , Mutación , Miocitos Cardíacos/metabolismo , Técnicas de Placa-Clamp , Linaje
20.
PLoS One ; 6(5): e19787, 2011 May 11.
Artículo en Inglés | MEDLINE | ID: mdl-21589943

RESUMEN

The cardioprotective effects of high-density lipoprotein cholesterol (HDL-C) and apolipoprotein A1 (apoA-I) are well documented, but their effects in the direction of the cardiac differentiation of embryonic stem cells are unknown. We evaluated the effects of exogenous apoA-I expression on cardiac differentiation of ESCs and maturation of ESC-derived cardiomyocytes. We stably over-expressed full-length human apoA-I cDNA with lentivirus (LV)-mediated gene transfer in undifferentiated mouse ESCs and human induced pluripotent stem cells. Upon cardiac differentiation, we observed a significantly higher percentage of beating embryoid bodies, an increased number of cardiomyocytes as determined by flow cytometry, and expression of cardiac markers including α-myosin heavy chain, ß-myosin heavy chain and myosin light chain 2 ventricular transcripts in LV-apoA-I transduced ESCs compared with control (LV-GFP). In the presence of noggin, a BMP4 antagonist, activation of BMP4-SMAD signaling cascade in apoA-I transduced ESCs completely abolished the apoA-I stimulated cardiac differentiation. Furthermore, co-application of recombinant apoA-I and BMP4 synergistically increased the percentage of beating EBs derived from untransduced D3 ESCs. These together suggests that that pro-cardiogenic apoA-I is mediated via the BMP4-SMAD signaling pathway. Functionally, cardiomyocytes derived from the apoA-I-transduced cells exhibited improved calcium handling properties in both non-caffeine and caffeine-induced calcium transient, suggesting that apoA-I plays a role in enhancing cardiac maturation. This increased cardiac differentiation and maturation has also been observed in human iPSCs, providing further evidence of the beneficial effects of apoA-I in promoting cardiac differentiation. In Conclusion, we present novel experimental evidence that apoA-I enhances cardiac differentiation of ESCs and iPSCs and promotes maturation of the calcium handling property of ESC-derived cardiomyocytes via the BMP4/SMAD signaling pathway.


Asunto(s)
Apolipoproteína A-I/metabolismo , Diferenciación Celular , Miocardio/citología , Células Madre Pluripotentes/citología , Animales , Proteína Morfogenética Ósea 4/metabolismo , Calcio/metabolismo , Citometría de Flujo , Homeostasis , Humanos , Ratones , Miocardio/metabolismo , Transducción de Señal , Proteínas Smad/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA