Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 99
Filtrar
Más filtros

Bases de datos
Tipo del documento
Intervalo de año de publicación
2.
Am J Hum Genet ; 102(3): 468-479, 2018 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-29429572

RESUMEN

Variants affecting the function of different subunits of the BAF chromatin-remodelling complex lead to various neurodevelopmental syndromes, including Coffin-Siris syndrome. Furthermore, variants in proteins containing PHD fingers, motifs recognizing specific histone tail modifications, have been associated with several neurological and developmental-delay disorders. Here, we report eight heterozygous de novo variants (one frameshift, two splice site, and five missense) in the gene encoding the BAF complex subunit double plant homeodomain finger 2 (DPF2). Affected individuals share common clinical features described in individuals with Coffin-Siris syndrome, including coarse facial features, global developmental delay, intellectual disability, speech impairment, and hypoplasia of fingernails and toenails. All variants occur within the highly conserved PHD1 and PHD2 motifs. Moreover, missense variants are situated close to zinc binding sites and are predicted to disrupt these sites. Pull-down assays of recombinant proteins and histone peptides revealed that a subset of the identified missense variants abolish or impaire DPF2 binding to unmodified and modified H3 histone tails. These results suggest an impairment of PHD finger structural integrity and cohesion and most likely an aberrant recognition of histone modifications. Furthermore, the overexpression of these variants in HEK293 and COS7 cell lines was associated with the formation of nuclear aggregates and the recruitment of both wild-type DPF2 and BRG1 to these aggregates. Expression analysis of truncating variants found in the affected individuals indicated that the aberrant transcripts escape nonsense-mediated decay. Altogether, we provide compelling evidence that de novo variants in DPF2 cause Coffin-Siris syndrome and propose a dominant-negative mechanism of pathogenicity.


Asunto(s)
Anomalías Múltiples/genética , Proteínas de Unión al ADN/genética , Cara/anomalías , Deformidades Congénitas de la Mano/genética , Discapacidad Intelectual/genética , Micrognatismo/genética , Mutación/genética , Cuello/anomalías , Subunidades de Proteína/genética , Adolescente , Secuencia de Aminoácidos , Animales , Células COS , Niño , Preescolar , Chlorocebus aethiops , Proteínas de Unión al ADN/química , Facies , Femenino , Células HEK293 , Histonas/metabolismo , Humanos , Masculino , Fenotipo , Factores de Transcripción
3.
Biol Chem ; 402(8): 953-972, 2021 07 27.
Artículo en Inglés | MEDLINE | ID: mdl-33951759

RESUMEN

In order to take advantage of the continuously increasing number of transcriptome studies, it is important to develop strategies that integrate multiple expression datasets addressing the same biological question to allow a robust analysis. Here, we propose a meta-analysis framework that integrates enriched pathways identified through the Gene Set Enrichment Analysis (GSEA) approach and calculates for each meta-pathway an empirical p-value. Validation of our approach on benchmark datasets showed comparable or even better performance than existing methods and an increase in robustness with increasing number of integrated datasets. We then applied the meta-analysis framework to 15 functional genomics datasets of physiological and pathological cardiac hypertrophy. Within these datasets we grouped expression sets measured at time points that represent the same hallmarks of heart tissue remodeling ('aggregated time points') and performed meta-analysis on the expression sets assigned to each aggregated time point. To facilitate biological interpretation, results were visualized as gene set enrichment networks. Here, our meta-analysis framework identified well-known biological mechanisms associated with pathological cardiac hypertrophy (e.g., cardiomyocyte apoptosis, cardiac contractile dysfunction, and alteration in energy metabolism). In addition, results highlighted novel, potentially cardioprotective mechanisms in physiological cardiac hypertrophy involving the down-regulation of immune cell response, which are worth further investigation.


Asunto(s)
Genómica , Transcriptoma , Cardiomegalia , Perfilación de la Expresión Génica , Análisis de Secuencia por Matrices de Oligonucleótidos
4.
Proc Natl Acad Sci U S A ; 114(30): 8029-8034, 2017 07 25.
Artículo en Inglés | MEDLINE | ID: mdl-28698371

RESUMEN

GAS2L3 is a recently identified cytoskeleton-associated protein that interacts with actin filaments and tubulin. The in vivo function of GAS2L3 in mammals remains unknown. Here, we show that mice deficient in GAS2L3 die shortly after birth because of heart failure. Mammalian cardiomyocytes lose the ability to proliferate shortly after birth, and further increase in cardiac mass is achieved by hypertrophy. The proliferation arrest of cardiomyocytes is accompanied by binucleation through incomplete cytokinesis. We observed that GAS2L3 deficiency leads to inhibition of cardiomyocyte proliferation and to cardiomyocyte hypertrophy during embryonic development. Cardiomyocyte-specific deletion of GAS2L3 confirmed that the phenotype results from the loss of GAS2L3 in cardiomyocytes. Cardiomyocytes from Gas2l3-deficient mice exhibit increased expression of a p53-transcriptional program including the cell cycle inhibitor p21. Furthermore, loss of GAS2L3 results in premature binucleation of cardiomyocytes accompanied by unresolved midbody structures. Together these results suggest that GAS2L3 plays a specific role in cardiomyocyte cytokinesis and proliferation during heart development.


Asunto(s)
Cardiomiopatía Dilatada/genética , Citocinesis , Proteínas del Citoesqueleto/fisiología , Miocitos Cardíacos/fisiología , Animales , Cardiomiopatía Dilatada/patología , Inhibidor p21 de las Quinasas Dependientes de la Ciclina/metabolismo , Citocinesis/genética , Proteínas del Citoesqueleto/genética , Fibrosis , Eliminación de Gen , Regulación de la Expresión Génica , Ratones , Ratones Noqueados , Miocardio/patología , Proteína p53 Supresora de Tumor/metabolismo
5.
J Mol Cell Cardiol ; 134: 69-73, 2019 09.
Artículo en Inglés | MEDLINE | ID: mdl-31301302

RESUMEN

BACKGROUND: The majority of adult human, mouse and rat cardiomyocytes is not diploid mononucleated. Nevertheless, the current literature on heart regeneration based on cardiomyocyte proliferation focuses mainly on the proliferation capacity of diploid mononucleated cardiomyocytes, instead of the more abundant mononucleated polyploid or binucleated cardiomyocytes. Here, we aimed at a better understanding of the process of mitosis and cell division in postnatal binucleated cardiomyocytes. METHODS AND RESULTS: Postnatal rat binucleated cardiomyocytes were stimulated to re-enter the cell cycle either by fetal bovine serum or a combination of fibroblast growth factor 1 and p38 MAP kinase inhibitor. Phase-contrast videos revealed that binucleated cardiomyocytes form one metaphase plate and preferentially undergo afterwards cytokinesis failure. The maximum rate of cell division of video-recorded binucleated cardiomyocytes was around 6%. Immunofluorescence analyses of centriole number in mitotic binucleated cardiomyocytes revealed that these cells contain more than four centrioles, which can be paired as well as unpaired. In agreement with multiple and/or unpaired centrioles, multipolar spindle formation was observed in mitotic binucleated cardiomyocytes using fluorescence live imaging of tubulin-GFP. Multipoles were transient and resolved into pseudo-bipolar spindles both in case of cell division and cytokinesis failure. Notably, centrioles were in most cases unevenly distributed among daughter cells. CONCLUSIONS: Our results indicate that postnatal binucleated cardiomyocytes upon stimulation can enter mitosis, cope with their multiple and/or unpaired centrioles by forming pseudo-bipolar spindles, and divide.


Asunto(s)
División Celular/fisiología , Miocitos Cardíacos/fisiología , Animales , Ciclo Celular/fisiología , Núcleo Celular/metabolismo , Núcleo Celular/fisiología , Centriolos/metabolismo , Centriolos/fisiología , Citocinesis/fisiología , Mitosis/fisiología , Miocitos Cardíacos/metabolismo , Ratas , Ratas Sprague-Dawley , Tubulina (Proteína)/metabolismo
6.
Clin Sci (Lond) ; 133(11): 1229-1253, 2019 06 14.
Artículo en Inglés | MEDLINE | ID: mdl-31175264

RESUMEN

One great achievement in medical practice is the reduction in acute mortality of myocardial infarction due to identifying risk factors, antiplatelet therapy, optimized hospitalization and acute percutaneous coronary intervention. Yet, the prevalence of heart failure is increasing presenting a major socio-economic burden. Thus, there is a great need for novel therapies that can reverse damage inflicted to the heart. In recent years, data have accumulated suggesting that induction of cardiomyocyte proliferation might be a future option for cardiac regeneration. Here, we review the relevant literature since September 2015 concluding that it remains a challenge to verify that a therapy induces indeed cardiomyocyte proliferation. Most importantly, it is unclear that the detected increase in cardiomyocyte cell cycle activity is required for an associated improved function. In addition, we review the literature regarding the evidence that binucleated and polyploid mononucleated cardiomyocytes can divide, and put this in context to other cell types. Our analysis shows that there is significant evidence that binucleated cardiomyocytes can divide. Yet, it remains elusive whether also polyploid mononucleated cardiomyocytes can divide, how efficient proliferation of binucleated cardiomyocytes can be induced, what mechanism regulates cell cycle progression in these cells, and what fate and physiological properties the daughter cells have. In summary, we propose to standardize and independently validate cardiac regeneration studies, encourage the field to study the proliferative potential of binucleated and polyploid mononucleated cardiomyocytes, and to determine whether induction of polyploidization can enhance cardiac function post-injury.


Asunto(s)
Proliferación Celular , Corazón/fisiología , Miocitos Cardíacos/fisiología , Regeneración , Animales , Núcleo Celular/fisiología , Proliferación Celular/fisiología , Humanos , Poliploidía , Regeneración/fisiología , Medicina Regenerativa/métodos
7.
Biochem J ; 475(18): 2955-2967, 2018 09 25.
Artículo en Inglés | MEDLINE | ID: mdl-30120107

RESUMEN

Interferon-gamma (IFN-γ) is a pleiotropic cytokine that exerts important functions in inflammation, infectious diseases, and cancer. The large GTPase human guanylate-binding protein 1 (GBP-1) is among the most strongly IFN-γ-induced cellular proteins. Previously, it has been shown that GBP-1 mediates manifold cellular responses to IFN-γ including the inhibition of proliferation, spreading, migration, and invasion and through this exerts anti-tumorigenic activity. However, the mechanisms of GBP-1 anti-tumorigenic activities remain poorly understood. Here, we elucidated the molecular mechanism of the human GBP-1-mediated suppression of proliferation by demonstrating for the first time a cross-talk between the anti-tumorigenic IFN-γ and Hippo pathways. The α9-helix of GBP-1 was found to be sufficient to inhibit proliferation. Protein-binding and molecular modeling studies revealed that the α9-helix binds to the DNA-binding domain of the Hippo signaling transcription factor TEA domain protein (TEAD) mediated by the 376VDHLFQK382 sequence at the N-terminus of the GBP-1-α9-helix. Mutation of this sequence resulted in abrogation of both TEAD interaction and suppression of proliferation. Further on, the interaction caused inhibition of TEAD transcriptional activity associated with the down-regulation of TEAD-target genes. In agreement with these results, IFN-γ treatment of the cells also impaired TEAD activity, and this effect was abrogated by siRNA-mediated inhibition of GBP-1 expression. Altogether, this demonstrated that the α9-helix is the proliferation inhibitory domain of GBP-1, which acts independent of the GTPase activity through the inhibition of the Hippo transcription factor TEAD in mediating the anti-proliferative cell response to IFN-γ.


Asunto(s)
Proliferación Celular , Proteínas de Unión al GTP/metabolismo , Interferón gamma/metabolismo , Mutación Missense , Factores de Transcripción/metabolismo , Proteínas de Unión al GTP/genética , Células HeLa , Humanos , Interferón gamma/genética , Dominios Proteicos , Estructura Secundaria de Proteína , Factores de Transcripción/genética
8.
Pharmacol Rev ; 67(2): 338-67, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25713288

RESUMEN

The Adhesion family forms a large branch of the pharmacologically important superfamily of G protein-coupled receptors (GPCRs). As Adhesion GPCRs increasingly receive attention from a wide spectrum of biomedical fields, the Adhesion GPCR Consortium, together with the International Union of Basic and Clinical Pharmacology Committee on Receptor Nomenclature and Drug Classification, proposes a unified nomenclature for Adhesion GPCRs. The new names have ADGR as common dominator followed by a letter and a number to denote each subfamily and subtype, respectively. The new names, with old and alternative names within parentheses, are: ADGRA1 (GPR123), ADGRA2 (GPR124), ADGRA3 (GPR125), ADGRB1 (BAI1), ADGRB2 (BAI2), ADGRB3 (BAI3), ADGRC1 (CELSR1), ADGRC2 (CELSR2), ADGRC3 (CELSR3), ADGRD1 (GPR133), ADGRD2 (GPR144), ADGRE1 (EMR1, F4/80), ADGRE2 (EMR2), ADGRE3 (EMR3), ADGRE4 (EMR4), ADGRE5 (CD97), ADGRF1 (GPR110), ADGRF2 (GPR111), ADGRF3 (GPR113), ADGRF4 (GPR115), ADGRF5 (GPR116, Ig-Hepta), ADGRG1 (GPR56), ADGRG2 (GPR64, HE6), ADGRG3 (GPR97), ADGRG4 (GPR112), ADGRG5 (GPR114), ADGRG6 (GPR126), ADGRG7 (GPR128), ADGRL1 (latrophilin-1, CIRL-1, CL1), ADGRL2 (latrophilin-2, CIRL-2, CL2), ADGRL3 (latrophilin-3, CIRL-3, CL3), ADGRL4 (ELTD1, ETL), and ADGRV1 (VLGR1, GPR98). This review covers all major biologic aspects of Adhesion GPCRs, including evolutionary origins, interaction partners, signaling, expression, physiologic functions, and therapeutic potential.


Asunto(s)
Moléculas de Adhesión Celular/metabolismo , AMP Cíclico/fisiología , Modelos Moleculares , Receptores Acoplados a Proteínas G/metabolismo , Sistemas de Mensajero Secundario , Animales , Adhesión Celular , Moléculas de Adhesión Celular/química , Membrana Celular/enzimología , Membrana Celular/metabolismo , Movimiento Celular , Humanos , Agencias Internacionales , Ligandos , Farmacología/tendencias , Farmacología Clínica/tendencias , Isoformas de Proteínas/agonistas , Isoformas de Proteínas/química , Isoformas de Proteínas/clasificación , Isoformas de Proteínas/metabolismo , Receptores Acoplados a Proteínas G/agonistas , Receptores Acoplados a Proteínas G/química , Receptores Acoplados a Proteínas G/clasificación , Transducción de Señal , Sociedades Científicas , Terminología como Asunto
9.
Int J Mol Sci ; 19(7)2018 07 10.
Artículo en Inglés | MEDLINE | ID: mdl-29996502

RESUMEN

In contrast to the general belief that regeneration is a rare event, mainly occurring in simple organisms, the ability of regeneration is widely distributed in the animal kingdom. Yet, the efficiency and extent of regeneration varies greatly. Humans can recover from blood loss as well as damage to tissues like bone and liver. Yet damage to the heart and brain cannot be reversed, resulting in scaring. Thus, there is a great interest in understanding the molecular mechanisms of naturally occurring regeneration and to apply this knowledge to repair human organs. During regeneration, injury-activated immune cells induce wound healing, extracellular matrix remodeling, migration, dedifferentiation and/or proliferation with subsequent differentiation of somatic or stem cells. An anti-inflammatory response stops the regenerative process, which ends with tissue remodeling to achieve the original functional state. Notably, many of these processes are associated with enhanced glycolysis. Therefore, peroxisome proliferator-activated receptor (PPAR) ß/δ­which is known to be involved for example in lipid catabolism, glucose homeostasis, inflammation, survival, proliferation, differentiation, as well as mammalian regeneration of the skin, bone and liver­appears to be a promising target to promote mammalian regeneration. This review summarizes our current knowledge of PPARß/δ in processes associated with wound healing and regeneration.


Asunto(s)
Metabolismo de los Lípidos , PPAR delta/metabolismo , PPAR-beta/metabolismo , Cicatrización de Heridas , Animales , Diferenciación Celular , Glucólisis , Humanos , Regeneración , Vía de Señalización Wnt
10.
Ann Neurol ; 79(5): 826-840, 2016 May.
Artículo en Inglés | MEDLINE | ID: mdl-26971897

RESUMEN

OBJECTIVE: Mutations in the spastic paraplegia gene 11 (SPG11), encoding spatacsin, cause the most frequent form of autosomal-recessive complex hereditary spastic paraplegia (HSP) and juvenile-onset amyotrophic lateral sclerosis (ALS5). When SPG11 is mutated, patients frequently present with spastic paraparesis, a thin corpus callosum, and cognitive impairment. We previously delineated a neurodegenerative phenotype in neurons of these patients. In the current study, we recapitulated early developmental phenotypes of SPG11 and outlined their cellular and molecular mechanisms in patient-specific induced pluripotent stem cell (iPSC)-derived cortical neural progenitor cells (NPCs). METHODS: We generated and characterized iPSC-derived NPCs and neurons from 3 SPG11 patients and 2 age-matched controls. RESULTS: Gene expression profiling of SPG11-NPCs revealed widespread transcriptional alterations in neurodevelopmental pathways. These include changes in cell-cycle, neurogenesis, cortical development pathways, in addition to autophagic deficits. More important, the GSK3ß-signaling pathway was found to be dysregulated in SPG11-NPCs. Impaired proliferation of SPG11-NPCs resulted in a significant diminution in the number of neural cells. The decrease in mitotically active SPG11-NPCs was rescued by GSK3 modulation. INTERPRETATION: This iPSC-derived NPC model provides the first evidence for an early neurodevelopmental phenotype in SPG11, with GSK3ß as a potential novel target to reverse the disease phenotype. Ann Neurol 2016;79:826-840.

11.
Eur Heart J ; 37(23): 1789-98, 2016 06 14.
Artículo en Inglés | MEDLINE | ID: mdl-27055812

RESUMEN

Despite improvements in modern cardiovascular therapy, the morbidity and mortality of ischaemic heart disease (IHD) and heart failure (HF) remain significant in Europe and worldwide. Patients with IHD may benefit from therapies that would accelerate natural processes of postnatal collateral vessel formation and/or muscle regeneration. Here, we discuss the use of cells in the context of heart repair, and the most relevant results and current limitations from clinical trials using cell-based therapies to treat IHD and HF. We identify and discuss promising potential new therapeutic strategies that include ex vivo cell-mediated gene therapy, the use of biomaterials and cell-free therapies aimed at increasing the success rates of therapy for IHD and HF. The overall aim of this Position Paper of the ESC Working Group Cellular Biology of the Heart is to provide recommendations on how to improve the therapeutic application of cell-based therapies for cardiac regeneration and repair.


Asunto(s)
Tratamiento Basado en Trasplante de Células y Tejidos/métodos , Insuficiencia Cardíaca/terapia , Corazón/fisiología , Isquemia Miocárdica/terapia , Rastreo Celular/métodos , Ensayos Clínicos como Asunto , Exactitud de los Datos , Ética Médica , Insuficiencia Cardíaca/fisiopatología , Humanos , Isquemia Miocárdica/fisiopatología , Seguridad del Paciente , Selección de Paciente , Regeneración/fisiología , Trasplante de Células Madre/métodos , Volumen Sistólico/fisiología , Resultado del Tratamiento
12.
Am J Physiol Heart Circ Physiol ; 310(9): H1045-54, 2016 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-26921436

RESUMEN

In recent years, there has been a dramatic increase in research aimed at regenerating the mammalian heart by promoting endogenous cardiomyocyte proliferation. Despite many encouraging successes, it remains unclear if we are any closer to achieving levels of mammalian cardiomyocyte proliferation for regeneration as seen during zebrafish regeneration. Furthermore, current cardiac regenerative approaches do not clarify whether the induced cardiomyocyte proliferation is an epiphenomena or responsible for the observed improvement in cardiac function. Moreover, due to the lack of standardized protocols to determine cardiomyocyte proliferation in vivo, it remains unclear if one mammalian regenerative factor is more effective than another. Here, we discuss current methods to identify and evaluate factors for the induction of cardiomyocyte proliferation and challenges therein. Addressing challenges in evaluating adult cardiomyocyte proliferation will assist in determining 1) which regenerative factors should be pursued in large animal studies; 2) if a particular level of cell cycle regulation presents a better therapeutic target than another (e.g., mitogenic receptors vs. cyclins); and 3) which combinatorial approaches offer the greatest likelihood of success. As more and more regenerative studies come to pass, progress will require a system that not only can evaluate efficacy in an objective manner but can also consolidate observations in a meaningful way.


Asunto(s)
Proliferación Celular , Cardiopatías/terapia , Miocitos Cardíacos/patología , Regeneración , Medicina Regenerativa/métodos , Animales , Ciclo Celular , Proteínas de Ciclo Celular/metabolismo , Cardiopatías/metabolismo , Cardiopatías/patología , Cardiopatías/fisiopatología , Humanos , Modelos Animales , Miocitos Cardíacos/metabolismo , Recuperación de la Función , Transducción de Señal
13.
Basic Res Cardiol ; 111(6): 69, 2016 11.
Artículo en Inglés | MEDLINE | ID: mdl-27743118

RESUMEN

In this meeting report, particularly addressing the topic of protection of the cardiovascular system from ischemia/reperfusion injury, highlights are presented that relate to conditioning strategies of the heart with respect to molecular mechanisms and outcome in patients' cohorts, the influence of co-morbidities and medications, as well as the contribution of innate immune reactions in cardioprotection. Moreover, developmental or systems biology approaches bear great potential in systematically uncovering unexpected components involved in ischemia-reperfusion injury or heart regeneration. Based on the characterization of particular platelet integrins, mitochondrial redox-linked proteins, or lipid-diol compounds in cardiovascular diseases, their targeting by newly developed theranostics and technologies opens new avenues for diagnosis and therapy of myocardial infarction to improve the patients' outcome.


Asunto(s)
Cardiología/tendencias , Enfermedades Cardiovasculares , Nanomedicina Teranóstica/tendencias , Animales , Cardiología/métodos , Humanos
14.
Handb Exp Pharmacol ; 234: 351-368, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27832496

RESUMEN

The cardiovascular system in adult organisms forms a network of interconnected endothelial cells, supported by mural cells and displaying a high degree of hierarchy: arteries emerging from the heart ramify into arterioles and then capillaries, which return to the venous systems through venules and veins. The cardiovascular system allows blood circulation, which in turn is essential for hemostasis through gas diffusion, nutrient distribution, and cell trafficking. In this chapter, we have summarized the current knowledge on how adhesion GPCRs (aGPCRs) impact heart development, followed by their role in modulating vascular angiogenesis.


Asunto(s)
Barrera Hematoencefálica/metabolismo , Adhesión Celular , Membrana Celular/metabolismo , Corazón/crecimiento & desarrollo , Neovascularización Fisiológica , Receptores Acoplados a Proteínas G/metabolismo , Animales , Sitios de Unión , Barrera Hematoencefálica/crecimiento & desarrollo , Regulación del Desarrollo de la Expresión Génica , Humanos , Modelos Moleculares , Unión Proteica , Dominios y Motivos de Interacción de Proteínas , Receptores Acoplados a Proteínas G/química , Receptores Acoplados a Proteínas G/genética , Transducción de Señal , Relación Estructura-Actividad
15.
Proc Natl Acad Sci U S A ; 110(42): 16898-903, 2013 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-24082093

RESUMEN

Despite their abundance and multiple functions in a variety of organ systems, the function and signaling mechanisms of adhesion G protein-coupled receptors (GPCRs) are poorly understood. Adhesion GPCRs possess large N termini containing various functional domains. In addition, many of them are autoproteolytically cleaved at their GPS sites into an N-terminal fragment (NTF) and C-terminal fragment. Here we demonstrate that Gpr126 is expressed in the endocardium during early mouse heart development. Gpr126 knockout in mice and knockdown in zebrafish caused hypotrabeculation and affected mitochondrial function. Ectopic expression of Gpr126-NTF that lacks the GPS motif (NTF(ΔGPS)) in zebrafish rescued the trabeculation but not the previously described myelination phenotype in the peripheral nervous system. These data support a model in which the NTF of Gpr126, in contrast to the C-terminal fragment, plays an important role in heart development. Collectively, our analysis provides a unique example of the versatile function and signaling properties of adhesion GPCRs in vertebrates.


Asunto(s)
Endocardio/embriología , Mitocondrias Cardíacas/metabolismo , Modelos Biológicos , Receptores Acoplados a Proteínas G/metabolismo , Proteínas de Pez Cebra/metabolismo , Pez Cebra/embriología , Animales , Endocardio/citología , Ratones , Ratones Noqueados , Mitocondrias Cardíacas/genética , Especificidad de Órganos/fisiología , Estructura Terciaria de Proteína , Receptores Acoplados a Proteínas G/genética , Pez Cebra/genética , Proteínas de Pez Cebra/genética
16.
Biochim Biophys Acta ; 1843(11): 2556-62, 2014 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-25014164

RESUMEN

Apoptosis, or programmed cell death, is an essential physiological process for proper embryogenesis as well as for homeostasis during aging. In addition, apoptosis is one of the major mechanisms causing cell loss in pathophysiological conditions such as heart failure. Thus, inhibition of apoptosis is an important approach for preventive and therapeutic strategies. Here we show that the histone 3 lysine 4- and lysine 36-specific methyltransferase Smyd2 acts as an endogenous antagonistic player of p53-dependent cardiomyocyte apoptosis. Smyd2 protein levels were significantly decreased in cardiomyocytes upon cobalt chloride-induced apoptosis or myocardial infarction, while p53 expression was enhanced. siRNA-mediated knockdown of Smyd2 in cultured cardiomyocytes further enhanced cobalt chloride-induced cardiomyocyte apoptosis. In contrast, Smyd2 overexpression resulted in marked methylation of p53 and prevented its accumulation as well as apoptotic cell death in an Hsp90-independent manner. Moreover, overexpression, of Smyd2, but not Smyd2Y240F lacking a methyl transferase activity, significantly rescued CoCl2-induced apoptosis in H9c2 cardioblasts. Finally, Smyd2 cardiomyocyte-specific deletion in vivo promoted apoptotic cell death upon myocardial infarction, which correlated with enhanced expression of p53 and pro-apoptotic Bax. Collectively, our data indicate Smyd2 as a cardioprotective protein by methylating p53.

17.
Am J Physiol Renal Physiol ; 309(2): F164-78, 2015 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-26017974

RESUMEN

Kidney aging is accompanied by characteristic changes in the glomerulus, but little is known about the effect of aging on glomerular parietal epithelial cells (PECs), nor if the characteristic glomerular changes in humans and rats also occur in very old mice. Accordingly, a descriptive analysis was undertaken in 27-mo-old C57B6 mice, considered advanced age. PEC density was significantly lower in older mice compared with young mice (aged 3 mo), and the decrease was more pronounced in juxtamedullary glomeruli compared with outer cortical glomeruli. In addition to segmental and global glomerulosclerosis in older mice, staining for matrix proteins collagen type IV and heparan sulfate proteoglycan were markedly increased in Bowman's capsules of older mouse glomeruli, consistent with increased extracellular matrix production by PECs. De novo staining for CD44, a marker of activated and profibrotic PECs, was significantly increased in aged glomeruli. CD44 staining was more pronounced in the juxtamedullary region and colocalized with phosphorylated ERK. Additionally, a subset of aged PECs de novo expressed the epithelial-to-mesenchymal transition markers α-smooth muscle and vimentin, with no changes in epithelial-to-mesenchymal transition markers E-cadherin and ß-catenin. The mural cell markers neural/glial antigen 2, PDGF receptor-ß, and CD146 as well as Notch 3 were also substantially increased in aged PECs. These data show that mice can be used to better understand the aging kidney and that PECs undergo substantial changes, especially in juxtamedullary glomeruli, that may participate in the overall decline in glomerular structure and function with advancing age.


Asunto(s)
Envejecimiento/patología , Células Epiteliales/patología , Glomérulos Renales/patología , Envejecimiento/metabolismo , Animales , Biomarcadores/metabolismo , Cápsula Glomerular/metabolismo , Células Epiteliales/metabolismo , Transición Epitelial-Mesenquimal , Proteínas de la Matriz Extracelular/metabolismo , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Femenino , Receptores de Hialuranos/metabolismo , Glomérulos Renales/metabolismo , Ratones Endogámicos C57BL , Pericitos/metabolismo , Fosforilación , Podocitos , Receptor Notch3 , Receptores Notch/metabolismo
18.
Artículo en Inglés | MEDLINE | ID: mdl-23605180

RESUMEN

Mammalian cardiomyocytes withdraw from the cell cycle shortly after birth. Although the adult heart is unable to regenerate, numerous reports have shown that adult cardiomyocytes exhibit a dynamic range of cell cycle activity under various physiological and pathological conditions. Reason and consequence of cardiomyocyte cell cycle activity remain unclear and have led to a number of misconceptions. Understanding the scenarios in which cycling happens may promote new perspectives on the differentiated state of cardiomyocytes, treatments for hypertrophy, heart regeneration and cancer therapy. In this review we discuss the result of cardiomyocyte cell cycle activity in aging and disease and studies manipulating cardiac cell cycle activity to promote cardiac regeneration. In addition, we focus on cardiomyocyte differentiation, cell cycle exit, and the relationship between ploidy and regenerative potential. Finally, we provide observations that may further advance the goal of inducing adult mammalian heart regeneration through cardiomyocyte proliferation.


Asunto(s)
Cardiomegalia/patología , Homeostasis , Miocitos Cardíacos/patología , Regeneración , Animales , Ciclo Celular , Diferenciación Celular , Proliferación Celular , Humanos , Miocitos Cardíacos/fisiología
19.
Am J Physiol Heart Circ Physiol ; 309(8): H1237-50, 2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-26342071

RESUMEN

The newt and the zebrafish have the ability to regenerate many of their tissues and organs including the heart. Thus, a major goal in experimental medicine is to elucidate the molecular mechanisms underlying the regenerative capacity of these species. A wide variety of experiments have demonstrated that naturally occurring heart regeneration relies on cardiomyocyte proliferation. Thus, major efforts have been invested to induce proliferation of mammalian cardiomyocytes in order to improve cardiac function after injury or to protect the heart from further functional deterioration. In this review, we describe and analyze methods currently used to evaluate cardiomyocyte proliferation. In addition, we summarize the literature on naturally occurring heart regeneration. Our analysis highlights that newt and zebrafish heart regeneration relies on factors that are also utilized in cardiomyocyte proliferation during mammalian fetal development. Most of these factors have, however, failed to induce adult mammalian cardiomyocyte proliferation. Finally, our analysis of mammalian neonatal heart regeneration indicates experiments that could resolve conflicting results in the literature, such as binucleation assays and clonal analysis. Collectively, cardiac regeneration based on cardiomyocyte proliferation is a promising approach for improving adult human cardiac function after injury, but it is important to elucidate the mechanisms arresting mammalian cardiomyocyte proliferation after birth and to utilize better assays to determine formation of new muscle mass.


Asunto(s)
Investigación Biomédica/métodos , Proliferación Celular , Cardiopatías/patología , Corazón/embriología , Corazón/crecimiento & desarrollo , Miocitos Cardíacos/patología , Regeneración , Animales , Bioensayo , Diferenciación Celular , Linaje de la Célula , Cardiopatías/metabolismo , Cardiopatías/fisiopatología , Humanos , Modelos Animales , Miocitos Cardíacos/metabolismo , Organogénesis , Salamandridae , Transducción de Señal , Pez Cebra
20.
Blood ; 121(15): 3041-50, 2013 04 11.
Artículo en Inglés | MEDLINE | ID: mdl-23386126

RESUMEN

Angiogenesis, defined as blood vessel formation from a preexisting vasculature, is governed by multiple signal cascades including integrin receptors, in particular integrin αVß3. Here we identify the endothelial cell (EC)-secreted factor epidermal growth factor-like protein 7 (EGFL7) as a novel specific ligand of integrin αVß3, thus providing mechanistic insight into its proangiogenic actions in vitro and in vivo. Specifically, EGFL7 attaches to the extracellular matrix and by its interaction with integrin αVß3 increases the motility of EC, which allows EC to move on a sticky underground during vessel remodeling. We provide evidence that the deregulation of EGFL7 in zebrafish embryos leads to a severe integrin-dependent malformation of the caudal venous plexus, pointing toward the significance of EGFL7 in vessel development. In biopsy specimens of patients with neurologic diseases, vascular EGFL7 expression rose with increasing EC proliferation. Further, EGFL7 became upregulated in vessels of the stroke penumbra using a mouse model of reversible middle cerebral artery occlusion. Our data suggest that EGFL7 expression depends on the remodeling state of the existing vasculature rather than on the phenotype of neurologic disease analyzed. In sum, our work sheds a novel light on the molecular mechanism EGFL7 engages to govern physiological and pathological angiogenesis.


Asunto(s)
Vasos Sanguíneos/metabolismo , Factores de Crecimiento Endotelial/metabolismo , Células Endoteliales de la Vena Umbilical Humana/metabolismo , Integrina alfaVbeta3/metabolismo , Secuencias de Aminoácidos/genética , Animales , Proteínas de Unión al Calcio , Adhesión Celular/genética , Movimiento Celular/genética , Familia de Proteínas EGF , Embrión no Mamífero/irrigación sanguínea , Embrión no Mamífero/metabolismo , Factores de Crecimiento Endotelial/genética , Factores de Crecimiento Endotelial/farmacología , Matriz Extracelular/metabolismo , Expresión Génica , Células HEK293 , Humanos , Inmunohistoquímica , Inmunoprecipitación , Infarto de la Arteria Cerebral Media/genética , Infarto de la Arteria Cerebral Media/metabolismo , Integrina alfaVbeta3/genética , Ratones , Ratones Desnudos , Fosforilación/efectos de los fármacos , Unión Proteica , Interferencia de ARN , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Pez Cebra
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA