Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
PLoS Genet ; 19(2): e1010659, 2023 02.
Artigo em Inglês | MEDLINE | ID: mdl-36848371

RESUMO

Copy number variations (CNVs) in the Neurexin 1 (NRXN1) gene, which encodes a presynaptic protein involved in neurotransmitter release, are some of the most frequently observed single-gene variants associated with autism spectrum disorder (ASD). To address the functional contribution of NRXN1 CNVs to behavioral phenotypes relevant to ASD, we carried out systematic behavioral phenotyping of an allelic series of Nrxn1 mouse models: one carrying promoter and exon 1 deletion abolishing Nrxn1α transcription, one carrying exon 9 deletion disrupting Nrxn1α protein translation, and one carrying an intronic deletion with no observable effect on Nrxn1α expression. We found that homozygous loss of Nrxn1α resulted in enhanced aggression in males, reduced affiliative social behaviors in females, and significantly altered circadian activities in both sexes. Heterozygous or homozygous loss of Nrxn1α affected the preference for social novelty in male mice, and notably, enhanced repetitive motor skills and motor coordination in both sexes. In contrast, mice bearing an intronic deletion of Nrxn1 did not display alterations in any of the behaviors assessed. These findings demonstrate the importance of Nrxn1α gene dosage in regulating social, circadian, and motor functions, and the variables of sex and genomic positioning of CNVs in the expression of autism-related phenotypes. Importantly, mice with heterozygous loss of Nrxn1, as found in numerous autistic individuals, show an elevated propensity to manifest autism-related phenotypes, supporting the use of models with this genomic architecture to study ASD etiology and assess additional genetic variants associated with autism.


Assuntos
Transtorno do Espectro Autista , Proteínas de Ligação ao Cálcio , Moléculas de Adesão de Célula Nervosa , Animais , Feminino , Masculino , Camundongos , Transtorno do Espectro Autista/genética , Variações do Número de Cópias de DNA/genética , Fenótipo , Comportamento Social , Moléculas de Adesão de Célula Nervosa/genética , Proteínas de Ligação ao Cálcio/genética
2.
J Clin Invest ; 131(20)2021 10 15.
Artigo em Inglês | MEDLINE | ID: mdl-34651584

RESUMO

CDKL5 deficiency disorder (CDD) is an early onset, neurodevelopmental syndrome associated with pathogenic variants in the X-linked gene encoding cyclin-dependent kinase-like 5 (CDKL5). CDKL5 has been implicated in neuronal synapse maturation, yet its postdevelopmental necessity and the reversibility of CDD-associated impairments remain unknown. We temporally manipulated endogenous Cdkl5 expression in male mice and found that postdevelopmental loss of CDKL5 disrupts numerous behavioral domains, hippocampal circuit communication, and dendritic spine morphology, demonstrating an indispensable role for CDKL5 in the adult brain. Accordingly, restoration of Cdkl5 after the early stages of brain development using a conditional rescue mouse model ameliorated CDD-related behavioral impairments and aberrant NMDA receptor signaling. These findings highlight the requirement of CDKL5 beyond early development, underscore the potential for disease reversal in CDD, and suggest that a broad therapeutic time window exists for potential treatment of CDD-related deficits.


Assuntos
Síndromes Epilépticas/psicologia , Proteínas Serina-Treonina Quinases/fisiologia , Espasmos Infantis/psicologia , Animais , Dendritos/patologia , Potenciais Evocados/fisiologia , Camundongos , Camundongos Endogâmicos C57BL , Receptores de N-Metil-D-Aspartato/fisiologia
3.
Protein Cell ; 11(8): 565-583, 2020 08.
Artigo em Inglês | MEDLINE | ID: mdl-32193873

RESUMO

The anterior pituitary gland drives highly conserved physiologic processes in mammalian species. These hormonally controlled processes are central to somatic growth, pubertal transformation, fertility, lactation, and metabolism. Current cellular models of mammalian anteiror pituitary, largely built on candidate gene based immuno-histochemical and mRNA analyses, suggest that each of the seven hormones synthesized by the pituitary is produced by a specific and exclusive cell lineage. However, emerging evidence suggests more complex relationship between hormone specificity and cell plasticity. Here we have applied massively parallel single-cell RNA sequencing (scRNA-seq), in conjunction with complementary imaging-based single-cell analyses of mRNAs and proteins, to systematically map both cell-type diversity and functional state heterogeneity in adult male and female mouse pituitaries at single-cell resolution and in the context of major physiologic demands. These quantitative single-cell analyses reveal sex-specific cell-type composition under normal pituitary homeostasis, identify an array of cells associated with complex complements of hormone-enrichment, and undercover non-hormone producing interstitial and supporting cell-types. Interestingly, we also identified a Pou1f1-expressing cell population that is characterized by a unique multi-hormone gene expression profile. In response to two well-defined physiologic stresses, dynamic shifts in cellular diversity and transcriptome profiles were observed for major hormone producing and the putative multi-hormone cells. These studies reveal unanticipated cellular complexity and plasticity in adult pituitary, and provide a rich resource for further validating and expanding our molecular understanding of pituitary gene expression programs and hormone production.


Assuntos
Plasticidade Celular/genética , Hipófise/citologia , Hipófise/metabolismo , RNA Mensageiro/genética , RNA-Seq , Caracteres Sexuais , Análise de Célula Única , Transcriptoma , Animais , Feminino , Homeostase/genética , Masculino , Camundongos , Camundongos Transgênicos
4.
Endocrinology ; 161(5)2020 05 01.
Artigo em Inglês | MEDLINE | ID: mdl-32188976

RESUMO

Differentiation of the hormone-producing cells of the pituitary represents an informative model of cell fate determination. The generation and maintenance of 2 pituitary lineages, the growth hormone (GH)- producing somatotropes and the prolactin (PRL)- producing lactotropes, are dependent on the pituitary-specific transcription factor, POU1F1. While POU1F1 is expressed in both cell types, and plays a role in activation of both the Gh and Prl genes, expression of Gh and Prl is restricted to somatotropes and lactotropes, respectively. These observations imply the existence of additional factors that contribute to the somatotrope and lactotrope identities and their hormone expressions. Prior transcriptome analysis of primary somatotropes and lactotropes isolated from the mouse pituitary identified enrichment of a transcription factor, Nr4a2, in the lactotropes. Nr4a2 was shown in a cell culture model to bind the Prl promoter at a position adjacent to Pou1f1 and to synergize with Pou1f1 in driving Prl transcription. Here we demonstrate in vivo the role of Nr4a2 as an enhancer of Prl expression by conditional gene inactivation of the Nr4a2 gene in mouse lactotropes. We demonstrate that nuclear orphan receptor transcription factor (NR4A2) binding at the Prl promoter is dependent on actions of POU1F1; while POU1F1 is essential to loading polymerase (Pol) II on the Prl promoter, Nr4a2 plays a role in enhancing Pol II release into the Prl gene body. These studies establish an in vivo role of Nr4a2 in enhancing Prl expression in mouse lactotropes, explore its mechanism of action, and establish a system for further study of the lactotrope lineage in the pituitary.


Assuntos
Regulação da Expressão Gênica , Lactotrofos/metabolismo , Membro 2 do Grupo A da Subfamília 4 de Receptores Nucleares/genética , Hipófise/metabolismo , Prolactina/genética , Animais , Células Cultivadas , Feminino , Lactotrofos/citologia , Camundongos Endogâmicos , Camundongos Knockout , Camundongos Transgênicos , Microscopia de Fluorescência , Membro 2 do Grupo A da Subfamília 4 de Receptores Nucleares/metabolismo , Hipófise/citologia , Prolactina/metabolismo , Regiões Promotoras Genéticas/genética , Ligação Proteica , Fator de Transcrição Pit-1/genética , Fator de Transcrição Pit-1/metabolismo
5.
Endocrinology ; 159(12): 3965-3980, 2018 12 01.
Artigo em Inglês | MEDLINE | ID: mdl-30247555

RESUMO

The differentiation of the hormone-producing cell lineages of the anterior pituitary represents an informative model of mammalian cell fate determination. The generation and maintenance of two of these lineages, the GH-producing somatotropes and prolactin (PRL)-producing lactotropes, are dependent on the pituitary-specific transcription factor POU1F1. Whereas POU1F1 is expressed in both cell types, and plays a direct role in the activation of both the Gh and Prl genes, GH expression is restricted to somatotropes and PRL expression is restricted to lactotropes. These observations imply the existence of additional, cell type-enriched factors that contribute to the somatotrope and lactotrope cell identities. In this study, we use transgenic mouse models to facilitate sorting of somatotrope and lactotrope populations based on the expression of fluorescent markers expressed under Gh and Prl gene transcriptional controls. The transcriptomic analyses reveal a concordance of gene expression profiles in the two populations. The limited number of divergent mRNAs between the two populations includes a set of transcription factors that may have roles in pituitary lineage divergence and/or in regulating expression of cell type-specific genes after differentiation. Four of these factors were validated for lineage enrichment at the level of protein expression, two somatotrope enriched and two lactotrope enriched. Three of these four factors were shown to have corresponding activities in appropriate enhancement or repression of landmark genes in a cell culture model system. These studies identify novel regulators of the somatotropes and lactotropes, and they establish a useful database for further study of these lineages in the anterior pituitary.


Assuntos
Diferenciação Celular/genética , Linhagem da Célula/genética , Lactotrofos/metabolismo , Somatotrofos/metabolismo , Transcriptoma , Animais , Células Cultivadas , Feminino , Perfilação da Expressão Gênica , Genes de Troca , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Hipófise/citologia , Hipófise/metabolismo
6.
Genes Dev ; 29(3): 298-307, 2015 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-25644604

RESUMO

PR (PRD1-BF1-RIZ1 homologous) domain-containing 16 (PRDM16) drives a brown fat differentiation program, but the mechanisms by which PRDM16 activates brown fat-selective genes have been unclear. Through chromatin immunoprecipitation (ChIP) followed by deep sequencing (ChIP-seq) analyses in brown adipose tissue (BAT), we reveal that PRDM16 binding is highly enriched at a broad set of brown fat-selective genes. Importantly, we found that PRDM16 physically binds to MED1, a component of the Mediator complex, and recruits it to superenhancers at brown fat-selective genes. PRDM16 deficiency in BAT reduces MED1 binding at PRDM16 target sites and causes a fundamental change in chromatin architecture at key brown fat-selective genes. Together, these data indicate that PRDM16 controls chromatin architecture and superenhancer activity in BAT.


Assuntos
Tecido Adiposo Marrom/metabolismo , Cromatina/metabolismo , Proteínas de Ligação a DNA/metabolismo , Subunidade 1 do Complexo Mediador/metabolismo , Fatores de Transcrição/metabolismo , Transcrição Gênica/fisiologia , Animais , Cromatina/química , Cromatina/genética , Elementos Facilitadores Genéticos , Camundongos
7.
Mol Cell Biol ; 35(9): 1523-32, 2015 May.
Artigo em Inglês | MEDLINE | ID: mdl-25691665

RESUMO

The transcription factor Pit-1 (POU1-F1) plays a dominant role in cell lineage expansion and differentiation in the anterior pituitary. Prior studies of the mouse Pit-1 (mPit-1) gene revealed that this master regulatory locus is activated at embryonic day 13.5 (E13.5) by an early enhancer (EE), whereas its subsequent expression throughout adult life is maintained by a more distal definitive enhancer (DE). Here, we demonstrate that the sequential actions of these two enhancers are linked to corresponding shifts in their proximities to the Pit-1 promoter. We further demonstrate that the looping of the definitive enhancer to the mPit-1 promoter is critically dependent on a self-sustaining autoregulatory mechanism mediated by the Pit-1 protein. These Pit-1-dependent actions are accompanied by localized recruitment of CBP and enrichment for H3K27 acetylation within the Pit-1 locus. These data support a model in which the sequential actions of two developmentally activated enhancers are linked to a corresponding shift in higher-order chromatin structures. This shift establishes an autoregulatory circuit that maintains durable expression of Pit-1 throughout adult life.


Assuntos
Cromatina/genética , Regiões Promotoras Genéticas , Fator de Transcrição Pit-1/genética , Acetilação , Animais , Linhagem Celular , Cromatina/química , Cromatina/metabolismo , Elementos Facilitadores Genéticos , Histonas/metabolismo , Camundongos , Fator de Transcrição Pit-1/metabolismo , Ativação Transcricional
8.
Proc Natl Acad Sci U S A ; 111(46): E4946-53, 2014 Nov 18.
Artigo em Inglês | MEDLINE | ID: mdl-25369933

RESUMO

Notch is needed for T-cell development and is a common oncogenic driver in T-cell acute lymphoblastic leukemia. The protooncogene c-Myc (Myc) is a critical target of Notch in normal and malignant pre-T cells, but how Notch regulates Myc is unknown. Here, we identify a distal enhancer located >1 Mb 3' of human and murine Myc that binds Notch transcription complexes and physically interacts with the Myc proximal promoter. The Notch1 binding element in this region activates reporter genes in a Notch-dependent, cell-context-specific fashion that requires a conserved Notch complex binding site. Acute changes in Notch activation produce rapid changes in H3K27 acetylation across the entire enhancer (a region spanning >600 kb) that correlate with Myc expression. This broad Notch-influenced region comprises an enhancer region containing multiple domains, recognizable as discrete H3K27 acetylation peaks. Leukemia cells selected for resistance to Notch inhibitors express Myc despite epigenetic silencing of enhancer domains near the Notch transcription complex binding sites. Notch-independent expression of Myc in resistant cells is highly sensitive to inhibitors of bromodomain containing 4 (Brd4), a change in drug sensitivity that is accompanied by preferential association of the Myc promoter with more 3' enhancer domains that are strongly dependent on Brd4 for function. These findings indicate that altered long-range enhancer activity can mediate resistance to targeted therapies and provide a mechanistic rationale for combined targeting of Notch and Brd4 in leukemia.


Assuntos
Elementos Facilitadores Genéticos/genética , Regulação Leucêmica da Expressão Gênica/genética , Genes myc , Proteínas de Neoplasias/genética , Leucemia-Linfoma Linfoblástico de Células T Precursoras/genética , Receptor Notch1/metabolismo , Animais , Sequência de Bases , Proteínas de Ciclo Celular , Linhagem Celular Tumoral , Imunoprecipitação da Cromatina , Genes Reporter , Estudo de Associação Genômica Ampla , Histonas/metabolismo , Humanos , Proteína de Ligação a Sequências Sinal de Recombinação J de Imunoglobina/metabolismo , Modelos Moleculares , Dados de Sequência Molecular , Proteínas de Neoplasias/metabolismo , Proteínas Nucleares/antagonistas & inibidores , Leucemia-Linfoma Linfoblástico de Células T Precursoras/metabolismo , Regiões Promotoras Genéticas/genética , Conformação Proteica , Receptor Notch1/antagonistas & inibidores , Alinhamento de Sequência , Homologia de Sequência do Ácido Nucleico , Fatores de Transcrição/antagonistas & inibidores , Transcrição Gênica
9.
Mol Cell Biol ; 33(9): 1723-34, 2013 May.
Artigo em Inglês | MEDLINE | ID: mdl-23428872

RESUMO

For many mammalian genes, initiation of transcription during embryonic development must be subsequently sustained over extensive periods of adult life. It remains unclear whether maintenance of gene expression reflects the same set of pathways as are involved in initial gene activation. The human pituitary growth hormone (hGH-N) locus is activated in the differentiating somatotrope midway through embryogenesis by a multicomponent locus control region (LCR). DNase I-hypersensitive site I (HSI) of the LCR is essential to full developmental activation of the hGH-N locus. Here we demonstrate that conditional deletion of HSI from the active hGH locus in the adult pituitary effectively silences hGH-N expression. Analyses of chromatin structure and locus positioning demonstrate that a specific subset of the HSI functions active in the embryo retain their HSI dependence in the adult pituitary. These functions sustain engagement of the hGH locus with polymerase II (Pol II) factories, histone acetylation at the hGH-N promoter, and looping of the LCR to its target promoter. These data reveal that HSI is essential to both the maintenance and the initiation phases of gene expression. These observations contribute to our mechanistic understanding of how stable patterns of mammalian gene expression are established in a terminally differentiated cell.


Assuntos
Cromatina/metabolismo , Inativação Gênica , Hormônio do Crescimento/genética , Região de Controle de Locus Gênico , Ativação Transcricional , Acetilação , Animais , Sítios de Ligação , Cromatina/química , Cromatina/genética , DNA Polimerase II/metabolismo , Desoxirribonuclease I/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Hormônio do Crescimento/metabolismo , Humanos , Camundongos , Camundongos Transgênicos , Hipófise/metabolismo , Regiões Promotoras Genéticas , Deleção de Sequência , Fator de Transcrição Pit-1/genética , Fator de Transcrição Pit-1/metabolismo , Transgenes
10.
J Biol Chem ; 287(30): 25454-65, 2012 Jul 20.
Artigo em Inglês | MEDLINE | ID: mdl-22669946

RESUMO

Locus control regions (LCRs) comprise sets of DNA elements capable of establishing autonomous chromatin domains that support robust and physiologically appropriate expression of target genes, often working over extensive distances. Human growth hormone (hGH-N) expression in the pituitary is under the regulation of a well characterized LCR containing four DNase I hypersensitive sites (HSs). The two pituitary-specific HS, HSI and HSII, are located 14.5 and 15.5 kb 5' to the hGH-N promoter. HSI is essential for activation of hGH-N during pituitary development and for sustaining robust activity in the adult. To determine whether the closely linked HSII has a role in hGH-N expression, it was deleted from a previously validated hGH/P1 transgene. Analysis of three independent hGH/P1(ΔHSII) transgenic mouse lines revealed that this deletion had no adverse effect on the formation of HSI, yet resulted in a substantial loss (70%) in hGH-N mRNA expression. This loss of expression was accompanied by a corresponding reduction in recruitment of the pituitary-specific transcription factor Pit-1 to the hGH-N promoter and a selective decrease in promoter occupancy of the elongation-linked isoform of RNA polymerase II. Sufficiency of HSI and HSII in LCR activity was explored by establishing two additional sets of mouse transgenic lines in which DNA segments containing these HS were positioned within the λ phage genome. In this "neutral" DNA context, HSII was required for the recruitment of HAT activity. These data establish HSII as a nonredundant component of the hGH LCR essential for establishment of robust levels of hGH-N gene expression.


Assuntos
DNA Intergênico/metabolismo , Regulação da Expressão Gênica/fisiologia , Loci Gênicos/fisiologia , Hormônio do Crescimento Humano/biossíntese , Região de Controle de Locus Gênico/fisiologia , Hipófise/metabolismo , Animais , Sequência de Bases , DNA Intergênico/genética , Desoxirribonuclease I/química , Hormônio do Crescimento Humano/genética , Humanos , Camundongos , Camundongos Transgênicos , Hipófise/crescimento & desenvolvimento , Deleção de Sequência
11.
Mol Endocrinol ; 25(5): 877-84, 2011 May.
Artigo em Inglês | MEDLINE | ID: mdl-21415161

RESUMO

Expression of mammalian GH is normally restricted to somatotropes and somatolactotropes (somatotrope lineages) in the anterior pituitary. The basis for this restriction remains incompletely understood. Recent studies indicate that deoxyribonuclease I hypersensitive site I (HSI) of the hGH locus control region, located at -14.5 kb relative to the hGH-N promoter, acts as a potent long-range enhancer of hGH-N transcription. Here we report that HSI is also critical to somatotrope-restriction of hGH-N expression. Loss of HSI activity, either by direct inactivation of HSI or by interference with HSI-dependent downstream events, results in a relaxation of hGH-N cell-type specification with expansion of hGH-N expression to the full spectrum of Pit-1 positive pituitary cell types. These findings expand the defined roles for HSI of the hGH locus control region to include somatotrope lineage restriction as well as transcriptional enhancement of hGH-N gene expression.


Assuntos
Regulação da Expressão Gênica , Hormônio do Crescimento Humano/genética , Região de Controle de Locus Gênico , Proteínas Recombinantes/genética , Somatotrofos/metabolismo , Animais , Desoxirribonucleases de Sítio Específico do Tipo I/genética , Desoxirribonucleases de Sítio Específico do Tipo I/metabolismo , Técnica Indireta de Fluorescência para Anticorpo , Hormônio do Crescimento Humano/metabolismo , Masculino , Camundongos , Camundongos Transgênicos , Prolactina/metabolismo , Regiões Promotoras Genéticas , Proteínas Recombinantes/metabolismo , Tireotropina Subunidade beta/metabolismo , Fator de Transcrição Pit-1/metabolismo
12.
Mol Endocrinol ; 24(11): 2232-40, 2010 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-20829390

RESUMO

We report the establishment of three distinct pituitary-derived murine cell lines generated by targeted T-antigen-induced transformation. The Pit1/0 line expresses pituitary-specific transcription factor-1 (Pit-1) but lacks expression of GH, prolactin (Prl), or TSH, and the Pit1/Prl line is selectively positive for Pit-1 and Prl. The third line, Pit1/Triple, expresses Pit-1 and all three of the Pit-1-dependent hormones: GH, Prl, and TSHß/glycoprotein hormone α-subunit. The three corresponding transformation events appear to have captured pituitary cells representing: 1) an initial step in the Pit-1(+) lineage, 2) a cell line that corresponds to the differentiated lactotrope, and 3) a novel tri-hormone intermediate that may represent a pivotal step in Pit-1(+) cell lineage differentiation. The documented dependence of the tri-hormone expression in the Pit-1/Triple line on Pit-1 activity supports its potential role in the pathway of pituitary cell differentiation. The presence of a 123-kb human transgene encompassing the hGH locus (hGH/bacterial artificial chromosome) in two of these lines, Pit1/0 and Pit1/Prl, further expands their potential utility to the analysis of gene activation within the hGH gene cluster.


Assuntos
Antígenos Virais de Tumores/metabolismo , Linhagem Celular Transformada/citologia , Linhagem Celular Transformada/patologia , Linhagem da Célula , Transformação Celular Neoplásica/patologia , Hipófise/patologia , Fator de Transcrição Pit-1/metabolismo , Animais , Imunofluorescência , Regulação Neoplásica da Expressão Gênica , Camundongos , Camundongos Transgênicos , Modelos Biológicos , Hipófise/metabolismo , Neoplasias Hipofisárias/genética , Neoplasias Hipofisárias/patologia , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Fator de Transcrição Pit-1/genética , Transgenes/genética
13.
EMBO Rep ; 9(9): 891-8, 2008 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-18636089

RESUMO

Nonlinear chromatin configurations can juxtapose widely separated elements within a genomic locus; however, it remains unclear how these structures are established and contribute to transcriptional control. A 5'-remote locus control region (LCR) regulates the human growth hormone (hGH-N) gene. HSI, a pituitary-specific component of the hGH LCR, establishes a domain of polymerase II (PolII) transcription 5' to hGH-N. Repression of this transcriptional domain by HSI deletion or PolII blockade decreases hGH-N expression. Here, we show that hGH-N activation is accompanied by positioning of the hGH-N promoter to this LCR transcriptional domain. Selectively blocking LCR transcription inhibits the formation of this active 'looped' conformation. Thus, HSI is crucial for establishing a domain of noncoding PolII transcription, and this domain is intimately linked with chromatin organization of the active hGH-N locus. This integration of LCR transcription with chromatin reconfiguration constitutes a robust pathway for long-range gene activation.


Assuntos
Regulação da Expressão Gênica , Região de Controle de Locus Gênico/genética , Regiões Promotoras Genéticas/genética , Transcrição Gênica/genética , Animais , Cromatina/genética , Cromatina/metabolismo , Imunoprecipitação da Cromatina , Hormônio do Crescimento Humano/genética , Humanos , Camundongos , Camundongos Transgênicos , Reação em Cadeia da Polimerase/métodos
14.
Mol Cell ; 23(3): 365-75, 2006 Aug 04.
Artigo em Inglês | MEDLINE | ID: mdl-16885026

RESUMO

Activation of eukaryotic genes often relies on remote chromatin determinants. How these determinants function remains poorly understood. The hGH gene is activated by a 5'-remote locus control region (LCR). Pituitary-specific DNase I hypersensitive site I (HSI), the dominant hGH LCR element, is separated from the hGH-N promoter by a 14.5 kb span that encompasses the B-lymphocyte-specific CD79b gene. Here, we describe a domain of noncoding Pol II transcription in pituitary somatotropes that includes the hGH LCR and adjacent CD79b locus. This entire "LCR domain of transcription" is HSI [corrected] dependent and terminates 3' to CD79b, leaving a gap in transcription between this domain and the target hGH-N promoter. Insertion of a Pol II terminator within the LCR blocks CD79b transcription and represses hGH-N expression. These data document an essential role for LCR transcription in long-range control, link "bystander"CD79b transcription to this process, and support a unique model for locus activation.


Assuntos
Regulação da Expressão Gênica , Hormônio do Crescimento Humano/genética , Região de Controle de Locus Gênico/fisiologia , Transcrição Gênica/genética , Acetilação , Animais , Antígenos CD79/genética , Imunoprecipitação da Cromatina , Deleção de Genes , Biblioteca Gênica , Hormônio Liberador de Hormônio do Crescimento/genética , Histonas/metabolismo , Humanos , Fígado/metabolismo , Região de Controle de Locus Gênico/genética , Camundongos , Camundongos Transgênicos , Modelos Genéticos , Hipófise/metabolismo , RNA Polimerase II/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Regiões Terminadoras Genéticas/genética , Ativação Transcricional
15.
Mol Cell Biol ; 26(17): 6535-46, 2006 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-16914737

RESUMO

Activation of the human growth hormone (hGH-N) gene in pituitary somatotropes is mediated by a locus control region (LCR). This LCR is composed of DNase I-hypersensitive sites (HS) located -14.5 kb to -32 kb relative to the hGH-N promoter. HSI, at -14.5 kb, is the dominant determinant of hGH-N expression and is essential for establishment of a 32-kb domain of histone acetylation that encompasses the active hGH locus. This activity is conferred by three binding sites for the POU domain transcription factor Pit-1. These Pit-1 elements are sufficient to activate hGH-N expression in the mouse pituitary. In contrast, Pit-1 sites at the hGH-N promoter are consistently unable to mediate similar activity. In the present study, we demonstrate that the functional difference between the promoter-proximal and the HSI Pit-1 binding sites can be attributed in part to a single base difference. This base affects the conformation of the Pit-1/DNA complex, and reciprocal exchange of the divergent bases between the two sets of Pit-1 elements results in a partial reversal of their transgenic activities. These data support a model in which the Pit-1 binding sites in the hGH LCR allosterically program the bound Pit-1 complex for chromatin activating functions.


Assuntos
Pareamento de Bases , Hormônio do Crescimento Humano/genética , Região de Controle de Locus Gênico/genética , Regiões Promotoras Genéticas/genética , Fator de Transcrição Pit-1/química , Fator de Transcrição Pit-1/metabolismo , Adenina/metabolismo , Animais , Sequência de Bases , Sítios de Ligação , Proteína de Ligação a CREB/metabolismo , Sequência Consenso/genética , Expressão Gênica , Camundongos , Camundongos Transgênicos , Dados de Sequência Molecular , Mutação/genética , Peptídeo Hidrolases/metabolismo , Hipófise/citologia , Ligação Proteica , Conformação Proteica , Ratos , Timina/metabolismo , Fator de Transcrição Pit-1/genética
16.
Trends Endocrinol Metab ; 15(1): 40-5, 2004.
Artigo em Inglês | MEDLINE | ID: mdl-14693425

RESUMO

The cluster of genes encoding the human growth hormone (GH) contains an array of five highly related genes. From 5' to 3' these are: GHN, CSL (encoding chorionic somatomammotropin-like gene), CSA, GHV (encoding GH-variant gene) and CSB. These five genes are expressed in mutually exclusive tissue distributions, GHN in pituitary somatotropes and the remaining four genes in placental villous syncytiotrophoblasts. The onset of GH expression during development is dependent upon epigenetic modifications at the GH locus under the control of its distal locus control region (LCR). A clear understanding of these normal epigenetic controls on the expression of GH could lead to new insights into the development and treatment of isolated GH deficiency in children. This review focuses on the role of the LCR in histone hyperacetylation at the GH locus and subsequent effects on the tissue-specific activation of these genes.


Assuntos
Cromatina/genética , Cromatina/metabolismo , Hormônio do Crescimento Humano/genética , Hormônio do Crescimento Humano/metabolismo , Região de Controle de Locus Gênico , Hipófise/metabolismo , Placenta/metabolismo , Acetiltransferases/metabolismo , Regulação da Expressão Gênica , Histona Acetiltransferases , Histonas/genética , Histonas/metabolismo , Humanos , Especificidade de Órgãos
17.
J Mol Biol ; 329(2): 239-51, 2003 May 30.
Artigo em Inglês | MEDLINE | ID: mdl-12758073

RESUMO

A large number of group I introns encode a family of homologous proteins that either promote intron splicing (maturases) or are site-specific DNA endonucleases that function in intron mobility (a process called "homing"). Genetic studies have shown that some of these proteins have both activities, yet how a single protein carries out both functions remains obscure. The similarity between respective DNA-binding sites and the RNA structure near the 5' and 3' splice sites has fueled speculation that such proteins may use analogous interactions to perform both functions. The Aspergillus nidulans mitochondrial COB group I intron encodes a bi-functional protein, I-AniI, that has both RNA maturase and site-specific DNA endonuclease activities in vitro. Here, we show that I-AniI shows distinctive features of the endonuclease family to which it belongs, including highly specific, tight binding and sequential DNA strand cleavage. Competition experiments demonstrate that I-AniI binds the COB intron RNA even in saturating concentrations of its DNA target site substrate, suggesting that the protein has a separate binding site for RNA. In addition, we provide evidence that two different DNA-binding site mutants of I-AniI have little effect on the protein's RNA maturation activity. Since RNA splicing is likely a secondary adaptation of the protein, these observations support a model in which homing endonucleases may have developed maturase function by utilizing a hitherto "non-functional" protein surface.


Assuntos
Aspergillus nidulans/enzimologia , DNA Fúngico/metabolismo , Endonucleases/metabolismo , Íntrons/genética , Splicing de RNA , RNA Fúngico/metabolismo , DNA Polimerase Dirigida por RNA/metabolismo , Substituição de Aminoácidos , Sítios de Ligação/fisiologia , Ligação Competitiva/fisiologia , Primers do DNA/química , Ativação Enzimática/fisiologia , Fragmentos de Peptídeos/química , Especificidade por Substrato
18.
Mol Cell ; 9(2): 291-302, 2002 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-11864603

RESUMO

Gene activation in higher eukaryotes is often under the control of regulatory elements quite distant from their target promoters. It is unclear how such long-range control is mediated. Here we show that a single determinant of the human growth hormone locus control region (hGH LCR) located 14.5 kb 5prime prime or minute to the hGH-N promoter has a critical, specific, and nonredundant role in facilitating promoter trans factor binding and activating hGH-N transcription. Significantly, this same determinant plays an essential role in establishing a 32 kb acetylated domain that encompasses the entire hGH LCR and the contiguous hGH-N promoter. These data support a model for long-range gene activation via LCR-mediated targeting and extensive spreading of core histone acetylation.


Assuntos
Proteínas de Ligação a DNA/metabolismo , Regulação da Expressão Gênica/genética , Histonas/metabolismo , Hormônio do Crescimento Humano/genética , Região de Controle de Locus Gênico/fisiologia , Processamento de Proteína Pós-Traducional , Fatores de Transcrição/metabolismo , Acetilação , Animais , Sítios de Ligação , Cromatina/genética , Cromatina/metabolismo , Feminino , Humanos , Camundongos , Camundongos Transgênicos , Especificidade de Órgãos , Adeno-Hipófise/metabolismo , Gravidez , Regiões Promotoras Genéticas , Deleção de Sequência , Fator de Transcrição Pit-1 , Transcrição Gênica , Ativação Transcricional , Transgenes , Trofoblastos/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...