Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 21
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Nat Commun ; 14(1): 4716, 2023 08 05.
Artículo en Inglés | MEDLINE | ID: mdl-37543677

RESUMEN

The inability of adult human cardiomyocytes to proliferate is an obstacle to efficient cardiac regeneration after injury. Understanding the mechanisms that drive postnatal cardiomyocytes to switch to a non-regenerative state is therefore of great significance. Here we show that Arid1a, a subunit of the switching defective/sucrose non-fermenting (SWI/SNF) chromatin remodeling complex, suppresses postnatal cardiomyocyte proliferation while enhancing maturation. Genome-wide transcriptome and epigenome analyses revealed that Arid1a is required for the activation of a cardiomyocyte maturation gene program by promoting DNA access to transcription factors that drive cardiomyocyte maturation. Furthermore, we show that ARID1A directly binds and inhibits the proliferation-promoting transcriptional coactivators YAP and TAZ, indicating ARID1A sequesters YAP/TAZ from their DNA-binding partner TEAD. In ischemic heart disease, Arid1a expression is enhanced in cardiomyocytes of the border zone region. Inactivation of Arid1a after ischemic injury enhanced proliferation of border zone cardiomyocytes. Our study illuminates the pivotal role of Arid1a in cardiomyocyte maturation, and uncovers Arid1a as a crucial suppressor of cardiomyocyte proliferation.


Asunto(s)
Miocitos Cardíacos , Transducción de Señal , Humanos , Miocitos Cardíacos/metabolismo , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , ADN/metabolismo , Proliferación Celular , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo
2.
Stem Cell Reports ; 18(3): 749-764, 2023 03 14.
Artículo en Inglés | MEDLINE | ID: mdl-36868229

RESUMEN

Arrhythmogenic cardiomyopathy (ACM) is an inherited progressive disease characterized by electrophysiological and structural remodeling of the ventricles. However, the disease-causing molecular pathways, as a consequence of desmosomal mutations, are poorly understood. Here, we identified a novel missense mutation within desmoplakin in a patient clinically diagnosed with ACM. Using CRISPR-Cas9, we corrected this mutation in patient-derived human induced pluripotent stem cells (hiPSCs) and generated an independent knockin hiPSC line carrying the same mutation. Mutant cardiomyocytes displayed a decline in connexin 43, NaV1.5, and desmosomal proteins, which was accompanied by a prolonged action potential duration. Interestingly, paired-like homeodomain 2 (PITX2), a transcription factor that acts a repressor of connexin 43, NaV1.5, and desmoplakin, was induced in mutant cardiomyocytes. We validated these results in control cardiomyocytes in which PITX2 was either depleted or overexpressed. Importantly, knockdown of PITX2 in patient-derived cardiomyocytes is sufficient to restore the levels of desmoplakin, connexin 43, and NaV1.5.


Asunto(s)
Cardiomiopatías , Células Madre Pluripotentes Inducidas , Humanos , Miocitos Cardíacos/metabolismo , Conexina 43/genética , Conexina 43/metabolismo , Desmoplaquinas/genética , Desmoplaquinas/metabolismo , Células Madre Pluripotentes Inducidas/metabolismo , Mutación
3.
Sci Transl Med ; 15(688): eadd4248, 2023 03 22.
Artículo en Inglés | MEDLINE | ID: mdl-36947592

RESUMEN

Arrhythmogenic cardiomyopathy (ACM) is an inherited progressive cardiac disease. Many patients with ACM harbor mutations in desmosomal genes, predominantly in plakophilin-2 (PKP2). Although the genetic basis of ACM is well characterized, the underlying disease-driving mechanisms remain unresolved. Explanted hearts from patients with ACM had less PKP2 compared with healthy hearts, which correlated with reduced expression of desmosomal and adherens junction (AJ) proteins. These proteins were also disorganized in areas of fibrotic remodeling. In vitro data from human-induced pluripotent stem cell-derived cardiomyocytes and microtissues carrying the heterozygous PKP2 c.2013delC pathogenic mutation also displayed impaired contractility. Knockin mice carrying the equivalent heterozygous Pkp2 c.1755delA mutation recapitulated changes in desmosomal and AJ proteins and displayed cardiac dysfunction and fibrosis with age. Global proteomics analysis of 4-month-old heterozygous Pkp2 c.1755delA hearts indicated involvement of the ubiquitin-proteasome system (UPS) in ACM pathogenesis. Inhibition of the UPS in mutant mice increased area composita proteins and improved calcium dynamics in isolated cardiomyocytes. Additional proteomics analyses identified lysine ubiquitination sites on the desmosomal proteins, which were more ubiquitinated in mutant mice. In summary, we show that a plakophilin-2 mutation can lead to decreased desmosomal and AJ protein expression through a UPS-dependent mechanism, which preceded cardiac remodeling. These findings suggest that targeting protein degradation and improving desmosomal protein stability may be a potential therapeutic strategy for the treatment of ACM.


Asunto(s)
Cardiomiopatías , Placofilinas , Humanos , Ratones , Animales , Lactante , Proteolisis , Placofilinas/genética , Placofilinas/metabolismo , Miocitos Cardíacos/metabolismo , Mutación/genética , Cardiomiopatías/genética
4.
Int J Mol Sci ; 24(6)2023 Mar 11.
Artículo en Inglés | MEDLINE | ID: mdl-36982449

RESUMEN

Chronic kidney disease (CKD) is represented by a diminished filtration capacity of the kidneys. End-stage renal disease patients need dialysis treatment to remove waste and toxins from the circulation. However, endogenously produced uremic toxins (UTs) cannot always be filtered during dialysis. UTs are among the CKD-related factors that have been linked to maladaptive and pathophysiological remodeling of the heart. Importantly, 50% of the deaths in dialysis patients are cardiovascular related, with sudden cardiac death predominating. However, the mechanisms responsible remain poorly understood. The current study aimed to assess the vulnerability of action potential repolarization caused by exposure to pre-identified UTs at clinically relevant concentrations. We exposed human induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs) and HEK293 chronically (48 h) to the UTs indoxyl sulfate, kynurenine, or kynurenic acid. We used optical and manual electrophysiological techniques to assess action potential duration (APD) in the hiPSC-CMs and recorded IKr currents in stably transfected HEK293 cells (HEK-hERG). Molecular analysis of KV11.1, the ion channel responsible for IKr, was performed to further understand the potential mechanism underlying the effects of the UTs. Chronic exposure to the UTs resulted in significant APD prolongation. Subsequent assessment of the repolarization current IKr, often most sensitive and responsible for APD alterations, showed decreased current densities after chronic exposure to the UTs. This outcome was supported by lowered protein levels of KV11.1. Finally, treatment with an activator of the IKr current, LUF7244, could reverse the APD prolongation, indicating the potential modulation of electrophysiological effects caused by these UTs. This study highlights the pro-arrhythmogenic potential of UTs and reveals a mode of action by which they affect cardiac repolarization.


Asunto(s)
Células Madre Pluripotentes Inducidas , Insuficiencia Renal Crónica , Humanos , Tóxinas Urémicas , Células HEK293 , Potenciales de Acción , Células Madre Pluripotentes Inducidas/metabolismo , Diálisis Renal , Miocitos Cardíacos , Insuficiencia Renal Crónica/metabolismo
5.
Sci Transl Med ; 13(612): eabf2750, 2021 Sep 22.
Artículo en Inglés | MEDLINE | ID: mdl-34550725

RESUMEN

Arrhythmogenic cardiomyopathy (ACM) is an inherited disorder often caused by pathogenic variants in desmosomal genes and characterized by progressive fibrotic and fat tissue accumulation in the heart. The cellular origin and responsible molecular mechanisms of fibro-fatty deposits have been a matter of debate, due to limitations in animal models recapitulating this phenotype. Here, we used human-induced pluripotent stem cell (hiPSC)­derived cardiac cultures, single-cell RNA sequencing (scRNA-seq), and explanted human ACM hearts to study the epicardial contribution to fibro-fatty remodeling in ACM. hiPSC-epicardial cells generated from patients with ACM showed spontaneous fibro-fatty cellular differentiation that was absent in isogenic controls. This was further corroborated upon siRNA-mediated targeting of desmosomal genes in hiPSC-epicardial cells generated from healthy donors. scRNA-seq analysis identified the transcription factor TFAP2A (activating enhancer-binding protein 2 alpha) as a key trigger promoting this process. Gain- and loss-of-function studies on hiPSC-epicardial cells and primary adult epicardial-derived cells demonstrated that TFAP2A mediated epicardial differentiation through enhancing epithelial-to-mesenchymal transition (EMT). Furthermore, examination of explanted hearts from patients with ACM revealed epicardial activation and expression of TFAP2A in the subepicardial mesenchyme. These data suggest that TFAP2A-mediated epicardial EMT underlies fibro-fatty remodeling in ACM, a process amenable to therapeutic intervention.


Asunto(s)
Displasia Ventricular Derecha Arritmogénica , Diferenciación Celular , Humanos
6.
Nat Commun ; 12(1): 84, 2021 01 04.
Artículo en Inglés | MEDLINE | ID: mdl-33398012

RESUMEN

The disruption in blood supply due to myocardial infarction is a critical determinant for infarct size and subsequent deterioration in function. The identification of factors that enhance cardiac repair by the restoration of the vascular network is, therefore, of great significance. Here, we show that the transcription factor Zinc finger E-box-binding homeobox 2 (ZEB2) is increased in stressed cardiomyocytes and induces a cardioprotective cross-talk between cardiomyocytes and endothelial cells to enhance angiogenesis after ischemia. Single-cell sequencing indicates ZEB2 to be enriched in injured cardiomyocytes. Cardiomyocyte-specific deletion of ZEB2 results in impaired cardiac contractility and infarct healing post-myocardial infarction (post-MI), while cardiomyocyte-specific ZEB2 overexpression improves cardiomyocyte survival and cardiac function. We identified Thymosin ß4 (TMSB4) and Prothymosin α (PTMA) as main paracrine factors released from cardiomyocytes to stimulate angiogenesis by enhancing endothelial cell migration, and whose regulation is validated in our in vivo models. Therapeutic delivery of ZEB2 to cardiomyocytes in the infarcted heart induces the expression of TMSB4 and PTMA, which enhances angiogenesis and prevents cardiac dysfunction. These findings reveal ZEB2 as a beneficial factor during ischemic injury, which may hold promise for the identification of new therapies.


Asunto(s)
Isquemia/patología , Miocitos Cardíacos/metabolismo , Neovascularización Fisiológica , Caja Homeótica 2 de Unión a E-Box con Dedos de Zinc/metabolismo , Animales , Movimiento Celular/genética , Proliferación Celular/genética , Dependovirus/metabolismo , Regulación de la Expresión Génica , Humanos , Isquemia/genética , Ratones Noqueados , Modelos Biológicos , Infarto del Miocardio/genética , Infarto del Miocardio/patología , Miocitos Cardíacos/patología , Neovascularización Fisiológica/genética , Precursores de Proteínas/genética , Precursores de Proteínas/metabolismo , ARN Mensajero/genética , ARN Mensajero/metabolismo , Timosina/análogos & derivados , Timosina/genética , Timosina/metabolismo , Caja Homeótica 2 de Unión a E-Box con Dedos de Zinc/genética
7.
Commun Biol ; 4(1): 146, 2021 01 29.
Artículo en Inglés | MEDLINE | ID: mdl-33514846

RESUMEN

The efficiency of the repair process following ischemic cardiac injury is a crucial determinant for the progression into heart failure and is controlled by both intra- and intercellular signaling within the heart. An enhanced understanding of this complex interplay will enable better exploitation of these mechanisms for therapeutic use. We used single-cell transcriptomics to collect gene expression data of all main cardiac cell types at different time-points after ischemic injury. These data unveiled cellular and transcriptional heterogeneity and changes in cellular function during cardiac remodeling. Furthermore, we established potential intercellular communication networks after ischemic injury. Follow up experiments confirmed that cardiomyocytes express and secrete elevated levels of beta-2 microglobulin in response to ischemic damage, which can activate fibroblasts in a paracrine manner. Collectively, our data indicate phase-specific changes in cellular heterogeneity during different stages of cardiac remodeling and allow for the identification of therapeutic targets relevant for cardiac repair.


Asunto(s)
Perfilación de la Expresión Génica , Daño por Reperfusión Miocárdica/genética , Miocitos Cardíacos/metabolismo , Análisis de la Célula Individual , Transcriptoma , Remodelación Ventricular , Cicatrización de Heridas , Microglobulina beta-2/genética , Animales , Línea Celular , Modelos Animales de Enfermedad , Fibroblastos/metabolismo , Fibroblastos/patología , Células Madre Pluripotentes Inducidas/metabolismo , Células Madre Pluripotentes Inducidas/patología , Macrófagos/metabolismo , Macrófagos/patología , Ratones Endogámicos C57BL , Daño por Reperfusión Miocárdica/metabolismo , Daño por Reperfusión Miocárdica/patología , Daño por Reperfusión Miocárdica/fisiopatología , Miocitos Cardíacos/patología , Comunicación Paracrina , Factores de Tiempo , Microglobulina beta-2/metabolismo
8.
PLoS One ; 12(12): e0189139, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-29206857

RESUMEN

AIMS: The Z-disc is a crucial structure of the sarcomere and is implicated in mechanosensation/transduction. Dysregulation of Z-disc proteins often result in cardiomyopathy. We have previously shown that the Z-disc protein Cytoskeletal Heart-enriched Actin-associated Protein (CHAP) is essential for cardiac and skeletal muscle development. Furthermore, the CHAP gene has been associated with atrial fibrillation in humans. Here, we studied the misregulated expression of CHAP isoforms in heart disease. METHODS AND RESULTS: Mice that underwent transverse aortic constriction and calcineurin transgenic (Tg) mice, both models of experimental heart failure, displayed a significant increase in cardiac expression of fetal isoform CHAPb. To investigate whether increased expression of CHAPb postnatally is sufficient to induce cardiomyopathy, we generated CHAPb Tg mice under the control of the cardiac-specific αMHC promoter. CHAPb Tg mice displayed cardiac hypertrophy, interstitial fibrosis and enlargement of the left atrium at three months, which was more pronounced at the age of six months. Hypertrophy and fibrosis were confirmed by evidence of activation of the hypertrophic gene program (Nppa, Nppb, Myh7) and increased collagen expression, respectively. Connexin40 and 43 were downregulated in the left atrium, which was associated with delayed atrioventricular conduction. Tg hearts displayed both systolic and diastolic dysfunction partly caused by impaired sarcomere function evident from a reduced force generating capacity of single cardiomyocytes. This co-incided with activation of the actin signalling pathway leading to the formation of stress fibers. CONCLUSION: This study demonstrated that the fetal isoform CHAPb initiates progression towards cardiac hypertrophy, which is accompanied by delayed atrioventricular conduction and diastolic dysfunction. Moreover, CHAP may be a novel therapeutic target or candidate gene for screening in cardiomyopathies and atrial fibrillation.


Asunto(s)
Cardiomiopatías/metabolismo , Proteínas de Microfilamentos/metabolismo , Proteínas Musculares/metabolismo , Contracción Miocárdica , Isoformas de Proteínas/metabolismo , Animales , Ratones , Ratones Transgénicos
9.
Stem Cell Reports ; 9(6): 1765-1779, 2017 12 12.
Artículo en Inglés | MEDLINE | ID: mdl-29173897

RESUMEN

Reporter cell lines have already proven valuable in identifying, tracking, and purifying cardiac subtypes and progenitors during differentiation of human pluripotent stem cells (hPSCs). We previously showed that chick ovalbumin upstream promoter transcription factor II (COUP-TFII) is highly enriched in human atrial cardiomyocytes (CMs), but not ventricular. Here, we targeted mCherry to the COUP-TFII genomic locus in hPSCs expressing GFP from the NKX2.5 locus. This dual atrial NKX2.5EGFP/+-COUP-TFIImCherry/+ reporter line allowed identification and selection of GFP+ (G+)/mCherry+ (M+) CMs following cardiac differentiation. These cells exhibited transcriptional and functional properties of atrial CMs, whereas G+/M- CMs displayed ventricular characteristics. Via CRISPR/Cas9-mediated knockout, we demonstrated that COUP-TFII is not required for atrial specification in hPSCs. This new tool allowed selection of human atrial and ventricular CMs from mixed populations, of relevance for studying cardiac specification, developing human atrial disease models, and examining distinct effects of drugs on the atrium versus ventricle.


Asunto(s)
Diferenciación Celular/genética , Atrios Cardíacos/citología , Células Madre Embrionarias Humanas/citología , Miocitos Cardíacos/citología , Células Madre Pluripotentes/citología , Animales , Factor de Transcripción COUP II/genética , Sistemas CRISPR-Cas/genética , Rastreo Celular/métodos , Embrión de Pollo , Genes Reporteros/genética , Proteínas Fluorescentes Verdes , Atrios Cardíacos/crecimiento & desarrollo , Atrios Cardíacos/metabolismo , Células Madre Embrionarias Humanas/metabolismo , Humanos , Ratones , Miocitos Cardíacos/metabolismo , Ovalbúmina/genética , Células Madre Pluripotentes/metabolismo , Regiones Promotoras Genéticas
10.
EMBO Mol Med ; 8(12): 1390-1408, 2016 12.
Artículo en Inglés | MEDLINE | ID: mdl-27861123

RESUMEN

Genetic causes of many familial arrhythmia syndromes remain elusive. In this study, whole-exome sequencing (WES) was carried out on patients from three different families that presented with life-threatening arrhythmias and high risk of sudden cardiac death (SCD). Two French Canadian probands carried identical homozygous rare variant in TECRL gene (p.Arg196Gln), which encodes the trans-2,3-enoyl-CoA reductase-like protein. Both patients had cardiac arrest, stress-induced atrial and ventricular tachycardia, and QT prolongation on adrenergic stimulation. A third patient from a consanguineous Sudanese family diagnosed with catecholaminergic polymorphic ventricular tachycardia (CPVT) had a homozygous splice site mutation (c.331+1G>A) in TECRL Analysis of intracellular calcium ([Ca2+]i) dynamics in human induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs) generated from this individual (TECRLHom-hiPSCs), his heterozygous but clinically asymptomatic father (TECRLHet-hiPSCs), and a healthy individual (CTRL-hiPSCs) from the same Sudanese family, revealed smaller [Ca2+]i transient amplitudes as well as elevated diastolic [Ca2+]i in TECRLHom-hiPSC-CMs compared with CTRL-hiPSC-CMs. The [Ca2+]i transient also rose markedly slower and contained lower sarcoplasmic reticulum (SR) calcium stores, evidenced by the decreased magnitude of caffeine-induced [Ca2+]i transients. In addition, the decay phase of the [Ca2+]i transient was slower in TECRLHom-hiPSC-CMs due to decreased SERCA and NCX activities. Furthermore, TECRLHom-hiPSC-CMs showed prolonged action potentials (APs) compared with CTRL-hiPSC-CMs. TECRL knockdown in control human embryonic stem cell-derived CMs (hESC-CMs) also resulted in significantly longer APs. Moreover, stimulation by noradrenaline (NA) significantly increased the propensity for triggered activity based on delayed afterdepolarizations (DADs) in TECRLHom-hiPSC-CMs and treatment with flecainide, a class Ic antiarrhythmic drug, significantly reduced the triggered activity in these cells. In summary, we report that mutations in TECRL are associated with inherited arrhythmias characterized by clinical features of both LQTS and CPVT Patient-specific hiPSC-CMs recapitulated salient features of the clinical phenotype and provide a platform for drug screening evidenced by initial identification of flecainide as a potential therapeutic. These findings have implications for diagnosis and treatment of inherited cardiac arrhythmias.


Asunto(s)
Arritmias Cardíacas/genética , Arritmias Cardíacas/patología , Predisposición Genética a la Enfermedad , Mutación , Oxidorreductasas/genética , Adolescente , Adulto , Células Cultivadas , Exoma , Femenino , Genoma Humano , Humanos , Masculino , Análisis de Secuencia de ADN , Adulto Joven
11.
Biomaterials ; 51: 138-150, 2015 May.
Artículo en Inglés | MEDLINE | ID: mdl-25771005

RESUMEN

Cardiomyocytes from human pluripotent stem cells (hPSC-CM) have many potential applications in disease modelling and drug target discovery but their phenotypic similarity to early fetal stages of cardiac development limits their applicability. In this study we compared contraction stresses of hPSC-CM to 2nd trimester human fetal derived cardiomyocytes (hFetal-CM) by imaging displacement of fluorescent beads by single contracting hPSC-CM, aligned by microcontact-printing on polyacrylamide gels. hPSC-CM showed distinctly lower contraction stress than cardiomyocytes isolated from hFetal-CM. To improve maturation of hPSC-CM in vitro we made use of commercial media optimized for cardiomyocyte maturation, which promoted significantly higher contraction stress in hPSC-compared with hFetal-CM. Accordingly, other features of cardiomyocyte maturation were observed, most strikingly increased upstroke velocities and action potential amplitudes, lower resting membrane potentials, improved sarcomeric organization and alterations in cardiac-specific gene expression. Performing contraction force and electrophysiology measurements on individual cardiomyocytes revealed strong correlations between an increase in contraction force and a rise of the upstroke velocity and action potential amplitude and with a decrease in the resting membrane potential. We showed that under standard differentiation conditions hPSC-CM display lower contractile force than primary hFetal-CM and identified conditions under which a commercially available culture medium could induce molecular, morphological and functional maturation of hPSC-CM in vitro. These results are an important contribution for full implementation of hPSC-CM in cardiac disease modelling and drug discovery.


Asunto(s)
Diferenciación Celular , Fenómenos Electrofisiológicos , Contracción Miocárdica , Miocitos Cardíacos/citología , Células Madre Pluripotentes/citología , Fenómenos Biomecánicos , Feto/citología , Regulación de la Expresión Génica , Células Madre Embrionarias Humanas/citología , Humanos , Sarcómeros/metabolismo , Estrés Fisiológico
12.
Stem Cells ; 33(1): 56-67, 2015 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-25187301

RESUMEN

Understanding early differentiation events leading to cardiogenesis is crucial for controlling fate of human pluripotent stem cells and developing protocols that yield sufficient cell numbers for use in regenerative medicine and drug screening. Here, we develop a new tool to visualize patterning of early cardiac mesoderm and cardiomyocyte development in vitro by generating a dual MESP1(mCherry/w)-NKX2-5(eGFP/w) reporter line in human embryonic stem cells (hESCs) and using it to examine signals that lead to formation of cardiac progenitors and subsequent differentiation. MESP1 is a pivotal transcription factor for precardiac mesoderm in the embryo, from which the majority of cardiovascular cells arise. Transcription factor NKX2-5 is expressed upon cardiac crescent formation. Induction of cardiac differentiation in this reporter line resulted in transient expression of MESP1-mCherry, followed by continuous expression of NKX2-5-eGFP. MESP1-mCherry cells showed increased expression of mesodermal and epithelial-mesenchymal-transition markers confirming their mesodermal identity. Whole-genome microarray profiling and fluorescence-activated cell sorting analysis of MESP1-mCherry cells showed enrichment for mesodermal progenitor cell surface markers PDGFR-α, CD13, and ROR-2. No enrichment was found for the previously described KDR+PDGFR-α+ progenitors. MESP1-mCherry derivatives contained an enriched percentage of NKX2-5-eGFP and Troponin T expressing cells, indicating preferential cardiac differentiation; this was enhanced by inhibition of the Wnt-pathway. Furthermore, MESP1-mCherry derivatives harbored smooth muscle cells and endothelial cells, demonstrating their cardiac and vascular differentiation potential under appropriate conditions. The MESP1-NKX2-5 hESC reporter line allows us to identify molecular cues crucial for specification and expansion of human cardiac mesoderm and early progenitors and their differentiation to specific cardiovascular derivatives.


Asunto(s)
Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Células Madre Embrionarias/fisiología , Proteínas de Homeodominio/genética , Miocardio/citología , Células Madre/metabolismo , Factores de Transcripción/genética , Animales , Diferenciación Celular/fisiología , Células Madre Embrionarias/citología , Transición Epitelial-Mesenquimal , Proteínas Fluorescentes Verdes/genética , Proteína Homeótica Nkx-2.5 , Humanos , Mesodermo/citología , Mesodermo/fisiología , Ratones , Proteínas Recombinantes de Fusión/genética , Transducción de Señal
13.
Int J Dev Biol ; 56(4): 301-9, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22562206

RESUMEN

SARCOSIN, also named Krp1, has been identified as a protein exclusively expressed in striated muscle tissue. Here we report on the role of SARCOSIN in skeletal muscle development and differentiation. We demonstrate, by means of whole-mount in situ hybridization, that Sarcosin mRNA is expressed in the myotome part of the mature somites in mouse embryos from embryonic day 9.5 onwards. Sarcosin is not expressed in the developing heart at these embryonic stages, and in adult tissues the mRNA expression levels are five times lower in the heart than in skeletal muscle. SARCOSIN protein partially co-localizes with the M-band protein myomesin and between and below laterally fusing myofibrils in adult skeletal muscle tissue. RNA interference mediated knock-down of SARCOSIN in the C2C12 myoblast cell line appeared to be stimulatory in the early phase of differentiation, but inhibitory at a later phase of differentiation.


Asunto(s)
Diferenciación Celular/genética , Desarrollo de Músculos/genética , Proteínas Musculares/genética , Músculo Esquelético/metabolismo , Animales , Línea Celular , Conectina , Embrión de Mamíferos/citología , Embrión de Mamíferos/embriología , Embrión de Mamíferos/metabolismo , Femenino , Perfilación de la Expresión Génica , Regulación del Desarrollo de la Expresión Génica , Immunoblotting , Hibridación in Situ , Masculino , Ratones , Ratones Endogámicos C57BL , Microscopía Fluorescente , Fibras Musculares Esqueléticas/citología , Fibras Musculares Esqueléticas/metabolismo , Proteínas Musculares/metabolismo , Músculo Esquelético/citología , Músculo Esquelético/embriología , Mioblastos/citología , Mioblastos/metabolismo , Interferencia de ARN , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Somitos/embriología , Somitos/metabolismo , Factores de Tiempo
14.
Int J Dev Biol ; 55(6): 649-55, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-21948713

RESUMEN

We recently identified a new Z-disc protein, CHAP (Cytoskeletal Heart-enriched Actin-associated Protein), which is expressed in striated muscle and plays an important role during embryonic muscle development in mouse and zebrafish. Here, we confirm and further extend these findings by (i) the identification and characterization of the CHAP orthologue in chick and (ii) providing a detailed analysis of CHAP expression in mouse during embryonic and adult stages. Chick CHAP contains a PDZ domain and a nuclear localization signal, resembling the human and mouse CHAPa. CHAP is expressed in the developing heart and somites, as well as muscle precursors of the limb buds in mouse and chick embryos. CHAP expression in heart and skeletal muscle is maintained in adult mice, both in slow and fast muscle fibers. Moreover, besides expression in striated muscle, we demonstrate that CHAP is expressed in smooth muscle cells of aorta, carotid and coronary arteries in adult mice, but not during embryonic development.


Asunto(s)
Proteínas del Citoesqueleto/metabolismo , Proteínas de Microfilamentos/metabolismo , Proteínas Musculares/metabolismo , Músculo Liso/metabolismo , Músculo Estriado/metabolismo , Citoesqueleto de Actina/metabolismo , Secuencia de Aminoácidos , Animales , Aorta/metabolismo , Proteínas Aviares/química , Proteínas Aviares/metabolismo , Arterias Carótidas/metabolismo , Diferenciación Celular , Embrión de Pollo , Pollos , Vasos Coronarios/metabolismo , Proteínas del Citoesqueleto/biosíntesis , Desarrollo Embrionario , Femenino , Corazón/embriología , Corazón/fisiología , Esbozos de los Miembros/embriología , Esbozos de los Miembros/metabolismo , Ratones , Proteínas de Microfilamentos/química , Datos de Secuencia Molecular , Desarrollo de Músculos , Proteínas Musculares/química , Músculo Esquelético/embriología , Músculo Liso/embriología , Músculo Estriado/embriología , Miocitos Cardíacos/metabolismo , Miocitos del Músculo Liso/metabolismo , Embarazo , Isoformas de Proteínas , Alineación de Secuencia , Análisis de Secuencia de Proteína , Somitos/embriología , Somitos/metabolismo
15.
J Cell Sci ; 123(Pt 7): 1141-50, 2010 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-20215401

RESUMEN

In recent years, the perception of Z-disc function has changed from a passive anchor for myofilaments that allows transmission of force, to a dynamic multicomplex structure, capable of sensing and transducing extracellular signals. Here, we describe a new Z-disc protein, which we named CHAP (cytoskeletal heart-enriched actin-associated protein), expressed in differentiating heart and skeletal muscle in vitro and in vivo. Interestingly, in addition to its sarcomeric localization, CHAP was also able to translocate to the nucleus. CHAP was associated with filamentous actin in the cytoplasm and the nucleus when expressed ectopically in vitro, but in rat neonatal cardiomyocytes, CHAP disrupted the subcellular localization of alpha-actinin, another Z-disc protein. More importantly, knockdown of CHAP in zebrafish resulted in aberrant cardiac and skeletal muscle development and function. These findings suggest that CHAP is a critical component of the sarcomere with an important role in muscle development.


Asunto(s)
Núcleo Celular/metabolismo , Proteínas de Microfilamentos/metabolismo , Miocitos Cardíacos/metabolismo , Sarcómeros/metabolismo , Transporte Activo de Núcleo Celular , Animales , Animales Modificados Genéticamente , Células COS , Chlorocebus aethiops , Embrión de Mamíferos , Técnicas de Silenciamiento del Gen , Corazón/embriología , Corazón/fisiología , Ratones , Proteínas de Microfilamentos/genética , Desarrollo de Músculos , Músculo Esquelético/embriología , Músculo Esquelético/fisiología , Músculo Esquelético/ultraestructura , Miocitos Cardíacos/ultraestructura , Ratas , Sarcómeros/ultraestructura , Pez Cebra , Proteínas de Pez Cebra/genética , Proteínas de Pez Cebra/metabolismo
16.
J Proteome Res ; 9(3): 1610-8, 2010 Mar 05.
Artículo en Inglés | MEDLINE | ID: mdl-20088484

RESUMEN

The absence of identified cell surface proteins and corresponding antibodies to most differentiated derivatives of human embryonic stem cells (hESCs) has largely limited selection of specific cell types from mixed cell populations to genetic approaches. Here, we describe the use of mass spectrometry (MS)-based proteomics on cell membrane proteins isolated from hESCs that were differentiated into cardiomyocytes to identify candidate proteins for this particular lineage. Quantitative MS distinguished cardiomyocyte-specific plasma membrane proteins that were highly enriched or detected only in cardiomyocytes derived from hESCs and human fetal hearts compared with a heterogeneous pool of hESC-derived differentiated cells. For several candidates, cardiomyocyte-specific expression and cell surface localization were verified by conventional antibody-based methodologies. Using an antibody against elastin microfibril interfacer 2 (EMILIN2), we demonstrate that cardiomyocytes can be sorted from live cell populations. Besides showing that MS-based membrane proteomics is a powerful tool to identify candidate proteins that allow purification of specific cell lineages from heterogeneous populations, this approach generated a plasma membrane proteome profile suggesting signaling pathways that control cell behavior.


Asunto(s)
Biomarcadores/análisis , Células Madre Embrionarias/metabolismo , Proteínas de la Membrana/análisis , Miocitos Cardíacos/metabolismo , Proteómica/métodos , Anticuerpos/metabolismo , Biomarcadores/metabolismo , Western Blotting , Diferenciación Celular , Separación Celular , Células Madre Embrionarias/citología , Fibrilinas , Glicoproteínas/metabolismo , Humanos , Marcaje Isotópico , Espectrometría de Masas , Proteínas de la Membrana/metabolismo , Proteínas de Microfilamentos/metabolismo , Microscopía Fluorescente , Miocitos Cardíacos/citología
17.
Dev Dyn ; 238(11): 2903-11, 2009 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-19806667

RESUMEN

Recently, we have performed a whole genome micro-array analysis on human embryonic stem cells differentiating toward cardiomyocytes, which resulted in the identification of novel genes that were highly up-regulated during differentiation. Here, we describe one of these novel genes annotated as KIAA0774. The predicted protein contains a leucine-zipper domain at the C-terminus and has at least two isoforms (358 and 1354 amino acids). Whole-mount in situ hybridization confirmed that the mRNA of both the mouse and chicken orthologs of KIAA0774 is expressed during early cardiac development. Hence, we named this protein CAZIP (cardiac zipper protein). Later during embryonic development, Cazip was also expressed in parts of the nervous system. Northern blot and real-time polymerase chain reaction analysis showed that Cazip is expressed in heart and brain in adult mice. These results suggest a role for CAZIP in development and function of the heart and nervous system in vertebrates.


Asunto(s)
Proteínas Portadoras/metabolismo , Embrión de Mamíferos/embriología , Embrión no Mamífero/embriología , Corazón/embriología , Sistema Nervioso/embriología , Secuencia de Aminoácidos , Animales , Células COS , Proteínas Portadoras/química , Proteínas Portadoras/genética , Línea Celular , Embrión de Pollo , Chlorocebus aethiops , Embrión de Mamíferos/metabolismo , Embrión no Mamífero/metabolismo , Células Madre Embrionarias/metabolismo , Corazón/fisiología , Humanos , Ratones , Proteínas Asociadas a Microtúbulos , Datos de Secuencia Molecular , Sistema Nervioso/metabolismo , ARN Mensajero/metabolismo , Alineación de Secuencia
18.
Stem Cell Res ; 3(2-3): 106-12, 2009.
Artículo en Inglés | MEDLINE | ID: mdl-19560991

RESUMEN

Transplantation of human embryonic stem cell-derived cardiomyocytes (hESC-CM) has been shown to improve the function of the rodent heart 1 month after myocardial infarction (MI). However, the mechanistic basis and optimal delivery strategies are unclear. We investigated the influence of the number of injected cells, resulting graft size, and possible paracrine mechanisms in this process. MI was induced in NOD-SCID mice (n=84) followed by injection of enriched hESC-CM at different dosages, hESC-non-CM derivatives, culture medium, or no injection. Cardiac function was monitored for 12 weeks with 9.4 T MRI (n=70). Grafts were identified by epifluorescence of a transgenic GFP marker and characterized by immunofluorescence. Vascularity and paracrine effects were investigated immunohistochemically. Transplantation of differentiated hESCs improved short, mid-, and long-term cardiac performance and survival, although only cardiomyocytes formed grafts. A mid-term (4 weeks) cardiomyocyte-specific enhancement was associated with elevated vascular density around the graft and attenuated compensatory remodeling. However, increasing the number of hESC-CM for injection did not enhance heart function further. Moreover, we observed that small graft size was associated with a better functional outcome. HESC-CM increased myocardial vascularization and enhanced heart function in mice after MI, but larger graft size was associated with reduced functional improvement. Future studies should focus on advanced delivery strategies and mechanisms of action rather than increasing graft size.


Asunto(s)
Células Madre Embrionarias/trasplante , Miocardio/citología , Miocitos Cardíacos/fisiología , Animales , Células Madre Embrionarias/citología , Supervivencia de Injerto , Proteínas Fluorescentes Verdes/metabolismo , Humanos , Ratones , Ratones Endogámicos NOD , Ratones SCID , Infarto del Miocardio/terapia , Miocardio/patología , Miocitos Cardíacos/citología , Trasplante Heterólogo
19.
Stem Cells ; 26(3): 724-33, 2008 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-18096723

RESUMEN

Human embryonic stem cells (hESC) can proliferate indefinitely while retaining the capacity to form derivatives of all three germ layers. We have reported previously that hESC differentiate into cardiomyocytes when cocultured with a visceral endoderm-like cell line (END-2). Insulin/insulin-like growth factors and their intracellular downstream target protein kinase Akt are known to protect many cell types from apoptosis and to promote proliferation, including hESC-derived cardiomyocytes. Here, we show that in the absence of insulin, a threefold increase in the number of beating areas was observed in hESC/END-2 coculture. In agreement, the addition of insulin strongly inhibited cardiac differentiation, as evidenced by a significant reduction in beating areas, as well as in alpha-actinin and beta-myosin heavy chain (beta-MHC)-expressing cells. Real-time reverse transcription-polymerase chain reaction and Western blot analysis showed that insulin inhibited cardiomyogenesis in the early phase of coculture by suppressing the expression of endoderm (Foxa2, GATA-6), mesoderm (brachyury T), and cardiac mesoderm (Nkx2.5, GATA-4). In contrast to previous reports, insulin was not sufficient to maintain hESC in an undifferentiated state, since expression of the pluripotency markers Oct3/4 and nanog declined independently of the presence of insulin during coculture. Instead, insulin promoted the expression of neuroectodermal markers. Since insulin triggered sustained phosphorylation of Akt in hESC, we analyzed the effect of an Akt inhibitor during coculture. Indeed, the inhibition of Akt or insulin-like growth factor-1 receptor reversed the insulin-dependent effects. We conclude that in hESC/END-2 cocultures, insulin does not prevent differentiation but favors the neuroectodermal lineage at the expense of mesendodermal lineages.


Asunto(s)
Diferenciación Celular/efectos de los fármacos , Células Madre Embrionarias/citología , Endodermo/citología , Insulina/farmacología , Mesodermo/citología , Miocitos Cardíacos/citología , Placa Neural/citología , Animales , Biomarcadores/metabolismo , Linaje de la Célula/efectos de los fármacos , Células Madre Embrionarias/efectos de los fármacos , Células Madre Embrionarias/enzimología , Endodermo/efectos de los fármacos , Humanos , Mesodermo/efectos de los fármacos , Ratones , Modelos Biológicos , Miocitos Cardíacos/efectos de los fármacos , Placa Neural/efectos de los fármacos , Proteínas Proto-Oncogénicas c-akt/antagonistas & inhibidores , Receptor IGF Tipo 1/metabolismo
20.
Nat Protoc ; 2(10): 2551-67, 2007.
Artículo en Inglés | MEDLINE | ID: mdl-17947998

RESUMEN

We have developed a mouse severe combined immunodeficient (SCID) model of myocardial infarction based on permanent coronary artery occlusion that allows long-term functional analysis of engrafted human embryonic stem cell-derived cardiomyocytes, genetically marked with green fluorescent protein (GFP), in the mouse heart. We describe methods for delivery of dissociated cardiomyocytes to the left ventricle that minimize scar formation and visualization and validation of the identity of the engrafted cells using the GFP emission spectrum, and histological techniques compatible with GFP epifluorescence, for monitoring phenotypic changes in the grafts in vivo. In addition, we describe how magnetic resonance imaging can be adapted for use in mice to monitor cardiac function non-invasively and repeatedly. The model can be adapted to include multiple control or other cell populations. The procedure for a cohort of six mice can be completed in a maximum of 13 weeks, depending on follow-up, with 30 h of hands-on time.


Asunto(s)
Modelos Animales de Enfermedad , Células Madre Embrionarias/trasplante , Imagen por Resonancia Magnética/métodos , Ratones , Infarto del Miocardio/terapia , Animales , Técnicas de Cultivo de Célula , Diferenciación Celular , Trasplante de Células/instrumentación , Trasplante de Células/métodos , Cicatriz , Oclusión Coronaria/inmunología , Oclusión Coronaria/terapia , Crioultramicrotomía , Células Madre Embrionarias/citología , Técnica del Anticuerpo Fluorescente , Proteínas Fluorescentes Verdes/análisis , Humanos , Huésped Inmunocomprometido , Infarto del Miocardio/inmunología , Miocardio/patología , Miocitos Cardíacos/citología , Miocitos Cardíacos/trasplante
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...