Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 21
Filtrar
1.
Am J Hum Genet ; 100(3): 555-561, 2017 Mar 02.
Artigo em Inglês | MEDLINE | ID: mdl-28257693

RESUMO

In two independent consanguineous families each with two children affected by mild intellectual disability and microcephaly, we identified two homozygous missense variants (c.119T>A [p.Met40Lys] and c.92T>A [p.Leu31His]) in TATA-box-binding-protein-associated factor 13 (TAF13). Molecular modeling suggested a pathogenic effect of both variants through disruption of the interaction between TAF13 and TAF11. These two proteins form a histone-like heterodimer that is essential for their recruitment into the general RNA polymerase II transcription factor IID (TFIID) complex. Co-immunoprecipitation in HeLa cells transfected with plasmids encoding TAF11 and TAF13 revealed that both variants indeed impaired formation of the TAF13-TAF11 heterodimer, thus confirming the protein modeling analysis. To further understand the functional role of TAF13, we performed RNA sequencing of neuroblastoma cell lines upon TAF13 knockdown. The transcriptional profile showed significant deregulation of gene expression patterns with an emphasis on genes related to neuronal and skeletal functions and those containing E-box motives in their promoters. Here, we expand the spectrum of TAF-associated phenotypes and highlight the importance of TAF13 in neuronal functions.


Assuntos
Deficiência Intelectual/genética , Microcefalia/genética , Fatores Associados à Proteína de Ligação a TATA/genética , Fator de Transcrição TFIID/genética , Alelos , Feminino , Variação Genética , Humanos , Imunoprecipitação , Lactente , Masculino , Linhagem , Regiões Promotoras Genéticas , Conformação Proteica , Transcrição Gênica
2.
PLoS Genet ; 13(2): e1006600, 2017 02.
Artigo em Inglês | MEDLINE | ID: mdl-28178271

RESUMO

The TEAD family of transcription factors (TEAD1-4) bind the MCAT element in the regulatory elements of both growth promoting and myogenic differentiation genes. Defining TEAD transcription factor function in myogenesis has proved elusive due to overlapping expression of family members and their functional redundancy. We show that silencing of either Tead1, Tead2 or Tead4 did not effect primary myoblast (PM) differentiation, but that their simultaneous knockdown strongly impaired differentiation. In contrast, Tead1 or Tead4 silencing impaired C2C12 differentiation showing their different contributions in PMs and C2C12 cells. Chromatin immunoprecipitation identified enhancers associated with myogenic genes bound by combinations of Tead4, Myod1 or Myog. Tead4 regulated distinct gene sets in C2C12 cells and PMs involving both activation of the myogenic program and repression of growth and signaling pathways. ChIP-seq from mature mouse muscle fibres in vivo identified a set of highly transcribed muscle cell-identity genes and sites bound by Tead1 and Tead4. Although inactivation of Tead4 in mature muscle fibres caused no obvious phenotype under normal conditions, notexin-induced muscle regeneration was delayed in Tead4 mutants suggesting an important role in myogenic differentiation in vivo. By combining knockdown in cell models in vitro with Tead4 inactivation in muscle in vivo, we provide the first comprehensive description of the specific and redundant roles of Tead factors in myogenic differentiation.


Assuntos
Diferenciação Celular/genética , Proteínas de Ligação a DNA/genética , Proteínas Musculares/genética , Músculos/metabolismo , Mioblastos/metabolismo , Regeneração/genética , Fatores de Transcrição/genética , Animais , Linhagem Celular , Proteínas de Ligação a DNA/metabolismo , Elementos Facilitadores Genéticos/genética , Perfilação da Expressão Gênica/métodos , Immunoblotting , Camundongos Endogâmicos C57BL , Camundongos Knockout , Microscopia de Fluorescência , Desenvolvimento Muscular/genética , Proteínas Musculares/metabolismo , Músculos/citologia , Músculos/fisiologia , Mutação , Mioblastos/citologia , Ligação Proteica , Interferência de RNA , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transdução de Sinais/genética , Fatores de Transcrição de Domínio TEA , Fatores de Transcrição/metabolismo
3.
Proc Natl Acad Sci U S A ; 113(5): E568-76, 2016 Feb 02.
Artigo em Inglês | MEDLINE | ID: mdl-26787865

RESUMO

There is a paucity of new therapeutic targets to control allergic reactions and forestall the rising trend of allergic diseases. Although a variety of immune cells contribute to allergy, cytokine-secreting αß(+)CD4(+) T-helper 2 (TH2) cells orchestrate the type-2-driven immune response in a large proportion of atopic asthmatics. To identify previously unidentified putative targets in pathogenic TH2 cells, we performed in silico analyses of recently published transcriptional data from a wide variety of pathogenic TH cells [Okoye IS, et al. (2014) Proc Natl Acad Sci USA 111(30):E3081-E3090] and identified that transcription intermediary factor 1 regulator-alpha (Tif1α)/tripartite motif-containing 24 (Trim24) was predicted to be active in house dust mite (HDM)- and helminth-elicited Il4(gfp+)αß(+)CD4(+) TH2 cells but not in TH1, TH17, or Treg cells. Testing this prediction, we restricted Trim24 deficiency to T cells by using a mixed bone marrow chimera system and found that T-cell-intrinsic Trim24 is essential for HDM-mediated airway allergy and antihelminth immunity. Mechanistically, HDM-elicited Trim24(-/-) T cells have reduced expression of many TH2 cytokines and chemokines and were predicted to have compromised IL-1-regulated signaling. Following this prediction, we found that Trim24(-/-) T cells have reduced IL-1 receptor (IL-1R) expression, are refractory to IL-1ß-mediated activation in vitro and in vivo, and fail to respond to IL-1ß-exacerbated airway allergy. Collectively, these data identify a previously unappreciated Trim24-dependent requirement for IL-1R expression on TH2 cells and an important nonredundant role for T-cell-intrinsic Trim24 in TH2-mediated allergy and antihelminth immunity.


Assuntos
Hipersensibilidade/imunologia , Proteínas Nucleares/fisiologia , Receptores de Interleucina-1/metabolismo , Células Th2/imunologia , Fatores de Transcrição/fisiologia , Animais , Helmintos/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Proteínas Nucleares/genética , Células Th2/metabolismo , Fatores de Transcrição/genética
4.
PLoS Genet ; 11(10): e1005555, 2015 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-26440048

RESUMO

MIcrophthalmia-associated Transcription Factor (MITF) regulates melanocyte and melanoma physiology. We show that MITF associates the NURF chromatin-remodelling factor in melanoma cells. ShRNA-mediated silencing of the NURF subunit BPTF revealed its essential role in several melanoma cell lines and in untransformed melanocytes in vitro. Comparative RNA-seq shows that MITF and BPTF co-regulate overlapping gene expression programs in cell lines in vitro. Somatic and specific inactivation of Bptf in developing murine melanoblasts in vivo shows that Bptf regulates their proliferation, migration and morphology. Once born, Bptf-mutant mice display premature greying where the second post-natal coat is white. This second coat is normally pigmented by differentiated melanocytes derived from the adult melanocyte stem cell (MSC) population that is stimulated to proliferate and differentiate at anagen. An MSC population is established and maintained throughout the life of the Bptf-mutant mice, but these MSCs are abnormal and at anagen, give rise to reduced numbers of transient amplifying cells (TACs) that do not express melanocyte markers and fail to differentiate into mature melanin producing melanocytes. MSCs display a transcriptionally repressed chromatin state and Bptf is essential for reactivation of the melanocyte gene expression program at anagen, the subsequent normal proliferation of TACs and their differentiation into mature melanocytes.


Assuntos
Antígenos Nucleares/genética , Montagem e Desmontagem da Cromatina/genética , Melanoma/genética , Células-Tronco Mesenquimais , Fator de Transcrição Associado à Microftalmia/genética , Proteínas do Tecido Nervoso/genética , Fatores de Transcrição/genética , Animais , Ciclo Celular/genética , Diferenciação Celular/genética , Divisão Celular/genética , Regulação da Expressão Gênica no Desenvolvimento , Folículo Piloso , Melanócitos/metabolismo , Melanoma/patologia , Camundongos
5.
Cell Death Differ ; 30(3): 839-853, 2023 03.
Artigo em Inglês | MEDLINE | ID: mdl-36639541

RESUMO

Taf4 (TATA-box binding protein-associated factor 4) is a subunit of the general transcription factor TFIID, a component of the RNA polymerase II pre-initiation complex that interacts with tissue-specific transcription factors to regulate gene expression. Properly regulated gene expression is particularly important in the intestinal epithelium that is constantly renewed from stem cells. Tissue-specific inactivation of Taf4 in murine intestinal epithelium during embryogenesis compromised gut morphogenesis and the emergence of adult-type stem cells. In adults, Taf4 loss impacted the stem cell compartment and associated Paneth cells in the stem cell niche, epithelial turnover and differentiation of mature cells, thus exacerbating the response to inflammatory challenge. Taf4 inactivation ex vivo in enteroids prevented budding formation and maintenance and caused broad chromatin remodeling and a strong reduction in the numbers of stem and progenitor cells with a concomitant increase in an undifferentiated cell population that displayed high activity of the Ezh2 and Suz12 components of Polycomb Repressive Complex 2 (PRC2). Treatment of Taf4-mutant enteroids with a specific Ezh2 inhibitor restored buddings, cell proliferation and the stem/progenitor compartment. Taf4 loss also led to increased PRC2 activity in cells of adult crypts associated with modification of the immune/inflammatory microenvironment that potentiated Apc-driven tumorigenesis. Our results reveal a novel function of Taf4 in antagonizing PRC2-mediated repression of the stem cell gene expression program to assure normal development, homeostasis, and immune-microenvironment of the intestinal epithelium.


Assuntos
Proteínas de Drosophila , Células-Tronco , Camundongos , Animais , Diferenciação Celular/genética , Células-Tronco/metabolismo , Fator de Transcrição TFIID/genética , Mucosa Intestinal/metabolismo , Proteínas de Drosophila/metabolismo , Complexo Repressor Polycomb 2/metabolismo , Epigênese Genética
6.
Cell Rep ; 42(11): 113363, 2023 11 28.
Artigo em Inglês | MEDLINE | ID: mdl-37924516

RESUMO

Super-enhancers (SEs) are stretches of enhancers ensuring a high level of expression of key genes associated with cell function. The identification of cancer-specific SE-driven genes is a powerful means for the development of innovative therapeutic strategies. Here, we identify a MITF/SOX10/TFIIH-dependent SE promoting the expression of BAHCC1 in a broad panel of melanoma cells. BAHCC1 is highly expressed in metastatic melanoma and is required for tumor engraftment, growth, and dissemination. Integrative genomics analyses reveal that BAHCC1 is a transcriptional regulator controlling expression of E2F/KLF-dependent cell-cycle and DNA-repair genes. BAHCC1 associates with BRG1-containing remodeling complexes at the promoters of these genes. BAHCC1 silencing leads to decreased cell proliferation and delayed DNA repair. Consequently, BAHCC1 deficiency cooperates with PARP inhibition to induce melanoma cell death. Our study identifies BAHCC1 as an SE-driven gene expressed in melanoma and demonstrates how its inhibition can be exploited as a therapeutic target.


Assuntos
Melanoma , Humanos , Linhagem Celular Tumoral , Melanoma/patologia , Sequências Reguladoras de Ácido Nucleico , Instabilidade Genômica , Proliferação de Células/genética , Regulação Neoplásica da Expressão Gênica , Elementos Facilitadores Genéticos , Proteínas/metabolismo
7.
Cancer Res ; 82(24): 4555-4570, 2022 12 16.
Artigo em Inglês | MEDLINE | ID: mdl-36214632

RESUMO

Tumor heterogeneity is a key feature of melanomas that hinders development of effective treatments. Aiming to overcome this, we identified LINC00518 (LENOX; lincRNA-enhancer of oxidative phosphorylation) as a melanoma-specific lncRNA expressed in all known melanoma cell states and essential for melanoma survival in vitro and in vivo. Mechanistically, LENOX promoted association of the RAP2C GTPase with mitochondrial fission regulator DRP1, increasing DRP1 S637 phosphorylation, mitochondrial fusion, and oxidative phosphorylation. LENOX expression was upregulated following treatment with MAPK inhibitors, facilitating a metabolic switch from glycolysis to oxidative phosphorylation and conferring resistance to MAPK inhibition. Consequently, combined silencing of LENOX and RAP2C synergized with MAPK inhibitors to eradicate melanoma cells. Melanomas are thus addicted to the lncRNA LENOX, which acts to optimize mitochondrial function during melanoma development and progression. SIGNIFICANCE: The lncRNA LENOX is a novel regulator of melanoma metabolism, which can be targeted in conjunction with MAPK inhibitors to eradicate melanoma cells.


Assuntos
Melanoma , Inibidores de Proteínas Quinases , RNA Longo não Codificante , Proteínas ras , Humanos , Linhagem Celular Tumoral , Melanoma/tratamento farmacológico , Melanoma/genética , Melanoma/metabolismo , Dinâmica Mitocondrial , Fosforilação Oxidativa , Inibidores de Proteínas Quinases/farmacologia , Proteínas ras/metabolismo , RNA Longo não Codificante/genética , RNA Longo não Codificante/metabolismo , Resistencia a Medicamentos Antineoplásicos
8.
JCI Insight ; 7(21)2022 11 08.
Artigo em Inglês | MEDLINE | ID: mdl-36107619

RESUMO

Malignant melanoma is a major public health issue displaying frequent resistance to targeted therapy and immunotherapy. A major challenge lies in better understanding how melanoma cells evade immune elimination and how tumor growth and metastasis is facilitated by the tumor microenvironment. Here, we show that expression of the cytokine thymic stromal lymphopoietin (TSLP) by epidermal keratinocytes is induced by cutaneous melanoma in both mice and humans. Using genetically engineered models of melanoma and tumor cell grafting combined with TSLP-KO or overexpression, we defined a crosstalk between melanoma cells, keratinocytes, and immune cells in establishing a tumor-promoting microenvironment. Keratinocyte-derived TSLP is induced by signals derived from melanoma cells and subsequently acts via immune cells to promote melanoma progression and metastasis. Furthermore, we show that TSLP signals through TSLP receptor-expressing (TSLPR-expressing) DCs to play an unrecognized role in promoting GATA3+ Tregs expressing a gene signature including ST2, CCR8, ICOS, PD-1, CTLA-4, and OX40 and exhibiting a potent suppressive activity on CD8+ T cell proliferation and IFN-γ production. An analogous population of GATA3-expressing Tregs was also identified in human melanoma tumors. Our study provides insights into the role of TSLP in programming a protumoral immune microenvironment in cutaneous melanoma.


Assuntos
Melanoma , Neoplasias Cutâneas , Humanos , Camundongos , Animais , Microambiente Tumoral , Melanoma/genética , Melanoma/metabolismo , Neoplasias Cutâneas/genética , Neoplasias Cutâneas/metabolismo , Citocinas/metabolismo , Queratinócitos/metabolismo , Linfopoietina do Estroma do Timo , Melanoma Maligno Cutâneo
9.
Cell Death Dis ; 12(8): 790, 2021 08 12.
Artigo em Inglês | MEDLINE | ID: mdl-34385420

RESUMO

Regulation of gene expression involves a complex and dynamic dialogue between transcription factors, chromatin remodelling and modification complexes and the basal transcription machinery. To address the function of the Taf4 subunit of general transcription factor TFIID in the regulation of insulin signalling, it was inactivated in adult murine pancreatic beta cells. Taf4 inactivation impacted the expression of critical genes involved in beta-cell function leading to increased glycaemia, lowered plasma insulin levels and defective glucose-stimulated insulin secretion. One week after Taf4-loss, single-cell RNA-seq revealed cells with mixed beta cell, alpha and/or delta cell identities as well as a beta cell population trans-differentiating into alpha-like cells. Computational analysis of single-cell RNA-seq defines how known critical beta cell and alpha cell determinants may act in combination with additional transcription factors and the NuRF chromatin remodelling complex to promote beta cell trans-differentiation.


Assuntos
Transdiferenciação Celular/genética , Perfilação da Expressão Gênica , Células Secretoras de Insulina/citologia , Células Secretoras de Insulina/metabolismo , Subunidades Proteicas/metabolismo , Análise de Célula Única , Fator de Transcrição TFIID/metabolismo , Envelhecimento/metabolismo , Animais , Peso Corporal/efeitos dos fármacos , Transdiferenciação Celular/efeitos dos fármacos , Regulação da Expressão Gênica/efeitos dos fármacos , Células Secretoras de Glucagon/efeitos dos fármacos , Células Secretoras de Glucagon/metabolismo , Glucose/farmacologia , Insulina/metabolismo , Secreção de Insulina/efeitos dos fármacos , Células Secretoras de Insulina/efeitos dos fármacos , Camundongos , Mutação/genética , Fator de Transcrição TFIID/deficiência , Fatores de Transcrição/metabolismo
10.
J Biol Chem ; 284(39): 26286-96, 2009 Sep 25.
Artigo em Inglês | MEDLINE | ID: mdl-19635797

RESUMO

The major core promoter-binding factor in polymerase II transcription machinery is TFIID, a complex consisting of TBP, the TATA box-binding protein, and 13 to 14 TBP-associated factors (TAFs). Previously we found that the histone H2A-like TAF paralogs TAF4 and TAF4b possess DNA-binding activity. Whether TAF4/TAF4b DNA binding directs TFIID to a specific core promoter element or facilitates TFIID binding to established core promoter elements is not known. Here we analyzed the mode of TAF4b.TAF12 DNA binding and show that this complex binds DNA with high affinity. The DNA length required for optimal binding is approximately 70 bp. Although the complex displays a weak sequence preference, the nucleotide composition is less important than the length of the DNA for high affinity binding. Comparative expression profiling of wild-type and a DNA-binding mutant of TAF4 revealed common core promoter features in the down-regulated genes that include a TATA-box and an Initiator. Further examination of the PEL98 gene from this group showed diminished Initiator activity and TFIID occupancy in TAF4 DNA-binding mutant cells. These findings suggest that DNA binding by TAF4/4b-TAF12 facilitates the association of TFIID with the core promoter of a subset of genes.


Assuntos
DNA/metabolismo , Regiões Promotoras Genéticas/genética , Fatores Associados à Proteína de Ligação a TATA/metabolismo , Fator de Transcrição TFIID/metabolismo , Animais , Sequência de Bases , Sítios de Ligação/genética , Ligação Competitiva , Bovinos , Linhagem Celular , Imunoprecipitação da Cromatina , DNA/genética , Perfilação da Expressão Gênica , Humanos , Dados de Sequência Molecular , Mutação , Ligação Proteica , Multimerização Proteica , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Fatores Associados à Proteína de Ligação a TATA/química , Fatores Associados à Proteína de Ligação a TATA/genética , Fator de Transcrição TFIID/química , Fator de Transcrição TFIID/genética , Transfecção
11.
Cell Death Differ ; 27(1): 29-43, 2020 01.
Artigo em Inglês | MEDLINE | ID: mdl-31065107

RESUMO

Somatic oncogenic mutation of BRAF coupled with inactivation of PTEN constitute a frequent combination of genomic alterations driving the development of human melanoma. Mice genetically engineered to conditionally express oncogenic BrafV600E and inactivate Pten in melanocytes following tamoxifen treatment rapidly develop melanoma. While early-stage melanomas comprised melanin-pigmented Mitf and Dct-expressing cells, expression of these and other melanocyte identity genes was lost in later stage tumours that showed histological and molecular characteristics of de-differentiated neural crest type cells. Melanocyte identity genes displayed loss of active chromatin marks and RNA polymerase II and gain of heterochromatin marks, indicating epigenetic reprogramming during tumour progression. Nevertheless, late-stage tumour cells grown in culture re-expressed Mitf, and melanocyte markers and Mitf together with Sox10 coregulated a large number of genes essential for their growth. In this melanoma model, somatic inactivation that the catalytic Brg1 (Smarca4) subunit of the SWI/SNF complex and the scaffolding Bptf subunit of the NuRF complex delayed tumour formation and deregulated large and overlapping gene expression programs essential for normal tumour cell growth. Moreover, we show that Brg1 and Bptf coregulated many genes together with Mitf and Sox10. Together these transcription factors and chromatin remodelling complexes orchestrate essential gene expression programs in mouse melanoma cells.


Assuntos
Antígenos Nucleares/fisiologia , DNA Helicases/fisiologia , Regulação Neoplásica da Expressão Gênica , Melanoma Experimental/genética , Proteínas do Tecido Nervoso/fisiologia , Proteínas Nucleares/fisiologia , Proteínas Proto-Oncogênicas B-raf/genética , Neoplasias Cutâneas/genética , Fatores de Transcrição/fisiologia , Animais , Antígenos Nucleares/genética , DNA Helicases/genética , Progressão da Doença , Epigênese Genética , Melanoma Experimental/metabolismo , Melanoma Experimental/patologia , Camundongos , Fator de Transcrição Associado à Microftalmia/genética , Fator de Transcrição Associado à Microftalmia/metabolismo , Mutação , Proteínas do Tecido Nervoso/genética , Proteínas Nucleares/genética , PTEN Fosfo-Hidrolase/genética , Fatores de Transcrição SOXE/genética , Fatores de Transcrição SOXE/metabolismo , Neoplasias Cutâneas/metabolismo , Neoplasias Cutâneas/patologia , Fatores de Transcrição/genética , Células Tumorais Cultivadas
12.
Eur J Hum Genet ; 14(10): 1090-6, 2006 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-16773126

RESUMO

Monozygotic twin brothers with a subtelomeric 6q deletion presented with mental retardation, microcephaly, seizures, an enlarged cisterna magna, dimpling at elbows, a high arched palate and a thin upper lip. The same subtelomeric deletion was detected in the mother of the patients, presenting with a milder phenotype. We narrowed down the breakpoint to a region of approximately 100 kb and estimated the size of the terminal deletion to be 1.2 Mb. This region contains four known and seven putative genes. Comparison of the deletion with other reported patients showed TBP was the most plausible candidate gene for the mental retardation in this syndrome. We verified that the TBP gene expression was halved in our patients using real-time PCR. Cognitive and behavioural tests performed on previously described heterozygous tbp mice suggested that TBP is potentially involved in cognitive development.


Assuntos
Deleção Cromossômica , Cromossomos Humanos Par 6/genética , Deficiência Intelectual/genética , Proteína de Ligação a TATA-Box/genética , Anormalidades Múltiplas/genética , Adolescente , Animais , Ansiedade/genética , Doenças em Gêmeos/genética , Feminino , Humanos , Masculino , Transtornos da Memória/genética , Camundongos , Linhagem , Gêmeos Monozigóticos/genética
13.
Nat Commun ; 7: 11063, 2016 Mar 30.
Artigo em Inglês | MEDLINE | ID: mdl-27026076

RESUMO

TAF4 (TATA-binding protein-associated factor 4) and its paralogue TAF4b are components of the TFIID core module. We inactivated the murine Taf4a gene to address Taf4 function during embryogenesis. Here we show that Taf4a(-/-) embryos survive until E9.5 where primary germ layers and many embryonic structures are identified showing Taf4 is dispensable for their specification. In contrast, Taf4 is required for correct patterning of the trunk and anterior structures, ventral morphogenesis and proper heart positioning. Overlapping expression of Taf4a and Taf4b during embryogenesis suggests their redundancy at early stages. In agreement with this, Taf4a(-/-) embryonic stem cells (ESCs) are viable and comprise Taf4b-containing TFIID. Nevertheless, Taf4a(-/-) ESCs do not complete differentiation into glutamatergic neurons and cardiomyocytes in vitro due to impaired preinitiation complex formation at the promoters of critical differentiation genes. We define an essential role of a core TFIID TAF in differentiation events during mammalian embryogenesis.


Assuntos
Diferenciação Celular , Desenvolvimento Embrionário , Células-Tronco Embrionárias Murinas/metabolismo , Subunidades Proteicas/metabolismo , Fator de Transcrição TFIID/metabolismo , Animais , Biomarcadores/metabolismo , Padronização Corporal/efeitos dos fármacos , Padronização Corporal/genética , Anormalidades Cardiovasculares/embriologia , Anormalidades Cardiovasculares/genética , Anormalidades Cardiovasculares/patologia , Diferenciação Celular/efeitos dos fármacos , Diferenciação Celular/genética , Sobrevivência Celular/efeitos dos fármacos , Perda do Embrião/genética , Perda do Embrião/patologia , Desenvolvimento Embrionário/efeitos dos fármacos , Desenvolvimento Embrionário/genética , Feminino , Regulação da Expressão Gênica no Desenvolvimento/efeitos dos fármacos , Células Germinativas/efeitos dos fármacos , Células Germinativas/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Células-Tronco Embrionárias Murinas/efeitos dos fármacos , Mutação , Contração Miocárdica/efeitos dos fármacos , Miócitos Cardíacos/efeitos dos fármacos , Miócitos Cardíacos/patologia , Crista Neural/efeitos dos fármacos , Crista Neural/patologia , Neurogênese/efeitos dos fármacos , Neurogênese/genética , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Fenótipo , Gravidez , Subunidades Proteicas/genética , Fator de Transcrição TFIID/deficiência , Fator de Transcrição TFIID/genética , Tretinoína/farmacologia
14.
Elife ; 42015 Mar 24.
Artigo em Inglês | MEDLINE | ID: mdl-25803486

RESUMO

Microphthalmia-associated transcription factor (MITF) is the master regulator of the melanocyte lineage. To understand how MITF regulates transcription, we used tandem affinity purification and mass spectrometry to define a comprehensive MITF interactome identifying novel cofactors involved in transcription, DNA replication and repair, and chromatin organisation. We show that MITF interacts with a PBAF chromatin remodelling complex comprising BRG1 and CHD7. BRG1 is essential for melanoma cell proliferation in vitro and for normal melanocyte development in vivo. MITF and SOX10 actively recruit BRG1 to a set of MITF-associated regulatory elements (MAREs) at active enhancers. Combinations of MITF, SOX10, TFAP2A, and YY1 bind between two BRG1-occupied nucleosomes thus defining both a signature of transcription factors essential for the melanocyte lineage and a specific chromatin organisation of the regulatory elements they occupy. BRG1 also regulates the dynamics of MITF genomic occupancy. MITF-BRG1 interplay thus plays an essential role in transcription regulation in melanoma.


Assuntos
Montagem e Desmontagem da Cromatina , Cromatina/metabolismo , DNA Helicases/metabolismo , Melanoma/genética , Fator de Transcrição Associado à Microftalmia/metabolismo , Proteínas Nucleares/metabolismo , Sequências Reguladoras de Ácido Nucleico/genética , Fatores de Transcrição/metabolismo , Animais , Linhagem Celular Tumoral , Proliferação de Células , Reparo do DNA/genética , Replicação do DNA/genética , Regulação Neoplásica da Expressão Gênica , Genoma , Humanos , Melanócitos/metabolismo , Melanoma/patologia , Camundongos , Modelos Biológicos , Complexos Multiproteicos/metabolismo , Ligação Proteica , Transporte Proteico , Transcrição Gênica
15.
Elife ; 3: e03613, 2014 Sep 10.
Artigo em Inglês | MEDLINE | ID: mdl-25209997

RESUMO

The functions of the TAF subunits of mammalian TFIID in physiological processes remain poorly characterised. In this study, we describe a novel function of TAFs in directing genomic occupancy of a transcriptional activator. Using liver-specific inactivation in mice, we show that the TAF4 subunit of TFIID is required for post-natal hepatocyte maturation. TAF4 promotes pre-initiation complex (PIC) formation at post-natal expressed liver function genes and down-regulates a subset of embryonic expressed genes by increased RNA polymerase II pausing. The TAF4-TAF12 heterodimer interacts directly with HNF4A and in vivo TAF4 is necessary to maintain HNF4A-directed embryonic gene expression at post-natal stages and promotes HNF4A occupancy of functional cis-regulatory elements adjacent to the transcription start sites of post-natal expressed genes. Stable HNF4A occupancy of these regulatory elements requires TAF4-dependent PIC formation highlighting that these are mutually dependent events. Local promoter-proximal HNF4A-TFIID interactions therefore act as instructive signals for post-natal hepatocyte differentiation.


Assuntos
Diferenciação Celular/genética , Fator 4 Nuclear de Hepatócito/metabolismo , Hepatócitos/citologia , Hepatócitos/metabolismo , Regiões Promotoras Genéticas , Subunidades Proteicas/metabolismo , Fator de Transcrição TFIID/metabolismo , Animais , Animais Recém-Nascidos , Ductos Biliares/metabolismo , Ductos Biliares/patologia , Comunicação Celular , Colestase/complicações , Colestase/metabolismo , Colestase/patologia , Regulação para Baixo/genética , Genoma , Fator 4 Nuclear de Hepatócito/química , Proteínas de Homeodomínio/metabolismo , Hipoglicemia/complicações , Hipoglicemia/patologia , Camundongos , Mutação/genética , Ligação Proteica/genética , Multimerização Proteica , Estrutura Terciária de Proteína , RNA Polimerase II/metabolismo , Fatores Associados à Proteína de Ligação a TATA , Proteína de Ligação a TATA-Box/metabolismo , Fator de Transcrição TFIID/deficiência , Regulação para Cima/genética
16.
Development ; 134(16): 2947-58, 2007 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-17626060

RESUMO

The TAF4 subunit of transcription factor TFIID was inactivated in the basal keratinocytes of foetal and adult mouse epidermis. Loss of TAF4 in the foetal epidermis results in reduced expression of the genes required for skin barrier function, leading to early neonatal death. By contrast, TAF4 inactivation in adult epidermis leads to extensive fur loss and an aberrant hair cycle characterised by a defective anagen phase. Although the mutant epidermis contains few normal anagen-phase hair follicles, many genes expressed at this stage are strongly upregulated indicating desynchronized and inappropriate gene expression. The TAF4 mutant adult epidermis also displays interfollicular hyperplasia associated with a potent upregulation of several members of the EGF family of mitogens. Moreover, loss of TAF4 leads to malignant transformation of chemically induced papillomas and the appearance of invasive melanocytic tumours. Together, our results show that TAF4 is an important regulator of keratinocyte proliferation and has cell-autonomous and non-cell-autonomous tumour suppressor activity.


Assuntos
Proliferação de Células , Epiderme/metabolismo , Queratinócitos/citologia , Fatores Associados à Proteína de Ligação a TATA/metabolismo , Fatores Associados à Proteína de Ligação a TATA/fisiologia , Fator de Transcrição TFIID/metabolismo , Fator de Transcrição TFIID/fisiologia , Proteínas Supressoras de Tumor/metabolismo , Animais , Diferenciação Celular/genética , Epiderme/embriologia , Epiderme/patologia , Feminino , Predisposição Genética para Doença , Cabelo/citologia , Cabelo/embriologia , Hiperplasia/induzido quimicamente , Masculino , Camundongos , Camundongos Knockout , Nevo Pigmentado/induzido quimicamente , Nevo Pigmentado/genética , Subunidades Proteicas/fisiologia , Neoplasias Cutâneas/induzido quimicamente , Neoplasias Cutâneas/patologia , Fatores Associados à Proteína de Ligação a TATA/genética , Fator de Transcrição TFIID/genética , Tretinoína/efeitos adversos , Proteínas Supressoras de Tumor/fisiologia
17.
Cell Cycle ; 4(11): 1486-90, 2005 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-16205117

RESUMO

RNA polymerase II general transcription factor TFIID is a macromolecular complex comprising the TATA-binding protein, TBP and 13-14 evolutionary conserved TBP-associated factors, TAFs. Although genetic experiments have shown that TAFs are essential for cell cycle progression in yeast and in rapidly proliferating vertebrate cells in vitro, new experiments indicate they may be dispensible in specific developmental and physiological processes. Moreover, the TAF4 subunit of TFIID negatively regulates proliferation by inhibiting activation of the TGFbeta signalling pathway by its paralogue TAF4b. TAF4 is however essential in the retinoic acid and cAMP signalling pathways acting as a cofactor for CREB and the retinoic acid receptor, but is a negative regulator of the ATF7 transcription factor.


Assuntos
Ciclo Celular/fisiologia , Transdução de Sinais/fisiologia , Fatores Associados à Proteína de Ligação a TATA/fisiologia , Animais , Ciclo Celular/genética , Humanos , Transdução de Sinais/genética
18.
EMBO J ; 24(15): 2753-67, 2005 Aug 03.
Artigo em Inglês | MEDLINE | ID: mdl-16015375

RESUMO

We have inactivated transcription factor TFIID subunit TBP-associated factor 4 (TAF4) in mouse embryonic fibroblasts. Mutant taf4(-/-) cells are viable and contain intact TFIID comprising the related TAF4b showing that TAF4 is not an essential protein. TAF4 inactivation deregulates more than 1000 genes indicating that TFIID complexes containing TAF4 and TAF4b have distinct target gene specificities. However, taf4(-/-) cell lines have altered morphology and exhibit serum-independent autocrine growth correlated with the induced expression of several secreted mitotic factors and activators of the transforming growth factor beta signalling pathway. In addition to TAF4 inactivation, many of these genes can also be induced by overexpression of TAF4b. A competitive equilibrium between TAF4 and TAF4b therefore regulates expression of genes controlling cell proliferation. We have further identified a set of genes that are regulated both by TAF4 and upon adaptation to serum starvation and which may be important downstream mediators of serum-independent growth. Our study also shows that TAF4 is an essential cofactor for activation by the retinoic acid receptor and CREB, but not for Sp1 and the vitamin D3 receptor.


Assuntos
Comunicação Autócrina/fisiologia , Fibroblastos/metabolismo , Transdução de Sinais/fisiologia , Fatores Associados à Proteína de Ligação a TATA/antagonistas & inibidores , Fatores Associados à Proteína de Ligação a TATA/metabolismo , Fator de Transcrição TFIID/antagonistas & inibidores , Fator de Transcrição TFIID/metabolismo , Fator de Crescimento Transformador beta/metabolismo , Animais , Sobrevivência Celular/fisiologia , Proteína de Ligação ao Elemento de Resposta ao AMP Cíclico/metabolismo , Regulação da Expressão Gênica , Immunoblotting , Camundongos , Estrutura Terciária de Proteína , Receptores do Ácido Retinoico/metabolismo , Fatores Associados à Proteína de Ligação a TATA/genética , Fator de Transcrição TFIID/genética
19.
EMBO J ; 23(11): 2258-68, 2004 Jun 02.
Artigo em Inglês | MEDLINE | ID: mdl-15141166

RESUMO

Dosage compensation in flies involves doubling the transcription of genes on the single male X chromosome to match the combined expression level of the two female X chromosomes. Crucial for this activation is the acetylation of histone H4 by the histone acetyltransferase (HAT) MOF. In male cells, MOF resides in a complex (dosage compensation complex, DCC) with MSL proteins and noncoding roX RNA. Previous studies suggested that MOF's localization to the X chromosome was largely RNA-mediated. We now found that contact of the MOF chromo-related domain with roX RNA plays only a minor role in correct targeting to the X chromosome in vivo. Instead, a strong, direct interaction between a conserved MSL1 domain and a zinc finger within MOF's HAT domain is crucial. The functional consequences of this interaction were studied in vitro. Simultaneous contact of MOF with MSL1 and MSL3 led to its recruitment to chromatin, a dramatic stimulation of HAT activity and to improved substrate specificity. Activation of MOF's HAT activity upon integration into the DCC may serve to restrict the critical histone modification to the male X chromosome.


Assuntos
Acetiltransferases/metabolismo , Mecanismo Genético de Compensação de Dose , Proteínas de Drosophila , Drosophila melanogaster/metabolismo , Proteínas de Saccharomyces cerevisiae , Acetilação , Acetiltransferases/química , Animais , Células Cultivadas , Cromatina/metabolismo , Drosophila melanogaster/citologia , Drosophila melanogaster/enzimologia , Ativação Enzimática , Feminino , Histona Acetiltransferases , Masculino , Estrutura Terciária de Proteína , Proteínas Recombinantes de Fusão/metabolismo , Especificidade por Substrato , Transcrição Gênica , Transfecção , Cromossomo X , Dedos de Zinco
20.
J Cell Sci ; 116(Pt 9): 1847-58, 2003 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-12665565

RESUMO

Transcription regulation in male germ cells can involve specialised mechanisms and testis-specific paralogues of the general transcription machinery. Here we describe TAF7L, a germ-cell-specific paralogue of the TFIID subunit TAF7. TAF7L is expressed through most of the male germ-cell differentiation programme, but its intracellular localisation is dynamically regulated from cytoplasmic in spermatogonia and early spermatocytes to nuclear in late pachytene spermatocytes and haploid round spermatids. Import of TAF7L into the nucleus coincides with decreased TAF7 expression and a strong increase in nuclear TBP expression, which suggests that TAF7L replaces TAF7 as a TFIID subunit in late pachytene spermatocytes and in haploid cells. In agreement with this, biochemical experiments indicate that a subpopulation of TAF7L is tightly associated with TBP in both pachytene and haploid cells and TAF7L interacts with the TFIID subunit TAF1. We further show that TAF3, TAF4 and TAF10 are all strongly expressed in early spermatocytes, but that in contrast to TBP and TAF7L, they are downregulated in haploid cells. Hence, different subunits of the TFIID complex are regulated in distinct ways during male germ-cell differentiation. These results show for the first time how the composition of a general transcription factor such as TFIID and other TAF-containing complexes are modulated during a differentiation programme highlighting the unique nature of the transcription regulatory machinery in spermatogenesis.


Assuntos
Espermatogênese , Espermatozoides/metabolismo , Fatores Associados à Proteína de Ligação a TATA/metabolismo , Fator de Transcrição TFIID/metabolismo , Sequência de Aminoácidos , Animais , Clonagem Molecular , Feminino , Expressão Gênica , Líquido Intracelular/metabolismo , Masculino , Camundongos , Dados de Sequência Molecular , Subunidades Proteicas , Homologia de Sequência de Aminoácidos , Espermatogênese/genética , Fatores Associados à Proteína de Ligação a TATA/química , Fatores Associados à Proteína de Ligação a TATA/genética , Proteína de Ligação a TATA-Box/metabolismo , Distribuição Tecidual , Fator de Transcrição TFIID/química , Fator de Transcrição TFIID/genética
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa