Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 41
Filtrar
Mais filtros








Base de dados
Intervalo de ano de publicação
1.
Nat Commun ; 10(1): 5579, 2019 12 06.
Artigo em Inglês | MEDLINE | ID: mdl-31811120

RESUMO

Although influenza viruses lead to severe illness in high-risk populations, host genetic factors associated with severe disease are largely unknown. As the HLA-A*68:01 allele can be linked to severe pandemic 2009-H1N1 disease, we investigate a potential impairment of HLA-A*68:01-restricted CD8+ T cells to mount robust responses. We elucidate the HLA-A*68:01+CD8+ T cell response directed toward an extended influenza-derived nucleoprotein (NP) peptide and show that only ~35% individuals have immunodominant A68/NP145+CD8+ T cell responses. Dissecting A68/NP145+CD8+ T cells in low vs. medium/high responders reveals that high responding donors have A68/NP145+CD8+ memory T cells with clonally expanded TCRαßs, while low-responders display A68/NP145+CD8+ T cells with predominantly naïve phenotypes and non-expanded TCRαßs. Single-cell index sorting and TCRαß analyses link expansion of A68/NP145+CD8+ T cells to their memory potential. Our study demonstrates the immunodominance potential of influenza-specific CD8+ T cells presented by a risk HLA-A*68:01 molecule and advocates for priming CD8+ T cell compartments in HLA-A*68:01-expressing individuals for establishment of pre-existing protective memory T cell pools.


Assuntos
Linfócitos T CD8-Positivos/imunologia , Antígenos HLA-A/imunologia , Antígenos HLA-A/metabolismo , Vírus da Influenza A/imunologia , Influenza Humana/imunologia , Apresentação de Antígeno , Antígenos Virais/química , Linhagem Celular , Proteção Cruzada , Reações Cruzadas/imunologia , Epitopos de Linfócito T/imunologia , Antígenos HLA-A/química , Antígenos HLA-A/genética , Humanos , Memória Imunológica/imunologia , Vírus da Influenza A Subtipo H1N1/imunologia , Modelos Moleculares , Nucleoproteínas/química , Orthomyxoviridae/genética , Orthomyxoviridae/imunologia , Fragmentos de Peptídeos/química , Fenótipo , Conformação Proteica , Receptores de Antígenos de Linfócitos T alfa-beta/metabolismo , Proteínas do Core Viral/genética
2.
J Neuroimmunol ; 332: 167-175, 2019 07 15.
Artigo em Inglês | MEDLINE | ID: mdl-31048269

RESUMO

Following the 2009 H1N1 influenza pandemic, an increased risk of narcolepsy type 1 was observed. Homology between an H1N1 hemagglutinin and two hypocretin sequences has been reported. T cell reactivity to these peptides was assessed in 81 narcolepsy type 1 patients and 19 HLA-DQ6-matched healthy controls. HLA-DQ6-restricted H1N1 hemagglutinin-specific T cell responses were detected in 28.4% of patients and 15.8% of controls. Despite structural homology between HLA-DQ6-hypocretin and -H1N1 peptide complexes, T cell cross-reactivity was not detected. These results indicate that it is unlikely that cross-reactivity between H1N1 hemagglutinin and hypocretin peptides presented by HLA-DQ6 is involved in the development of narcolepsy.


Assuntos
Linfócitos T CD4-Positivos/imunologia , Antígenos HLA-DQ/imunologia , Glicoproteínas de Hemaglutininação de Vírus da Influenza/imunologia , Narcolepsia/imunologia , Orexinas/imunologia , Subpopulações de Linfócitos T/imunologia , Adolescente , Adulto , Proteínas do Líquido Cefalorraquidiano/análise , Criança , Cristalografia por Raios X , Feminino , Antígenos HLA-DQ/química , Cadeias alfa de HLA-DQ/análise , Cadeias beta de HLA-DQ/análise , Glicoproteínas de Hemaglutininação de Vírus da Influenza/química , Humanos , Vírus da Influenza A Subtipo H1N1 , Masculino , Pessoa de Meia-Idade , Modelos Moleculares , Mimetismo Molecular , Narcolepsia/etiologia , Orexinas/líquido cefalorraquidiano , Orexinas/química , Pandemias , Fragmentos de Peptídeos/química , Fragmentos de Peptídeos/imunologia , Conformação Proteica , Adulto Jovem
3.
Mucosal Immunol ; 10(1): 58-68, 2017 01.
Artigo em Inglês | MEDLINE | ID: mdl-27143301

RESUMO

Despite recent breakthroughs in identifying mucosal-associated invariant T (MAIT) cell antigens (Ags), the precise requirements for in vivo MAIT cell responses to infection remain unclear. Using major histocompatibility complex-related protein 1 (MR1) tetramers, the MAIT cell response was investigated in a model of bacterial lung infection employing riboflavin gene-competent and -deficient bacteria. MAIT cells were rapidly enriched in the lungs of C57BL/6 mice infected with Salmonella Typhimurium, comprising up to 50% of αß-T cells after 1 week. MAIT cell accumulation was MR1-dependent, required Ag derived from the microbial riboflavin synthesis pathway, and did not occur in response to synthetic Ag, unless accompanied by a Toll-like receptor agonist or by co-infection with riboflavin pathway-deficient S. Typhimurium. The MAIT cell response was associated with their long-term accumulation in the lungs, draining lymph nodes and spleen. Lung MAIT cells from infected mice displayed an activated/memory phenotype, and most expressed the transcription factor retinoic acid-related orphan receptor γt. T-bet expression increased following infection. The majority produced interleukin-17 while smaller subsets produced interferon-γ or tumor necrosis factor, detected directly ex vivo. Thus the activation and expansion of MAIT cells coupled with their pro-inflammatory cytokine production occurred in response to Ags derived from microbial riboflavin synthesis and was augmented by co-stimulatory signals.


Assuntos
Antígenos de Histocompatibilidade Classe I/metabolismo , Pulmão/imunologia , Antígenos de Histocompatibilidade Menor/metabolismo , Mucosa/imunologia , Células T Matadoras Naturais/imunologia , Infecções por Salmonella/imunologia , Salmonella typhimurium/imunologia , Linfócitos T/imunologia , Animais , Antígenos de Bactérias/imunologia , Células Cultivadas , Receptores Coestimuladores e Inibidores de Linfócitos T/metabolismo , Interferon gama/metabolismo , Interleucina-17/metabolismo , Pulmão/microbiologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Membro 3 do Grupo F da Subfamília 1 de Receptores Nucleares/genética , Membro 3 do Grupo F da Subfamília 1 de Receptores Nucleares/metabolismo , Receptores de Antígenos de Linfócitos T alfa-beta/metabolismo , Riboflavina/biossíntese , Riboflavina/imunologia , Transdução de Sinais , Proteínas com Domínio T/genética , Proteínas com Domínio T/metabolismo , Linfócitos T/microbiologia , Fator de Necrose Tumoral alfa/metabolismo
4.
Mucosal Immunol ; 9(3): 583-6, 2016 05.
Artigo em Inglês | MEDLINE | ID: mdl-27007675

RESUMO

Celiac disease is an autoimmune-like disorder that is triggered by dietary gluten and has a strong genetic association with the human leukocyte antigen locus, specifically, HLA-DQ2.5/DQ8. Here, Dahai-Koirala et al. apply ex vivo single-cell sequencing of TCRs from celiac disease patients, and show that biased T-cell receptor usage underpins the response to two gluten epitopes, which has implications for disease pathogenesis, diagnosis, and treatment.


Assuntos
Doença Celíaca , Epitopos , Glutens , Humanos , Neuroglia , Receptores de Antígenos de Linfócitos T
5.
Tissue Antigens ; 78(1): 11-20, 2011 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-21501118

RESUMO

HLA-B57 and HLA-B58 are major histocompatibility class (MHC)-I allotypes that are potentially predictive of important clinical immune phenotypes. HLA-B*5701 is strongly associated with hypersensitivity to the HIV drug abacavir, liver toxicity from the antibiotic flucloxacillin and is a marker for slow progression of HIV AIDS. HLA-B*5801 is associated with hypersensitivity to allopurinol used to treat hyperuricaemia and recurrent gout. Here we describe a monoclonal antibody (mAb) specific for HLA-B57 and HLA-B58 that provides an inexpensive and sensitive screen for these MHC-I allotypes. The usefulness of HLA-B57 screening for prediction of abacavir hypersensitivity was shown in three independent laboratories, including confirmation of the mAb sensitivity and specificity in a cohort of patients enrolled in the PREDICT-1 trial. Our data show that patients who test negative by mAb screening comprise 90%-95% of all individuals in most human populations and require no further human leukocyte antigen (HLA) typing. Patients who test positive by mAb screening should proceed to high-resolution typing to ascertain the presence of HLA-B*5701 or HLA-B*5801. Hence, mAb screening provides a low-cost alternative to high-resolution typing of all patients and lends itself to point-of-care diagnostics and rapid ascertainment of low-risk patients who can begin immediate therapy with abacavir, flucloxacillin or allopurinol.


Assuntos
Hipersensibilidade a Drogas/prevenção & controle , Antígenos HLA-B/análise , Programas de Rastreamento/métodos , Sequência de Aminoácidos , Animais , Anticorpos Monoclonais/biossíntese , Anticorpos Monoclonais/isolamento & purificação , Anticorpos Monoclonais/metabolismo , Anticorpos Monoclonais/farmacologia , Formação de Anticorpos , Especificidade de Anticorpos , Células Cultivadas , Hipersensibilidade a Drogas/genética , Hipersensibilidade a Drogas/imunologia , Ensaio de Imunoadsorção Enzimática , Antígenos HLA-B/genética , Antígenos HLA-B/imunologia , Teste de Histocompatibilidade/métodos , Humanos , Camundongos , Camundongos Endogâmicos BALB C , Modelos Moleculares , Dados de Sequência Molecular , Fatores de Tempo
6.
Tissue Antigens ; 72(5): 415-24, 2008 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-18946929

RESUMO

The non-classical major histocompatibility complex (MHC) class I molecule human leucocyte antigen (HLA)-E is the least polymorphic of all the MHC class I molecules and acts as a ligand for receptors of both the innate and the adaptive immune systems. The recognition of self-peptides complexed to HLA-E by the CD94-NKG2A receptor expressed by natural killer (NK) cells represents a crucial checkpoint for immune surveillance by NK cells. However, HLA-E can also be recognised by the T-cell receptor expressed by alphabeta CD8 T cells and therefore can play a role in the adaptive immune response to invading pathogens. The recent resolution of HLA-E in complex with both innate and adaptive ligands has provided insight into the dual role of this molecule in immunity.


Assuntos
Linfócitos T CD8-Positivos/imunologia , Antígenos HLA/metabolismo , Antígenos de Histocompatibilidade Classe I/metabolismo , Células Matadoras Naturais/imunologia , Subfamília D de Receptores Semelhantes a Lectina de Células NK/metabolismo , Receptores de Antígenos de Linfócitos T/metabolismo , Linfócitos T CD8-Positivos/metabolismo , Antígenos HLA/química , Antígenos HLA/imunologia , Transplante de Células-Tronco Hematopoéticas , Antígenos de Histocompatibilidade Classe I/química , Antígenos de Histocompatibilidade Classe I/imunologia , Humanos , Imunidade Ativa/imunologia , Imunidade Inata/imunologia , Células Matadoras Naturais/metabolismo , Subfamília D de Receptores Semelhantes a Lectina de Células NK/imunologia , Polimorfismo Genético , Domínios e Motivos de Interação entre Proteínas/fisiologia , Receptores de Antígenos de Linfócitos T/imunologia , Receptores de Células Matadoras Naturais/imunologia , Receptores de Células Matadoras Naturais/metabolismo , Antígenos HLA-E
7.
J Biol Chem ; 276(45): 42138-45, 2001 Nov 09.
Artigo em Inglês | MEDLINE | ID: mdl-11533048

RESUMO

S-Nitrosoglutathione and the dinitrosyl-diglutathionyl iron complex are involved in the storage and transport of NO in biological systems. Their interactions with the human glutathione transferase P1-1 may reveal an additional physiological role for this enzyme. In the absence of GSH, S-nitrosoglutathione causes rapid and stable S-nitrosylation of both the Cys(47) and Cys(101) residues. Ion spray ionization-mass spectrometry ruled out the possibility of S-glutathionylation and confirms the occurrence of a poly-S-nitrosylation in GST P1-1. S-Nitrosylation of Cys(47) lowers the affinity 10-fold for GSH, but this negative effect is minimized by a half-site reactivity mechanism that protects one Cys(47)/dimer from nitrosylation. Thus, glutathione transferase P1-1, retaining most of its original activity, may act as a NO carrier protein when GSH depletion occurs in the cell. The dinitrosyl-diglutathionyl iron complex, which is formed by S-nitrosoglutathione decomposition in the presence of physiological concentrations of GSH and traces of ferrous ions, binds with extraordinary affinity to one active site of this dimeric enzyme (K(i) < 10(-12) m) and triggers negative cooperativity in the vacant subunit (K(i) = 10(-9) m). The complex bound to the enzyme is stable for hours, whereas in the free form and at low concentrations, its life time is only a few minutes. ESR and molecular modeling studies provide a reasonable explanation of this strong interaction, suggesting that Tyr(7) and enzyme-bound GSH could be involved in the coordination of the iron atom. All of the observed findings suggest that glutathione transferase P1-1, by means of an intersubunit communication, may act as a NO carrier under different cellular conditions while maintaining its well known detoxificating activity toward dangerous compounds.


Assuntos
Glutationa Transferase/fisiologia , Isoenzimas/fisiologia , Óxido Nítrico/metabolismo , Ligação Competitiva , Espectroscopia de Ressonância de Spin Eletrônica , Glutationa/metabolismo , Glutationa/farmacologia , Glutationa S-Transferase pi , Humanos , Ferro/metabolismo , Espectrometria de Massas , Óxidos de Nitrogênio/metabolismo , Ligação Proteica , S-Nitrosoglutationa/metabolismo , S-Nitrosoglutationa/farmacologia , Albumina Sérica/metabolismo
8.
Eur J Biochem ; 268(14): 4001-10, 2001 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-11453994

RESUMO

Methylobacterium dichloromethanicum DM4 is able to grow with dichloromethane as the sole carbon and energy source by using a dichloromethane dehalogenase/glutathione S-transferase (GST) for the conversion of dichloromethane to formaldehyde. Mammalian homologs of this bacterial enzyme are also known to catalyze this reaction. However, the dehalogenation of dichloromethane by GST T1-1 from rat was highly mutagenic and toxic to methylotrophic bacteria. Plasmid-driven expression of rat GST T1-1 in strain DM4-2cr, a mutant of strain DM4 lacking dichloromethane dehalogenase, reduced cell viability 10(5)-fold in the presence of dichloromethane. This effect was exploited to select dichloromethane-resistant transconjugants of strain DM4-2cr carrying a plasmid-encoded rGSTT1 gene. Transconjugants that still expressed the GST T1 protein after dichloromethane treatment included rGSTT1 mutants encoding protein variants with sequence changes from the wild-type ranging from single residue exchanges to large insertions and deletions. A structural model of rat GST T1-1 suggested that sequence variation was clustered around the glutathione activation site and at the protein C-terminus believed to cap the active site. The enzymatic activity of purified His-tagged GST T1-1 variants expressed in Escherichia coli was markedly reduced with both dichloromethane and the alternative substrate 1,2-epoxy-3-(4'-nitrophenoxy)propane. These results provide the first experimental evidence for the involvement of Gln102 and Arg107 in catalysis, and illustrate the potential of in vivo approaches to identify catalytic residues in GSTs whose activity leads to toxic effects.


Assuntos
Domínio Catalítico , Glutationa Transferase/genética , Glutationa Transferase/metabolismo , Cloreto de Metileno/metabolismo , Mutagênicos/metabolismo , Sequência de Aminoácidos , Animais , Variação Genética , Inativação Metabólica , Liases/metabolismo , Methylobacterium/enzimologia , Methylobacterium/genética , Modelos Moleculares , Dados de Sequência Molecular , Mutagênese , Mutação , Ratos , Proteínas Recombinantes , Seleção Genética , Homologia de Sequência de Aminoácidos
9.
J Biol Chem ; 276(11): 8261-8, 2001 Mar 16.
Artigo em Inglês | MEDLINE | ID: mdl-11102453

RESUMO

Perfringolysin O (PFO), a member of the cholesterol-dependent cytolysin family of pore-forming toxins, forms large oligomeric complexes comprising up to 50 monomers. In the present study, a disulfide bridge was introduced between cysteine-substituted serine 190 of transmembrane hairpin 1 (TMH1) and cysteine-substituted glycine 57 of domain 2 of PFO. The resulting disulfide-trapped mutant (PFO(C190-C57)) was devoid of hemolytic activity and could not insert either of its transmembrane beta-hairpins (TMHs) into the membrane unless the disulfide was reduced. Both the size of the oligomer formed on the membrane and its rate of formation were unaffected by the oxidation state of the Cys(190)-Cys(57) disulfide bond; thus, the disulfide-trapped PFO was assembled into a prepore complex on the membrane. The conversion of this prepore to the pore complex was achieved by reducing the C190-C57 disulfide bond. PFO(C190-C57) that was allowed to form the prepore prior to the reduction of the disulfide exhibited a dramatic increase in the rate of PFO-dependent hemolysis and the membrane insertion of its TMHs when compared with toxin that had the disulfide reduced prior mixing the toxin with membranes. Therefore, the rate-limiting step in pore formation is prepore assembly, not TMH insertion. These data demonstrate that the prepore is a legitimate intermediate during the insertion of the large transmembrane beta-sheet of the PFO oligomer. Finally, the PFO TMHs do not appear to insert independently, but instead their insertion is coupled.


Assuntos
Toxinas Bacterianas/química , Membrana Celular/química , Dissulfetos/química , Estrutura Secundária de Proteína , Ditiotreitol/farmacologia , Proteínas Hemolisinas , Hemólise , Microscopia Eletrônica
10.
Biochemistry ; 39(51): 15961-70, 2000 Dec 26.
Artigo em Inglês | MEDLINE | ID: mdl-11123923

RESUMO

We have probed the electrophilic binding site (H-site) of human glutathione transferase P1-1 through mutagenesis of two valines, Val 10 and Val 35, into glycine and alanine, respectively. These two residues were previously shown to be the only conformationally variable residues in the H-site and hence may play important roles in cosubstrate recognition and/or product dissociation. Both of these mutant enzymes have been expressed in Escherichia coli and purified and their kinetic properties characterized. The results demonstrate that Val35Ala behaves similarly to wild-type, whereas Val10Gly exhibits a strong decrease of k(cat) and k(cat)/K(m) (cosub) toward two selected cosubstrates: ethacrynic acid and 1-chloro-2,4-dinitrobenzene. Pre-steady-state kinetic analysis of the GSH conjugation with ethacrynic acid shows that both wild-type and Val10Gly mutant enzymes exhibit the same rate-limiting step: the dissociation of product. However, in the Val10Gly mutant there is an increased energetic barrier which renders the dissociation of product more difficult. Similar results are found for the Val10Gly mutant with 1-chloro-2,4-dinitrobenzene as cosubstrate. With this latter cosubstrate, Val 10 also exerts a positive role in the conformational transitions of the ternary complex before the chemical event. Crystallographic analysis of the Val10Gly mutant in complex with the inhibitor S-hexyl-GSH suggests that Val 10 optimally orientates products, thus promoting their exit from the active site.


Assuntos
Glutationa Transferase/metabolismo , Isoenzimas/metabolismo , Valina/metabolismo , Alanina/genética , Substituição de Aminoácidos/genética , Sítios de Ligação/genética , Cristalografia por Raios X , Dinitroclorobenzeno/metabolismo , Ácido Etacrínico/metabolismo , Glutationa/metabolismo , Glutationa S-Transferase pi , Glutationa Transferase/química , Glutationa Transferase/genética , Glicina/genética , Humanos , Isoenzimas/química , Isoenzimas/genética , Cinética , Modelos Moleculares , Mutagênese Sítio-Dirigida , Espectrofotometria , Especificidade por Substrato/genética , Valina/química , Valina/genética
11.
Biochem J ; 351 Pt 2: 341-6, 2000 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-11023819

RESUMO

Glutathione S-transferases (GSTs) normally use hydroxy-group-containing residues in the N-terminal domain of the enzyme for stabilizing the activated form of the co-substrate, glutathione. However, previous mutagenesis studies have shown that this is not true for Beta class GSTs and thus the origin of the stabilization remains a mystery. The recently determined crystal structure of Proteus mirabilis GST B1-1 (PmGST B1-1) suggested that the stabilizing role might be fulfilled in Beta class GSTs by one or more residues in the C-terminal domain of the enzyme. To test this hypothesis we mutated His(106) and Lys(107) of PmGST B1-1 to investigate their possible role in the enzyme's catalytic activity. His(106) was mutated to Ala, Asn and Phe, and Lys(107) to Ala and Arg. The effects of the replacement on the activity, thermal stability and antibiotic-binding capacity of the enzyme were examined. The results are consistent with the involvement of His(106) and Lys(107) in interacting with glutathione at the active site but these residues do not contribute significantly to catalysis, folding or antibiotic binding.


Assuntos
Glutationa Transferase/química , Alanina/química , Sequência de Aminoácidos , Antibacterianos/farmacologia , Arginina/química , Asparagina/química , Sítios de Ligação , Catálise , Sequência Conservada , Relação Dose-Resposta a Droga , Escherichia coli/metabolismo , Guanidina/farmacologia , Histidina/química , Lisina/química , Dados de Sequência Molecular , Mutagênese Sítio-Dirigida , Fenilalanina/química , Ligação Proteica , Conformação Proteica , Dobramento de Proteína , Estrutura Secundária de Proteína , Proteus mirabilis/enzimologia , Rifamicinas/farmacologia , Espectrometria de Fluorescência , Temperatura , Fatores de Tempo
12.
J Mol Biol ; 302(2): 295-302, 2000 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-10970734

RESUMO

An N-capping box motif (Ser/Thr-Xaa-Xaa-Asp) is strictly conserved at the beginning of helix alpha6 in the core of virtually all glutathione transferases (GST) and GST-related proteins. It has been demonstrated that this local motif is important in determining the alpha-helical propensity of the isolated alpha6-peptide and plays a crucial role in the folding and stability of GSTs. Its removal by site-directed mutagenesis generated temperature-sensitive folding mutants unable to refold at physiological temperature (37 degrees C). In the present work, variants of human GSTP1-1 (S150A and D153A), in which the capping residues have been substituted by alanine, have been generated and purified for structural analysis. Thus, for the first time, temperature-sensitive folding mutants of an enzyme, expressed at a permissive temperature, have been crystallized and their three-dimensional structures determined by X-ray crystallography. The crystal structures of human pi class GST temperature-sensitive mutants provide a basis for understanding the structural origin of the dramatic effects observed on the overall stability of the enzyme at higher temperatures upon single substitution of a capping residue.


Assuntos
Glutationa Transferase/química , Mutação/genética , Substituição de Aminoácidos/genética , Cristalografia por Raios X , Estabilidade Enzimática , Glutationa Transferase/genética , Glutationa Transferase/metabolismo , Humanos , Modelos Moleculares , Estrutura Secundária de Proteína
13.
Protein Sci ; 9(2): 417-20, 2000 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-10716194

RESUMO

Alpha1-antitrypsin deficiency, which can lead to both emphysema and liver disease, is a result of the accumulation of alpha1-antitrypsin polymers within the hepatocyte. A wealth of biochemical and biophysical data suggests that alpha1-antitrypsin polymers form via insertion of residues from the reactive center loop of one molecule into the beta-sheet of another. However, this long-standing hypothesis has not been confirmed by direct structural evidence. Here, we describe the first crystallographic evidence of a beta-strand linked polymer form of alpha1-antitrypsin: the crystal structure of a cleaved alpha1-antitrypsin polymer.


Assuntos
alfa 1-Antitripsina/química , Biopolímeros/química , Cristalografia por Raios X , Humanos , Modelos Moleculares , Estrutura Quaternária de Proteína , Estrutura Secundária de Proteína , Deficiência de alfa 1-Antitripsina/metabolismo
14.
Blood ; 95(8): 2491-8, 2000 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-10753826

RESUMO

Heterodimeric cytokine receptors generally consist of a major cytokine-binding subunit and a signaling subunit. The latter can transduce signals by more than 1 cytokine, as exemplified by the granulocyte-macrophage colony-stimulating factor (GM-CSF), interleukin-2 (IL-2), and IL-6 receptor systems. However, often the signaling subunits in isolation are unable to bind cytokines, a fact that has made it more difficult to obtain structural definition of their ligand-binding sites. This report details the crystal structure of the ligand-binding domain of the GM-CSF/IL-3/IL-5 receptor beta-chain (beta(c)) signaling subunit in complex with the Fab fragment of the antagonistic monoclonal antibody, BION-1. This is the first single antagonist of all 3 known eosinophil-producing cytokines, and it is therefore capable of regulating eosinophil-related diseases such as asthma. The structure reveals a fibronectin type III domain, and the antagonist-binding site involves major contributions from the loop between the B and C strands and overlaps the cytokine-binding site. Furthermore, tyrosine(421) (Tyr(421)), a key residue involved in receptor activation, lies in the neighboring loop between the F and G strands, although it is not immediately adjacent to the cytokine-binding residues in the B-C loop. Interestingly, functional experiments using receptors mutated across these loops demonstrate that they are cooperatively involved in full receptor activation. The experiments, however, reveal subtle differences between the B-C loop and Tyr(421), which is suggestive of distinct functional roles. The elucidation of the structure of the ligand-binding domain of beta(c) also suggests how different cytokines recognize a single receptor subunit, which may have implications for homologous receptor systems. (Blood. 2000;95:2491-2498)


Assuntos
Receptores de Fator Estimulador das Colônias de Granulócitos e Macrófagos/química , Receptores de Interleucina-3/química , Receptores de Interleucina/química , Anticorpos Monoclonais/metabolismo , Anticorpos Monoclonais/farmacologia , Sítios de Ligação , Linhagem Celular , Mapeamento de Epitopos , Humanos , Ligantes , Dados de Sequência Molecular , Ligação Proteica , Conformação Proteica , Receptores de Fator Estimulador das Colônias de Granulócitos e Macrófagos/antagonistas & inibidores , Receptores de Fator Estimulador das Colônias de Granulócitos e Macrófagos/metabolismo , Receptores de Interleucina/antagonistas & inibidores , Receptores de Interleucina/metabolismo , Receptores de Interleucina-3/antagonistas & inibidores , Receptores de Interleucina-3/metabolismo , Receptores de Interleucina-5
15.
Protein Sci ; 8(10): 2205-12, 1999 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-10548067

RESUMO

The human Theta class glutathione transferase GSTT2-2 has a novel sulfatase activity that is not dependent on the presence of a conserved hydrogen bond donor in the active site. Initial homology modeling and the crystallographic studies have identified three conserved Arg residues that contribute to the formation of (Arg107 and Arg239), and entry to (Arg242), a sulfate binding pocket. These residues have been individually mutated to Ala to investigate their potential role in substrate binding and catalysis. The mutation of Arg107 had a significant detrimental effect on the sulfatase reaction, while the Arg242 mutation caused only a small reduction in sulfatase activity. Surprisingly, the Arg239 had an increased activity resulting from a reduction in stability. Thus, Arg239 appears to play a role in maintaining the architecture of the active site. Electrostatic calculations performed on the wild-type and mutant forms of the enzyme are in good agreement with the experimental results. These findings, along with docking studies, suggest that prior to conjugation, the location of 1-menaphthyl sulfate, a model substrate for the sulfatase reaction, is approximately midway between the position ultimately occupied by the naphthalene ring of 1-menaphthylglutathione and the free sulfate. It is further proposed that the Arg residues in and around the sulfate binding pocket have a role in electrostatic substrate recognition.


Assuntos
Arginina/metabolismo , Glutationa Transferase/metabolismo , Sulfatos/metabolismo , Arginina/química , Sequência de Bases , Sítios de Ligação , Primers do DNA , Glutationa Transferase/química , Glutationa Transferase/genética , Humanos , Cinética , Modelos Moleculares , Mutagênese Sítio-Dirigida , Eletricidade Estática
16.
Cell ; 99(3): 293-9, 1999 Oct 29.
Artigo em Inglês | MEDLINE | ID: mdl-10555145

RESUMO

Perfringolysin O (PFO), a water-soluble monomeric cytolysin secreted by pathogenic Clostridium perfringens, oligomerizes and forms large pores upon encountering cholesterol-containing membranes. Whereas all pore-forming bacterial toxins examined previously have been shown to penetrate the membrane using a single amphipathic beta hairpin per polypeptide, cysteine-scanning mutagenesis and multiple independent fluorescence techniques here reveal that each PFO monomer contains a second domain involved in pore formation, and that each of the two amphipathic beta hairpins completely spans the membrane. In the soluble monomer, these transmembrane segments are folded into six alpha helices. The insertion of two transmembrane hairpins per toxin monomer and the major change in secondary structure are striking and define a novel paradigm for the mechanism of membrane insertion by a cytolytic toxin.


Assuntos
Toxinas Bacterianas/química , Toxinas Bacterianas/metabolismo , Clostridium perfringens/fisiologia , Lipossomos , Sequência de Aminoácidos , Substituição de Aminoácidos , Toxinas Bacterianas/genética , Cisteína , Corantes Fluorescentes , Proteínas Hemolisinas/metabolismo , Modelos Biológicos , Modelos Moleculares , Dados de Sequência Molecular , Mutagênese Sítio-Dirigida , Fosfatidilcolinas , Estrutura Secundária de Proteína , Proteínas Recombinantes/química , Proteínas Recombinantes/metabolismo , Marcadores de Spin
17.
J Mol Biol ; 293(5): 1145-60, 1999 Nov 12.
Artigo em Inglês | MEDLINE | ID: mdl-10547292

RESUMO

Pneumolysin, an important virulence factor of the human pathogen Streptococcus pneumoniae, is a pore-forming toxin which also possesses the ability to activate the complement system directly. Pneumolysin binds to cholesterol in cell membrane surfaces as a prelude to pore formation, which involves the oligomerization of the protein. Two important aspects of the pore-forming activity of pneumolysin are therefore the effect of the toxin on bilayer membrane structure and the nature of the self-association into oligomers undergone by it. We have used analytical ultracentrifugation (AUC) to investigate oligomerization and small-angle neutron scattering (SANS) to investigate the changes in membrane structure accompanying pore formation. Pneumolysin self-associates in solution to form oligomeric structures apparently similar to those which appear on the membrane coincident with pore formation. It has previously been demonstrated by us using site-specific chemical derivatization of the protein that the self-interaction preceding oligomerization involves its C-terminal domain. The AUC experiments described here involved pneumolysin toxoids harbouring mutations in different domains, and support our previous conclusions that self-interaction via the C-terminal domain leads to oligomerization and that this may be related to the mechanism by which pneumolysin activates the complement system.SANS data at a variety of neutron contrasts were obtained from liposomes used as model cell membranes in the absence of pneumolysin, and following the addition of toxin at a number of concentrations. These experiments were designed to allow visualization of the effect that pneumolysin has on bilayer membrane structure resulting from oligomerization into a pore-forming complex. The structure of the liposomal membrane alone and following addition of pneumolysin was calculated by the fitting of scattering equations directly to the scattering curves. The fitting equations describe scattering from simple three-dimensional scattering volume models for the structures present in the sample, whose dimensions were varied iteratively within the fitting program. The overall trend was a thinning of the liposome surface on toxin attack, which was countered by the formation of localized structures thicker than the liposome bilayer itself, in a manner dependent on pneumolysin concentration. At the neutron contrast match point of the liposomes, pneumolysin oligomers were observed. Inactive toxin appeared to bind to the liposome but not to cause membrane alteration; subsequent activation of pneumolysin in situ brought about changes in liposome structure similar to those seen in the presence of active toxin. We propose that the changes in membrane structure on toxin attack which we have observed are related to the mechanism by which pneumolysin forms pores and provide an important perspective on protein/membrane interactions in general. We discuss these results in the light of published data concerning the interaction of gramicidin with bilayers and the hydrophobic mismatch effect.


Assuntos
Citotoxinas/química , Citotoxinas/metabolismo , Bicamadas Lipídicas/metabolismo , Lipossomos/metabolismo , Estreptolisinas/química , Estreptolisinas/metabolismo , Sequência de Aminoácidos , Proteínas de Bactérias , Citotoxinas/genética , Dimerização , Bicamadas Lipídicas/química , Lipossomos/química , Modelos Moleculares , Dados de Sequência Molecular , Mutação/genética , Nêutrons , Ligação Proteica , Estrutura Quaternária de Proteína , Espalhamento de Radiação , Soluções , Streptococcus pneumoniae/química , Streptococcus pneumoniae/genética , Estreptolisinas/genética , Relação Estrutura-Atividade , Ultracentrifugação , Água/metabolismo
18.
Cell ; 97(5): 647-55, 1999 May 28.
Artigo em Inglês | MEDLINE | ID: mdl-10367893

RESUMO

The human pathogen Streptococcus pneumoniae produces soluble pneumolysin monomers that bind host cell membranes to form ring-shaped, oligomeric pores. We have determined three-dimensional structures of a helical oligomer of pneumolysin and of a membrane-bound ring form by cryo-electron microscopy. Fitting the four domains from the crystal structure of the closely related perfringolysin reveals major domain rotations during pore assembly. Oligomerization results in the expulsion of domain 3 from its original position in the monomer. However, domain 3 reassociates with the other domains in the membrane pore form. The base of domain 4 contacts the bilayer, possibly along with an extension of domain 3. These results reveal a two-stage mechanism for pore formation by the cholesterol-binding toxins.


Assuntos
Membrana Celular/ultraestrutura , Streptococcus pneumoniae/fisiologia , Streptococcus pneumoniae/ultraestrutura , Estreptolisinas/química , Proteínas de Bactérias , Toxinas Bacterianas/química , Membrana Celular/fisiologia , Microscopia Crioeletrônica , Citotoxinas/química , Proteínas Hemolisinas/química , Humanos , Modelos Moleculares , Modelos Estruturais , Estrutura Secundária de Proteína
19.
EMBO J ; 18(12): 3204-13, 1999 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-10369661

RESUMO

Glutathione synthetase (GS) catalyses the production of glutathione from gamma-glutamylcysteine and glycine in an ATP-dependent manner. Malfunctioning of GS results in disorders including metabolic acidosis, 5-oxoprolinuria, neurological dysfunction, haemolytic anaemia and in some cases is probably lethal. Here we report the crystal structure of human GS (hGS) at 2.1 A resolution in complex with ADP, two magnesium ions, a sulfate ion and glutathione. The structure indicates that hGS belongs to the recently identified ATP-grasp superfamily, although it displays no detectable sequence identity with other family members including its bacterial counterpart, Escherichia coli GS. The difficulty in identifying hGS as a member of the family is due in part to a rare gene permutation which has resulted in a circular shift of the conserved secondary structure elements in hGS with respect to the other known ATP-grasp proteins. Nevertheless, it appears likely that the enzyme shares the same general catalytic mechanism as other ligases. The possibility of cyclic permutations provides an insight into the evolution of this family and will probably lead to the identification of new members. Mutations that lead to GS deficiency have been mapped onto the structure, providing a molecular basis for understanding their effects.


Assuntos
Glutationa Sintase/química , Glutationa Sintase/genética , Mutação , Difosfato de Adenosina/química , Difosfato de Adenosina/metabolismo , Trifosfato de Adenosina/química , Trifosfato de Adenosina/metabolismo , Sequência de Aminoácidos , Sítios de Ligação , Sequência Conservada/genética , Cristalização , Cristalografia por Raios X , Dimerização , Evolução Molecular , Genes/genética , Glutationa/química , Glutationa/metabolismo , Glutationa Sintase/deficiência , Glutationa Sintase/metabolismo , Humanos , Magnésio/química , Magnésio/metabolismo , Modelos Moleculares , Dados de Sequência Molecular , Fosfatos/química , Fosfatos/metabolismo , Estrutura Secundária de Proteína , Sulfatos/química , Sulfatos/metabolismo
20.
Nat Struct Biol ; 6(4): 327-31, 1999 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-10201399

RESUMO

Amyloid precursor protein (APP) plays a central role in Alzheimer disease. A proteolytic-breakdown product of APP, called beta-amyloid, is a major component of the diffuse and fibrillar deposits found in Alzheimer diseased brains. The normal physiological role of APP remains largely unknown despite much work. A knowledge of its function will not only provide insights into the genesis of the disease but may also prove vital in the development of an effective therapy. Here we describe the 1.8 A resolution crystal structure of the N-terminal, heparin-binding domain of APP (residues 28-123), which is responsible, among other things, for stimulation of neurite outgrowth. The structure reveals a highly charged basic surface that may interact with glycosaminoglycans in the brain and an abutting hydrophobic surface that is proposed to play an important functional role such as dimerization or ligand binding. Structural similarities with cysteine-rich growth factors, taken together with its known growth-promoting properties, suggests the APP N-terminal domain could function as a growth factor in vivo.


Assuntos
Precursor de Proteína beta-Amiloide/química , Precursor de Proteína beta-Amiloide/metabolismo , Doença de Alzheimer/metabolismo , Sequência de Aminoácidos , Sítios de Ligação , Cristalografia por Raios X , Substâncias de Crescimento/química , Heparina/metabolismo , Fator de Crescimento de Hepatócito/química , Modelos Moleculares , Dados de Sequência Molecular , Fragmentos de Peptídeos/química , Conformação Proteica , Homologia de Sequência de Aminoácidos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA