Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 46
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Proc Natl Acad Sci U S A ; 116(32): 16046-16055, 2019 08 06.
Artículo en Inglés | MEDLINE | ID: mdl-31341086

RESUMEN

Epstein-Barr virus (EBV) is a human tumor virus and a model of herpesviral latency. The virus efficiently infects resting human B lymphocytes and induces their continuous proliferation in vitro, which mimics certain aspects of EBV's oncogenic potential in vivo. How lymphoblastoid cell lines (LCLs) evolve from the infected lymphocytes is uncertain. We conducted a systematic time-resolved longitudinal study of cellular functions and transcriptional profiles of newly infected naïve primary B lymphocytes. EBV reprograms the cells comprehensively and globally. Rapid and extensive transcriptional changes occur within 24 h and precede any metabolic and phenotypic changes. Within 72 h, the virus activates the cells, changes their phenotypes with respect to cell size, RNA, and protein content, and induces metabolic pathways to cope with the increased demand for energy, supporting an efficient cell cycle entry on day 3 postinfection. The transcriptional program that EBV initiates consists of 3 waves of clearly discernable clusters of cellular genes that peak on day 2, 3, or 4 and regulate RNA synthesis, metabolic pathways, and cell division, respectively. Upon onset of cell doublings on day 4, the cellular transcriptome appears to be completely reprogrammed to support the proliferating cells, but 3 additional clusters of EBV-regulated genes fine-tune cell signaling, migration, and immune response pathways, eventually. Our study reveals that more than 11,000 genes are regulated upon EBV infection as naïve B cells exit quiescence to enter a germinal center-like differentiation program, which culminates in immortalized, proliferating cells that partially resemble plasmablasts and early plasma cells.


Asunto(s)
Linfocitos B/virología , Infecciones por Virus de Epstein-Barr/virología , Herpesvirus Humano 4/fisiología , Linfocitos B/metabolismo , Infecciones por Virus de Epstein-Barr/genética , Regulación Viral de la Expresión Génica , Células HEK293 , Humanos , Activación de Linfocitos/genética , Familia de Multigenes , Fenotipo , Factores de Tiempo , Transcriptoma/genética
2.
Am J Hum Genet ; 102(6): 1018-1030, 2018 06 07.
Artículo en Inglés | MEDLINE | ID: mdl-29754768

RESUMEN

Coenzyme A (CoA) is an essential metabolic cofactor used by around 4% of cellular enzymes. Its role is to carry and transfer acetyl and acyl groups to other molecules. Cells can synthesize CoA de novo from vitamin B5 (pantothenate) through five consecutive enzymatic steps. Phosphopantothenoylcysteine synthetase (PPCS) catalyzes the second step of the pathway during which phosphopantothenate reacts with ATP and cysteine to form phosphopantothenoylcysteine. Inborn errors of CoA biosynthesis have been implicated in neurodegeneration with brain iron accumulation (NBIA), a group of rare neurological disorders characterized by accumulation of iron in the basal ganglia and progressive neurodegeneration. Exome sequencing in five individuals from two unrelated families presenting with dilated cardiomyopathy revealed biallelic mutations in PPCS, linking CoA synthesis with a cardiac phenotype. Studies in yeast and fruit flies confirmed the pathogenicity of identified mutations. Biochemical analysis revealed a decrease in CoA levels in fibroblasts of all affected individuals. CoA biosynthesis can occur with pantethine as a source independent from PPCS, suggesting pantethine as targeted treatment for the affected individuals still alive.


Asunto(s)
Cardiomiopatía Dilatada/enzimología , Cardiomiopatía Dilatada/genética , Genes Recesivos , Mutación/genética , Péptido Sintasas/genética , Secuencia de Aminoácidos , Animales , Vías Biosintéticas , Cardiomiopatía Dilatada/diagnóstico , Carnitina/análogos & derivados , Carnitina/metabolismo , Preescolar , Coenzima A/biosíntesis , Demografía , Drosophila , Estabilidad de Enzimas , Femenino , Fibroblastos/metabolismo , Corazón/fisiopatología , Secuenciación de Nucleótidos de Alto Rendimiento , Homocigoto , Humanos , Lactante , Recién Nacido , Imagen por Resonancia Magnética , Masculino , Panteteína/administración & dosificación , Panteteína/análogos & derivados , Linaje , Péptido Sintasas/sangre , Péptido Sintasas/química , Péptido Sintasas/deficiencia , Reproducibilidad de los Resultados , Saccharomyces cerevisiae/genética
3.
Am J Physiol Endocrinol Metab ; 316(5): E866-E879, 2019 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-30620635

RESUMEN

Intermuscular adipose tissue (IMAT) is negatively related to insulin sensitivity, but a causal role of IMAT in the development of insulin resistance is unknown. IMAT was sampled in humans to test for the ability to induce insulin resistance in vitro and characterize gene expression to uncover how IMAT may promote skeletal muscle insulin resistance. Human primary muscle cells were incubated with conditioned media from IMAT, visceral (VAT), or subcutaneous adipose tissue (SAT) to evaluate changes in insulin sensitivity. RNAseq analysis was performed on IMAT with gene expression compared with skeletal muscle and SAT, and relationships to insulin sensitivity were determined in men and women spanning a wide range of insulin sensitivity measured by hyperinsulinemic-euglycemic clamp. Conditioned media from IMAT and VAT decreased insulin sensitivity similarly compared with SAT. Multidimensional scaling analysis revealed distinct gene expression patterns in IMAT compared with SAT and muscle. Pathway analysis revealed that IMAT expression of genes in insulin signaling, oxidative phosphorylation, and peroxisomal metabolism related positively to donor insulin sensitivity, whereas expression of macrophage markers, inflammatory cytokines, and secreted extracellular matrix proteins were negatively related to insulin sensitivity. Perilipin 5 gene expression suggested greater IMAT lipolysis in insulin-resistant individuals. Combined, these data show that factors secreted from IMAT modulate muscle insulin sensitivity, possibly via secretion of inflammatory cytokines and extracellular matrix proteins, and by increasing local FFA concentration in humans. These data suggest IMAT may be an important regulator of skeletal muscle insulin sensitivity and could be a novel therapeutic target for skeletal muscle insulin resistance.


Asunto(s)
Tejido Adiposo/metabolismo , Resistencia a la Insulina/genética , Fibras Musculares Esqueléticas/metabolismo , Músculo Esquelético/metabolismo , Adulto , Atletas , Diabetes Mellitus Tipo 2/metabolismo , Ácidos Grasos no Esterificados/metabolismo , Femenino , Perfilación de la Expresión Génica , Regulación de la Expresión Génica , Técnica de Clampeo de la Glucosa , Humanos , Grasa Intraabdominal/metabolismo , Masculino , Persona de Mediana Edad , Obesidad/metabolismo , Cultivo Primario de Células , Conducta Sedentaria , Análisis de Secuencia de ARN , Grasa Subcutánea/metabolismo
4.
Am J Hum Genet ; 98(6): 1130-1145, 2016 Jun 02.
Artículo en Inglés | MEDLINE | ID: mdl-27259049

RESUMEN

Multiple acyl-CoA dehydrogenase deficiencies (MADDs) are a heterogeneous group of metabolic disorders with combined respiratory-chain deficiency and a neuromuscular phenotype. Despite recent advances in understanding the genetic basis of MADD, a number of cases remain unexplained. Here, we report clinically relevant variants in FLAD1, which encodes FAD synthase (FADS), as the cause of MADD and respiratory-chain dysfunction in nine individuals recruited from metabolic centers in six countries. In most individuals, we identified biallelic frameshift variants in the molybdopterin binding (MPTb) domain, located upstream of the FADS domain. Inasmuch as FADS is essential for cellular supply of FAD cofactors, the finding of biallelic frameshift variants was unexpected. Using RNA sequencing analysis combined with protein mass spectrometry, we discovered FLAD1 isoforms, which only encode the FADS domain. The existence of these isoforms might explain why affected individuals with biallelic FLAD1 frameshift variants still harbor substantial FADS activity. Another group of individuals with a milder phenotype responsive to riboflavin were shown to have single amino acid changes in the FADS domain. When produced in E. coli, these mutant FADS proteins resulted in impaired but detectable FADS activity; for one of the variant proteins, the addition of FAD significantly improved protein stability, arguing for a chaperone-like action similar to what has been reported in other riboflavin-responsive inborn errors of metabolism. In conclusion, our studies identify FLAD1 variants as a cause of potentially treatable inborn errors of metabolism manifesting with MADD and shed light on the mechanisms by which FADS ensures cellular FAD homeostasis.


Asunto(s)
Mutación del Sistema de Lectura/genética , Enfermedades Mitocondriales/genética , Deficiencia Múltiple de Acil Coenzima A Deshidrogenasa/genética , Nucleotidiltransferasas/genética , Riboflavina/farmacología , Complejo Vitamínico B/farmacología , Adulto , Western Blotting , Estudios de Casos y Controles , Células Cultivadas , Transporte de Electrón , Femenino , Fibroblastos/efectos de los fármacos , Fibroblastos/metabolismo , Fibroblastos/patología , Flavina-Adenina Dinucleótido/metabolismo , Perfilación de la Expresión Génica , Humanos , Lactante , Recién Nacido , Hígado/efectos de los fármacos , Hígado/metabolismo , Hígado/patología , Masculino , Enfermedades Mitocondriales/tratamiento farmacológico , Enfermedades Mitocondriales/patología , Deficiencia Múltiple de Acil Coenzima A Deshidrogenasa/tratamiento farmacológico , Deficiencia Múltiple de Acil Coenzima A Deshidrogenasa/patología , Músculo Esquelético/efectos de los fármacos , Músculo Esquelético/metabolismo , Músculo Esquelético/patología , Mutagénesis Sitio-Dirigida , Unión Proteica , ARN Mensajero/genética , Reacción en Cadena en Tiempo Real de la Polimerasa , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Piel/efectos de los fármacos , Piel/metabolismo , Piel/patología , Espectrometría de Masa por Láser de Matriz Asistida de Ionización Desorción , Adulto Joven
5.
Nature ; 501(7468): 506-11, 2013 Sep 26.
Artículo en Inglés | MEDLINE | ID: mdl-24037378

RESUMEN

Genome sequencing projects are discovering millions of genetic variants in humans, and interpretation of their functional effects is essential for understanding the genetic basis of variation in human traits. Here we report sequencing and deep analysis of messenger RNA and microRNA from lymphoblastoid cell lines of 462 individuals from the 1000 Genomes Project--the first uniformly processed high-throughput RNA-sequencing data from multiple human populations with high-quality genome sequences. We discover extremely widespread genetic variation affecting the regulation of most genes, with transcript structure and expression level variation being equally common but genetically largely independent. Our characterization of causal regulatory variation sheds light on the cellular mechanisms of regulatory and loss-of-function variation, and allows us to infer putative causal variants for dozens of disease-associated loci. Altogether, this study provides a deep understanding of the cellular mechanisms of transcriptome variation and of the landscape of functional variants in the human genome.


Asunto(s)
Variación Genética/genética , Genoma Humano/genética , Secuenciación de Nucleótidos de Alto Rendimiento , Análisis de Secuencia de ARN , Transcriptoma/genética , Alelos , Línea Celular Transformada , Exones/genética , Perfilación de la Expresión Génica , Humanos , Polimorfismo de Nucleótido Simple/genética , Sitios de Carácter Cuantitativo/genética , ARN Mensajero/análisis , ARN Mensajero/genética
6.
Am J Hum Genet ; 96(5): 826-31, 2015 May 07.
Artículo en Inglés | MEDLINE | ID: mdl-25913036

RESUMEN

Methionyl-tRNA synthetase (MARS) catalyzes the ligation of methionine to tRNA and is critical for protein biosynthesis. We identified biallelic missense mutations in MARS in a specific form of pediatric pulmonary alveolar proteinosis (PAP), a severe lung disorder that is prevalent on the island of Réunion and the molecular basis of which is unresolved. Mutations were found in 26 individuals from Réunion and nearby islands and in two families from other countries. Functional consequences of the mutated alleles were assessed by growth of wild-type and mutant strains and methionine-incorporation assays in yeast. Enzyme activity was attenuated in a liquid medium without methionine but could be restored by methionine supplementation. In summary, identification of a founder mutation in MARS led to the molecular definition of a specific type of PAP and will enable carrier screening in the affected community and possibly open new treatment opportunities.


Asunto(s)
Metionina-ARNt Ligasa/genética , Proteinosis Alveolar Pulmonar/genética , Adolescente , Alelos , Niño , Preescolar , Retículo Endoplásmico/genética , Retículo Endoplásmico/patología , Femenino , Aparato de Golgi/genética , Aparato de Golgi/patología , Humanos , Masculino , Mutación Missense , Biosíntesis de Proteínas , Proteinosis Alveolar Pulmonar/patología , Adulto Joven
7.
Am J Hum Genet ; 97(1): 163-9, 2015 Jul 02.
Artículo en Inglés | MEDLINE | ID: mdl-26073778

RESUMEN

Acute liver failure (ALF) in infancy and childhood is a life-threatening emergency. Few conditions are known to cause recurrent acute liver failure (RALF), and in about 50% of cases, the underlying molecular cause remains unresolved. Exome sequencing in five unrelated individuals with fever-dependent RALF revealed biallelic mutations in NBAS. Subsequent Sanger sequencing of NBAS in 15 additional unrelated individuals with RALF or ALF identified compound heterozygous mutations in an additional six individuals from five families. Immunoblot analysis of mutant fibroblasts showed reduced protein levels of NBAS and its proposed interaction partner p31, both involved in retrograde transport between endoplasmic reticulum and Golgi. We recommend NBAS analysis in individuals with acute infantile liver failure, especially if triggered by fever.


Asunto(s)
Fallo Hepático Agudo/genética , Proteínas de Neoplasias/genética , Secuencia de Bases , Transporte Biológico/genética , Exoma/genética , Fibroblastos/metabolismo , Frecuencia de los Genes , Alemania , Humanos , Immunoblotting , Lactante , Datos de Secuencia Molecular , Proteínas de Neoplasias/metabolismo , Linaje , Recurrencia , Análisis de Secuencia de ADN
8.
Am J Hum Genet ; 94(1): 11-22, 2014 Jan 02.
Artículo en Inglés | MEDLINE | ID: mdl-24360804

RESUMEN

Neurodegeneration with brain iron accumulation (NBIA) comprises a clinically and genetically heterogeneous group of disorders with progressive extrapyramidal signs and neurological deterioration, characterized by iron accumulation in the basal ganglia. Exome sequencing revealed the presence of recessive missense mutations in COASY, encoding coenzyme A (CoA) synthase in one NBIA-affected subject. A second unrelated individual carrying mutations in COASY was identified by Sanger sequence analysis. CoA synthase is a bifunctional enzyme catalyzing the final steps of CoA biosynthesis by coupling phosphopantetheine with ATP to form dephospho-CoA and its subsequent phosphorylation to generate CoA. We demonstrate alterations in RNA and protein expression levels of CoA synthase, as well as CoA amount, in fibroblasts derived from the two clinical cases and in yeast. This is the second inborn error of coenzyme A biosynthesis to be implicated in NBIA.


Asunto(s)
Encéfalo/efectos de los fármacos , Exoma , Hierro/metabolismo , Degeneración Nerviosa/patología , Encéfalo/patología , Clonación Molecular , Coenzima A/metabolismo , Escherichia coli/genética , Femenino , Fibroblastos/metabolismo , Regulación de la Expresión Génica , Humanos , Masculino , Mitocondrias/enzimología , Mitocondrias/genética , Mutación Missense , Panteteína/análogos & derivados , Panteteína/metabolismo , Linaje , Fosforilación , Saccharomyces cerevisiae/genética , Transferasas/genética , Transferasas/metabolismo
9.
Am J Hum Genet ; 95(6): 698-707, 2014 Dec 04.
Artículo en Inglés | MEDLINE | ID: mdl-25434003

RESUMEN

Mutations in components of the major spliceosome have been described in disorders with craniofacial anomalies, e.g., Nager syndrome and mandibulofacial dysostosis type Guion-Almeida. The U5 spliceosomal complex of eight highly conserved proteins is critical for pre-mRNA splicing. We identified biallelic mutations in TXNL4A, a member of this complex, in individuals with Burn-McKeown syndrome (BMKS). This rare condition is characterized by bilateral choanal atresia, hearing loss, cleft lip and/or palate, and other craniofacial dysmorphisms. Mutations were found in 9 of 11 affected families. In 8 families, affected individuals carried a rare loss-of-function mutation (nonsense, frameshift, or microdeletion) on one allele and a low-frequency 34 bp deletion (allele frequency 0.76%) in the core promoter region on the other allele. In a single highly consanguineous family, formerly diagnosed as oculo-oto-facial dysplasia, the four affected individuals were homozygous for a 34 bp promoter deletion, which differed from the promoter deletion in the other families. Reporter gene and in vivo assays showed that the promoter deletions led to reduced expression of TXNL4A. Depletion of TXNL4A (Dib1) in yeast demonstrated reduced assembly of the tri-snRNP complex. Our results indicate that BMKS is an autosomal-recessive condition, which is frequently caused by compound heterozygosity of low-frequency promoter deletions in combination with very rare loss-of-function mutations.


Asunto(s)
Atresia de las Coanas/genética , Sordera/congénito , Eliminación de Gen , Cardiopatías Congénitas/genética , Regiones Promotoras Genéticas/genética , Ribonucleoproteína Nuclear Pequeña U5/genética , Empalmosomas/genética , Alelos , Preescolar , Atresia de las Coanas/diagnóstico , Sordera/diagnóstico , Sordera/genética , Exosomas/genética , Facies , Femenino , Perfilación de la Expresión Génica , Frecuencia de los Genes , Genes Reporteros , Cardiopatías Congénitas/diagnóstico , Heterocigoto , Homocigoto , Humanos , Masculino , Mutación , Análisis de Secuencia por Matrices de Oligonucleótidos , Linaje , Fenotipo , Ribonucleoproteína Nuclear Pequeña U5/metabolismo , Análisis de Secuencia de ADN , Empalmosomas/metabolismo
10.
Am J Hum Genet ; 95(6): 708-20, 2014 Dec 04.
Artículo en Inglés | MEDLINE | ID: mdl-25434004

RESUMEN

Respiratory chain deficiencies exhibit a wide variety of clinical phenotypes resulting from defective mitochondrial energy production through oxidative phosphorylation. These defects can be caused by either mutations in the mtDNA or mutations in nuclear genes coding for mitochondrial proteins. The underlying pathomechanisms can affect numerous pathways involved in mitochondrial physiology. By whole-exome and candidate gene sequencing, we identified 11 individuals from 9 families carrying compound heterozygous or homozygous mutations in GTPBP3, encoding the mitochondrial GTP-binding protein 3. Affected individuals from eight out of nine families presented with combined respiratory chain complex deficiencies in skeletal muscle. Mutations in GTPBP3 are associated with a severe mitochondrial translation defect, consistent with the predicted function of the protein in catalyzing the formation of 5-taurinomethyluridine (τm(5)U) in the anticodon wobble position of five mitochondrial tRNAs. All case subjects presented with lactic acidosis and nine developed hypertrophic cardiomyopathy. In contrast to individuals with mutations in MTO1, the protein product of which is predicted to participate in the generation of the same modification, most individuals with GTPBP3 mutations developed neurological symptoms and MRI involvement of thalamus, putamen, and brainstem resembling Leigh syndrome. Our study of a mitochondrial translation disorder points toward the importance of posttranscriptional modification of mitochondrial tRNAs for proper mitochondrial function.


Asunto(s)
Acidosis Láctica/genética , Encefalopatías/genética , Cardiomiopatía Hipertrófica/genética , Proteínas de Unión al GTP/genética , Procesamiento Proteico-Postraduccional , Acidosis Láctica/fisiopatología , Secuencia de Aminoácidos , Encéfalo/patología , Encefalopatías/fisiopatología , Cardiomiopatía Hipertrófica/fisiopatología , Línea Celular , Niño , Preescolar , Consanguinidad , Femenino , Fibroblastos , Proteínas de Unión al GTP/metabolismo , Humanos , Lactante , Recién Nacido , Masculino , Datos de Secuencia Molecular , Mutación , Linaje , Biosíntesis de Proteínas , Interferencia de ARN , ARN de Transferencia/genética , ARN de Transferencia/metabolismo , Alineación de Secuencia
11.
N Engl J Med ; 370(11): 1019-28, 2014 Mar 13.
Artículo en Inglés | MEDLINE | ID: mdl-24571724

RESUMEN

BACKGROUND: Corticotropin-independent Cushing's syndrome is caused by tumors or hyperplasia of the adrenal cortex. The molecular pathogenesis of cortisol-producing adrenal adenomas is not well understood. METHODS: We performed exome sequencing of tumor-tissue specimens from 10 patients with cortisol-producing adrenal adenomas and evaluated recurrent mutations in candidate genes in an additional 171 patients with adrenocortical tumors. We also performed genomewide copy-number analysis in 35 patients with cortisol-secreting bilateral adrenal hyperplasias. We studied the effects of these genetic defects both clinically and in vitro. RESULTS: Exome sequencing revealed somatic mutations in PRKACA, which encodes the catalytic subunit of cyclic AMP-dependent protein kinase (protein kinase A [PKA]), in 8 of 10 adenomas (c.617A→C in 7 and c.595_596insCAC in 1). Overall, PRKACA somatic mutations were identified in 22 of 59 unilateral adenomas (37%) from patients with overt Cushing's syndrome; these mutations were not detectable in 40 patients with subclinical hypercortisolism or in 82 patients with other adrenal tumors. Among 35 patients with cortisol-producing hyperplasias, 5 (including 2 first-degree relatives) carried a germline copy-number gain (duplication) of the genomic region on chromosome 19 that includes PRKACA. In vitro studies showed impaired inhibition of both PKA catalytic subunit mutants by the PKA regulatory subunit, whereas cells from patients with germline chromosomal gains showed increased protein levels of the PKA catalytic subunit; in both instances, basal PKA activity was increased. CONCLUSIONS: Genetic alterations of the catalytic subunit of PKA were found to be associated with human disease. Germline duplications of this gene resulted in bilateral adrenal hyperplasias, whereas somatic PRKACA mutations resulted in unilateral cortisol-producing adrenal adenomas. (Funded by the European Commission Seventh Framework Program and others.).


Asunto(s)
Adenoma/genética , Neoplasias de las Glándulas Suprarrenales/genética , Hiperplasia Suprarrenal Congénita/genética , Síndrome de Cushing/etiología , Proteínas Quinasas Dependientes de AMP Cíclico/genética , Mutación de Línea Germinal , Adenoma/complicaciones , Adenoma/enzimología , Neoplasias de las Glándulas Suprarrenales/complicaciones , Neoplasias de las Glándulas Suprarrenales/enzimología , Adulto , Dominio Catalítico , Síndrome de Cushing/enzimología , Proteínas Quinasas Dependientes de AMP Cíclico/química , Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Exoma , Humanos , Hidrocortisona/biosíntesis , Persona de Mediana Edad , Mutación , Conformación Proteica , Análisis de Secuencia de ADN
12.
J Mol Cell Cardiol ; 98: 103-7, 2016 09.
Artículo en Inglés | MEDLINE | ID: mdl-27476877

RESUMEN

Deep sequencing techniques and advanced data analysis methods recently enabled the characterization of thousands of circular RNA isoforms (circRNAs) from a number of tissues and organisms. There is emerging evidence that some circRNAs may have important biological functions or serve as diagnostic biomarkers in disease conditions. In order to analyze circRNA expression in the heart and its changes in different conditions we performed RNA-Seq analysis of ribosome-depleted libraries from rats (neonatal and adult), mice (sham or after transverse aortic constriction, TAC) and humans (failing, non-failing). All samples were sequenced after treatment with exonuclease RNase R or a mock treatment and >9000 candidate circRNAs were detected for each species. Additionally, we performed separate isolation of nuclear and cytoplasmic RNA and co-immunoprecipitated RNA interacting with endogenous argonaute 2 (Ago2) in primary cardiac myocytes. We found circRNAs to be significantly enriched in the cytoplasm compared to linear transcripts and to have a similar level of association with Ago2. Notably in all three species we observed dozens of circRNAs arising from the titin (Ttn) gene, which is known to undergo highly complex alternative splicing during heart maturation. Correspondingly we observed extensive differential regulation of Ttn circRNAs between neonatal and adult rat hearts, suggesting that circRNA formation could be involved in the regulation of titin splicing. We expect that our inventory of cardiac circRNAs, as well as the information on their conservation and differential expression will provide an important basis for further studies addressing their function and suitability as biomarkers.


Asunto(s)
Miocardio/metabolismo , ARN/genética , Animales , Biomarcadores , Expresión Génica , Regulación de la Expresión Génica , Humanos , Espacio Intracelular/metabolismo , Ratones , ARN Circular , Complejo Silenciador Inducido por ARN/metabolismo , Ratas , Análisis de Secuencia de ARN , Transcripción Genética
13.
Mamm Genome ; 27(3-4): 111-21, 2016 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-26803617

RESUMEN

We performed exome sequencing for mutation discovery of an ENU (N-ethyl-N-nitrosourea)-derived mouse model characterized by significant elevated plasma alkaline phosphatase (ALP) activities in female and male mutant mice, originally named BAP014 (bone screen alkaline phosphatase #14). We identified a novel loss-of-function mutation within the Fam46a (family with sequence similarity 46, member A) gene (NM_001160378.1:c.469G>T, NP_001153850.1:p.Glu157*). Heterozygous mice of this mouse line (renamed Fam46a (E157*Mhda)) had significantly high ALP activities and apparently no other differences in morphology compared to wild-type mice. In contrast, homozygous Fam46a (E157*Mhda) mice showed severe morphological and skeletal abnormalities including short stature along with limb, rib, pelvis, and skull deformities with minimal trabecular bone and reduced cortical bone thickness in long bones. ALP activities of homozygous mutants were almost two-fold higher than in heterozygous mice. Fam46a is weakly expressed in most adult and embryonic tissues with a strong expression in mineralized tissues as calvaria and femur. The FAM46A protein is computationally predicted as a new member of the superfamily of nucleotidyltransferase fold proteins, but little is known about its function. Fam46a (E157*Mhda) mice are the first mouse model for a mutation within the Fam46a gene.


Asunto(s)
Enfermedades del Desarrollo Óseo/genética , Enfermedades del Desarrollo Óseo/patología , Codón sin Sentido , Exoma , Fosfatasa Alcalina/metabolismo , Animales , Huesos/metabolismo , Huesos/patología , Análisis Mutacional de ADN , Modelos Animales de Enfermedad , Activación Enzimática , Femenino , Expresión Génica , Genotipo , Secuenciación de Nucleótidos de Alto Rendimiento , Masculino , Ratones , Ratones Noqueados , Fenotipo
14.
Mol Syst Biol ; 11(7): 819, 2015 Jul 14.
Artículo en Inglés | MEDLINE | ID: mdl-26174933

RESUMEN

The extracellular matrix (ECM) is a key regulator of tissue morphogenesis and repair. However, its composition and architecture are not well characterized. Here, we monitor remodeling of the extracellular niche in tissue repair in the bleomycin-induced lung injury mouse model. Mass spectrometry quantified 8,366 proteins from total tissue and bronchoalveolar lavage fluid (BALF) over the course of 8 weeks, surveying tissue composition from the onset of inflammation and fibrosis to its full recovery. Combined analysis of proteome, secretome, and transcriptome highlighted post-transcriptional events during tissue fibrogenesis and defined the composition of airway epithelial lining fluid. To comprehensively characterize the ECM, we developed a quantitative detergent solubility profiling (QDSP) method, which identified Emilin-2 and collagen-XXVIII as novel constituents of the provisional repair matrix. QDSP revealed which secreted proteins interact with the ECM, and showed drastically altered association of morphogens to the insoluble matrix upon injury. Thus, our proteomic systems biology study assigns proteins to tissue compartments and uncovers their dynamic regulation upon lung injury and repair, potentially contributing to the development of anti-fibrotic strategies.


Asunto(s)
Matriz Extracelular/metabolismo , Lesión Pulmonar/inducido químicamente , Lesión Pulmonar/metabolismo , Proteómica/métodos , Animales , Bleomicina/toxicidad , Líquido del Lavado Bronquioalveolar , Colágeno/metabolismo , Modelos Animales de Enfermedad , Femenino , Regulación de la Expresión Génica , Glicoproteínas/metabolismo , Pulmón/metabolismo , Pulmón/fisiopatología , Lesión Pulmonar/patología , Ratones
15.
Am J Hum Genet ; 91(6): 1144-9, 2012 Dec 07.
Artículo en Inglés | MEDLINE | ID: mdl-23176820

RESUMEN

Neurodegeneration with brain iron accumulation (NBIA) is a group of genetic disorders characterized by abnormal iron deposition in the basal ganglia. We report that de novo mutations in WDR45, a gene located at Xp11.23 and encoding a beta-propeller scaffold protein with a putative role in autophagy, cause a distinctive NBIA phenotype. The clinical features include early-onset global developmental delay and further neurological deterioration (parkinsonism, dystonia, and dementia developing by early adulthood). Brain MRI revealed evidence of iron deposition in the substantia nigra and globus pallidus. Males and females are phenotypically similar, an observation that might be explained by somatic mosaicism in surviving males and germline or somatic mutations in females, as well as skewing of X chromosome inactivation. This clinically recognizable disorder is among the more common forms of NBIA, and we suggest that it be named accordingly as beta-propeller protein-associated neurodegeneration.


Asunto(s)
Encéfalo/metabolismo , Proteínas Portadoras/genética , Exoma , Genes Ligados a X , Sobrecarga de Hierro/genética , Mutación , Fenotipo , Alelos , Secuencia de Aminoácidos , Secuencia de Bases , Encéfalo/patología , Femenino , Orden Génico , Secuenciación de Nucleótidos de Alto Rendimiento , Humanos , Sobrecarga de Hierro/diagnóstico , Imagen por Resonancia Magnética , Masculino , Datos de Secuencia Molecular
16.
J Hepatol ; 61(6): 1312-20, 2014 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-25135868

RESUMEN

BACKGROUND & AIMS: Hepatoblastoma (HB) is the most common childhood liver cancer and occasionally presents with histological and clinical features reminiscent of hepatocellular carcinoma (HCC). Identification of molecular mechanisms that drive the neoplastic continuation towards more aggressive HCC phenotypes may help to guide the new stage of targeted therapies. METHODS: We performed comprehensive studies on genetic and chromosomal alterations as well as candidate gene function and their clinical relevance. RESULTS: Whole-exome sequencing identified HB as a genetically very simple tumour (2.9 mutations per tumour) with recurrent mutations in ß-catenin (CTNNB1) (12/15 cases) and the transcription factor NFE2L2 (2/15 cases). Their HCC-like progenies share the common CTNNB1 mutation, but additionally exhibit a significantly increased mutation number and chromosomal instability due to deletions of the genome guardians RAD17 and TP53, accompanied by telomerase reverse-transcriptase (TERT) promoter mutations. Targeted genotyping of 33 primary tumours and cell lines revealed CTNNB1, NFE2L2, and TERT mutations in 72.5%, 9.8%, and 5.9% of cases, respectively. All NFE2L2 mutations affected residues of the NFE2L2 protein that are recognized by the KEAP1/CUL3 complex for proteasomal degradation. Consequently, cells transfected with mutant NFE2L2 were insensitive to KEAP1-mediated downregulation of NFE2L2 signalling. Clinically, overexpression of the NFE2L2 target gene NQO1 in tumours was significantly associated with metastasis, vascular invasion, the adverse prognostic C2 gene signature, as well as poor outcome. CONCLUSIONS: Our study demonstrates the importance of CTNNB1 mutations and NFE2L2-KEAP1 pathway activation in HB development and defines loss of genomic stability and TERT promoter mutations as prominent characteristics of aggressive HB with HCC features.


Asunto(s)
Carcinoma Hepatocelular/genética , Genómica , Hepatoblastoma/genética , Neoplasias Hepáticas/genética , Análisis de Secuencia de ADN , Adulto , Biopsia , Carcinoma Hepatocelular/patología , Línea Celular Tumoral , Niño , ADN de Neoplasias/genética , Hepatoblastoma/patología , Humanos , Péptidos y Proteínas de Señalización Intracelular/genética , Proteína 1 Asociada A ECH Tipo Kelch , Neoplasias Hepáticas/patología , Mutación/genética , Factor 2 Relacionado con NF-E2/genética , Estudios Retrospectivos , Telomerasa/genética , beta Catenina/genética
17.
Am J Med Genet A ; 164A(8): 1976-80, 2014 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-24798461

RESUMEN

In patients with genetically heterogeneous disorders such as intellectual disability or epilepsy, exome sequencing is a powerful tool to elucidate the underlying genetic cause. Homozygous and compound heterozygous mutations in C12orf57 have recently been described to cause an autosomal recessive syndromic form of intellectual disability, including agenesis/hypoplasia of the corpus callosum, optic coloboma, and intractable seizures. Here, we report on two siblings from nonconsanguineous parents harboring two compound heterozygous loss-of-function mutations in C12orf57 identified by exome sequencing, including a novel nonsense mutation, and review the patients described in the literature.


Asunto(s)
Anomalías Congénitas/diagnóstico , Anomalías Congénitas/genética , Exoma , Heterocigoto , Mutación , Fenotipo , Hermanos , Niño , Coloboma/diagnóstico , Coloboma/genética , Cuerpo Calloso/patología , Facies , Femenino , Secuenciación de Nucleótidos de Alto Rendimiento , Humanos , Discapacidad Intelectual/diagnóstico , Discapacidad Intelectual/genética , Masculino , Convulsiones/diagnóstico , Convulsiones/genética , Síndrome
18.
Lancet ; 380(9854): 1674-82, 2012 Nov 10.
Artículo en Inglés | MEDLINE | ID: mdl-23020937

RESUMEN

BACKGROUND: The genetic cause of intellectual disability in most patients is unclear because of the absence of morphological clues, information about the position of such genes, and suitable screening methods. Our aim was to identify de-novo variants in individuals with sporadic non-syndromic intellectual disability. METHODS: In this study, we enrolled children with intellectual disability and their parents from ten centres in Germany and Switzerland. We compared exome sequences between patients and their parents to identify de-novo variants. 20 children and their parents from the KORA Augsburg Diabetes Family Study were investigated as controls. FINDINGS: We enrolled 51 participants from the German Mental Retardation Network. 45 (88%) participants in the case group and 14 (70%) in the control group had de-novo variants. We identified 87 de-novo variants in the case group, with an exomic mutation rate of 1·71 per individual per generation. In the control group we identified 24 de-novo variants, which is 1·2 events per individual per generation. More participants in the case group had loss-of-function variants than in the control group (20/51 vs 2/20; p=0·022), suggesting their contribution to disease development. 16 patients carried de-novo variants in known intellectual disability genes with three recurrently mutated genes (STXBP1, SYNGAP1, and SCN2A). We deemed at least six loss-of-function mutations in six novel genes to be disease causing. We also identified several missense alterations with potential pathogenicity. INTERPRETATION: After exclusion of copy-number variants, de-novo point mutations and small indels are associated with severe, sporadic non-syndromic intellectual disability, accounting for 45-55% of patients with high locus heterogeneity. Autosomal recessive inheritance seems to contribute little in the outbred population investigated. The large number of de-novo variants in known intellectual disability genes is only partially attributable to known non-specific phenotypes. Several patients did not meet the expected syndromic manifestation, suggesting a strong bias in present clinical syndrome descriptions. FUNDING: German Ministry of Education and Research, European Commission 7th Framework Program, and Swiss National Science Foundation.


Asunto(s)
Exoma/genética , Discapacidad Intelectual/genética , Mutación/genética , Estudios de Casos y Controles , Niño , Femenino , Humanos , Masculino
19.
Cell Rep ; 42(6): 112533, 2023 06 27.
Artículo en Inglés | MEDLINE | ID: mdl-37257449

RESUMEN

The acquisition of mesenchymal traits is considered a hallmark of breast cancer progression. However, the functional relevance of epithelial-to-mesenchymal transition (EMT) remains controversial and context dependent. Here, we isolate epithelial and mesenchymal populations from human breast cancer metastatic biopsies and assess their functional potential in vivo. Strikingly, progressively decreasing epithelial cell adhesion molecule (EPCAM) levels correlate with declining disease propagation. Mechanistically, we find that persistent EPCAM expression marks epithelial clones that resist EMT induction and propagate competitively. In contrast, loss of EPCAM defines clones arrested in a mesenchymal state, with concomitant suppression of tumorigenicity and metastatic potential. This dichotomy results from distinct clonal trajectories impacting global epigenetic programs that are determined by the interplay between human ZEB1 and its target GRHL2. Collectively, our results indicate that susceptibility to irreversible EMT restrains clonal propagation, whereas resistance to mesenchymal reprogramming sustains disease spread in multiple models of human metastatic breast cancer, including patient-derived cells in vivo.


Asunto(s)
Neoplasias de la Mama , Humanos , Femenino , Molécula de Adhesión Celular Epitelial , Neoplasias de la Mama/patología , Línea Celular Tumoral , Mama/metabolismo , Células Clonales/metabolismo , Transición Epitelial-Mesenquimal
20.
iScience ; 25(1): 103596, 2022 Jan 21.
Artículo en Inglés | MEDLINE | ID: mdl-34988410

RESUMEN

Childhood-onset myocardial hypertrophy and cardiomyopathic changes are associated with significant morbidity and mortality in early life, particularly in patients with Noonan syndrome, a multisystemic genetic disorder caused by autosomal dominant mutations in genes of the Ras-MAPK pathway. Although the cardiomyopathy associated with Noonan syndrome (NS-CM) shares certain cardiac features with the hypertrophic cardiomyopathy caused by mutations in sarcomeric proteins (HCM), such as pathological myocardial remodeling, ventricular dysfunction, and increased risk for malignant arrhythmias, the clinical course of NS-CM significantly differs from HCM. This suggests a distinct pathophysiology that remains to be elucidated. Here, through analysis of sarcomeric myosin conformational states, histopathology, and gene expression in left ventricular myocardial tissue from NS-CM, HCM, and normal hearts complemented with disease modeling in cardiomyocytes differentiated from patient-derived PTPN11 N308S/+ induced pluripotent stem cells, we demonstrate distinct disease phenotypes between NS-CM and HCM and uncover cell cycle defects as a potential driver of NS-CM.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA