Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
1.
Genomics Proteomics Bioinformatics ; 18(4): 384-396, 2020 08.
Artículo en Inglés | MEDLINE | ID: mdl-33338663

RESUMEN

Muscle formation is a coordinated process driven by extensive gene expression changes where single cells fuse together to form multinucleated muscle fibers. Newly synthesized mRNAs are then regulated by RNA binding proteins (RBPs), affecting post-transcriptional transcript metabolism. Here, we determined how large-scale gene expression changes affect the catalog of RBPs by studying proliferating and differentiated muscle cells in healthy and dystrophic conditions. Transcriptomic analysis showed that the expression of more than 7000 genes was affected during myogenesis. We identified 769 RBPs, of which 294 were muscle-specific and 49 were uniquely shared with cardiomyocytes. A subset of 32 RBPs (half of which were muscle-specific) was found to be preferentially associated with target mRNAs in either myoblasts (MBs) or myotubes (MTs). A large proportion of catalytic proteins were bound to mRNAs even though they lack classical RNA binding domains. Finally, we showed how the identification of cell-specific RBPs enabled the identification of biomarkers that can separate healthy individuals from dystrophic patients. Our data show how interactome data can shed light on new basic RNA biology as well as provide cell-specific data that can be used for diagnostic purposes.


Asunto(s)
Proteoma , Proteínas de Unión al ARN , Perfilación de la Expresión Génica , Humanos , Miocitos Cardíacos/metabolismo , Proteoma/metabolismo , ARN Mensajero/genética , Proteínas de Unión al ARN/genética
2.
Proc Natl Acad Sci U S A ; 117(28): 16456-16464, 2020 07 14.
Artículo en Inglés | MEDLINE | ID: mdl-32616572

RESUMEN

Duchenne muscular dystrophy (DMD) is caused by mutations in the DMD gene leading to the presence of premature termination codons (PTC). Previous transcriptional studies have shown reduced DMD transcript levels in DMD patient and animal model muscles when PTC are present. Nonsense-mediated decay (NMD) has been suggested to be responsible for the observed reduction, but there is no experimental evidence supporting this claim. In this study, we aimed to investigate the mechanism responsible for the drop in DMD expression levels in the presence of PTC. We observed that the inhibition of NMD does not normalize DMD gene expression in DMD. Additionally, in situ hybridization showed that DMD messenger RNA primarily localizes in the nuclear compartment, confirming that a cytoplasmic mechanism like NMD indeed cannot be responsible for the observed reduction. Sequencing of nascent RNA to explore DMD transcription dynamics revealed a lower rate of DMD transcription in patient-derived myotubes compared to healthy controls, suggesting a transcriptional mechanism involved in reduced DMD transcript levels. Chromatin immunoprecipitation in muscle showed increased levels of the repressive histone mark H3K9me3 in mdx mice compared to wild-type mice, indicating a chromatin conformation less prone to transcription in mdx mice. In line with this finding, treatment with the histone deacetylase inhibitor givinostat caused a significant increase in DMD transcript expression in mdx mice. Overall, our findings show that transcription dynamics across the DMD locus are affected by the presence of PTC, hinting at a possible epigenetic mechanism responsible for this process.


Asunto(s)
Codón sin Sentido/genética , Distrofina/genética , Distrofia Muscular de Duchenne/genética , ARN Mensajero/genética , Animales , Codón sin Sentido/metabolismo , Modelos Animales de Enfermedad , Distrofina/metabolismo , Humanos , Ratones , Ratones Endogámicos mdx , Distrofia Muscular de Duchenne/metabolismo , Degradación de ARNm Mediada por Codón sin Sentido , ARN Mensajero/metabolismo
3.
Eur J Hum Genet ; 28(6): 815-825, 2020 06.
Artículo en Inglés | MEDLINE | ID: mdl-31896777

RESUMEN

Duchenne muscular dystrophy (DMD) is caused by pathogenic variants in the DMD gene leading to the lack of dystrophin. Variability in the disease course suggests that other factors influence disease progression. With this study we aimed to identify genetic factors that may account for some of the variability in the clinical presentation. We compared whole-exome sequencing (WES) data in 27 DMD patients with extreme phenotypes to identify candidate variants that could affect disease progression. Validation of the candidate SNPs was performed in two independent cohorts including 301 (BIO-NMD cohort) and 109 (CINRG cohort of European ancestry) DMD patients, respectively. Variants in the Tctex1 domain containing 1 (TCTEX1D1) gene on chromosome 1 were associated with age of ambulation loss. The minor alleles of two independent variants, known to affect TCTEX1D1 coding sequence and induce skipping of its exon 4, were associated with earlier loss of ambulation. Our data show that disease progression of DMD is affected by a new locus on chromosome 1 and demonstrate the possibility to identify genetic modifiers in rare diseases by studying WES data in patients with extreme phenotypes followed by multiple layers of validation.


Asunto(s)
Genes Modificadores , Distrofia Muscular de Duchenne/genética , Adolescente , Niño , Progresión de la Enfermedad , Humanos , Masculino , Distrofia Muscular de Duchenne/patología , Fenotipo , Polimorfismo de Nucleótido Simple
4.
PLoS One ; 13(10): e0204485, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30278058

RESUMEN

BACKGROUND: Duchenne muscular dystrophy is a lethal disease caused by lack of dystrophin. Skipping of exons adjacent to out-of-frame deletions has proven to restore dystrophin expression in Duchenne patients. Exon 51 has been the most studied target in both preclinical and clinical settings and the availability of standardized procedures to quantify exon skipping would be advantageous for the evaluation of preclinical and clinical data. OBJECTIVE: To compare methods currently used to quantify antisense oligonucleotide-induced exon 51 skipping in the DMD transcript and to provide guidance about the method to use. METHODS: Six laboratories shared blinded RNA samples from Duchenne patient-derived muscle cells treated with different amounts of exon 51 targeting antisense oligonucleotide. Exon 51 skipping levels were quantified using five different techniques: digital droplet PCR, single PCR assessed with Agilent bioanalyzer, nested PCR with agarose gel image analysis by either ImageJ or GeneTools software and quantitative real-time PCR. RESULTS: Differences in mean exon skipping levels and dispersion around the mean were observed across the different techniques. Results obtained by digital droplet PCR were reproducible and showed the smallest dispersion. Exon skipping quantification with the other methods showed overestimation of exon skipping or high data variation. CONCLUSIONS: Our results suggest that digital droplet PCR was the most precise and quantitative method. The quantification of exon 51 skipping by Agilent bioanalyzer after a single round of PCR was the second-best choice with a 2.3-fold overestimation of exon 51 skipping levels compared to digital droplet PCR.


Asunto(s)
Distrofina/genética , Distrofia Muscular de Duchenne/genética , Distrofia Muscular de Duchenne/metabolismo , Oligonucleótidos Antisentido , Reacción en Cadena de la Polimerasa/métodos , Empalme del ARN , Línea Celular , Distrofina/metabolismo , Exones , Humanos , Mioblastos/metabolismo
5.
Methods Mol Biol ; 1828: 249-262, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30171546

RESUMEN

Duchenne muscular dystrophy (DMD) is a severe, neuromuscular disorder caused by mutations in the DMD gene, precluding synthesis of functional dystrophin protein. Antisense oligonucleotide (AON)-mediated exon skipping has been developed as a method to restore the reading frame, which allows the synthesis of internally truncated, but partially functional dystrophin proteins, as found in the less severe Becker muscular dystrophy (BMD). This approach is species specific, since AONs targeting human exons often will not have full homology to mouse exons. As such, mouse models with mutations in the murine Dmd gene are of limited use to study human specific AONs in vivo. However, our del52hDMD/mdx mouse model contains mutated copies of both the mouse (nonsense mutation in exon 23) and human (deletion of exon 52) dystrophin-encoding genes. This model allows for testing effects of treatment with human specific exon 51 or 53 targeting AONs on RNA, protein, histological, and functional levels. Therefore, the model can be used to optimize human specific AONs, e.g., by comparing dystrophin protein and exon skipping levels.Absolute quantification of exon skipping levels can be obtained by digital droplet PCR (ddPCR). This method compartmentalizes samples into thousands of droplets that represent individual micro PCR reactions, and can be either positive or negative after amplification depending on whether there was a template molecule present or not. This allows for precise determination of the copy numbers of template molecules. The protocol described here uses probes binding to exon-exon junctions (EEJs) of human DMD transcripts with and without skipping of exon 51. We report that this method is specific for human transcripts so that exon skipping levels can be quantified accurately by ddPCR in del52hDMD/mdx mice.


Asunto(s)
Distrofina/genética , Exones , Distrofia Muscular de Duchenne/genética , Empalme del ARN , Eliminación de Secuencia , Animales , Modelos Animales de Enfermedad , Humanos , Ratones , Ratones Endogámicos mdx , Oligonucleótidos Antisentido , Reacción en Cadena de la Polimerasa , Transfección
6.
J Neuromuscul Dis ; 4(4): 327-335, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-29172005

RESUMEN

BACKGROUND: Inclusion body myositis is a late onset inflammatory myopathy lacking reliable serum biomarkers for diagnosis and for disease progression. OBJECTIVE: To identify diagnostic and predictive biomarkers, cytokine profiling is used to assess the potential of cytokines to discriminate between cases and controls and to assess whether treatment with methotrexate can influence biomarkers associated with disease progression. METHODS: The diagnostic and follow-up potential of 48 cytokines was tested using Bioplex-assay and ELISA in sera of healthy individuals, IBM patients and patients with other neuromuscular disorders. RESULTS: Ten cytokines (TRAIL, IL-8, MIF, MCP-1, LIF, IP-10, IFN-α2, MIG, bNGF and IL-3) were identified to be good to excellent markers to discern IBM patients from healthy controls. Three cytokines (IP-10, Eotaxin and SDF1A) changed significantly upon methotrexate treatment as compared with the natural clinical course. Muscle strength loss was associated with changes in IL-8 and SDF1A levels. IFN-γ levels were only associated with survival of IBM patients before correction for multiple comparisons. DISCUSSION: Cytokine profiling can discriminate IBM patients from healthy controls and other neuromuscular disorders. Immunosuppression with methotrexate affects cytokine levels in IBM. IL-8 and SDF1A could serve as biomarkers for disease progression.


Asunto(s)
Citocinas/sangre , Miositis por Cuerpos de Inclusión/sangre , Anciano , Anciano de 80 o más Años , Biomarcadores/sangre , Progresión de la Enfermedad , Femenino , Estudios de Seguimiento , Humanos , Inmunosupresores/uso terapéutico , Masculino , Metotrexato/uso terapéutico , Persona de Mediana Edad , Fuerza Muscular , Miositis por Cuerpos de Inclusión/tratamiento farmacológico , Miositis por Cuerpos de Inclusión/inmunología , Miositis por Cuerpos de Inclusión/mortalidad , Análisis de Supervivencia
7.
Proteomics Clin Appl ; 10(3): 290-9, 2016 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-26680509

RESUMEN

PURPOSE: Duchenne muscular dystrophy (DMD) is a severe and fatal neuromuscular disease. With the current developments on novel therapeutic strategies for DMD, the need to carefully monitor disease progression or regression upon treatment using molecular markers has become urgent. EXPERIMENTAL DESIGN: 2D LC protein fractionation was performed on patient serum samples, followed by LC-MS/MS-based identifications with label-free quantifications. RESULTS: Protein signatures were compared between patients and healthy (child and adult) controls and between ambulant and nonambulant patients. Various myofibrillar proteins demonstrated differences between DMD patients and controls, likely due to leakiness and breakdown of muscle fibers. Previously reported biomarkers, such as muscle-derived titin, myosin, and carbonic anhydrase I (CA1), were verified. MS-based results were compared with ELISA for vitamin D binding protein (GC), fibulin-1 (FBLN1), gelsolin (GSN), and carbonic anhydrase 1 (CA1). CONCLUSIONS AND CLINICAL RELEVANCE: The combined results of MS- and ELISA-based quantifications indicated more studies are needed to validate this serum protein signature for DMD patients. With these data promising candidate biomarkers have been identified for a rare genetic disease using serum proteome analysis.


Asunto(s)
Proteínas Sanguíneas/análisis , Ensayo de Inmunoadsorción Enzimática , Distrofia Muscular de Duchenne/sangre , Proteoma/análisis , Adolescente , Adulto , Biomarcadores/sangre , Niño , Cromatografía Líquida de Alta Presión , Biología Computacional , Humanos , Espectrometría de Masas , Proteómica , Adulto Joven
8.
Mol Ther Nucleic Acids ; 4: e265, 2015 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-26623937

RESUMEN

Duchenne muscular dystrophy (DMD) is a severe muscle wasting disorder typically caused by frame-shifting mutations in the DMD gene. Restoration of the reading frame would allow the production of a shorter but partly functional dystrophin protein as seen in Becker muscular dystrophy patients. This can be achieved with antisense oligonucleotides (AONs) that induce skipping of specific exons during pre-mRNA splicing. Different chemical modifications have been developed to improve AON properties. The 2'-deoxy-2'-fluoro (2F) RNA modification is attractive for exon skipping due to its ability to recruit ILF2/3 proteins to the 2F/pre-mRNA duplex, which resulted in enhanced exon skipping in spinal muscular atrophy models. In this study, we examined the effect of two different 2'-substituted AONs (2'-F phosphorothioate (2FPS) and 2'-O-Me phosphorothioate (2OMePS)) on exon skipping in DMD cell and animal models. In human cell cultures, 2FPS AONs showed higher exon skipping levels than their isosequential 2OMePS counterparts. Interestingly, in the mdx mouse model, 2FPS was less efficient than 2OMePS and suggested safety issues as evidenced by increased spleen size and weight loss. Our results do not support a clinical application for 2FPS AON.

9.
J Neurol Neurosurg Psychiatry ; 86(10): 1060-5, 2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-25476005

RESUMEN

OBJECTIVE: Duchenne muscular dystrophy (DMD) is characterised by progressive muscle weakness. It has recently been reported that single nucleotide polymorphisms (SNPs) located in the SPP1 and LTBP4 loci can account for some of the inter-individual variability observed in the clinical disease course. The validation of genetic association in large independent cohorts is a key process for rare diseases in order to qualify prognostic biomarkers and stratify patients in clinical trials. METHODS: Duchenne patients from five European neuromuscular centres were included. Information about age at wheelchair dependence and steroid use was gathered. Melting curve analysis of PCR fragments or Sanger sequencing were used to genotype SNP rs28357094 in the SPP1 gene in 336 patients. The genotype of SNPs rs2303729, rs1131620, rs1051303 and rs10880 in the LTBP4 locus was determined in 265 patients by mass spectrometry. For both loci, a multivariate analysis was performed, using genotype/haplotype, steroid use and cohort as covariates. RESULTS: We show that corticosteroid treatment and the IAAM haplotype of the LTBP4 gene are significantly associated with prolonged ambulation in patients with DMD. There was no significant association between the SNP rs28357094 in the SPP1 gene and the age of ambulation loss. CONCLUSIONS: This study underlines the importance of replicating genetic association studies for rare diseases in large independent cohorts to identify the most robust associations. We anticipate that genotyping of validated genetic associations will become important for the design and interpretation of clinical trials.


Asunto(s)
Proteínas de Unión a TGF-beta Latente/genética , Distrofia Muscular de Duchenne/genética , Osteopontina/genética , Factores de Edad , Niño , Estudios de Cohortes , Progresión de la Enfermedad , Europa (Continente) , Femenino , Genotipo , Humanos , Masculino , Polimorfismo de Nucleótido Simple , Pronóstico , Reproducibilidad de los Resultados , Esteroides/uso terapéutico , Caminata , Silla de Ruedas
10.
EMBO Mol Med ; 6(7): 918-36, 2014 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-24920607

RESUMEN

Despite the recent progress in the broad-scaled analysis of proteins in body fluids, there is still a lack in protein profiling approaches for biomarkers of rare diseases. Scarcity of samples is the main obstacle hindering attempts to apply discovery driven protein profiling in rare diseases. We addressed this challenge by combining samples collected within the BIO-NMD consortium from four geographically dispersed clinical sites to identify protein markers associated with muscular dystrophy using an antibody bead array platform with 384 antibodies. Based on concordance in statistical significance and confirmatory results obtained from analysis of both serum and plasma, we identified eleven proteins associated with muscular dystrophy, among which four proteins were elevated in blood from muscular dystrophy patients: carbonic anhydrase III (CA3) and myosin light chain 3 (MYL3), both specifically expressed in slow-twitch muscle fibers and mitochondrial malate dehydrogenase 2 (MDH2) and electron transfer flavoprotein A (ETFA). Using age-matched sub-cohorts, 9 protein profiles correlating with disease progression and severity were identified, which hold promise for the development of new clinical tools for management of dystrophinopathies.


Asunto(s)
Proteínas Sanguíneas/análisis , Distrofia Muscular de Duchenne/sangre , Biomarcadores/análisis , Biomarcadores/sangre , Estudios de Cohortes , Femenino , Humanos , Masculino , Distrofia Muscular de Duchenne/diagnóstico , Análisis por Matrices de Proteínas/métodos , Proteómica/métodos , Enfermedades Raras/sangre , Enfermedades Raras/diagnóstico
11.
PLoS One ; 9(5): e96290, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24789369

RESUMEN

MicroRNAs (miRNAs) regulate many aspects of cellular function and their deregulation has been implicated in heart disease. MiRNA-30c is differentially expressed in the heart during the progression towards heart failure and in vitro studies hint to its importance in cellular physiology. As little is known about the in vivo function of miRNA-30c in the heart, we generated transgenic mice that specifically overexpress miRNA-30c in cardiomyocytes. We show that these mice display no abnormalities until about 6 weeks of age, but subsequently develop a severely dilated cardiomyopathy. Gene expression analysis of the miRNA-30c transgenic hearts before onset of the phenotype indicated disturbed mitochondrial function. This was further evident by the downregulation of mitochondrial oxidative phosphorylation (OXPHOS) complexes III and IV at the protein level. Taken together these data indicate impaired mitochondrial function due to OXPHOS protein depletion as a potential cause for the observed dilated cardiomyopathic phenotype in miRNA-30c transgenic mice. We thus establish an in vivo role for miRNA-30c in cardiac physiology, particularly in mitochondrial function.


Asunto(s)
Cardiomiopatía Dilatada/genética , Perfilación de la Expresión Génica , MicroARNs/genética , Miocitos Cardíacos/metabolismo , Animales , Cardiomiopatía Dilatada/metabolismo , Cardiomiopatía Dilatada/fisiopatología , Ecocardiografía , Ratones Endogámicos C57BL , Ratones Transgénicos , Microscopía Electrónica de Transmisión , Mitocondrias/genética , Mitocondrias/metabolismo , Mitocondrias/ultraestructura , Proteínas Mitocondriales/genética , Proteínas Mitocondriales/metabolismo , Miocitos Cardíacos/ultraestructura , Análisis de Secuencia por Matrices de Oligonucleótidos , Fosforilación Oxidativa , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Proteínas Ribosómicas/genética , Proteínas Ribosómicas/metabolismo , Factores de Tiempo
12.
Proteomics Clin Appl ; 8(3-4): 269-78, 2014 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-24458521

RESUMEN

PURPOSE: To identify and validate serum biomarkers for the progression of Duchenne muscular dystrophy (DMD) using a MS-based bottom-up pipeline. EXPERIMENTAL DESIGN: We used a bottom-up proteomics approach, including a protein concentration equalization step, different proteolytic digestions, and MS detection schemes, to identify candidate biomarkers in serum samples from control subjects and DMD patients. Fibronectin was chosen for follow-up based on the differences in peptide spectral counts and sequence coverage observed between the DMD and control groups. Subsequently, fibronectin levels were determined with ELISA in 68 DMD patients, 38 milder Becker muscular dystrophy patients, 33 patients with other neuromuscular disorders, and 15 age-matched adult and child controls. RESULTS: There was a significant increase in fibronectin levels in DMD patients compared to age-matched controls. Fibronectin levels in patients with Becker muscular dystrophy, Bethlem myopathy, or myasthenia gravis were comparable to control levels. Progressive elevation in fibronectin levels was observed in longitudinal samples from 22 DMD patients followed up for a period of 6 months up to 4 years. CONCLUSION AND CLINICAL RELEVANCE: This study suggests that serum fibronectin levels may constitute a promising biomarker to monitor disease progression in DMD patients.


Asunto(s)
Biomarcadores/sangre , Fibronectinas/sangre , Músculo Esquelético/metabolismo , Distrofia Muscular de Duchenne/sangre , Adolescente , Adulto , Niño , Preescolar , Contractura/sangre , Contractura/genética , Contractura/patología , Femenino , Fibronectinas/genética , Humanos , Masculino , Espectrometría de Masas , Persona de Mediana Edad , Músculo Esquelético/patología , Distrofias Musculares/sangre , Distrofias Musculares/congénito , Distrofias Musculares/genética , Distrofias Musculares/patología , Distrofia Muscular de Duchenne/genética , Distrofia Muscular de Duchenne/patología , Miastenia Gravis/sangre , Miastenia Gravis/genética , Miastenia Gravis/patología
13.
PLoS Curr ; 52013 Nov 22.
Artículo en Inglés | MEDLINE | ID: mdl-24292657

RESUMEN

Background Duchenne muscular dystrophy is a lethal, progressive, muscle-wasting disease caused by mutations in the DMD gene. Structural remodelling processes are responsible for muscle atrophy and replacement of myofibers by fibrotic and adipose tissues. Molecular interventions modulating catabolic pathways, such as the ubiquitin-proteasome and the autophagy-lysosome systems, are under development for Duchenne and other muscular dystrophies. The Akt signaling cascade is one of the main pathways involved in protein synthesis and autophagy repression and is known to be up-regulated in dystrophin null mdx mice. Results We report that autophagy is triggered by fasting in the tibialis anterior muscle of control mice but not in mdx mice. Mdx mice show persistent Akt activation upon fasting and failure to increase the expression of FoxO3 regulated autophagy and atrophy genes, such as Bnip3 and Atrogin1. We also provide evidence that autophagy is differentially regulated in mdx tibialis anterior and diaphragm muscles. Conclusions Our data support the concept that autophagy is impaired in the tibialis anterior muscle of mdx mice and that the regulation of autophagy is muscle type dependent. Differences between muscle groups should be considered during the pre-clinical development of therapeutic strategies addressing muscle metabolism.

14.
PLoS One ; 7(5): e36754, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22586493

RESUMEN

The Kruppel-like factor (KLF) family of transcription factors regulates diverse cell biological processes including proliferation, differentiation, survival and growth. Previous studies have shown that KLF15 inhibits cardiac hypertrophy by repressing the activity of pivotal cardiac transcription factors such as GATA4, MEF2 and myocardin. We set out this study to characterize the interaction of KLF15 with putative other transcription factors. We first show that KLF15 interacts with myocardin-related transcription factors (MRTFs) and strongly represses the transcriptional activity of MRTF-A and MRTF-B. Second, we identified a region within the C-terminal zinc fingers of KLF15 that contains the nuclear localization signal. Third, we investigated whether overexpression of KLF15 in the heart would have therapeutic potential. Using recombinant adeno-associated viruses (rAAV) we have overexpressed KLF15 specifically in the mouse heart and provide the first evidence that elevation of cardiac KLF15 levels prevents the development of cardiac hypertrophy in a model of Angiotensin II induced hypertrophy.


Asunto(s)
Cardiomegalia , Proteínas de Unión al ADN , Proteínas Nucleares/metabolismo , Proteínas de Fusión Oncogénica , Transactivadores/metabolismo , Factores de Transcripción , Angiotensina II/farmacología , Animales , Sitios de Unión , Células COS , Cardiomegalia/genética , Cardiomegalia/metabolismo , Cardiomegalia/terapia , Diferenciación Celular , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Modelos Animales de Enfermedad , Regulación de la Expresión Génica/efectos de los fármacos , Terapia Genética , Humanos , Factores de Transcripción de Tipo Kruppel , Ratones , Proteínas Mutantes/metabolismo , Proteínas Nucleares/genética , Proteínas de Fusión Oncogénica/genética , Proteínas de Fusión Oncogénica/metabolismo , Regiones Promotoras Genéticas , Transactivadores/genética , Factores de Transcripción/genética , Factores de Transcripción/metabolismo
15.
J Biol Chem ; 285(35): 27449-27456, 2010 Aug 27.
Artículo en Inglés | MEDLINE | ID: mdl-20566642

RESUMEN

Pathological forms of left ventricular hypertrophy (LVH) often progress to heart failure. Specific transcription factors have been identified that activate the gene program to induce pathological forms of LVH. It is likely that apart from activating transcriptional inducers of LVH, constitutive transcriptional repressors need to be removed during the development of cardiac hypertrophy. Here, we report that the constitutive presence of Krüppel-like factor 15 (KLF15) is lost in pathological hypertrophy and that this loss precedes progression toward heart failure. We show that transforming growth factor-beta-mediated activation of p38 MAPK is necessary and sufficient to decrease KLF15 expression. We further show that KLF15 robustly inhibits myocardin, a potent transcriptional activator. Loss of KLF15 during pathological LVH relieves the inhibitory effects on myocardin and stimulates the expression of serum response factor target genes, such as atrial natriuretic factor. This uncovers a novel mechanism where activated p38 MAPK decreases KLF15, an important constitutive transcriptional repressor whose removal seems a vital step to allow the induction of pathological LVH.


Asunto(s)
Proteínas de Unión al ADN/metabolismo , Regulación de la Expresión Génica , Hipertrofia Ventricular Izquierda/metabolismo , Factores de Transcripción de Tipo Kruppel/metabolismo , Miocardio/metabolismo , Proteínas Nucleares/metabolismo , Proteínas Represoras/metabolismo , Transactivadores/metabolismo , Factores de Transcripción/metabolismo , Animales , Factor Natriurético Atrial/metabolismo , Células COS , Chlorocebus aethiops , Activación Enzimática , Ratones , Ratas , Ratas Endogámicas Lew , Factor de Crecimiento Transformador beta/metabolismo , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...