Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 139
Filtrar
1.
Genes (Basel) ; 14(1)2023 01 14.
Artigo em Inglês | MEDLINE | ID: mdl-36672963

RESUMO

The SOX transcription factor family is pivotal in controlling aspects of development. To identify genotype-phenotype relationships of SOX proteins, we performed a non-biased study of SOX using 1890 open-reading frame and 6667 amino acid sequences in combination with structural dynamics to interpret 3999 gnomAD, 485 ClinVar, 1174 Geno2MP, and 4313 COSMIC human variants. We identified, within the HMG (High Mobility Group)- box, twenty-seven amino acids with changes in multiple SOX proteins annotated to clinical pathologies. These sites were screened through Geno2MP medical phenotypes, revealing novel SOX15 R104G associated with musculature abnormality and SOX8 R159G with intellectual disability. Within gnomAD, SOX18 E137K (rs201931544), found within the HMG box of ~0.8% of Latinx individuals, is associated with seizures and neurological complications, potentially through blood-brain barrier alterations. A total of 56 highly conserved variants were found at sites outside the HMG-box, including several within the SOX2 HMG-box-flanking region with neurological associations, several in the SOX9 dimerization region associated with Campomelic Dysplasia, SOX14 K88R (rs199932938) flanking the HMG box associated with cardiovascular complications within European populations, and SOX7 A379V (rs143587868) within an SOXF conserved far C-terminal domain heterozygous in 0.716% of African individuals with associated eye phenotypes. This SOX data compilation builds a robust genotype-to-phenotype association for a gene family through more robust ortholog data integration.


Assuntos
Proteínas de Grupo de Alta Mobilidade , Fatores de Transcrição SOX , Humanos , Proteínas de Grupo de Alta Mobilidade/química , Proteínas de Grupo de Alta Mobilidade/genética , Proteínas de Grupo de Alta Mobilidade/metabolismo , Fatores de Transcrição SOX/genética , Sequência de Aminoácidos , Dimerização , Genótipo , Fatores de Transcrição SOXF/genética , Fatores de Transcrição SOXF/metabolismo , Fatores de Transcrição SOXB2/genética , Fatores de Transcrição SOXB2/metabolismo , Fatores de Transcrição SOXE/genética
2.
Cells ; 11(19)2022 09 20.
Artigo em Inglês | MEDLINE | ID: mdl-36230893

RESUMO

Yeast Hmo1 is a high mobility group B (HMGB) protein that participates in the transcription of ribosomal protein genes and rDNA, and also stimulates the activities of some ATP-dependent remodelers. Hmo1 binds both DNA and nucleosomes and has been proposed to be a functional yeast analog of mammalian linker histones. We used EMSA and single particle Förster resonance energy transfer (spFRET) microscopy to characterize the effects of Hmo1 on nucleosomes alone and with the histone chaperone FACT. Hmo1 induced a significant increase in the distance between the DNA gyres across the nucleosomal core, and also caused the separation of linker segments. This was opposite to the effect of the linker histone H1, which enhanced the proximity of linkers. Similar to Nhp6, another HMGB factor, Hmo1, was able to support large-scale, ATP-independent, reversible unfolding of nucleosomes by FACT in the spFRET assay and partially support FACT function in vivo. However, unlike Hmo1, Nhp6 alone does not affect nucleosome structure. These results suggest physiological roles for Hmo1 that are distinct from Nhp6 and possibly from other HMGB factors and linker histones, such as H1.


Assuntos
Nucleossomos , Proteínas de Saccharomyces cerevisiae , Trifosfato de Adenosina/metabolismo , Animais , DNA Ribossômico/metabolismo , Proteínas de Ligação a DNA/metabolismo , Proteínas HMGB/genética , Proteínas HMGB/metabolismo , Proteínas de Grupo de Alta Mobilidade/química , Proteínas de Grupo de Alta Mobilidade/genética , Proteínas de Grupo de Alta Mobilidade/metabolismo , Chaperonas de Histonas/genética , Chaperonas de Histonas/metabolismo , Histonas/metabolismo , Mamíferos/metabolismo , Nucleossomos/metabolismo , Proteínas Ribossômicas/metabolismo , Saccharomyces cerevisiae/metabolismo , Proteínas de Saccharomyces cerevisiae/genética , Fatores de Elongação da Transcrição
3.
Transcription ; 13(1-3): 16-38, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35485711

RESUMO

The identification of FACT as a histone chaperone enabling transcription through chromatin in vitro has strongly shaped how its roles are envisioned. However, FACT has been implicated in essentially all aspects of chromatin biology, from transcription to DNA replication, DNA repair, and chromosome segregation. In this review, we focus on recent literature describing the role and mechanisms of FACT during transcription. We highlight the prime importance of FACT in preserving chromatin integrity during transcription and challenge its role as an elongation factor. We also review evidence for FACT's role as a cell-type/gene-specific regulator of gene expression and briefly summarize current efforts at using FACT inhibition as an anti-cancer strategy.


Assuntos
Proteínas de Grupo de Alta Mobilidade , Chaperonas de Histonas , Cromatina/genética , Proteínas de Ligação a DNA/metabolismo , Proteínas de Grupo de Alta Mobilidade/química , Proteínas de Grupo de Alta Mobilidade/genética , Proteínas de Grupo de Alta Mobilidade/metabolismo , Chaperonas de Histonas/química , Chaperonas de Histonas/genética , Chaperonas de Histonas/metabolismo , Nucleossomos , Fatores de Elongação da Transcrição/metabolismo
4.
Commun Biol ; 5(1): 2, 2022 01 10.
Artigo em Inglês | MEDLINE | ID: mdl-35013515

RESUMO

FACT is a histone chaperone that participates in nucleosome removal and reassembly during transcription and replication. We used electron microscopy to study FACT, FACT:Nhp6 and FACT:Nhp6:nucleosome complexes, and found that all complexes adopt broad ranges of configurations, indicating high flexibility. We found unexpectedly that the DNA binding protein Nhp6 also binds to the C-terminal tails of FACT subunits, inducing more open geometries of FACT even in the absence of nucleosomes. Nhp6 therefore supports nucleosome unfolding by altering both the structure of FACT and the properties of nucleosomes. Complexes formed with FACT, Nhp6, and nucleosomes also produced a broad range of structures, revealing a large number of potential intermediates along a proposed unfolding pathway. The data suggest that Nhp6 has multiple roles before and during nucleosome unfolding by FACT, and that the process proceeds through a series of energetically similar intermediate structures, ultimately leading to an extensively unfolded form.


Assuntos
Trifosfato de Adenosina/química , Proteínas de Ligação a DNA/química , Proteínas de Grupo de Alta Mobilidade/química , Nucleossomos/química , Proteínas de Saccharomyces cerevisiae/química , Fatores de Elongação da Transcrição/química , Humanos , Microscopia Eletrônica de Transmissão , Dobramento de Proteína , Saccharomyces cerevisiae/genética
5.
Trends Genet ; 37(11): 986-994, 2021 11.
Artigo em Inglês | MEDLINE | ID: mdl-34311989

RESUMO

High mobility group proteins (HMGs) are the most abundant nuclear proteins next to histones and are robustly expressed across tissues and organs. HMGs can uniquely bend or bind distorted DNA, and are central to such processes as transcription, recombination, and DNA repair. However, their dynamic association with chromatin renders capturing HMGs on chromosomes challenging. Recent work has changed this and now implicates these factors in spatial genome organization. Here, I revisit older and review recent literature to describe how HMGs rewire spatial chromatin interactions to sustain homeostasis or promote cellular aging. I propose a 'rheostat' model to explain how HMG-box proteins (HMGBs), and to some extent HMG A proteins (HMGAs), may control cellular aging and, likely, cancer progression.


Assuntos
Cromatina , Proteínas de Grupo de Alta Mobilidade , Proliferação de Células/genética , Cromatina/genética , DNA/metabolismo , Proteínas de Grupo de Alta Mobilidade/química , Proteínas de Grupo de Alta Mobilidade/metabolismo , Histonas/metabolismo
6.
J Mol Neurosci ; 71(8): 1714-1722, 2021 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-33595778

RESUMO

The P53N gene maps precisely to human chromosome sub-band 22q12.1-12.3, a region where loss of heterozygosity has been reported in 30% of astrocytic tumors and associated with progression to anaplasia. Moreover, a putative tumor suppressor gene has been indicated on 22q11 region involved in pathogenesis of ependymal tumors. Our objectives to examine the expression level of novel membrane-associated protein (termed P53N) encoded by a novel human gene on chromosome 22q12.1-12.3 in glioblastomas and ependymomas. Serial analysis of gene expression (SAGE) and immunofluorescence analysis of the P53N in the brain tumor tissues were performed. Our analysis revealed that there was high expression of the P53N mRNA in brain ependymoma and brain well-differentiated astrocytoma libraries. The P53N protein. P53N protein contains a high mobility group (HMG) domain at amino acid positions 301 to 360 expressed highly in glioblastoma and ependymoma specimens. Anti-P53N carboxyl-terminal peptide antibody localized the P53N protein to the cytoplasmic membranes of protoplasmic astrocytes in the glioblastoma and ependymoma specimens. These results are in good agreement with the SAGE analysis and the predicted transmembrane topology for the P53N protein and support a possible transmembrane model in which the P53N contains a predicted transmembrane region with its amino terminus localized to the inside of the cytoplasmic membrane.


Assuntos
Neoplasias Encefálicas/metabolismo , Cromossomos Humanos Par 22/genética , Ependimoma/metabolismo , Glioblastoma/metabolismo , Proteínas de Grupo de Alta Mobilidade/genética , Neoplasias Encefálicas/genética , Clonagem Molecular , Ependimoma/genética , Glioblastoma/genética , Proteínas de Grupo de Alta Mobilidade/química , Proteínas de Grupo de Alta Mobilidade/metabolismo , Humanos , Domínios Proteicos
7.
Int J Biol Macromol ; 160: 1144-1157, 2020 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-32454109

RESUMO

Affinity for G-quadruplex (G4) structures may be a common feature of transcription-facilitating histone chaperons (HCs). This assumption is based on previous unmatched studies of HCs FACT, nucleolin (NCL), BRD3, and ATRX. We verified this assumption and considered its implications for the therapeutic applications of synthetic (exogenous) G4s and the biological significance of genomic G4s. First, we questioned whether exogenous G4s that recognize cell-surface NCL and could trap other HCs in the nucleus are usable as anticancer agents. We performed in vitro binding assays and selected leading multi-targeted G4s. They exhibited minor effects on cell viability. The presumed NCL-regulated intracellular transport of G4s was inefficient or insufficient for tumor-specific G4 delivery. Next, to clarify whether G4s in the human genome could recruit HCs, we compared available HC ChIP-seq data with G4-seq/G4-ChIP-seq data. Several G4s, including the well-known c-Myc quadruplex structure, were found to be colocalized with HC occupancy sites in cancer cell lines. As evidenced by our molecular modeling data, c-Myc G4 might interfere with the HC function of BRD3 but is unlikely to prevent the BRD3-driven assembly of the chromatin remodeling complex. The c-Myc case illustrates the intricate role of genomic G4s in chromatin remodeling, nucleosome remodeling, and transcription.


Assuntos
Quadruplex G , Chaperonas de Histonas/química , Simulação de Acoplamento Molecular , Células A549 , Sítios de Ligação , Células Cultivadas , DNA/química , DNA/metabolismo , Proteínas de Ligação a DNA/química , Proteínas de Ligação a DNA/metabolismo , Genes myc , Células HT29 , Células Hep G2 , Proteínas de Grupo de Alta Mobilidade/química , Proteínas de Grupo de Alta Mobilidade/metabolismo , Chaperonas de Histonas/metabolismo , Humanos , Fosfoproteínas/química , Fosfoproteínas/metabolismo , Ligação Proteica , Proteínas de Ligação a RNA/química , Proteínas de Ligação a RNA/metabolismo , Fatores de Transcrição/química , Fatores de Transcrição/metabolismo , Fatores de Elongação da Transcrição/química , Fatores de Elongação da Transcrição/metabolismo , Proteína Nuclear Ligada ao X/química , Proteína Nuclear Ligada ao X/metabolismo , Nucleolina
8.
DNA Repair (Amst) ; 89: 102825, 2020 05.
Artigo em Inglês | MEDLINE | ID: mdl-32109764

RESUMO

Proteins form adducts with nucleic acids in a variety of contexts, and these adducts may be cytotoxic if not repaired. Here we apply a proteomic approach to identification of proteins adducted to DNA or RNA in normally proliferating cells. This approach combines RADAR fractionation of proteins covalently bound to nucleic acids with quantitative mass spectrometry (MS). We demonstrate that "RADAR-MS" can quantify induction of TOP1- or TOP2-DNA adducts in cells treated with topotecan or etoposide, respectively, and also identify intermediates in physiological adduct repair. We validate RADAR-MS for discovery of previously unknown adducts by determining the repertoires of adducted proteins in two different normally proliferating human cell lines, CCRF-CEM T cells and GM639 fibroblasts. These repertoires are significantly similar with one another and exhibit robust correlations in their quantitative profiles (Spearman r = 0.52). A very similar repertoire is identified by the classical approach of CsCl buoyant density gradient centrifugation. We find that in normally proliferating human cells, the repertoire of adducted proteins - the "adductome" - is comprised of a limited number of proteins belonging to specific functional groups, and that it is greatly enriched for histones, HMG proteins and proteins involved in RNA splicing. Treatment with low concentrations of formaldehyde caused little change in the composition of the repertoire of adducted proteins, suggesting that reactive aldehydes generated by ongoing metabolic processes may contribute to protein adduction in normally proliferating cells. The identification of an endogenous adductome highlights the importance of adduct repair in maintaining genomic structure and the potential for deficiencies in adduct repair to contribute to cancer.


Assuntos
Adutos de DNA/análise , Proteínas de Ligação a DNA/química , DNA/química , Espectrometria de Massas , Proteínas de Ligação a RNA/química , Linhagem Celular , Reagentes de Ligações Cruzadas/química , Reagentes de Ligações Cruzadas/farmacologia , DNA Topoisomerases Tipo I/química , Proteínas de Ligação a DNA/análise , Etoposídeo/química , Etoposídeo/farmacologia , Formaldeído/química , Formaldeído/farmacologia , Proteínas de Grupo de Alta Mobilidade/química , Histonas/química , Humanos , Proteômica , Proteínas de Ligação a RNA/análise , Topotecan/química , Topotecan/farmacologia
9.
Molecules ; 24(19)2019 Sep 24.
Artigo em Inglês | MEDLINE | ID: mdl-31554191

RESUMO

Cutaneous T-cell lymphomas (CTCL) are the most common primary lymphomas of the skin. We have previously identified thymocyte selection-associated high mobility group (HMG) box protein (TOX) as a promising drug target in CTCL; however, there are currently no small molecules able to directly inhibit TOX. We aimed to address this unmet opportunity by developing anti-TOX therapeutics with the use of computer-aided drug discovery methods. The available NMR-resolved structure of the TOX protein was used to model its DNA-binding HMG-box domain. To investigate the druggability of the corresponding protein-DNA interface on TOX, we performed a pilot virtual screening of 200,000 small molecules using in silico docking and identified 'hot spots' for drug-binding on the HMG-box domain. We then performed a large-scale virtual screening of 7.6 million drug-like compounds that were available from the ZINC15 database. As a result, a total of 140 top candidate compounds were selected for subsequent in vitro validation. Of those, 18 small molecules have been characterized as selective TOX inhibitors.


Assuntos
Antineoplásicos/química , Antineoplásicos/farmacologia , Desenho de Fármacos , Descoberta de Drogas/métodos , Proteínas de Grupo de Alta Mobilidade/antagonistas & inibidores , Proteínas de Grupo de Alta Mobilidade/química , Animais , Antineoplásicos/uso terapêutico , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Humanos , Linfoma Cutâneo de Células T/tratamento farmacológico , Camundongos , Simulação de Acoplamento Molecular , Simulação de Dinâmica Molecular , Ligação Proteica , Relação Quantitativa Estrutura-Atividade , Bibliotecas de Moléculas Pequenas
10.
Mol Biol (Mosk) ; 52(5): 737-749, 2018.
Artigo em Russo | MEDLINE | ID: mdl-30363049

RESUMO

HMGB proteins are involved in structural rearrangements caused by regulatory chromatin remodeling factors. Particular interest is attracted to a DNA chaperone mechanism, suggesting that the HMGB proteins introduce bends into the double helix, thus rendering DNA accessible to effector proteins and facilitating their activity. The review discusses the role that the HMBG proteins play in key intranuclear processes, including assembly of the preinitiation complex during transcription of ribosomal genes; transcription by RNA polymerases I, II, and III; recruitment of the SWI/SNF complex during transcription of nonribosomal genes; DNA repair; etc. The functions of the HMGB proteins are considered in detail with the examples of yeast HMO1 and NHP6. The two proteins possess unique features in adition to properties characteristic of the HMGB proteins. Thus, NHP6 stimulates a large-scale ATP-independent unwrapping of nucleosomal DNA by the FACT complex, while in its absence FACT stabilizes the nucleosome. HMO1 acts as an alternative linker histone. Both HMO1 and NHP6 are of applied interest primarly because they are homologs of human HMGB1, an important therapeutic target of anticancer and anti-inflammatory treatments.


Assuntos
Cromatina/química , Proteínas HMGB/química , Proteínas HMGN/química , Proteínas de Grupo de Alta Mobilidade/química , Chaperonas Moleculares/química , Proteínas de Saccharomyces cerevisiae/química , DNA/química , Histonas/química , Humanos , Nucleossomos/química , Saccharomyces cerevisiae/química
11.
Nat Struct Mol Biol ; 23(12): 1111-1116, 2016 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-27820806

RESUMO

DNA accessibility to regulatory proteins is substantially influenced by nucleosome structure and dynamics. The facilitates chromatin transcription (FACT) complex increases the accessibility of nucleosomal DNA, but the mechanism and extent of its nucleosome reorganization activity are unknown. Here we determined the effects of FACT from the yeast Saccharomyces cerevisiae on single nucleosomes by using single-particle Förster resonance energy transfer (spFRET) microscopy. FACT binding results in dramatic ATP-independent, symmetrical and reversible DNA uncoiling that affects at least 70% of the DNA within a nucleosome, occurs without apparent loss of histones and proceeds via an 'all-or-none' mechanism. A mutated version of FACT is defective in uncoiling, and a histone mutation that suppresses phenotypes caused by this FACT mutation in vivo restores the uncoiling activity in vitro. Thus, FACT-dependent nucleosome unfolding modulates the accessibility of nucleosomal DNA, and this activity is an important function of FACT in vivo.


Assuntos
Trifosfato de Adenosina/metabolismo , DNA Fúngico/metabolismo , Proteínas de Ligação a DNA/metabolismo , Proteínas de Grupo de Alta Mobilidade/metabolismo , Histonas/metabolismo , Nucleossomos/metabolismo , Proteínas de Saccharomyces cerevisiae/metabolismo , Saccharomyces cerevisiae/metabolismo , Fatores de Transcrição/metabolismo , Fatores de Elongação da Transcrição/metabolismo , DNA Fúngico/química , Proteínas de Ligação a DNA/química , Transferência Ressonante de Energia de Fluorescência , Proteínas de Grupo de Alta Mobilidade/química , Histonas/química , Modelos Moleculares , Chaperonas Moleculares/química , Chaperonas Moleculares/metabolismo , Conformação de Ácido Nucleico , Nucleossomos/química , Ligação Proteica , Saccharomyces cerevisiae/química , Proteínas de Saccharomyces cerevisiae/química , Fatores de Transcrição/química , Fatores de Elongação da Transcrição/química
12.
J Mol Model ; 20(8): 2357, 2014 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-25056123

RESUMO

NUPR1, a small chromatin protein, plays a critical role in cancer development, progression, and resistance to therapy. Here, using a combination of structural bioinformatics and molecular modeling methods, we report several novel findings that enhance our understanding of the biochemical function of this protein. We find that NUPR1 has been conserved throughout evolution, and over time it has undergone duplications and transpositions to form other transcriptional regulators. Using threading, homology-based molecular modeling, molecular mechanics calculations, and molecular dynamics simulations, we generated structural models for four of these proteins: NUPR1a, NUPR1b, NUPR2, and the NUPR-like domain of GTF2-I. Comparative analyses of these models combined with extensive linear motif identification reveal that these four proteins, though similar in their propensities for folding, differ in size, surface changes, and sites amenable for posttranslational modification. Lastly, taking NUPR1a as the paradigm for this family, we built models of a NUPR-DNA complex. Additional structural comparisons revealed that NUPR1 defines a new family of small-groove-binding proteins that share structural features with, yet are distinct from, helix-loop-helix AT-hook-containing HMG proteins. These models and inferences should lead to a better understanding of the function of this group of chromatin proteins, which play a critical role in the development of human malignant diseases.


Assuntos
Motivos AT-Hook , Fatores de Transcrição Hélice-Alça-Hélice Básicos/química , Sequências Hélice-Volta-Hélice , Proteínas de Grupo de Alta Mobilidade/química , Proteínas de Neoplasias/química , Sequência de Aminoácidos , Sequência Conservada , DNA/metabolismo , Humanos , Modelos Moleculares , Dados de Sequência Molecular , Proteínas Mutantes/química , Conformação de Ácido Nucleico , Ligação Proteica , Processamento de Proteína Pós-Traducional , Estrutura Terciária de Proteína , Transcrição Gênica
13.
Cancer Biother Radiopharm ; 27(5): 329-35, 2012 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-22655796

RESUMO

Our previous studies have shown that high-mobility group box 1 (HMGB1) could physically associate with the retinoblastoma (RB) protein via an LXCXE (leucine-X-cysteine-X-glutamic; X=any amino acid) motif. An identical LXCXE motif is present in the HMGB1-3 protein sequences, whereas a near-consensus LXCXD (leucine-X-cysteine-X-asparagine; X=any amino acid) motif is found in the HMGB4 protein. In this study, we have demonstrated that like HMGB1, HMGB2-3 also associated with the RB in vitro and in vivo, as evidenced by glutathione-s-transferase capture and immunoprecipitation-Western blot assays. A point mutation of the LXCXE or LXCXD motif led to disruption of RB:HMGB1-4 interactions. Enforced expression of HMGB1-3 or HMGB4 by adenoviral-vector-mediated gene transfer resulted in significant inhibition of breast cancer cell proliferation through an LXCXE- or LXCXD-dependent mechanism and an increased radiosensitivity through an LXCXE- or LXCXD-independent mechanism. These results suggest an important role of the LXCXE/D motif in RB:HMGB1-4 association and modulation of cancer cell growth, but not radiosensitivity.


Assuntos
Neoplasias da Mama/metabolismo , Proteínas de Grupo de Alta Mobilidade/metabolismo , Proteína do Retinoblastoma/metabolismo , Sequência de Aminoácidos , Sítios de Ligação , Neoplasias da Mama/genética , Neoplasias da Mama/patologia , Neoplasias da Mama/radioterapia , Processos de Crescimento Celular/fisiologia , Linhagem Celular Tumoral , Feminino , Proteínas de Grupo de Alta Mobilidade/química , Proteínas de Grupo de Alta Mobilidade/genética , Humanos , Dados de Sequência Molecular , Ligação Proteica , Tolerância a Radiação , Retinoblastoma/genética , Retinoblastoma/metabolismo , Retinoblastoma/patologia , Proteína do Retinoblastoma/genética , Transfecção
14.
Proc Natl Acad Sci U S A ; 109(21): 8085-90, 2012 May 22.
Artigo em Inglês | MEDLINE | ID: mdl-22570500

RESUMO

The LSD1-CoREST histone demethylase complex is required to repress neuronal genes in nonneuronal tissues. Here we show that sumoylation of Braf35, one of the subunits of the complex, is required to maintain full repression of neuron-specific genes and for occupancy of the LSD1-CoREST complex at its gene targets. Interestingly, expression of Braf35 was sufficient to prevent neuronal differentiation induced by bHLH neurogenic transcription factors in P19 cells and in neuronal progenitors of the chicken embryo neural tube. Sumoylation of Braf35 is required for this antineurogenic activity. We also show that iBraf, a paralogue of Braf35, forms heterodimers with Braf35. Braf35-iBraf heterodimerization impairs Braf35 interaction with the LSD1-CoREST complex and inhibits Braf35 sumoylation. Consistent with these results, iBraf prevents the antineurogenic activity of Braf35 in vivo. Our data uncover a mechanism of regulation of the LSD1-CoREST complex and provide a molecular explanation for the antagonism between Braf35 and iBraf in neuronal differentiation.


Assuntos
Proteínas de Grupo de Alta Mobilidade/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Neurogênese/fisiologia , Neurônios/citologia , Oxirredutases N-Desmetilantes/metabolismo , Proteínas Repressoras/metabolismo , Animais , Proteínas de Ciclo Celular , Diferenciação Celular/fisiologia , Embrião de Galinha , Galinhas , Proteínas Correpressoras , Proteínas de Ligação a DNA , Dimerização , Células-Tronco de Carcinoma Embrionário/citologia , Regulação da Expressão Gênica/fisiologia , Células HEK293 , Células HeLa , Proteínas de Grupo de Alta Mobilidade/química , Proteínas de Grupo de Alta Mobilidade/genética , Histona Desmetilases/química , Histona Desmetilases/metabolismo , Humanos , Camundongos , Complexos Multiproteicos/química , Complexos Multiproteicos/metabolismo , Proteínas do Tecido Nervoso/química , Oxirredutases N-Desmetilantes/química , Proteínas Repressoras/química , Sumoilação/fisiologia
15.
Nucleic Acids Res ; 40(3): 981-95, 2012 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-21967847

RESUMO

HBP1 is a sequence-specific DNA-binding transcription factor with many important biological roles. It activates or represses the expression of some specific genes during cell growth and differentiation. Previous studies have exhibited that HBP1 binds to p16(INK4A) promoter and activates p16(INK4A) expression. We found that trichostatin A (TSA), an inhibitor of HDAC (histone deacetylase), induces p16(INK4A) expression in an HBP1-dependent manner. This result was drawn from a transactivation experiment by measuring relative luciferase activities of p16(INK4A) promoter with HBP1-binding site in comparison with that of the wild-type p16(INK4A) promoter by transient cotransfection with HBP1 into HEK293T cells and 2BS cells. HBP1 acetylation after TSA treatment was confirmed by immunoprecipitation assay. Our data showed that HBP1 interacted with histone acetyltransferase p300 and CREB-binding protein (CBP) and also recruited p300/CBP to p16(INK4A) promoter. HBP1 was acetylated by p300/CBP in two regions: repression domain (K297/305/307) and P domain (K171/419). Acetylation of Repression domain was not required for HBP1 transactivation on p16(INK4A). However, luciferase assay and western blotting results indicate that acetylation of P domain, especially K419 acetylation is essential for HBP1 transactivation on p16(INK4A). As assayed by SA-beta-gal staining, the acetylation of HBP1 at K419 enhanced HBP1-induced premature senescence in 2BS cells. In addition, HDAC4 repressed HBP1-induced premature senescence through permanently deacetylating HBP1. We conclude that our data suggest that HBP1 acetylation at K419 plays an important role in HBP1-induced p16(INK4A) expression.


Assuntos
Proteína de Ligação a CREB/metabolismo , Inibidor p16 de Quinase Dependente de Ciclina/genética , Proteínas de Grupo de Alta Mobilidade/metabolismo , Proteínas Repressoras/metabolismo , Ativação Transcricional , Fatores de Transcrição de p300-CBP/metabolismo , Acetilação , Linhagem Celular , Senescência Celular , DNA/metabolismo , Proteínas de Grupo de Alta Mobilidade/química , Inibidores de Histona Desacetilases/farmacologia , Histona Desacetilases/metabolismo , Humanos , Ácidos Hidroxâmicos/farmacologia , Lisina/metabolismo , Regiões Promotoras Genéticas , Estrutura Terciária de Proteína , Proteínas Repressoras/química
17.
Biochemistry ; 50(17): 3462-8, 2011 May 03.
Artigo em Inglês | MEDLINE | ID: mdl-21417337

RESUMO

High mobility group A proteins (HMGA1 and HMGA2) are architectural factors involved in chromatin remodelling and regulation of gene expression. HMGA are highly expressed during embryogenesis and in cancer cells and are involved in development and cell differentiation as well as cancer formation and progression. These factors, by binding to DNA and interacting with other nuclear proteins, can organize macromolecular complexes involved in transcription, chromatin dynamics, RNA processing, and DNA repair. The identification of protein partners for HMGA has greatly contributed to our understanding of their multiple functions. He we report the identification of HMGA molecular partners using a gene fragment library in a phage display screening. Using an ORF-enriched cDNA library, we have isolated several HMGA1 interacting clones and for two of them, TBP associated factor 3 (TAF3) and chromatin assembly factor 1 p150/CAF-1, have demonstrated an in vivo association with HMGA1. The identification of these new partners suggests that HMGA can also influence general aspects of transcription and once more underlines their involvement in chromatin remodelling and dynamics.


Assuntos
Proteínas de Grupo de Alta Mobilidade/química , Biblioteca de Peptídeos , Sequência de Aminoácidos , Animais , Fator 1 de Modelagem da Cromatina/química , DNA Complementar/química , Células HEK293 , Proteínas de Grupo de Alta Mobilidade/genética , Proteínas de Grupo de Alta Mobilidade/isolamento & purificação , Humanos , Camundongos , Dados de Sequência Molecular , Mutação , Fases de Leitura Aberta , Mapeamento de Interação de Proteínas , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/isolamento & purificação , Deleção de Sequência , Fatores Associados à Proteína de Ligação a TATA/química
18.
Biochim Biophys Acta ; 1784(9): 1159-66, 2008 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-18513496

RESUMO

The high mobility group (HMG) proteins, including HMGA, HMGB and HMGN, are abundant and ubiquitous nuclear proteins that bind to DNA, nucleosome and other multi-protein complexes in a dynamic and reversible fashion to regulate DNA processing in the context of chromatin. All HMG proteins, like histone proteins, are subjected to extensive post-translational modifications (PTMs), such as lysine acetylation, arginine/lysine methylation and serine/threonine phosphorylation, to modulate their interactions with DNA and other proteins. There is a growing appreciation for the complex relationship between the PTMs of HMG proteins and their diverse biological activities. Here, we reviewed the identified covalent modifications of HMG proteins, and highlighted how these PTMs affect the functions of HMG proteins in a variety of cellular processes.


Assuntos
Proteínas de Grupo de Alta Mobilidade/química , Proteínas de Grupo de Alta Mobilidade/metabolismo , Acetilação , Transporte Ativo do Núcleo Celular , Sequência de Aminoácidos , Animais , Proteínas de Grupo de Alta Mobilidade/genética , Histonas/metabolismo , Humanos , Metilação , Modelos Moleculares , Dados de Sequência Molecular , Neoplasias/genética , Neoplasias/metabolismo , Fosforilação , Processamento de Proteína Pós-Traducional , Estrutura Terciária de Proteína
19.
Nucleic Acids Res ; 36(9): 3011-24, 2008 May.
Artigo em Inglês | MEDLINE | ID: mdl-18390577

RESUMO

Sox9 is a transcription factor of the SRY family required for several steps of chondrogenesis. It activates the expression of various chondrocyte-specific genes, but the mechanisms and role of cofactors involved in Sox9-regulated gene transcription are not fully understood. Here, we report on the characterization of a Tat interactive protein-60 (Tip60) as Sox9-associated protein identified in a yeast two-hybrid screen. Both in vitro and in vivo assays confirmed the specificity of interactions between Sox9 and Tip60 including the existence of an endogenous complex containing both polypeptides in chondrocytes. Gel shift assays showed the presence of a complex containing Sox9, Tip60 and the DNA of an enhancer region of the Col2a1 promoter. Reporter assays using a Col2a1 promoter with multimerized Col2a1 Sox9-binding sites indicated that Tip60 enhanced the transcriptional activity of Sox9. A larger Col2a1 promoter showed that Tip60 increased the activity of this promoter in the presence of both Sox9 and Sox5. Ectopic expression of Sox9 and transient-cotransfection with Tip60 in COS7 cells showed a more diffuse subnuclear colocalization, suggesting changes in the chromatin structure. Chromatin immunoprecipitation assays showed that Tip60, Sox9 and Sox5 associated with the same Col2a1 enhancer region. Consistent with a role of Tip60 in chondrogenesis, addition of Tip60 siRNA to limb-bud micromass cultures delayed chondrocyte differention. Tip60 enhances acetylation of Sox9 mainly through K61, 253, 398 residues; however, the K61/253/398A mutant of Sox9 still exhibited enhanced transcriptional activity by Tip60. Our results support the hypothesis that Tip60 is a coactivator of Sox9 in chondrocytes.


Assuntos
Condrogênese/genética , Cromatina/metabolismo , Proteínas de Ligação a DNA/metabolismo , Proteínas de Grupo de Alta Mobilidade/metabolismo , Histona Acetiltransferases/metabolismo , Proteínas Nucleares/metabolismo , Fatores de Transcrição/metabolismo , Ativação Transcricional , Acetilação , Animais , Células COS , Núcleo Celular/química , Células Cultivadas , Chlorocebus aethiops , Condrócitos/metabolismo , Imunoprecipitação da Cromatina , Colágeno Tipo II/genética , Elementos Facilitadores Genéticos , Proteínas de Grupo de Alta Mobilidade/química , Histona Acetiltransferases/análise , Humanos , Lisina/metabolismo , Lisina Acetiltransferase 5 , Camundongos , Fatores de Transcrição SOX9 , Fatores de Transcrição SOXD , Fatores de Transcrição/química , Técnicas do Sistema de Duplo-Híbrido
20.
Nucleic Acids Res ; 36(2): 570-7, 2008 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-18056081

RESUMO

Post-translational modification of histones and other chromosomal proteins regulates chromatin conformation and gene activity. Methylation and acetylation of lysyl residues are among the most frequently described modifications in these proteins. Whereas these modifications have been studied in detail, very little is known about a recently discovered chemical modification, the N(epsilon)-lysine formylation, in histones and other nuclear proteins. Here we mapped, for the first time, the sites of lysine formylation in histones and several other nuclear proteins. We found that core and linker histones are formylated at multiple lysyl residues located both in the tails and globular domains of histones. In core histones, formylation was found at lysyl residues known to be involved in organization of nucleosomal particles that are frequently acetylated and methylated. In linker histones and high mobility group proteins, multiple formylation sites were mapped to residues with important role in DNA binding. N(epsilon)-lysine formylation in chromosomal proteins is relatively abundant, suggesting that it may interfere with epigenetic mechanisms governing chromatin function, which could lead to deregulation of the cell and disease.


Assuntos
Proteínas Cromossômicas não Histona/química , Proteínas Cromossômicas não Histona/metabolismo , Histonas/química , Histonas/metabolismo , Lisina/química , Processamento de Proteína Pós-Traducional , Aldeídos/química , Animais , Linhagem Celular Tumoral , Cromatina/metabolismo , Proteínas de Grupo de Alta Mobilidade/química , Proteínas de Grupo de Alta Mobilidade/metabolismo , Humanos , Lisina/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Proteínas Nucleares/química , Proteínas Nucleares/metabolismo , Espectrometria de Massas em Tandem
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA