Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 60
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Biomed Pharmacother ; 165: 115118, 2023 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-37437373

RESUMO

Cardiovascular disease (CVD) is the most fatal disease that causes sudden death, and inflammation contributes substantially to its occurrence and progression. The prevalence of CVD increases as the population ages, and the pathophysiology is complex. Anti-inflammatory and immunological modulation are the potential methods for CVD prevention and treatment. High-Mobility Group (HMG) chromosomal proteins are one of the most abundant nuclear nonhistone proteins which act as inflammatory mediators in DNA replication, transcription, and repair by producing cytokines and serving as damage-associated molecular patterns in inflammatory responses. The most common and well-studied HMG proteins are those with an HMGB domain, which participate in a variety of biological processes. HMGB1 and HMGB2 were the first members of the HMGB family to be identified and are present in all investigated eukaryotes. Our review is primarily concerned with the involvement of HMGB1 and HMGB2 in CVD. The purpose of this review is to provide a theoretical framework for diagnosing and treating CVD by discussing the structure and function of HMGB1 and HMGB2.


Assuntos
Doenças Cardiovasculares , Proteína HMGB1 , Humanos , Proteína HMGB1/genética , Proteína HMGB1/metabolismo , Proteína HMGB2/genética , Proteína HMGB2/metabolismo , Proteínas HMGB/química , Proteínas HMGB/metabolismo , Biomarcadores
2.
Wiley Interdiscip Rev RNA ; 14(5): e1778, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-36646476

RESUMO

Nucleic acid binding proteins regulate transcription, splicing, RNA stability, RNA localization, and translation, together tailoring gene expression in response to stimuli. Upon discovery, these proteins are typically classified as either DNA or RNA binding as defined by their in vivo functions; however, recent evidence suggests dual DNA and RNA binding by many of these proteins. High mobility group box (HMGB) proteins have a DNA binding HMGB domain, act as transcription factors and chromatin remodeling proteins, and are increasingly understood to interact with RNA as means to regulate gene expression. Herein, multiple layers of evidence that the HMGB family are dual DNA and RNA binding proteins is comprehensively reviewed. For example, HMGB proteins directly interact with RNA in vitro and in vivo, are localized to RNP granules involved in RNA processing, and their protein interactors are enriched in RNA binding proteins involved in RNA metabolism. Importantly, in cell-based systems, HMGB-RNA interactions facilitate protein-protein interactions, impact splicing outcomes, and modify HMGB protein genomic or cellular localization. Misregulation of these HMGB-RNA interactions are also likely involved in human disease. This review brings to light that as a family, HMGB proteins are likely to bind RNA which is essential to HMGB protein biology. This article is categorized under: RNA Interactions with Proteins and Other Molecules > Protein-RNA Recognition RNA Interactions with Proteins and Other Molecules > RNA-Protein Complexes RNA Interactions with Proteins and Other Molecules > Protein-RNA Interactions: Functional Implications.


Assuntos
Proteínas HMGB , RNA , Humanos , RNA/genética , RNA/metabolismo , Proteínas HMGB/química , Proteínas HMGB/genética , Proteínas HMGB/metabolismo , Proteínas de Ligação a DNA/genética , Fatores de Transcrição/metabolismo , DNA/metabolismo , Proteínas de Ligação a RNA/metabolismo , Ligação Proteica
3.
Curr Biol ; 31(23): 5227-5237.e7, 2021 12 06.
Artigo em Inglês | MEDLINE | ID: mdl-34666003

RESUMO

Virus infection necessarily requires redirecting cellular resources toward viral progeny production. Adenovirus encodes the histone-like protein VII, which causes catastrophic global reorganization of host chromatin to promote virus infection. Protein VII recruits the family of high mobility group box (HMGB) proteins to chromatin along with the histone chaperone SET. As a consequence of this recruitment, we find that protein VII causes chromatin depletion of several linker histone H1 isoforms. The relationship between linker histone H1 and the functionally opposite HMGB proteins is critical for higher-order chromatin structure. However, the physiological consequences of perturbing this relationship are largely unknown. Here, we employ complementary systems in Saccharomyces cerevisiae and human cells to demonstrate that adenovirus protein VII disrupts the H1-HMGB balance to obstruct the cell cycle. We find that protein VII causes an accumulation of G2/M cells both in yeast and human systems, underscoring the high conservation of this chromatin vulnerability. In contrast, adenovirus E1A and E1B proteins are well established to override cell cycle regulation and promote transformation of human cells. Strikingly, we find that protein VII obstructs the cell cycle, even in the presence of E1A and E1B. We further show that, in a protein-VII-deleted infection, several cell cycle markers are regulated differently compared to wild-type infection, supporting our model that protein VII plays an integral role in hijacking cell cycle regulation during infection. Together, our results demonstrate that protein VII targets H1-HMGB1 antagonism to obstruct cell cycle progression, revealing an unexpected chromatin vulnerability exploited for viral benefit.


Assuntos
Proteínas HMGB , Histonas , Ciclo Celular , Cromatina , Proteínas HMGB/química , Proteínas HMGB/metabolismo , Histonas/genética , Histonas/metabolismo , Humanos , Saccharomyces cerevisiae/genética , Saccharomyces cerevisiae/metabolismo , Proteínas Virais/metabolismo
4.
Biomolecules ; 11(9)2021 09 21.
Artigo em Inglês | MEDLINE | ID: mdl-34572607

RESUMO

In the traditional fermentative model yeast Saccharomyces cerevisiae, ScIxr1 is an HMGB (High Mobility Group box B) protein that has been considered as an important regulator of gene transcription in response to external changes like oxygen, carbon source, or nutrient availability. Kluyveromyces lactis is also a useful eukaryotic model, more similar to many human cells due to its respiratory metabolism. We cloned and functionally characterized by different methodologies KlIXR1, which encodes a protein with only 34.4% amino acid sequence similarity to ScIxr1. Our data indicate that both proteins share common functions, including their involvement in the response to hypoxia or oxidative stress induced by hydrogen peroxide or metal treatments, as well as in the control of key regulators for maintenance of the dNTP (deoxyribonucleotide triphosphate) pool and ribosome synthesis. KlIxr1 is able to bind specific regulatory DNA sequences in the promoter of its target genes, which are well conserved between S. cerevisiae and K. lactis. Oppositely, we found important differences between ScIrx1 and KlIxr1 affecting cellular responses to cisplatin or cycloheximide in these yeasts, which could be dependent on specific and non-conserved domains present in these two proteins.


Assuntos
Desoxirribonucleotídeos/metabolismo , Proteínas Fúngicas/metabolismo , Regulação Fúngica da Expressão Gênica , Proteínas HMGB/metabolismo , Kluyveromyces/crescimento & desenvolvimento , Kluyveromyces/genética , Sequência de Bases , Cádmio/toxicidade , Carbono/farmacologia , Ciclo Celular/efeitos dos fármacos , Cisplatino/farmacologia , Resistência a Medicamentos/efeitos dos fármacos , Proteínas Fúngicas/química , Deleção de Genes , Regulação Fúngica da Expressão Gênica/efeitos dos fármacos , Proteínas HMGB/química , Heme/biossíntese , Peróxido de Hidrogênio/toxicidade , Kluyveromyces/efeitos dos fármacos , Mutação/genética , Oxirredução/efeitos dos fármacos , Fenótipo , Regiões Promotoras Genéticas , Ligação Proteica/efeitos dos fármacos , Processamento Pós-Transcricional do RNA/efeitos dos fármacos , RNA Ribossômico/genética , Ribossomos/efeitos dos fármacos , Ribossomos/metabolismo , Saccharomyces cerevisiae/metabolismo
5.
Biomolecules ; 10(8)2020 08 16.
Artigo em Inglês | MEDLINE | ID: mdl-32824374

RESUMO

Mitochondrial DNA (mtDNA) molecules are packaged into compact nucleo-protein structures called mitochondrial nucleoids (mt-nucleoids). Their compaction is mediated in part by high-mobility group (HMG)-box containing proteins (mtHMG proteins), whose additional roles include the protection of mtDNA against damage, the regulation of gene expression and the segregation of mtDNA into daughter organelles. The molecular mechanisms underlying these functions have been identified through extensive biochemical, genetic, and structural studies, particularly on yeast (Abf2) and mammalian mitochondrial transcription factor A (TFAM) mtHMG proteins. The aim of this paper is to provide a comprehensive overview of the biochemical properties of mtHMG proteins, the structural basis of their interaction with DNA, their roles in various mtDNA transactions, and the evolutionary trajectories leading to their rapid diversification. We also describe how defects in the maintenance of mtDNA in cells with dysfunctional mtHMG proteins lead to different pathologies at the cellular and organismal level.


Assuntos
DNA Mitocondrial/genética , Proteínas HMGB/metabolismo , Doenças Mitocondriais/genética , DNA Mitocondrial/metabolismo , Regulação da Expressão Gênica , Proteínas HMGB/química , Humanos , Mitocôndrias/genética , Mitocôndrias/metabolismo , Ligação Proteica
6.
Genes (Basel) ; 10(10)2019 10 12.
Artigo em Inglês | MEDLINE | ID: mdl-31614829

RESUMO

Development requires the careful orchestration of several biological events in order to create any structure and, eventually, to build an entire organism. On the other hand, the fate transformation of terminally differentiated cells is a consequence of erroneous development, and ultimately leads to cancer. In this review, we elaborate how development and cancer share several biological processes, including molecular controls. Transcription factors (TF) are at the helm of both these processes, among many others, and are evolutionarily conserved, ranging from yeast to humans. Here, we discuss four families of TFs that play a pivotal role and have been studied extensively in both embryonic development and cancer-high mobility group box (HMG), GATA, paired box (PAX) and basic helix-loop-helix (bHLH) in the context of their role in development, cancer, and their conservation across several species. Finally, we review TFs as possible therapeutic targets for cancer and reflect on the importance of natural resistance against cancer in certain organisms, yielding knowledge regarding TF function and cancer biology.


Assuntos
Desenvolvimento Embrionário , Neoplasias/metabolismo , Fatores de Transcrição/metabolismo , Fatores de Transcrição Hélice-Alça-Hélice Básicos/química , Fatores de Transcrição Hélice-Alça-Hélice Básicos/genética , Fatores de Transcrição Hélice-Alça-Hélice Básicos/metabolismo , Movimento Celular/genética , Movimento Celular/imunologia , Desenvolvimento Embrionário/genética , Transição Epitelial-Mesenquimal/genética , Transição Epitelial-Mesenquimal/fisiologia , Fatores de Transcrição GATA/química , Fatores de Transcrição GATA/genética , Fatores de Transcrição GATA/metabolismo , Proteínas HMGB/química , Proteínas HMGB/genética , Proteínas HMGB/metabolismo , Humanos , Neoplasias/tratamento farmacológico , Neoplasias/genética , Fatores de Transcrição Box Pareados/química , Fatores de Transcrição Box Pareados/genética , Fatores de Transcrição Box Pareados/metabolismo , Fatores de Transcrição/química , Fatores de Transcrição/efeitos dos fármacos , Fatores de Transcrição/genética
7.
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
8.
Gene ; 671: 152-160, 2018 Sep 10.
Artigo em Inglês | MEDLINE | ID: mdl-29859286

RESUMO

High-mobility group B (HMGB) proteins have highly conserved, unique DNA-binding domains, HMG boxes, that can bind non-B-type DNA structures, such as bent, kinked and unwound structures, with high affinity. HMGB proteins also promote DNA bending, looping and unwinding. In this study, we determined the role of the Aedes aegypti single HMG-box domain protein AaHMGB; characterized its structure, spatiotemporal expression levels, subcellular localization, and nucleic acid binding activities; and compared these properties with those of its double-HMG-box counterpart protein, AaHMGB1. Via qRT-PCR, we showed that AaHMGB is expressed at much higher levels than AaHMGB1 throughout mosquito development. In situ hybridization results suggested a role for AaHMGB and AaHMGB1 during embryogenesis. Immunolocalization in the midgut revealed that AaHMGB is exclusively nuclear. Circular dichroism and fluorescence spectroscopy analyses showed that AaHMGB exhibits common features of α-helical structures and is more stably folded than AaHMGB1, likely due to the presence of one or two HMG boxes. Using several DNA substrates or single-stranded RNAs as probes, we observed significant differences between AaHMGB and AaHMGB1 in terms of their binding patterns, activity and/or specificity. Importantly, we showed that the phosphorylation of AaHMGB plays a critical role in its DNA-binding activity. Our study provides additional insight into the roles of single- versus double-HMG-box-containing proteins in nucleic acid interactions for better understanding of mosquito development, physiology and homeostasis.


Assuntos
Aedes/crescimento & desenvolvimento , Proteínas HMGB/química , Proteínas HMGB/genética , Proteínas HMGB/metabolismo , Aedes/genética , Aedes/metabolismo , Animais , Núcleo Celular/metabolismo , Dicroísmo Circular , Regulação da Expressão Gênica no Desenvolvimento , Proteínas de Insetos/química , Proteínas de Insetos/genética , Proteínas de Insetos/metabolismo , Modelos Moleculares , Fosforilação , Estrutura Secundária de Proteína , Distribuição Tecidual
9.
FEMS Yeast Res ; 18(2)2018 03 01.
Artigo em Inglês | MEDLINE | ID: mdl-29438513

RESUMO

Ixr1 is a Saccharomyces cerevisiae transcriptional factor that extensively regulates the response to hypoxia and controls other important cellular functions and DNA repair. During aerobic growth, the Ixr1 repressor function is predominant on regulated promoters of hypoxic genes, although activator effects are also observed on other genes. During hypoxia, Ixr1 expression increases and the number of genes activated by Ixr1 also increase. In this work we demonstrate that the NH2-terminal region of Ixr1 is involved in transcriptional activation. We also present the first analysis about Ixr1 interactions with three factors that have been previously identified as important players in the yeast hypoxic response, Cyc8, Tup1 and Ssn8; results demonstrate that only Ssn8 binds to Ixr1. We have also looked for other Ixr1-binding proteins associated with transcriptional regulation, by co-purification and mass spectrometry identification. Tdh3, a protein involved in transcriptional silencing, is among the new identified Ixr1-binding proteins. Differential phosphorylation of Ixr1 is found when comparing aerobic and hypoxic yeast growth. Implication of these results in transcriptional regulation mediated by Ixr1 is discussed.


Assuntos
Proteínas Fúngicas/metabolismo , Regulação Fúngica da Expressão Gênica , Proteínas HMGB/metabolismo , Transcrição Gênica , Sequência de Aminoácidos , Proteínas Fúngicas/química , Ordem dos Genes , Vetores Genéticos/genética , Proteínas HMGB/química , Hipóxia , Regiões Promotoras Genéticas , Ligação Proteica , Domínios e Motivos de Interação entre Proteínas , Processamento de Proteína Pós-Traducional , Ativação Transcricional
10.
J Mol Biol ; 430(5): 655-667, 2018 03 02.
Artigo em Inglês | MEDLINE | ID: mdl-29307468

RESUMO

Architectural DNA-binding proteins function to regulate diverse DNA reactions and have the defining property of significantly changing DNA conformation. Although the 1D movement along DNA by other types of DNA-binding proteins has been visualized, the mobility of architectural DNA-binding proteins on DNA remains unknown. Here, we applied single-molecule fluorescence imaging on arrays of extended DNA molecules to probe the binding dynamics of three structurally distinct architectural DNA-binding proteins: Nhp6A, HU, and Fis. Each of these proteins was observed to move along DNA, and the salt concentration independence of the 1D diffusion implies sliding with continuous contact to DNA. Nhp6A and HU exhibit a single sliding mode, whereas Fis exhibits two sliding modes. Based on comparison of the diffusion coefficients and sizes of many DNA binding proteins, the architectural proteins are categorized into a new group distinguished by an unusually high free-energy barrier for 1D diffusion. The higher free-energy barrier for 1D diffusion by architectural proteins can be attributed to the large DNA conformational changes that accompany binding and impede rotation-coupled movement along the DNA grooves.


Assuntos
Proteínas de Ligação a DNA/química , DNA/química , Proteínas de Bactérias/química , Proteínas de Bactérias/metabolismo , DNA/metabolismo , Proteínas de Ligação a DNA/metabolismo , Difusão , Entropia , Proteínas HMGB/química , Modelos Moleculares , Conformação de Ácido Nucleico , Ligação Proteica
11.
Cell Mol Life Sci ; 74(17): 3149-3162, 2017 09.
Artigo em Inglês | MEDLINE | ID: mdl-28597298

RESUMO

For many proteins, biological function requires the folding of the polypeptide chain into a unique and persistent tertiary structure. This review concerns proteins that adopt a specific tertiary structure to function, but are otherwise partially or completely disordered. The biological cue for protein folding is environmental perturbation or minor post-translational modification. Hence, we term these proteins conditionally disordered. Many of these proteins recognize and bind other molecules, and conditional disorder has been hypothesized to allow for more nuanced control and regulation of binding processes. However, this remains largely unproven. The sequences of conditionally disordered proteins suggest their propensity to fold; yet, under the standard laboratory conditions, they do not do so, which may appear surprising. We argue that the surprise results from the failure to consider the role of the environment in protein structure formation and that conditional disorder arises as a natural consequence of the marginal stability of the folded state.


Assuntos
Proteínas Intrinsicamente Desordenadas/metabolismo , Bactérias/metabolismo , Proteínas de Bactérias/química , Proteínas de Bactérias/metabolismo , Fator de Iniciação 4E em Eucariotos/química , Fator de Iniciação 4E em Eucariotos/metabolismo , Proteínas HMGB/química , Proteínas HMGB/metabolismo , Humanos , Proteínas Intrinsicamente Desordenadas/química , Ligação Proteica , Dobramento de Proteína , Estrutura Terciária de Proteína , Receptores de Glucocorticoides/química , Receptores de Glucocorticoides/metabolismo , Ribonuclease P/química , Ribonuclease P/metabolismo , Fatores de Transcrição/química , Fatores de Transcrição/metabolismo
12.
Int J Parasitol ; 46(4): 253-62, 2016 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-26820302

RESUMO

The Schistosoma mansoni High Mobility Group Box (HMGB) proteins SmHMGB1, SmHMGB2 and SmHMGB3 share highly conserved HMG box DNA binding domains but have significantly different C-terminal acidic tails. Here, we used three full-length and tailless forms of the S. mansoni HMGB proteins to examine the functional roles of their acidic tails. DNA binding assays revealed that the different lengths of the acidic tails among the three SmHMGB proteins significantly and distinctively influenced their DNA transactions. Spectroscopic analyses indicated that the longest acidic tail of SmHMGB3 contributes to the structural stabilisation of this protein. Using immunohistochemical analysis, we showed distinct patterns of SmHMGB1, SmHMGB2 and SmHMGB3 expression in different tissues of adult worms. RNA interference approaches indicated a role for SmHMGB2 and SmHMGB3 in the reproductive system of female worms, whereas for SmHMGB1 no clear phenotype was observed. Schistosome HMGB proteins can be phosphorylated, acetylated and methylated. Importantly, the acetylation and methylation of schistosome HMGBs were greatly enhanced upon removal of the acidic tail. These data support the notion that the C-terminal acidic tails dictate the differences in the structure, expression and function of schistosome HMGB proteins.


Assuntos
Proteínas HMGB/química , Schistosoma mansoni/química , Sequência de Aminoácidos , Animais , Dicroísmo Circular , DNA de Helmintos/metabolismo , Feminino , Proteínas HMGB/genética , Proteínas HMGB/fisiologia , Imuno-Histoquímica , Masculino , Especificidade de Órgãos , Domínios e Motivos de Interação entre Proteínas/fisiologia , Processamento de Proteína Pós-Traducional/fisiologia , Estrutura Secundária de Proteína , Estrutura Terciária de Proteína , Interferência de RNA , Schistosoma mansoni/genética
13.
Proc Natl Acad Sci U S A ; 112(28): 8590-5, 2015 Jul 14.
Artigo em Inglês | MEDLINE | ID: mdl-26124095

RESUMO

Extracellular signal-regulated kinase (ERK) coordinates cellular responses to a range of stimuli by phosphorylating its numerous substrates. One of these substrates, Capicua (Cic), is a transcriptional repressor that was first identified in Drosophila and has been implicated in a number of human diseases. Here we use a chemical biology approach to map the binding interface of ERK and Cic. The noncanonical amino acid p-azidophenylalanine (AzF) was introduced into the ERK-binding region of Drosophila Cic, and photocrosslinking and tandem mass spectrometry were used to pinpoint its binding site on ERK. We also identified the ERK-binding region of human Cic and showed that it binds to the same site on ERK despite lacking conservation with the Drosophila Cic binding region. Finally, we mapped the amino acids involved in human Cic binding to ERK using AzF-labeled ERK. These results reveal the molecular details of the ERK-Cic interaction and demonstrate that the photocrosslinking approach is complementary to existing methods for mapping kinase-substrate binding interfaces.


Assuntos
Proteínas de Drosophila/metabolismo , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Proteínas HMGB/metabolismo , Proteínas Repressoras/metabolismo , Sequência de Aminoácidos , Animais , Sítios de Ligação , Drosophila , Proteínas de Drosophila/química , Proteínas HMGB/química , Humanos , Espectrometria de Massas , Dados de Sequência Molecular , Processos Fotoquímicos , Proteínas Repressoras/química
14.
Structure ; 22(9): 1274-1286, 2014 Sep 02.
Artigo em Inglês | MEDLINE | ID: mdl-25126959

RESUMO

In pluripotent cells, OCT4 associates with SOX2 to maintain pluripotency or with SOX17 to induce primitive endoderm commitment. The OCT4-SOX2 and OCT4-SOX17 combinations bind mutually exclusive to two distinct composite DNA elements, known as the "canonical" and "compressed" motifs, respectively. The structural basis for the OCT4-SOX17 cooperativity is unknown. Whereas SOX17 has been engineered to replace SOX2 in the pluripotency circuitry, all generated SOX2 mutants have failed to act like SOX17. From molecular simulations, we revealed the OCT4-SOX17 interaction interface and elucidated the SOX-dependent motif preference of OCT4. Moreover, we designed a SOX2 mutant that we predicted and confirmed experimentally to bind cooperatively with OCT4 to the compressed motif. Ultimately, we found a strong correlation between the experimental and calculated relative cooperative-binding free energies of 12 OCT4-SOX-DNA complexes. Therefore, we validated the OCT4-SOX interfaces and demonstrated that in silico design of DNA-binding cooperativity is suitable for altering transcriptional circuitries.


Assuntos
Proteínas HMGB/química , Fator 3 de Transcrição de Octâmero/química , Fatores de Transcrição SOXB1/química , Fatores de Transcrição SOXF/química , Células-Tronco/fisiologia , Sequência de Aminoácidos , Animais , Sequência de Bases , Diferenciação Celular , Sequência Consenso , DNA/química , Proteínas HMGB/genética , Interações Hidrofóbicas e Hidrofílicas , Camundongos , Simulação de Dinâmica Molecular , Dados de Sequência Molecular , Fator 3 de Transcrição de Octâmero/genética , Ligação Proteica , Domínios e Motivos de Interação entre Proteínas , Fatores de Transcrição SOXB1/genética , Fatores de Transcrição SOXF/genética , Termodinâmica
15.
Nucleic Acids Res ; 41(22): 10135-49, 2013 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-24021628

RESUMO

Ribosome biogenesis is a major metabolic effort for growing cells. In Saccharomyces cerevisiae, Hmo1, an abundant high-mobility group box protein (HMGB) binds to the coding region of the RNA polymerase I transcribed ribosomal RNAs genes and the promoters of ∼70% of ribosomal protein genes. In this study, we have demonstrated the functional conservation of eukaryotic HMGB proteins involved in ribosomal DNA (rDNA) transcription. We have shown that when expressed in budding yeast, human UBF1 and a newly identified Sp-Hmo1 (Schizosaccharomyces pombe) localize to the nucleolus and suppress growth defect of the RNA polymerase I mutant rpa49-Δ. Owing to the multiple functions of both proteins, Hmo1 and UBF1 are not fully interchangeable. By deletion and domains swapping in Hmo1, we identified essential domains that stimulate rDNA transcription but are not fully required for stimulation of ribosomal protein genes expression. Hmo1 is organized in four functional domains: a dimerization module, a canonical HMGB motif followed by a conserved domain and a C-terminal nucleolar localization signal. We propose that Hmo1 has acquired species-specific functions and shares with UBF1 and Sp-Hmo1 an ancestral function to stimulate rDNA transcription.


Assuntos
DNA Ribossômico/metabolismo , Proteínas HMGB/química , Proteínas HMGB/metabolismo , Proteínas de Grupo de Alta Mobilidade/química , Proteínas de Grupo de Alta Mobilidade/metabolismo , Proteínas de Saccharomyces cerevisiae/química , Proteínas de Saccharomyces cerevisiae/metabolismo , Transcrição Gênica , Motivos de Aminoácidos , Sequência de Aminoácidos , Nucléolo Celular/metabolismo , Sequência Conservada , Proteínas HMGB/genética , Proteínas de Grupo de Alta Mobilidade/genética , Humanos , Proteínas Pol1 do Complexo de Iniciação de Transcrição/química , Proteínas Pol1 do Complexo de Iniciação de Transcrição/metabolismo , Estrutura Terciária de Proteína , RNA Polimerase I/metabolismo , Proteínas Ribossômicas/genética , Saccharomyces cerevisiae/genética , Saccharomyces cerevisiae/metabolismo , Proteínas de Saccharomyces cerevisiae/genética , Proteínas de Schizosaccharomyces pombe/metabolismo , Relação Estrutura-Atividade
16.
Nucleic Acids Res ; 41(1): 167-81, 2013 Jan 07.
Artigo em Inglês | MEDLINE | ID: mdl-23143110

RESUMO

Eukaryotic High-Mobility Group B (HMGB) proteins alter DNA elasticity while facilitating transcription, replication and DNA repair. We developed a new single-molecule method to probe non-specific DNA interactions for two HMGB homologs: the human HMGB2 box A domain and yeast Nhp6Ap, along with chimeric mutants replacing neutral N-terminal residues of the HMGB2 protein with cationic sequences from Nhp6Ap. Surprisingly, HMGB proteins constrain DNA winding, and this torsional constraint is released over short timescales. These measurements reveal the microscopic dissociation rates of HMGB from DNA. Separate microscopic and macroscopic (or local and non-local) unbinding rates have been previously proposed, but never independently observed. Microscopic dissociation rates for the chimeric mutants (~10 s(-1)) are higher than those observed for wild-type proteins (~0.1-1.0 s(-1)), reflecting their reduced ability to bend DNA through short-range interactions, despite their increased DNA-binding affinity. Therefore, transient local HMGB-DNA contacts dominate the DNA-bending mechanism used by these important architectural proteins to increase DNA flexibility.


Assuntos
DNA/química , Proteínas HMGB/química , Sequência de Aminoácidos , Pareamento de Bases , DNA/metabolismo , DNA de Forma B/química , Elasticidade , Domínios HMG-Box , Proteínas HMGB/metabolismo , Proteína HMGB2/química , Proteína HMGB2/metabolismo , Proteínas HMGN/metabolismo , Humanos , Cinética , Dados de Sequência Molecular , Ligação Proteica , Proteínas de Saccharomyces cerevisiae/metabolismo
17.
PLoS One ; 7(11): e50789, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-23209826

RESUMO

BACKGROUND: Crassostrea ariakensis Gould is a representative bivalve species and an economically important oyster in China, but suffers severe mortalities in recent years that are caused by rickettsia-like organism (RLO). Prevention and control of this disease is a priority for the development of oyster aquaculture. It has been proven that mammalian HMGB (high mobility group box) can be released extracellularly and acts as an important pro-inflammatory cytokine and late mediator of inflammatory reactions. In vertebrates, HMGB's antibody (anti-HMGB) has been shown to confer significant protection against certain local and systemic inflammatory diseases. Therefore, we investigated the functions of Ca-HMGB (oyster HMGB) and anti-CaHMGB (Ca-HMGB's antibody) in oyster RLO/LPS (RLO or LPS)-induced disease or inflammation. METHODOLOGY/PRINCIPAL FINDINGS: Sequencing analysis revealed Ca-HMGB shares conserved structures with mammalians. Tissue-specific expression indicates that Ca-HMGB has higher relative expression in hemocytes. Significant continuous up-regulation of Ca-HMGB was detected when the hemocytes were stimulated with RLO/LPS. Recombinant Ca-HMGB protein significantly up-regulated the expression levels of some cytokines. Indirect immunofluorescence study revealed that Ca-HMGB localized both in the hemocyte nucleus and cytoplasm before RLO challenge, but mainly in the cytoplasm 12 h after challenge. Western blot analysis demonstrated Ca-HMGB was released extracellularly 4-12 h after RLO challenge. Anti-CaHMGB was added to the RLO/LPS-challenged hemocyte monolayer and real-time RT-PCR showed that administration of anti-CaHMGB dramatically reduced the rate of RLO/LPS-induced up-regulation of LITAF at 4-12 h after treatment. Flow cytometry analysis indicated that administration of anti-CaHMGB reduced RLO/LPS-induced hemocyte apoptosis and necrosis rates. CONCLUSIONS/SIGNIFICANCE: Ca-HMGB can be released extracellularly and its subcellular localization varies when stimulated with RLO. Ca-HMGB is involved in oyster immune reactions and functions as a pro-inflammatory cytokine. Anti-CaHMGB can significantly suppress RLO/LPS-induced inflammatory responses and hemocyte necrosis and apoptosis, suggesting that Ca-HMGB is a potential target to prevent and control RLO/LPS-induced disease or inflammation.


Assuntos
Anticorpos/imunologia , Crassostrea/imunologia , Crassostrea/metabolismo , Proteínas HMGB/química , Proteínas HMGB/metabolismo , Animais , Western Blotting , Citocinas/metabolismo , Citometria de Fluxo , Proteínas HMGB/genética , Proteínas HMGB/imunologia , Filogenia , Reação em Cadeia da Polimerase em Tempo Real
18.
Biochemistry ; 51(34): 6728-37, 2012 Aug 28.
Artigo em Inglês | MEDLINE | ID: mdl-22901013

RESUMO

Proteins in the HMG family are important transcription factors. They recognize cisplatin-damaged DNA lesions with a structure-specific preference and account for more than 70% of all proteins that interact with the cisplatin 1,2-intrastrand d(GpG) cross-link. HMGB4, a new member of the mammalian HMGB protein family expressed preferentially in the testis, was generated recombinantly, and its interactions with cisplatin-modified DNA were investigated in vitro. The binding affinities of the two individual DNA-binding domains of HMGB4 to DNA carrying a cisplatin 1,2-intrastrand d(GpG) cross-link are weaker than those of the DNA-binding domains of HMGB1. Full-length HMGB4, however, has a 28-fold stronger binding affinity (K(d) = 4.35 nM) for the platinated adduct compared to that of HMGB1 (K(d) = 120 nM), presumably because the former lacks a C-terminal acidic tail. The residue Phe37 plays a critical role in stabilizing the binding complex of HMGB4 with the cisplatin-modified DNA, as it does for HMGB1. Hydroxyl radical footprinting analysis of the HMGB4/platinated DNA complex reveals a footprinting pattern very different from that of HMGB1, however, revealing very little binding asymmetry with respect to the platinated lesion. An in vitro repair assay revealed that HMGB4, at 1 µM, interferes with repair of cisplatin 1,2-intrastrand cross-link damage by >90% compared to control, whereas HMGB1 at the same concentration inhibits repair by 45%. This repair inhibition capability is highly dependent on both the binding affinity and the size of the proteins. The putative role of HMGB4 in the mechanism of action of cisplatin, and especially its potential relevance to the hypersensitivity of testicular germ cell tumors to cisplatin, are discussed.


Assuntos
Cisplatino/química , DNA/metabolismo , Proteínas HMGB/metabolismo , Motivos de Aminoácidos , Animais , Sequência de Bases , Cisplatino/metabolismo , DNA/química , Adutos de DNA/química , Adutos de DNA/metabolismo , Reparo do DNA , Proteínas HMGB/química , Proteínas HMGB/genética , Cinética , Camundongos , Dados de Sequência Molecular , Ligação Proteica
19.
Biochem J ; 443(1): 39-47, 2012 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-22181698

RESUMO

It has recently been proposed that the sequence preferences of DNA-binding TFs (transcription factors) can be well described by models that include the positional interdependence of the nucleotides of the target sites. Such binding models allow for multiple motifs to be invoked, such as principal and secondary motifs differing at two or more nucleotide positions. However, the structural mechanisms underlying the accommodation of such variant motifs by TFs remain elusive. In the present study we examine the crystal structure of the HMG (high-mobility group) domain of Sox4 [Sry (sex-determining region on the Y chromosome)-related HMG box 4] bound to DNA. By comparing this structure with previously solved structures of Sox17 and Sox2, we observed subtle conformational differences at the DNA-binding interface. Furthermore, using quantitative electrophoretic mobility-shift assays we validated the positional interdependence of two nucleotides and the presence of a secondary Sox motif in the affinity landscape of Sox4. These results suggest that a concerted rearrangement of two interface amino acids enables Sox4 to accommodate primary and secondary motifs. The structural adaptations lead to altered dinucleotide preferences that mutually reinforce each other. These analyses underline the complexity of the DNA recognition by TFs and provide an experimental validation for the conceptual framework of positional interdependence and secondary binding motifs.


Assuntos
DNA/química , Substâncias Macromoleculares/química , Fatores de Transcrição SOXC/química , Motivos de Aminoácidos , Sequência de Aminoácidos , Animais , Sítios de Ligação , Sequência Conservada , Cristalografia por Raios X , Ensaio de Desvio de Mobilidade Eletroforética , Elementos Facilitadores Genéticos , Proteínas HMGB/química , Laminina/genética , Camundongos , Modelos Moleculares , Dados de Sequência Molecular , Conformação de Ácido Nucleico , Ligação Proteica , Estrutura Terciária de Proteína , Fatores de Transcrição SOXF/química
20.
Annu Rev Genet ; 45: 167-202, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-21854230

RESUMO

V(D)J recombination assembles immunoglobulin and T cell receptor genes during lymphocyte development through a series of carefully orchestrated DNA breakage and rejoining events. DNA cleavage requires a series of protein-DNA complexes containing the RAG1 and RAG2 proteins and recombination signals that flank the recombining gene segments. In this review, we discuss recent advances in our understanding of the function and domain organization of the RAG proteins, the composition and structure of RAG-DNA complexes, and the pathways that lead to the formation of these complexes. We also consider the functional significance of RAG-mediated histone recognition and ubiquitin ligase activities, and the role played by RAG in ensuring proper repair of DNA breaks made during V(D)J recombination. Finally, we propose a model for the formation of RAG-DNA complexes that involves anchoring of RAG1 at the recombination signal nonamer and RAG2-dependent surveillance of adjoining DNA for suitable spacer and heptamer sequences.


Assuntos
Proteínas de Ligação a DNA/química , Proteínas de Homeodomínio/química , Recombinação V(D)J , Animais , Clivagem do DNA , Fragmentação do DNA , Reparo do DNA , Proteínas de Ligação a DNA/isolamento & purificação , Proteínas HMGB/química , Proteínas de Homeodomínio/isolamento & purificação , Camundongos , Complexos Multiproteicos/química , Ligação Proteica , Conformação Proteica , Transdução de Sinais , Ubiquitina-Proteína Ligases/química , Ubiquitinação
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...