Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 28
Filtrar
Más filtros

Bases de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
PLoS Genet ; 19(4): e1010710, 2023 04.
Artículo en Inglés | MEDLINE | ID: mdl-37068109

RESUMEN

Prader-Willi syndrome (PWS) is a multisystem disorder with neurobehavioral, metabolic, and hormonal phenotypes, caused by loss of expression of a paternally-expressed imprinted gene cluster. Prior evidence from a PWS mouse model identified abnormal pancreatic islet development with retention of aged insulin and deficient insulin secretion. To determine the collective roles of PWS genes in ß-cell biology, we used genome-editing to generate isogenic, clonal INS-1 insulinoma lines having 3.16 Mb deletions of the silent, maternal- (control) and active, paternal-allele (PWS). PWS ß-cells demonstrated a significant cell autonomous reduction in basal and glucose-stimulated insulin secretion. Further, proteomic analyses revealed reduced levels of cellular and secreted hormones, including all insulin peptides and amylin, concomitant with reduction of at least ten endoplasmic reticulum (ER) chaperones, including GRP78 and GRP94. Critically, differentially expressed genes identified by whole transcriptome studies included reductions in levels of mRNAs encoding these secreted peptides and the group of ER chaperones. In contrast to the dosage compensation previously seen for ER chaperones in Grp78 or Grp94 gene knockouts or knockdown, compensation is precluded by the stress-independent deficiency of ER chaperones in PWS ß-cells. Consistent with reduced ER chaperones levels, PWS INS-1 ß-cells are more sensitive to ER stress, leading to earlier activation of all three arms of the unfolded protein response. Combined, the findings suggest that a chronic shortage of ER chaperones in PWS ß-cells leads to a deficiency of protein folding and/or delay in ER transit of insulin and other cargo. In summary, our results illuminate the pathophysiological basis of pancreatic ß-cell hormone deficits in PWS, with evolutionary implications for the multigenic PWS-domain, and indicate that PWS-imprinted genes coordinate concerted regulation of ER chaperone biosynthesis and ß-cell secretory pathway function.


Asunto(s)
Síndrome de Prader-Willi , Ratones , Animales , Síndrome de Prader-Willi/genética , Síndrome de Prader-Willi/metabolismo , Secreción de Insulina/genética , Chaperón BiP del Retículo Endoplásmico , Regulación hacia Abajo , Proteómica , Chaperonas Moleculares/genética , Chaperonas Moleculares/metabolismo , Insulina/genética , Insulina/metabolismo , Retículo Endoplásmico/genética , Retículo Endoplásmico/metabolismo
2.
Am J Hum Genet ; 104(5): 815-834, 2019 05 02.
Artículo en Inglés | MEDLINE | ID: mdl-31031012

RESUMEN

We identified individuals with variations in ACTL6B, a component of the chromatin remodeling machinery including the BAF complex. Ten individuals harbored bi-allelic mutations and presented with global developmental delay, epileptic encephalopathy, and spasticity, and ten individuals with de novo heterozygous mutations displayed intellectual disability, ambulation deficits, severe language impairment, hypotonia, Rett-like stereotypies, and minor facial dysmorphisms (wide mouth, diastema, bulbous nose). Nine of these ten unrelated individuals had the identical de novo c.1027G>A (p.Gly343Arg) mutation. Human-derived neurons were generated that recaptured ACTL6B expression patterns in development from progenitor cell to post-mitotic neuron, validating the use of this model. Engineered knock-out of ACTL6B in wild-type human neurons resulted in profound deficits in dendrite development, a result recapitulated in two individuals with different bi-allelic mutations, and reversed on clonal genetic repair or exogenous expression of ACTL6B. Whole-transcriptome analyses and whole-genomic profiling of the BAF complex in wild-type and bi-allelic mutant ACTL6B neural progenitor cells and neurons revealed increased genomic binding of the BAF complex in ACTL6B mutants, with corresponding transcriptional changes in several genes including TPPP and FSCN1, suggesting that altered regulation of some cytoskeletal genes contribute to altered dendrite development. Assessment of bi-alleic and heterozygous ACTL6B mutations on an ACTL6B knock-out human background demonstrated that bi-allelic mutations mimic engineered deletion deficits while heterozygous mutations do not, suggesting that the former are loss of function and the latter are gain of function. These results reveal a role for ACTL6B in neurodevelopment and implicate another component of chromatin remodeling machinery in brain disease.


Asunto(s)
Actinas/genética , Proteínas Cromosómicas no Histona/genética , Proteínas de Unión al ADN/genética , Dendritas/patología , Epilepsia/etiología , Células Madre Pluripotentes Inducidas/patología , Mutación , Trastornos del Neurodesarrollo/etiología , Neuronas/patología , Adulto , Niño , Preescolar , Cromatina/genética , Cromatina/metabolismo , Dendritas/metabolismo , Epilepsia/patología , Femenino , Humanos , Células Madre Pluripotentes Inducidas/metabolismo , Lactante , Masculino , Trastornos del Neurodesarrollo/patología , Neuronas/metabolismo , Adulto Joven
3.
Am J Med Genet A ; 185(7): 2046-2055, 2021 07.
Artículo en Inglés | MEDLINE | ID: mdl-33949097

RESUMEN

Guanylate cyclase 2C (GC-C), encoded by the GUCY2C gene, is implicated in hereditary early onset chronic diarrhea. Several families with chronic diarrhea symptoms have been identified with autosomal dominant, gain-of-function mutations in GUCY2C. We have identified a Mennonite patient with a novel GUCY2C variant (c.2381A > T; p.Asp794Val) with chronic diarrhea and an extensive maternal family history of chronic diarrhea and bowel dilatation. Functional studies including co-segregation analysis showed that all family members who were heterozygous for this variant had GI-related symptoms. HEK-293 T cells expressing the Asp794Val GC-C variant showed increased cGMP production when stimulated with Escherichia coli heat-stable enterotoxin STp (HST), which was reversed when 5-(3-Bromophenyl)-5,11-dihydro-1,3-dimethyl-1H-indeno[2',1':5,6]pyrido[2,3-d]pyrimidine-2,4,6(3H)-trione (BPIPP; a GC-C inhibitor) was used. In addition, cystic fibrosis transmembrane conductance regulator (CFTR) activity measured with SPQ fluorescence assay was increased in these cells after treatment with HST, indicating a crucial role for CFTR activity in the pathogenesis of this disorder. These results support pathogenicity of the GC-C Asp794Val variant as a cause of chronic diarrhea in this family. Furthermore, this work identifies potential candidate drug, GC-C inhibitor BPIPP, to treat diarrhea caused by this syndrome.


Asunto(s)
Regulador de Conductancia de Transmembrana de Fibrosis Quística/genética , Diarrea/genética , Predisposición Genética a la Enfermedad , Receptores de Enterotoxina/genética , Adolescente , Toxinas Bacterianas/antagonistas & inhibidores , Toxinas Bacterianas/genética , Niño , Diarrea/tratamiento farmacológico , Diarrea/patología , Enterotoxinas/antagonistas & inhibidores , Enterotoxinas/genética , Proteínas de Escherichia coli/antagonistas & inhibidores , Proteínas de Escherichia coli/genética , Femenino , Mutación con Ganancia de Función/genética , Células HEK293 , Compuestos Heterocíclicos de 4 o más Anillos/farmacología , Humanos , Masculino , Linaje , Adulto Joven
4.
Hum Mol Genet ; 24(11): 3238-47, 2015 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-25721401

RESUMEN

Acyl-CoA dehydrogenase 9 (ACAD9) is an assembly factor for mitochondrial respiratory chain Complex I (CI), and ACAD9 mutations are recognized as a frequent cause of CI deficiency. ACAD9 also retains enzyme ACAD activity for long-chain fatty acids in vitro, but the biological relevance of this function remains controversial partly because of the tissue specificity of ACAD9 expression: high in liver and neurons and minimal in skin fibroblasts. In this study, we hypothesized that this enzymatic ACAD activity is required for full fatty acid oxidation capacity in cells expressing high levels of ACAD9 and that loss of this function is important in determining phenotype in ACAD9-deficient patients. First, we confirmed that HEK293 cells express ACAD9 abundantly. Then, we showed that ACAD9 knockout in HEK293 cells affected long-chain fatty acid oxidation along with Cl, both of which were rescued by wild type ACAD9. Further, we evaluated whether the loss of ACAD9 enzymatic fatty acid oxidation affects clinical severity in patients with ACAD9 mutations. The effects on ACAD activity of 16 ACAD9 mutations identified in 24 patients were evaluated using a prokaryotic expression system. We showed that there was a significant inverse correlation between residual enzyme ACAD activity and phenotypic severity of ACAD9-deficient patients. These results provide evidence that in cells where it is strongly expressed, ACAD9 plays a physiological role in fatty acid oxidation, which contributes to the severity of the phenotype in ACAD9-deficient patients. Accordingly, treatment of ACAD9 patients should aim at counteracting both CI and fatty acid oxidation dysfunctions.


Asunto(s)
Acil-CoA Deshidrogenasas/genética , Complejo I de Transporte de Electrón/metabolismo , Ácidos Grasos/metabolismo , Enfermedades Mitocondriales/enzimología , Acil-CoA Deshidrogenasas/deficiencia , Animales , Estudios de Asociación Genética , Células HEK293 , Humanos , Ratones , Enfermedades Mitocondriales/patología , Mutación Missense , Oxidación-Reducción , Multimerización de Proteína , Índice de Severidad de la Enfermedad
5.
Mamm Genome ; 24(5-6): 165-78, 2013 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-23609791

RESUMEN

Prader-Willi syndrome (PWS) occurs in about 1 in 15,000 individuals and is a contiguous gene disorder causing developmental disability, hyperphagia usually with obesity, and behavioral problems, including an increased incidence of psychiatric illness. The genomic imprinting that regulates allele-specific expression of PWS candidate genes, the fact that multiple genes are typically inactivated, and the presence of many genes that produce functional RNAs rather than proteins has complicated the identification of the underlying genetic pathophysiology of PWS. Over 30 genetically modified mouse strains that have been developed and characterized have been instrumental in elucidating the genetic and epigenetic mechanisms for the regulation of PWS genes and in discovering their physiological functions. In 2011, a PWS Animal Models Working Group (AMWG) was established to generate discussions and facilitate exchange of ideas regarding the best use of PWS animal models. Here, we summarize the goals of the AMWG, describe current animal models of PWS, and make recommendations for strategies to maximize the utility of animal models and for the development and use of new animal models of PWS.


Asunto(s)
Modelos Animales de Enfermedad , Ratones , Síndrome de Prader-Willi/genética , Animales , Humanos , Ratones/genética , Ratones/metabolismo , Síndrome de Prader-Willi/metabolismo
6.
Nature ; 447(7141): 167-77, 2007 May 10.
Artículo en Inglés | MEDLINE | ID: mdl-17495919

RESUMEN

We report a high-quality draft of the genome sequence of the grey, short-tailed opossum (Monodelphis domestica). As the first metatherian ('marsupial') species to be sequenced, the opossum provides a unique perspective on the organization and evolution of mammalian genomes. Distinctive features of the opossum chromosomes provide support for recent theories about genome evolution and function, including a strong influence of biased gene conversion on nucleotide sequence composition, and a relationship between chromosomal characteristics and X chromosome inactivation. Comparison of opossum and eutherian genomes also reveals a sharp difference in evolutionary innovation between protein-coding and non-coding functional elements. True innovation in protein-coding genes seems to be relatively rare, with lineage-specific differences being largely due to diversification and rapid turnover in gene families involved in environmental interactions. In contrast, about 20% of eutherian conserved non-coding elements (CNEs) are recent inventions that postdate the divergence of Eutheria and Metatheria. A substantial proportion of these eutherian-specific CNEs arose from sequence inserted by transposable elements, pointing to transposons as a major creative force in the evolution of mammalian gene regulation.


Asunto(s)
Evolución Molecular , Genoma/genética , Genómica , Zarigüeyas/genética , Animales , Composición de Base , Secuencia Conservada/genética , Elementos Transponibles de ADN/genética , Humanos , Polimorfismo de Nucleótido Simple/genética , Biosíntesis de Proteínas , Sintenía/genética , Inactivación del Cromosoma X/genética
7.
Hum Mol Genet ; 19(7): 1153-64, 2010 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-20053671

RESUMEN

The loss of HBII-52 and related C/D box small nucleolar RNA (snoRNA) expression units have been implicated as a cause for the Prader-Willi syndrome (PWS). We recently found that the C/D box snoRNA HBII-52 changes the alternative splicing of the serotonin receptor 2C pre-mRNA, which is different from the traditional C/D box snoRNA function in non-mRNA methylation. Using bioinformatic predictions and experimental verification, we identified five pre-mRNAs (DPM2, TAF1, RALGPS1, PBRM1 and CRHR1) containing alternative exons that are regulated by MBII-52, the mouse homolog of HBII-52. Analysis of a single member of the MBII-52 cluster of snoRNAs by RNase protection and northern blot analysis shows that the MBII-52 expressing unit generates shorter RNAs that originate from the full-length MBII-52 snoRNA through additional processing steps. These novel RNAs associate with hnRNPs and not with proteins associated with canonical C/D box snoRNAs. Our data indicate that not a traditional C/D box snoRNA MBII-52, but a processed version lacking the snoRNA stem is the predominant MBII-52 RNA missing in PWS. This processed snoRNA functions in alternative splice-site selection. Its substitution could be a therapeutic principle for PWS.


Asunto(s)
Empalme Alternativo , Síndrome de Prader-Willi/genética , ARN Nucleolar Pequeño , Receptor de Serotonina 5-HT2C/genética , Animales , Regulación de la Expresión Génica , Ribonucleoproteínas Nucleares Heterogéneas/metabolismo , Ratones , Edición de ARN , Precursores del ARN
8.
Am J Physiol Endocrinol Metab ; 300(5): E909-22, 2011 May.
Artículo en Inglés | MEDLINE | ID: mdl-21343540

RESUMEN

Prader-Willi syndrome (PWS) is a multisystem disorder caused by genetic loss of function of a cluster of imprinted, paternally expressed genes. Neonatal failure to thrive in PWS is followed by childhood-onset hyperphagia and obesity among other endocrine and behavioral abnormalities. PWS is typically assumed to be caused by an unknown hypothalamic-pituitary dysfunction, but the underlying pathogenesis remains unknown. A transgenic deletion mouse model (TgPWS) has severe failure to thrive, with very low levels of plasma insulin and glucagon in fetal and neonatal life prior to and following onset of progressive hypoglycemia. In this study, we tested the hypothesis that primary deficits in pancreatic islet development or function may play a fundamental role in the TgPWS neonatal phenotype. Major pancreatic islet hormones (insulin, glucagon) were decreased in TgPWS mice, consistent with plasma levels. Immunohistochemical analysis of the pancreas demonstrated disrupted morphology of TgPWS islets, with reduced α- and ß-cell mass arising from an increase in apoptosis. Furthermore, in vivo and in vitro studies show that the rate of insulin secretion is significantly impaired in TgPWS ß-cells. In TgPWS pancreas, mRNA levels for genes encoding all pancreatic hormones, other secretory factors, and the ISL1 transcription factor are upregulated by either a compensatory response to plasma hormone deficiencies or a primary effect of a deleted gene. Our findings identify a cluster of imprinted genes required for the development, survival, coordinate regulation of genes encoding hormones, and secretory function of pancreatic endocrine cells, which may underlie the neonatal phenotype of the TgPWS mouse model.


Asunto(s)
Islotes Pancreáticos/crecimiento & desarrollo , Islotes Pancreáticos/fisiología , Síndrome de Prader-Willi/patología , Animales , Glucemia/metabolismo , Péptido C/metabolismo , Caspasa 3/metabolismo , Proliferación Celular , Femenino , Eliminación de Gen , Glucagón/sangre , Células Secretoras de Glucagón/fisiología , Inmunohistoquímica , Insulina/sangre , Insulina/metabolismo , Secreción de Insulina , Células Secretoras de Insulina/fisiología , Islotes Pancreáticos/metabolismo , Ratones , Ratones Noqueados , Análisis por Micromatrices , Síndrome de Prader-Willi/genética , Síndrome de Prader-Willi/metabolismo , Embarazo , ARN Mensajero/biosíntesis , ARN Mensajero/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Somatostatina/metabolismo
9.
J Clin Invest ; 118(6): 2316-24, 2008 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-18464933

RESUMEN

Intrauterine growth retardation (IUGR) has been linked to the onset of diseases in adulthood, including type 2 diabetes, and has been proposed to result from altered gene regulation patterns due to epigenetic modifications of developmental genes. To determine whether epigenetic modifications may play a role in the development of adult diabetes following IUGR, we used a rodent model of IUGR that expresses lower levels of Pdx1, a pancreatic and duodenal homeobox 1 transcription factor critical for beta cell function and development, which develops diabetes in adulthood. We found that expression of Pdx1 was permanently reduced in IUGR beta cells and underwent epigenetic modifications throughout development. The fetal IUGR state was characterized by loss of USF-1 binding at the proximal promoter of Pdx1, recruitment of the histone deacetylase 1 (HDAC1) and the corepressor Sin3A, and deacetylation of histones H3 and H4. Following birth, histone 3 lysine 4 (H3K4) was demethylated and histone 3 lysine 9 (H3K9) was methylated. During the neonatal period, these epigenetic changes and the reduction in Pdx1 expression could be reversed by HDAC inhibition. After the onset of diabetes in adulthood, the CpG island in the proximal promoter was methylated, resulting in permanent silencing of the Pdx1 locus. These results provide insight into the development of type 2 diabetes following IUGR and we believe they are the first to describe the ontogeny of chromatin remodeling in vivo from the fetus to the onset of disease in adulthood.


Asunto(s)
Diabetes Mellitus Tipo 2/genética , Epigénesis Genética , Retardo del Crecimiento Fetal/genética , Proteínas de Homeodominio/genética , Proteínas de Homeodominio/metabolismo , Transactivadores/genética , Transactivadores/metabolismo , Animales , Cromatina/química , Islas de CpG , Metilación de ADN , Histonas/metabolismo , Regiones Promotoras Genéticas , ARN Mensajero/metabolismo , Ratas , Ratas Sprague-Dawley , Factores de Transcripción/metabolismo
10.
Sci Rep ; 10(1): 13026, 2020 08 03.
Artículo en Inglés | MEDLINE | ID: mdl-32747801

RESUMEN

Prader-Willi (PWS) and Angelman (AS) syndromes are two clinically distinct imprinted disorders characterized by genetic abnormalities at 15q11-q13. Early diagnosis of both syndromes provides improved treatment and accurate genetic counseling. Whole blood (WB) is the most common DNA source of many methodologies to detect PWS and AS, however, the need of WB makes a massive screening difficult in newborns due to economic and technical limitations. The aim of this study was to adapt a Methylation-sensitive High-Resolution Melting (MS-HRM) approach from dried blood spot (DBS) samples, assessing the different DNA isolation techniques and diagnostic performance. Over a 1-year period, we collected 125 DBS cards, of which 45 had already been diagnosed by MS-HRM (20 PWS, 1 AS, and 24 healthy individuals). We tested three different DBS-DNA extraction techniques assessing the DNA concentration and quality, followed by MS-HRM and statistical comparison. Each DBS-DNA extraction method was capable of accuracy in detecting all PWS and AS individuals. However, the efficiency to detect healthy individuals varied according to methodology. In our experience, DNA extracted from DBS analyzed by the MS-HRM methodology provides an accurate approach for genetic screening of imprinting related disorders in newborns, offering several benefits compared to traditional whole blood methods.


Asunto(s)
Síndrome de Angelman/sangre , Síndrome de Angelman/genética , Metilación de ADN/genética , Pruebas con Sangre Seca , Tamizaje Neonatal , Desnaturalización de Ácido Nucleico/genética , Síndrome de Prader-Willi/sangre , Síndrome de Prader-Willi/genética , Autoantígenos/genética , Humanos , Recién Nacido , Proyectos Piloto , Ribonucleasa P/genética
11.
JCI Insight ; 5(20)2020 10 15.
Artículo en Inglés | MEDLINE | ID: mdl-33055427

RESUMEN

Phenylalanine hydroxylase-deficient (PAH-deficient) phenylketonuria (PKU) results in systemic hyperphenylalaninemia, leading to neurotoxicity with severe developmental disabilities. Dietary phenylalanine (Phe) restriction prevents the most deleterious effects of hyperphenylalaninemia, but adherence to diet is poor in adult and adolescent patients, resulting in characteristic neurobehavioral phenotypes. Thus, an urgent need exists for new treatments. Additionally, rodent models of PKU do not adequately reflect neurocognitive phenotypes, and thus there is a need for improved animal models. To this end, we have developed PAH-null pigs. After selection of optimal CRISPR/Cas9 genome-editing reagents by using an in vitro cell model, zygote injection of 2 sgRNAs and Cas9 mRNA demonstrated deletions in preimplantation embryos, with embryo transfer to a surrogate leading to 2 founder animals. One pig was heterozygous for a PAH exon 6 deletion allele, while the other was compound heterozygous for deletions of exon 6 and of exons 6-7. The affected pig exhibited hyperphenylalaninemia (2000-5000 µM) that was treatable by dietary Phe restriction, consistent with classical PKU, along with juvenile growth retardation, hypopigmentation, ventriculomegaly, and decreased brain gray matter volume. In conclusion, we have established a large-animal preclinical model of PKU to investigate pathophysiology and to assess new therapeutic interventions.


Asunto(s)
Hígado/metabolismo , Fenilalanina Hidroxilasa/genética , Fenilalanina/genética , Fenilcetonurias/genética , Adolescente , Adulto , Animales , Sistemas CRISPR-Cas/genética , Dieta , Modelos Animales de Enfermedad , Edición Génica , Humanos , Hígado/efectos de los fármacos , Fenotipo , Fenilalanina/metabolismo , Fenilalanina/farmacología , Fenilcetonurias/dietoterapia , Fenilcetonurias/metabolismo , Fenilcetonurias/patología , Porcinos
12.
PLoS Genet ; 2(10): e182, 2006 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-17069464

RESUMEN

Genomic imprinting, representing parent-specific expression of alleles at a locus, raises many questions about how--and especially why--epigenetic silencing of mammalian genes evolved. We present the first in-depth study of how a human imprinted domain evolved, analyzing a domain containing several imprinted genes that are involved in human disease. Using comparisons of orthologous genes in humans, marsupials, and the platypus, we discovered that the Prader-Willi/Angelman syndrome region on human Chromosome 15q was assembled only recently (105-180 million years ago). This imprinted domain arose after a region bearing UBE3A (Angelman syndrome) fused with an unlinked region bearing SNRPN (Prader-Willi syndrome), which had duplicated from the non-imprinted SNRPB/B'. This region independently acquired several retroposed gene copies and arrays of small nucleolar RNAs from different parts of the genome. In their original configurations, SNRPN and UBE3A are expressed from both alleles, implying that acquisition of imprinting occurred after their rearrangement and required the evolution of a control locus. Thus, the evolution of imprinting in viviparous mammals is ongoing.


Asunto(s)
Impresión Genómica/genética , Marsupiales/genética , Ornitorrinco/genética , Alelos , Animales , Autoantígenos/genética , Mapeo Cromosómico , Cromosomas de los Mamíferos/genética , Genoma Humano/genética , Humanos , Hibridación Fluorescente in Situ , Ratones , Ribonucleoproteínas Nucleares Pequeñas/genética , Análisis de Secuencia de ADN , Homología de Secuencia , Ubiquitina-Proteína Ligasas/genética , Proteínas Nucleares snRNP
13.
Nucleic Acids Res ; 34(4): 1205-15, 2006.
Artículo en Inglés | MEDLINE | ID: mdl-16500891

RESUMEN

FMR1 encodes an RNA-binding protein whose absence results in fragile X mental retardation. In most patients, the FMR1 gene is cytosine-methylated and transcriptionally inactive. NRF-1 and Sp1 are known to bind and stimulate the active, but not the methylated/silenced, FMR1 promoter. Prior analysis has implicated a CRE site in regulation of FMR1 in neural cells but the role of this site is controversial. We now show that a phospho-CREB/ATF family member is bound to this site in vivo. We also find that the histone acetyltransferases CBP and p300 are associated with active FMR1 but are lost at the hypoacetylated fragile X allele. Surprisingly, FMR1 is not cAMP-inducible and resides in a newly recognized subclass of CREB-regulated genes. We have also elucidated a role for NRF-2 as a regulator of FMR1 in vivo through a previously unrecognized and highly conserved recognition site in FMR1. NRF-1 and NRF-2 act additively while NRF-2 synergizes with CREB/ATF at FMR1's promoter. These data add FMR1 to the collection of genes controlled by both NRF-1 and NRF-2 and disfavor its membership in the immediate early response group of genes.


Asunto(s)
Factores de Transcripción Activadores/metabolismo , Proteína de Unión a Elemento de Respuesta al AMP Cíclico/metabolismo , Proteína de la Discapacidad Intelectual del Síndrome del Cromosoma X Frágil/genética , Factor de Transcripción de la Proteína de Unión a GA/metabolismo , Activación Transcripcional , Animales , Secuencia de Bases , Sitios de Unión , Línea Celular , Secuencia de Consenso , Secuencia Conservada , AMP Cíclico/metabolismo , Humanos , Datos de Secuencia Molecular , Fosforilación , Regiones Promotoras Genéticas , Ratas , Proteínas Represoras/metabolismo , Factores Estimuladores hacia 5'/metabolismo , Factores de Transcripción p300-CBP/metabolismo
14.
Nucleic Acids Res ; 33(15): 4740-53, 2005.
Artículo en Inglés | MEDLINE | ID: mdl-16116039

RESUMEN

The imprinted SNRPN locus is a complex transcriptional unit that encodes the SNURF and SmN polypeptides as well as multiple non-coding RNAs. SNRPN is located within the Prader-Willi and Angelman syndrome (PWS/AS) region that contains multiple imprinted genes, which are coordinately regulated by a bipartite imprinting center (IC). The SNRPN 5' region co-localizes with the PWS-IC and contains two DNase I hypersensitive sites, DHS1 at the SNRPN promoter, and DHS2 within intron 1, exclusively on the paternally inherited chromosome. We have examined DHS1 and DHS2 to identify cis- and trans-acting regulatory elements within the endogenous SNRPN 5' region. Analysis of DHS1 by in vivo footprinting and chromatin immunoprecipitation identified allele-specific interaction with multiple regulatory proteins, including NRF-1, which regulates genes involved in mitochondrial and metabolic functions. DHS2 acted as an enhancer of the SNRPN promoter and contained a highly conserved region that showed allele-specific interaction with unphosphorylated RNA polymerase II, YY1, Sp1 and NRF-1, further suggesting a key role for NRF-1 in regulation of the SNRPN locus. We propose that one or more of the regulatory elements identified in this study may also contribute to PWS-IC function.


Asunto(s)
Impresión Genómica , Proteínas Nucleares/genética , Elementos de Respuesta , Ribonucleoproteínas Nucleares Pequeñas/genética , Factores de Transcripción/metabolismo , Región de Flanqueo 5' , Síndrome de Angelman/genética , Autoantígenos , Secuencia de Bases , Sitios de Unión , Huella de ADN , Desoxirribonucleasa I/metabolismo , Elementos de Facilitación Genéticos , Histonas/metabolismo , Humanos , Intrones , Datos de Secuencia Molecular , Síndrome de Prader-Willi/genética , Regiones Promotoras Genéticas , ARN Polimerasa II/metabolismo , Proteínas Nucleares snRNP
15.
BMC Genomics ; 6: 157, 2005 Nov 09.
Artículo en Inglés | MEDLINE | ID: mdl-16280085

RESUMEN

BACKGROUND: Prader-Willi and Angelman syndrome (PWS and AS) patients typically have an approximately 5 Mb deletion of human chromosome 15q11-q13, of opposite parental origin. A mouse model of PWS and AS has a transgenic insertion-deletion (TgPWS/TgAS) of chromosome 7B/C subsequent to paternal or maternal inheritance, respectively. In this study, we define the deletion endpoints and examine the impact on expression of flanking genes. RESULTS: Using molecular and cytological methods we demonstrate that 13 imprinted and 11 non-imprinted genes are included in the TgPWS/TgAS deletion. Normal expression levels were found in TgPWS brain for genes extending 9.1- or 5.6-Mb centromeric or telomeric of the deletion, respectively. Our molecular cytological studies map the proximal deletion breakpoint between the Luzp2 and Siglec-H loci, and we show that overall mRNA levels of Luzp2 in TgPWS and TgAS brain are significantly reduced by 17%. Intriguingly, 5' Chrna7 shows 1.7-fold decreased levels in TgPWS and TgAS brain whereas there is a > or =15-fold increase in expression in neonatal liver and spleen of these mouse models. By isolating a Chrna7-Tg fusion transcript from TgAS mice, we mapped the telomeric deletion breakpoint in Chrna7 intron 4. CONCLUSION: Based on the extent of the deletion, TgPWS/TgAS mice are models for PWS/AS class I deletions. Other than for the first gene promoters immediately outside the deletion, since genes extending 5.6-9.1 Mb away from each end of the deletion show normal expression levels in TgPWS brain, this indicates that the transgene array does not induce silencing and there are no additional linked rearrangements. Using gene expression, non-coding conserved sequence (NCCS) and synteny data, we have genetically mapped a putative Luzp2 neuronal enhancer responsible for approximately 33% of allelic transcriptional activity. The Chrna7 results are explained by hypothesizing loss of an essential neuronal transcriptional enhancer required for approximately 80% of allelic Chrna7 promoter activity, while the Chrna7 promoter is upregulated in B lymphocytes by the transgene immunoglobulin enhancer. The mapping of a putative Chrna7 neuronal enhancer inside the deletion has significant implications for understanding the transcriptional regulation of this schizophrenia-susceptibility candidate gene.


Asunto(s)
Síndrome de Angelman/genética , Mapeo Cromosómico/métodos , Proteínas de Unión al ADN/genética , Elementos de Facilitación Genéticos , Eliminación de Gen , Neuronas/metabolismo , Síndrome de Prader-Willi/genética , Receptores Nicotínicos/genética , Transgenes , Alelos , Animales , Encéfalo/metabolismo , Centrómero/ultraestructura , Modelos Animales de Enfermedad , Regulación de la Expresión Génica , Silenciador del Gen , Impresión Genómica , Inmunoglobulinas/metabolismo , Hibridación Fluorescente in Situ , Intrones , Ratones , Modelos Genéticos , Análisis de Secuencia por Matrices de Oligonucleótidos , Mapeo Físico de Cromosoma , Regiones Promotoras Genéticas , Esquizofrenia/genética , Telómero/ultraestructura , Distribución Tisular , Transcripción Genética , Regulación hacia Arriba , Receptor Nicotínico de Acetilcolina alfa 7
16.
Brain Res ; 957(1): 42-5, 2002 Dec 06.
Artículo en Inglés | MEDLINE | ID: mdl-12443978

RESUMEN

As in Prader-Willi syndrome (PWS) infants, mouse models of PWS display failure-to-thrive during the neonatal period. In rodents, the hypothalamic neuropeptide, Neuropeptide Y (NPY) and Agouti-related peptide (AgrP) stimulate while alpha-melanocyte stimulating hormone (alpha-MSH) inhibits appetite. We hypothesized that altered expression of these neuropeptides in the hypothalamus may underlie the failure-to-thrive in PWS neonatal mice. To test this hypothesis we evaluated mRNA expression of Npy, Agrp, and Pomc by in situ hybridization in the hypothalamic arcuate nucleus (ARC) of 3-day-old female and male PWS neonates. The results showed that Agrp mRNA expression was decreased relative to wild-type (WT) controls in neonates of both sexes, while mRNA expression of Pomc was upregulated in PWS neonates. Since AgrP and the Pomc-derived peptide, alpha-MSH, are functional antagonists at melanocortin 4 receptors in the hypothalamic regulation of appetitive behavior, these results show that robust anorexigenic melanocortin signaling, may contribute to the failure-to-thrive in PWS neonatal mice.


Asunto(s)
Anorexia/metabolismo , Núcleo Arqueado del Hipotálamo/metabolismo , Neuropéptido Y/metabolismo , Síndrome de Prader-Willi/metabolismo , Proopiomelanocortina/metabolismo , Proteínas/metabolismo , Transducción de Señal , Proteína Relacionada con Agouti , Animales , Animales Recién Nacidos , Modelos Animales de Enfermedad , Femenino , Hibridación in Situ , Péptidos y Proteínas de Señalización Intercelular , Masculino , Ratones , Ratones Transgénicos , Neuropéptido Y/genética , Proopiomelanocortina/genética , Proteínas/genética , ARN Mensajero/metabolismo , Regulación hacia Arriba
17.
DNA Seq ; 15(4): 306-9, 2004 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-15620220

RESUMEN

Phenotypic analyses of a set of homozygous-lethal deletion mutants at the pink-eyed dilution (p) locus has resulted in the identification of p-linked obesity locus 1 (plo 1), distal to the p locus, as a locus involved in the modulation of body fat and/or affecting lipid metabolism in these mice. The plo 1 region maps to mouse chromosome 7 (MMU 7) between two genes, Gabrb3 and Ube3a, which have been used as anchor points to generate an integrated deletion and physical map of plo 1 that encompasses about 1.2-1.3 Mb. A deletion/physical map was constructed and the genomic DNA between the two loci was sequenced to identify genes mapping to this region. Data show that Atp10c, a novel type IV ATPase a putative phospholipid transporter, is the only coding unit in this region of the chromosome.


Asunto(s)
Adenosina Trifosfatasas/genética , Proteínas Portadoras/genética , Proteínas de la Membrana/genética , Proteínas de Transporte de Membrana/genética , Ubiquitina-Proteína Ligasas/genética , Animales , Deleción Cromosómica , Cromosomas Artificiales Bacterianos , Cromosomas Artificiales de Levadura , Cruzamientos Genéticos , Ratones , Mapeo Físico de Cromosoma
18.
PLoS One ; 7(5): e37138, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22623986

RESUMEN

The growth of a malignant tumor beyond a certain, limited size requires that it first develop an independent blood supply. In addition to providing metabolic support, this neovasculature also allows tumor cells to access the systemic circulation, thus facilitating metastatic dissemination. The neovasculature may originate either from normal blood vessels in close physical proximity to the tumor and/or from the recruitment of bone marrow-derived endothelial cell (EC) precursors. Recent studies have shown that human tumor vasculature ECs may also arise directly from tumor cells themselves and that the two populations have highly similar or identical karyotypes. We now show that, during the course of serial in vivo passage, these tumor-derived ECs (TDECs) progressively acquire more pronounced EC-like properties. These include higher-level expression of EC-specific genes and proteins, a greater capacity for EC-like behavior in vitro, and a markedly enhanced propensity to incorporate into the tumor vasculature. In addition, both vessel density and size are significantly increased in neoplasms derived from mixtures of tumor cells and serially passaged TDECs. A comparison of early- and late-passage TDECs using whole-genome single nucleotide polymorphism profiling showed the latter cells to have apparently evolved by a process of clonal expansion of a population with a distinct pattern of interstitial chromosomal gains and losses affecting a relatively small number of genes. The majority of these have established roles in vascular development, tumor suppression or epithelial-mesenchymal transition. These studies provide direct evidence that TDECs have a strong evolutionary capacity as a result of their inherent genomic instability. Consequently such cells might be capable of escaping anti-angiogenic cancer therapies by generating resistant populations.


Asunto(s)
Diferenciación Celular/fisiología , Linaje de la Célula/fisiología , Células Endoteliales/fisiología , Regulación Neoplásica de la Expresión Génica/fisiología , Neoplasias/irrigación sanguínea , Neovascularización Patológica/fisiopatología , Análisis Citogenético , Células Endoteliales/metabolismo , Regulación Neoplásica de la Expresión Génica/genética , Inestabilidad Genómica/fisiología , Humanos , Polimorfismo de Nucleótido Simple/genética , Reacción en Cadena en Tiempo Real de la Polimerasa
19.
PLoS One ; 7(5): e36505, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22574173

RESUMEN

Hereditary spastic paraplegias (HSPs) comprise a group of neurodegenerative disorders that are characterized by progressive spasticity of the lower extremities, due to axonal degeneration in the corticospinal motor tracts. HSPs are genetically heterogeneous and show autosomal dominant inheritance in ∼70-80% of cases, with additional cases being recessive or X-linked. The most common type of HSP is SPG4 with mutations in the SPAST gene, encoding spastin, which occurs in 40% of dominantly inherited cases and in ∼10% of sporadic cases. Both loss-of-function and dominant-negative mutation mechanisms have been described for SPG4, suggesting that precise or stoichiometric levels of spastin are necessary for biological function. Therefore, we hypothesized that regulatory mechanisms controlling expression of SPAST are important determinants of spastin biology, and if altered, could contribute to the development and progression of the disease. To examine the transcriptional and post-transcriptional regulation of SPAST, we used molecular phylogenetic methods to identify conserved sequences for putative transcription factor binding sites and miRNA targeting motifs in the SPAST promoter and 3'-UTR, respectively. By a variety of molecular methods, we demonstrate that SPAST transcription is positively regulated by NRF1 and SOX11. Furthermore, we show that miR-96 and miR-182 negatively regulate SPAST by effects on mRNA stability and protein level. These transcriptional and miRNA regulatory mechanisms provide new functional targets for mutation screening and therapeutic targeting in HSP.


Asunto(s)
Adenosina Trifosfatasas/genética , Regulación de la Expresión Génica , Tasa de Mutación , Paraplejía/enzimología , Paraplejía/genética , Transcripción Genética , Regiones no Traducidas 3'/genética , Animales , Secuencia de Bases , Línea Celular , Secuencia Conservada/genética , Sitios Genéticos/genética , Humanos , MicroARNs/genética , MicroARNs/metabolismo , Datos de Secuencia Molecular , Factor Nuclear 1 de Respiración/metabolismo , Motivos de Nucleótidos/genética , Paraplejía/diagnóstico , Paraplejía/terapia , Primates/genética , Regiones Promotoras Genéticas/genética , Proproteína Convertasas/metabolismo , Factores de Transcripción SOXC/metabolismo , Serina Endopeptidasas/metabolismo , Espastina
20.
Proc Natl Acad Sci U S A ; 103(32): 12039-44, 2006 Aug 08.
Artículo en Inglés | MEDLINE | ID: mdl-16882727

RESUMEN

A recently promoted genome evolution model posits that mammalian pseudogenes can regulate their founding source genes, and it thereby ascribes an important function to "junk DNA." This model arose from analysis of a serendipitous mouse mutant in which a transgene insertion/deletion caused severe polycystic kidney disease and osteogenesis imperfecta with approximately 80% perinatal lethality, when inherited paternally [Hirotsune, S., et al. (2003) Nature 423, 91-96]. The authors concluded that the transgene reduced the expression of a nearby transcribed and imprinted pseudogene, Mkrn1-p1. This reduction in chromosome 5-imprinted Mkrn1-p1 transcripts was proposed to destabilize the cognate chromosome 6 Mkrn1 source gene mRNA, with a partial reduction in one Mkrn1 isoform leading to the imprinted phenotype. Here, we show that 5' Mkrn1-p1 is fully methylated on both alleles, a pattern indicative of silenced chromatin, and that Mkrn1-p1 is not transcribed and therefore cannot stabilize Mkrn1 transcripts in trans. A small, truncated, rodent-specific Mkrn1 transcript explains the product erroneously attributed to Mkrn1-p1. Additionally, Mkrn1 expression is not imprinted, and 5' Mkrn1 is fully unmethylated. Finally, mice in which Mkrn1 has been directly disrupted show none of the phenotypes attributed to a partial reduction of Mkrn1. These data contradict the previous suggestions that Mkrn1-p1 is imprinted, and that either it or its source Mkrn1 gene relates to the original imprinted transgene phenotype. This study invalidates the data upon which the pseudogene trans-regulation model is based and therefore strongly supports the view that mammalian pseudogenes are evolutionary relics.


Asunto(s)
Regulación de la Expresión Génica , Impresión Genómica , Proteínas del Tejido Nervioso/genética , Seudogenes , Ribonucleoproteínas/genética , Animales , Secuencia de Bases , Cromatina/metabolismo , Epigénesis Genética , Evolución Molecular , Eliminación de Gen , Ratones , Modelos Genéticos , Datos de Secuencia Molecular , Transgenes
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA