Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Mol Ther Nucleic Acids ; 35(2): 102206, 2024 Jun 11.
Artículo en Inglés | MEDLINE | ID: mdl-38803421

RESUMEN

Huntington's disease (HD) is an incurable neurodegenerative disorder caused by genetic expansion of a CAG repeat sequence in one allele of the huntingtin (HTT) gene. Reducing expression of the mutant HTT (mutHTT) protein has remained a clear therapeutic goal, but reduction of wild-type HTT (wtHTT) is undesirable, as it compromises gene function and potential therapeutic efficacy. One promising allele-selective approach involves targeting the CAG repeat expansion with steric binding small RNAs bearing central mismatches. However, successful genetic encoding requires consistent placement of mismatches to the target within the small RNA guide sequence, which involves 5' processing precision by cellular enzymes. Here, we used small RNA sequencing (RNA-seq) to monitor the processing precision of a limited set of CAG repeat-targeted small RNAs expressed from multiple scaffold contexts. Small RNA-seq identified expression constructs with high-guide strand 5' processing precision and promising allele-selective inhibition of mutHTT. Transcriptome-wide mRNA-seq also identified an allele-selective small RNA with a favorable off-target profile. These results support continued investigation and optimization of genetically encoded repeat-targeted small RNAs for allele-selective HD gene therapy and underscore the value of sequencing methods to balance specificity with allele selectivity during the design and selection process.

2.
Mol Cell Endocrinol ; 583: 112128, 2024 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-38142853

RESUMEN

Pituitary gland function is regulated by the activity of various transcription factors that control cell fate decisions leading to cellular differentiation and hormone production. FOXO1 is necessary for normal somatotrope differentiation and function. Recent in vivo data implicate FOXO1 in the regulation of genes important for somatotrope differentiation including Gh1, Neurod4, and Pou1f1. In the current study, the somatotrope-like cell line GH3 was treated with a FOXO1 inhibitor, resulting in significant reduction in Neurod4 and Gh1 expression. Consistent with these findings, CRISPR/Cas9-mediated deletion of Foxo1 in GH3 cells significantly reduced expression of Gh1 and Neurod4. Chromatin immunoprecipitation sequencing identifies novel FOXO1 binding sites associated with the Neurod4, Gh1, and Pou1f1 genes. The FOXO1 binding site in the Neurod4 gene exhibits enhancer activity in somatotrope-like cells but not in gonadotrope-like cells. These data strongly suggest FOXO1 directly contributes to the transcriptional control of genes important for somatotrope differentiation.


Asunto(s)
Gonadotrofos , Hipófisis , Hipófisis/metabolismo , Proteína Forkhead Box O1/genética , Proteína Forkhead Box O1/metabolismo , Regulación de la Expresión Génica , Diferenciación Celular/genética , Factores de Transcripción/metabolismo , Gonadotrofos/metabolismo
3.
Hum Mol Genet ; 32(3): 386-401, 2023 01 13.
Artículo en Inglés | MEDLINE | ID: mdl-35981081

RESUMEN

De novo deleterious and heritable biallelic mutations in the DNA binding domain (DBD) of the transcription factor deformed epidermal autoregulatory factor 1 (DEAF1) result in a phenotypic spectrum of disorders termed DEAF1-associated neurodevelopmental disorders (DAND). RNA-sequencing using hippocampal RNA from mice with conditional deletion of Deaf1 in the central nervous system indicate that loss of Deaf1 activity results in the altered expression of genes involved in neuronal function, dendritic spine maintenance, development, and activity, with reduced dendritic spines in hippocampal regions. Since DEAF1 is not a dosage-sensitive gene, we assessed the dominant negative activity of previously identified de novo variants and a heritable recessive DEAF1 variant on selected DEAF1-regulated genes in 2 different cell models. While no altered gene expression was observed in cells over-expressing the recessive heritable variant, the gene expression profiles of cells over-expressing de novo variants resulted in similar gene expression changes as observed in CRISPR-Cas9-mediated DEAF1-deleted cells. Altered expression of DEAF1-regulated genes was rescued by exogenous expression of WT-DEAF1 but not by de novo variants in cells lacking endogenous DEAF1. De novo heterozygous variants within the DBD of DEAF1 were identified in 10 individuals with a phenotypic spectrum including autism spectrum disorder, developmental delays, sleep disturbance, high pain tolerance, and mild dysmorphic features. Functional assays demonstrate these variants alter DEAF1 transcriptional activity. Taken together, this study expands the clinical phenotypic spectrum of individuals with DAND, furthers our understanding of potential roles of DEAF1 on neuronal function, and demonstrates dominant negative activity of identified de novo variants.


Asunto(s)
Trastorno del Espectro Autista , Trastornos del Neurodesarrollo , Animales , Ratones , Proteínas de Unión al ADN/genética , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Trastornos del Neurodesarrollo/genética , ARN
4.
Hum Gene Ther ; 33(15-16): 829-839, 2022 08.
Artículo en Inglés | MEDLINE | ID: mdl-35726380

RESUMEN

Short hairpin RNAs, or short hairpin RNAs (shRNAs), are a proven tool for gene knockdown and a promising therapeutic approach for suppression of disease-associated genes. The efficient preparation of shRNA-expressing vectors can sometimes become a bottleneck due to the complexity of shRNA hairpin sequence and structure, especially for repetitive or high GC-content targets. Here, we present improved shRNA cloning and validation methods that enabled efficient and rapid cloning of several shRNAs targeting disease-associated repeat expansions, including GGGGCC, CAG, CTG, CCTG, and CGG into modified pLKO.1 vectors. Improvements included shRNA insert design and preparation, recombination-based cloning, and sequencing-based validation that included Sanger and nanopore long-read sequencing. This improved method should enable practical, efficient cloning of nearly any shRNA sequence.


Asunto(s)
Vectores Genéticos , Clonación Molecular , Técnicas de Silenciamiento del Gen , Vectores Genéticos/genética , ARN Interferente Pequeño/genética
5.
Nat Commun ; 12(1): 6591, 2021 11 15.
Artículo en Inglés | MEDLINE | ID: mdl-34782635

RESUMEN

CRISPR-Cas12a is a leading technology for development of model organisms, therapeutics, and diagnostics. These applications could benefit from chemical modifications that stabilize or tune enzyme properties. Here we chemically modify ribonucleotides of the AsCas12a CRISPR RNA 5' handle, a pseudoknot structure that mediates binding to Cas12a. Gene editing in human cells required retention of several native RNA residues corresponding to predicted 2'-hydroxyl contacts. Replacing these RNA residues with a variety of ribose-modified nucleotides revealed 2'-hydroxyl sensitivity. Modified 5' pseudoknots with as little as six out of nineteen RNA residues, with phosphorothioate linkages at remaining RNA positions, yielded heavily modified pseudoknots with robust cell-based editing. High trans activity was usually preserved with cis activity. We show that the 5' pseudoknot can tolerate near complete modification when design is guided by structural and chemical compatibility. Rules for modification of the 5' pseudoknot should accelerate therapeutic development and be valuable for CRISPR-Cas12a diagnostics.


Asunto(s)
Proteínas Bacterianas/genética , Proteínas Bacterianas/metabolismo , Proteínas Asociadas a CRISPR/química , Sistemas CRISPR-Cas , Endodesoxirribonucleasas/genética , Endodesoxirribonucleasas/metabolismo , Edición Génica , Ribosa/metabolismo , Proteínas Bacterianas/química , Proteínas Asociadas a CRISPR/genética , Proteínas Asociadas a CRISPR/metabolismo , Endodesoxirribonucleasas/química , Células HEK293 , Humanos , Ácidos Nucleicos , Patología Molecular/métodos , ARN , ARN Guía de Kinetoplastida/genética , Ribosa/química
6.
Behav Brain Res ; 380: 112383, 2020 02 17.
Artículo en Inglés | MEDLINE | ID: mdl-31783086

RESUMEN

Deleterious mutations within the DNA binding domain of the transcription factor deformed epidermal autoregulatory factor 1 (DEAF1) result in a phenotypic spectrum of neurodevelopmental disorders including intellectual disabilities and autism spectrum disorders. While whole animal deletion of Deaf1 in mice is lethal, mice with conditional disruption of the gene in neuronal precursor cells can display memory deficits and increased anxiety-like behavior. This study aimed to further characterize learning and memory alterations and assess changes in marble burying activity and hippocampal size in mice with conditional deletion of Deaf1. Mice lacking DEAF1 in the CNS (NKO) displayed reduced memory in both contextual fear conditioning and a 3-day massed trials Morris water maze paradigm. NKO mice had reduced marble burying activity in full cage marble burying tests. Using a half-cage marble test, NKO mice again buried fewer marbles and spent significantly more time on the side of the cage away from the marbles compared to control animals. The area of the dorsal hippocampus of NKO mice was decreased compared to control and animals with a single Deaf1 allele. These results continue to establish the importance of DEAF1 in cognitive behavior and provide new evidence that DEAF1 regulates hippocampal morphology.


Asunto(s)
Conducta Animal/fisiología , Condicionamiento Clásico/fisiología , Proteínas de Unión al ADN/fisiología , Hipocampo/patología , Aprendizaje por Laberinto/fisiología , Trastornos de la Memoria/genética , Trastornos de la Memoria/fisiopatología , Factores de Transcripción/fisiología , Animales , Proteínas de Unión al ADN/genética , Modelos Animales de Enfermedad , Masculino , Ratones , Ratones Noqueados , Factores de Transcripción/genética
7.
Genet Med ; 21(9): 2059-2069, 2019 09.
Artículo en Inglés | MEDLINE | ID: mdl-30923367

RESUMEN

PURPOSE: To investigate the effect of different DEAF1 variants on the phenotype of patients with autosomal dominant and recessive inheritance patterns and on DEAF1 activity in vitro. METHODS: We assembled a cohort of 23 patients with de novo and biallelic DEAF1 variants, described the genotype-phenotype correlation, and investigated the differential effect of de novo and recessive variants on transcription assays using DEAF1 and Eif4g3 promoter luciferase constructs. RESULTS: The proportion of the most prevalent phenotypic features, including intellectual disability, speech delay, motor delay, autism, sleep disturbances, and a high pain threshold, were not significantly different in patients with biallelic and pathogenic de novo DEAF1 variants. However, microcephaly was exclusively observed in patients with recessive variants (p < 0.0001). CONCLUSION: We propose that different variants in the DEAF1 gene result in a phenotypic spectrum centered around neurodevelopmental delay. While a pathogenic de novo dominant variant would also incapacitate the product of the wild-type allele and result in a dominant-negative effect, a combination of two recessive variants would result in a partial loss of function. Because the clinical picture can be nonspecific, detailed phenotype information, segregation, and functional analysis are fundamental to determine the pathogenicity of novel variants and to improve the care of these patients.


Asunto(s)
Proteínas de Unión al ADN/genética , Discapacidades del Desarrollo/genética , Discapacidad Intelectual/genética , Microcefalia/genética , Factores de Transcripción/genética , Adolescente , Adulto , Alelos , Trastorno Autístico/genética , Trastorno Autístico/patología , Niño , Preescolar , Discapacidades del Desarrollo/patología , Exoma/genética , Femenino , Estudios de Asociación Genética , Humanos , Discapacidad Intelectual/patología , Trastornos del Desarrollo del Lenguaje/genética , Trastornos del Desarrollo del Lenguaje/patología , Masculino , Microcefalia/patología , Mutación Missense/genética , Adulto Joven
8.
Hum Mutat ; 38(12): 1774-1785, 2017 12.
Artículo en Inglés | MEDLINE | ID: mdl-28940898

RESUMEN

Deformed epidermal autoregulatory factor-1 (DEAF1), a transcription factor essential for central nervous system and early embryonic development, has recently been implicated in a series of intellectual disability-related neurodevelopmental anomalies termed, in this study, as DEAF1-associated neurodevelopmental disorder (DAND). We identified six potentially deleterious DEAF1 variants in a cohort of individuals with DAND via clinical exome sequencing (CES) and in silico analysis, including two novel de novo variants: missense variant c.634G > A p.Gly212Ser in the SAND domain and deletion variant c.913_915del p.Lys305del in the NLS domain, as well as c.676C > T p.Arg226Trp, c.700T > A p.Trp234Arg, c.737G > C p.Arg246Thr, and c.791A > C p.Gln264Pro. Luciferase reporter, immunofluorescence staining, and electrophoretic mobility shift assays revealed that these variants had decreased transcriptional repression activity at the DEAF1 promoter and reduced affinity to consensus DEAF1 DNA binding sequences. In addition, c.913_915del p.K305del localized primarily to the cytoplasm and interacted with wild-type DEAF1. Our results demonstrate that variants located within the SAND or NLS domains significantly reduce DEAF1 transcriptional regulatory activities and are thus, likely to contribute to the underlying clinical concerns in DAND patients. These findings illustrate the importance of experimental characterization of variants with uncertain significance identified by CES to assess their potential clinical significance and possible use in diagnosis.


Asunto(s)
Exoma/genética , Discapacidad Intelectual/genética , Trastornos del Neurodesarrollo/genética , Proteínas Nucleares/genética , Secuencia de Aminoácidos , Estudios de Cohortes , Proteínas de Unión al ADN , Humanos , Mutación , Proteínas Nucleares/metabolismo , Fenotipo , Regiones Promotoras Genéticas/genética , Alineación de Secuencia , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Secuenciación Completa del Genoma
9.
Mol Cell Endocrinol ; 401: 130-41, 2015 Feb 05.
Artículo en Inglés | MEDLINE | ID: mdl-25448846

RESUMEN

Cytokine-inducible SH2-containing protein (CIS) inhibits prolactin receptor (PRLR) signaling and acts as part of an E3 ubiquitin ligase complex through interactions with Elongin B/C proteins. This study aimed to identify CIS lysine ubiquitination sites and determine roles of ubiquitination and Elongin B/C interactions on CIS protein stability and PRLR signaling inhibition. Site-directed mutations revealed that CIS can be ubiquitinated on all six lysine residues. Elongin B/C interaction box mutation had no influence on CIS ubiquitination. CIS stability was increased by mutation of lysine residues and further enhanced by co-mutation of Elongin B/C interaction domain. CIS inhibition of STAT5B phosphorylation and casein promoter activation was dependent on CIS interactions with Elongin B/C, but not on CIS ubiquitination. These data indicate CIS protein stability is regulated through multiple mechanisms, including ubiquitination and interaction with Elongin B/C proteins, whereas CIS functional inhibition of PRLR signaling is dependent on the Elongin B/C interaction.


Asunto(s)
Lisina/genética , Receptores de Prolactina/metabolismo , Proteínas Supresoras de la Señalización de Citocinas/química , Proteínas Supresoras de la Señalización de Citocinas/metabolismo , Factores de Transcripción/metabolismo , Caseínas/genética , Elonguina , Células HEK293 , Humanos , Mutagénesis Sitio-Dirigida , Mutación , Estabilidad Proteica , Factor de Transcripción STAT5/metabolismo , Transducción de Señal , Proteínas Supresoras de la Señalización de Citocinas/genética , Ubiquitinación
10.
PLoS One ; 9(12): e115908, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25531106

RESUMEN

DEAF1 is a transcriptional regulator associated with autoimmune and neurological disorders and is known to bind TTCG motifs. To further ascertain preferred DEAF1 DNA ligands, we screened a random oligonucleotide library containing an "anchored" CpG motif. We identified a binding consensus that generally conformed to a repeated TTCGGG motif, with the two invariant CpG dinucleotides separated by 6-11 nucleotides. Alteration of the consensus surrounding the dual CpG dinucleotides, or cytosine methylation of a single CpG half-site, eliminated DEAF1 binding. A sequence within the Htr1a promoter that resembles the binding consensus but contains a single CpG motif was confirmed to have low affinity binding with DEAF1. A DEAF1 binding consensus was identified in the EIF4G3 promoter and ChIP assay showed endogenous DEAF1 was bound to the region. We conclude that DEAF1 preferentially binds variably spaced and unmethylated CpG-containing half-sites when they occur within an appropriate consensus.


Asunto(s)
Islas de CpG/genética , Metilación de ADN , Factor 4G Eucariótico de Iniciación/genética , Regulación de la Expresión Génica , Proteínas Nucleares/metabolismo , Motivos de Nucleótidos/genética , Regiones Promotoras Genéticas/genética , Sitios de Unión , Inmunoprecipitación de Cromatina , Proteínas de Unión al ADN , Factor 4G Eucariótico de Iniciación/metabolismo , Humanos , Reacción en Cadena de la Polimerasa , Unión Proteica , Factores de Transcripción
11.
Am J Hum Genet ; 94(5): 649-61, 2014 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-24726472

RESUMEN

Recently, we identified in two individuals with intellectual disability (ID) different de novo mutations in DEAF1, which encodes a transcription factor with an important role in embryonic development. To ascertain whether these mutations in DEAF1 are causative for the ID phenotype, we performed targeted resequencing of DEAF1 in an additional cohort of over 2,300 individuals with unexplained ID and identified two additional individuals with de novo mutations in this gene. All four individuals had severe ID with severely affected speech development, and three showed severe behavioral problems. DEAF1 is highly expressed in the CNS, especially during early embryonic development. All four mutations were missense mutations affecting the SAND domain of DEAF1. Altered DEAF1 harboring any of the four amino acid changes showed impaired transcriptional regulation of the DEAF1 promoter. Moreover, behavioral studies in mice with a conditional knockout of Deaf1 in the brain showed memory deficits and increased anxiety-like behavior. Our results demonstrate that mutations in DEAF1 cause ID and behavioral problems, most likely as a result of impaired transcriptional regulation by DEAF1.


Asunto(s)
Discapacidad Intelectual/genética , Trastornos Mentales/genética , Proteínas Nucleares/genética , Trastornos del Habla/genética , Secuencia de Aminoácidos , Animales , Niño , Estudios de Cohortes , Análisis Mutacional de ADN , Proteínas de Unión al ADN , Femenino , Humanos , Masculino , Ratones , Ratones Noqueados , Datos de Secuencia Molecular , Mutación , Estructura Terciaria de Proteína/genética , Factores de Transcripción
12.
Cell Tissue Res ; 355(1): 103-9, 2014 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-24057878

RESUMEN

The epithelial sodium channel (ENaC) is involved in Na(+) responses such as Na(+) absorption and salt taste. The alpha ENaC subunit (α-ENaC) is expressed in the skin of both the adult and larval (tadpole) bullfrog. α-ENaC expression in the developing bullfrog embryo has not been previously investigated. In this study, the expression of α-ENaC at various stages (Sts.) of bullfrog embryonic development is assessed by western blot and immunofluorescence analysis. Bullfrog α-ENaC (α-fENaC) protein was detected by western blot in embryos at Sts. (Gosner/Shumway) 19, 21 and 25. Immunofluorescence studies indicate that α-fENaC was localized to the embryonic cement glands at St. 18 (muscular response), St. 19 (heart beat) and St. 21 (mouth open and/or cornea transparent), to the external gills at St. 21 and to the outermost cell-layer of the skin at St. 25 (operculum complete). The function(s) of ENaC in these embryonic structures remain to be elucidated.


Asunto(s)
Embrión no Mamífero/metabolismo , Canales Epiteliales de Sodio/análisis , Branquias/embriología , Rana catesbeiana/embriología , Piel/embriología , Animales , Western Blotting , Embrión no Mamífero/química , Embrión no Mamífero/citología , Canales Epiteliales de Sodio/genética , Técnica del Anticuerpo Fluorescente , Expresión Génica , Branquias/química , Branquias/ultraestructura , Piel/química , Piel/ultraestructura
13.
Acta Histochem ; 115(4): 357-62, 2013 May.
Artículo en Inglés | MEDLINE | ID: mdl-23072797

RESUMEN

The epithelial Na channel (ENaC) plays an essential role in sodium transport across epithelia such as adult frog skin. Transport across the skin, measured as short-circuit current (SCC), is blocked by amiloride. Bullfrog alpha-ENaC (α-fENaC) is expressed in adult bullfrog skin, and the SCC across this skin is blocked by amiloride. In contrast, an amiloride-blockable SCC is not detected in larval bullfrog skin, even though it expresses α-fENaC. We examined the subcellular localization of α-ENaC in such larval and adult skins. Immunofluorescent and immunoelectron microscopy of apical cells in the larval epidermis revealed α-fENaC localization within intracellular vesicles, but not in the plasma membrane. In contrast, in adult skin α-fENaC was localized to the apical-side membrane and to intracellular vesicles in Stratum granulosum cells. This may support the view that amiloride-blockable SCC is absent from larval skin, but is present in adult skin.


Asunto(s)
Amilorida/farmacología , Vesículas Citoplasmáticas/metabolismo , Rana catesbeiana/metabolismo , Piel/metabolismo , Animales , Western Blotting , Membrana Celular/metabolismo , Células Cultivadas , Células Epiteliales/efectos de los fármacos , Células Epiteliales/metabolismo , Células Epiteliales/ultraestructura , Canales Epiteliales de Sodio/efectos de los fármacos , Canales Epiteliales de Sodio/metabolismo , Canales Epiteliales de Sodio/ultraestructura , Técnica del Anticuerpo Fluorescente , Larva , Piel/efectos de los fármacos , Piel/ultraestructura
14.
PLoS One ; 7(3): e33404, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22442688

RESUMEN

Deformed Epidermal Autoregulatory Factor 1 (DEAF1) is a transcription factor linked to suicide, cancer, autoimmune disorders and neural tube defects. To better understand the role of DEAF1 in protein interaction networks, a GST-DEAF1 fusion protein was used to isolate interacting proteins in mammalian cell lysates, and the XRCC6 (Ku70) and the XRCC5 (Ku80) subunits of DNA dependent protein kinase (DNA-PK) complex were identified by mass spectrometry, and the DNA-PK catalytic subunit was identified by immunoblotting. Interaction of DEAF1 with Ku70 and Ku80 was confirmed to occur within cells by co-immunoprecipitation of epitope-tagged proteins, and was mediated through interaction with the Ku70 subunit. Using in vitro GST-pulldowns, interaction between DEAF1 and the Ku70 subunit was mapped to the DEAF1 DNA binding domain and the C-terminal Bax-binding region of Ku70. In transfected cells, DEAF1 and Ku70 colocalized to the nucleus, but Ku70 could not relocalize a mutant cytoplasmic form of DEAF1 to the nucleus. Using an in vitro kinase assay, DEAF1 was phosphorylated by DNA-PK in a DNA-independent manner. Electrophoretic mobility shift assays showed that DEAF1 or Ku70/Ku80 did not interfere with the DNA binding of each other, but DNA containing DEAF1 binding sites inhibited the DEAF1-Ku70 interaction. The data demonstrates that DEAF1 can interact with the DNA-PK complex through interactions of its DNA binding domain with the carboxy-terminal region of Ku70 that contains the Bax binding domain, and that DEAF1 is a potential substrate for DNA-PK.


Asunto(s)
Antígenos Nucleares/metabolismo , ADN Helicasas/metabolismo , Proteína Quinasa Activada por ADN/metabolismo , Proteínas de Unión al ADN/metabolismo , Proteínas Nucleares/metabolismo , Animales , Antígenos Nucleares/genética , Línea Celular , Núcleo Celular/genética , Núcleo Celular/metabolismo , ADN Helicasas/genética , Proteína Quinasa Activada por ADN/genética , Proteínas de Unión al ADN/genética , Haplorrinos , Humanos , Autoantígeno Ku , Mutación , Proteínas Nucleares/genética , Mapeo Peptídico , Fosforilación/genética , Unión Proteica/genética , Estructura Terciaria de Proteína , Factores de Transcripción
15.
J Comp Physiol B ; 176(4): 287-93, 2006 May.
Artículo en Inglés | MEDLINE | ID: mdl-16308722

RESUMEN

Amiloride-blockable Na(+) transport, measured as an amiloride-blockable short-circuit current (Am-SCC), is mediated by the epithelial Na(+) channel (ENaC). Am-SCC is not normally present in bullfrog tadpole skin, but when such skin is cultured with corticoids an amiloride-blockable Na transport appears. Prolactin (PRL) inhibits its corticoid-induced development. Using specific PCR primers for adult frog ENaC and RT-PCR, we investigated whether corticoids can induce all three ENaC subunits, and whether this expression of ENaC subunit(s) can be blocked by adding PRL with the corticoids. We found that (1) the sequences of the RT-PCR products obtained using primers for alpha-ENaC were identical between larval and adult skins, (2) the mRNAs for all three ENaC subunits were expressed in larval skin under normal conditions despite no amiloride-blockable Na(+) transport being detectable, (3) all three subunits were expressed in larval skins whether they were cultured with corticoids (amiloride-blockable Na transport present) or with corticoids supplemented with PRL (no amiloride-blockable Na transport present). An antibody against a peptide from the alpha-ENaC of adult bullfrog was localized to the apical cells of both larval and adult skins. Since no amiloride-blockable Na transport exists across larval skin under these conditions, these results suggest that ENaC protein was expressed prior to the onset of transport. ENaC may be in the plasma membrane in an inactivated form or, alternatively, within vesicles waiting to be inserted.


Asunto(s)
ARN Mensajero/metabolismo , Rana catesbeiana/metabolismo , Piel/metabolismo , Canales de Sodio/metabolismo , Amilorida/farmacología , Animales , Anticuerpos , Canales Epiteliales de Sodio , Larva/metabolismo , Receptores Purinérgicos P2/metabolismo , Receptores Purinérgicos P2X , Piel/embriología , Bloqueadores de los Canales de Sodio/farmacología , Canales de Sodio/genética
16.
J Biol Chem ; 279(31): 32692-9, 2004 Jul 30.
Artículo en Inglés | MEDLINE | ID: mdl-15161925

RESUMEN

Deformed epidermal autoregulatory factor-1 (DEAF-1) is a DNA-binding protein required for embryonic development and linked to clinical depression and suicidal behavior in humans. Although primarily nuclear, cytoplasmic localization of DEAF-1 has been observed, and this suggests the presence of a nuclear export signal (NES). Using a series of fluorescent fusion proteins, an NES with a novel spacing of leucines (LXLX(6)LLX(5)LX(2)L) was identified near the COOH-terminal MYND domain at amino acids 454-476. The NES was leptomycin B-sensitive and mutation of the leucine residues decreased or eliminated nuclear export activity. In vitro pull downs and an in vivo fluorescent protein interaction assay identified a DEAF-1/DEAF-1 protein interaction domain within the NES region. DNA binding had been previously mapped to a positively charged surface patch in the novel DNA binding fold called the "SAND" domain. A second protein-protein interaction domain was identified at amino acids 243-306 that contains the DNA-binding SAND domain and also an adjacent zinc binding motif and a monopartite nuclear localization signal (NLS). Deletion of these adjacent sequences or mutation of the conserved cysteines or histidine in the zinc binding motif not only inhibits protein interaction but also eliminates DNA binding, demonstrating that DEAF-1 protein-protein interaction is required for DNA recognition. The identification of an NES and NLS provides a basis for the control of DEAF-1 subcellular localization and function, whereas the requirement of protein-protein interaction by the SAND domain appears to be unique among this class of transcription factors.


Asunto(s)
Núcleo Celular/metabolismo , Proteínas de Unión al ADN/metabolismo , Transporte Activo de Núcleo Celular , Secuencias de Aminoácidos , Secuencia de Aminoácidos , Animales , Secuencia de Bases , Western Blotting , Cisteína/química , Citoplasma/metabolismo , ADN/química , ADN/metabolismo , Ácidos Grasos Insaturados/farmacología , Glutatión Transferasa/metabolismo , Histidina/química , Humanos , Leucina/química , Microscopía Fluorescente , Datos de Secuencia Molecular , Mutación , Señales de Localización Nuclear , Proteínas Nucleares , Péptidos/química , Plásmidos/metabolismo , Pruebas de Precipitina , Unión Proteica , Conformación Proteica , Estructura Terciaria de Proteína , Proteínas Recombinantes de Fusión/metabolismo , Factores de Transcripción , Transcripción Genética , Transfección , Zinc/química
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...