Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 93
Filtrar
Mais filtros

Bases de dados
Tipo de documento
Intervalo de ano de publicação
1.
J Biol Chem ; 299(12): 105382, 2023 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-37866628

RESUMO

Proteomic studies have identified moesin (MSN), a protein containing a four-point-one, ezrin, radixin, moesin (FERM) domain, and the receptor CD44 as hub proteins found within a coexpression module strongly linked to Alzheimer's disease (AD) traits and microglia. These proteins are more abundant in Alzheimer's patient brains, and their levels are positively correlated with cognitive decline, amyloid plaque deposition, and neurofibrillary tangle burden. The MSN FERM domain interacts with the phospholipid phosphatidylinositol 4,5-bisphosphate (PIP2) and the cytoplasmic tail of CD44. Inhibiting the MSN-CD44 interaction may help limit AD-associated neuronal damage. Here, we investigated the feasibility of developing inhibitors that target this protein-protein interaction. We have employed structural, mutational, and phage-display studies to examine how CD44 binds to the FERM domain of MSN. Interestingly, we have identified an allosteric site located close to the PIP2 binding pocket that influences CD44 binding. These findings suggest a mechanism in which PIP2 binding to the FERM domain stimulates CD44 binding through an allosteric effect, leading to the formation of a neighboring pocket capable of accommodating a receptor tail. Furthermore, high-throughput screening of a chemical library identified two compounds that disrupt the MSN-CD44 interaction. One compound series was further optimized for biochemical activity, specificity, and solubility. Our results suggest that the FERM domain holds potential as a drug development target. Small molecule preliminary leads generated from this study could serve as a foundation for additional medicinal chemistry efforts with the goal of controlling microglial activity in AD by modifying the MSN-CD44 interaction.


Assuntos
Doença de Alzheimer , Ligação Proteica , Humanos , Doença de Alzheimer/tratamento farmacológico , Doença de Alzheimer/metabolismo , Doença de Alzheimer/patologia , Domínios FERM , Receptores de Hialuronatos/metabolismo , Ligação Proteica/efeitos dos fármacos , Proteômica
2.
Mol Cell ; 64(4): 704-719, 2016 11 17.
Artigo em Inglês | MEDLINE | ID: mdl-27871366

RESUMO

The cytotoxicity of DNA-protein crosslinks (DPCs) is largely ascribed to their ability to block the progression of DNA replication. DPCs frequently occur in cells, either as a consequence of metabolism or exogenous agents, but the mechanism of DPC repair is not completely understood. Here, we characterize SPRTN as a specialized DNA-dependent and DNA replication-coupled metalloprotease for DPC repair. SPRTN cleaves various DNA binding substrates during S-phase progression and thus protects proliferative cells from DPC toxicity. Ruijs-Aalfs syndrome (RJALS) patient cells with monogenic and biallelic mutations in SPRTN are hypersensitive to DPC-inducing agents due to a defect in DNA replication fork progression and the inability to eliminate DPCs. We propose that SPRTN protease represents a specialized DNA replication-coupled DPC repair pathway essential for DNA replication progression and genome stability. Defective SPRTN-dependent clearance of DPCs is the molecular mechanism underlying RJALS, and DPCs are contributing to accelerated aging and cancer.


Assuntos
Reparo do DNA , Replicação do DNA , Proteínas de Ligação a DNA/metabolismo , DNA/química , Instabilidade Genômica , Sequência de Aminoácidos , Sítios de Ligação , Reagentes de Ligações Cruzadas/química , DNA/genética , DNA/metabolismo , Dano ao DNA , Proteínas de Ligação a DNA/genética , Etoposídeo/química , Formaldeído/química , Expressão Gênica , Humanos , Cinética , Mutação , Ligação Proteica , Alinhamento de Sequência , Homologia de Sequência de Aminoácidos , Especificidade por Substrato , Síndrome , Raios Ultravioleta
3.
Nucleic Acids Res ; 50(3): 1484-1500, 2022 02 22.
Artigo em Inglês | MEDLINE | ID: mdl-35037045

RESUMO

The SARS-CoV-2 coronavirus is the causal agent of the current global pandemic. SARS-CoV-2 belongs to an order, Nidovirales, with very large RNA genomes. It is proposed that the fidelity of coronavirus (CoV) genome replication is aided by an RNA nuclease complex, comprising the non-structural proteins 14 and 10 (nsp14-nsp10), an attractive target for antiviral inhibition. Our results validate reports that the SARS-CoV-2 nsp14-nsp10 complex has RNase activity. Detailed functional characterization reveals nsp14-nsp10 is a versatile nuclease capable of digesting a wide variety of RNA structures, including those with a blocked 3'-terminus. Consistent with a role in maintaining viral genome integrity during replication, we find that nsp14-nsp10 activity is enhanced by the viral RNA-dependent RNA polymerase complex (RdRp) consisting of nsp12-nsp7-nsp8 (nsp12-7-8) and demonstrate that this stimulation is mediated by nsp8. We propose that the role of nsp14-nsp10 in maintaining replication fidelity goes beyond classical proofreading by purging the nascent replicating RNA strand of a range of potentially replication-terminating aberrations. Using our developed assays, we identify drug and drug-like molecules that inhibit nsp14-nsp10, including the known SARS-CoV-2 major protease (Mpro) inhibitor ebselen and the HIV integrase inhibitor raltegravir, revealing the potential for multifunctional inhibitors in COVID-19 treatment.


Assuntos
Antivirais/farmacologia , Avaliação Pré-Clínica de Medicamentos , Exorribonucleases/metabolismo , Genoma Viral/genética , Instabilidade Genômica , SARS-CoV-2/enzimologia , SARS-CoV-2/genética , Proteínas não Estruturais Virais/metabolismo , Proteínas Virais Reguladoras e Acessórias/metabolismo , RNA-Polimerase RNA-Dependente de Coronavírus/metabolismo , Exorribonucleases/antagonistas & inibidores , Genoma Viral/efeitos dos fármacos , Instabilidade Genômica/efeitos dos fármacos , Instabilidade Genômica/genética , Inibidores de Integrase de HIV/farmacologia , Isoindóis/farmacologia , Complexos Multienzimáticos/antagonistas & inibidores , Complexos Multienzimáticos/metabolismo , Compostos Organosselênicos/farmacologia , RNA Viral/biossíntese , RNA Viral/genética , Raltegravir Potássico/farmacologia , SARS-CoV-2/efeitos dos fármacos , Proteínas não Estruturais Virais/antagonistas & inibidores , Proteínas Virais Reguladoras e Acessórias/antagonistas & inibidores , Replicação Viral/efeitos dos fármacos , Replicação Viral/genética
4.
Nucleic Acids Res ; 49(16): 9294-9309, 2021 09 20.
Artigo em Inglês | MEDLINE | ID: mdl-34387694

RESUMO

The SNM1 nucleases which help maintain genome integrity are members of the metallo-ß-lactamase (MBL) structural superfamily. Their conserved MBL-ß-CASP-fold SNM1 core provides a molecular scaffold forming an active site which coordinates the metal ions required for catalysis. The features that determine SNM1 endo- versus exonuclease activity, and which control substrate selectivity and binding are poorly understood. We describe a structure of SNM1B/Apollo with two nucleotides bound to its active site, resembling the product state of its exonuclease reaction. The structure enables definition of key SNM1B residues that form contacts with DNA and identifies a 5' phosphate binding pocket, which we demonstrate is important in catalysis and which has a key role in determining endo- versus exonucleolytic activity across the SNM1 family. We probed the capacity of SNM1B to digest past sites of common endogenous DNA lesions and find that base modifications planar to the nucleobase can be accommodated due to the open architecture of the active site, but lesions axial to the plane of the nucleobase are not well tolerated due to constriction around the altered base. We propose that SNM1B/Apollo might employ its activity to help remove common oxidative lesions from telomeres.


Assuntos
Endonucleases/química , Exodesoxirribonucleases/química , Exonucleases/química , beta-Lactamases/genética , Sítios de Ligação/genética , Catálise , Domínio Catalítico/genética , Proteínas de Ligação a DNA , Endonucleases/genética , Exodesoxirribonucleases/genética , Exodesoxirribonucleases/ultraestrutura , Exonucleases/genética , Humanos , Metais , Fosfatos/química , beta-Lactamases/química
5.
Nucleic Acids Res ; 49(16): 9310-9326, 2021 09 20.
Artigo em Inglês | MEDLINE | ID: mdl-34387696

RESUMO

Artemis (SNM1C/DCLRE1C) is an endonuclease that plays a key role in development of B- and T-lymphocytes and in dsDNA break repair by non-homologous end-joining (NHEJ). Artemis is phosphorylated by DNA-PKcs and acts to open DNA hairpin intermediates generated during V(D)J and class-switch recombination. Artemis deficiency leads to congenital radiosensitive severe acquired immune deficiency (RS-SCID). Artemis belongs to a superfamily of nucleases containing metallo-ß-lactamase (MBL) and ß-CASP (CPSF-Artemis-SNM1-Pso2) domains. We present crystal structures of the catalytic domain of wildtype and variant forms of Artemis, including one causing RS-SCID Omenn syndrome. The catalytic domain of the Artemis has similar endonuclease activity to the phosphorylated full-length protein. Our structures help explain the predominantly endonucleolytic activity of Artemis, which contrasts with the predominantly exonuclease activity of the closely related SNM1A and SNM1B MBL fold nucleases. The structures reveal a second metal binding site in its ß-CASP domain unique to Artemis, which is amenable to inhibition by compounds including ebselen. By combining our structural data with that from a recently reported Artemis structure, we were able model the interaction of Artemis with DNA substrates. The structures, including one of Artemis with the cephalosporin ceftriaxone, will help enable the rational development of selective SNM1 nuclease inhibitors.


Assuntos
Proteínas de Ciclo Celular/ultraestrutura , Proteínas de Ligação a DNA/ultraestrutura , Endonucleases/ultraestrutura , Exodesoxirribonucleases/ultraestrutura , Imunodeficiência Combinada Severa/genética , Linfócitos B/enzimologia , Domínio Catalítico/genética , Proteínas de Ciclo Celular/química , Proteínas de Ciclo Celular/genética , Cristalografia por Raios X , Reparo do DNA por Junção de Extremidades/genética , Reparo do DNA/genética , Proteínas de Ligação a DNA/antagonistas & inibidores , Proteínas de Ligação a DNA/química , Proteínas de Ligação a DNA/genética , Endonucleases/antagonistas & inibidores , Endonucleases/química , Endonucleases/genética , Inibidores Enzimáticos/química , Exodesoxirribonucleases/química , Exodesoxirribonucleases/genética , Humanos , Fosforilação/genética , Dobramento de Proteína , Imunodeficiência Combinada Severa/enzimologia , Imunodeficiência Combinada Severa/patologia , Linfócitos T/enzimologia
6.
J Biol Chem ; 295(10): 2948-2958, 2020 03 06.
Artigo em Inglês | MEDLINE | ID: mdl-31914405

RESUMO

Forkhead box N1 (FOXN1) is a member of the forkhead box family of transcription factors and plays an important role in thymic epithelial cell differentiation and development. FOXN1 mutations in humans and mice give rise to the "nude" phenotype, which is marked by athymia. FOXN1 belongs to a subset of the FOX family that recognizes an alternative forkhead-like (FHL) consensus sequence (GACGC) that is different from the more widely recognized forkhead (FKH) sequence RYAAAYA (where R is purine, and Y is pyrimidine). Here, we present the FOXN1 structure in complex with DNA containing an FHL motif at 1.6 Å resolution, in which the DNA sequence is recognized by a mixture of direct and water-mediated contacts provided by residues in an α-helix inserted in the DNA major groove (the recognition helix). Comparisons with the structure of other FOX family members revealed that the FKH and FHL DNA sequences are bound in two distinct modes, with partially different registers for the protein DNA contacts. We identified a single alternative rotamer within the recognition helix itself as an important determinant of DNA specificity and found protein sequence features in the recognition helix that could be used to predict the specificity of other FOX family members. Finally, we demonstrate that the C-terminal region of FOXN1 is required for high-affinity DNA binding and that FOXN1 has a significantly reduced affinity for DNA that contains 5'-methylcytosine, which may have implications for the role of FOXN1 in thymic involution.


Assuntos
DNA/metabolismo , Fatores de Transcrição Forkhead/metabolismo , Sequência de Aminoácidos , Sequência de Bases , Sítios de Ligação , Cristalografia por Raios X , DNA/química , Metilação de DNA , Ensaio de Desvio de Mobilidade Eletroforética , Fatores de Transcrição Forkhead/química , Fatores de Transcrição Forkhead/genética , Humanos , Ligação Proteica , Conformação Proteica em alfa-Hélice , Estrutura Terciária de Proteína , Proteínas Recombinantes/biossíntese , Proteínas Recombinantes/química , Proteínas Recombinantes/isolamento & purificação , Alinhamento de Sequência
7.
Nucleic Acids Res ; 45(7): 4231-4243, 2017 04 20.
Artigo em Inglês | MEDLINE | ID: mdl-28100692

RESUMO

RecQ helicases are important maintainers of genome integrity with distinct roles in almost every cellular process requiring access to DNA. RECQL5 is one of five human RecQ proteins and is particularly versatile in this regard, forming protein complexes with a diverse set of cellular partners in order to coordinate its helicase activity to various processes including replication, recombination and DNA repair. In this study, we have determined crystal structures of the core helicase domain of RECQL5 both with and without the nucleotide ADP in two distinctly different ('Open' and 'Closed') conformations. Small angle X-ray scattering studies show that the 'Open' form of the protein predominates in solution and we discuss implications of this with regards to the RECQL5 mechanism and conformational changes. We have measured the ATPase, helicase and DNA binding properties of various RECQL5 constructs and variants and discuss the role of these regions and residues in the various RECQL5 activities. Finally, we have performed a systematic comparison of the RECQL5 structures with other RecQ family structures and based on these comparisons we have constructed a model for the mechano-chemical cycle of the common catalytic core of these helicases.


Assuntos
Modelos Moleculares , RecQ Helicases/química , Domínio Catalítico , Humanos , Mutação , Conformação Proteica em alfa-Hélice , RecQ Helicases/genética , RecQ Helicases/metabolismo , Espalhamento a Baixo Ângulo , Difração de Raios X
8.
Genes Dev ; 25(17): 1859-70, 2011 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-21896658

RESUMO

One of the major DNA interstrand cross-link (ICL) repair pathways in mammalian cells is coupled to replication, but the mechanistic roles of the critical factors involved remain largely elusive. Here, we show that purified human SNM1A (hSNM1A), which exhibits a 5'-3' exonuclease activity, can load from a single DNA nick and digest past an ICL on its substrate strand. hSNM1A-depleted cells are ICL-sensitive and accumulate replication-associated DNA double-strand breaks (DSBs), akin to ERCC1-depleted cells. These DSBs are Mus81-induced, indicating that replication fork cleavage by Mus81 results from the failure of the hSNM1A- and XPF-ERCC1-dependent ICL repair pathway. Our results reveal how collaboration between hSNM1A and XPF-ERCC1 is necessary to initiate ICL repair in replicating human cells.


Assuntos
Enzimas Reparadoras do DNA/metabolismo , Reparo do DNA/genética , Proteínas de Ligação a DNA/metabolismo , DNA/metabolismo , Endonucleases/metabolismo , Proteínas Nucleares/metabolismo , Linhagem Celular Tumoral , Quebras de DNA de Cadeia Dupla , Quebras de DNA de Cadeia Simples , Enzimas Reparadoras do DNA/genética , Proteínas de Ligação a DNA/genética , Endonucleases/genética , Exodesoxirribonucleases , Células HeLa , Humanos , Proteínas Nucleares/genética
9.
Nucleic Acids Res ; 44(4): 1813-32, 2016 Feb 29.
Artigo em Inglês | MEDLINE | ID: mdl-26826708

RESUMO

Post-transcriptional control of mitochondrial gene expression, including the processing and generation of mature transcripts as well as their degradation, is a key regulatory step in gene expression in human mitochondria. Consequently, identification of the proteins responsible for RNA processing and degradation in this organelle is of great importance. The metallo-ß-lactamase (MBL) is a candidate protein family that includes ribo- and deoxyribonucleases. In this study, we discovered a function for LACTB2, an orphan MBL protein found in mammalian mitochondria. Solving its crystal structure revealed almost perfect alignment of the MBL domain with CPSF73, as well as to other ribonucleases of the MBL superfamily. Recombinant human LACTB2 displayed robust endoribonuclease activity on ssRNA with a preference for cleavage after purine-pyrimidine sequences. Mutational analysis identified an extended RNA-binding site. Knockdown of LACTB2 in cultured cells caused a moderate but significant accumulation of many mitochondrial transcripts, and its overexpression led to the opposite effect. Furthermore, manipulation of LACTB2 expression resulted in cellular morphological deformation and cell death. Together, this study discovered that LACTB2 is an endoribonuclease that is involved in the turnover of mitochondrial RNA, and is essential for mitochondrial function in human cells.


Assuntos
Endorribonucleases/química , Metaloproteínas/química , Mitocôndrias/enzimologia , Proteínas de Ligação a RNA/química , beta-Lactamases/química , Sítios de Ligação , Cristalografia por Raios X , Endorribonucleases/genética , Endorribonucleases/isolamento & purificação , Humanos , Metaloproteínas/genética , Estrutura Terciária de Proteína , RNA/genética , RNA Mitocondrial , Proteínas de Ligação a RNA/genética , Proteínas de Ligação a RNA/isolamento & purificação , beta-Lactamases/genética , beta-Lactamases/isolamento & purificação
10.
Proc Natl Acad Sci U S A ; 112(14): 4286-91, 2015 Apr 07.
Artigo em Inglês | MEDLINE | ID: mdl-25831490

RESUMO

RecQ helicases are a widely conserved family of ATP-dependent motors with diverse roles in nearly every aspect of bacterial and eukaryotic genome maintenance. However, the physical mechanisms by which RecQ helicases recognize and process specific DNA replication and repair intermediates are largely unknown. Here, we solved crystal structures of the human RECQ1 helicase in complexes with tailed-duplex DNA and ssDNA. The structures map the interactions of the ssDNA tail and the branch point along the helicase and Zn-binding domains, which, together with reported structures of other helicases, define the catalytic stages of helicase action. We also identify a strand-separating pin, which (uniquely in RECQ1) is buttressed by the protein dimer interface. A duplex DNA-binding surface on the C-terminal domain is shown to play a role in DNA unwinding, strand annealing, and Holliday junction (HJ) branch migration. We have combined EM and analytical ultracentrifugation approaches to show that RECQ1 can form what appears to be a flat, homotetrameric complex and propose that RECQ1 tetramers are involved in HJ recognition. This tetrameric arrangement suggests a platform for coordinated activity at the advancing and receding duplexes of an HJ during branch migration.


Assuntos
DNA Helicases/química , DNA/química , RecQ Helicases/química , Animais , Cromatografia em Gel , Cristalização , Cristalografia por Raios X , DNA Cruciforme/fisiologia , DNA de Cadeia Simples/química , Escherichia coli/metabolismo , Humanos , Insetos , Conformação Molecular , Desnaturação de Ácido Nucleico , Nucleotídeos/química , Ligação Proteica , Estrutura Terciária de Proteína , Zinco/química
11.
Nucleic Acids Res ; 43(22): 11047-60, 2015 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-26582912

RESUMO

The human SNM1A and SNM1B/Apollo proteins are members of an extended family of eukaryotic nuclease containing a motif related to the prokaryotic metallo-ß-lactamase (MBL) fold. SNM1A is a key exonuclease during replication-dependent and transcription-coupled interstrand crosslink repair, while SNM1B/Apollo is required for maintaining telomeric overhangs. Here, we report the crystal structures of SNM1A and SNM1B at 2.16 Å. While both proteins contain a typical MBL-ß-CASP domain, a region of positive charge surrounds the active site of SNM1A, which is absent in SNM1B and explains the greater apparent processivity of SNM1A. The structures of both proteins also reveal a putative, wide DNA-binding groove. Extensive mutagenesis of this groove, coupled with detailed biochemical analysis, identified residues that did not impact on SNM1A catalytic activity, but drastically reduced its processivity. Moreover, we identified a key role for this groove for efficient digestion past DNA interstrand crosslinks, facilitating the key DNA repair reaction catalysed by SNM1A. Together, the architecture and dimensions of this groove, coupled to the surrounding region of high positive charge, explain the remarkable ability of SNM1A to accommodate and efficiently digest highly distorted DNA substrates, such as those containing DNA lesions.


Assuntos
Enzimas Reparadoras do DNA/química , DNA/metabolismo , Exodesoxirribonucleases/química , Proteínas Nucleares/química , Domínio Catalítico , Proteínas de Ciclo Celular , Dano ao DNA , Enzimas Reparadoras do DNA/metabolismo , Exodesoxirribonucleases/metabolismo , Humanos , Modelos Moleculares , Proteínas Nucleares/metabolismo , Ligação Proteica , Estrutura Terciária de Proteína
12.
Nucleic Acids Res ; 43(10): 5221-35, 2015 May 26.
Artigo em Inglês | MEDLINE | ID: mdl-25901030

RESUMO

Bloom's syndrome helicase (BLM) is a member of the RecQ family of DNA helicases, which play key roles in the maintenance of genome integrity in all organism groups. We describe crystal structures of the BLM helicase domain in complex with DNA and with an antibody fragment, as well as SAXS and domain association studies in solution. We show an unexpected nucleotide-dependent interaction of the core helicase domain with the conserved, poorly characterized HRDC domain. The BLM-DNA complex shows an unusual base-flipping mechanism with unique positioning of the DNA duplex relative to the helicase core domains. Comparison with other crystal structures of RecQ helicases permits the definition of structural transitions underlying ATP-driven helicase action, and the identification of a nucleotide-regulated tunnel that may play a role in interactions with complex DNA substrates.


Assuntos
RecQ Helicases/química , Difosfato de Adenosina/química , Cristalografia por Raios X , DNA/química , Modelos Moleculares , Mutação , Conformação Proteica , Estrutura Terciária de Proteína , RecQ Helicases/genética , RecQ Helicases/metabolismo , Anticorpos de Domínio Único/química , Zinco/química
13.
Nucleic Acids Res ; 43(1): 247-58, 2015 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-25505141

RESUMO

Cockayne syndrome (CS) is a premature aging disorder characterized by photosensitivity, impaired development and multisystem progressive degeneration, and consists of two strict complementation groups, A and B. Using a yeast two-hybrid approach, we identified the 5'-3' exonuclease SNM1A as one of four strong interacting partners of CSB. This direct interaction was confirmed using purified recombinant proteins-with CSB able to modulate the exonuclease activity of SNM1A on oligonucleotide substrates in vitro-and the two proteins were shown to exist in a common complex in human cell extracts. CSB and SNM1A were also found, using fluorescently tagged proteins in combination with confocal microscopy and laser microirradiation, to be recruited to localized trioxsalen-induced ICL damage in human cells, with accumulation being suppressed by transcription inhibition. Moreover, SNM1A recruitment was significantly reduced in CSB-deficient cells, suggesting coordination between the two proteins in vivo. CSB-deficient neural cells exhibited increased sensitivity to DNA crosslinking agents, particularly, in a non-cycling, differentiated state, as well as delayed ICL processing as revealed by a modified Comet assay and γ-H2AX foci persistence. The results indicate that CSB coordinates the resolution of ICLs, possibly in a transcription-associated repair mechanism involving SNM1A, and that defects in the process could contribute to the post-mitotic degenerative pathologies associated with CS.


Assuntos
DNA Helicases/metabolismo , Enzimas Reparadoras do DNA/metabolismo , Reparo do DNA , Proteínas de Ligação a DNA/metabolismo , Proteínas de Ciclo Celular , Linhagem Celular , Reagentes de Ligações Cruzadas , DNA/metabolismo , Dano ao DNA , Exodesoxirribonucleases , Exonucleases/metabolismo , Células HeLa , Humanos , Proteínas de Ligação a Poli-ADP-Ribose
14.
J Biol Chem ; 290(22): 13692-709, 2015 May 29.
Artigo em Inglês | MEDLINE | ID: mdl-25866208

RESUMO

Ets transcription factors, which share the conserved Ets DNA-binding domain, number nearly 30 members in humans and are particularly involved in developmental processes. Their deregulation following changes in expression, transcriptional activity, or by chromosomal translocation plays a critical role in carcinogenesis. Ets DNA binding, selectivity, and regulation have been extensively studied; however, questions still arise regarding binding specificity outside the core GGA recognition sequence and the mode of action of Ets post-translational modifications. Here, we report the crystal structures of Etv1, Etv4, Etv5, and Fev, alone and in complex with DNA. We identify previously unrecognized features of the protein-DNA interface. Interactions with the DNA backbone account for most of the binding affinity. We describe a highly coordinated network of water molecules acting in base selection upstream of the GGAA core and the structural features that may account for discrimination against methylated cytidine residues. Unexpectedly, all proteins crystallized as disulfide-linked dimers, exhibiting a novel interface (distant to the DNA recognition helix). Homodimers of Etv1, Etv4, and Etv5 could be reduced to monomers, leading to a 40-200-fold increase in DNA binding affinity. Hence, we present the first indication of a redox-dependent regulatory mechanism that may control the activity of this subset of oncogenic Ets transcription factors.


Assuntos
Proteínas E1A de Adenovirus/química , Proteínas de Ligação a DNA/química , Dissulfetos/química , Proteínas Nucleares/química , Proteínas Proto-Oncogênicas c-ets/química , Proteínas Proto-Oncogênicas/química , Fatores de Transcrição/química , Sequência de Aminoácidos , Cromatografia Líquida , DNA/química , Regulação da Expressão Gênica , Humanos , Modelos Moleculares , Dados de Sequência Molecular , Mutagênese Sítio-Dirigida , Neoplasias/metabolismo , Oxirredução , Oxigênio/química , Ligação Proteica , Conformação Proteica , Multimerização Proteica , Proteínas Recombinantes/química , Homologia de Sequência de Aminoácidos , Espectrometria de Massas por Ionização por Electrospray
15.
J Biol Chem ; 290(13): 8539-49, 2015 Mar 27.
Artigo em Inglês | MEDLINE | ID: mdl-25670864

RESUMO

Ets-2, like its closely related homologue Ets-1, is a member of the Ets family of DNA binding transcription factors. Both proteins are subject to multiple levels of regulation of their DNA binding and transactivation properties. One such regulatory mechanism is the presence of an autoinhibitory module, which in Ets-1 allosterically inhibits the DNA binding activity. This inhibition can be relieved by interaction with protein partners or cooperative binding to closely separated Ets binding sites in a palindromic arrangement. In this study we describe the 2.5 Å resolution crystal structure of a DNA complex of the Ets-2 Ets domain. The Ets domain crystallized with two distinct species in the asymmetric unit, which closely resemble the autoinhibited and DNA bound forms of Ets-1. This discovery prompted us to re-evaluate the current model for the autoinhibitory mechanism and the structural basis for cooperative DNA binding. In contrast to Ets-1, in which the autoinhibition is caused by a combination of allosteric and steric mechanisms, we were unable to find clear evidence for the allosteric mechanism in Ets-2. We also demonstrated two possibly distinct types of cooperative binding to substrates with Ets binding motifs separated by four and six base pairs and suggest possible molecular mechanisms for this behavior.


Assuntos
Proteína Proto-Oncogênica c-ets-2/química , Regulação Alostérica , Cristalografia por Raios X , DNA/química , Humanos , Modelos Moleculares , Ligação Proteica , Estrutura Secundária de Proteína , Estrutura Terciária de Proteína
16.
Antimicrob Agents Chemother ; 60(1): 142-50, 2016 01.
Artigo em Inglês | MEDLINE | ID: mdl-26482303

RESUMO

ß-Lactams are the most successful antibacterials, but their effectiveness is threatened by resistance, most importantly by production of serine- and metallo-ß-lactamases (MBLs). MBLs are of increasing concern because they catalyze the hydrolysis of almost all ß-lactam antibiotics, including recent-generation carbapenems. Clinically useful serine-ß-lactamase inhibitors have been developed, but such inhibitors are not available for MBLs. l-Captopril, which is used to treat hypertension via angiotensin-converting enzyme inhibition, has been reported to inhibit MBLs by chelating the active site zinc ions via its thiol(ate). We report systematic studies on B1 MBL inhibition by all four captopril stereoisomers. High-resolution crystal structures of three MBLs (IMP-1, BcII, and VIM-2) in complex with either the l- or d-captopril stereoisomer reveal correlations between the binding mode and inhibition potency. The results will be useful in the design of MBL inhibitors with the breadth of selectivity required for clinical application against carbapenem-resistant Enterobacteriaceae and other organisms causing MBL-mediated resistant infections.


Assuntos
Inibidores da Enzima Conversora de Angiotensina/farmacologia , Captopril/farmacologia , Inibidores de beta-Lactamases/farmacologia , beta-Lactamases/química , Inibidores da Enzima Conversora de Angiotensina/química , Antibacterianos/farmacologia , Captopril/química , Carbapenêmicos/farmacologia , Clonagem Molecular , Cristalografia por Raios X , Reposicionamento de Medicamentos , Enterobacteriaceae/efeitos dos fármacos , Enterobacteriaceae/enzimologia , Enterobacteriaceae/genética , Expressão Gênica , Hidrólise , Cinética , Testes de Sensibilidade Microbiana , Modelos Moleculares , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Estereoisomerismo , Relação Estrutura-Atividade , Resistência beta-Lactâmica/efeitos dos fármacos , Resistência beta-Lactâmica/genética , Inibidores de beta-Lactamases/química , beta-Lactamases/genética , beta-Lactamases/metabolismo
17.
Proc Natl Acad Sci U S A ; 110(24): 9710-5, 2013 Jun 11.
Artigo em Inglês | MEDLINE | ID: mdl-23716676

RESUMO

ABCB10 is one of the three ATP-binding cassette (ABC) transporters found in the inner membrane of mitochondria. In mammals ABCB10 is essential for erythropoiesis, and for protection of mitochondria against oxidative stress. ABCB10 is therefore a potential therapeutic target for diseases in which increased mitochondrial reactive oxygen species production and oxidative stress play a major role. The crystal structure of apo-ABCB10 shows a classic exporter fold ABC transporter structure, in an open-inwards conformation, ready to bind the substrate or nucleotide from the inner mitochondrial matrix or membrane. Unexpectedly, however, ABCB10 adopts an open-inwards conformation when complexed with nonhydrolysable ATP analogs, in contrast to other transporter structures which adopt an open-outwards conformation in complex with ATP. The three complexes of ABCB10/ATP analogs reported here showed varying degrees of opening of the transport substrate binding site, indicating that in this conformation there is some flexibility between the two halves of the protein. These structures suggest that the observed plasticity, together with a portal between two helices in the transmembrane region of ABCB10, assist transport substrate entry into the substrate binding cavity. These structures indicate that ABC transporters may exist in an open-inwards conformation when nucleotide is bound. We discuss ways in which this observation can be aligned with the current views on mechanisms of ABC transporters.


Assuntos
Transportadores de Cassetes de Ligação de ATP/química , Conformação Molecular , Nucleotídeos/química , Estrutura Terciária de Proteína , Transportadores de Cassetes de Ligação de ATP/genética , Transportadores de Cassetes de Ligação de ATP/metabolismo , Adenosina Trifosfatases/química , Adenosina Trifosfatases/genética , Adenosina Trifosfatases/metabolismo , Trifosfato de Adenosina/química , Trifosfato de Adenosina/metabolismo , Sequência de Aminoácidos , Animais , Sítios de Ligação/genética , Cristalografia por Raios X , Humanos , Bicamadas Lipídicas/química , Bicamadas Lipídicas/metabolismo , Modelos Moleculares , Simulação de Dinâmica Molecular , Dados de Sequência Molecular , Mutação , Nucleotídeos/metabolismo , Fosfatidilcolinas/química , Fosfatidiletanolaminas/química , Ligação Proteica , Homologia de Sequência de Aminoácidos , Células Sf9
18.
Am J Hum Genet ; 91(5): 897-905, 2012 Nov 02.
Artigo em Inglês | MEDLINE | ID: mdl-23063620

RESUMO

Carpenter syndrome is an autosomal-recessive multiple-congenital-malformation disorder characterized by multisuture craniosynostosis and polysyndactyly of the hands and feet; many other clinical features occur, and the most frequent include obesity, umbilical hernia, cryptorchidism, and congenital heart disease. Mutations of RAB23, encoding a small GTPase that regulates vesicular transport, are present in the majority of cases. Here, we describe a disorder caused by mutations in multiple epidermal-growth-factor-like-domains 8 (MEGF8), which exhibits substantial clinical overlap with Carpenter syndrome but is frequently associated with abnormal left-right patterning. We describe five affected individuals with similar dysmorphic facies, and three of them had either complete situs inversus, dextrocardia, or transposition of the great arteries; similar cardiac abnormalities were previously identified in a mouse mutant for the orthologous Megf8. The mutant alleles comprise one nonsense, three missense, and two splice-site mutations; we demonstrate in zebrafish that, in contrast to the wild-type protein, the proteins containing all three missense alterations provide only weak rescue of an early gastrulation phenotype induced by Megf8 knockdown. We conclude that mutations in MEGF8 cause a Carpenter syndrome subtype frequently associated with defective left-right patterning, probably through perturbation of signaling by hedgehog and nodal family members. We did not observe any subject with biallelic loss-of function mutations, suggesting that some residual MEGF8 function might be necessary for survival and might influence the phenotypes observed.


Assuntos
Acrocefalossindactilia/genética , Estudos de Associação Genética , Proteínas de Membrana/genética , Mutação , Acrocefalossindactilia/diagnóstico , Alelos , Animais , Animais Geneticamente Modificados , Criança , Pré-Escolar , Fácies , Feminino , Genótipo , Humanos , Masculino , Proteínas de Membrana/química , Peixe-Zebra/genética
19.
J Biol Chem ; 288(11): 7803-7814, 2013 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-23349464

RESUMO

Cullin-RING ligases are multisubunit E3 ubiquitin ligases that recruit substrate-specific adaptors to catalyze protein ubiquitylation. Cul3-based Cullin-RING ligases are uniquely associated with BTB adaptors that incorporate homodimerization, Cul3 assembly, and substrate recognition into a single multidomain protein, of which the best known are BTB-BACK-Kelch domain proteins, including KEAP1. Cul3 assembly requires a BTB protein "3-box" motif, analogous to the F-box and SOCS box motifs of other Cullin-based E3s. To define the molecular basis for this assembly and the overall architecture of the E3, we determined the crystal structures of the BTB-BACK domains of KLHL11 both alone and in complex with Cul3, along with the Kelch domain structures of KLHL2 (Mayven), KLHL7, KLHL12, and KBTBD5. We show that Cul3 interaction is dependent on a unique N-terminal extension sequence that packs against the 3-box in a hydrophobic groove centrally located between the BTB and BACK domains. Deletion of this N-terminal region results in a 30-fold loss in affinity. The presented data offer a model for the quaternary assembly of this E3 class that supports the bivalent capture of Nrf2 and reveals potential new sites for E3 inhibitor design.


Assuntos
Proteínas de Transporte/química , Proteínas Culina/química , Ubiquitina-Proteína Ligases/química , Sítios de Ligação , Calorimetria/métodos , Cristalografia por Raios X/métodos , Dimerização , Humanos , Modelos Moleculares , Conformação Molecular , Filogenia , Complexo de Endopeptidases do Proteassoma/metabolismo , Ligação Proteica , Conformação Proteica , Mapeamento de Interação de Proteínas , Estrutura Secundária de Proteína , Estrutura Terciária de Proteína , Especificidade por Substrato , Ubiquitina/química , Ubiquitina-Proteína Ligases/metabolismo
20.
Biochem Soc Trans ; 42(1): 108-13, 2014 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-24450636

RESUMO

Activation of cGMP synthesis leads to vasodilation, and is an important mechanism in clinical treatment of angina, heart failure, and severe peripheral and pulmonary hypertension. The nitric oxide-responsive sGC (soluble guanylate cyclase) has been the target of recent drug discovery efforts. The present review surveys recent data on the structure and regulation of sGC, and the prospects of new avenues for therapeutic intervention.


Assuntos
Doenças Cardiovasculares/tratamento farmacológico , Guanilato Ciclase/química , Regulação Alostérica , Animais , Domínio Catalítico , Descoberta de Drogas , Guanilato Ciclase/antagonistas & inibidores , Guanilato Ciclase/metabolismo , Humanos , Estrutura Secundária de Proteína
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA