Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 9 de 9
Filtrar
1.
Nat Commun ; 12(1): 5887, 2021 10 07.
Artigo em Inglês | MEDLINE | ID: mdl-34620853

RESUMO

TRIP6, a member of the ZYXIN-family of LIM domain proteins, is a focal adhesion component. Trip6 deletion in the mouse, reported here, reveals a function in the brain: ependymal and choroid plexus epithelial cells are carrying, unexpectedly, fewer and shorter cilia, are poorly differentiated, and the mice develop hydrocephalus. TRIP6 carries numerous protein interaction domains and its functions require homodimerization. Indeed, TRIP6 disruption in vitro (in a choroid plexus epithelial cell line), via RNAi or inhibition of its homodimerization, confirms its function in ciliogenesis. Using super-resolution microscopy, we demonstrate TRIP6 localization at the pericentriolar material and along the ciliary axoneme. The requirement for homodimerization which doubles its interaction sites, its punctate localization along the axoneme, and its co-localization with other cilia components suggest a scaffold/co-transporter function for TRIP6 in cilia. Thus, this work uncovers an essential role of a LIM-domain protein assembly factor in mammalian ciliogenesis.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/genética , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Encéfalo/metabolismo , Proteínas com Domínio LIM/genética , Proteínas com Domínio LIM/metabolismo , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo , Animais , Encéfalo/patologia , Epêndima/patologia , Adesões Focais/metabolismo , Regulação da Expressão Gênica , Camundongos , Camundongos Knockout , Interferência de RNA , Transcriptoma
2.
Elife ; 102021 02 17.
Artigo em Inglês | MEDLINE | ID: mdl-33594975

RESUMO

Brain homeostasis is regulated by the viability and functionality of neurons. HAT (histone acetyltransferase) and HDAC (histone deacetylase) inhibitors have been applied to treat neurological deficits in humans; yet, the epigenetic regulation in neurodegeneration remains elusive. Mutations of HAT cofactor TRRAP (transformation/transcription domain-associated protein) cause human neuropathies, including psychosis, intellectual disability, autism, and epilepsy, with unknown mechanism. Here we show that Trrap deletion in Purkinje neurons results in neurodegeneration of old mice. Integrated transcriptomics, epigenomics, and proteomics reveal that TRRAP via SP1 conducts a conserved transcriptomic program. TRRAP is required for SP1 binding at the promoter proximity of target genes, especially microtubule dynamics. The ectopic expression of Stathmin3/4 ameliorates defects of TRRAP-deficient neurons, indicating that the microtubule dynamics is particularly vulnerable to the action of SP1 activity. This study unravels a network linking three well-known, but up-to-date unconnected, signaling pathways, namely TRRAP, HAT, and SP1 with microtubule dynamics, in neuroprotection.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/genética , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Fator de Transcrição Sp1/genética , Fator de Transcrição Sp1/metabolismo , Envelhecimento , Animais , Epigênese Genética , Deleção de Genes , Regulação da Expressão Gênica , Camundongos , Camundongos Mutantes , Microtúbulos/metabolismo , Células de Purkinje/patologia , Transdução de Sinais
3.
Genomics Proteomics Bioinformatics ; 15(1): 19-36, 2017 02.
Artigo em Inglês | MEDLINE | ID: mdl-28161493

RESUMO

Embryonic development is critical for the final functionality and maintenance of the adult brain. Brain development is tightly regulated by intracellular and extracellular signaling. Lysine acetylation and deacetylation are posttranslational modifications that are able to link extracellular signals to intracellular responses. A wealth of evidence indicates that lysine acetylation and deacetylation are critical for brain development and functionality. Indeed, mutations of the enzymes and cofactors responsible for these processes are often associated with neurodevelopmental and psychiatric disorders. Lysine acetylation and deacetylation are involved in all levels of brain development, starting from neuroprogenitor survival and proliferation, cell fate decisions, neuronal maturation, migration, and synaptogenesis, as well as differentiation and maturation of astrocytes and oligodendrocytes, to the establishment of neuronal circuits. Hence, fluctuations in the balance between lysine acetylation and deacetylation contribute to the final shape and performance of the brain. In this review, we summarize the current basic knowledge on the specific roles of lysine acetyltransferase (KAT) and lysine deacetylase (KDAC) complexes in brain development and the different neurodevelopmental disorders that are associated with dysfunctional lysine (de)acetylation machineries.


Assuntos
Encéfalo/metabolismo , Lisina/metabolismo , Acetilação , Encéfalo/crescimento & desenvolvimento , Carboxiliases/metabolismo , Montagem e Desmontagem da Cromatina , Humanos , Lisina Acetiltransferases/metabolismo , Células-Tronco Neurais/citologia , Células-Tronco Neurais/metabolismo , Transtornos do Neurodesenvolvimento/metabolismo , Transtornos do Neurodesenvolvimento/patologia , Neurogênese
4.
Cell Stem Cell ; 14(5): 632-43, 2014 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-24792116

RESUMO

Fate decisions in neural progenitor cells are orchestrated via multiple pathways, and the role of histone acetylation in these decisions has been ascribed to a general function promoting gene activation. Here, we show that the histone acetyltransferase (HAT) cofactor transformation/transcription domain-associated protein (Trrap) specifically regulates activation of cell-cycle genes, thereby integrating discrete cell-intrinsic programs of cell-cycle progression and epigenetic regulation of gene transcription in order to control neurogenesis. Deletion of Trrap impairs recruitment of HATs and transcriptional machinery specifically to E2F cell-cycle target genes, disrupting their transcription with consequent cell-cycle lengthening specifically within cortical apical neural progenitors (APs). Consistently, Trrap conditional mutants exhibit microcephaly because of premature differentiation of APs into intermediate basal progenitors and neurons, and overexpressing cell-cycle regulators in vivo can rescue these premature differentiation defects. These results demonstrate an essential and highly specific role for Trrap-mediated histone regulation in controlling cell-cycle progression and neurogenesis.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Proteínas de Ciclo Celular/metabolismo , Proteínas Nucleares/metabolismo , Proteínas Adaptadoras de Transdução de Sinal/genética , Animais , Ciclo Celular , Proteínas de Ciclo Celular/genética , Diferenciação Celular/genética , Diferenciação Celular/fisiologia , Células Cultivadas , Imunoprecipitação da Cromatina , Feminino , Histona Acetiltransferases/genética , Histona Acetiltransferases/metabolismo , Immunoblotting , Marcação In Situ das Extremidades Cortadas , Masculino , Camundongos , Camundongos Transgênicos , Modelos Teóricos , Proteínas Nucleares/genética
5.
DNA Repair (Amst) ; 12(8): 645-55, 2013 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-23683352

RESUMO

MCPH1 encodes BRCT-containing protein MCPH1/Microcephalin/BRIT1, mutations of which in humans cause autosomal recessive disorder primary microcephaly type 1 (MCPH1), characterized by a congenital reduction of brain size particularly in the cerebral cortex. We have shown previously that a deletion of Mcph1 in mice results in microcephaly because of a premature switch from symmetric to asymmetric division of the neuroprogenitors, which is regulated by MCPH1's function in the centrosome. Because MCPH1 has been implicated in ATM and ATR-mediated DNA damage response (DDR) and defective DDR is often associated with neurodevelopmental diseases, we wonder whether the DDR-related function of MCPH1 prevents microcephaly. Here, we show that a deletion of Mcph1 results in a specific reduction of the cerebral cortex at birth, which is persistent through life. Due to an effect on premature neurogenic production, Mcph1-deficient progenitors give rise to a high level of early-born neurons that form deep layers (IV-VI), while generate less late-born neurons that form a thinner outer layer (II-III) of the cortex. However, neuronal migration seems to be unaffected by Mcph1 deletion. Ionizing radiation (IR) induces a massive apoptosis in the Mcph1-null neocortex and also embryonic lethality. Finally, Mcph1 deletion compromises homologous recombination repair and increases genomic instability. Altogether, our data suggest that MCPH1 ensures proper neuroprogenitor expansion and differentiation not only through its function in the centrosome, but also in the DDR.


Assuntos
Proteínas Cromossômicas não Histona/genética , Dano ao DNA , Microcefalia/genética , Animais , Apoptose/efeitos da radiação , Proteínas de Ciclo Celular , Diferenciação Celular , Centrossomo/metabolismo , Centrossomo/patologia , Proteínas Cromossômicas não Histona/metabolismo , Proteínas do Citoesqueleto , Reparo do DNA , Modelos Animais de Doenças , Deleção de Genes , Técnicas de Inativação de Genes , Instabilidade Genômica , Camundongos , Microcefalia/embriologia , Microcefalia/patologia , Neocórtex/embriologia , Neocórtex/patologia , Neocórtex/efeitos da radiação , Neurônios/citologia , Neurônios/patologia , Radiação Ionizante , Recombinação Genética
6.
Biochem Pharmacol ; 84(12): 1581-91, 2012 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-23018034

RESUMO

Sp1 is a transcription factor regulating many genes through its DNA binding domain, containing three zinc fingers. We were interested in identifying target genes regulated by Sp1, particularly those involved in proliferation and cancer. Our approach was to treat HeLa cells with a siRNA directed against Sp1 mRNA to decrease the expression of Sp1 and, in turn, the genes activated by this transcription factor. Sp1-siRNA treatment led to a great number of differentially expressed genes as determined by whole genome cDNA microarray analysis. Underexpressed genes were selected since they represent putative genes activated by Sp1 and classified in six Gene Onthology categories, namely proliferation and cancer, mRNA processing, lipid metabolism, glucidic metabolism, transcription and translation. Putative Sp1 binding sites were found in the promoters of the selected genes using the Match™ software. After literature mining, 11 genes were selected for further validation. Underexpression by qRT-PCR was confirmed for the 11 genes plus Sp1 in HeLa cells after Sp1-siRNA treatment. EMSA and ChIP assays were performed to test for binding of Sp1 to the promoters of these genes. We observed binding of Sp1 to the promoters of RAB20, FGF21, IHPK2, ARHGAP18, NPM3, SRSF7, CALM3, PGD and Sp1 itself. Furthermore, the mRNA levels of RAB20, FGF21 and IHPK2 and luciferase activity for these three genes related to proliferation and cancer, were determined after overexpression of Sp1 in HeLa cells, to confirm their regulation by Sp1. Involvement of these three genes in proliferation was validated by gene silencing using polypurine reverse hoogsteen hairpins.


Assuntos
Proliferação de Células , Genômica , Neoplasias/patologia , Fator de Transcrição Sp1/genética , Sequência de Bases , Western Blotting , Imunoprecipitação da Cromatina , Primers do DNA , Células HeLa , Humanos , Análise de Sequência com Séries de Oligonucleotídeos , Regiões Promotoras Genéticas , Reação em Cadeia da Polimerase em Tempo Real , Fator de Transcrição Sp1/metabolismo
7.
Cell Cycle ; 7(18): 2856-67, 2008 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-18769160

RESUMO

Sp1 transcription factor regulates the expression of multiple genes, including the Sp1 gene itself. We analyzed the ability of different cell cycle regulatory proteins to interact with Sp1 and to affect Sp1 promoter activity. Using an antibody array, we observed that CDK4, SKP2, Rad51, BRCA2 and p21 could interact with Sp1 and we confirmed these interactions by co-immunoprecipitation. CDK4, SKP2, Rad51, BRCA2 and p21 also activated the Sp1 promoter. Among the known Sp1-interacting proteins, E2F-DP1, Cyclin D1, Stat3 and Rb activated the Sp1 promoter, whereas p53 and NF kappaB inhibited it. The proteins that regulated Sp1 gene expression were shown by positive chromatin immunoprecipitation to be bound to the Sp1 promoter. Moreover, SKP2, BRCA2, p21, E2F-DP1, Stat3, Rb, p53 and NF kappaB had similar effects on an artificial promoter containing only Sp1 binding sites. Transient transfections of CDK4, Rad51, E2F-DP1, p21 and Stat3 increased mRNA expression from the endogenous Sp1 gene in HeLa cells whereas overexpression of NF kappaB, and p53 decreased Sp1 mRNA levels. p21 expression from a stably integrated inducible promoter in HT1080 cells activated Sp1 expression at the promoter and mRNA levels, but at the same time it decreased Sp1 protein levels due to the activation of Sp1 degradation. The observed multiple effects of cell cycle regulators on Sp1 suggest that Sp1 may be a key mediator of cell cycle associated changes in gene expression.


Assuntos
Proteínas de Ciclo Celular/metabolismo , Fator de Transcrição Sp1/metabolismo , Sítios de Ligação , Imunoprecipitação da Cromatina , Inibidor de Quinase Dependente de Ciclina p21/metabolismo , Regulação Neoplásica da Expressão Gênica , Células HeLa , Humanos , Modelos Biológicos , Regiões Promotoras Genéticas/genética , Ligação Proteica , Fator de Transcrição Sp1/genética , Transcrição Gênica
8.
Biochim Biophys Acta ; 1779(5): 318-29, 2008 May.
Artigo em Inglês | MEDLINE | ID: mdl-18342022

RESUMO

We analyzed in detail the proximal promoter of transcription factor Sp3, which expands 281 bp from the translational start. This sequence contains putative binding sites for Sp1, NF-Y, NF-1, Myb, AP-1 and E2F transcription factors. In this work, we further explored the role of these boxes on the regulation of the Sp3 gene. Gel-shift and competition assays showed specific binding of NF-1, Myb, AP-1 and E2F. Furthermore, chromatin immunoprecipitation assays demonstrated that Sp1, Sp3, NF-Y, NF-1, c-Myb, B-Myb, c-Jun and E2F1 actually occupied the Sp3 promoter in HeLa cells. Transient transfections and luciferase assays revealed activation of the Sp3 proximal promoter upon overexpression of NF-1, c-Myb, B-Myb, c-Jun and c-Fos, and repression after overexpression of E2F/DP1. Point mutation of the binding sites for NF1, Myb, AP1 and E2F and cell incubation with specific siRNAs further confirmed the role of these transcription factors in the regulation of the Sp3 promoter. The regulation of the endogenous Sp3 gene was also observed at the mRNA level when the studied transcription factors were overexpressed or knocked down by siRNA incubation. These results help to explain the complex regulation of the Sp3 gene, which depends, at least in part, on the relative amount of Sp1, Sp3, NF-Y, NF-1, c-Myb, B-Myb, AP-1, and E2F proteins in the cell.


Assuntos
Regulação da Expressão Gênica , Regiões Promotoras Genéticas , Fator de Transcrição Sp3/genética , Fatores de Transcrição/metabolismo , Região 5'-Flanqueadora , Sequência de Bases , Proteínas de Ciclo Celular/metabolismo , Linhagem Celular Tumoral , Fatores de Transcrição E2F/metabolismo , Células HeLa , Humanos , Dados de Sequência Molecular , Neurofibromina 1/metabolismo , Proteínas Proto-Oncogênicas c-myb/metabolismo , Fatores de Transcrição Sp/metabolismo , Transativadores/metabolismo , Fator de Transcrição AP-1/metabolismo , Transcrição Gênica
9.
Biochim Biophys Acta ; 1730(2): 126-36, 2005 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-16024108

RESUMO

A fragment of 1079 bp from the 5'-flanking region of the human Sp3 gene was isolated and characterized. The Sp3 promoter is a GC-rich region that contains putative binding sites for Elk-1, c-Myb, NF-1, Ap1, Sp1, NF-Y, Ap2 and USF. Several transcriptional start sites located between 70 and 132 bp upstream of the translational start site were identified. The proximal promoter was contained in the first 281 bp 5' of the translational start, whereas the region including up to -225 relative to the translational start was referred as the minimal promoter. Transient transfections and luciferase assays revealed activation of the Sp3 proximal promoter upon overexpression of either Sp1 or Sp3, alone or in combination. Gel-shift and supershift assays demonstrated specific binding of Sp1 and Sp3 proteins to the GC box located in the proximal promoter of Sp3. Overexpression of NF-YA had a synergistic effect on Sp1 overexpression and an additive effect on Sp3 overexpression. Additionally, overexpression of NF-YA, Sp1 and Sp3 altogether had a synergistic effect on Sp3 promoter activity. Furthermore, binding of the NF-Y complex to the CCAAT box located in the proximal promoter of Sp3 was observed in gel-shift assays.


Assuntos
Região 5'-Flanqueadora/genética , Proteínas de Ligação a DNA/genética , Fatores de Transcrição/genética , Sequência de Aminoácidos , Sequência de Bases , Fator de Ligação a CCAAT/fisiologia , Clonagem Molecular , Perfilação da Expressão Gênica , Regulação da Expressão Gênica , Células HeLa , Humanos , Dados de Sequência Molecular , Regiões Promotoras Genéticas , Homologia de Sequência do Ácido Nucleico , Fator de Transcrição Sp1/fisiologia , Fator de Transcrição Sp3 , Transcrição Gênica
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA