Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
1.
Circulation ; 148(9): 778-797, 2023 08 29.
Artículo en Inglés | MEDLINE | ID: mdl-37427428

RESUMEN

BACKGROUND: Cardiac fibroblasts have crucial roles in the heart. In particular, fibroblasts differentiate into myofibroblasts in the damaged myocardium, contributing to scar formation and interstitial fibrosis. Fibrosis is associated with heart dysfunction and failure. Myofibroblasts therefore represent attractive therapeutic targets. However, the lack of myofibroblast-specific markers has precluded the development of targeted therapies. In this context, most of the noncoding genome is transcribed into long noncoding RNAs (lncRNAs). A number of lncRNAs have pivotal functions in the cardiovascular system. lncRNAs are globally more cell-specific than protein-coding genes, supporting their importance as key determinants of cell identity. METHODS: In this study, we evaluated the value of the lncRNA transcriptome in very deep single-cell RNA sequencing. We profiled the lncRNA transcriptome in cardiac nonmyocyte cells after infarction and probed heterogeneity in the fibroblast and myofibroblast populations. In addition, we searched for subpopulation-specific markers that can constitute novel targets in therapy for heart disease. RESULTS: We demonstrated that cardiac cell identity can be defined by the sole expression of lncRNAs in single-cell experiments. In this analysis, we identified lncRNAs enriched in relevant myofibroblast subpopulations. Selecting 1 candidate we named FIXER (fibrogenic LOX-locus enhancer RNA), we showed that its silencing limits fibrosis and improves heart function after infarction. Mechanitically, FIXER interacts with CBX4, an E3 SUMO protein ligase and transcription factor, guiding CBX4 to the promoter of the transcription factor RUNX1 to control its expression and, consequently, the expression of a fibrogenic gene program.. FIXER is conserved in humans, supporting its translational value. CONCLUSIONS: Our results demonstrated that lncRNA expression is sufficient to identify the various cell types composing the mammalian heart. Focusing on cardiac fibroblasts and their derivatives, we identified lncRNAs uniquely expressed in myofibroblasts. In particular, the lncRNA FIXER represents a novel therapeutic target for cardiac fibrosis.


Asunto(s)
Cardiomiopatías , ARN Largo no Codificante , Animales , Humanos , Transcriptoma , ARN Largo no Codificante/genética , ARN Largo no Codificante/metabolismo , Cardiomiopatías/genética , Fibrosis , Análisis de Secuencia de ARN , Factores de Transcripción/genética , Infarto , Mamíferos/genética , Mamíferos/metabolismo , Ligasas/genética , Ligasas/metabolismo , Proteínas del Grupo Polycomb/genética , Proteínas del Grupo Polycomb/metabolismo
2.
J Mol Cell Cardiol ; 89(Pt A): 98-112, 2015 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-26423156

RESUMEN

Long noncoding RNAs (lncRNAs) are emerging as important regulators of developmental pathways. However, their roles in human cardiac precursor cell (CPC) remain unexplored. To characterize the long noncoding transcriptome during human CPC cardiac differentiation, we profiled the lncRNA transcriptome in CPCs isolated from the human fetal heart and identified 570 lncRNAs that were modulated during cardiac differentiation. Many of these were associated with active cardiac enhancer and super enhancers (SE) with their expression being correlated with proximal cardiac genes. One of the most upregulated lncRNAs was a SE-associated lncRNA that was named CARMEN, (CAR)diac (M)esoderm (E)nhancer-associated (N)oncoding RNA. CARMEN exhibits RNA-dependent enhancing activity and is upstream of the cardiac mesoderm-specifying gene regulatory network. Interestingly, CARMEN interacts with SUZ12 and EZH2, two components of the polycomb repressive complex 2 (PRC2). We demonstrate that CARMEN knockdown inhibits cardiac specification and differentiation in cardiac precursor cells independently of MIR-143 and -145 expression, two microRNAs located proximal to the enhancer sequences. Importantly, CARMEN expression was activated during pathological remodeling in the mouse and human hearts, and was necessary for maintaining cardiac identity in differentiated cardiomyocytes. This study demonstrates therefore that CARMEN is a crucial regulator of cardiac cell differentiation and homeostasis.


Asunto(s)
Tipificación del Cuerpo/genética , Diferenciación Celular/genética , Corazón/embriología , Homeostasis/genética , ARN Largo no Codificante/metabolismo , Animales , Linaje de la Célula/genética , Elementos de Facilitación Genéticos/genética , Proteína Potenciadora del Homólogo Zeste 2 , Perfilación de la Expresión Génica , Regulación del Desarrollo de la Expresión Génica , Técnicas de Silenciamiento del Gen , Humanos , Ratones , Miocardio/patología , Complejo Represivo Polycomb 2/metabolismo , ARN Largo no Codificante/genética , Células Madre/citología , Transcriptoma/genética
3.
Eur Heart J ; 35(32): 2174-85, 2014 Aug 21.
Artículo en Inglés | MEDLINE | ID: mdl-23166366

RESUMEN

AIMS: In the adult heart, Notch signalling regulates the response to injury. Notch inhibition leads to increased cardiomyocyte apoptosis, and exacerbates the development of cardiac hypertrophy and fibrosis. The role of Notch in the mesenchymal stromal cell fraction, which contains cardiac fibroblasts and cardiac precursor cells, is, however, largely unknown. In the present study, we evaluate, therefore, whether forced activation of the Notch pathway in mesenchymal stromal cells regulates pathological cardiac remodelling. METHODS AND RESULTS: We generated transgenic mice overexpressing the Notch ligand Jagged1 on the surface of cardiomyocytes to activate Notch signalling in adjacent myocyte and non-myocyte cells. In neonatal transgenic mice, activated Notch sustained cardiac precursor and myocyte proliferation after birth, and led to increased numbers of cardiac myocytes in adult mice. In the adult heart under pressure overload, Notch inhibited the development of cardiomyocyte hypertrophy and transforming growth factor-ß/connective tissue growth factor-mediated cardiac fibrosis. Most importantly, Notch activation in the stressed adult heart reduced the proliferation of myofibroblasts and stimulated the expansion of stem cell antigen-1-positive cells, and in particular of Nkx2.5-positive cardiac precursor cells. CONCLUSIONS: We conclude that Notch is pivotal in the healing process of the injured heart. Specifically, Notch regulates key cellular mechanisms in the mesenchymal stromal cell population, and thereby controls the balance between fibrotic and regenerative repair in the adult heart. Altogether, these findings indicate that Notch represents a unique therapeutic target for inducing regeneration in the adult heart via mobilization of cardiac precursor cells.


Asunto(s)
Receptores Notch/fisiología , Transducción de Señal/fisiología , Remodelación Ventricular/fisiología , Animales , Proteínas de Unión al Calcio/metabolismo , Cardiomegalia/fisiopatología , Cardiomegalia/terapia , Proliferación Celular/fisiología , Tamaño de la Célula , Constricción , Fibrosis/metabolismo , Corazón/fisiología , Humanos , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Proteína Jagged-1 , Proteínas de la Membrana/metabolismo , Ratones Transgénicos , Miocitos Cardíacos/citología , Miocitos Cardíacos/metabolismo , Fosfohidrolasa PTEN/metabolismo , Fosfatidilinositol 3-Quinasas/metabolismo , Regeneración , Proteínas Serrate-Jagged , Serina-Treonina Quinasas TOR/metabolismo , Factores de Crecimiento Transformadores/metabolismo
4.
Cardiovasc Res ; 119(6): 1361-1376, 2023 06 13.
Artículo en Inglés | MEDLINE | ID: mdl-36537036

RESUMEN

AIMS: The major cardiac cell types composing the adult heart arise from common multipotent precursor cells. Cardiac lineage decisions are guided by extrinsic and cell-autonomous factors, including recently discovered long noncoding RNAs (lncRNAs). The human lncRNA CARMEN, which is known to dictate specification toward the cardiomyocyte (CM) and the smooth muscle cell (SMC) fates, generates a diversity of alternatively spliced isoforms. METHODS AND RESULTS: The CARMEN locus can be manipulated to direct human primary cardiac precursor cells (CPCs) into specific cardiovascular fates. Investigating CARMEN isoform usage in differentiating CPCs represents therefore a unique opportunity to uncover isoform-specific functions in lncRNAs. Here, we identify one CARMEN isoform, CARMEN-201, to be crucial for SMC commitment. CARMEN-201 activity is encoded within an alternatively spliced exon containing a MIRc short interspersed nuclear element. This element binds the transcriptional repressor REST (RE1 Silencing Transcription Factor), targets it to cardiogenic loci, including ISL1, IRX1, IRX5, and SFRP1, and thereby blocks the CM gene program. In turn, genes regulating SMC differentiation are induced. CONCLUSIONS: These data show how a critical physiological switch is wired by alternative splicing and functional transposable elements in a long noncoding RNA. They further demonstrated the crucial importance of the lncRNA isoform CARMEN-201 in SMC specification during heart development.


Asunto(s)
ARN Largo no Codificante , Humanos , ARN Largo no Codificante/genética , Elementos Transponibles de ADN , Corazón , Diferenciación Celular/genética , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo
5.
Circulation ; 123(10): 1073-82, 2011 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-21357822

RESUMEN

BACKGROUND: Cardiac hypertrophy involves growth responses to a variety of stimuli triggered by increased workload. It is an independent risk factor for heart failure and sudden death. Mammalian target of rapamycin (mTOR) plays a key role in cellular growth responses by integrating growth factor and energy status signals. It is found in 2 structurally and functionally distinct multiprotein complexes called mTOR complex (mTORC) 1 and mTORC2. The role of each of these branches of mTOR signaling in the adult heart is currently unknown. METHODS AND RESULTS: We generated mice with deficient myocardial mTORC1 activity by targeted ablation of raptor, which encodes an essential component of mTORC1, during adulthood. At 3 weeks after the deletion, atrial and brain natriuretic peptides and ß-myosin heavy chain were strongly induced, multiple genes involved in the regulation of energy metabolism were altered, but cardiac function was normal. Function deteriorated rapidly afterward, resulting in dilated cardiomyopathy and high mortality within 6 weeks. Aortic banding-induced pathological overload resulted in severe dilated cardiomyopathy already at 1 week without a prior phase of adaptive hypertrophy. The mechanism involved a lack of adaptive cardiomyocyte growth via blunted protein synthesis capacity, as supported by reduced phosphorylation of ribosomal S6 kinase 1 and 4E-binding protein 1. In addition, reduced mitochondrial content, a shift in metabolic substrate use, and increased apoptosis and autophagy were observed. CONCLUSIONS: Our results demonstrate an essential function for mTORC1 in the heart under physiological and pathological conditions and are relevant for the understanding of disease states in which the insulin/insulin-like growth factor signaling axis is affected such as diabetes mellitus and heart failure or after cancer therapy.


Asunto(s)
Cardiomegalia/genética , Cardiomegalia/fisiopatología , Proteínas Portadoras/genética , Proteínas Portadoras/fisiología , Insuficiencia Cardíaca/etiología , Frecuencia Cardíaca/fisiología , Proteínas Adaptadoras Transductoras de Señales , Animales , Apoptosis , Factor Natriurético Atrial/análisis , Factor Natriurético Atrial/metabolismo , Autofagia , Proteínas Portadoras/metabolismo , Proteínas de Ciclo Celular , Metabolismo Energético/genética , Metabolismo Energético/fisiología , Factores Eucarióticos de Iniciación , Expresión Génica/fisiología , Insuficiencia Cardíaca/genética , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Mitocondrias Cardíacas/metabolismo , Mitocondrias Cardíacas/fisiología , Cadenas Pesadas de Miosina/análisis , Cadenas Pesadas de Miosina/metabolismo , Péptido Natriurético Encefálico/análisis , Péptido Natriurético Encefálico/metabolismo , Miosina Tipo IIB no Muscular/análisis , Miosina Tipo IIB no Muscular/metabolismo , Fosfoproteínas/metabolismo , Fosforilación , Proteína Reguladora Asociada a mTOR , Proteínas Quinasas S6 Ribosómicas 90-kDa/metabolismo
6.
J Cardiovasc Dev Dis ; 9(4)2022 Apr 07.
Artículo en Inglés | MEDLINE | ID: mdl-35448087

RESUMEN

Cardiac pathologies lead to an acute or gradual loss of cardiomyocytes. Because of the limited regenerative capacity of the mammalian heart, cardiomyocytes are only replaced by fibrotic tissue. Excessive fibrosis contributes to the deterioration of cardiac function and the transition to heart failure, which is the leading cause of morbidity and mortality worldwide. Currently, no treatments can promote replenishment of the injured heart with newly formed cardiomyocytes. In this context, regenerative strategies explore the possibility to promote recovery through induction of cardiomyocyte production from pre-existing cardiomyocytes. On the other hand, cardiac non-myocyte cells can be directly reprogrammed into induced cardiac precursor cells and cardiomyocytes, suggesting that these cells could be exploited to produce cardiomyocytes in vivo. Here, we provide evidence that the sequential activation and inhibition of the NOTCH1 signaling pathway in the stressed heart decreases fibrosis and improves cardiac function in the stressed heart. This is accompanied by the emergence of new cardiomyocytes from non-myocyte origin. Overall, our data show how a developmental pathway such as the NOTCH pathway can be manipulated to provide therapeutic benefit in the damaged heart.

7.
Biochem J ; 424(1): 119-27, 2009 Oct 23.
Artículo en Inglés | MEDLINE | ID: mdl-19708858

RESUMEN

Ucn2 (urocortin 2) has been shown to exert potent beneficial effects in the cardiovascular system, including inhibition of apoptosis, improvement of cardiomyocyte contractility and decrease of oxidative stress. The mechanisms that contribute to the regulation of hUcn2 (human Ucn2) expression in cardiovascular pathologies are not known. In the present study, we analysed the mechanism by which hypoxia, a major stimulus in ischaemic heart disease, regulates Ucn2 gene expression. Hypoxia and CPX (ciclopirox olamine), which prevents proteolytic degradation of HIF (hypoxia-inducible factor), significantly increased hUcn2 mRNA levels in TE-671 cells. Gene silencing of endogenous HIF1alpha abolishes this increase. Hypoxia and CPX activated a luciferase-linked fragment of the 3'FLR (3'-flanking region) of the hUcn2 gene containing two putative HREs (hypoxia-response elements), HRE1 and HRE2. Site-directed mutagenesis experiments demonstrated that HRE1 is required for HIF1alpha-dependent luciferase activation. This activation was conserved in constructs with the 3'FLR fragment placed upstream of the luciferase gene, indicating an enhancer function for HRE1. Competition assays revealed direct binding between HRE1 and HIF1alpha. Regulation of Ucn2 by hypoxia was confirmed in rat neonatal cardiomyocytes and in cardiac-derived H9c2 cells transfected with constructs of the 3'FLR of the hUcn2 gene. In conclusion, our study demonstrates that hypoxia induces hUcn2 expression via a specific HRE in the 3'FLR of the hUcn2 gene, which interacts with the transcription factor HIF1alpha. Hypoxia-mediated stimulation of cardioprotective Ucn2 may help to preserve cardiac function and prevent apoptosis in ischaemic conditions in the heart.


Asunto(s)
Hipoxia de la Célula/fisiología , Elementos de Respuesta/genética , Urocortinas/genética , Urocortinas/metabolismo , Animales , Animales Recién Nacidos , Antifúngicos/farmacología , Hipoxia de la Célula/genética , Línea Celular , Línea Celular Tumoral , Células Cultivadas , Ciclopirox , Regulación de la Expresión Génica , Humanos , Subunidad alfa del Factor 1 Inducible por Hipoxia/genética , Subunidad alfa del Factor 1 Inducible por Hipoxia/fisiología , Immunoblotting , Mutagénesis Sitio-Dirigida , Piridonas/farmacología , Ratas , Ratas Wistar , Elementos de Respuesta/fisiología
8.
Front Cardiovasc Med ; 5: 90, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30087899

RESUMEN

High aldehyde dehydrogenase (ALDHhi) activity has been reported in normal and cancer stem cells. We and others have shown previously that human ALDHhi cardiac atrial appendage cells are enriched with stem/progenitor cells. The role of ALDH in these cells is poorly understood but it may come down to the specific ALDH isoform(s) expressed. This study aimed to compare ALDHhi and ALDHlo atrial cells and to identify the isoform(s) that contribute to ALDH activity, and their functional role. Methods and Results: Cells were isolated from atrial appendage specimens from patients with ischemic and/or valvular heart disease undergoing heart surgery. ALDHhi activity assessed with the Aldefluor reagent coincided with primitive surface marker expression (CD34+). Depending on their ALDH activity, RT-PCR analysis of ALDHhi and ALDHlo cells demonstrated a differential pattern of pluripotency genes (Oct 4, Nanog) and genes for more established cardiac lineages (Nkx2.5, Tbx5, Mef2c, GATA4). ALDHhi cells, but not ALDHlo cells, formed clones and were culture-expanded. When cultured under cardiac differentiation conditions, ALDHhi cells gave rise to a higher number of cardiomyocytes compared with ALDHlo cells. Among 19 ALDH isoforms known in human, ALDH1A3 was most highly expressed in ALDHhi atrial cells. Knocking down ALDH1A3, but not ALDH1A1, ALDH1A2, ALDH2, ALDH4A1, or ALDH8A1 using siRNA decreased ALDH activity and cell proliferation in ALDHhi cells. Conversely, overexpressing ALDH1A3 with a retroviral vector increased proliferation in ALDHlo cells. Conclusions: ALDH1A3 is the key isoform responsible for ALDH activity in ALDHhi atrial appendage cells, which have a propensity to differentiate into cardiomyocytes. ALDH1A3 affects in vitro proliferation of these cells.

9.
Sci Transl Med ; 9(395)2017 06 21.
Artículo en Inglés | MEDLINE | ID: mdl-28637928

RESUMEN

Long noncoding RNAs (lncRNAs) are emerging as powerful regulators of cardiac development and disease. However, our understanding of the importance of these molecules in cardiac fibrosis is limited. Using an integrated genomic screen, we identified Wisper (Wisp2 super-enhancer-associated RNA) as a cardiac fibroblast-enriched lncRNA that regulates cardiac fibrosis after injury. Wisper expression was correlated with cardiac fibrosis both in a murine model of myocardial infarction (MI) and in heart tissue from human patients suffering from aortic stenosis. Loss-of-function approaches in vitro using modified antisense oligonucleotides (ASOs) demonstrated that Wisper is a specific regulator of cardiac fibroblast proliferation, migration, and survival. Accordingly, ASO-mediated silencing of Wisper in vivo attenuated MI-induced fibrosis and cardiac dysfunction. Functionally, Wisper regulates cardiac fibroblast gene expression programs critical for cell identity, extracellular matrix deposition, proliferation, and survival. In addition, its association with TIA1-related protein allows it to control the expression of a profibrotic form of lysyl hydroxylase 2, implicated in collagen cross-linking and stabilization of the matrix. Together, our findings identify Wisper as a cardiac fibroblast-enriched super-enhancer-associated lncRNA that represents an attractive therapeutic target to reduce the pathological development of cardiac fibrosis in response to MI and prevent adverse remodeling in the damaged heart.


Asunto(s)
Cardiomiopatías/genética , ARN Largo no Codificante/genética , Cardiomiopatías/patología , Fibroblastos/metabolismo , Fibroblastos/patología , Fibrosis/genética , Fibrosis/patología , Humanos , ARN Largo no Codificante/fisiología , Remodelación Ventricular
10.
Nat Commun ; 8(1): 1806, 2017 11 27.
Artículo en Inglés | MEDLINE | ID: mdl-29180618

RESUMEN

Enhancers and long noncoding RNAs (lncRNAs) are key determinants of lineage specification during development. Here, we evaluate remodeling of the enhancer landscape and modulation of the lncRNA transcriptome during mesendoderm specification. We sort mesendodermal progenitors from differentiating embryonic stem cells (ESCs) according to Eomes expression, and find that enhancer usage is coordinated with mesendoderm-specific expression of key lineage-determining transcription factors. Many of these enhancers are associated with the expression of lncRNAs. Examination of ESC-specific enhancers interacting in three-dimensional space with mesendoderm-specifying transcription factor loci identifies MesEndoderm Transcriptional Enhancer Organizing Region (Meteor). Genetic and epigenetic manipulation of the Meteor enhancer reveal its indispensable role during mesendoderm specification and subsequent cardiogenic differentiation via transcription-independent and -dependent mechanisms. Interestingly, Meteor-deleted ESCs are epigenetically redirected towards neuroectodermal lineages. Loci, topologically associating a transcribed enhancer and its cognate protein coding gene, appear to represent therefore a class of genomic elements controlling developmental competence in pluripotency.


Asunto(s)
Ectodermo/fisiología , Células Madre Embrionarias/fisiología , Elementos de Facilitación Genéticos/fisiología , Mesodermo/fisiología , ARN Largo no Codificante/fisiología , Animales , Diferenciación Celular/genética , Línea Celular , Linaje de la Célula/genética , Ectodermo/citología , Perfilación de la Expresión Génica/métodos , Regulación del Desarrollo de la Expresión Génica/fisiología , Humanos , Células Madre Pluripotentes Inducidas , Mesodermo/citología , Ratones , Placa Neural/citología , Placa Neural/fisiología
11.
JACC Basic Transl Sci ; 1(6): 472-493, 2016 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-29707678

RESUMEN

The mechanisms controlling differentiation in adult cardiac precursor cells (CPCs) are still largely unknown. In this study, CPCs isolated from the human heart were found to produce predominantly smooth muscle cells but could be redirected to the cardiomyocyte fate by transient activation followed by inhibition of NOTCH signaling. NOTCH inhibition repressed MIR-143/145 expression, and blocked smooth muscle differentiation. Expression of the microRNAs is under control of CARMEN, a long noncoding RNA associated with an enhancer located in the MIR-143/145 locus and target of NOTCH signaling. The CARMEN/MIR-145/143 axis represents, therefore, a promising target to favor production of cardiomyocytes in cell replacement therapies.

12.
Free Radic Biol Med ; 51(3): 609-18, 2011 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-21620958

RESUMEN

ß(1)-Integrin mediates cardiomyocyte growth and survival and its proper regulation is essential for the structural and functional integrity of the heart. ß(1)-Integrin expression is enhanced in hypertrophy, but the mechanism and significance of its up-regulation are unknown. Because reactive oxygen species (ROS) are important mediators of myocardial remodeling we examined their role in regulated ß(1)-integrin expression. Hypertrophy was induced in neonatal cardiomyocytes by endothelin-1 (ET-1), which activated the regulatory NADPH oxidase subunit Rac1, evoked ROS, and enhanced fetal gene expression and cardiomyocyte size. ET-1 also enhanced cell adhesion and FAK phosphorylation and inhibited oxidative stress-induced cardiomyocyte apoptosis. Further, ET-1 increased ß(1)-integrin mRNA and protein expression via Rac1-ROS-dependent MEK/ERK and EGF receptor-PI3K/Akt activation as shown by adenoviral dominant-negative Rac1 or overexpression of copper/zinc-superoxide dismutase. The relevance of regulated ß(1)-integrin expression was examined in cardiomyocytes, in which targeting siRNA impeded the ET-1-induced ß(1)-integrin up-regulation. In these cells, ET-1-induced cell adhesion, FAK phosphorylation, and hypertrophic response were significantly blunted, whereas its antiapoptotic effect was predominantly unchanged, suggesting at least partial dissociation of prohypertrophic and prosurvival signaling elicited by ET-1. In conclusion, ß(1)-integrin up-regulation in response to ET-1 is mediated via Rac1-ROS-dependent activation of prohypertrophic pathways and is mandatory for ET-1-induced FAK activation, cell adhesion, and hypertrophic response.


Asunto(s)
Hiperplasia/metabolismo , Integrina beta1/metabolismo , Miocardio/patología , Miocitos Cardíacos/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Proteína de Unión al GTP rac1/metabolismo , Animales , Apoptosis/efectos de los fármacos , Apoptosis/genética , Adhesión Celular/efectos de los fármacos , Adhesión Celular/genética , Células Cultivadas , Endotelina-1/farmacología , Regulación del Desarrollo de la Expresión Génica/efectos de los fármacos , Hiperplasia/genética , Integrina beta1/genética , Mutación/genética , Miocitos Cardíacos/efectos de los fármacos , Miocitos Cardíacos/patología , ARN Interferente Pequeño/genética , Ratas , Ratas Wistar , Transducción de Señal/efectos de los fármacos , Transgenes/genética , Regulación hacia Arriba/efectos de los fármacos , Remodelación Ventricular/genética , Proteína de Unión al GTP rac1/genética
13.
Am J Physiol Endocrinol Metab ; 294(2): E241-50, 2008 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-17971516

RESUMEN

Recent evidence supports that TNF-alpha, long considered a catabolic factor, may also have a physiological function in skeletal muscle. The catabolic view, mainly based on correlative studies in human and in vivo animal models, was challenged by experiments with myoblasts, in which TNF-alpha induced differentiation. The biological effects of TNF-alpha in differentiated muscle, however, remain poorly understood. In the present study, we tested whether TNF-alpha has growth-promoting effects in myotubes, and we characterized the mechanisms leading to these effects. Treatment of C(2)C(12) myotubes with TNF-alpha for 24 h increased protein synthesis (PS) and enhanced cellular dehydrogenase activity by 22 and 26%, respectively, without changing cell numbers. These effects were confirmed in myotubes differentiated from primary rat myoblasts. TNF-alpha activated two signaling cascades: 1) ERK1/2 and its target eIF4E and 2) Akt and its downstream effectors GSK-3, p70(S6K), and 4E-BP1. TNF-alpha-induced phosphorylation of Akt, and ERK1/2 was inhibited by an antibody against TNF-alpha receptor 1 (TNF-R1). PD-98059 pretreatment abolished TNF-alpha-induced phosphorylation of ERK1/2 and eIF4E, whereas PS was only partially inhibited. LY-294002 completely abolished TNF-alpha-induced stimulation of PS as well as phosphorylation of Akt and its downstream targets GSK-3, p70(S6K), and 4E-BP1. Rapamycin inhibited TNF-alpha-induced phosphorylation of the mTOR C1 target p70(S6K) without altering TNF-alpha-induced PS and 4E-BP1 phosphorylation. In conclusion, our results provide evidence that TNF-alpha enhances PS in myotubes and that this is based on enhanced protein translation mediated by the TNF-R1 and PI3K-Akt and MEK-ERK signaling cascades.


Asunto(s)
Proteínas Quinasas Activadas por Mitógenos/fisiología , Fibras Musculares Esqueléticas/metabolismo , Proteínas Musculares/biosíntesis , Fosfatidilinositol 3-Quinasas/fisiología , Receptores Tipo I de Factores de Necrosis Tumoral/fisiología , Factor de Necrosis Tumoral alfa/farmacología , Animales , Western Blotting , Línea Celular , Supervivencia Celular/efectos de los fármacos , Ratones , Mitocondrias Musculares/efectos de los fármacos , Mitocondrias Musculares/enzimología , Proteínas Quinasas Activadas por Mitógenos/genética , Fibras Musculares Esqueléticas/efectos de los fármacos , Proteína Oncogénica v-akt/metabolismo , Oxidorreductasas/metabolismo , Fosfatidilinositol 3-Quinasas/genética , Fosforilación , Biosíntesis de Proteínas/efectos de los fármacos , Receptores Tipo I de Factores de Necrosis Tumoral/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transducción de Señal/efectos de los fármacos
14.
J Biol Chem ; 283(45): 30754-65, 2008 Nov 07.
Artículo en Inglés | MEDLINE | ID: mdl-18667438

RESUMEN

Gap junctions are clusters of transmembrane channels allowing a passive diffusion of ions and small molecules between adjacent cells. Connexin43, the main channel-forming protein expressed in ventricular myocytes, can associate with zonula occludens-1, a scaffolding protein linked to the actin cytoskeleton and to signal transduction molecules. The possible influence of Rho GTPases, major regulators of cellular junctions and of the actin cytoskeleton, in the modulation of gap junctional intercellular communication (GJIC) was examined. The activation of RhoA by cytoxic necrotizing factor 1 markedly enhanced GJIC, whereas its specific inhibition by the Clostridium botulinum C3 exoenzyme significantly reduced it. RhoA activity affects GJIC without major cellular redistribution of junctional plaques or changes in the Cx43 phosphorylation pattern. As these GTPases frequently act via the cortical cytoskeleton, the importance of F-actin in the modulation of GJIC was investigated by means of agents interfering with actin polymerization. Cytoskeleton stabilization by phalloidin slowed down the kinetics of channel rundown in the absence of ATP, whereas its disruption by cytochalasin D rapidly and markedly reduced GJIC despite ATP presence. Cytoskeleton stabilization by phalloidin markedly reduced the consequences of RhoA activation or inactivation. This mechanism appears to be the first described capable to both up- or down-regulate GJIC through RhoA activation or, conversely, inhibition. The inhibition of Rho downstream kinase effectors had no effect on GJIC. The present results provide further insight into the gating and regulation of junctional channels and identify a new downstream target for the small G-protein RhoA.


Asunto(s)
Actinas/metabolismo , Permeabilidad de la Membrana Celular/fisiología , Conexina 43/metabolismo , Uniones Comunicantes/metabolismo , Miocitos Cardíacos/metabolismo , Proteína de Unión al GTP rhoA/metabolismo , ADP Ribosa Transferasas/farmacología , Adenosina Trifosfato/metabolismo , Animales , Toxinas Bacterianas/farmacología , Toxinas Botulínicas/farmacología , Permeabilidad de la Membrana Celular/efectos de los fármacos , Citocalasina D/farmacología , Citoesqueleto/metabolismo , Activación Enzimática/efectos de los fármacos , Activación Enzimática/fisiología , Proteínas de Escherichia coli/farmacología , Cinética , Proteínas de la Membrana/metabolismo , Inhibidores de la Síntesis del Ácido Nucleico/farmacología , Faloidina/farmacología , Fosfoproteínas/metabolismo , Fosforilación/efectos de los fármacos , Fosforilación/fisiología , Venenos/farmacología , Ratas , Transducción de Señal/efectos de los fármacos , Transducción de Señal/fisiología , Proteína de la Zonula Occludens-1
15.
Pflugers Arch ; 447(2): 181-94, 2003 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-14504927

RESUMEN

In cardiac myocytes of new-born rats, the degree of intercellular communication through gap junctional channels closely depends on the metabolic state of the cells. In contrast, in stably transfected HeLa cells expressing rat cardiac connexin43 (Cx43, the main channel-forming protein present in ventricular myocytes), a major part of junctional communication persisted in ATP-depleted conditions, in the presence of a metabolic inhibitor (KCN) or of a broad spectrum inhibitor of protein kinases (H7). However, another metabolic inhibitor, antimycin A, which like cyanide inhibits electron transfer in the respiratory chain, totally interrupted cell-to-cell communication between Cx43-HeLa cells, even in whole-cell conditions, when ATP (5 mM) was present. Antimycin A caused a modest increase in cytosolic calcium concentration; however, junctional uncoupling still occurred when this rise was prevented. Conditions of ischemic insult (e.g. ischemia or chemical hypoxia) frequently cause the activation of protein kinases, particularly of Src and MAP kinases, and such activations are known to markedly disrupt gap junctional communication. Antimycin-induced junctional uncoupling occurred even in the presence of inhibitors of these kinases. Antimycin A appears able to cause junctional uncoupling either through the ATP depletion it induces as a metabolic poison or via a direct action on gap junction constituents.


Asunto(s)
Adenosina Trifosfato/fisiología , Antimetabolitos/farmacología , Antimicina A/farmacología , Calcio/fisiología , Comunicación Celular/efectos de los fármacos , Miocitos Cardíacos/fisiología , Adenosina Trifosfato/deficiencia , Animales , Calcio/metabolismo , Células Cultivadas , Conexina 43/metabolismo , Citosol/metabolismo , Activación Enzimática , Inhibidores Enzimáticos/farmacología , Células HeLa/metabolismo , Células HeLa/fisiología , Heptanol/farmacología , Humanos , Modelos Biológicos , Concentración Osmolar , Técnicas de Placa-Clamp , Fosforilación , Cianuro de Potasio/farmacología , Inhibidores de Proteínas Quinasas , Proteínas Quinasas/metabolismo , Ratas , Ratas Wistar
16.
Eur Biophys J ; 33(3): 201-10, 2004 May.
Artículo en Inglés | MEDLINE | ID: mdl-14745523

RESUMEN

Gap junctions, specialized membrane structures that mediate cell-to-cell communication in almost all animal tissues, are composed of channel-forming integral membrane proteins termed connexins. Most of them, particularly connexin43 (Cx43), the most ubiquitous connexin, the major connexin present in cardiac myocytes, are phosphoproteins. Connexin phosphorylation has been thought to regulate gap junctional protein trafficking, gap junction assembly, channel gating, and turnover. Some connexins, including Cx43, show mobility shifts in gel electrophoresis when cells are exposed to phosphorylating or dephosphorylating treatments. However, after exposure of rat cardiac myocytes to different uncoupling dephosphorylating agents such as H7 or butanedione monoxime, no modification in the Cx43 phosphorylation profile was generally observed. The lack of direct correlation between the inhibition of cell-to-cell communication and changes in the phosphorylation pattern of Cx43 or, conversely, modifications of the latter without modifications of the intercellular coupling degree, suggest that the functional state of junctional channels might rather be determined by regulatory proteins associated with Cx43. The modulation of the activity of junctional channels by protein phosphorylation/dephosphorylation processes very likely requires (as for several other membrane channels) the formation of a multiprotein complex, where pore-forming subunits bind to auxiliary proteins (e.g. scaffolding proteins, enzymes, cytoskeleton elements) that play essential roles in channel localization and activity. Such regulatory proteins, behaving as targets for phosphorylation/dephosphorylation catalysers, might in particular control the open probability of junctional channels. A schematic illustration of the regulation of Cx43-made channels by protein phosphorylation involving a partner phosphoprotein is proposed.


Asunto(s)
Conexina 43/metabolismo , Uniones Comunicantes/fisiología , Ventrículos Cardíacos/citología , Canales Iónicos/fisiología , Miocardio/metabolismo , Animales , Encéfalo/fisiología , Comunicación Celular/fisiología , Células Cultivadas , Cucarachas/fisiología , Electrofisiología , Ganglios de Invertebrados/metabolismo , Homeostasis/fisiología , Miocardio/citología , Neuronas/fisiología , Fosforilación , Ratas , Sinapsis/fisiología , Xenopus/fisiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA