Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 127
Filtrar
Mais filtros

Base de dados
País/Região como assunto
Tipo de documento
Intervalo de ano de publicação
1.
Bioconjug Chem ; 31(5): 1562-1574, 2020 05 20.
Artigo em Inglês | MEDLINE | ID: mdl-32369345

RESUMO

Exosomes are cellular components with promising uses in cancer diagnostics and therapeutics, and their imaging and tracking are essential to study their biological properties. Herein, we report on an in situ one-step fluorescence labeling strategy for exosomes via bioorthogonal click chemistry. First, exosome donor cancer cells were treated with tetraacetylated N-azidoacetyl-d-mannosamine (Ac4ManNAz) to generate unnatural azide groups (-N3) on their surface via metabolic glycoengineering. Then, the azide groups were labeled with near-infrared fluorescent dye-conjugated dibenzylcyclooctyne (DBCO-Cy5) via bioorthogonal click chemistry. After 2 days of incubation, the DBCO-Cy5-labeled exosomes (Cy5-Exo) were successfully secreted from the donor cancer cells and were isolated via classical ultracentrifugation, providing a high-yield of fluorescent dye-labeled exosomes. This in situ one-step bioorthogonal click chemistry offers improved labeling efficiency, biocompatibility, and imaging sensitivy compared to standard exosomes (ST-Exo), purified with classical ultracentrifugation or carbocyanine lipophilic dye (DiD)-labeled exosomes (DiD-Exo) in vitro. In particular, the Cy5-Exo were successfully taken up by A549 cells in a time-dependent manner, and they could escape from lysosome confinement, showing their possible use as a delivery carrier of therapeutic drugs or imaging agents. Finally, intraveneously injected Cy5-Exo were noninvasively tracked and imaged via near-infrared fluorescence (NIRF) imaging in tumor-bearing mice. This new fluorescence labeling strategy for natural exosomes may be useful to provide better understanding of their theranostic effects in many biomedical applications.


Assuntos
Exossomos/metabolismo , Corantes Fluorescentes/química , Corantes Fluorescentes/metabolismo , Imagem Óptica/métodos , Animais , Linhagem Celular Tumoral , Química Click , Cicloparafinas/química , Humanos , Camundongos
2.
Biochem Biophys Res Commun ; 503(2): 770-775, 2018 09 05.
Artigo em Inglês | MEDLINE | ID: mdl-29909012

RESUMO

Glucose isomerase (GI) catalyzes the reversible enzymatic isomerization of d-glucose and d-xylose to d-fructose and d-xylulose, respectively. This is one of the most important enzymes in the production of high-fructose corn syrup (HFCS) and biofuel. We recently determined the crystal structure of GI from S. rubiginosus (SruGI) complexed with a xylitol inhibitor in one metal binding mode. Although we assessed inhibitor binding at the M1 site, the metal binding at the M2 site and the substrate recognition mechanism for SruGI remains the unclear. Here, we report the crystal structure of the two metal binding modes of SruGI and its complex with glucose. This study provides a snapshot of metal binding at the SruGI M2 site in the presence of Mn2+, but not in the presence of Mg2+. Metal binding at the M2 site elicits a configuration change at the M1 site. Glucose molecule can only bind to the M1 site in presence of Mn2+ at the M2 site. Glucose and Mn2+ at the M2 site were bridged by water molecules using a hydrogen bonding network. The metal binding geometry of the M2 site indicates a distorted octahedral coordination with an angle of 55-110°, whereas the M1 site has a relatively stable octahedral coordination with an angle of 85-95°. We suggest a two-step sequential process for SruGI substrate recognition, in Mn2+ binding mode, at the M2 site. Our results provide a better understanding of the molecular role of the M2 site in GI substrate recognition.


Assuntos
Aldose-Cetose Isomerases/metabolismo , Glucose/metabolismo , Manganês/metabolismo , Streptomyces/enzimologia , Aldose-Cetose Isomerases/química , Sítios de Ligação , Cátions Bivalentes/metabolismo , Cristalografia por Raios X , Modelos Moleculares , Conformação Proteica , Streptomyces/química , Streptomyces/metabolismo , Especificidade por Substrato
3.
Amino Acids ; 50(11): 1583-1594, 2018 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-30105541

RESUMO

Previously we have demonstrated transglutaminase 2 (TGase 2) inhibition abrogated renal cell carcinoma (RCC) using GK921 (3-(phenylethynyl)-2-(2-(pyridin-2-yl)ethoxy)pyrido[3,2-b]pyrazine), although the mechanism of TGase 2 inhibition remains unsolved. Recently, we found that the increase of TGase 2 expression is required for p53 depletion in RCC by transporting the TGase 2 (1-139 a.a)-p53 complex to the autophagosome, through TGase 2 (472-687 a.a) binding p62. In this study, mass analysis revealed that GK921 bound to the N terminus of TGase 2 (81-116 a.a), which stabilized p53 by blocking TGase 2 binding. This suggests that RCC survival can be stopped by p53-induced cell death through blocking the p53-TGase 2 complex formation using GK921. Although GK921 does not bind to the active site of TGase 2, GK921 binding to the N terminus of TGase 2 also inactivated TGase 2 activity through acceleration of non-covalent self-polymerization of TGase 2 via conformational change. This suggests that TGase 2 has an allosteric binding site (81-116 a.a) which changes the conformation of TGase 2 enough to accelerate inactivation through self-polymer formation.


Assuntos
Carcinoma de Células Renais/enzimologia , Proteínas de Ligação ao GTP/metabolismo , Neoplasias Renais/enzimologia , Proteínas de Neoplasias/metabolismo , Transglutaminases/metabolismo , Regulação Alostérica , Carcinoma de Células Renais/tratamento farmacológico , Linhagem Celular Tumoral , Proteínas de Ligação ao GTP/antagonistas & inibidores , Proteínas de Ligação ao GTP/genética , Células HEK293 , Humanos , Neoplasias Renais/tratamento farmacológico , Proteínas de Neoplasias/antagonistas & inibidores , Proteínas de Neoplasias/genética , Domínios Proteicos , Proteína 2 Glutamina gama-Glutamiltransferase , Pirazinas/farmacologia , Transglutaminases/antagonistas & inibidores , Transglutaminases/genética
4.
Nucleic Acids Res ; 44(19): 9483-9493, 2016 Nov 02.
Artigo em Inglês | MEDLINE | ID: mdl-27596595

RESUMO

GTP and branched-chain amino acids (BCAAs) are metabolic sensors that are indispensable for the determination of the metabolic status of cells. However, their molecular sensing mechanism remains unclear. CodY is a unique global transcription regulator that recognizes GTP and BCAAs as specific signals and affects expression of more than 100 genes associated with metabolism. Herein, we report the first crystal structures of the full-length CodY complex with sensing molecules and describe their functional states. We observed two different oligomeric states of CodY: a dimeric complex of CodY from Staphylococcus aureus with the two metabolites GTP and isoleucine, and a tetrameric form (apo) of CodY from Bacillus cereus Notably, the tetrameric state shows in an auto-inhibitory manner by blocking the GTP-binding site, whereas the binding sites of GTP and isoleucine are clearly visible in the dimeric state. The GTP is located at a hinge site between the long helical region and the metabolite-binding site. Together, data from structural and electrophoretic mobility shift assay analyses improve understanding of how CodY senses GTP and operates as a DNA-binding protein and a pleiotropic transcription regulator.


Assuntos
Proteínas de Bactérias/química , Guanosina Trifosfato/química , Modelos Moleculares , Proteínas Repressoras/química , Fatores de Transcrição/química , Sequência de Aminoácidos , Proteínas de Bactérias/metabolismo , Sítios de Ligação , DNA/química , DNA/metabolismo , Guanosina Trifosfato/metabolismo , Conformação Proteica , Domínios e Motivos de Interação entre Proteínas , Proteínas Repressoras/metabolismo , Relação Estrutura-Atividade , Fatores de Transcrição/metabolismo
5.
Biochem Biophys Res Commun ; 490(2): 400-405, 2017 08 19.
Artigo em Inglês | MEDLINE | ID: mdl-28623126

RESUMO

tRNA maturation involves several steps, including processing, splicing, CCA addition, and posttranscriptional modifications. tRNAHis guanylyltransferase (Thg1) is the only enzyme known to catalyze templated nucleotide addition in the 3'-5' direction, unlike other DNA and RNA polymerases. For a better understanding of its unique catalytic mechanism at the molecular level, we determined the crystal structure of GTP-bound Thg1 from Saccharomyces cerevisiae at the maximum resolution of 3.0 Å. The structure revealed the enzyme to have a tetrameric conformation that is well conserved among different species, and the GTP molecule was clearly bound at the active site, coordinating with two magnesium ions. In addition, two flexible protomers at the potential binding site (PBS) for tRNAHis were observed. We suggest that the PBS of the tetramer could also be one of the sites for interaction with partner proteins.


Assuntos
Nucleotidiltransferases/química , Proteínas de Saccharomyces cerevisiae/química , Saccharomyces cerevisiae/química , Cristalografia por Raios X , Guanosina Trifosfato/metabolismo , Simulação de Acoplamento Molecular , Nucleotidiltransferases/metabolismo , Conformação Proteica , Saccharomyces cerevisiae/metabolismo , Proteínas de Saccharomyces cerevisiae/metabolismo
6.
Biochem Biophys Res Commun ; 493(1): 28-33, 2017 11 04.
Artigo em Inglês | MEDLINE | ID: mdl-28935372

RESUMO

Enoyl-ACP reductase (ENR, also known as FabI) has received considerable interest as an anti-bacterial target due to its essentiality in fatty acid synthesis. All the FabI structures reported to date, regardless of the organism, are composed of homo-tetramers, except for two structures: Bacillus cereus and Staphylococcus aureus FabI (bcFabI and saFabI, respectively), which have been reported as dimers. However, the reason for the existence of the dimeric form in these organisms and the biological meaning of dimeric and tetrameric forms of FabI are ambiguous. Herein, we report the high-resolution crystal structure of a dimeric form of Bacillus anthracis FabI (baFabI) and the crystal structures of tetrameric forms of baFabI in the apo state and in complex with NAD+ and with NAD+-triclosan, at 1.7 Å, 1.85 Å, 1.96 Å, and 1.95 Å, respectively. Interestingly, we found that baFabI with a His6-tag at its C-terminus exists as a dimer, whereas untagged-baFabI exists as a tetramer. The His6-tag may block the dimer-tetramer transition, since baFabI has relatively short-length amino acids (255LG256) after the 310-helix η7 compared to those of FabI of other organisms. The dimeric form of baFabI is catalytically inactive, because the α-helix α5 occupies the NADH-binding site. During the process of dimer-tetramer transition, this α5 helix rotates about 55° toward the tetramer interface and the active site is established. Therefore, tetramerization of baFabI is required for cofactor binding and catalytic activity.


Assuntos
Bacillus anthracis/química , Proteínas de Bactérias/química , Proteínas de Bactérias/ultraestrutura , Enoil-(Proteína de Transporte de Acila) Redutase (NADH)/química , Enoil-(Proteína de Transporte de Acila) Redutase (NADH)/ultraestrutura , Modelos Químicos , Simulação de Acoplamento Molecular , NAD/química , NAD/ultraestrutura , Sítios de Ligação , Cristalografia por Raios X , Dimerização , Ligação Proteica , Conformação Proteica , Relação Estrutura-Atividade
7.
Proc Natl Acad Sci U S A ; 111(42): 15084-9, 2014 Oct 21.
Artigo em Inglês | MEDLINE | ID: mdl-25288775

RESUMO

In higher eukaryotes, one of the two arginyl-tRNA synthetases (ArgRSs) has evolved to have an extended N-terminal domain that plays a crucial role in protein synthesis and cell growth and in integration into the multisynthetase complex (MSC). Here, we report a crystal structure of the MSC subcomplex comprising ArgRS, glutaminyl-tRNA synthetase (GlnRS), and the auxiliary factor aminoacyl tRNA synthetase complex-interacting multifunctional protein 1 (AIMP1)/p43. In this complex, the N-terminal domain of ArgRS forms a long coiled-coil structure with the N-terminal helix of AIMP1 and anchors the C-terminal core of GlnRS, thereby playing a central role in assembly of the three components. Mutation of AIMP1 destabilized the N-terminal helix of ArgRS and abrogated its catalytic activity. Mutation of the N-terminal helix of ArgRS liberated GlnRS, which is known to control cell death. This ternary complex was further anchored to AIMP2/p38 through interaction with AIMP1. These findings demonstrate the importance of interactions between the N-terminal domains of ArgRS and AIMP1 for the catalytic and noncatalytic activities of ArgRS and for the assembly of the higher-order MSC protein complex.


Assuntos
Aminoacil-tRNA Sintetases/química , Arginina-tRNA Ligase/química , Citocinas/química , Proteínas de Neoplasias/química , Proteínas de Ligação a RNA/química , Sítios de Ligação , Cromatografia em Gel , Dicroísmo Circular , Cristalografia por Raios X , Escherichia coli/metabolismo , Glutationa Transferase/química , Humanos , Modelos Moleculares , Complexos Multiproteicos , Mutagênese , Mutação , Biossíntese de Proteínas , Estrutura Secundária de Proteína , Estrutura Terciária de Proteína , Espalhamento de Radiação
8.
Biochemistry ; 55(36): 5117-27, 2016 09 13.
Artigo em Inglês | MEDLINE | ID: mdl-27551953

RESUMO

Many bacteria, particularly pathogens, possess methionine sulfoxide reductase A (MsrA) and B (MsrB) as a fusion form (MsrAB). However, it is not clear why they possess a fusion MsrAB form rather than the separate enzymes that exist in most organisms. In this study, we performed biochemical and kinetic analyses of MsrAB from Treponema denticola (TdMsrAB), single-domain forms (TdMsrA and TdMsrB), and catalytic Cys mutants (TdMsrAB(C11S) and TdMsrAB(C285S)). We found that the catalytic efficiency of both MsrA and MsrB increased after fusion of the domains and that the linker region (iloop) that connects TdMsrA and TdMsrB is required for the higher catalytic efficiency of TdMsrAB. We also determined the crystal structure of TdMsrAB at 2.3 Å, showing that the iloop mainly interacts with TdMsrB via hydrogen bonds. Further kinetic analysis using the iloop mutants revealed that the iloop-TdMsrB interactions are critical to MsrB and MsrA activities. We also report the structure in which an oxidized form of dithiothreitol, an in vitro reductant for MsrA and MsrB, is present in the active site of TdMsrA. Collectively, the results of this study reveal an essential role of the iloop in maintaining the higher catalytic efficiency of the MsrAB fusion enzyme and provide a better understanding of why the MsrAB enzyme exists as a fused form.


Assuntos
Proteínas de Bactérias/metabolismo , Proteínas Recombinantes de Fusão/metabolismo , Sequência de Aminoácidos , Proteínas de Bactérias/química , Biocatálise , Dicroísmo Circular , Proteínas Recombinantes de Fusão/química , Homologia de Sequência de Aminoácidos
9.
Anal Chem ; 88(3): 1516-20, 2016 Feb 02.
Artigo em Inglês | MEDLINE | ID: mdl-26713895

RESUMO

The crystallinity of polyethylene, which significantly affects the properties of the polymer, is quite sensitive to the concentration of its branches. Thus, it is necessary to estimate branch concentration with reasonable accuracy. Currently, (13)C NMR and gel permeation chromatography-Fourier transform infrared spectroscopy are widely-used analysis methods for the analysis of branch concentration. Despite several advantages, these methods sometimes have limitations. For instance, the preparation of samples for (13)C- NMR is tedious because high-concentration samples are required and the time for analysis is greater than 12 h. To more efficiently estimate the branch concentration of polyethylene, we developed a new high-field (1)H NMR method with an improved peak resolution by employing (1) homonuclear decoupling and (2) 2D heteronuclear correlation. The new method was observed to significantly reduce the experimental time to ∼ 30 min; furthermore, sample preparation was relatively simple because the method did not require high-concentration samples.

10.
Biochem Biophys Res Commun ; 478(1): 1-6, 2016 09 09.
Artigo em Inglês | MEDLINE | ID: mdl-27475498

RESUMO

Polmacoxib is not only a selective COX-2 inhibitor but also a potent inhibitor of carbonic anhydrases (CAs). Both CA I and CA II are highly expressed in the GI tract and kidneys, organs that are also thought to be the sites at which selective COX-2 inhibitors show their side effects. By inhibition assays, we show that both CA I and CA II are strongly inhibited by polmacoxib, while CA II also demonstrates direct competition with COX-2. To understand, at the molecular level, how polmacoxib interacts with CA I and II, we solved the first crystal structures of CA I and CA II in complex with polmacoxib, at 2.0 Å and 1.8 Å, respectively. Interestingly, three polmacoxib molecules bind to the active site of CA I, whereas only one molecule binds CA II. In the active site, the three molecules of polmacoxib organize itself along hydrophobic interaction as "stack-on-formation", and fully occupy a cone-shaped active pocket in CA I. The binding mode of polmacoxib to CA II was found different than its binding to celecoxib and valdecoxib. Our results provide structural insight into inhibition of CA I and CA II by polmacoxib, to assess its potential clinical efficacy.


Assuntos
Anidrase Carbônica II/antagonistas & inibidores , Anidrase Carbônica II/química , Anidrase Carbônica I/antagonistas & inibidores , Anidrase Carbônica I/química , Inibidores da Anidrase Carbônica/farmacologia , Inibidores de Ciclo-Oxigenase 2/farmacologia , Furanos/farmacocinética , Sulfonamidas/farmacocinética , Anidrase Carbônica I/metabolismo , Anidrase Carbônica II/metabolismo , Domínio Catalítico/efeitos dos fármacos , Cristalografia por Raios X , Humanos , Modelos Moleculares , Conformação Proteica/efeitos dos fármacos , Multimerização Proteica/efeitos dos fármacos
11.
Arch Biochem Biophys ; 606: 143-50, 2016 09 15.
Artigo em Inglês | MEDLINE | ID: mdl-27495739

RESUMO

RNase H fold protein PF2046 of Pyrococcus furiosus is a 3'-5' ssDNA exonuclease that cleaves after the second nucleotide from the 3' end of ssDNA and prefers poly-dT over poly-dA as a substrate. In our crystal structure of PF2046 complexed with an oligonucleotide of four thymidine nucleotides (dT4), PF2046 accommodates dT4 tightly in a groove and imposes steric hindrance on dT4 mainly by Phe220 such that dT4 assumes the A-form. As poly-dA prefer B-form due to the stereochemical restrictions, the A-form ssDNA binding by PF2046 should disfavor the processing of poly-dA. Phe220 variants display reduced activity toward poly-dA and the A-form appears to be a prerequisite for the processing by PF2046.


Assuntos
Proteínas de Bactérias/química , DNA de Cadeia Simples/química , Pyrococcus furiosus/enzimologia , Ribonuclease H/química , Domínio Catalítico , Cristalização , Cristalografia por Raios X , Reparo do DNA , Exonucleases/química , Concentração de Íons de Hidrogênio , Mutação , Conformação de Ácido Nucleico , Oligodesoxirribonucleotídeos , Especificidade por Substrato , Difração de Raios X
12.
Protein Expr Purif ; 126: 122-126, 2016 10.
Artigo em Inglês | MEDLINE | ID: mdl-27371359

RESUMO

In enterohemorrhagic Escherichia coli (EHEC), the QseEF two-component system causes attaching and effacing (AE) lesion on epithelial cells. QseE histidine kinase senses the host hormone epinephrine, sulfate, and phosphate; it also regulates QseF response regulator, which activates LEE gene that encodes AE lesion. In order to understand the recognition of ligand molecules and signal transfer mechanism in pathogenic bacteria, structural studies of the sensor domain of QseE of Escherichia coli should be conducted. In this study, we describe the overexpression, purification, and structural and biophysical properties of the sensor domain of QseE. The fusion protein had a 6×His tag at its N-terminus; this protein was overexpressed as inclusion bodies in E. coli BL21 (DE3). The protein was denatured in 7M guanidine hydrochloride and refolded by dialysis. The purification of the refolded protein was carried out using Ni-NTA affinity column and size-exclusion chromatography. Thereafter, the characteristics of the refolded protein were determined from NMR, CD, and MALS spectroscopies. In a pH range of 7.4-5.0, the folded protein existed in a monomeric form with a predominantly helical structure. (1)H-(15)N HSQC NMR spectra shows that approximately 93% backbone amide peaks are detected at pH 5.0, suggesting that the number of backbone signals is sufficient for NMR studies. These data might provide an opportunity for structural and functional studies of the sensor domain of QseE.


Assuntos
Escherichia coli Êntero-Hemorrágica/enzimologia , Proteínas de Escherichia coli , Expressão Gênica , Redobramento de Proteína , Receptores Adrenérgicos , Escherichia coli Êntero-Hemorrágica/genética , Proteínas de Escherichia coli/biossíntese , Proteínas de Escherichia coli/genética , Proteínas de Escherichia coli/isolamento & purificação , Concentração de Íons de Hidrogênio , Ressonância Magnética Nuclear Biomolecular , Domínios Proteicos , Estrutura Secundária de Proteína , Receptores Adrenérgicos/biossíntese , Receptores Adrenérgicos/genética , Receptores Adrenérgicos/isolamento & purificação , Proteínas Recombinantes de Fusão/biossíntese , Proteínas Recombinantes de Fusão/química , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/isolamento & purificação
13.
Biochem Biophys Res Commun ; 465(4): 817-24, 2015 Oct 02.
Artigo em Inglês | MEDLINE | ID: mdl-26315269

RESUMO

The SET domain of LegAS4, a type IV secretion system effector of Legionella pneumophila, is a eukaryotic protein motif involved in histone methylation and epigenetic modulation. The SET domain of LegAS4 is involved in the modification of Lys4 of histone H3 (H3K4) in the nucleolus of the host cell, thereby enhancing heterochromatic rDNA transcription. Moreover, LegAS4 contains an ankyrin repeat domain of unknown function at its C-terminal region. Here, we report the crystal structure of LegAS4 in complex with S-adenosyl-l-methionine (SAM). Our data indicate that the ankyrin repeats interact extensively with the SET domain, especially with the SAM-binding amino acids, through conserved residues. Conserved surface analysis marks Glu159, Glu203, and Glu206 on the SET domain serve as candidate residues involved in interaction with the positively charged histone tail. Conserved surface residues on the ankyrin repeat domain surround a small pocket, which is suspected to serve as a binding site for an unknown ligand.


Assuntos
Proteínas de Bactérias/química , Legionella pneumophila/química , Sistemas de Secreção Tipo IV/química , Sequência de Aminoácidos , Repetição de Anquirina , Proteínas de Bactérias/genética , Proteínas de Bactérias/metabolismo , Sítios de Ligação , Cristalografia por Raios X , Histonas/genética , Histonas/metabolismo , Legionella pneumophila/genética , Legionella pneumophila/metabolismo , Modelos Moleculares , Dados de Sequência Molecular , Domínios e Motivos de Interação entre Proteínas , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , S-Adenosilmetionina/química , S-Adenosilmetionina/metabolismo , Homologia Estrutural de Proteína , Sistemas de Secreção Tipo IV/genética , Sistemas de Secreção Tipo IV/metabolismo
14.
Biochem Biophys Res Commun ; 461(4): 648-52, 2015 Jun 12.
Artigo em Inglês | MEDLINE | ID: mdl-25912135

RESUMO

Thioredoxin (Trx) is a major thiol-disulfide reductase that plays a role in many biological processes, including DNA replication and redox signaling. Although selenocysteine (Sec)-containing Trxs have been identified in certain bacteria, their enzymatic properties have not been characterized. In this study, we expressed a selenoprotein Trx from Treponema denticola, an oral spirochete, in Escherichia coli and characterized this selenoenzyme and its natural cysteine (Cys) homologue using E. coli Trx1 as a positive control. (75)Se metabolic labeling and mutation analyses showed that the SECIS (Sec insertion sequence) of T. denticola selenoprotein Trx is functional in the E. coli Sec insertion system with specific selenium incorporation into the Sec residue. The selenoprotein Trx exhibited approximately 10-fold higher catalytic activity than the Sec-to-Cys version and natural Cys homologue and E. coli Trx1, suggesting that Sec confers higher catalytic activity on this thiol-disulfide reductase. Kinetic analysis also showed that the selenoprotein Trx had a 30-fold higher Km than Cys-containing homologues, suggesting that this selenoenzyme is adapted to work efficiently with high concentrations of substrate. Collectively, the results of this study support the hypothesis that selenium utilization in oxidoreductase systems is primarily due to the catalytic advantage provided by the rare amino acid, Sec.


Assuntos
Selênio/química , Selenocisteína/química , Tiorredoxinas/química , Treponema denticola/enzimologia , Sítios de Ligação , Catálise , Ativação Enzimática , Ligação Proteica , Relação Estrutura-Atividade , Especificidade por Substrato
15.
Biochem Biophys Res Commun ; 457(4): 567-71, 2015 Feb 20.
Artigo em Inglês | MEDLINE | ID: mdl-25600814

RESUMO

Glutaredoxin (Grx), a major redox regulator, can act as a reductant of methionine sulfoxide reductase A (MsrA). However, the biochemical mechanisms involved in MsrA activity regeneration by Grx remain largely unknown. In this study, we investigated the regeneration mechanism of 1-Cys type Clostridium oremlandii MsrA (cMsrA) lacking a resolving Cys residue in a Grx-dependent assay. Kinetic analysis showed that cMsrA could be reduced by both monothiol and dithiol Grxs as efficiently as by in vitro reductant dithiothreitol. Our data revealed that the catalytic Cys sulfenic acid intermediate is not glutathionylated in the presence of the substrate, and that Grx instead directly formed a complex with cMsrA. Mass spectrometry analysis identified a disulfide bond between the N-terminal catalytic Cys of the active site of Grx and the catalytic Cys of cMsrA. This mixed disulfide bond could be resolved by glutathione. Based on these findings, we propose a model for regeneration of 1-Cys type cMsrA by Grx that involves no glutathionylation on the catalytic Cys of cMsrA. This mechanism contrasts with that of the previously known 1-Cys type MsrB.


Assuntos
Clostridium/enzimologia , Glutarredoxinas/metabolismo , Metionina Sulfóxido Redutases/metabolismo , Sequência de Aminoácidos , Clostridium/química , Clostridium/metabolismo , Cisteína/análogos & derivados , Cisteína/química , Cisteína/metabolismo , Glutarredoxinas/química , Glutationa/metabolismo , Cinética , Metionina Sulfóxido Redutases/química , Dados de Sequência Molecular , Ácidos Sulfênicos/química , Ácidos Sulfênicos/metabolismo
16.
J Lipid Res ; 55(6): 1098-110, 2014 06.
Artigo em Inglês | MEDLINE | ID: mdl-24752549

RESUMO

We investigated the hypotriglyceridemic mechanism of action of linalool, an aromatic monoterpene present in teas and fragrant herbs. Reporter gene and time-resolved fluorescence resonance energy transfer assays demonstrated that linalool is a direct ligand of PPARα. Linalool stimulation reduced cellular lipid accumulation regulating PPARα-responsive genes and significantly induced FA oxidation, and its effects were markedly attenuated by silencing PPARα expression. In mice, the oral administration of linalool for 3 weeks reduced plasma TG concentrations in Western-diet-fed C57BL/6J mice (31%, P < 0.05) and human apo E2 mice (50%, P < 0.05) and regulated hepatic PPARα target genes. However, no such effects were seen in PPARα-deficient mice. Transcriptome profiling revealed that linalool stimulation rewired global gene expression in lipid-loaded hepatocytes and that the effects of 1 mM linalool were comparable to those of 0.1 mM fenofibrate. Metabolomic analysis of the mouse plasma revealed that the global metabolite profiles were significantly distinguishable between linalool-fed mice and controls. Notably, the concentrations of saturated FAs were significantly reduced in linalool-fed mice. These findings suggest that the appropriate intake of a natural aromatic compound could exert beneficial metabolic effects by regulating a cellular nutrient sensor.


Assuntos
Fígado/metabolismo , Metaboloma/efeitos dos fármacos , Monoterpenos/farmacologia , PPAR alfa/biossíntese , Transcriptoma/efeitos dos fármacos , Triglicerídeos/sangue , Monoterpenos Acíclicos , Animais , Hepatócitos/metabolismo , Masculino , Camundongos , Camundongos Mutantes , PPAR alfa/agonistas , PPAR alfa/genética , Triglicerídeos/genética
17.
Biochim Biophys Acta ; 1831(4): 698-708, 2013 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-23228689

RESUMO

To investigate the underlying mechanism of targets of cyanidin, a flavonoid, which exhibits potent anti-atherogenic activities in vitro and in vivo, a natural chemical library that identified potent agonistic activity between cyanidin and peroxisome proliferator-activated receptors (PPAR) was performed. Cyanidin induced transactivation activity in all three PPAR subtypes in a reporter gene assay and time-resolved fluorescence energy transfer analyses. Cyanidin also bound directly to all three subtypes, as assessed by surface plasmon resonance experiments, and showed the greatest affinity to PPARα. These effects were confirmed by measuring the expression of unique genes of each PPAR subtype. Cyanidin significantly reduced cellular lipid concentrations in lipid-loaded steatotic hepatocytes. In addition, transcriptome profiling in lipid-loaded primary hepatocytes revealed that the net effects of stimulation with cyanidin on lipid metabolic pathways were similar to those elicited by hypolipidemic drugs. Cyanidin likely acts as a physiological PPARα agonist and potentially for PPARß/δ and γ, and reduces hepatic lipid concentrations by rewiring the expression of genes involved in lipid metabolic pathways.


Assuntos
Antocianinas/farmacologia , Hepatócitos/efeitos dos fármacos , Hepatócitos/metabolismo , PPAR alfa/agonistas , Animais , Células CHO , Células Cultivadas , Cricetinae , Células Hep G2 , Humanos , Metabolismo dos Lipídeos/efeitos dos fármacos , PPAR gama/agonistas , PPAR beta/agonistas
18.
Acta Crystallogr D Biol Crystallogr ; 70(Pt 4): 922-31, 2014 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-24699638

RESUMO

The aconitase family of proteins includes three classes of hydro-lyase enzymes: aconitases, homoaconitases and isopropylmalate (IPM) isomerases. They have a common Fe-S cluster-binding site and catalyze the isomerization of specific substrates by sequential dehydration and hydration. The archaeon Methanococcus jannaschii contains two aconitase family proteins, IPM isomerase and homoaconitase, which have 50% sequence identity. These two enzymes are heterodimeric proteins composed of large and small subunits encoded by separate genes. Although structures have been reported for the small subunits of the two enzymes, the first structures of oxidized and reduced forms of the large subunit of IPM isomerase (ox-MJ0499 and red-MJ0499, respectively) from M. jannaschii are reported here at 1.8 and 2.7 Šresolution, respectively, together with the structure of the large subunit of homoaconitase (MJ1003) at 2.5 Šresolution. The structures of both proteins have unbound Fe-S clusters and contain a fourth cysteine in the active site. The active site of MJ1003 is homologous to that of aconitase, whereas MJ0499 has significant structural distortion at the active site compared with aconitase. In addition, significant large conformational changes were observed in the active site of red-MJ0499 when compared with ox-MJ0499. The active sites of the two proteins adopt two different states before changing to the Fe-S cluster-bound `activated' state observed in aconitase. MJ1003 has an `open' active site, which forms an active pocket for the cluster, while ox-MJ0499 has a `closed' active site, with four cysteines in disulfide bonds. These data will be helpful in understanding the biochemical mechanism of clustering of the Fe-S protein family.


Assuntos
Hidroliases/química , Isomerases/química , Methanocaldococcus/enzimologia , Modelos Moleculares , Estrutura Quaternária de Proteína , Estrutura Terciária de Proteína , Homologia Estrutural de Proteína
19.
Acta Crystallogr D Biol Crystallogr ; 70(Pt 11): 2863-74, 2014 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-25372678

RESUMO

Muskelin is an intracellular kelch-repeat protein comprised of discoidin, LisH, CTLH and kelch-repeat domains. It is involved in cell adhesion and the regulation of cytoskeleton dynamics as well as being a component of a putative E3 ligase complex. Here, the first crystal structure of mouse muskelin discoidin domain (MK-DD) is reported at 1.55 Šresolution, which reveals a distorted eight-stranded ß-barrel with two short α-helices at one end of the barrel. Interestingly, the N- and C-termini are not linked by the disulfide bonds found in other eukaryotic discoidin structures. A highly conserved MIND motif appears to be the determinant for MK-DD specific interaction together with the spike loops. Analysis of interdomain interaction shows that MK-DD binds the kelch-repeat domain directly and that this interaction depends on the presence of the LisH domain.


Assuntos
Moléculas de Adesão Celular/química , Peptídeos e Proteínas de Sinalização Intracelular/química , Lectinas/química , Proteínas de Protozoários/química , Sequência de Aminoácidos , Animais , Moléculas de Adesão Celular/metabolismo , Cristalografia por Raios X , Discoidinas , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Lectinas/metabolismo , Camundongos , Modelos Moleculares , Dados de Sequência Molecular , Estrutura Secundária de Proteína , Estrutura Terciária de Proteína , Proteínas de Protozoários/metabolismo , Alinhamento de Sequência
20.
Acta Crystallogr D Biol Crystallogr ; 70(Pt 7): 1944-53, 2014 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-25004971

RESUMO

Despite recent progress in research on the Hippo signalling pathway, the structural information available in this area is extremely limited. Intriguingly, the homodimeric and heterodimeric interactions of mammalian sterile 20-like (MST) kinases through the so-called `SARAH' (SAV/RASSF/HPO) domains play a critical role in cellular homeostasis, dictating the fate of the cell regarding cell proliferation or apoptosis. To understand the mechanism of the heterodimerization of SARAH domains, the three-dimensional structures of an MST1-RASSF5 SARAH heterodimer and an MST2 SARAH homodimer were determined by X-ray crystallography and were analysed together with that previously determined for the MST1 SARAH homodimer. While the structure of the MST2 homodimer resembled that of the MST1 homodimer, the MST1-RASSF5 heterodimer showed distinct structural features. Firstly, the six N-terminal residues (Asp432-Lys437), which correspond to the short N-terminal 310-helix h1 kinked from the h2 helix in the MST1 homodimer, were disordered. Furthermore, the MST1 SARAH domain in the MST1-RASSF5 complex showed a longer helical structure (Ser438-Lys480) than that in the MST1 homodimer (Val441-Lys480). Moreover, extensive polar and nonpolar contacts in the MST1-RASSF5 SARAH domain were identified which strengthen the interactions in the heterodimer in comparison to the interactions in the homodimer. Denaturation experiments performed using urea also indicated that the MST-RASSF heterodimers are substantially more stable than the MST homodimers. These findings provide structural insights into the role of the MST1-RASSF5 SARAH domain in apoptosis signalling.


Assuntos
MAP Quinase Quinase Quinases/metabolismo , Proteínas Serina-Treonina Quinases/metabolismo , Transdução de Sinais , Sequência de Aminoácidos , Cristalografia por Raios X , Dimerização , Via de Sinalização Hippo , Humanos , Dados de Sequência Molecular , Conformação Proteica , Homologia de Sequência de Aminoácidos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA