Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 56
Filtrar
Mais filtros

Métodos Terapêuticos e Terapias MTCI
Base de dados
País/Região como assunto
Tipo de documento
Intervalo de ano de publicação
1.
Orig Life Evol Biosph ; 52(1-3): 149-163, 2022 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-36071304

RESUMO

The origin of the genetic code is probably the central problem of the studies on the origin of life. The key question to answer is the molecular mechanism that allows the association of the amino acids with their triplet codons. We proposed that the codon-anticodon duplex located in the acceptor stem of primitive tRNAs would facilitate the chemical reactions required to synthesize cognate amino acids from simple amino acids (glycine, valine, and aspartic acid) linked to the 3' acceptor end. In our view, various nucleotide-A-derived cofactors (with reactive chemical groups) may be attached to the codon-anticodon duplex, which allows group-transferring reactions from cofactors to simple amino acids, thereby producing the final amino acid. The nucleotide-A-derived cofactors could be incorporated into the RNA duplex (helix) by docking Adenosine (cofactor) into the minor groove via an interaction similar to the A-minor motif, forming a base triple between Adenosine and one complementary base pair of the duplex. Furthermore, we propose that this codon-anticodon duplex could initially catalyze a self-aminoacylation reaction with a simple amino acid. Therefore, the sequence of bases in the codon-anticodon duplex would determine the reactions that occurred during the formation of new amino acids for selective binding of nucleotide-A-derived cofactors.


Assuntos
Anticódon , Ácido Aspártico , Adenosina , Aminoácidos/química , Ácido Aspártico/genética , Códon , Código Genético , Glicina , Nucleotídeos , RNA/química , RNA de Transferência/química , RNA de Transferência/genética , Valina
2.
New Phytol ; 216(3): 814-828, 2017 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-28722263

RESUMO

Little is known about the molecular mechanism of the R2R3-MYB transcriptional repressors involved in plant phenylpropanoid metabolism. Here, we describe one R2R3-type MYB repressor, FtMYB11 from Fagopyrum tataricum. It contains the SID-like motif GGDFNFDL and it is regulated by both the importin protein 'Sensitive to ABA and Drought 2' (SAD2) and the jasmonates signalling cascade repressor JAZ protein. Yeast two hybrid and bimolecular fluorescence complementation assays demonstrated that FtMYB11 interacts with SAD2 and FtJAZ1. Protoplast transactivation assays demonstrated that FtMYB11 acts synergistically with FtSAD2 or FtJAZ1 and directly represses its target genes via the MYB-core element AATAGTT. Changing the Asp122 residue to Asn in the SID-like motif results in cytoplasmic localization of FtMYB11 because of loss of interaction with SAD2, while changing the Asp126 residue to Asn results in the loss of interaction with FtJAZ1. Overexpression of FtMYB11or FtMYB11D126N in F. tataricum hairy roots resulted in reduced accumulation of rutin, while overexpression of FtMYB11D122N in hairy roots did not lead to such a change. The results indicate that FtMYB11 acts as a regulator via interacting with FtSAD2 or FtJAZ1 to repress phenylpropanoid biosynthesis, and this repression depends on two conserved Asp residues of its SID-like motif.


Assuntos
Fagopyrum/metabolismo , Regulação da Expressão Gênica de Plantas , Proteínas de Plantas/metabolismo , Motivos de Aminoácidos , Arabidopsis/genética , Ácido Aspártico/genética , Ácido Aspártico/metabolismo , Citoplasma/metabolismo , Fagopyrum/genética , Teste de Complementação Genética , Mutação , Fenilpropionatos/metabolismo , Proteínas de Plantas/genética , Raízes de Plantas/genética , Raízes de Plantas/metabolismo , Plantas Geneticamente Modificadas , Proteínas Repressoras/genética , Proteínas Repressoras/metabolismo , Rutina/biossíntese , Rutina/genética , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo
3.
J Mol Biol ; 427(22): 3491-3500, 2015 Nov 06.
Artigo em Inglês | MEDLINE | ID: mdl-26196441

RESUMO

Hfq facilitates gene regulation by small non-coding RNAs (sRNAs), thereby affecting bacterial attributes such as biofilm formation and virulence. Escherichia coli Hfq recognizes specific U-rich and AAN motifs in sRNAs and target mRNAs, after which an arginine patch on the rim promotes base pairing between their complementary sequences. In the cell, Hfq must discriminate between many similar RNAs. Here, we report that acidic amino acids lining the sRNA binding channel between the inner pore and rim of the Hfq hexamer contribute to the selectivity of Hfq's chaperone activity. RNase footprinting, in vitro binding and stopped-flow fluorescence annealing assays showed that alanine substitution of D9, E18 or E37 strengthened RNA interactions with the rim of Hfq and increased annealing of non-specific or U-tailed RNA oligomers. Although the mutants were less able than wild-type Hfq to anneal sRNAs with wild-type rpoS mRNA, the D9A mutation bypassed recruitment of Hfq to an (AAN)4 motif in rpoS, both in vitro and in vivo. These results suggest that acidic residues normally modulate access of RNAs to the arginine patch. We propose that this selectivity limits indiscriminate target selection by E. coli Hfq and enforces binding modes that favor genuine sRNA and mRNA pairs.


Assuntos
Proteínas de Escherichia coli/metabolismo , Escherichia coli/metabolismo , Regulação Bacteriana da Expressão Gênica , Fator Proteico 1 do Hospedeiro/metabolismo , RNA Bacteriano/metabolismo , RNA Mensageiro/metabolismo , Pequeno RNA não Traduzido/metabolismo , Arginina/química , Arginina/genética , Arginina/metabolismo , Ácido Aspártico/química , Ácido Aspártico/genética , Ácido Aspártico/metabolismo , Pareamento de Bases , Sequência de Bases , Pegada de DNA , Ensaio de Desvio de Mobilidade Eletroforética , Escherichia coli/genética , Proteínas de Escherichia coli/genética , Ácido Glutâmico/química , Ácido Glutâmico/genética , Ácido Glutâmico/metabolismo , Fator Proteico 1 do Hospedeiro/genética , Dados de Sequência Molecular , Mutação/genética , Conformação de Ácido Nucleico , RNA Bacteriano/genética , RNA Mensageiro/genética , Pequeno RNA não Traduzido/genética
4.
PLoS One ; 9(4): e93550, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24695807

RESUMO

The common, co-segregating Toll-like receptor 4 (TLR4) non-synonymous single nucleotide polymorphisms (SNPs), Asp299Gly and Thr399Ile, are associated with hyporesponsiveness to inhaled lipopolysaccharide (LPS) and increased susceptibility to Gram negative pathogens in humans. The purpose of this study was to identify the relative contributions of the Asp299Gly and the Thr399Ile variants in inhibiting the function of TLR4. 293/hMD2-CD14 cell line was transfected with lentiviral constructs containing human wild type (WT) TLR4-EGFP or TLR4-EGFP with Asp299Gly, Thr399Ile or Asp299Gly/Thr399Ile complementary DNA (cDNA). Multiple stable cell lines were established for each construct: three for WT TLR4, Asp299Gly, and Thr399Ile, and only two for Asp299Gly/Thr399Ile mutants and EGFP control. We did not observe a significant effect of polymorphisms on cell surface and intracellular TLR4 expression nor were there any significant differences in TLR4 and EGFP protein levels assessed by Western blotting and confocal microscopy among the multiple cell lines of each of the constructs. All cell lines had a dose-dependent responsiveness to LPS stimulation. However, compared to the WT TLR4, cells expressing TLR4 with Asp299Gly but not Thr399Ile polymorphism produced significantly less (P<0.05) IL-8 following LPS stimulation. Similarly, cells expressing TLR4 Asp299Gly but not Thr399Ile allele had significantly lower percentage of phosphorylated and total NF-κB P65 following LPS stimulation. While we could not do statistics on the Asp299Gly/Thr399Ile group, we observed a reduced responsiveness to LPS compared to WT TLR4. Taken together, we observed that the TLR4 Asp299Gly variant, but not the Thr399Ile variant, is responsible for impaired responsiveness of TLR4 to LPS and corresponding activation of NF-κB.


Assuntos
Ácido Aspártico/genética , Glicina/genética , Polimorfismo Genético , Transdução de Sinais , Receptor 4 Toll-Like/metabolismo , Linhagem Celular , Humanos , Isoleucina/genética , NF-kappa B/metabolismo , Treonina/genética , Receptor 4 Toll-Like/genética
5.
Mol Plant Microbe Interact ; 27(2): 150-62, 2014 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-24111920

RESUMO

Viral determinants and mechanisms involved in extension of host range of monocot-infecting viruses are poorly understood. Viral coat proteins (CP) serve many functions in almost every aspect of the virus life cycle. The role of the C-terminal region of Wheat streak mosaic virus (WSMV) CP in virus biology was examined by mutating six negatively charged aspartic acid residues at positions 216, 289, 290, 326, 333, and 334. All of these amino acid residues are dispensable for virion assembly, and aspartic acid residues at positions 216, 333, and 334 are expendable for normal infection of wheat and maize. However, mutants D289N, D289A, D290A, DD289/290NA, and D326A exhibited slow cell-to-cell movement in wheat, which resulted in delayed onset of systemic infection, followed by a rapid recovery of genomic RNA accumulation and symptom development. Mutants D289N, D289A, and D326A inefficiently infected maize, eliciting milder symptoms, while D290A and DD289/290NA failed to infect systemically, suggesting that the C-terminus of CP is involved in differential infection of wheat and maize. Mutation of aspartic acid residues at amino acid positions 289, 290, and 326 severely debilitated virus ingress into the vascular system of maize but not wheat, suggesting that these amino acids facilitate expansion of WSMV host range through host-specific long-distance transport.


Assuntos
Proteínas do Capsídeo/metabolismo , Especificidade de Hospedeiro , Doenças das Plantas/virologia , Potyviridae/metabolismo , Triticum/virologia , Zea mays/virologia , Sequência de Aminoácidos , Ácido Aspártico/genética , Ácido Aspártico/metabolismo , Transporte Biológico , Proteínas do Capsídeo/genética , DNA Complementar/genética , Proteínas de Fluorescência Verde , Dados de Sequência Molecular , Mutação , Feixe Vascular de Plantas/virologia , Potyviridae/genética , Potyviridae/ultraestrutura , RNA Viral/genética , RNA Viral/metabolismo , Plântula/virologia , Vírion , Montagem de Vírus
6.
J Biol Chem ; 288(28): 20758-67, 2013 Jul 12.
Artigo em Inglês | MEDLINE | ID: mdl-23709221

RESUMO

Previous studies have identified two salt bridges in human CFTR chloride ion channels, Arg(352)-Asp(993) and Arg(347)-Asp(924), that are required for normal channel function. In the present study, we determined how the two salt bridges cooperate to maintain the open pore architecture of CFTR. Our data suggest that Arg(347) not only interacts with Asp(924) but also interacts with Asp(993). The tripartite interaction Arg(347)-Asp(924)-Asp(993) mainly contributes to maintaining a stable s2 open subconductance state. The Arg(352)-Asp(993) salt bridge, in contrast, is involved in stabilizing both the s2 and full (f) open conductance states, with the main contribution being to the f state. The s1 subconductance state does not require either salt bridge. In confirmation of the role of Arg(352) and Asp(993), channels bearing cysteines at these sites could be latched into a full open state using the bifunctional cross-linker 1,2-ethanediyl bismethanethiosulfonate, but only when applied in the open state. Channels remained latched open even after washout of ATP. The results suggest that these interacting residues contribute differently to stabilizing the open pore in different phases of the gating cycle.


Assuntos
Arginina/metabolismo , Ácido Aspártico/metabolismo , Regulador de Condutância Transmembrana em Fibrose Cística/metabolismo , Ativação do Canal Iônico , Trifosfato de Adenosina/farmacologia , Algoritmos , Sequência de Aminoácidos , Animais , Arginina/genética , Ácido Aspártico/genética , Sítios de Ligação/genética , Cloretos/farmacologia , Regulador de Condutância Transmembrana em Fibrose Cística/genética , Feminino , Humanos , Cinética , Potenciais da Membrana/efeitos dos fármacos , Potenciais da Membrana/fisiologia , Mutação , Oócitos/metabolismo , Oócitos/fisiologia , Técnicas de Patch-Clamp , RNA Complementar/genética , Xenopus laevis
7.
Inorg Chem ; 51(7): 4066-75, 2012 Apr 02.
Artigo em Inglês | MEDLINE | ID: mdl-22432748

RESUMO

Hard-ligand, high-potential copper sites have been characterized in double mutants of Pseudomonas aeruginosa azurin (C112D/M121X (X = L, F, I)). These sites feature a small A(zz)(Cu) splitting in the EPR spectrum together with enhanced electron transfer activity. Due to these unique properties, these constructs have been called "type zero" copper sites. In contrast, the single mutant, C112D, features a large A(zz)(Cu) value characteristic of the typical type 2 Cu(II). In general, A(zz)(Cu) comprises contributions from Fermi contact, spin dipolar, and orbital dipolar terms. In order to understand the origin of the low A(zz)(Cu) value of type zero Cu(II), we explored in detail its degree of covalency, as manifested by spin delocalization over its ligands, which affects A(zz)(Cu) through the Fermi contact and spin dipolar contributions. This was achieved by the application of several complementary EPR hyperfine spectroscopic techniques at X- and W-band (∼9.5 and 95 GHz, respectively) frequencies to map the ligand hyperfine couplings. Our results show that spin delocalization over the ligands in type zero Cu(II) is different from that of type 2 Cu(II) in the single C112D mutant. The (14)N hyperfine couplings of the coordinated histidine nitrogens are smaller by about 25-40%, whereas that of the (13)C carboxylate of D112 is about 50% larger. From this comparison, we concluded that the spin delocalization of type zero copper over its ligands is not dramatically larger than in type 2 C112D. Therefore, the reduced A(zz)(Cu) value of type zero Cu(II) is largely attributable to an increased orbital dipolar contribution that is related to its larger g(zz) value, as a consequence of the distorted tetrahedral geometry. The increased spin delocalization over the D112 carboxylate in type zero mutants compared to type 2 C112D suggests that electron transfer paths involving this residue are enhanced.


Assuntos
Azurina/química , Cobre/química , Elétrons , Pseudomonas aeruginosa/química , Ácido Aspártico/química , Ácido Aspártico/genética , Azurina/genética , Azurina/metabolismo , Cisteína/química , Cisteína/genética , Espectroscopia de Ressonância de Spin Eletrônica , Transporte de Elétrons , Escherichia coli , Histidina/química , Leucina/química , Leucina/genética , Ligantes , Espectroscopia de Ressonância Magnética , Metionina/química , Metionina/genética , Modelos Moleculares , Mutação , Oxirredução , Pseudomonas aeruginosa/metabolismo , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo
8.
Proc Natl Acad Sci U S A ; 107(17): 7987-92, 2010 Apr 27.
Artigo em Inglês | MEDLINE | ID: mdl-20385800

RESUMO

Proper regulation of neurotransmission requires that ligand-activated ion channels remain closed until agonist binds. How channels then open remains poorly understood. Glycine receptor (GlyR) gating is initiated by agonist binding at interfaces between adjacent subunits in the extracellular domain. Aspartate-97, located at the alpha1 GlyR interface, is a conserved residue in the cys-loop receptor superfamily. The mutation of D97 to arginine (D97R) causes spontaneous channel opening, with open and closed dwell times similar to those of maximally activated WT GlyR. Using a model of the N-terminal domain of the alpha1 GlyR, we hypothesized that an arginine-119 residue was forming intersubunit electrostatic bonds with D97. The D97R/R119E charge reversal restored this interaction, stabilizing channels in their closed states. Cysteine substitution shows that this link occurs between adjacent subunits. This intersubunit electrostatic interaction among GlyR subunits thus contributes to the stabilization of the closed channel state, and its disruption represents a critical step in GlyR activation.


Assuntos
Ativação do Canal Iônico/fisiologia , Modelos Moleculares , Receptores de Glicina/metabolismo , Transmissão Sináptica/fisiologia , Sequência de Aminoácidos , Animais , Arginina/metabolismo , Ácido Aspártico/genética , Ácido Aspártico/metabolismo , Sequência de Bases , DNA Complementar/genética , Eletrofisiologia , Dados de Sequência Molecular , Mutação/genética , Técnicas de Patch-Clamp , Receptores de Glicina/química , Receptores de Glicina/genética , Análise de Sequência de DNA , Eletricidade Estática , Xenopus
9.
Mol Cancer Ther ; 9(3): 673-81, 2010 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-20197397

RESUMO

K-ras is the most frequently mutated oncogene in non-small cell lung cancer (NSCLC), the most common form of lung cancer. Recent studies indicate that NSCLC patients with mutant K-ras do not respond to epidermal growth factor receptor inhibitors. In the attempt to find alternative therapeutic regimes for such patients, we tested PHA-848125, an oral pan cyclin-dependent kinase inhibitor currently under evaluation in phase II clinical trial, on a transgenic mouse model, K-Ras(G12D)LA2, which develops pulmonary cancerous lesions reminiscent of human lung adenocarcinomas. We used magnetic resonance imaging and positron emission tomography to follow longitudinally disease progression and evaluate therapeutic efficacy in this model. Treatment of K-Ras(G12D)LA2 mice with 40 mg/kg twice daily for 10 days with PHA-848125 induced a significant tumor growth inhibition at the end of treatment (P < 0.005) and this was accompanied by a reduction in the cell membrane turnover, as seen by 11C-Choline-positron emission tomography (P < 0.05). Magnetic resonance imaging data were validated versus histology and the mechanism of action of the compound was verified by immunohistochemistry, using cyclin-dependent kinase-related biomarkers phospho-Retinoblastoma and cyclin A. In this study, multimodality imaging was successfully used for the preclinical assessment of PHA-848125 therapeutic efficacy on a lung adenocarcinoma mouse model. This compound induced a volumetric and metabolic anticancer effect and could represent a valid therapeutic approach for NSCLC patients with mutant K-ras.


Assuntos
Adenocarcinoma/tratamento farmacológico , Antineoplásicos/uso terapêutico , Quinases Ciclina-Dependentes/antagonistas & inibidores , Diagnóstico por Imagem/métodos , Neoplasias Pulmonares/tratamento farmacológico , Pirazóis/uso terapêutico , Quinazolinas/uso terapêutico , Adenocarcinoma/diagnóstico , Adenocarcinoma/genética , Adenocarcinoma/patologia , Substituição de Aminoácidos/genética , Substituição de Aminoácidos/fisiologia , Animais , Antineoplásicos/farmacologia , Ácido Aspártico/genética , Diagnóstico por Imagem/veterinária , Modelos Animais de Doenças , Avaliação Pré-Clínica de Medicamentos , Genes ras , Glicina/genética , Humanos , Neoplasias Pulmonares/diagnóstico , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/patologia , Camundongos , Camundongos Transgênicos , Proteínas Mutantes/genética , Pirazóis/farmacologia , Quinazolinas/farmacologia , Resultado do Tratamento
10.
Nature ; 462(7274): 808-12, 2009 Dec 10.
Artigo em Inglês | MEDLINE | ID: mdl-20010690

RESUMO

Mistranslation arising from confusion of serine for alanine by alanyl-tRNA synthetases (AlaRSs) has profound functional consequences. Throughout evolution, two editing checkpoints prevent disease-causing mistranslation from confusing glycine or serine for alanine at the active site of AlaRS. In both bacteria and mice, Ser poses a bigger challenge than Gly. One checkpoint is the AlaRS editing centre, and the other is from widely distributed AlaXps-free-standing, genome-encoded editing proteins that clear Ser-tRNA(Ala). The paradox of misincorporating both a smaller (glycine) and a larger (serine) amino acid suggests a deep conflict for nature-designed AlaRS. Here we show the chemical basis for this conflict. Nine crystal structures, together with kinetic and mutational analysis, provided snapshots of adenylate formation for each amino acid. An inherent dilemma is posed by constraints of a structural design that pins down the alpha-amino group of the bound amino acid by using an acidic residue. This design, dating back more than 3 billion years, creates a serendipitous interaction with the serine OH that is difficult to avoid. Apparently because no better architecture for the recognition of alanine could be found, the serine misactivation problem was solved through free-standing AlaXps, which appeared contemporaneously with early AlaRSs. The results reveal unconventional problems and solutions arising from the historical design of the protein synthesis machinery.


Assuntos
Alanina-tRNA Ligase/metabolismo , Alanina/metabolismo , Escherichia coli/enzimologia , Biossíntese de Proteínas , Serina/metabolismo , Alanina-tRNA Ligase/química , Alanina-tRNA Ligase/genética , Ácido Aspártico/genética , Ácido Aspártico/metabolismo , Domínio Catalítico , Cristalização , Cinética , Modelos Moleculares , Mutação , Conformação Proteica , RNA de Transferência de Alanina/metabolismo , Relação Estrutura-Atividade
11.
Biochem Biophys Res Commun ; 388(2): 450-5, 2009 Oct 16.
Artigo em Inglês | MEDLINE | ID: mdl-19679105

RESUMO

The cytoplasmic tail of the amyloid precursor protein (APP) contains two putatively cytotoxic peptides, Jcasp and C31, derived by caspase cleavage of APP. Jcasp is a fragment starting from the epsilon-secretase site to position 664, while C31 is a fragment from position 665 to the C-terminus. Our studies now showed that compared to C31, Jcasp appeared to play a minor role in cytotoxicity. In particular, inhibition of Jcasp generation by treatment of gamma-secretase inhibitor did not lead to any attenuation of C31-induced toxicity. Secondly, because C31 toxicity is largely absent in cells lacking endogenous APP, we determined, using a split beta-galactosidase complementary assay to monitor protein-protein interactions, the presence of APP associated complexes. Our results demonstrated that both APP homomeric and C31/APP heteromeric complexes were correlated with cell death, indicating that C31 complexes with APP to recruit the interacting partners that initiate the signals related to cellular toxicity.


Assuntos
Precursor de Proteína beta-Amiloide/biossíntese , Citotoxinas/biossíntese , Fragmentos de Peptídeos/biossíntese , Doença de Alzheimer/metabolismo , Sequência de Aminoácidos , Secretases da Proteína Precursora do Amiloide/antagonistas & inibidores , Secretases da Proteína Precursora do Amiloide/metabolismo , Precursor de Proteína beta-Amiloide/genética , Animais , Apoptose , Ácido Aspártico/genética , Linhagem Celular Tumoral , Citotoxinas/genética , Dipeptídeos/farmacologia , Camundongos , Dados de Sequência Molecular , Fragmentos de Peptídeos/genética , Multimerização Proteica , Estrutura Terciária de Proteína , Ratos
12.
Biochemistry ; 48(39): 9266-77, 2009 Oct 06.
Artigo em Inglês | MEDLINE | ID: mdl-19705835

RESUMO

The chemoreceptors of Escherichia coli and Salmonella typhimurium form stable oligomers that associate with the coupling protein CheW and the histidine kinase CheA to form an ultrasensitive, ultrastable signaling lattice. Attractant binding to the periplasmic domain of a given receptor dimer triggers a transmembrane conformational change transmitted through the receptor to its cytoplasmic kinase control module, a long four-helix bundle that binds and regulates CheA kinase. The kinase control module comprises three functional regions: the adaptation region possessing the receptor adaptation sites, a coupling region that transmits signals between other regions, and the protein interaction region possessing contact sites for receptor oligomerization and for CheA-CheW binding. On the basis of the spatial clustering of known signal locking Cys substitutions and engineered disulfide bonds, this study develops the yin-yang hypothesis for signal transmission through the kinase control module. This hypothesis proposes that signals are transmitted through the four-helix bundle via changes in helix-helix packing and that the helix packing changes in the adaptation and protein interaction regions are tightly and antisymmetrically coupled. Specifically, strong helix packing in the adaptation region stabilizes the receptor on state, while strong helix packing in the protein interaction region stabilizes the off state. To test the yin-yang hypothesis, conserved sockets likely to strengthen specific helix-helix contacts via knob-in-hole packing interactions were identified in the adaptation, coupling, and protein interaction regions. For 32 sockets, the knob side chain was truncated to Ala to weaken the knob-in-hole packing and thereby destabilize the local helix-helix interaction provided by that socket. We term this approach a "knob truncation scan". Of the 32 knob truncations, 28 yielded stable receptors. Functional analysis of the signaling state of these receptors revealed seven lock-off knob truncations, all located in the adaptation region, that trap the receptor in its "off" signaling state (low kinase activity, high methylation activity). Also revealed were five lock-on knob truncations, all located in the protein interaction region, that trap the "on" state (high kinase activity, low methylation activity). These findings provide strong evidence that a yin-yang coupling mechanism generates concerted, antisymmetric helix-helix packing changes within the adaptation and protein interaction regions during receptor on-off switching. Conserved sockets that stabilize local helix-helix interactions play a central role in this mechanism: in the on state, sockets are formed in the adaptation region and disrupted in the protein interaction region, while the opposite is true in the off state.


Assuntos
Proteínas de Bactérias/química , Quimiotaxia , Proteínas de Escherichia coli/química , Proteínas de Membrana/química , Modelos Químicos , Mutação Puntual , Mapeamento de Interação de Proteínas/métodos , Receptores de Aminoácido/química , Transdução de Sinais/fisiologia , Motivos de Aminoácidos/genética , Ácido Aspártico/genética , Ácido Aspártico/metabolismo , Proteínas de Bactérias/genética , Proteínas de Bactérias/metabolismo , Quimiotaxia/genética , Proteínas de Escherichia coli/genética , Proteínas de Escherichia coli/fisiologia , Histidina Quinase , Proteínas de Membrana/genética , Proteínas de Membrana/metabolismo , Proteínas Quimiotáticas Aceptoras de Metil , Metiltransferases/química , Mutagênese Sítio-Dirigida , Conformação Proteica , Estrutura Secundária de Proteína/genética , Receptores de Aminoácido/genética , Receptores de Aminoácido/fisiologia , Salmonella typhimurium/química , Salmonella typhimurium/genética , Transdução de Sinais/genética
13.
Life Sci ; 85(7-8): 339-44, 2009 Aug 12.
Artigo em Inglês | MEDLINE | ID: mdl-19580817

RESUMO

AIMS: The study was designed to examine the internalization of Asp104Lys mutant of beta(1)-adrenergic receptor (beta(1)-AR) and compared to other mutant (Asp104Ala) and wild type receptors. Moreover, this study needs to perform the role of GRK2 (betaARK1) and beta-arrestin1 on this internalization of Asp104Lys mutant of beta(1)-AR. MAIN METHODS: Binding affinity, functional potency of agonist and agonist-induced internalization were determined for wild type and both mutants of beta(1)-ARs stably expressed in HEK 293 cells as assessed by [(3)H] CGP12177 radioligand. We have performed GRK2 and beta-arrestin1 expression levels by western blot analysis and also performed internalization of this mutant receptor after over expression and deletion of beta-arrestin1 gene. KEY FINDINGS: In the present study, the binding affinity of (-)-isoproterenol for both mutants were significantly decreased compared to wild type. Though the mutant Asp104Ala showed agonist-induced receptor activation, interestingly this mutant was not internalized. However, the mutant Asp104Lys, which showed uncoupling with G protein, was internalized 31.77+/-3.13% from cell surface. Asp104Lys mutant produced the same level of GRK2 expression in (-)-isoproterenol induced stimulation of wild type receptor and addition of (-)-isoproterenol further increased GRK2 expression in mutant receptors. In addition, overexpression of beta-arrestin1 in mutant Asp104Lys promoted (39.75+/-2.19%) and knockdown of beta-arrestin1 by siRNA decreased (3.55+/-1.75%) internalization compared to Asp104Lys mutant of beta(1)-ARs. SIGNIFICANCE: The present studies suggest that Asp104Lys mutant beta(1)-ARs triggers unconventional homologous internalization induced by G protein independent signals, where GRK2 and beta-arrestin1 play an important role for beta(1)-AR internalization.


Assuntos
Ácido Aspártico/genética , Lisina/genética , Mutação , Receptores Adrenérgicos beta 1/metabolismo , Agonistas Adrenérgicos beta/metabolismo , Antagonistas Adrenérgicos beta/metabolismo , Ligação Competitiva , Western Blotting , Linhagem Celular , Membrana Celular/metabolismo , AMP Cíclico/metabolismo , Humanos , Isoproterenol/metabolismo , Ligantes , Mutagênese Sítio-Dirigida , Propranolol/metabolismo , Ensaio Radioligante , Receptores Adrenérgicos beta 1/genética , Transfecção
14.
J Biol Chem ; 284(10): 6530-5, 2009 Mar 06.
Artigo em Inglês | MEDLINE | ID: mdl-19116204

RESUMO

The single nucleotide polymorphism 118A>G of the human micro-opioid receptor gene OPRM1, which leads to an exchange of the amino acid asparagine (N) to aspartic acid (D) at position 40 of the extracellular receptor region, alters the in vivo effects of opioids to different degrees in pain-processing brain regions. The most pronounced N40D effects were found in brain regions involved in the sensory processing of pain intensity. Using the mu-opioid receptor-specific agonist DAMGO, we analyzed the micro-opioid receptor signaling, expression, and binding affinity in human brain tissue sampled postmortem from the secondary somatosensory area (SII) and from the ventral posterior part of the lateral thalamus, two regions involved in the sensory processing and transmission of nociceptive information. We show that the main effect of the N40D micro-opioid receptor variant is a reduction of the agonist-induced receptor signaling efficacy. In the SII region of homo- and heterozygous carriers of the variant 118G allele (n=18), DAMGO was only 62% as efficient (p=0.002) as in homozygous carriers of the wild-type 118A allele (n=15). In contrast, the number of [3H]DAMGO binding sites was unaffected. Hence, the micro-opioid receptor G-protein coupling efficacy in SII of carriers of the 118G variant was only 58% as efficient as in homozygous carriers of the 118A allele (p<0.001). The thalamus was unaffected by the OPRM1 118A>G SNP. In conclusion, we provide a molecular basis for the reduced clinical effects of opioid analgesics in carriers of mu-opioid receptor variant N40D.


Assuntos
Substituição de Aminoácidos , Dor/metabolismo , Polimorfismo de Nucleotídeo Único , Receptores Opioides mu/metabolismo , Córtex Somatossensorial/metabolismo , Tálamo/metabolismo , Alelos , Analgésicos Opioides/farmacologia , Asparagina/genética , Asparagina/metabolismo , Ácido Aspártico/genética , Ácido Aspártico/metabolismo , Ala(2)-MePhe(4)-Gly(5)-Encefalina/farmacologia , Regulação da Expressão Gênica/efeitos dos fármacos , Regulação da Expressão Gênica/fisiologia , Homozigoto , Humanos , Dor/genética , Estrutura Terciária de Proteína , Receptores Opioides mu/agonistas , Receptores Opioides mu/genética
15.
FEBS J ; 275(6): 1131-9, 2008 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-18248458

RESUMO

The lugworm Arenicola marina inhabits marine sediments in which sulfide concentrations can reach up to 2 mM. Although sulfide is a potent toxin for humans and most animals, because it inhibits mitochondrial cytochrome c oxidase at micromolar concentrations, A. marina can use electrons from sulfide for mitochondrial ATP production. In bacteria, electron transfer from sulfide to quinone is catalyzed by the membrane-bound flavoprotein sulfide : quinone oxidoreductase (SQR). A cDNA from A. marina was isolated and expressed in Saccharomyces cerevisiae, which lacks endogenous SQR. The heterologous enzyme was active in mitochondrial membranes. After affinity purification, Arenicola SQR isolated from yeast mitochondria reduced decyl-ubiquinone (K(m) = 6.4 microm) after the addition of sulfide (K(m) = 23 microm) only in the presence of cyanide (K(m) = 2.6 mM). The end product of the reaction was thiocyanate. When cyanide was substituted by Escherichia coli thioredoxin and sulfite, SQR exhibited one-tenth of the cyanide-dependent activity. Six amino acids known to be essential for bacterial SQR were exchanged by site-directed mutagenesis. None of the mutant enzymes was active after expression in yeast, implicating these amino acids in the catalytic mechanism of the eukaryotic enzyme.


Assuntos
Cianetos/química , Poliquetos/enzimologia , Quinona Redutases/química , Tiorredoxinas/química , Sequência de Aminoácidos , Animais , Ácido Aspártico/química , Ácido Aspártico/genética , Catálise , DNA Complementar/genética , Mitocôndrias/enzimologia , Membranas Mitocondriais/enzimologia , Dados de Sequência Molecular , Mutação , Quinona Redutases/biossíntese , Quinona Redutases/genética , Proteínas Recombinantes/biossíntese , Proteínas Recombinantes/química , Saccharomyces cerevisiae/genética , Sulfetos/química , Tiocianatos/análise , Tiossulfatos/metabolismo
16.
Appl Environ Microbiol ; 73(18): 5789-96, 2007 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-17660309

RESUMO

We investigated the carbon dioxide metabolism of Streptococcus thermophilus, evaluating the phenotype of a phosphoenolpyruvate carboxylase-negative mutant obtained by replacement of a functional ppc gene with a deleted and inactive version, Deltappc. The growth of the mutant was compared to that of the parent strain in a chemically defined medium and in milk, supplemented or not with L-aspartic acid, the final product of the metabolic pathway governed by phosphoenolpyruvate carboxylase. It was concluded that aspartate present in milk is not sufficient for the growth of S. thermophilus. As a consequence, phosphoenolpyruvate carboxylase activity was considered fundamental for the biosynthesis of L-aspartic acid in S. thermophilus metabolism. This enzymatic activity is therefore essential for growth of S. thermophilus in milk even if S. thermophilus was cultured in association with proteinase-positive Lactobacillus delbrueckii subsp. bulgaricus. It was furthermore observed that the supplementation of milk with aspartate significantly affected the level of urease activity. Further experiments, carried out with a p(ureI)-gusA recombinant strain, revealed that expression of the urease operon was sensitive to the aspartate concentration in milk and to the cell availability of glutamate, glutamine, and ammonium ions.


Assuntos
Ácido Aspártico/biossíntese , Genes Bacterianos , Leite/microbiologia , Streptococcus thermophilus/metabolismo , Streptococcus/genética , Urease/genética , Animais , Ácido Aspártico/genética , Microbiologia de Alimentos , Regulação Bacteriana da Expressão Gênica , Redes e Vias Metabólicas , Streptococcus/enzimologia , Streptococcus/crescimento & desenvolvimento , Iogurte/microbiologia
17.
Acta Crystallogr D Biol Crystallogr ; 63(Pt 8): 906-22, 2007 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-17642517

RESUMO

The protonation states of aspartic acids and glutamic acids as well as histidine are investigated in four X-ray cases: Ni,Ca concanavalin A at 0.94 A, a thrombin-hirugen binary complex at 1.26 A resolution and two thrombin-hirugen-inhibitor ternary complexes at 1.32 and 1.39 A resolution. The truncation of the Ni,Ca concanavalin A data at various test resolutions between 0.94 and 1.50 A provided a test comparator for the ;unknown' thrombin-hirugen carboxylate bond lengths. The protonation states of aspartic acids and glutamic acids can be determined (on the basis of convincing evidence) even to the modest resolution of 1.20 A as exemplified by our X-ray crystal structure refinements of Ni and Mn concanavalin A and also as indicated in the 1.26 A structure of thrombin, both of which are reported here. The protonation-state indication of an Asp or a Glu is valid provided that the following criteria are met (in order of importance). (i) The acidic residue must have a single occupancy. (ii) Anisotropic refinement at a minimum diffraction resolution of 1.20 A (X-ray data-to-parameter ratio of approximately 3.5:1) is required. (iii) Both of the bond lengths must agree with the expectation (i.e. dictionary values), thus allowing some relaxation of the bond-distance standard uncertainties required to approximately 0.025 A for a '3sigma' determination or approximately 0.04 A for a '2sigma' determination, although some variation of the expected bond-distance values must be allowed according to the microenvironment of the hydrogen of interest. (iv) Although the F(o) - F(c) map peaks are most likely to be unreliable at the resolution range around 1.20 A, if admitted as evidence the peak at the hydrogen position must be greater than or equal to 2.5 sigma and in the correct geometry. (v) The atomic B factors need to be less than 10 A(2) for bond-length differentiation; furthermore, the C=O bond can also be expected to be observed with continuous 2F(o) - F(c) electron density and the C-OH bond with discontinuous electron density provided that the atomic B factors are less than approximately 20 A(2) and the contour level is increased. The final decisive option is to carry out more than one experiment, e.g. multiple X-ray crystallography experiments and ideally neutron crystallography. The complementary technique of neutron protein crystallography has provided evidence of the protonation states of histidine and acidic residues in concanavalin A and also the correct orientations of asparagine and glutamine side chains. Again, the truncation of the neutron data at various test resolutions between 2.5 and 3.0 A, even 3.25 and 3.75 A resolution, examines the limits of the neutron probe. These various studies indicate a widening of the scope of both X-ray and neutron probes in certain circumstances to elucidate the protonation states in proteins.


Assuntos
Proteínas/química , Prótons , Ácido Aspártico/química , Ácido Aspártico/genética , Ácido Aspártico/metabolismo , Concanavalina A/química , Concanavalina A/genética , Concanavalina A/metabolismo , Cristalografia por Raios X , Ácido Glutâmico/química , Ácido Glutâmico/genética , Ácido Glutâmico/metabolismo , Hirudinas/química , Hirudinas/genética , Hirudinas/metabolismo , Histidina/química , Histidina/genética , Histidina/metabolismo , Ligação de Hidrogênio , Modelos Moleculares , Fragmentos de Peptídeos/química , Fragmentos de Peptídeos/genética , Fragmentos de Peptídeos/metabolismo , Estrutura Terciária de Proteína , Proteínas/genética , Proteínas/metabolismo , Trombina/química , Trombina/genética , Trombina/metabolismo
18.
Biochemistry ; 43(46): 14821-31, 2004 Nov 23.
Artigo em Inglês | MEDLINE | ID: mdl-15544353

RESUMO

Fumagillin, an angiogenic inhibitor, binds to methionine aminopeptidase 2, which is the same as eukaryotic initiation factor 2-associated glycoprotein, p67. p67 protects eIF2alpha from phosphorylation by its kinases. To understand the importance of fumagillin binding to p67, we measured the level of p67 in mouse C2C12 myoblasts treated with fumagillin. We show that fumagillin increases the stability of p67 by decreasing its turnover rate. The increased levels of p67 result in inhibition of phosphorylation of extracellular signal-regulated kinases 1 and 2 (ERKs 1 and 2). p67 binds to these ERKs, and the 108-480 amino acid segment is sufficient for this binding. p67's affinity to ERKs 1 and 2 also increases in fumagillin-treated myoblasts while its affinity for eIF2alpha remains unchanged. A mutant at the conserved amino acid residue D251A increases the phosphorylation of ERKs 1 and 2 without affecting the binding to p67, thus indicating the importance of this residue in the regulation of the phosphorylation of these ERKs. These results suggest that fumagillin increases the stability of p67 and its affinity to ERKs 1 and 2 and causes the inhibition of the phosphorylation of ERKs 1 and 2.


Assuntos
Aminopeptidases/metabolismo , Inibidores da Angiogênese/farmacologia , Fator de Iniciação 2 em Eucariotos/metabolismo , Ácidos Graxos Insaturados/farmacologia , Glicoproteínas/metabolismo , Proteína Quinase 1 Ativada por Mitógeno/metabolismo , Proteína Quinase 3 Ativada por Mitógeno/metabolismo , Alanina/genética , Substituição de Aminoácidos/genética , Aminopeptidases/biossíntese , Aminopeptidases/genética , Inibidores da Angiogênese/metabolismo , Animais , Ácido Aspártico/genética , Linhagem Celular , Cicloexanos , Inibidores Enzimáticos/metabolismo , Estabilidade Enzimática/efeitos dos fármacos , Estabilidade Enzimática/genética , Ácidos Graxos Insaturados/metabolismo , Feminino , Glicoproteínas/biossíntese , Glicoproteínas/genética , Concentração Inibidora 50 , Metionil Aminopeptidases , Camundongos , Camundongos Endogâmicos BALB C , Proteína Quinase 1 Ativada por Mitógeno/antagonistas & inibidores , Proteína Quinase 3 Ativada por Mitógeno/antagonistas & inibidores , Mioblastos Esqueléticos/efeitos dos fármacos , Mioblastos Esqueléticos/enzimologia , Mioblastos Esqueléticos/metabolismo , Fragmentos de Peptídeos/metabolismo , Fosforilação/efeitos dos fármacos , Ligação Proteica/efeitos dos fármacos , Ligação Proteica/genética , Sesquiterpenos , Transfecção
19.
J Neurol ; 251(6): 715-24, 2004 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-15311348

RESUMO

Fatal familial insomnia (FFI) is a prion disease exhibiting the PRNP D178N/129M genotype. Features of this autosomal dominant illness are progressive insomnia, dysautonomia, myoclonus, cognitive decline and motor signs associated with thalamic nerve cell loss and gliosis. In contrast to the new variant of Creutzfeldt-Jakob disease (vCJD) the onset of FFI is in middle to late adulthood. We report two male patients who belong to a large German FFI kindred. They were examined clinically, and postmortem neuropathological examination was carried out in collaboration with the German reference centre for prion disease. Additionally, the prion protein gene (PRNP) was analysed. To identify further patients with disease onset under 30 years of age a comprehensive literature review was carried out. Two male patients presented with typical symptoms of FFI at the age of 23 and 24 years. In their kindred, the age of onset has never before been under 44 years of age. Our literature review identified five additional early onset cases who died at age 21 to 25 years. In all 22 reviewed FFI families the median manifestation age was 49.5 years. Although phenotypic variability of FFI is common, age of onset under 30 years has been considered to be a hallmark of vCJD with a mean manifestation at 27 years of age. Our findings underline that in addition to vCJD, FFI must be considered in cases of young-onset prion disease. This has considerable impact on clinical management and genetic counselling.


Assuntos
Saúde da Família , Insônia Familiar Fatal/genética , Insônia Familiar Fatal/fisiopatologia , Adulto , Idade de Início , Asparagina/genética , Ácido Aspártico/genética , Análise Mutacional de DNA/métodos , Aconselhamento Genético/métodos , Glucose/metabolismo , Humanos , Imuno-Histoquímica/métodos , Insônia Familiar Fatal/metabolismo , Insônia Familiar Fatal/patologia , Masculino , Metionina/metabolismo , Pessoa de Meia-Idade , Exame Neurológico , Linhagem , Mudanças Depois da Morte , Príons/genética , Príons/metabolismo , Literatura de Revisão como Assunto , Tálamo/metabolismo , Tálamo/patologia , Tomografia Computadorizada de Emissão/métodos
20.
Hum Mutat ; 22(6): 496-7, 2003 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-14635113

RESUMO

In a patient with fatal neonatal lactic acidosis due to pyruvate dehydrogenase deficiency, the only potential mutation detected was c.888C>G in PDHA1, the gene for the E1alpha subunit of the complex. This would result in a substitution of glutamate for aspartate (D296E). Pathogenicity of this minor alteration in amino acid sequence was demonstrated by expression studies. By comparing the mutant sequence with the known structures of the E1 components of pyruvate dehydrogenase and the closely related branched chain alpha-ketoacid dehydrogenase, an explanation for the profound consequences of the mutation can be proposed.


Assuntos
Substituição de Aminoácidos/genética , Piruvato Desidrogenase (Lipoamida)/genética , Doença da Deficiência do Complexo de Piruvato Desidrogenase/genética , Ácido Aspártico/genética , Domínio Catalítico/genética , Análise Mutacional de DNA , DNA Complementar/química , DNA Complementar/genética , Evolução Fatal , Feminino , Fibroblastos/enzimologia , Fibroblastos/metabolismo , Ácido Glutâmico/genética , Humanos , Recém-Nascido , Masculino , Modelos Moleculares , Mutação , Piruvato Desidrogenase (Lipoamida)/química , Piruvato Desidrogenase (Lipoamida)/metabolismo , Doença da Deficiência do Complexo de Piruvato Desidrogenase/patologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA