Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 24
Filtrar
1.
Genes Dev ; 28(8): 841-57, 2014 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-24682306

RESUMEN

Fibro-adipogenic progenitors (FAPs) are important components of the skeletal muscle regenerative environment. Whether FAPs support muscle regeneration or promote fibro-adipogenic degeneration is emerging as a key determinant in the pathogenesis of muscular diseases, including Duchenne muscular dystrophy (DMD). However, the molecular mechanism that controls FAP lineage commitment and activity is currently unknown. We show here that an HDAC-myomiR-BAF60 variant network regulates the fate of FAPs in dystrophic muscles of mdx mice. Combinatorial analysis of gene expression microarray, genome-wide chromatin remodeling by nuclease accessibility (NA) combined with next-generation sequencing (NA-seq), small RNA sequencing (RNA-seq), and microRNA (miR) high-throughput screening (HTS) against SWI/SNF BAF60 variants revealed that HDAC inhibitors (HDACis) derepress a "latent" myogenic program in FAPs from dystrophic muscles at early stages of disease. Specifically, HDAC inhibition induces two core components of the myogenic transcriptional machinery, MYOD and BAF60C, and up-regulates the myogenic miRs (myomiRs) (miR-1.2, miR-133, and miR-206), which target the alternative BAF60 variants BAF60A and BAF60B, ultimately directing promyogenic differentiation while suppressing the fibro-adipogenic phenotype. In contrast, FAPs from late stage dystrophic muscles are resistant to HDACi-induced chromatin remodeling at myogenic loci and fail to activate the promyogenic phenotype. These results reveal a previously unappreciated disease stage-specific bipotency of mesenchimal cells within the regenerative environment of dystrophic muscles. Resolution of such bipotency by epigenetic intervention with HDACis provides a molecular rationale for the in situ reprogramming of target cells to promote therapeutic regeneration of dystrophic muscles.


Asunto(s)
Histona Desacetilasas/metabolismo , MicroARNs/metabolismo , Músculo Esquelético/fisiología , Distrofias Musculares/genética , Distrofias Musculares/fisiopatología , Células Madre/metabolismo , Animales , Reprogramación Celular/genética , Cromatina/genética , Ensamble y Desensamble de Cromatina/fisiología , Proteínas Cromosómicas no Histona/genética , Proteínas Cromosómicas no Histona/metabolismo , Perfilación de la Expresión Génica , Regulación de la Expresión Génica/efectos de los fármacos , Inhibidores de Histona Desacetilasas/farmacología , Histona Desacetilasas/genética , Ácidos Hidroxámicos/farmacología , Ratones , Ratones Endogámicos mdx , Proteínas Musculares/genética , Proteínas Musculares/metabolismo
2.
RNA ; 18(3): 472-84, 2012 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-22282338

RESUMEN

In order to understand the role of microRNAs (miRNAs) in vascular physiopathology, we took advantage of deep-sequencing techniques to accurately and comprehensively profile the entire miRNA population expressed by endothelial cells exposed to hypoxia. SOLiD sequencing of small RNAs derived from human umbilical vein endothelial cells (HUVECs) exposed to 1% O2 or normoxia for 24 h yielded more than 22 million reads per library. A customized bioinformatic pipeline identified more than 400 annotated microRNA/microRNA* species with a broad abundance range: miR-21 and miR-126 totaled almost 40% of all miRNAs. A complex repertoire of isomiRs was found, displaying also 5' variations, potentially affecting target recognition. High-stringency bioinformatic analysis identified microRNA candidates, whose predicted pre-miRNAs folded into a stable hairpin. Validation of a subset by qPCR identified 18 high-confidence novel miRNAs as detectable in independent HUVEC cultures and associated to the RISC complex. The expression of two novel miRNAs was significantly down-modulated by hypoxia, while miR-210 was significantly induced. Gene ontology analysis of their predicted targets revealed a significant association to hypoxia-inducible factor signaling, cardiovascular diseases, and cancer. Overexpression of the novel miRNAs in hypoxic endothelial cells affected cell growth and confirmed the biological relevance of their down-modulation. In conclusion, deep-sequencing accurately profiled known, variant, and novel microRNAs expressed by endothelial cells in normoxia and hypoxia.


Asunto(s)
Células Endoteliales/metabolismo , Secuenciación de Nucleótidos de Alto Rendimiento , MicroARNs/análisis , MicroARNs/química , Carboxipeptidasas/metabolismo , Hipoxia de la Célula , Proliferación Celular , Perfilación de la Expresión Génica , Regulación de la Expresión Génica , Biblioteca de Genes , Células HEK293 , Humanos , MicroARNs/metabolismo , Anotación de Secuencia Molecular , Conformación de Ácido Nucleico , ARN Bicatenario , Análisis de Secuencia de ARN , Transducción de Señal
3.
Mol Ther ; 21(10): 1841-51, 2013 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-23760446

RESUMEN

Exogenous high-mobility group box 1 protein (HMGB1) administration to the mouse heart, during acute myocardial infarction (MI), results in cardiac regeneration via resident c-kit(+) cell (CPC) activation. Aim of the present study was to identify the molecular pathways involved in HMGB1-induced heart repair. Gene expression profiling was performed to identify differentially expressed genes in the infarcted and bordering regions of untreated and HMGB1-treated mouse hearts, 3 days after MI. Functional categorization of the transcripts, accomplished using Ingenuity Pathway Analysis software (IPA), revealed that genes involved in tissue regeneration, that is, cardiogenesis, vasculogenesis and angiogenesis, were present both in the infarcted area and in the peri-infarct zone; HMGB1 treatment further increased the expression of these genes. IPA revealed the involvement of Notch signaling pathways in HMGB1-treated hearts. Importantly, HMGB1 determined a 35 and 58% increase in cardiomyocytes and CPCs expressing Notch intracellular cytoplasmic domain, respectively. Further, Notch inhibition by systemic treatment with the γ-secretase inhibitor DAPT, which blocked the proteolytic activation of Notch receptors, reduced the number of CPCs, their proliferative fraction, and cardiomyogenic differentiation in HMGB1-treated infarcted hearts. The present study gives insight into the molecular processes involved in HMGB1-mediated cardiac regeneration and indicates Notch signaling as a key player.


Asunto(s)
Perfilación de la Expresión Génica , Proteína HMGB1/farmacología , Infarto del Miocardio/metabolismo , Miocardio/metabolismo , Receptores Notch/metabolismo , Regeneración/genética , Transducción de Señal , Animales , Diferenciación Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Células Cultivadas , Femenino , Proteína HMGB1/administración & dosificación , Corazón/efectos de los fármacos , Ratones , Ratones Endogámicos C57BL , Infarto del Miocardio/fisiopatología , Miocitos Cardíacos/efectos de los fármacos , Miocitos Cardíacos/metabolismo , Regeneración/efectos de los fármacos , Transducción de Señal/efectos de los fármacos
4.
J Biol Chem ; 287(53): 44761-71, 2012 Dec 28.
Artículo en Inglés | MEDLINE | ID: mdl-23148210

RESUMEN

MicroRNA-210 (miR-210) induction is a virtually constant feature of the hypoxic response in both normal and transformed cells, regulating several key aspects of cardiovascular diseases and cancer. We found that miR-210 was induced in normoxic myoblasts upon myogenic differentiation both in vitro and in vivo. miR-210 transcription was activated in an hypoxia-inducible factor 1-α (Hif1a)-dependent manner, and chromatin immunoprecipitation experiments show that Hif1a bound to the miR-210 promoter only in differentiated myotubes. Accordingly, luciferase reporter assays demonstrated the functional relevance of the Hif1a binding site for miR-210 promoter activation in differentiating myoblasts. To investigate the functional relevance of increased miR-210 levels in differentiated myofibers, we blocked miR-210 with complementary locked nucleic acid oligonucleotides (anti-miR-210). We found that C2C12 myoblast cell line differentiation was largely unaffected by anti-miR-210. Likewise, miR-210 inhibition did not affect skeletal muscle regeneration following cardiotoxin damage. However, we found that miR-210 blockade greatly increased myotube sensitivity to oxidative stress and mitochondrial dysfunction. In conclusion, miR-210 is induced in normoxic myofibers, playing a cytoprotective role.


Asunto(s)
Diferenciación Celular , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , MicroARNs/genética , Mioblastos/citología , Mioblastos/metabolismo , Oxígeno/metabolismo , Animales , Secuencia de Bases , Línea Celular , Subunidad alfa del Factor 1 Inducible por Hipoxia/genética , Masculino , Ratones , MicroARNs/metabolismo , Datos de Secuencia Molecular , Regiones Promotoras Genéticas
5.
Circulation ; 122(11 Suppl): S124-31, 2010 Sep 14.
Artículo en Inglés | MEDLINE | ID: mdl-20837903

RESUMEN

BACKGROUND: MicroRNAs are involved in various critical functions, including the regulation of cellular differentiation, proliferation, angiogenesis, and apoptosis. We hypothesize that microRNA-210 can rescue cardiac function after myocardial infarction by upregulation of angiogenesis and inhibition of cellular apoptosis in the heart. METHODS AND RESULTS: Using microRNA microarrays, we first showed that microRNA-210 was highly expressed in live mouse HL-1 cardiomyocytes compared with apoptotic cells after 48 hours of hypoxia exposure. We confirmed by polymerase chain reaction that microRNA-210 was robustly induced in these cells. Gain-of-function and loss-of-function approaches were used to investigate microRNA-210 therapeutic potential in vitro. After transduction, microRNA-210 can upregulate several angiogenic factors, inhibit caspase activity, and prevent cell apoptosis compared with control. Afterward, adult FVB mice underwent intramyocardial injections with minicircle vector carrying microRNA-210 precursor, minicircle carrying microRNA-scramble, or sham surgery. At 8 weeks, echocardiography showed a significant improvement of left ventricular fractional shortening in the minicircle vector carrying microRNA-210 precursor group compared with the minicircle carrying microRNA-scramble control. Histological analysis confirmed decreased cellular apoptosis and increased neovascularization. Finally, 2 potential targets of microRNA-210, Efna3 and Ptp1b, involved in angiogenesis and apoptosis were confirmed through additional experimental validation. CONCLUSIONS: MicroRNA-210 can improve angiogenesis, inhibit apoptosis, and improve cardiac function in a murine model of myocardial infarction. It represents a potential novel therapeutic approach for treatment of ischemic heart disease.


Asunto(s)
Apoptosis , Terapia Genética/métodos , MicroARNs/biosíntesis , Infarto del Miocardio/terapia , Miocitos Cardíacos/metabolismo , Neovascularización Fisiológica , Animales , Hipoxia de la Célula/genética , Línea Celular , Modelos Animales de Enfermedad , Femenino , Perfilación de la Expresión Génica , Regulación de la Expresión Génica/genética , Humanos , Ratones , MicroARNs/genética , Infarto del Miocardio/genética , Infarto del Miocardio/metabolismo , Miocitos Cardíacos/patología , Análisis de Secuencia por Matrices de Oligonucleótidos , Proteína Tirosina Fosfatasa no Receptora Tipo 1/biosíntesis , Proteína Tirosina Fosfatasa no Receptora Tipo 1/genética , Factores de Tiempo , Transducción Genética/métodos , Función Ventricular Izquierda/genética
6.
J Biol Chem ; 284(50): 35134-43, 2009 Dec 11.
Artículo en Inglés | MEDLINE | ID: mdl-19826008

RESUMEN

miR-210 is a key player of cell response to hypoxia, modulating cell survival, VEGF-driven endothelial cell migration, and the ability of endothelial cells to form capillary-like structures. A crucial step in understanding microRNA (miRNA) function is the identification of their targets. However, only few miR-210 targets have been identified to date. Here, we describe an integrated strategy for large-scale identification of new miR-210 targets by combining transcriptomics and proteomics with bioinformatic approaches. To experimentally validate candidate targets, the RNA-induced silencing complex (RISC) loaded with miR-210 was purified by immunoprecipitation along with its mRNA targets. The complex was significantly enriched in mRNAs of 31 candidate targets, such as BDNF, GPD1L, ISCU, NCAM, and the non-coding RNA Xist. A subset of the newly identified targets was further confirmed by 3'-untranslated region (UTR) reporter assays, and hypoxia induced down-modulation of their expression was rescued blocking miR-210, providing support for the approach validity. In the case of 9 targets, such as PTPN1 and P4HB, miR-210 seed-pairing sequences localized in the coding sequence or in the 5'-UTR, in line with recent data extending miRNA targeting beyond the "classic" 3'-UTR recognition. Finally, Gene Ontology analysis of the targets highlights known miR-210 impact on cell cycle regulation and differentiation, and predicts a new role of this miRNA in RNA processing, DNA binding, development, membrane trafficking, and amino acid catabolism. Given the complexity of miRNA actions, we view such a multiprong approach as useful to adequately describe the multiple pathways regulated by miR-210 during physiopathological processes.


Asunto(s)
Regulación de la Expresión Génica , Hipoxia/metabolismo , MicroARNs/metabolismo , Animales , Secuencia de Bases , Células Cultivadas , Análisis por Conglomerados , Perfilación de la Expresión Génica , Humanos , MicroARNs/genética , Proteoma/análisis , Proteómica/métodos
7.
FASEB J ; 23(10): 3335-46, 2009 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-19528256

RESUMEN

The aim of this work was to identify micro-RNAs (miRNAs) involved in the pathological pathways activated in skeletal muscle damage and regeneration by both dystrophin absence and acute ischemia. Eleven miRNAs were deregulated both in MDX mice and in Duchenne muscular dystrophy patients (DMD signature). Therapeutic interventions ameliorating the mdx-phenotype rescued DMD-signature alterations. The significance of DMD-signature changes was characterized using a damage/regeneration mouse model of hind-limb ischemia and newborn mice. According to their expression, DMD-signature miRNAs were divided into 3 classes. 1) Regeneration miRNAs, miR-31, miR-34c, miR-206, miR-335, miR-449, and miR-494, which were induced in MDX mice and in DMD patients, but also in newborn mice and in newly formed myofibers during postischemic regeneration. Notably, miR-206, miR-34c, and miR-335 were up-regulated following myoblast differentiation in vitro. 2) Degenerative-miRNAs, miR-1, miR-29c, and miR-135a, that were down-modulated in MDX mice, in DMD patients, in the degenerative phase of the ischemia response, and in newborn mice. Their down-modulation was linked to myofiber loss and fibrosis. 3) Inflammatory miRNAs, miR-222 and miR-223, which were expressed in damaged muscle areas, and their expression correlated with the presence of infiltrating inflammatory cells. These findings show an important role of miRNAs in physiopathological pathways regulating muscle response to damage and regeneration.


Asunto(s)
Isquemia/metabolismo , MicroARNs/biosíntesis , Músculo Esquelético/irrigación sanguínea , Músculo Esquelético/metabolismo , Distrofia Muscular de Duchenne/metabolismo , Regeneración , Animales , Humanos , Isquemia/patología , Ratones , Ratones Endogámicos mdx , Músculo Esquelético/patología , Distrofia Muscular de Duchenne/patología
8.
FASEB J ; 20(8): 1242-4, 2006 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-16603604

RESUMEN

The understanding of endothelial cell responses to oxidative stress may provide insights into aging mechanisms and into the pathogenesis of numerous cardiovascular diseases. In this study, we examined the regulation and the functional role of cyclin D1, a crucial player in cell proliferation and survival. On H2O2 treatment, endothelial cells showed a rapid down-modulation of cyclin D1. Other D-cyclins were similarly regulated, and this decrease was also observed after exposure to other oxidative stress-inducing stimuli, namely 1,3-bis (2 chloroethyl)-1 nitrosourea treatment and ischemia. H2O2 treatment induced cyclin D1 ubiquitination followed by proteasome degradation. Phospholipase C inhibition prevented cyclin D1 degradation, and its activation triggered cyclin D1 down-modulation in the absence of oxidative stress. Activated phospholipase C generates inositol-1,4,5-trisphosphate (IP3) and Ca2+ release from internal stores. We found that both IP3-receptor inhibition and intracellular Ca2+ chelation prevented cyclin D1 degradation induced by oxidative stress. Furthermore, Ca2+ increase was transduced by Ca2+/calmodulin-dependent protein kinase (CaMK). In fact, H2O2 stimulated CaMK activity, CaMK inhibitors prevented H2O2-induced cyclin D1 down-modulation, and CaMK overexpression induced cyclin D1 degradation. Finally, overriding of cyclin D1 down-modulation via its forced overexpression or via CaMK inhibition increased cell sensitivity to H2O2-induced apoptotic cell death. Thus, cyclin D1 degradation enhances endothelial cell survival on oxidative stress.


Asunto(s)
Ciclina D1/metabolismo , Endotelio Vascular/metabolismo , Estrés Oxidativo , Apoptosis , Calcio/metabolismo , Proteínas Quinasas Dependientes de Calcio-Calmodulina/antagonistas & inhibidores , Proteínas Quinasas Dependientes de Calcio-Calmodulina/metabolismo , Supervivencia Celular , Regulación hacia Abajo , Células Endoteliales/efectos de los fármacos , Células Endoteliales/metabolismo , Endotelio Vascular/citología , Endotelio Vascular/efectos de los fármacos , Humanos , Peróxido de Hidrógeno/farmacología , Complejo de la Endopetidasa Proteasomal/metabolismo , Fosfolipasas de Tipo C/metabolismo , Ubiquitina/metabolismo
9.
Circulation ; 109(23): 2917-23, 2004 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-15173034

RESUMEN

BACKGROUND: Oxidative stress plays a pivotal role in ischemia and ischemia/reperfusion injury. Because p66(ShcA)-null (p66(ShcA)-/-) mice exhibit both lower levels of intracellular reactive oxygen species and increased resistance to cell death induced by oxidative stress, we investigated whether tissue damage that follows acute ischemia or ischemia/reperfusion was altered in p66(ShcA)-/- mice. METHODS AND RESULTS: Unilateral hindlimb ischemia was induced by femoral artery dissection, and ischemia/reperfusion was induced with an elastic tourniquet. Both procedures caused similar changes in blood perfusion in p66(ShcA) wild-type (p66(ShcA)wt) and p66(ShcA)-/- mice. However, significant differences in tissue damage were found: p66(ShcA)wt mice displayed marked capillary density decrease and muscle fiber necrosis. In contrast, in p66(ShcA)-/- mice, minimal capillary density decrease and myofiber death were present. When apoptosis after ischemia was assayed, significantly lower levels of apoptotic endothelial cells and myofibers were found in p66(ShcA)-/- mice. In agreement with these data, both satellite muscle cells and endothelial cells isolated from p66(ShcA)-/- mice were resistant to apoptosis induced by simulated ischemia in vitro. Lower apoptosis levels after ischemia in p66(ShcA)-/- cells correlated with decreased levels of oxidative stress both in vivo and in vitro. CONCLUSIONS: p66(ShcA) plays a crucial role in the cell death pathways activated by acute ischemia and ischemia/reperfusion, indicating p66(ShcA) as a potential therapeutic target for prevention and treatment of ischemic tissue damage.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/fisiología , Miembro Posterior/irrigación sanguínea , Isquemia/fisiopatología , Proteínas Adaptadoras Transductoras de Señales/deficiencia , Proteínas Adaptadoras Transductoras de Señales/genética , Animales , Apoptosis , Capilares/patología , Células Cultivadas/efectos de los fármacos , Células Endoteliales/efectos de los fármacos , Isquemia/patología , Masculino , Ratones , Ratones Noqueados , Células Musculares/efectos de los fármacos , Fibras Musculares Esqueléticas/patología , Necrosis , Estrés Oxidativo , Fosforilación , Procesamiento Proteico-Postraduccional , Especies Reactivas de Oxígeno , Daño por Reperfusión/fisiopatología , Proteínas Adaptadoras de la Señalización Shc , Proteína Transformadora 1 que Contiene Dominios de Homología 2 de Src , Sustancias Reactivas al Ácido Tiobarbitúrico/análisis , Factor A de Crecimiento Endotelial Vascular/biosíntesis , Factor A de Crecimiento Endotelial Vascular/genética
10.
Curr Vasc Pharmacol ; 13(3): 305-15, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-23713865

RESUMEN

microRNAs (miRNAs), small non-coding RNA molecules that act as negative regulators of gene expression, are involved in a wide range of biological functions and control several cellular processes. This review illustrates miRNA regulation and function in tissue response to acute ischemia, focusing on miRNA role in acute myocardial infarction and describing a subset of miRNAs de-regulated upon cardiac ischemia. These miRNAs may represent "master ischemic" miRNAs, playing a pathogenetic role in one of the different components of tissue response to ischemia. Moreover, circulating miRNAs correlated to myocardial infarction and examples of miRNA involvement in ischemic diseases different from cardiac ischemia are also discussed. The identification of specific miRNAs as key regulators of cell biology has opened new clinical avenues, and may allow new diagnostic and/or prognostic tools development, as much as innovative therapeutic strategies. Two paradigmatic reports, in which miRNAs have been targeted to improve cardiac function in pre-clinical models of myocardial infarction, are described in detail and confirmed the efficacy of these strategies.


Asunto(s)
Biomarcadores/análisis , MicroARNs/efectos de los fármacos , MicroARNs/metabolismo , Isquemia Miocárdica/diagnóstico , Isquemia Miocárdica/tratamiento farmacológico , Síndrome Coronario Agudo/tratamiento farmacológico , Animales , Humanos
11.
Int J Cardiol ; 197: 333-47, 2015 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-26159041

RESUMEN

BACKGROUND: We recently demonstrated that epicardial progenitor cells participate in the regenerative response to myocardial infarction (MI) and factors released in the pericardial fluid (PF) may play a key role in this process. Exosomes are secreted nanovesicles of endocytic origin, identified in most body fluids, which may contain molecules able to modulate a variety of cell functions. Here, we investigated whether exosomes are present in the PF and their potential role in cardiac repair. METHODS AND RESULTS: Early gene expression studies in 3day-infarcted mouse hearts showed that PF induces epithelial-to-mesenchymal transition (EMT) in epicardial cells. Exosomes were identified in PFs from non-infarcted patients (PFC) and patients with acute MI (PFMI). A shotgun proteomics analysis identified clusterin in exosomes isolated from PFMI but not from PFC. Notably, clusterin has a protective effect on cardiomyocytes after acute MI in vivo and is an important mediator of TGFß-induced. Clusterin addition to the pericardial sac determined an increase in epicardial cells expressing the EMT marker α-SMA and, interestingly, an increase in the number of epicardial cells ckit(+)/α-SMA(+), 7days following MI. Importantly, clusterin treatment enhanced arteriolar length density and lowered apoptotic rates in the peri-infarct area. Hemodynamic studies demonstrated an improvement in cardiac function in clusterin-treated compared to untreated infarcted hearts. CONCLUSIONS: Exosomes are present and detectable in the PFs. Clusterin was identified in PFMI-exosomes and might account for an improvement in myocardial performance following MI through a framework including EMT-mediated epicardial activation, arteriogenesis and reduced cardiomyocyte apoptosis.


Asunto(s)
Clusterina/metabolismo , Vasos Coronarios/metabolismo , Exosomas/metabolismo , Infarto del Miocardio/metabolismo , Líquido Pericárdico/metabolismo , Pericardio/metabolismo , Anciano , Anciano de 80 o más Años , Animales , Apoptosis/fisiología , Biomarcadores/análisis , Biomarcadores/metabolismo , Clusterina/análisis , Vasos Coronarios/química , Exosomas/química , Femenino , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Persona de Mediana Edad , Infarto del Miocardio/diagnóstico , Miocardio/química , Miocardio/metabolismo , Miocardio/patología , Líquido Pericárdico/química , Pericardio/química , Pericardio/patología
12.
J Dermatol Sci ; 73(1): 3-9, 2014 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-24012494

RESUMEN

The wound healing process is characterized by a series of overlapping phases, such as coagulation, inflammation, reepithelialization/granulation tissue generation and remodeling. It is important to obtain a deeper insight into the cutaneous wound repair mechanisms, in order to develop novel pharmacological tools for the treatment of chronic non-healing ulcers which are a frequent and high morbidity complication of diabetes, ischaemia, venous insufficiency, and other local or systemic factors. Several transcription factors, many of which belong to gene families, are known to play a role in cutaneous wound repair through the orchestration of cellular responses which promote the reconstitution of skin integrity. The aim of this review is to provide an updated analysis of the transcription factor role in the reepithelialization process, in the context of skin wound repair.


Asunto(s)
Repitelización/genética , Piel/metabolismo , Factores de Transcripción/metabolismo , Transcripción Genética , Animales , Regulación de la Expresión Génica , Redes Reguladoras de Genes , Humanos , Piel/patología , Piel/fisiopatología , Factores de Transcripción/genética
13.
Antioxid Redox Signal ; 21(8): 1177-88, 2014 Sep 10.
Artículo en Inglés | MEDLINE | ID: mdl-23931770

RESUMEN

AIMS: Peripheral artery disease is caused by the restriction or occlusion of arteries supplying the leg. Better understanding of the molecular mechanisms underpinning tissue response to ischemia is urgently needed to improve therapeutic options. The aim of this study is to investigate hypoxia-induced miR-210 regulation and its role in a mouse model of hindlimb ischemia. RESULTS: miR-210 expression was induced by femoral artery dissection. To study the role of miR-210, its function was inhibited by the systemic administration of a miR-210 complementary locked nucleic acid (LNA)-oligonucleotide (anti-miR-210). In the ischemic skeletal muscle, anti-miR-210 caused a marked decrease of miR-210 compared with LNA-scramble control, while miR-210 target expression increased accordingly. Histological evaluation of acute tissue damage showed that miR-210 inhibition increased both apoptosis at 1 day and necrosis at 3 days. Capillary density decrease caused by ischemia was significantly more pronounced in anti-miR-210-treated mice; residual limb perfusion decreased accordingly. To investigate the molecular mechanisms underpinning the increased damage triggered by miR-210 blockade, we tested the impact of anti-miR-210 treatment on the transcriptome. Gene expression analysis highlighted the deregulation of mitochondrial function and redox balance. Accordingly, oxidative damage was more severe in the ischemic limb of anti-miR-210-treated mice and miR-210 inhibition increased oxidative metabolism. Further, oxidative-stress resistant p66(Shc)-null mice displayed decreased tissue damage following ischemia. INNOVATION: This study identifies miR-210 as a crucial element in the adaptive mechanisms to acute peripheral ischemia. CONCLUSIONS: The physiopathological significance of miR-210 is context dependent. In the ischemic skeletal muscle it seems to be cytoprotective, regulating oxidative metabolism and oxidative stress.


Asunto(s)
Isquemia/metabolismo , MicroARNs/fisiología , Enfermedad Aguda , Animales , Apoptosis , Hipoxia de la Célula , Línea Celular , Expresión Génica , Glucólisis , Miembro Posterior/irrigación sanguínea , Isquemia/genética , Masculino , Ratones de la Cepa 129 , Ratones Noqueados , Músculo Esquelético/irrigación sanguínea , Músculo Esquelético/metabolismo , Músculo Esquelético/patología , Estrés Oxidativo , Interferencia de ARN
14.
PLoS One ; 9(4): e93983, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24722564

RESUMEN

Myotonic dystrophy type 2 (DM2) is a genetic, autosomal dominant disease due to expansion of tetraplet (CCTG) repetitions in the first intron of the ZNF9/CNBP gene. DM2 is a multisystemic disorder affecting the skeletal muscle, the heart, the eye and the endocrine system. According to the proposed pathological mechanism, the expanded tetraplets have an RNA toxic effect, disrupting the splicing of many mRNAs. Thus, the identification of aberrantly spliced transcripts is instrumental for our understanding of the molecular mechanisms underpinning the disease. The aim of this study was the identification of new aberrant alternative splicing events in DM2 patients. By genome wide analysis of 10 DM2 patients and 10 controls (CTR), we identified 273 alternative spliced exons in 218 genes. While many aberrant splicing events were already identified in the past, most were new. A subset of these events was validated by qPCR assays in 19 DM2 and 15 CTR subjects. To gain insight into the molecular pathways involving the identified aberrantly spliced genes, we performed a bioinformatics analysis with Ingenuity system. This analysis indicated a deregulation of development, cell survival, metabolism, calcium signaling and contractility. In conclusion, our genome wide analysis provided a database of aberrant splicing events in the skeletal muscle of DM2 patients. The affected genes are involved in numerous pathways and networks important for muscle physio-pathology, suggesting that the identified variants may contribute to DM2 pathogenesis.


Asunto(s)
Empalme Alternativo , Genoma Humano , Distrofia Miotónica/genética , Proteínas Adaptadoras Transductoras de Señales/genética , Biopsia , Señalización del Calcio , Supervivencia Celular , Biología Computacional , Exones , Femenino , Proteínas Fetales/genética , Forminas , Regulación de la Expresión Génica , Humanos , Proteínas con Dominio LIM/genética , Masculino , Persona de Mediana Edad , Músculo Esquelético/metabolismo , Factores de Transcripción NFI/genética , Proteínas Nucleares/genética , Reacción en Cadena de la Polimerasa , ARN/metabolismo , Proteínas de Unión al ARN/genética , Proteínas tau/genética
15.
PLoS One ; 7(9): e44651, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-23024754

RESUMEN

Most metazoan microRNA (miRNA) target sites have perfect pairing to the "seed" sequence, a highly conserved region centering on miRNA nucleotides 2-7. Thus, complementarity to this region is a necessary requirement for target prediction algorithms. However, also non-canonical miRNA binding can confer target regulation. Here, we identified a seedless target of miR-210, a master miRNA of the hypoxic response. We analyzed 20 genes that were inversely correlated to miR-210 expression and did not display any complementarity with miR-210 seed sequence. We validated ROD1 (Regulator of Differentiation 1, also named PTBP3, Polypyrimidine Tract Binding protein 3) as a miR-210 seedless transcript enriched in miR-210-containing RNA-induced silencing complexes. ROD1 was not indirectly targeted by a miR-210-induced miRNA. Conversely, we identified a "centered" miR-210 binding site in ROD1 involving 10 consecutive bases in the central portion of miR-210. Reporter assays showed that miR-210 inhibited ROD1 by the direct binding to this sequence, demonstrating that ROD1 is a bona fide seedless target of miR-210. As expected, both ROD1 mRNA and protein were down-modulated upon hypoxia in a miR-210 dependent manner. ROD1 targeting by miR-210 was biologically significant: the rescue of ROD1 inhibition significantly increased hypoxia-induced cell death. These data highlight the importance of ROD1 regulation by miR-210 for cell homeostasis.


Asunto(s)
Regulación de la Expresión Génica , Hipoxia , MicroARNs/genética , Proteína de Unión al Tracto de Polipirimidina/genética , Apoptosis , Emparejamiento Base , Secuencia de Bases , Sitios de Unión , Supervivencia Celular/genética , Células HEK293 , Humanos , MicroARNs/química , Proteína de Unión al Tracto de Polipirimidina/química , Interferencia de ARN , Reproducibilidad de los Resultados , Transcripción Genética
16.
PLoS One ; 7(6): e39732, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22768114

RESUMEN

Myotonic Dystrophy Type-2 (DM2) is an autosomal dominant disease caused by the expansion of a CCTG tetraplet repeat. It is a multisystemic disorder, affecting skeletal muscles, the heart, the eye, the central nervous system and the endocrine system. Since microRNA (miRNA) expression is disrupted in Myotonic Dystrophy Type-1 and many other myopathies, miRNAs deregulation was studied in skeletal muscle biopsies of 13 DM2 patients and 13 controls. Eleven miRNAs were deregulated: 9 displayed higher levels compared to controls (miR-34a-5p, miR-34b-3p, miR-34c-5p, miR-146b-5p, miR-208a, miR-221-3p and miR-381), while 4 were decreased (miR-125b-5p, miR-193a-3p, miR-193b-3p and miR-378a-3p). To explore the relevance of DM2 miRNA deregulation, the predicted interactions between miRNA and mRNA were investigated. Global gene expression was analyzed in DM2 and controls and bioinformatic analysis identified more than 1,000 miRNA/mRNA interactions. Pathway and function analysis highlighted the involvement of the miRNA-deregulated mRNAs in multiple aspects of DM2 pathophysiology. In conclusion, the observed miRNA dysregulations may contribute to DM2 pathogenetic mechanisms.


Asunto(s)
Regulación de la Expresión Génica , MicroARNs/genética , Trastornos Miotónicos/genética , Atrofia , Estudios de Casos y Controles , Estudios de Cohortes , Femenino , Perfilación de la Expresión Génica , Redes Reguladoras de Genes/genética , Humanos , Hipertrofia , Masculino , MicroARNs/metabolismo , Persona de Mediana Edad , Fibras Musculares Esqueléticas/metabolismo , Fibras Musculares Esqueléticas/patología , Distrofia Miotónica , ARN Mensajero/genética , ARN Mensajero/metabolismo , Reproducibilidad de los Resultados , Transcriptoma/genética
17.
Diabetes ; 61(6): 1633-41, 2012 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-22427379

RESUMEN

Increased morbidity and mortality associated with ischemic heart failure (HF) in type 2 diabetic patients requires a deeper understanding of the underpinning pathogenetic mechanisms. Given the implication of microRNAs (miRNAs) in HF, we investigated their regulation and potential role. miRNA expression profiles were measured in left ventricle biopsies from 10 diabetic HF (D-HF) and 19 nondiabetic HF (ND-HF) patients affected by non-end stage dilated ischemic cardiomyopathy. The HF groups were compared with each other and with 16 matched nondiabetic, non-HF control subjects. A total of 17 miRNAs were modulated in D-HF and/or ND-HF patients when compared with control subjects. miR-216a, strongly increased in both D-HF and ND-HF patients, negatively correlated with left ventricular ejection fraction. Six miRNAs were differently expressed when comparing D-HF and ND-HF patients: miR-34b, miR-34c, miR-199b, miR-210, miR-650, and miR-223. Bioinformatic analysis of their modulated targets showed the enrichment of cardiac dysfunctions and HF categories. Moreover, the hypoxia-inducible factor pathway was activated in the noninfarcted, vital myocardium of D-HF compared with ND-HF patients, indicating a dysregulation of the hypoxia response mechanisms. Accordingly, miR-199a, miR-199b, and miR-210 were modulated by hypoxia and high glucose in cardiomyocytes and endothelial cells cultured in vitro. In conclusion, these findings show a dysregulation of miRNAs in HF, shedding light on the specific disease mechanisms differentiating diabetic patients.


Asunto(s)
Diabetes Mellitus Tipo 2/genética , Cardiomiopatías Diabéticas/genética , Insuficiencia Cardíaca/genética , MicroARNs/genética , Isquemia Miocárdica/genética , Anciano , Diabetes Mellitus Tipo 2/metabolismo , Cardiomiopatías Diabéticas/metabolismo , Femenino , Insuficiencia Cardíaca/metabolismo , Humanos , Masculino , MicroARNs/metabolismo , Persona de Mediana Edad , Isquemia Miocárdica/metabolismo , Miocardio/metabolismo , Miocitos Cardíacos/metabolismo
18.
Cardiovasc Res ; 85(2): 272-80, 2010 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-19617222

RESUMEN

Degradation of poly-ubiquitinated proteins by the 26S-proteasome complex represents a crucial quantitative control mechanism. The ubiquitin-proteasome system (UPS) plays a pivotal role in the complex molecular network regulating the progression both between and within each cell-cycle phase. Two major complexes are involved: the SKP1-CUL1-F-box-protein complex (SCF) and the anaphase-promoting complex/cyclosome (APC/C). Notwithstanding structural similarities, SCF and APC/C display different cellular functions and mechanisms of action. SCF modulates all cell-cycle stages and plays a prominent role at G1/S transition mainly through three regulatory subunits: Skp2, Fbw7, and beta-TRCP. APC/C, regulated by Cdc20 or Cdh1 subunits, has a crucial role in mitosis. In this review, we will describe how the endothelial cell cycle is regulated by the UPS. We will illustrate the principal SCF- and APC/C-dependent molecular mechanisms that modulate cell growth, allowing a unidirectional cell-cycle progression. Then, we will focus our attention on UPS modulation by oxidative stress, a pathogenic stimulus that causes endothelial dysfunction and is involved in numerous cardiovascular diseases.


Asunto(s)
Ciclo Celular , Células Endoteliales/citología , Complejo de la Endopetidasa Proteasomal/fisiología , Proteínas Ligasas SKP Cullina F-box/fisiología , Complejos de Ubiquitina-Proteína Ligasa/fisiología , Ubiquitina/metabolismo , Ciclosoma-Complejo Promotor de la Anafase , Animales , Enfermedades Cardiovasculares/etiología , Humanos , Factor 2 Relacionado con NF-E2/fisiología , Estrés Oxidativo , Proteína p53 Supresora de Tumor/fisiología
19.
Pharmacol Ther ; 125(1): 92-104, 2010 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-19896977

RESUMEN

microRNAs (miRNAs) are 21-23-nucleotide non-protein-coding RNA molecules that act as negative regulators of gene expression, modulating the stability and/or the translational efficiency of target messenger RNAs. This review describes miRNA regulation and function in tissue response to acute ischemia. We focused our attention on a subset of miRNAs that have been found de-regulated in different studies, suggesting that they may represent "master ischemic" miRNAs, playing a pathogenetic role in different components of tissue response to ischemia. First, we analyzed the role of miRNAs in cell response to hypoxia, a crucial component of ischemia, and in angiogenesis. Then, we describe miRNAs role in acute myocardial infarction as much as in hindlimb, cerebral, hepatic and retinal ischemia. The role played by specific miRNAs in the regulation of apoptosis, fibrosis, regeneration and myocardial arrhythmias is illustrated. The identification of specific miRNAs as key regulators of the response to ischemia has opened new clinical avenues. miRNAs may constitute excellent non-invasive disease biomarkers. Furthermore, innovative strategies targeting miRNAs, aimed to reduce the levels of pathogenic or aberrantly expressed miRNAs or to elevate the levels of miRNAs with beneficial functions, have been developed and could be applied in the treatment of ischemic diseases.


Asunto(s)
Terapia Genética , Isquemia/terapia , MicroARNs/metabolismo , Enfermedad Aguda , Animales , Marcadores Genéticos , Pruebas Genéticas , Humanos , Hipoxia/genética , Isquemia/genética , Isquemia/patología , Isquemia/fisiopatología , Neovascularización Fisiológica/genética , Estabilidad del ARN
20.
PLoS One ; 4(10): e7607, 2009 Oct 27.
Artículo en Inglés | MEDLINE | ID: mdl-19859555

RESUMEN

BACKGROUND: MicroRNAs (miRNAs) are a class of small non-coding RNAs that have recently emerged as important regulators of gene expression. They negatively regulate gene expression post-transcriptionally by translational repression and target mRNA degradation. miRNAs have been shown to play crucial roles in muscle development and in regulation of muscle cell proliferation and differentiation. METHODOLOGY/PRINCIPAL FINDINGS: By comparing miRNA expression profiling of proliferating myoblasts versus differentiated myotubes, a number of modulated miRNAs, not previously implicated in regulation of myogenic differentiation, were identified. Among these, miR-221 and miR-222 were strongly down-regulated upon differentiation of both primary and established myogenic cells. Conversely, miR-221 and miR-222 expression was restored in post-mitotic, terminally differentiated myotubes subjected to Src tyrosine kinase activation. By the use of specific inhibitors we provide evidence that expression of miR-221 and miR-222 is under the control of the Ras-MAPK pathway. Both in myoblasts and in myotubes, levels of the cell cycle inhibitor p27 inversely correlated with miR-221 and miR-222 expression, and indeed we show that p27 mRNA is a direct target of these miRNAs in myogenic cells. Ectopic expression of miR-221 and miR-222 in myoblasts undergoing differentiation induced a delay in withdrawal from the cell cycle and in myogenin expression, followed by inhibition of sarcomeric protein accumulation. When miR-221 and miR-222 were expressed in myotubes undergoing maturation, a profound alteration of myofibrillar organization was observed. CONCLUSIONS/SIGNIFICANCE: miR-221 and miR-222 have been found to be modulated during myogenesis and to play a role both in the progression from myoblasts to myocytes and in the achievement of the fully differentiated phenotype. Identification of miRNAs modulating muscle gene expression is crucial for the understanding of the circuits controlling skeletal muscle differentiation and maintenance.


Asunto(s)
Regulación de la Expresión Génica , MicroARNs/genética , Músculo Esquelético/metabolismo , Mioblastos/citología , Regiones no Traducidas 3' , Animales , Diferenciación Celular , Proliferación Celular , Sistema de Señalización de MAP Quinasas , Ratones , Microscopía Fluorescente/métodos , Modelos Biológicos , Desarrollo de Músculos , Fenotipo , Codorniz
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA