Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 41
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Development ; 148(14)2021 07 15.
Artículo en Inglés | MEDLINE | ID: mdl-34897388

RESUMEN

Terminally differentiated cells are generally thought to have arrived at their final form and function. Many terminally differentiated cell types are polyploid, i.e. they have multiple copies of the normally diploid genome. Mammalian heart muscle cells, termed cardiomyocytes, are one such example of polyploid cells. Terminally differentiated cardiomyocytes are bi- or multi-nucleated, or have polyploid nuclei. Recent mechanistic studies of polyploid cardiomyocytes indicate that they can limit cellular proliferation and, hence, heart regeneration. In this short Spotlight, we present the mechanisms generating bi- and multi-nucleated cardiomyocytes, and the mechanisms generating polyploid nuclei. Our aim is to develop hypotheses about how these mechanisms might relate to cardiomyocyte proliferation and cardiac regeneration. We also discuss how these new findings could be applied to advance cardiac regeneration research, and how they relate to studies of other polyploid cells, such as cancer cells.


Asunto(s)
Corazón/fisiología , Miocitos Cardíacos/metabolismo , Poliploidía , Regeneración , Animales , Humanos
2.
Cell ; 138(2): 257-70, 2009 Jul 23.
Artículo en Inglés | MEDLINE | ID: mdl-19632177

RESUMEN

Many organs rely on undifferentiated stem and progenitor cells for tissue regeneration. Whether differentiated cells themselves can contribute to cell replacement and tissue regeneration is a controversial question. Here, we show that differentiated heart muscle cells, cardiomyocytes, can be induced to proliferate and regenerate. We identify an underlying molecular mechanism for controlling this process that involves the growth factor neuregulin1 (NRG1) and its tyrosine kinase receptor, ErbB4. NRG1 induces mononucleated, but not binucleated, cardiomyocytes to divide. In vivo, genetic inactivation of ErbB4 reduces cardiomyocyte proliferation, whereas increasing ErbB4 expression enhances it. Injecting NRG1 in adult mice induces cardiomyocyte cell-cycle activity and promotes myocardial regeneration, leading to improved function after myocardial infarction. Undifferentiated progenitor cells did not contribute to NRG1-induced cardiomyocyte proliferation. Thus, increasing the activity of the NRG1/ErbB4 signaling pathway may provide a molecular strategy to promote myocardial regeneration.


Asunto(s)
Proliferación Celular , Receptores ErbB/metabolismo , Miocardio/citología , Miocitos Cardíacos/citología , Neurregulina-1/metabolismo , Transducción de Señal , Animales , Ciclo Celular , Citocinesis , Lesiones Cardíacas/terapia , Humanos , Masculino , Ratones , Infarto del Miocardio/terapia , Ratas , Ratas Wistar , Receptor ErbB-4 , Células Madre
3.
PLoS Genet ; 17(11): e1009890, 2021 11.
Artículo en Inglés | MEDLINE | ID: mdl-34723970

RESUMEN

In contrast to mammals, the zebrafish maintains its cardiomyocyte proliferation capacity throughout adulthood. However, neither the molecular mechanisms that orchestrate the proliferation of cardiomyocytes during developmental heart growth nor in the context of regeneration in the adult are sufficiently defined yet. We identified in a forward genetic N-ethyl-N-nitrosourea (ENU) mutagenesis screen the recessive, embryonic-lethal zebrafish mutant baldrian (bal), which shows severely impaired developmental heart growth due to diminished cardiomyocyte proliferation. By positional cloning, we identified a missense mutation in the zebrafish histone deacetylase 1 (hdac1) gene leading to severe protein instability and the loss of Hdac1 function in vivo. Hdac1 inhibition significantly reduces cardiomyocyte proliferation, indicating a role of Hdac1 during developmental heart growth in zebrafish. To evaluate whether developmental and regenerative Hdac1-associated mechanisms of cardiomyocyte proliferation are conserved, we analyzed regenerative cardiomyocyte proliferation after Hdac1 inhibition at the wound border zone in cryoinjured adult zebrafish hearts and we found that Hdac1 is also essential to orchestrate regenerative cardiomyocyte proliferation in the adult vertebrate heart. In summary, our findings suggest an important and conserved role of Histone deacetylase 1 (Hdac1) in developmental and adult regenerative cardiomyocyte proliferation in the vertebrate heart.


Asunto(s)
Corazón/fisiología , Histona Desacetilasa 1/metabolismo , Miocitos Cardíacos/citología , Regeneración/fisiología , Proteínas de Pez Cebra/metabolismo , Pez Cebra/embriología , Animales , Proliferación Celular
4.
Am J Physiol Heart Circ Physiol ; 322(4): H579-H596, 2022 04 01.
Artículo en Inglés | MEDLINE | ID: mdl-35179974

RESUMEN

During the past two decades, the field of mammalian myocardial regeneration has grown dramatically, and with this expanded interest comes increasing claims of experimental manipulations that mediate bona fide proliferation of cardiomyocytes. Too often, however, insufficient evidence or improper controls are provided to support claims that cardiomyocytes have definitively proliferated, a process that should be strictly defined as the generation of two de novo functional cardiomyocytes from one original cardiomyocyte. Throughout the literature, one finds inconsistent levels of experimental rigor applied, and frequently the specific data supplied as evidence of cardiomyocyte proliferation simply indicate cell-cycle activation or DNA synthesis, which do not necessarily lead to the generation of new cardiomyocytes. In this review, we highlight potential problems and limitations faced when characterizing cardiomyocyte proliferation in the mammalian heart, and summarize tools and experimental standards, which should be used to support claims of proliferation-based remuscularization. In the end, definitive establishment of de novo cardiomyogenesis can be difficult to prove; therefore, rigorous experimental strategies should be used for such claims.


Asunto(s)
Miocitos Cardíacos , Regeneración , Animales , Ciclo Celular , Proliferación Celular , Corazón/fisiología , Mamíferos , Miocitos Cardíacos/fisiología
5.
Development ; 145(5)2018 03 06.
Artículo en Inglés | MEDLINE | ID: mdl-29444893

RESUMEN

Zebrafish regenerate cardiac tissue through proliferation of pre-existing cardiomyocytes and neovascularization. Secreted growth factors such as FGFs, IGF, PDGFs and Neuregulin play essential roles in stimulating cardiomyocyte proliferation. These factors activate the Ras/MAPK pathway, which is tightly controlled by the feedback attenuator Dual specificity phosphatase 6 (Dusp6), an ERK phosphatase. Here, we show that suppressing Dusp6 function enhances cardiac regeneration. Inactivation of Dusp6 by small molecules or by gene inactivation increased cardiomyocyte proliferation, coronary angiogenesis, and reduced fibrosis after ventricular resection. Inhibition of Erbb or PDGF receptor signaling suppressed cardiac regeneration in wild-type zebrafish, but had a milder effect on regeneration in dusp6 mutants. Moreover, in rat primary cardiomyocytes, NRG1-stimulated proliferation can be enhanced upon chemical inhibition of Dusp6 with BCI. Our results suggest that Dusp6 attenuates Ras/MAPK signaling during regeneration and that suppressing Dusp6 can enhance cardiac repair.


Asunto(s)
Fosfatasa 6 de Especificidad Dual/fisiología , Corazón/fisiología , Sistema de Señalización de MAP Quinasas/fisiología , Proteínas Proto-Oncogénicas p21(ras)/metabolismo , Regeneración/genética , Pez Cebra/fisiología , Animales , Animales Modificados Genéticamente , Proliferación Celular/genética , Regulación hacia Abajo/genética , Miocitos Cardíacos/fisiología , Transducción de Señal/genética , Proteínas de Pez Cebra/fisiología
6.
Curr Cardiol Rep ; 19(2): 13, 2017 02.
Artículo en Inglés | MEDLINE | ID: mdl-28185170

RESUMEN

PURPOSE OF REVIEW: Congenital heart disease is the most common birth defect and acquired heart disease is the leading cause of death in adults. Understanding the mechanisms that drive cardiomyocyte proliferation and differentiation has the potential to advance the understanding and potentially the treatment of different cardiac pathologies, ranging from myopathies and heart failure to myocardial infarction. This review focuses on studies aimed at elucidating signal transduction pathways and molecular mechanisms that promote proliferation, differentiation, and regeneration of differentiated heart muscle cells, cardiomyocytes. RECENT FINDINGS: There is now significant evidence that demonstrates cardiomyocytes continue to proliferate into adulthood. Potential regulators have been identified, including cell cycle regulators, extracellular ligands such as neuregulin, epigenetic targets, reactive oxygen species, and microRNA. The necessary steps should involve validating and applying the new knowledge about cardiomyocyte regeneration towards the development of therapeutic targets for patients. This will be facilitated by the application of standardized pre-clinical models to study cardiomyocyte regeneration.


Asunto(s)
Diferenciación Celular , Proliferación Celular , Miocitos Cardíacos/citología , Regeneración , Enfermedades Cardiovasculares/fisiopatología , Ciclo Celular , Humanos , Transducción de Señal
7.
Circ Res ; 124(12): 1725-1726, 2019 06 07.
Artículo en Inglés | MEDLINE | ID: mdl-31170042
8.
Proc Natl Acad Sci U S A ; 110(4): 1446-51, 2013 Jan 22.
Artículo en Inglés | MEDLINE | ID: mdl-23302686

RESUMEN

The human heart is believed to grow by enlargement but not proliferation of cardiomyocytes (heart muscle cells) during postnatal development. However, recent studies have shown that cardiomyocyte proliferation is a mechanism of cardiac growth and regeneration in animals. Combined with evidence for cardiomyocyte turnover in adult humans, this suggests that cardiomyocyte proliferation may play an unrecognized role during the period of developmental heart growth between birth and adolescence. We tested this hypothesis by examining the cellular growth mechanisms of the left ventricle on a set of healthy hearts from humans aged 0-59 y (n = 36). The percentages of cardiomyocytes in mitosis and cytokinesis were highest in infants, decreasing to low levels by 20 y. Although cardiomyocyte mitosis was detectable throughout life, cardiomyocyte cytokinesis was not evident after 20 y. Between the first year and 20 y of life, the number of cardiomyocytes in the left ventricle increased 3.4-fold, which was consistent with our predictions based on measured cardiomyocyte cell cycle activity. Our findings show that cardiomyocyte proliferation contributes to developmental heart growth in young humans. This suggests that children and adolescents may be able to regenerate myocardium, that abnormal cardiomyocyte proliferation may be involved in myocardial diseases that affect this population, and that these diseases might be treatable through stimulation of cardiomyocyte proliferation.


Asunto(s)
Corazón/crecimiento & desarrollo , Miocitos Cardíacos/citología , Adolescente , Adulto , Ciclo Celular , Aumento de la Célula , Proliferación Celular , Niño , Preescolar , Femenino , Fibrosis , Corazón/fisiología , Humanos , Lactante , Recién Nacido , Masculino , Persona de Mediana Edad , Miocardio/citología , Miocardio/patología , Miocitos Cardíacos/patología , Ploidias , Regeneración , Adulto Joven
9.
Naturwissenschaften ; 102(7-8): 43, 2015 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-26185112

RESUMEN

When disturbed, adults of the Death's-head hawkmoth (Lepidoptera, Sphingidae: Acherontia atropos) produce short squeaks by drawing in and deflating air into and out of the pharynx as a defence mechanism. We took a new look at Prell's hypothesis of a two-phase mechanism by providing new insights into the functional morphology behind the pharyngeal sound production of this species. First, we compared the head anatomy of A. atropos with another sphingid species, Manduca sexta, by using micro-computed tomography (CT) and 3D reconstruction methods. Despite differences in feeding behaviour and capability of sound production in the two species, the musculature in the head is surprisingly similar. However, A. atropos has a much shorter proboscis and a modified epipharynx with a distinct sclerotised lobe projecting into the opening of the pharynx. Second, we observed the sound production in vivo with X-ray videography, mammography CT and high-speed videography. Third, we analysed acoustic pressure over time and spectral frequency composition of six A. atropos specimens, both intact and with a removed proboscis. Single squeaks of A. atropos last for ca. 200 ms and consist of an inflation phase, a short pause and a deflation phase. The inflation phase is characterised by a burst of ca. 50 pulses with decreasing pulse frequency and a major frequency peak at ca. 8 kHz, followed by harmonics ranging up to more than 60 kHz. The deflation phase is characterised by a less clear acoustic pattern, a lower amplitude and more pronounced peaks in the same frequency range. The removal of the proboscis resulted in a significantly shortened squeak, a lower acoustic pressure level and a slightly more limited frequency spectrum. We hypothesise that the uptake of viscous honey facilitated the evolution of an efficient valve at the opening of the pharynx (i.e. a modified epipharynx), and that sound production could relatively easily have evolved based on this morphological pre-adaptation.


Asunto(s)
Comunicación Animal , Mariposas Nocturnas/anatomía & histología , Animales , Manduca/anatomía & histología , Tomografía Computarizada por Rayos X , Grabación en Video
10.
Nat Med ; 13(8): 962-9, 2007 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-17632525

RESUMEN

Adult mammalian hearts respond to injury with scar formation and not with cardiomyocyte proliferation, the cellular basis of regeneration. Although cardiogenic progenitor cells may maintain myocardial turnover, they do not give rise to a robust regenerative response. Here we show that extracellular periostin induced reentry of differentiated mammalian cardiomyocytes into the cell cycle. Periostin stimulated mononucleated cardiomyocytes to go through the full mitotic cell cycle. Periostin activated alphaV, beta1, beta3 and beta5 integrins located in the cardiomyocyte cell membrane. Activation of phosphatidylinositol-3-OH kinase was required for periostin-induced reentry of cardiomyocytes into the cell cycle and was sufficient for cell-cycle reentry in the absence of periostin. After myocardial infarction, periostin-induced cardiomyocyte cell-cycle reentry and mitosis were associated with improved ventricular remodeling and myocardial function, reduced fibrosis and infarct size, and increased angiogenesis. Thus, periostin and the pathway that it regulates may provide a target for innovative strategies to treat heart failure.


Asunto(s)
Moléculas de Adhesión Celular/farmacología , Diferenciación Celular , Miocitos Cardíacos/citología , Miocitos Cardíacos/efectos de los fármacos , Regeneración/efectos de los fármacos , Animales , Moléculas de Adhesión Celular/uso terapéutico , Diferenciación Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Células Cultivadas , ADN/biosíntesis , Fibrosis/tratamiento farmacológico , Fibrosis/patología , Humanos , Hipertrofia/tratamiento farmacológico , Hipertrofia/patología , Integrinas/metabolismo , Masculino , Infarto del Miocardio/tratamiento farmacológico , Infarto del Miocardio/fisiopatología , Miocitos Cardíacos/fisiología , Fosfatidilinositol 3-Quinasas/metabolismo , Ratas , Ratas Wistar
11.
PLoS One ; 19(1): e0295651, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38271331

RESUMEN

BACKGROUND: We have developed a new clinical research approach for the quantification of cellular proliferation in human infants to address unanswered questions about tissue renewal and regeneration. The approach consists of oral 15N-thymidine administration to label cells in S-phase, followed by Multi-isotope Imaging Mass Spectrometry for detection of the incorporated label in cell nuclei. To establish the approach, we performed an observational study to examine uptake and elimination of 15N-thymidine. We compared at-home label administration with in-hospital administration in infants with tetralogy of Fallot, a form of congenital heart disease, and infants with heart failure. METHODS: We examined urine samples from 18 infants who received 15N-thymidine (50 mg/kg body weight) by mouth for five consecutive days. We used Isotope Ratio Mass Spectrometry to determine enrichment of 15N relative to 14N (%) in urine. RESULTS/FINDINGS: 15N-thymidine dose administration produced periodic rises of 15N enrichment in urine. Infants with tetralogy of Fallot had a 3.2-fold increase and infants with heart failure had a 4.3-fold increase in mean peak 15N enrichment over baseline. The mean 15N enrichment was not statistically different between the two patient populations (p = 0.103). The time to peak 15N enrichment in tetralogy of Fallot infants was 6.3 ± 1 hr and in infants with heart failure 7.5 ± 2 hr (mean ± SEM). The duration of significant 15N enrichment after a dose was 18.5 ± 1.7 hr in tetralogy of Fallot and in heart failure 18.2 ± 1.8 hr (mean ± SEM). The time to peak enrichment and duration of enrichment were also not statistically different (p = 0.617 and p = 0.887). CONCLUSIONS: The presented results support two conclusions of significance for future applications: (1) Demonstration that 15N-thymidine label administration at home is equivalent to in-hospital administration. (2) Two different types of heart disease show no differences in 15N-thymidine absorption and elimination. This enables the comparative analysis of cellular proliferation between different types of heart disease.


Asunto(s)
Insuficiencia Cardíaca , Tetralogía de Fallot , Humanos , Tetralogía de Fallot/tratamiento farmacológico , Isótopos de Nitrógeno , Administración Oral , Boca , Insuficiencia Cardíaca/tratamiento farmacológico
12.
STAR Protoc ; 4(3): 102552, 2023 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-37651236

RESUMEN

Nuclear pore complexes are pathways for nuclear-cytoplasmic communication that participate in chromatin organization. Here, we present a protocol to image and quantify the number of nuclear pore complexes in cells. We describe steps for cell plating and culture, immunofluorescence detection, and confocal microscopy visualization of nuclear pore complexes. We then detail quantification and 3D data analysis. This protocol utilizes digital thresholding under human supervision for quantification of nuclear pore complexes. For complete details on the use and execution of this protocol, please refer to Han et al.1.


Asunto(s)
Análisis de Datos , Poro Nuclear , Humanos , Citoplasma , Citosol , Microscopía Confocal
13.
Nucleus ; 14(1): 2246310, 2023 12.
Artículo en Inglés | MEDLINE | ID: mdl-37606283

RESUMEN

In adult mammals, many heart muscle cells (cardiomyocytes) are polyploid, do not proliferate (post-mitotic), and, consequently, cannot contribute to heart regeneration. In contrast, fetal and neonatal heart muscle cells are diploid, proliferate, and contribute to heart regeneration. We have identified interdependent changes of the nuclear lamina, nuclear pore complexes, and DNA-content (ploidy) in heart muscle cell maturation. These results offer new perspectives on how cells alter their nuclear transport and, with that, their gene regulation in response to extracellular signals. We present how changes of the nuclear lamina alter nuclear pore complexes in heart muscle cells. The consequences of these changes for cellular regeneration and stress response in the heart are discussed.


Asunto(s)
Lámina Nuclear , Poro Nuclear , Animales , Ploidias , Diferenciación Celular , Laminas , Mamíferos
14.
Nat Metab ; 5(12): 2184-2205, 2023 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-37996701

RESUMEN

Barth syndrome (BTHS) is a life-threatening genetic disorder with unknown pathogenicity caused by mutations in TAFAZZIN (TAZ) that affect remodeling of mitochondrial cardiolipin (CL). TAZ deficiency leads to accumulation of mono-lyso-CL (MLCL), which forms a peroxidase complex with cytochrome c (cyt c) capable of oxidizing polyunsaturated fatty acid-containing lipids. We hypothesized that accumulation of MLCL facilitates formation of anomalous MLCL-cyt c peroxidase complexes and peroxidation of polyunsaturated fatty acid phospholipids as the primary BTHS pathogenic mechanism. Using genetic, biochemical/biophysical, redox lipidomic and computational approaches, we reveal mechanisms of peroxidase-competent MLCL-cyt c complexation and increased phospholipid peroxidation in different TAZ-deficient cells and animal models and in pre-transplant biopsies from hearts of patients with BTHS. A specific mitochondria-targeted anti-peroxidase agent inhibited MLCL-cyt c peroxidase activity, prevented phospholipid peroxidation, improved mitochondrial respiration of TAZ-deficient C2C12 myoblasts and restored exercise endurance in a BTHS Drosophila model. Targeting MLCL-cyt c peroxidase offers therapeutic approaches to BTHS treatment.


Asunto(s)
Síndrome de Barth , Animales , Humanos , Síndrome de Barth/genética , Síndrome de Barth/patología , Citocromos c , Fosfolípidos , Cardiolipinas , Ácidos Grasos Insaturados , Peroxidasas
15.
Am J Physiol Heart Circ Physiol ; 302(11): H2139-47, 2012 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-22427524

RESUMEN

The signaling complex consisting of the growth factor neuregulin-1 (NRG1) and its tyrosine kinase receptors ErbB2 and ErbB4 has a critical role in cardiac development and homeostasis of the structure and function of the adult heart. Recent research results suggest that targeting this signaling complex may provide a viable strategy for treating heart failure. Clinical trials are currently evaluating the effectiveness and safety of intravenous administration of recombinant NRG1 formulations in heart failure patients. Endogenous as well as administered NRG1 has multiple possible activities in the adult heart, but how these are related is unknown. It has recently been demonstrated that NRG1 administration can stimulate proliferation of cardiomyocytes, which may contribute to repair failing hearts. This review summarizes the current knowledge of how NRG1 and its receptors control cardiac physiology and biology, with special emphasis on its role in cardiomyocyte proliferation during myocardial growth and regeneration.


Asunto(s)
Proliferación Celular , Receptores ErbB/fisiología , Miocitos Cardíacos/fisiología , Neurregulinas/fisiología , Receptor ErbB-2/fisiología , Transducción de Señal/fisiología , Animales , Insuficiencia Cardíaca/tratamiento farmacológico , Humanos , Ratones , Modelos Animales , Miocitos Cardíacos/citología , Neurregulina-1/uso terapéutico , Neurregulinas/uso terapéutico , Ratas , Receptor ErbB-4 , Proteínas Recombinantes/uso terapéutico
16.
Dev Cell ; 57(20): 2397-2411.e9, 2022 10 24.
Artículo en Inglés | MEDLINE | ID: mdl-36283391

RESUMEN

Nuclear pores are essential for nuclear-cytoplasmic transport. Whether and how cells change nuclear pores to alter nuclear transport and cellular function is unknown. Here, we show that rat heart muscle cells (cardiomyocytes) undergo a 63% decrease in nuclear pore numbers during maturation, and this changes their responses to extracellular signals. The maturation-associated decline in nuclear pore numbers is associated with lower nuclear import of signaling proteins such as mitogen-activated protein kinase (MAPK). Experimental reduction of nuclear pore numbers decreased nuclear import of signaling proteins, resulting in decreased expression of immediate-early genes. In a mouse model of high blood pressure, reduction of nuclear pore numbers improved adverse heart remodeling and reduced progression to lethal heart failure. The decrease in nuclear pore numbers in cardiomyocyte maturation and resulting functional changes demonstrate how terminally differentiated cells permanently alter their handling of information flux across the nuclear envelope and, with that, their behavior.


Asunto(s)
Membrana Nuclear , Poro Nuclear , Ratones , Ratas , Animales , Poro Nuclear/metabolismo , Transporte Activo de Núcleo Celular , Membrana Nuclear/metabolismo , Proteínas de Complejo Poro Nuclear/genética , Proteínas de Complejo Poro Nuclear/metabolismo , Proteínas Quinasas Activadas por Mitógenos/metabolismo
19.
Methods Mol Biol ; 2158: 125-139, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-32857370

RESUMEN

Failure to regenerate myocardium after injury is a major cause of mortality and morbidity in humans. Direct differentiation of human induced pluripotent stem cells (iPSCs) into cardiomyocytes provides an invaluable resource to pursue cardiac regeneration based on cellular transplantation. Beyond the potential for clinical therapies, iPSC technology also enables the generation of cardiomyocytes to recapitulate patient-specific phenotypes, thus presenting a powerful in vitro cell-based model to understand disease pathology and guide precision medicine. Here, we describe protocols for reprogramming of human dermal fibroblasts and blood cells into iPSCs using the non-integrative Sendai virus system and for the monolayer differentiation of iPSCs to cardiomyocytes using chemically defined media.


Asunto(s)
Diferenciación Celular , Reprogramación Celular , Dermis/citología , Fibroblastos/citología , Células Madre Pluripotentes Inducidas/citología , Miocitos Cardíacos/citología , Regeneración , Humanos
20.
Methods Mol Biol ; 2158: 199-210, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-32857375

RESUMEN

Procurement and characterization of intact human cells are essential for studies in regenerative medicine and translational medical research. The selection of the currently available approaches to isolate intact cells depends on the age of the hearts. To isolate cardiomyocytes from the fetal or neonatal myocardium, the myocardium can be minced into small tissue blocks followed by enzyme incubation. However, the fetal and neonatal cardiomyocytes are very soft and the morphology changes from long rod or spindle shape to spheres after isolation. Because of the dense packing of the cardiomyocytes and the strong cell-cell connection in adult myocardium, it is difficult to isolate the cardiomyocytes from adult myocardium by enzyme incubation only. A perfusion method is necessary to deliver the enzyme solution to the deep layers of the myocardium. However, intact hearts, which are very rare, are required for the perfusion method. Therefore, lacking methods to efficiently isolate cardiomyocytes from myocardium of various ages builds a barrier between basic research and clinical studies. Here, we describe a method for the isolation of intact cardiomyocytes from fresh or frozen human myocardium or fresh mouse hearts and the quantification of multinucleation, cardiomyocyte size, cell cycle activity, and total cardiomyocyte count per heart. We generalize this fixation-digestion method by isolating cells from a variety of mouse organs, including the liver, lung, and thymus.


Asunto(s)
Técnicas de Cultivo de Célula/métodos , Separación Celular/métodos , Corazón/crecimiento & desarrollo , Imagen Molecular/métodos , Miocardio , Miocitos Cardíacos/citología , Animales , Células Cultivadas , Humanos , Ratones , Miocitos Cardíacos/fisiología , Perfusión
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA