Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 32
Filtrar
1.
Genes Dev ; 31(17): 1770-1783, 2017 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-28982760

RESUMEN

Direct reprogramming of fibroblasts to cardiomyocytes represents a potential means of restoring cardiac function following myocardial injury. AKT1 in the presence of four cardiogenic transcription factors, GATA4, HAND2, MEF2C, and TBX5 (AGHMT), efficiently induces the cardiac gene program in mouse embryonic fibroblasts but not adult fibroblasts. To identify additional regulators of adult cardiac reprogramming, we performed an unbiased screen of transcription factors and cytokines for those that might enhance or suppress the cardiogenic activity of AGHMT in adult mouse fibroblasts. Among a collection of inducers and repressors of cardiac reprogramming, we discovered that the zinc finger transcription factor 281 (ZNF281) potently stimulates cardiac reprogramming by genome-wide association with GATA4 on cardiac enhancers. Concomitantly, ZNF281 suppresses expression of genes associated with inflammatory signaling, suggesting the antagonistic convergence of cardiac and inflammatory transcriptional programs. Consistent with an inhibitory influence of inflammatory pathways on cardiac reprogramming, blockade of these pathways with anti-inflammatory drugs or components of the nucleosome remodeling deacetylase (NuRD) complex, which associate with ZNF281, stimulates cardiac gene expression. We conclude that ZNF281 acts at a nexus of cardiac and inflammatory gene programs, which exert opposing influences on fibroblast to cardiac reprogramming.


Asunto(s)
Reprogramación Celular/genética , Regulación de la Expresión Génica/genética , Factores de Transcripción/metabolismo , Antiinflamatorios/farmacología , Reprogramación Celular/efectos de los fármacos , Fibroblastos/fisiología , Factor de Transcripción GATA4/metabolismo , Regulación de la Expresión Génica/efectos de los fármacos , Estudio de Asociación del Genoma Completo , Complejo Desacetilasa y Remodelación del Nucleosoma Mi-2/metabolismo , Miocitos Cardíacos/efectos de los fármacos , Miocitos Cardíacos/fisiología , Proteínas Represoras , Transcriptoma
2.
Biochem Biophys Res Commun ; 632: 181-188, 2022 12 03.
Artículo en Inglés | MEDLINE | ID: mdl-36215905

RESUMEN

The number of patients with heart failure and related deaths is rapidly increasing worldwide, making it a major problem. Cardiac hypertrophy is a crucial preliminary step in heart failure, but its treatment has not yet been fully successful. In this study, we established a system to evaluate cardiomyocyte hypertrophy using a deep learning-based high-throughput screening system and identified drugs that inhibit it. First, primary cultured cardiomyocytes from neonatal rats were stimulated by both angiotensin II and endothelin-1, and cellular images were captured using a phase-contrast microscope. Subsequently, we used a deep learning model for instance segmentation and established a system to automatically and unbiasedly evaluate the cardiomyocyte size and perimeter. Using this system, we screened 100 FDA-approved drugs library and identified 12 drugs that inhibited cardiomyocyte hypertrophy. We focused on ezetimibe, a cholesterol absorption inhibitor, that inhibited cardiomyocyte hypertrophy in a dose-dependent manner in vitro. Additionally, ezetimibe improved the cardiac dysfunction induced by pressure overload in mice. These results suggest that the deep learning-based system is useful for the evaluation of cardiomyocyte hypertrophy and drug screening, leading to the development of new treatments for heart failure.


Asunto(s)
Cardiomegalia , Aprendizaje Profundo , Evaluación Preclínica de Medicamentos , Insuficiencia Cardíaca , Animales , Ratones , Ratas , Angiotensina II/farmacología , Cardiomegalia/diagnóstico por imagen , Cardiomegalia/tratamiento farmacológico , Células Cultivadas , Colesterol , Evaluación Preclínica de Medicamentos/métodos , Endotelina-1 , Ezetimiba , Insuficiencia Cardíaca/tratamiento farmacológico , Miocitos Cardíacos/citología , Miocitos Cardíacos/efectos de los fármacos
3.
Int J Mol Sci ; 21(5)2020 Mar 06.
Artículo en Inglés | MEDLINE | ID: mdl-32155842

RESUMEN

Skeletal muscle comprises 30-40% of the weight of a healthy human body and is required for voluntary movements in humans. Mature skeletal muscle is formed by multinuclear cells, which are called myofibers. Formation of myofibers depends on the proliferation, differentiation, and fusion of muscle progenitor cells during development and after injury. Muscle progenitor cells are derived from muscle satellite (stem) cells (MuSCs), which reside on the surface of the myofiber but beneath the basement membrane. MuSCs play a central role in postnatal maintenance, growth, repair, and regeneration of skeletal muscle. In sedentary adult muscle, MuSCs are mitotically quiescent, but are promptly activated in response to muscle injury. Physiological and chronological aging induces MuSC aging, leading to an impaired regenerative capability. Importantly, in pathological situations, repetitive muscle injury induces early impairment of MuSCs due to stem cell aging and leads to early impairment of regeneration ability. In this review, we discuss (1) the role of MuSCs in muscle regeneration, (2) stem cell aging under physiological and pathological conditions, and (3) prospects related to clinical applications of controlling MuSCs.


Asunto(s)
Envejecimiento/fisiología , Senescencia Celular , Desarrollo de Músculos , Músculo Esquelético/citología , Enfermedades Musculares/terapia , Regeneración , Células Madre/citología , Animales , Diferenciación Celular , Humanos , Músculo Esquelético/fisiología , Enfermedades Musculares/fisiopatología , Células Madre/fisiología
4.
EMBO J ; 33(14): 1565-81, 2014 Jul 17.
Artículo en Inglés | MEDLINE | ID: mdl-24920580

RESUMEN

Fibroblasts can be directly reprogrammed into cardiomyocyte-like cells (iCMs) by overexpression of cardiac transcription factors or microRNAs. However, induction of functional cardiomyocytes is inefficient, and molecular mechanisms of direct reprogramming remain undefined. Here, we demonstrate that addition of miR-133a (miR-133) to Gata4, Mef2c, and Tbx5 (GMT) or GMT plus Mesp1 and Myocd improved cardiac reprogramming from mouse or human fibroblasts by directly repressing Snai1, a master regulator of epithelial-to-mesenchymal transition. MiR-133 overexpression with GMT generated sevenfold more beating iCMs from mouse embryonic fibroblasts and shortened the duration to induce beating cells from 30 to 10 days, compared to GMT alone. Snai1 knockdown suppressed fibroblast genes, upregulated cardiac gene expression, and induced more contracting iCMs with GMT transduction, recapitulating the effects of miR-133 overexpression. In contrast, overexpression of Snai1 in GMT/miR-133-transduced cells maintained fibroblast signatures and inhibited generation of beating iCMs. MiR-133-mediated Snai1 repression was also critical for cardiac reprogramming in adult mouse and human cardiac fibroblasts. Thus, silencing fibroblast signatures, mediated by miR-133/Snai1, is a key molecular roadblock during cardiac reprogramming.


Asunto(s)
Transdiferenciación Celular/fisiología , Fibroblastos/metabolismo , Regulación de la Expresión Génica/fisiología , MicroARNs/metabolismo , Miocitos Cardíacos/metabolismo , Factores de Transcripción/genética , Análisis de Varianza , Animales , Western Blotting , Transdiferenciación Celular/genética , Clonación Molecular , Fibroblastos/citología , Citometría de Flujo , Técnicas de Silenciamiento del Gen , Proteínas Fluorescentes Verdes , Humanos , Inmunohistoquímica , Ratones , Análisis por Micromatrices , Miocitos Cardíacos/citología , Reacción en Cadena en Tiempo Real de la Polimerasa , Factores de Transcripción de la Familia Snail , Factores de Transcripción/metabolismo
5.
Biochem Biophys Res Commun ; 464(4): 1000-1007, 2015 Sep 04.
Artículo en Inglés | MEDLINE | ID: mdl-26168730

RESUMEN

The precise assemblage of several types of cardiac precursors controls heart organogenesis. The cardiac precursors show dynamic movement during early development and then form the complicated heart structure. However, cardiomyocyte movements inside the newly organized mammalian heart remain unclear. We previously established the method of ex vivo time-lapse imaging of the murine heart to study cardiomyocyte behavior by using the Fucci (fluorescent ubiquitination-based cell cycle indicator) system, which can effectively label individual G1, S/G2/M, and G1/S-transition phase nuclei in living cardiomyocytes as red, green, and yellow, respectively. Global analysis of gene expression in Fucci green positive ventricular cardiomyocytes confirmed that cell cycle regulatory genes expressed in G1/S, S, G2/M, and M phase transitions were upregulated. Interestingly, pathway analysis revealed that many genes related to the cell cycle were significantly upregulated in the Fucci green positive ventricular cardiomyocytes, while only a small number of genes related to cell motility were upregulated. Time-lapse imaging showed that murine proliferating cardiomyocytes did not exhibit dynamic movement inside the heart, but stayed on site after entering the cell cycle.


Asunto(s)
Corazón Fetal/citología , Miocardio/citología , Miocitos Cardíacos/citología , Miocitos Cardíacos/fisiología , Animales , Puntos de Control del Ciclo Celular/genética , Movimiento Celular , Proliferación Celular , Femenino , Corazón Fetal/embriología , Regulación del Desarrollo de la Expresión Génica , Genes Reporteros , Corazón/crecimiento & desarrollo , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Miocardio/metabolismo , Análisis de Secuencia por Matrices de Oligonucleótidos , Embarazo
6.
J Mol Cell Cardiol ; 72: 241-9, 2014 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-24704900

RESUMEN

Mammalian cardiomyocytes withdraw from the cell cycle shortly after birth, although it remains unclear how cardiomyocyte cell cycles behave during development. Compared to conventional immunohistochemistry in static observation, time-lapse imaging can reveal comprehensive data in hard-to-understand biological phenomenon. However, there are no reports of an established protocol of successful time-lapse imaging in mammalian heart. Thus, it is valuable to establish a time-lapse imaging system to enable the observation of cell cycle dynamics in living murine cardiomyocytes. This study sought to establish time-lapse imaging of murine heart to study cardiomyocyte cell cycle behavior. The Fucci (fluorescent ubiquitination-based cell cycle indicator) system can effectively label individual G1, S/G2/M, and G1/S-transition phase nuclei red, green and yellow, respectively, in living mammalian cells, and could therefore be useful to visualize the real-time cell cycle transitions in living murine heart. To establish a similar system for time-lapse imaging of murine heart, we first developed an ex vivo culture system, with the culture conditions determined in terms of sample state, serum concentration, and oxygen concentration. The optimal condition (slice culture, oxygen concentration 20%, serum concentration 10%) successfully mimicked physiological cardiomyocyte proliferation in vivo. Time-lapse imaging of cardiac slices from E11.5, E14.5, E18.5, and P1 Fucci-expressing transgenic mice revealed an elongated S/G2/M phase in cardiomyocytes during development. Our time-lapse imaging of murine heart revealed a gradual elongation of the S/G2/M phase during development in living cardiomyocytes.


Asunto(s)
Ciclo Celular/fisiología , Desarrollo Embrionario/fisiología , Miocitos Cardíacos/citología , Imagen de Lapso de Tiempo , Animales , Proliferación Celular , Embrión de Mamíferos , Femenino , Colorantes Fluorescentes , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Microscopía Fluorescente/métodos , Miocitos Cardíacos/fisiología , Embarazo , Técnicas de Cultivo de Tejidos , Ubiquitinación
7.
J Cardiol ; 82(2): 128-133, 2023 08.
Artículo en Inglés | MEDLINE | ID: mdl-37141938

RESUMEN

Machine learning is a subfield of artificial intelligence. The quality and versatility of machine learning have been rapidly improving and playing a critical role in many aspects of social life. This trend is also observed in the medical field. Generally, there are three main types of machine learning: supervised, unsupervised, and reinforcement learning. Each type of learning is adequately selected for the purpose and type of data. In the field of medicine, various types of information are collected and used, and research using machine learning is becoming increasingly relevant. Many clinical studies are conducted using electronic health and medical records, including in the cardiovascular area. Machine learning has also been applied in basic research. Machine learning has been widely used for several types of data analysis, such as clustering of microarray analysis and RNA sequence analysis. Machine learning is essential for genome and multi-omics analyses. This review summarizes the recent advancements in the use of machine learning in clinical applications and basic cardiovascular research.


Asunto(s)
Inteligencia Artificial , Cardiología , Humanos , Aprendizaje Automático
8.
Stem Cell Reports ; 18(6): 1274-1283, 2023 06 13.
Artículo en Inglés | MEDLINE | ID: mdl-37315521

RESUMEN

Cardiac transcription factors (TFs) directly reprogram fibroblasts into induced cardiomyocytes (iCMs), where MEF2C acts as a pioneer factor with GATA4 and TBX5 (GT). However, the generation of functional and mature iCMs is inefficient, and the molecular mechanisms underlying this process remain largely unknown. Here, we found that the overexpression of transcriptionally activated MEF2C via fusion of the powerful MYOD transactivation domain combined with GT increased the generation of beating iCMs by 30-fold. Activated MEF2C with GT generated iCMs that were transcriptionally, structurally, and functionally more mature than those generated by native MEF2C with GT. Mechanistically, activated MEF2C recruited p300 and multiple cardiogenic TFs to cardiac loci to induce chromatin remodeling. In contrast, p300 inhibition suppressed cardiac gene expression, inhibited iCM maturation, and decreased the beating iCM numbers. Splicing isoforms of MEF2C with similar transcriptional activities did not promote functional iCM generation. Thus, MEF2C/p300-mediated epigenetic remodeling promotes iCM maturation.


Asunto(s)
Ensamble y Desensamble de Cromatina , Factores de Transcripción MEF2 , Miocitos Cardíacos , Factores de Transcripción p300-CBP , Epigénesis Genética , Epigenómica , Fibroblastos , Factores de Transcripción MEF2/genética , Factores de Transcripción p300-CBP/genética
9.
Biochem Biophys Res Commun ; 425(4): 755-61, 2012 Sep 07.
Artículo en Inglés | MEDLINE | ID: mdl-22884798

RESUMEN

MicroRNAs (miRNAs) play a pivotal role during embryonic development and are required for proper organogenesis, including hematopoiesis. Recent studies suggest that, in the early mesoderm, there is an interaction between the hematopoietic and cardiac lineages. However, whether miRNAs can affect other lineages remains unknown. Therefore, we investigated whether hematopoietic miR-142-3p modulated the mesoderm formation. We report that knockdown (KD) of miR-142-3p, a hematopoietic-specific miRNA, in zebrafish resulted in loss of hematopoiesis during embryonic development. Intriguingly, we observed abnormal cardiac phenotypes and insufficiency of somitegenesis in KD-morphants. In the early developmental stage, a tiny heart, contractile dysfunction in the ventricle, cardiac arrhythmia (e.g. a 2:1 ratio of atrial:ventricular beating), and bradycardia were consistently observed. Histological examination revealed severe hypoplasia of the ventricle and disrupted muscle alignment. To determine the mechanism, we performed DNA microarray analysis. The results revealed that the expression of several mesodermal genes essential for the formation of cardiac and somatic mesoderm, such as no tail, T-box gene 16, mesoderm posterior a, one eye pinhead, and rho-associated, coiled-coil containing protein kinase (Rock2a), were increased in miR-142-3p KD-morphants. The luciferase reporter assay revealed that miR-142-3p repressed luciferase activity on the Rock2a 3'-UTR. The findings of the present study indicate that miR-142-3p plays a critical role in hematopoiesis, cardiogenesis, and somitegenesis in the early stage of mesoderm formation via regulation of Rock2a.


Asunto(s)
Corazón/embriología , Hematopoyesis , MicroARNs/metabolismo , Organogénesis , Pez Cebra/embriología , Animales , Regulación del Desarrollo de la Expresión Génica , Técnicas de Silenciamiento del Gen , Genes Reporteros , Corazón/fisiología , Luciferasas/biosíntesis , Luciferasas/genética , MicroARNs/genética , Pez Cebra/genética , Pez Cebra/metabolismo
10.
Circ Res ; 106(6): 1083-91, 2010 Apr 02.
Artículo en Inglés | MEDLINE | ID: mdl-20167925

RESUMEN

RATIONALE: The transcriptional networks guiding heart development remain poorly understood, despite the identification of several essential cardiac transcription factors. OBJECTIVE: To isolate novel cardiac transcription factors, we performed gene chip analysis and found that Zac1, a zinc finger-type transcription factor, was strongly expressed in the developing heart. This study was designed to investigate the molecular and functional role of Zac1 as a cardiac transcription factor. METHODS AND RESULTS: Zac1 was strongly expressed in the heart from cardiac crescent stages and in the looping heart showed a chamber-restricted pattern. Zac1 stimulated luciferase reporter constructs driven by ANF, BNP, or alphaMHC promoters. Strong functional synergy was seen between Zac1 and Nkx2-5 on the ANF promoter, which carries adjacent Zac1 and Nkx2-5 DNA-binding sites. Zac1 directly associated with the ANF promoter in vitro and in vivo, and Zac1 and Nkx2-5 physically associated through zinc fingers 5 and 6 in Zac1, and the homeodomain in Nkx2-5. Zac1 is a maternally imprinted gene and is the first such gene found to be involved in heart development. Homozygous and paternally derived heterozygous mice carrying an interruption in the Zac1 locus showed decreased levels of chamber and myofilament genes, increased apoptotic cells, partially penetrant lethality and morphological defects including atrial and ventricular septal defects, and thin ventricular walls. CONCLUSIONS: Zac1 plays an essential role in the cardiac gene regulatory network. Our data provide a potential mechanistic link between Zac1 in cardiogenesis and congenital heart disease manifestations associated with genetic or epigenetic defects in an imprinted gene network.


Asunto(s)
Proteínas de Ciclo Celular/genética , Regulación del Desarrollo de la Expresión Génica , Redes Reguladoras de Genes , Cardiopatías Congénitas/genética , Corazón/embriología , Factores de Transcripción/genética , Animales , Apoptosis/genética , Factor Natriurético Atrial/genética , Factor Natriurético Atrial/metabolismo , Sitios de Unión , Células COS , Proteínas de Ciclo Celular/metabolismo , Chlorocebus aethiops , Perfilación de la Expresión Génica/métodos , Genes Supresores de Tumor , Impresión Genómica , Edad Gestacional , Cardiopatías Congénitas/embriología , Cardiopatías Congénitas/metabolismo , Cardiopatías Congénitas/patología , Proteína Homeótica Nkx-2.5 , Proteínas de Homeodominio/genética , Proteínas de Homeodominio/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos ICR , Ratones Mutantes , Morfogénesis/genética , Mutación , Péptido Natriurético Tipo-C/genética , Péptido Natriurético Tipo-C/metabolismo , Análisis de Secuencia por Matrices de Oligonucleótidos , Regiones Promotoras Genéticas , Precursores de Proteínas/genética , Precursores de Proteínas/metabolismo , Ratas , Factores de Transcripción/metabolismo , Activación Transcripcional , Transfección
11.
Nat Commun ; 13(1): 5409, 2022 09 15.
Artículo en Inglés | MEDLINE | ID: mdl-36109509

RESUMEN

Failure of the right ventricle plays a critical role in any type of heart failure. However, the mechanism remains unclear, and there is no specific therapy. Here, we show that the right ventricle predominantly expresses alternative complement pathway-related genes, including Cfd and C3aR1. Complement 3 (C3)-knockout attenuates right ventricular dysfunction and fibrosis in a mouse model of right ventricular failure. C3a is produced from C3 by the C3 convertase complex, which includes the essential component complement factor D (Cfd). Cfd-knockout mice also show attenuation of right ventricular failure. Moreover, the plasma concentration of CFD correlates with the severity of right ventricular failure in patients with chronic right ventricular failure. A C3a receptor (C3aR) antagonist dramatically improves right ventricular dysfunction in mice. In summary, we demonstrate the crucial role of the C3-Cfd-C3aR axis in right ventricular failure and highlight potential therapeutic targets for right ventricular failure.


Asunto(s)
Insuficiencia Cardíaca , Disfunción Ventricular Derecha , Animales , Complemento C3/genética , Convertasas de Complemento C3-C5 , Factor D del Complemento , Insuficiencia Cardíaca/genética , Ratones , Ratones Noqueados , Remodelación Ventricular
12.
Nat Commun ; 12(1): 257, 2021 01 11.
Artículo en Inglés | MEDLINE | ID: mdl-33431893

RESUMEN

Advances in deep learning technology have enabled complex task solutions. The accuracy of image classification tasks has improved owing to the establishment of convolutional neural networks (CNN). Cellular senescence is a hallmark of ageing and is important for the pathogenesis of ageing-related diseases. Furthermore, it is a potential therapeutic target. Specific molecular markers are used to identify senescent cells. Moreover senescent cells show unique morphology, which can be identified. We develop a successful morphology-based CNN system to identify senescent cells and a quantitative scoring system to evaluate the state of endothelial cells by senescence probability output from pre-trained CNN optimised for the classification of cellular senescence, Deep Learning-Based Senescence Scoring System by Morphology (Deep-SeSMo). Deep-SeSMo correctly evaluates the effects of well-known anti-senescent reagents. We screen for drugs that control cellular senescence using a kinase inhibitor library by Deep-SeSMo-based drug screening and identify four anti-senescent drugs. RNA sequence analysis reveals that these compounds commonly suppress senescent phenotypes through inhibition of the inflammatory response pathway. Thus, morphology-based CNN system can be a powerful tool for anti-senescent drug screening.


Asunto(s)
Forma de la Célula , Senescencia Celular , Aprendizaje Profundo , Evaluación Preclínica de Medicamentos , Células Endoteliales de la Vena Umbilical Humana/citología , Células Endoteliales de la Vena Umbilical Humana/efectos de los fármacos , Células Endoteliales de la Vena Umbilical Humana/metabolismo , Humanos , Peróxido de Hidrógeno/toxicidad , Redes Neurales de la Computación , beta-Galactosidasa/metabolismo
13.
Nat Cell Biol ; 23(5): 467-475, 2021 05.
Artículo en Inglés | MEDLINE | ID: mdl-33941892

RESUMEN

Direct cardiac reprogramming of fibroblasts to cardiomyocytes presents an attractive therapeutic strategy to restore cardiac function following injury. Cardiac reprogramming was initially achieved through overexpression of the transcription factors Gata4, Mef2c and Tbx5; later, Hand2 and Akt1 were found to further enhance this process1-5. Yet, staunch epigenetic barriers severely limit the ability of these cocktails to reprogramme adult fibroblasts6,7. We undertook a screen of mammalian gene regulatory factors to discover novel regulators of cardiac reprogramming in adult fibroblasts and identified the histone reader PHF7 as the most potent activating factor8. Mechanistically, PHF7 localizes to cardiac super enhancers in fibroblasts, and through cooperation with the SWI/SNF complex, it increases chromatin accessibility and transcription factor binding at these sites. Furthermore, PHF7 recruits cardiac transcription factors to activate a positive transcriptional autoregulatory circuit in reprogramming. Importantly, PHF7 achieves efficient reprogramming in the absence of Gata4. Here, we highlight the underexplored necessity of cardiac epigenetic readers, such as PHF7, in harnessing chromatin remodelling and transcriptional complexes to overcome critical barriers to direct cardiac reprogramming.


Asunto(s)
Factor de Transcripción GATA4/metabolismo , Histonas/metabolismo , Transducción de Señal/fisiología , Ubiquitina-Proteína Ligasas/metabolismo , Animales , Reprogramación Celular , Fibroblastos/metabolismo , Factor de Transcripción GATA4/genética , Miocitos Cardíacos/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Secuencias Reguladoras de Ácidos Nucleicos/fisiología , Factores de Transcripción/metabolismo , Ubiquitina-Proteína Ligasas/genética
14.
Sci Rep ; 10(1): 21467, 2020 12 08.
Artículo en Inglés | MEDLINE | ID: mdl-33293623

RESUMEN

In deep burns, early wound closure is important for healing, and skin grafting is mainly used for wound closure. However, it is difficult to achieve early wound closure in extensive total body surface area deep burns due to the lack of donor sites. Dermal fibroblasts, responsible for dermis formation, may be lost in deep burns. However, fat layers composed of adipocytes, lying underneath the dermis, are retained even in such cases. Direct reprogramming is a novel method for directly reprograming some cells into other types by introducing specific master regulators; it has exhibited appreciable success in various fields. In this study, we aimed to assess whether the transfection of master regulators (ELF4, FOXC2, FOXO1, IRF1, PRRX1, and ZEB1) could reprogram mouse adipocytes into dermal fibroblast-like cells. Our results indicated the shrinkage of fat droplets in reprogrammed mouse adipocytes and their transformation into spindle-shaped dermal fibroblasts. Reduced expression of PPAR-2, c/EBP, aP2, and leptin, the known markers of adipocytes, in RT-PCR, and enhanced expression of anti-ER-TR7, the known anti-fibroblast marker, in immunocytochemistry, were confirmed in the reprogrammed mouse adipocytes. The dermal fibroblast-like cells, reported here, may open up a new treatment mode for enabling early closure of deep burn wounds.


Asunto(s)
Adipocitos/citología , Técnicas de Reprogramación Celular/métodos , Dermis/citología , Fibroblastos/citología , Adipocitos/metabolismo , Animales , Células Cultivadas , Dermis/metabolismo , Fibroblastos/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Transfección/métodos
15.
Cell Stem Cell ; 25(1): 69-86.e5, 2019 07 03.
Artículo en Inglés | MEDLINE | ID: mdl-31080136

RESUMEN

The cardiogenic transcription factors (TFs) Mef2c, Gata4, and Tbx5 can directly reprogram fibroblasts to induced cardiac-like myocytes (iCLMs), presenting a potential source of cells for cardiac repair. While activity of these TFs is enhanced by Hand2 and Akt1, their genomic targets and interactions during reprogramming are not well studied. We performed genome-wide analyses of cardiogenic TF binding and enhancer profiling during cardiac reprogramming. We found that these TFs synergistically activate enhancers highlighted by Mef2c binding sites and that Hand2 and Akt1 coordinately recruit other TFs to enhancer elements. Intriguingly, these enhancer landscapes collectively resemble patterns of enhancer activation during embryonic cardiogenesis. We further constructed a cardiac reprogramming gene regulatory network and found repression of EGFR signaling pathway genes. Consistently, chemical inhibition of EGFR signaling augmented reprogramming. Thus, by defining epigenetic landscapes these findings reveal synergistic transcriptional activation across a broad landscape of cardiac enhancers and key signaling pathways that govern iCLM reprogramming.


Asunto(s)
Receptores ErbB/metabolismo , Fibroblastos/fisiología , Miocitos Cardíacos/fisiología , Animales , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Células Cultivadas , Reprogramación Celular , Receptores ErbB/genética , Factor de Transcripción GATA4/genética , Redes Reguladoras de Genes , Estudio de Asociación del Genoma Completo , Factores de Transcripción MEF2/genética , Ratones , Ratones Endogámicos C57BL , Transducción de Señal , Proteínas de Dominio T Box/genética
17.
Nat Rev Cardiol ; 15(10): 585-600, 2018 10.
Artículo en Inglés | MEDLINE | ID: mdl-29872165

RESUMEN

Ischaemic heart disease is a leading cause of death worldwide. Injury to the heart is followed by loss of the damaged cardiomyocytes, which are replaced with fibrotic scar tissue. Depletion of cardiomyocytes results in decreased cardiac contraction, which leads to pathological cardiac dilatation, additional cardiomyocyte loss, and mechanical dysfunction, culminating in heart failure. This sequential reaction is defined as cardiac remodelling. Many therapies have focused on preventing the progressive process of cardiac remodelling to heart failure. However, after patients have developed end-stage heart failure, intervention is limited to heart transplantation. One of the main reasons for the dramatic injurious effect of cardiomyocyte loss is that the adult human heart has minimal regenerative capacity. In the past 2 decades, several strategies to repair the injured heart and improve heart function have been pursued, including cellular and noncellular therapies. In this Review, we discuss current therapeutic approaches for cardiac repair and regeneration, describing outcomes, limitations, and future prospects of preclinical and clinical trials of heart regeneration. Substantial progress has been made towards understanding the cellular and molecular mechanisms regulating heart regeneration, offering the potential to control cardiac remodelling and redirect the adult heart to a regenerative state.


Asunto(s)
Terapia Genética/métodos , Cardiopatías/terapia , Ventrículos Cardíacos/patología , Ventrículos Cardíacos/fisiopatología , Miocardio/patología , Regeneración , Trasplante de Células Madre/métodos , Remodelación Ventricular , Animales , Cardiopatías/genética , Cardiopatías/patología , Cardiopatías/fisiopatología , Humanos , Recuperación de la Función , Resultado del Tratamiento
18.
Stem Cell Reports ; 8(3): 548-560, 2017 03 14.
Artículo en Inglés | MEDLINE | ID: mdl-28262548

RESUMEN

Conversion of fibroblasts into functional cardiomyocytes represents a potential means of restoring cardiac function after myocardial infarction, but so far this process remains inefficient and little is known about its molecular mechanisms. Here we show that DAPT, a classical Notch inhibitor, enhances the conversion of mouse fibroblasts into induced cardiac-like myocytes by the transcription factors GATA4, HAND2, MEF2C, and TBX5. DAPT cooperates with AKT kinase to further augment this process, resulting in up to 70% conversion efficiency. Moreover, DAPT promotes the acquisition of specific cardiomyocyte features, substantially increasing calcium flux, sarcomere structure, and the number of spontaneously beating cells. Transcriptome analysis shows that DAPT induces genetic programs related to muscle development, differentiation, and excitation-contraction coupling. Mechanistically, DAPT increases binding of the transcription factor MEF2C to the promoter regions of cardiac structural genes. These findings provide mechanistic insights into the reprogramming process and may have important implications for cardiac regeneration therapies.


Asunto(s)
Reprogramación Celular/genética , Diaminas/farmacología , Factores de Transcripción MEF2/metabolismo , Miocitos Cardíacos/citología , Miocitos Cardíacos/metabolismo , Receptores Notch/metabolismo , Transducción de Señal , Tiazoles/farmacología , Activación Transcripcional , Animales , Señalización del Calcio/efectos de los fármacos , Diferenciación Celular , Reprogramación Celular/efectos de los fármacos , Fenómenos Electrofisiológicos/efectos de los fármacos , Perfilación de la Expresión Génica , Regulación del Desarrollo de la Expresión Génica , Ratones , Miocitos Cardíacos/efectos de los fármacos , Proteínas Proto-Oncogénicas c-akt/genética , Proteínas Proto-Oncogénicas c-akt/metabolismo , Sarcómeros/efectos de los fármacos , Sarcómeros/metabolismo , Transcriptoma
19.
FEBS Lett ; 591(18): 2879-2889, 2017 09.
Artículo en Inglés | MEDLINE | ID: mdl-28805244

RESUMEN

The expression of pluripotency genes fluctuates in a population of embryonic stem (ES) cells and the fluctuations in the expression of some pluripotency genes correlate. However, no correlation in the fluctuation of Pou5f1, Zfp42, and Nanog expression was observed in ES cells. Correlation between Pou5f1 and Zfp42 fluctuations was demonstrated in ES cells containing a knockout in the NuRD component Mbd3. ES cells containing a triple knockout in the DNA methyltransferases Dnmt1, Dnmt3a, and Dnmt3b showed correlation between the fluctuation of Pou5f1, Zfp42, and Nanog gene expression. We suggest that an epigenetic barrier is key to preventing the propagation of fluctuating pluripotency gene expression in ES cells.


Asunto(s)
Células Madre Embrionarias/metabolismo , Animales , Epigenómica , Expresión Génica/genética , Ratones , Proteína Homeótica Nanog/genética , Factor 3 de Transcripción de Unión a Octámeros/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Factores de Transcripción SOXB1/genética
20.
Sci Rep ; 7: 44312, 2017 03 14.
Artículo en Inglés | MEDLINE | ID: mdl-28290476

RESUMEN

Alteration of the nuclear Ca2+ transient is an early event in cardiac remodeling. Regulation of the nuclear Ca2+ transient is partly independent of the cytosolic Ca2+ transient in cardiomyocytes. One nuclear membrane protein, emerin, is encoded by EMD, and an EMD mutation causes Emery-Dreifuss muscular dystrophy (EDMD). It remains unclear whether emerin is involved in nuclear Ca2+ homeostasis. The aim of this study is to elucidate the role of emerin in rat cardiomyocytes by means of hypertrophic stimuli and in EDMD induced pluripotent stem (iPS) cell-derived cardiomyocytes in terms of nuclear structure and the Ca2+ transient. The cardiac hypertrophic stimuli increased the nuclear area, decreased nuclear invagination, and increased the half-decay time of the nuclear Ca2+ transient in cardiomyocytes. Emd knockdown cardiomyocytes showed similar properties after hypertrophic stimuli. The EDMD-iPS cell-derived cardiomyocytes showed increased nuclear area, decreased nuclear invagination, and increased half-decay time of the nuclear Ca2+ transient. An autopsied heart from a patient with EDMD also showed increased nuclear area and decreased nuclear invagination. These data suggest that Emerin plays a crucial role in nuclear structure and in the nuclear Ca2+ transient. Thus, emerin and the nuclear Ca2+ transient are possible therapeutic targets in heart failure and EDMD.


Asunto(s)
Calcio/metabolismo , Cardiomegalia/genética , Proteínas de la Membrana/genética , Distrofia Muscular de Emery-Dreifuss/genética , Miocardio/metabolismo , Miocitos Cardíacos/metabolismo , Membrana Nuclear/metabolismo , Proteínas Nucleares/genética , Transporte Activo de Núcleo Celular/efectos de los fármacos , Angiotensina II/farmacología , Compuestos de Anilina/química , Animales , Remodelación Atrial , Cardiomegalia/metabolismo , Cardiomegalia/patología , Citoplasma/efectos de los fármacos , Citoplasma/metabolismo , Citoplasma/ultraestructura , Modelos Animales de Enfermedad , Endotelina-1/farmacología , Colorantes Fluorescentes/química , Regulación de la Expresión Génica , Compuestos Heterocíclicos con 3 Anillos/química , Humanos , Proteínas de la Membrana/antagonistas & inhibidores , Proteínas de la Membrana/metabolismo , Distrofia Muscular de Emery-Dreifuss/metabolismo , Distrofia Muscular de Emery-Dreifuss/patología , Miocardio/patología , Miocitos Cardíacos/efectos de los fármacos , Miocitos Cardíacos/ultraestructura , Membrana Nuclear/efectos de los fármacos , Membrana Nuclear/ultraestructura , Proteínas Nucleares/antagonistas & inhibidores , Proteínas Nucleares/metabolismo , Fenilefrina/farmacología , Cultivo Primario de Células , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/metabolismo , Ratas , Ratas Sprague-Dawley , Remodelación Ventricular , Xantenos/química
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA