Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 17 de 17
Filtrar
Mais filtros

Base de dados
País/Região como assunto
Tipo de documento
Intervalo de ano de publicação
1.
Eur J Clin Microbiol Infect Dis ; 41(3): 445-454, 2022 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-34997388

RESUMO

This st udy aims to evaluate the prevalence of SARS-CoV-2 antibodies in locked-down family households to determine viral dynamics and immunity acquisition. COVID-19 individuals and their households in lockdown under the same roof during early spring 2020 were interviewed and tested using rapid immunochromatographic lateral flow antibodies assays (LFA) between July and September 2020. Outcomes were secondary infection rate (SIR) among contacts, household infection rate, and predictors of transmission. We enrolled 87 households including 87 COVID-19 index cases (female 78.2%; median age: 47.0 years, IQR: 42.0-51.5) and 255 contacts (males: 52.9%; median age: 19.0 years, IQR: 11.0-43.5) consisting of their children (42%) or spouses/partners (28.2%). A total of 95/255 contacts were SARS-CoV-2 antibody positive leading to a SIR of 37.3% (95% confidence interval (CI): 31.3-43.5%). Viral transmission was observed in 54 households (62%). SARS-CoV-2 infection was asymptomatic in 33/95 (34.7%) of SARS-CoV-2-positive contacts. Independent predictors of virus transmission from index to contacts were housing surface area < 60 m2 (OR: 5.6 [1.1; 28.2] and a four-member family compared to five (OR: 3.6 [1.2; 10.3]). Households represent a high-risk setting for SARS-CoV-2 transmission through close contact within the family amplified by the number of family members and the housing surface area.


Assuntos
COVID-19 , Adulto , Criança , Controle de Doenças Transmissíveis , Características da Família , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Paris , SARS-CoV-2 , Adulto Jovem
2.
Nature ; 520(7545): 109-13, 2015 Apr 02.
Artigo em Inglês | MEDLINE | ID: mdl-25581790

RESUMO

Dengue disease is caused by four different flavivirus serotypes, which infect 390 million people yearly with 25% symptomatic cases and for which no licensed vaccine is available. Recent phase III vaccine trials showed partial protection, and in particular no protection for dengue virus serotype 2 (refs 3, 4). Structural studies so far have characterized only epitopes recognized by serotype-specific human antibodies. We recently isolated human antibodies potently neutralizing all four dengue virus serotypes. Here we describe the X-ray structures of four of these broadly neutralizing antibodies in complex with the envelope glycoprotein E from dengue virus serotype 2, revealing that the recognition determinants are at a serotype-invariant site at the E-dimer interface, including the exposed main chain of the E fusion loop and the two conserved glycan chains. This 'E-dimer-dependent epitope' is also the binding site for the viral glycoprotein prM during virus maturation in the secretory pathway of the infected cell, explaining its conservation across serotypes and highlighting an Achilles' heel of the virus with respect to antibody neutralization. These findings will be instrumental for devising novel immunogens to protect simultaneously against all four serotypes of dengue virus.


Assuntos
Anticorpos Neutralizantes/química , Anticorpos Neutralizantes/imunologia , Anticorpos Antivirais/química , Anticorpos Antivirais/imunologia , Vírus da Dengue/química , Vírus da Dengue/imunologia , Anticorpos Neutralizantes/genética , Anticorpos Antivirais/genética , Reações Cruzadas/imunologia , Cristalografia por Raios X , Vírus da Dengue/classificação , Epitopos/química , Epitopos/imunologia , Humanos , Modelos Moleculares , Dados de Sequência Molecular , Mutação/genética , Conformação Proteica , Multimerização Proteica , Solubilidade , Especificidade da Espécie , Proteínas do Envelope Viral/química , Proteínas do Envelope Viral/imunologia
3.
J Antimicrob Chemother ; 75(5): 1321-1323, 2020 05 01.
Artigo em Inglês | MEDLINE | ID: mdl-31977046

RESUMO

BACKGROUND: Several studies have shown that NNRTI/PI-based triple therapy could be safely administered as a 4 days (4D) or 5 days (5D) a week maintenance strategy. We report here our experience of using an integrase inhibitor (INSTI)-based 4D/5D regimen in virologically suppressed HIV patients. METHODS: This cohort study enrolled adult patients on ART with viral load (VL) <50 copies/mL for >1 year, who switched to an INSTI-based triple regimen given 4D/5D a week. The primary endpoint was the virological efficacy rate at Week (W) 48, with virological failure defined as confirmed VL ≥50 copies/mL. RESULTS: A total of 73 patients were included (n = 28 for 4D, n = 45 for 5D): 54 men (74%), median (IQR) age 51 (45-57) years, ART duration 10 (6-18) years and duration of viral suppression 5 (2-9) years at baseline. As of 25 March 2019, the median follow-up was 21 (14-35) months, with a total of 161 patient-years of follow-up; all patients had reached the W24 visit, 66 (90%) W48 and 34 (47%) W96. Four patients discontinued the strategy: virological failure (n = 2) at W60 and W67, respectively, switch for renal toxicity (n = 1) at W28 and switch to rilpivirine/dolutegravir (n = 1) at W65. Overall the rate of virological success (95% CI) was 100% (94%-100%) at W24 and W48 and 93.7% (79.8%-98.2%) at W96. CONCLUSIONS: While waiting for the final results of the large randomized QUATUOR ANRS-170 study, our real-life results suggest that the use of an intermittent maintenance triple-drug regimen given as a weekend (2 or 3 days) off is as effective with an INSTI-based regimen as with a PI or an NNRTI.


Assuntos
Fármacos Anti-HIV , Infecções por HIV , Inibidores de Integrase de HIV , Adulto , Fármacos Anti-HIV/uso terapêutico , Estudos de Coortes , Infecções por HIV/tratamento farmacológico , Inibidores de Integrase de HIV/uso terapêutico , Humanos , Masculino , Pessoa de Meia-Idade , Rilpivirina/uso terapêutico , Resultado do Tratamento , Carga Viral
4.
Proc Natl Acad Sci U S A ; 112(9): 2865-70, 2015 Mar 03.
Artigo em Inglês | MEDLINE | ID: mdl-25730860

RESUMO

The glycine receptor (GlyR) is a pentameric ligand-gated ion channel (pLGIC) mediating inhibitory transmission in the nervous system. Its transmembrane domain (TMD) is the target of allosteric modulators such as general anesthetics and ethanol and is a major locus for hyperekplexic congenital mutations altering the allosteric transitions of activation or desensitization. We previously showed that the TMD of the human α1GlyR could be fused to the extracellular domain of GLIC, a bacterial pLGIC, to form a functional chimera called Lily. Here, we overexpress Lily in Schneider 2 insect cells and solve its structure by X-ray crystallography at 3.5 Å resolution. The TMD of the α1GlyR adopts a closed-channel conformation involving a single ring of hydrophobic residues at the center of the pore. Electrophysiological recordings show that the phenotypes of key allosteric mutations of the α1GlyR, scattered all along the pore, are qualitatively preserved in this chimera, including those that confer decreased sensitivity to agonists, constitutive activity, decreased activation kinetics, or increased desensitization kinetics. Combined structural and functional data indicate a pore-opening mechanism for the α1GlyR, suggesting a structural explanation for the effect of some key hyperekplexic allosteric mutations. The first X-ray structure of the TMD of the α1GlyR solved here using GLIC as a scaffold paves the way for mechanistic investigation and design of allosteric modulators of a human receptor.


Assuntos
Receptores de Glicina/química , Regulação Alostérica/fisiologia , Animais , Cristalografia por Raios X , Drosophila melanogaster , Humanos , Estrutura Terciária de Proteína , Receptores de Glicina/genética , Receptores de Glicina/metabolismo , Relação Estrutura-Atividade
5.
J Virol ; 90(6): 3112-22, 2016 Jan 06.
Artigo em Inglês | MEDLINE | ID: mdl-26739044

RESUMO

UNLABELLED: Hypervariable region 1 (HVR1) (amino acids [aa] 384 to 410) on the E2 glycoprotein of hepatitis C virus contributes to persistent infection by evolving escape mutations that attenuate binding of inhibitory antibodies and by blocking access of broadly neutralizing antibodies to their epitopes. A third proposed mechanism of immune antagonism is that poorly neutralizing antibodies binding to HVR1 interfere with binding of other superior neutralizing antibodies. Epitope mapping of human monoclonal antibodies (HMAbs) that bind to an adjacent, conserved domain on E2 encompassing aa 412 to 423 revealed two subsets, designated HC33 HMAbs. While both subsets have contact residues within aa 412 to 423, alanine-scanning mutagenesis suggested that one subset, which includes HC33.8, has an additional contact residue within HVR1. To test for interference of anti-HVR1 antibodies with binding of antibodies to aa 412 to 423 and other E2 determinants recognized by broadly neutralizing HMAbs, two murine MAbs against HVR1 (H77.16) and aa 412 to 423 (H77.39) were studied. As expected, H77.39 inhibited the binding of all HC33 HMAbs. Unexpectedly, H77.16 also inhibited the binding of both subsets of HC33 HMAbs. This inhibition also was observed against other broadly neutralizing HMAbs to epitopes outside aa 412 to 423. Combination antibody neutralization studies by the median-effect analysis method with H77.16 and broadly reactive HMAbs revealed antagonism between these antibodies. Structural studies demonstrated conformational flexibility in this antigenic region, which supports the possibility of anti-HVR1 antibodies hindering the binding of broadly neutralizing MAbs. These findings support the hypothesis that anti-HVR1 antibodies can interfere with a protective humoral response against HCV infection. IMPORTANCE: HVR1 contributes to persistent infection by evolving mutations that escape from neutralizing antibodies to HVR1 and by shielding broadly neutralizing antibodies from their epitopes. This study provides insight into a new immune antagonism mechanism by which the binding of antibodies to HVR1 blocks the binding and activity of broadly neutralizing antibodies to HCV. Immunization strategies that avoid the induction of HVR1 antibodies should increase the inhibitory activity of broadly neutralizing anti-HCV antibodies elicited by candidate vaccines.


Assuntos
Anticorpos Neutralizantes/imunologia , Hepacivirus/imunologia , Anticorpos Anti-Hepatite C/imunologia , Proteínas do Envelope Viral/imunologia , Animais , Anticorpos Monoclonais/imunologia , Mapeamento de Epitopos , Epitopos de Linfócito B/imunologia , Camundongos , Ligação Proteica
6.
Nature ; 468(7324): 709-12, 2010 Dec 02.
Artigo em Inglês | MEDLINE | ID: mdl-21124458

RESUMO

Chikungunya virus (CHIKV) is an emerging mosquito-borne alphavirus that has caused widespread outbreaks of debilitating human disease in the past five years. CHIKV invasion of susceptible cells is mediated by two viral glycoproteins, E1 and E2, which carry the main antigenic determinants and form an icosahedral shell at the virion surface. Glycoprotein E2, derived from furin cleavage of the p62 precursor into E3 and E2, is responsible for receptor binding, and E1 for membrane fusion. In the context of a concerted multidisciplinary effort to understand the biology of CHIKV, here we report the crystal structures of the precursor p62-E1 heterodimer and of the mature E3-E2-E1 glycoprotein complexes. The resulting atomic models allow the synthesis of a wealth of genetic, biochemical, immunological and electron microscopy data accumulated over the years on alphaviruses in general. This combination yields a detailed picture of the functional architecture of the 25 MDa alphavirus surface glycoprotein shell. Together with the accompanying report on the structure of the Sindbis virus E2-E1 heterodimer at acidic pH (ref. 3), this work also provides new insight into the acid-triggered conformational change on the virus particle and its inbuilt inhibition mechanism in the immature complex.


Assuntos
Vírus Chikungunya/química , Glicoproteínas de Membrana/química , Proteínas do Envelope Viral/química , Vírion/química , Animais , Linhagem Celular , Microscopia Crioeletrônica , Cristalografia por Raios X , Drosophila melanogaster , Concentração de Íons de Hidrogênio , Modelos Moleculares , Complexos Multiproteicos/química , Multimerização Proteica , Precursores de Proteínas/química , Estrutura Quaternária de Proteína , Proteínas Virais de Fusão/química
7.
J Struct Biol ; 188(1): 71-8, 2014 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-25172991

RESUMO

Nuclear magnetic resonance spectroscopy is a powerful tool to study structural and functional properties of proteins, provided that they can be enriched in stable isotopes such as (15)N, (13)C and (2)H. This is usually easy and inexpensive when the proteins are expressed in Escherichiacoli, but many eukaryotic (human in particular) proteins cannot be produced this way. An alternative is to express them in insect cells. Labeled insect cell growth media are commercially available but at prohibitive prices, limiting the NMR studies to only a subset of biologically important proteins. Non-commercial solutions from academic institutions have been proposed, but none of them is really satisfying. We have developed a (15)N-labeling procedure based on the use of a commercial medium depleted of all amino acids and supplemented with a (15)N-labeled yeast autolysate for a total cost about five times lower than that of the currently available solutions. We have applied our procedure to the production of a non-polymerizable mutant of actin in Sf9 cells and of fragments of eukaryotic and viral membrane fusion proteins in S2 cells, which typically cannot be produced in E. coli, with production yields comparable to those obtained with standard commercial media. Our results support, in particular, the putative limits of a self-folding domain within a viral glycoprotein of unknown structure.


Assuntos
Perfilação da Expressão Gênica/métodos , Imageamento por Ressonância Magnética , Biossíntese de Proteínas , Proteínas da Matriz Viral/química , Aminoácidos/química , Animais , Drosophila/química , Drosophila/genética , Humanos , Radioisótopos de Nitrogênio/química , Células Sf9 , Spodoptera
8.
J Biol Chem ; 288(25): 18561-73, 2013 Jun 21.
Artigo em Inglês | MEDLINE | ID: mdl-23653352

RESUMO

Widespread drug resistance calls for the urgent development of new antimalarials that target novel steps in the life cycle of Plasmodium falciparum and Plasmodium vivax. The essential subtilisin-like serine protease SUB1 of Plasmodium merozoites plays a dual role in egress from and invasion into host erythrocytes. It belongs to a new generation of attractive drug targets against which specific potent inhibitors are actively searched. We characterize here the P. vivax SUB1 enzyme and show that it displays a typical auto-processing pattern and apical localization in P. vivax merozoites. To search for small PvSUB1 inhibitors, we took advantage of the similarity of SUB1 with bacterial subtilisins and generated P. vivax SUB1 three-dimensional models. The structure-based virtual screening of a large commercial chemical compounds library identified 306 virtual best hits, of which 37 were experimentally confirmed inhibitors and 5 had Ki values of <50 µM for PvSUB1. Interestingly, they belong to different chemical families. The most promising competitive inhibitor of PvSUB1 (compound 2) was equally active on PfSUB1 and displayed anti-P. falciparum and Plasmodium berghei activity in vitro and in vivo, respectively. Compound 2 inhibited the endogenous PfSUB1 as illustrated by the inhibited maturation of its natural substrate PfSERA5 and inhibited parasite egress and subsequent erythrocyte invasion. These data indicate that the strategy of in silico screening of three-dimensional models to select for virtual inhibitors combined with stringent biological validation successfully identified several inhibitors of the PvSUB1 enzyme. The most promising hit proved to be a potent cross-inhibitor of PlasmodiumSUB1, laying the groundwork for the development of a globally active small compound antimalarial.


Assuntos
Plasmodium vivax/enzimologia , Estrutura Terciária de Proteína , Proteínas de Protozoários/química , Serina Proteases/química , Sequência de Aminoácidos , Animais , Antimaláricos/química , Antimaláricos/farmacologia , Sítios de Ligação/genética , Biocatálise/efeitos dos fármacos , Relação Dose-Resposta a Droga , Eritrócitos/efeitos dos fármacos , Eritrócitos/parasitologia , Feminino , Cinética , Malária/parasitologia , Malária/prevenção & controle , Merozoítos/efeitos dos fármacos , Merozoítos/enzimologia , Camundongos , Modelos Moleculares , Dados de Sequência Molecular , Estrutura Molecular , Plasmodium berghei/efeitos dos fármacos , Plasmodium berghei/enzimologia , Plasmodium vivax/efeitos dos fármacos , Plasmodium vivax/genética , Proteínas de Protozoários/genética , Proteínas de Protozoários/metabolismo , Homologia de Sequência de Aminoácidos , Serina Proteases/genética , Serina Proteases/metabolismo , Inibidores de Serina Proteinase/química , Inibidores de Serina Proteinase/farmacologia , Células Sf9 , Especificidade por Substrato
9.
Nat Struct Mol Biol ; 2024 Jun 04.
Artigo em Inglês | MEDLINE | ID: mdl-38834913

RESUMO

The hallmark of non-selective autophagy is the formation of cup-shaped phagophores that capture bulk cytoplasm. The process is accompanied by the conjugation of LC3B to phagophores by an E3 ligase complex comprising ATG12-ATG5 and ATG16L1. Here we combined two complementary reconstitution approaches to reveal the function of LC3B and its ligase complex during phagophore expansion. We found that LC3B forms together with ATG12-ATG5-ATG16L1 a membrane coat that remodels flat membranes into cups that closely resemble phagophores. Mechanistically, we revealed that cup formation strictly depends on a close collaboration between LC3B and ATG16L1. Moreover, only LC3B, but no other member of the ATG8 protein family, promotes cup formation. ATG16L1 truncates that lacked the C-terminal membrane binding domain catalyzed LC3B lipidation but failed to assemble coats, did not promote cup formation and inhibited the biogenesis of non-selective autophagosomes. Our results thus demonstrate that ATG16L1 and LC3B induce and stabilize the characteristic cup-like shape of phagophores.

10.
Materials (Basel) ; 15(20)2022 Oct 13.
Artigo em Inglês | MEDLINE | ID: mdl-36295185

RESUMO

Materials produced by additive manufacturing (AM) often have different microstructures from those obtained using conventional metallurgy (CM), which can have significant impacts on the materials' durability, and in particular, resistance to corrosion. In this study, we were concerned with the susceptibility to pitting and environmentally assisted cracking (EAC) of 17-4PH martensitic stainless steel (MSS). We focused on the evolution from pitting to EAC, and the behaviour of MSS produced by AM was compared with that of its CM counterpart. Potentiodynamic polarisation tests were combined with chronoamperometry measurements performed without and with mechanical loading to study both stable and metastable pitting and the influence of stress on these processes. EAC tests were carried out and combined with observations of fracture surfaces. MSS produced by AM was more resistant to pit initiation due to fewer and finer NbC particles. However, the propagation kinetics of stable pits were higher for this MSS due to a higher amount of reversed austenite. The stress was found to stabilise the metastable pits and to accelerate the propagation of stable pits, which resulted in an increased susceptibility to EAC of the MSS produced by AM. These results clearly highlighted the fact that the reversed austenite amount has to be perfectly controlled in AM processes.

11.
J Immunother Cancer ; 7(1): 20, 2019 01 28.
Artigo em Inglês | MEDLINE | ID: mdl-30691536

RESUMO

BACKGROUND: Enadenotucirev is a chimeric adenovirus with demonstrated preclinical tumor-selective cytotoxicity and a short half-life. Further clinical mechanism of action data showed that enadenotucirev can gain access to and replicate within different types of epithelial tumors. This phase 1 dose escalation study assessed intravenous (IV) dose escalation with enadenotucirev to establish the maximum tolerated dose (MTD) and subsequently identify a suitable schedule for repeated cycles. METHODS: Sixty-one patients with advanced epithelial tumors unresponsive to conventional therapy were enrolled and received enadenotucirev monotherapy as part of this study. During the phase 1a dose escalation (n = 22) and expansion (n = 9), delivery of enadenotucirev between 1 × 1010 and 1 × 1013 viral particles (vp) on days 1, 3, and 5 (single cycle) was used to determine an appropriate MTD. Subsequent treatment cohorts (phase 1a, n = 6 and phase 1b, n = 24) examined the feasibility of repeated dosing cycles in either 3-weekly or weekly dosing regimens. RESULTS: Enadenotucirev displayed a predictable and manageable safety profile at doses up to the MTD of 3 × 1012 vp, irrespective of infusion time or dosing schedule. The most commonly reported treatment-emergent adverse events (TEAEs) of grade 3 or higher were hypoxia, lymphopenia, and neutropenia. The frequency of all TEAEs (notably pyrexia and chills) was highest within 24 h of the first enadenotucirev infusion and decreased upon subsequent dosing. Additionally, delivery of three doses of enadenotucirev over 5 days optimized pharmacokinetic and chemokine profiles in the circulation over time. CONCLUSIONS: This study provides key clinical data in patients with solid epithelial tumors following treatment with IV enadenotucirev monotherapy and supports further investigation of enadenotucirev in combination with other therapeutic agents at doses up to the MTD of 3 × 1012 vp. TRIAL REGISTRATION: ( ClinicalTrials.gov Identifier: NCT02028442 ). Trial registration date: 07 January 2014 - Retrospectively registered.


Assuntos
Adenoviridae , Neoplasias Epiteliais e Glandulares/terapia , Terapia Viral Oncolítica , Vírus Oncolíticos , Administração Intravenosa , Adulto , Idoso , Citocinas/sangue , Feminino , Humanos , Masculino , Dose Máxima Tolerável , Pessoa de Meia-Idade , Neoplasias Epiteliais e Glandulares/sangue , Neoplasias Epiteliais e Glandulares/virologia
12.
J Immunother Cancer ; 5(1): 71, 2017 09 19.
Artigo em Inglês | MEDLINE | ID: mdl-28923104

RESUMO

BACKGROUND: Enadenotucirev (formerly ColoAd1) is a tumor-selective chimeric adenovirus with demonstrated preclinical activity. This phase 1 Mechanism of Action study assessed intravenous (IV) delivery of enadenotucirev in patients with resectable colorectal cancer (CRC), non-small-cell lung cancer (NSCLC), urothelial cell cancer (UCC), and renal cell cancer (RCC) with a comparator intratumoral (IT) dosed CRC patient cohort. METHODS: Seventeen patients scheduled for primary tumor resection were enrolled. IT injection of enadenotucirev (CRC only) was administered as a single dose (≤ 3 × 1011 viral particles [vp]) on day 1, followed by resection during days 8-15. IV infusion of enadenotucirev was administered by three separate doses (1 × 1012 vp) on days 1, 3, and 5, followed by resection during days 8-15 (CRC) or days 10-25 (NSCLC, UCC, and RCC). Enadenotucirev activity was measured using immunohistochemical staining of nuclear viral hexon and quantitative polymerase chain reaction for viral genomic DNA. RESULTS: Delivery of enadenotucirev was observed in most tumor samples following IV infusion, with little or no demonstrable activity in normal tissue. This virus delivery (by both IV and IT dosing) was accompanied by high local CD8+ cell infiltration in 80% of tested tumor samples, suggesting a potential enadenotucirev-driven immune response. Both methods of enadenotucirev delivery were well tolerated, with no treatment-associated serious adverse events. CONCLUSIONS: This study provides key delivery and feasibility data to support the use of IV infusion of enadenotucirev, or therapeutic transgene-bearing derivatives of it, in clinical trials across a range of epithelial tumors, including the ongoing combination study of enadenotucirev with the checkpoint inhibitor nivolumab. It also provides insights into the potential immune-stimulating properties of enadenotucirev. TRIAL REGISTRATION: This MOA study was a phase 1, multicenter, non-randomized, open-label study to investigate the administration of enadenotucirev in a preoperative setting (ClinicalTrials.gov: NCT02053220).


Assuntos
Adenovírus Humanos/fisiologia , Carcinoma Pulmonar de Células não Pequenas/terapia , Carcinoma de Células Renais/terapia , Carcinoma de Células de Transição/terapia , Neoplasias Colorretais/terapia , Neoplasias Pulmonares/terapia , Adenovírus Humanos/genética , Administração Intravenosa , Linfócitos T CD8-Positivos/metabolismo , Carcinoma Pulmonar de Células não Pequenas/imunologia , Carcinoma de Células Renais/imunologia , Carcinoma de Células de Transição/imunologia , Neoplasias Colorretais/imunologia , Terapia Combinada , DNA Viral/genética , Procedimentos Cirúrgicos do Sistema Digestório , Humanos , Neoplasias Pulmonares/imunologia , Terapia Viral Oncolítica , Vírus Oncolíticos/genética , Vírus Oncolíticos/fisiologia , Procedimentos Cirúrgicos Pulmonares , Resultado do Tratamento , Procedimentos Cirúrgicos Urológicos
13.
FEBS Lett ; 590(20): 3690-3699, 2016 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-27680677

RESUMO

Understanding the structural basis of actin cytoskeleton remodeling requires stabilization of actin monomers, oligomers, and filaments in complex with partner proteins, using various biochemical strategies. Here, we report a dramatic destabilization of the dynamic interaction with a model ß-thymosin/WH2 domain induced by mutations in actin. This result underlines that mutant actins should be used with prudence to characterize interactions with intrinsically disordered partners as destabilization of dynamic interactions, although identifiable by NMR, may be invisible to other structural techniques. It also highlights how both ß-thymosin/WH2 domains and actin tune local structure and dynamics in regulatory processes involving intrinsically disordered domains.


Assuntos
Actinas/genética , Mutação , Timosina/química , Actinas/química , Animais , Cristalografia por Raios X , Modelos Moleculares , Ligação Proteica , Conformação Proteica , Coelhos , Homologia de Sequência de Aminoácidos
14.
Nat Commun ; 5: 4833, 2014 Sep 10.
Artigo em Inglês | MEDLINE | ID: mdl-25204226

RESUMO

The Plasmodium subtilase SUB1 plays a pivotal role during the egress of malaria parasites from host hepatocytes and erythrocytes. Here we report the crystal structure of full-length SUB1 from the human-infecting parasite Plasmodium vivax, revealing a bacterial-like catalytic domain in complex with a Plasmodium-specific prodomain. The latter displays a novel architecture with an amino-terminal insertion that functions as a 'belt', embracing the catalytic domain to further stabilize the quaternary structure of the pre-protease, and undergoes calcium-dependent autoprocessing during subsequent activation. Although dispensable for recombinant enzymatic activity, the SUB1 'belt' could not be deleted in Plasmodium berghei, suggesting an essential role of this domain for parasite development in vivo. The SUB1 structure not only provides a valuable platform to develop new anti-malarial candidates against this promising drug target, but also defines the Plasmodium-specific 'belt' domain as a key calcium-dependent regulator of SUB1 during parasite egress from host cells.


Assuntos
Plasmodium berghei , Plasmodium vivax , Proteínas de Protozoários/metabolismo , Subtilisinas/metabolismo , Sequência de Aminoácidos , Antimaláricos/uso terapêutico , Cristalografia , Humanos , Malária Vivax/tratamento farmacológico , Dados de Sequência Molecular , Terapia de Alvo Molecular , Estrutura Terciária de Proteína
15.
Antimicrob Agents Chemother ; 52(2): 491-6, 2008 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-18039922

RESUMO

The objective of this study was to characterize the mutations selected by darunavir (DRV) use in protease inhibitor (PI)-experienced patients and the associated factors. We analyzed treatment failure in 54 PI-experienced human immunodeficiency virus (HIV)-infected patients on a DRV- and ritonavir-containing regimen. Viral genotyping was carried out at the baseline, at between 1 and 3 months of treatment, and at between 3 and 6 months of treatment to search for the selection of mutations conferring resistance to PIs. The median baseline HIV RNA level was 4.9 log(10) copies/ml, and the median CD4 count was 87 cells/mm(3). At the baseline, the median numbers of resistance mutations were as follows: 3 DRV resistance mutations, 4 major PI resistance mutations, and 10 minor PI resistance mutations. The most common mutations that emerged at rebound included V32I (44%), I54M/L (24%), L33F (25%), I84V (21%), and L89V (12%). Multivariate analysis showed that higher baseline HIV RNA levels and smaller numbers of nucleoside reverse transcriptase inhibitor simultaneously used with DRV were associated with a higher risk of DRV resistance mutation selection. By contrast, L76V, a known DRV resistance mutation, was found to decrease the risk of selection of another DRV resistance mutation. The occurrence of virological failure while a patient was on DRV was associated with the selection of mutations that increased the level of DRV resistance without affecting susceptibility to tipranavir (TPV). In these PI-treated patients who displayed treatment failure while they were on a DRV-containing regimen, we confirmed the set of emerging mutations associated with DRV failure and identified the factors associated with the selection of these mutations. TPV susceptibility does not seem to be affected by the selection of a DRV resistance mutation.


Assuntos
Farmacorresistência Viral/genética , Infecções por HIV/tratamento farmacológico , Inibidores da Protease de HIV/uso terapêutico , Protease de HIV/genética , Mutação , Sulfonamidas/uso terapêutico , Falha de Tratamento , Darunavir , Quimioterapia Combinada , Feminino , Infecções por HIV/virologia , Protease de HIV/efeitos dos fármacos , Inibidores da Protease de HIV/farmacologia , HIV-1/efeitos dos fármacos , HIV-1/enzimologia , HIV-1/genética , Humanos , Masculino , Testes de Sensibilidade Microbiana/métodos , RNA Viral/sangue , Inibidores da Transcriptase Reversa/uso terapêutico , Fatores de Risco , Seleção Genética , Sulfonamidas/farmacologia
16.
Biochemistry ; 42(51): 15300-9, 2003 Dec 30.
Artigo em Inglês | MEDLINE | ID: mdl-14690440

RESUMO

In bacteria, regulatory phosphorylation of proteins at serine and/or threonine residues by Ser/Thr protein kinase (STPK) is an emerging theme in prokaryotic signaling, particularly since the prediction of the occurrence of several STPKs from genome sequencing and sequence surveys. Here we show that protein PknH possesses an autokinase activity and belongs to the large STPK family found in Mycobacterium tuberculosis. Evidence is presented that PknH can also phosphorylate EmbR, a protein suspected to modulate the level of arabinosyltransferase activity involved in arabinan biosynthesis of arabinogalactan, a key molecule of the mycobacterial cell wall. Interestingly, EmbR possesses an FHA (forkhead-associated) domain, a newly described phosphoprotein recognition domain, which plays an essential role in PknH-EmbR interaction and phosphorylation of EmbR by PknH. It is demonstrated that mutation of each of three particular residues of this FHA domain, Arg312, Ser326, and Asn348, totally abolishes the PknH-mediated phosphorylation of EmbR, thus highlighting the critical role of this domain in the direct interaction between EmbR and PknH.


Assuntos
Proteínas de Bactérias/metabolismo , Mycobacterium tuberculosis/enzimologia , Proteínas Nucleares/fisiologia , Pentosiltransferases/metabolismo , Fosfoproteínas/metabolismo , Proteínas Serina-Treonina Quinases/metabolismo , Fatores de Transcrição/fisiologia , Proteínas de Bactérias/química , Proteínas de Bactérias/genética , Proteínas de Bactérias/isolamento & purificação , Catálise , Citosol/química , Citosol/metabolismo , Ativação Enzimática , Fatores de Transcrição Forkhead , Mycobacterium tuberculosis/genética , Proteínas Nucleares/química , Proteínas Nucleares/metabolismo , Proteínas Serina-Treonina Quinases/química , Proteínas Serina-Treonina Quinases/genética , Proteínas Serina-Treonina Quinases/isolamento & purificação , Estrutura Terciária de Proteína/fisiologia , Serina/metabolismo , Especificidade por Substrato , Treonina/metabolismo , Fatores de Transcrição/química , Fatores de Transcrição/metabolismo
17.
Biochem Biophys Res Commun ; 308(4): 820-5, 2003 Sep 05.
Artigo em Inglês | MEDLINE | ID: mdl-12927792

RESUMO

Protein PknE from Mycobacterium tuberculosis has been overproduced and purified, and its biochemical properties have been analyzed. This protein is shown to be a eukaryotic-like (Hanks'-type) protein kinase with a structural organization similar to that of membrane-bound eukaryotic sensor serine/threonine kinases. It consists of a N-terminal catalytic domain located in the cytoplasm, linked via a single transmembrane-spanning region to an extracellular C-terminal domain. The full-length enzyme, as well as the cytosolic domain alone, can autophosphorylate on serine and threonine residues. Such autokinase activity requires the presence of a lysine residue at position 45 in subdomain II, which is known to be essential also for eukaryotic kinase activity. Involvement of PknE in the transduction of external signals into the cytosol of bacteria is proposed.


Assuntos
Mycobacterium tuberculosis/metabolismo , Proteínas Serina-Treonina Quinases/química , Proteínas Serina-Treonina Quinases/fisiologia , Motivos de Aminoácidos , Domínio Catalítico , Membrana Celular/metabolismo , Citoplasma/metabolismo , Citosol/metabolismo , Glutationa Transferase/metabolismo , Lisina/química , Modelos Genéticos , Mutagênese Sítio-Dirigida , Mutação , Fosforilação , Plasmídeos/metabolismo , Proteínas Quinases/metabolismo , Proteínas Serina-Treonina Quinases/metabolismo , Estrutura Terciária de Proteína , Proteínas Recombinantes de Fusão/metabolismo , Serina/química , Transdução de Sinais , Treonina/química
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa