Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 34
Filtrar
1.
Mol Cell ; 63(1): 86-96, 2016 07 07.
Artigo em Inglês | MEDLINE | ID: mdl-27264868

RESUMO

The recalcitrance of many bacterial infections to antibiotic treatment is thought to be due to the presence of persisters that are non-growing, antibiotic-insensitive cells. Eventually, persisters resume growth, accounting for relapses of infection. Salmonella is an important pathogen that causes disease through its ability to survive inside macrophages. After macrophage phagocytosis, a significant proportion of the Salmonella population forms non-growing persisters through the action of toxin-antitoxin modules. Here we reveal that one such toxin, TacT, is an acetyltransferase that blocks the primary amine group of amino acids on charged tRNA molecules, thereby inhibiting translation and promoting persister formation. Furthermore, we report the crystal structure of TacT and note unique structural features, including two positively charged surface patches that are essential for toxicity. Finally, we identify a detoxifying mechanism in Salmonella wherein peptidyl-tRNA hydrolase counteracts TacT-dependent growth arrest, explaining how bacterial persisters can resume growth.


Assuntos
Acetiltransferases/metabolismo , Proteínas de Bactérias/metabolismo , Toxinas Bacterianas/metabolismo , Farmacorresistência Bacteriana , RNA Bacteriano/metabolismo , RNA de Transferência/metabolismo , Salmonella typhimurium/enzimologia , Aminoacilação de RNA de Transferência , Acetiltransferases/química , Proteínas de Bactérias/química , Toxinas Bacterianas/química , Hidrolases de Éster Carboxílico/metabolismo , Farmacorresistência Bacteriana/genética , Modelos Moleculares , Conformação Proteica , RNA Bacteriano/genética , RNA de Transferência/genética , Salmonella typhimurium/genética , Salmonella typhimurium/crescimento & desenvolvimento , Salmonella typhimurium/patogenicidade , Relação Estrutura-Atividade , Propriedades de Superfície , Fatores de Tempo , Virulência
2.
Nat Chem Biol ; 17(12): 1296-1304, 2021 12.
Artigo em Inglês | MEDLINE | ID: mdl-34556858

RESUMO

Toxin-antitoxin (TA) systems are a large family of genes implicated in the regulation of bacterial growth and its arrest in response to attacks. These systems encode nonsecreted toxins and antitoxins that specifically pair, even when present in several paralogous copies per genome. Salmonella enterica serovar Typhimurium contains three paralogous TacAT systems that block bacterial translation. We determined the crystal structures of the three TacAT complexes to understand the structural basis of specific TA neutralization and the evolution of such specific pairing. In the present study, we show that alteration of a discrete structural add-on element on the toxin drives specific recognition by their cognate antitoxin underpinning insulation of the three pairs. Similar to other TA families, the region supporting TA-specific pairing is key to neutralization. Our work reveals that additional TA interfaces beside the main neutralization interface increase the safe space for evolution of pairing specificity.


Assuntos
Antitoxinas/química , Toxinas Bacterianas/química , Proteínas Recombinantes/química , Sequência de Aminoácidos , Antitoxinas/genética , Bactérias , Cristalização , Escherichia coli/genética , Modelos Moleculares , Ligação Proteica , Conformação Proteica , Proteínas Recombinantes/genética , Sistemas Toxina-Antitoxina
3.
Int J Mol Sci ; 24(13)2023 Jun 28.
Artigo em Inglês | MEDLINE | ID: mdl-37445934

RESUMO

The process of human embryonic mammary development gives rise to the structures in which mammary cells share a developmental lineage with skin epithelial cells such as keratinocytes. As some breast carcinomas have previously been shown to express high levels of involucrin, a marker of keratinocyte differentiation, we hypothesised that some breast tumours may de-differentiate to a keratinocyte-derived 'evolutionary history'. To confirm our hypothesis, we investigated the frequency of involucrin expression along with that of Brk, a tyrosine kinase expressed in up to 86% of breast carcinomas whose normal expression patterns are restricted to differentiating epithelial cells, most notably those in the skin (keratinocytes) and the gastrointestinal tract. We found that involucrin, a keratinocyte differentiation marker, was expressed in a high proportion (78%) of breast carcinoma samples and cell lines. Interestingly, tumour samples found to express high levels of involucrin were also shown to express Brk. 1,25-dihydroxyvitamin D3, a known differentiation agent and potential anti-cancer agent, decreased proliferation in the breast cancer cell lines that expressed both involucrin and Brk, whereas the Brk/involucrin negative cell lines tested were less susceptible. In addition, responses to 1,25-dihydroxyvitamin D3 were not correlated with vitamin D receptor expression. These data contribute to the growing body of evidence suggesting that cellular responses to 1,25-dihydroxyvitamin D3 are potentially independent of vitamin D receptor status and provide an insight into potential markers, such as Brk and/or involucrin that could predict therapeutic responses to 1,25-dihydroxyvitamin D3.


Assuntos
Neoplasias da Mama , Receptores de Calcitriol , Humanos , Feminino , Neoplasias da Mama/metabolismo , Colecalciferol , Calcitriol , Proteínas de Neoplasias/metabolismo , Proteínas Tirosina Quinases
4.
Nature ; 523(7560): 366-9, 2015 Jul 16.
Artigo em Inglês | MEDLINE | ID: mdl-26061770

RESUMO

Retroviral integration is catalysed by a tetramer of integrase (IN) assembled on viral DNA ends in a stable complex, known as the intasome. How the intasome interfaces with chromosomal DNA, which exists in the form of nucleosomal arrays, is currently unknown. Here we show that the prototype foamy virus (PFV) intasome is proficient at stable capture of nucleosomes as targets for integration. Single-particle cryo-electron microscopy reveals a multivalent intasome-nucleosome interface involving both gyres of nucleosomal DNA and one H2A-H2B heterodimer. While the histone octamer remains intact, the DNA is lifted from the surface of the H2A-H2B heterodimer to allow integration at strongly preferred superhelix location ±3.5 positions. Amino acid substitutions disrupting these contacts impinge on the ability of the intasome to engage nucleosomes in vitro and redistribute viral integration sites on the genomic scale. Our findings elucidate the molecular basis for nucleosome capture by the viral DNA recombination machinery and the underlying nucleosome plasticity that allows integration.


Assuntos
Nucleossomos/química , Nucleossomos/virologia , Spumavirus/metabolismo , Integração Viral , Substituição de Aminoácidos , Sítios de Ligação/genética , Microscopia Crioeletrônica , DNA/genética , DNA/metabolismo , DNA/ultraestrutura , Genoma/genética , Histonas/química , Histonas/metabolismo , Histonas/ultraestrutura , Integrases/metabolismo , Modelos Moleculares , Nucleossomos/genética , Nucleossomos/ultraestrutura , Multimerização Proteica , Recombinação Genética , Spumavirus/química , Spumavirus/genética , Spumavirus/ultraestrutura
5.
J Biol Chem ; 291(50): 25853-25863, 2016 Dec 09.
Artigo em Inglês | MEDLINE | ID: mdl-27789710

RESUMO

Many bacterial pathogens secrete virulence (effector) proteins that interfere with immune signaling in their host. SpvD is a Salmonella enterica effector protein that we previously demonstrated to negatively regulate the NF-κB signaling pathway and promote virulence of S. enterica serovar Typhimurium in mice. To shed light on the mechanistic basis for these observations, we determined the crystal structure of SpvD and show that it adopts a papain-like fold with a characteristic cysteine-histidine-aspartate catalytic triad comprising Cys-73, His-162, and Asp-182. SpvD possessed an in vitro deconjugative activity on aminoluciferin-linked peptide and protein substrates in vitro A C73A mutation abolished SpvD activity, demonstrating that an intact catalytic triad is required for its function. Taken together, these results strongly suggest that SpvD is a cysteine protease. The amino acid sequence of SpvD is highly conserved across different S. enterica serovars, but residue 161, located close to the catalytic triad, is variable, with serovar Typhimurium SpvD having an arginine and serovar Enteritidis a glycine at this position. This variation affected hydrolytic activity of the enzyme on artificial substrates and can be explained by substrate accessibility to the active site. Interestingly, the SpvDG161 variant more potently inhibited NF-κB-mediated immune responses in cells in vitro and increased virulence of serovar Typhimurium in mice. In summary, our results explain the biochemical basis for the effect of virulence protein SpvD and demonstrate that a single amino acid polymorphism can affect the overall virulence of a bacterial pathogen in its host.


Assuntos
Antígenos de Bactérias/metabolismo , Proteínas de Bactérias/metabolismo , Mutação de Sentido Incorreto , Salmonella enteritidis , Salmonella typhimurium , Fatores de Virulência/metabolismo , Substituição de Aminoácidos , Animais , Antígenos de Bactérias/genética , Proteínas de Bactérias/genética , Catálise , Células HEK293 , Humanos , Camundongos , Infecções por Salmonella/genética , Infecções por Salmonella/metabolismo , Salmonella enteritidis/genética , Salmonella enteritidis/metabolismo , Salmonella enteritidis/patogenicidade , Salmonella typhimurium/genética , Salmonella typhimurium/metabolismo , Salmonella typhimurium/patogenicidade , Especificidade da Espécie , Fatores de Virulência/genética
6.
Biochim Biophys Acta Proteins Proteom ; 1865(4): 422-433, 2017 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-28130069

RESUMO

The critical need for iron presents a challenge for pathogenic bacteria that must survive in an environment bereft of accessible iron due to a natural low bioavailability and their host's nutritional immunity. Appropriating haem, either direct from host haemoproteins or by secreting haem-scavenging haemophores, is one way pathogenic bacteria can overcome this challenge. After capturing their target, haem appropriation systems must remove haem from a high-affinity binding site (on the host haemoprotein or bacterial haemophore) and transfer it to a binding site of lower affinity on a bacterial receptor. Structural information is now available to show how, using a combination of induced structural changes and steric clashes, bacteria are able to extract haem from haemophores, haemopexin and haemoglobin. This review focuses on structural descriptions of these bacterial haem acquisition systems, summarising how they bind haem and their target haemoproteins with particularly emphasis on the mechanism of haem extraction.


Assuntos
Bactérias , Infecções Bacterianas , Proteínas de Bactérias , Heme/metabolismo , Animais , Bactérias/genética , Bactérias/metabolismo , Bactérias/patogenicidade , Infecções Bacterianas/genética , Infecções Bacterianas/metabolismo , Proteínas de Bactérias/genética , Proteínas de Bactérias/metabolismo , Humanos
7.
Proc Natl Acad Sci U S A ; 111(7): 2728-33, 2014 Feb 18.
Artigo em Inglês | MEDLINE | ID: mdl-24449914

RESUMO

Transportin 3 (Tnpo3, Transportin-SR2) is implicated in nuclear import of splicing factors and HIV-1 replication. Herein, we show that the majority of cellular Tnpo3 binding partners contain arginine-serine (RS) repeat domains and present crystal structures of human Tnpo3 in its free as well as GTPase Ran- and alternative splicing factor/splicing factor 2 (ASF/SF2)-bound forms. The flexible ß-karyopherin fold of Tnpo3 embraces the RNA recognition motif and RS domains of the cargo. A constellation of charged residues on and around the arginine-rich helix of Tnpo3 HEAT repeat 15 engage the phosphorylated RS domain and are critical for the recognition and nuclear import of ASF/SF2. Mutations in the same region of Tnpo3 impair its interaction with the cleavage and polyadenylation specificity factor 6 (CPSF6) and its ability to support HIV-1 replication. Steric incompatibility of the RS domain and RanGTP engagement by Tnpo3 provides the mechanism for cargo release in the nucleus. Our results elucidate the structural bases for nuclear import of splicing factors and the Tnpo3-CPSF6 nexus in HIV-1 biology.


Assuntos
Núcleo Celular/metabolismo , HIV-1/fisiologia , Modelos Moleculares , Proteínas Nucleares/metabolismo , Conformação Proteica , beta Carioferinas/química , beta Carioferinas/metabolismo , Transporte Ativo do Núcleo Celular/fisiologia , Western Blotting , Cromatografia em Gel , Cromatografia por Troca Iônica , Cristalografia por Raios X , Células HEK293 , HIV-1/metabolismo , Humanos , Imunoprecipitação , Oligonucleotídeos/genética , Ligação Proteica , Replicação Viral/fisiologia , Difração de Raios X , Fatores de Poliadenilação e Clivagem de mRNA/metabolismo
8.
EMBO J ; 31(13): 3020-8, 2012 Jun 29.
Artigo em Inglês | MEDLINE | ID: mdl-22580823

RESUMO

Retroviral integrase (IN) is responsible for two consecutive reactions, which lead to insertion of a viral DNA copy into a host cell chromosome. Initially, the enzyme removes di- or trinucleotides from viral DNA ends to expose 3'-hydroxyls attached to the invariant CA dinucleotides (3'-processing reaction). Second, it inserts the processed 3'-viral DNA ends into host chromosomal DNA (strand transfer). Herein, we report a crystal structure of prototype foamy virus IN bound to viral DNA prior to 3'-processing. Furthermore, taking advantage of its dependence on divalent metal ion cofactors, we were able to freeze trap the viral enzyme in its ground states containing all the components necessary for 3'-processing or strand transfer. Our results shed light on the mechanics of retroviral DNA integration and explain why HIV IN strand transfer inhibitors are ineffective against the 3'-processing step of integration. The ground state structures moreover highlight a striking substrate mimicry utilized by the inhibitors in their binding to the IN active site and suggest ways to improve upon this clinically relevant class of small molecules.


Assuntos
Integrases/química , Spumavirus/enzimologia , Catálise , Domínio Catalítico , Cátions Bivalentes/química , DNA Viral/metabolismo , HIV/enzimologia , Integração Viral , Difração de Raios X
9.
Nature ; 468(7321): 326-9, 2010 Nov 11.
Artigo em Inglês | MEDLINE | ID: mdl-21068843

RESUMO

To establish productive infection, a retrovirus must insert a DNA replica of its genome into host cell chromosomal DNA. This process is operated by the intasome, a nucleoprotein complex composed of an integrase tetramer (IN) assembled on the viral DNA ends. The intasome engages chromosomal DNA within a target capture complex to carry out strand transfer, irreversibly joining the viral and cellular DNA molecules. Although several intasome/transpososome structures from the DDE(D) recombinase superfamily have been reported, the mechanics of target DNA capture and strand transfer by these enzymes remained unclear. Here we report crystal structures of the intasome from prototype foamy virus in complex with target DNA, elucidating the pre-integration target DNA capture and post-catalytic strand transfer intermediates of the retroviral integration process. The cleft between IN dimers within the intasome accommodates chromosomal DNA in a severely bent conformation, allowing widely spaced IN active sites to access the scissile phosphodiester bonds. Our results resolve the structural basis for retroviral DNA integration and provide a framework for the design of INs with altered target sequences.


Assuntos
Spumavirus/química , Spumavirus/fisiologia , Integração Viral , Sequência de Bases , Domínio Catalítico , Cristalografia por Raios X , DNA/química , DNA/genética , DNA/metabolismo , Integrases/genética , Integrases/metabolismo , Modelos Moleculares , Conformação Molecular , Spumavirus/enzimologia
10.
Nature ; 464(7286): 232-6, 2010 Mar 11.
Artigo em Inglês | MEDLINE | ID: mdl-20118915

RESUMO

Integrase is an essential retroviral enzyme that binds both termini of linear viral DNA and inserts them into a host cell chromosome. The structure of full-length retroviral integrase, either separately or in complex with DNA, has been lacking. Furthermore, although clinically useful inhibitors of HIV integrase have been developed, their mechanism of action remains speculative. Here we present a crystal structure of full-length integrase from the prototype foamy virus in complex with its cognate DNA. The structure shows the organization of the retroviral intasome comprising an integrase tetramer tightly associated with a pair of viral DNA ends. All three canonical integrase structural domains are involved in extensive protein-DNA and protein-protein interactions. The binding of strand-transfer inhibitors displaces the reactive viral DNA end from the active site, disarming the viral nucleoprotein complex. Our findings define the structural basis of retroviral DNA integration, and will allow modelling of the HIV-1 intasome to aid in the development of antiretroviral drugs.


Assuntos
DNA Viral/metabolismo , Integrases/química , Modelos Moleculares , Retroviridae/enzimologia , Retroviridae/genética , Domínio Catalítico , HIV-1/enzimologia , HIV-1/genética , Integrases/metabolismo , Estrutura Terciária de Proteína
11.
Nat Struct Mol Biol ; 2024 Mar 27.
Artigo em Inglês | MEDLINE | ID: mdl-38538913

RESUMO

Transcription factors control gene expression; among these, transcriptional repressors must liberate the promoter for derepression to occur. Toxin-antitoxin (TA) modules are bacterial elements that autoregulate their transcription by binding the promoter in a T:A ratio-dependent manner, known as conditional cooperativity. The molecular basis of how excess toxin triggers derepression has remained elusive, largely because monitoring the rearrangement of promoter-repressor complexes, which underpin derepression, is challenging. Here, we dissect the autoregulation of the Salmonella enterica tacAT3 module. Using a combination of assays targeting DNA binding and promoter activity, as well as structural characterization, we determine the essential TA and DNA elements required to control transcription, and we reconstitute a repression-to-derepression path. We demonstrate that excess toxin triggers molecular stripping of the repressor complex off the DNA through multiple allosteric changes causing DNA distortion and ultimately leading to derepression. Thus, our work provides important insight into the mechanisms underlying conditional cooperativity.

12.
Proc Natl Acad Sci U S A ; 107(36): 15910-5, 2010 Sep 07.
Artigo em Inglês | MEDLINE | ID: mdl-20733078

RESUMO

The intasome is the basic recombination unit of retroviral integration, comprising the integrase protein and the ends of the viral DNA made by reverse transcription. Clinical inhibitors preferentially target the DNA-bound form of integrase as compared with the free protein, highlighting the critical requirement for detailed understanding of HIV-1 intasome structure and function. Although previous biochemical studies identified integrase residues that contact the DNA, structural details of protein-protein and protein-DNA interactions within the functional intasome were lacking. The recent crystal structure of the prototype foamy virus (PFV) integrase-viral DNA complex revealed numerous details of this related integration machine. Structures of drug-bound PFV intasomes moreover elucidated the mechanism of inhibitor action. Herein we present a model for the HIV-1 intasome assembled using the PFV structure as template. Our results pinpoint previously identified protein-DNA contacts within the quaternary structure and reveal hitherto unknown roles for Arg20 and Lys266 in DNA binding and integrase function. Models for clinical inhibitors bound at the HIV-1 integrase active site were also constructed and compared with previous studies. Our findings highlight the structural basis for HIV-1 integration and define the mechanism of its inhibition, which should help in formulating new drugs to inhibit viruses resistant to first-in-class compounds.


Assuntos
HIV-1/fisiologia , HIV-1/química , Humanos , Modelos Teóricos , Conformação Proteica
13.
Proc Natl Acad Sci U S A ; 107(46): 20057-62, 2010 Nov 16.
Artigo em Inglês | MEDLINE | ID: mdl-21030679

RESUMO

The development of HIV integrase (IN) strand transfer inhibitors (INSTIs) and our understanding of viral resistance to these molecules have been hampered by a paucity of available structural data. We recently reported cocrystal structures of the prototype foamy virus (PFV) intasome with raltegravir and elvitegravir, establishing the general INSTI binding mode. We now present an expanded set of cocrystal structures containing PFV intasomes complexed with first- and second-generation INSTIs at resolutions of up to 2.5 Å. Importantly, the improved resolution allowed us to refine the complete coordination spheres of the catalytic metal cations within the INSTI-bound intasome active site. We show that like the Q148H/G140S and N155H HIV-1 IN variants, the analogous S217H and N224H PFV INs display reduced sensitivity to raltegravir in vitro. Crystal structures of the mutant PFV intasomes in INSTI-free and -bound forms revealed that the amino acid substitutions necessitate considerable conformational rearrangements within the IN active site to accommodate an INSTI, thus explaining their adverse effects on raltegravir antiviral activity. Furthermore, our structures predict physical proximity and an interaction between HIV-1 IN mutant residues His148 and Ser/Ala140, rationalizing the coevolution of Q148H and G140S/A mutations in drug-resistant viral strains.


Assuntos
Farmacorresistência Viral/genética , Evolução Molecular , Inibidores de Integrase de HIV/farmacologia , Integrases/metabolismo , Retroviridae/enzimologia , Substituição de Aminoácidos/genética , Domínio Catalítico , Inibidores de Integrase de HIV/química , Inibidores de Integrase de HIV/metabolismo , HIV-1/enzimologia , HIV-1/genética , Concentração Inibidora 50 , Mutação/genética , Pirrolidinonas/química , Pirrolidinonas/farmacologia , Raltegravir Potássico
14.
Microb Cell ; 9(4): 84-102, 2022 Apr 04.
Artigo em Inglês | MEDLINE | ID: mdl-35434122

RESUMO

Not all treasure is silver and gold; for pathogenic bacteria, iron is the most precious and the most pillaged of metallic elements. Iron is essential for the survival and growth of all life; however free iron is scarce for bacteria inside human hosts. As a mechanism of defence, humans have evolved ways to store iron so as to render it inaccessible for invading pathogens, such as keeping the metal bound to iron-carrying proteins. For bacteria to survive within humans, they must therefore evolve counters to this defence to compete with these proteins for iron binding, or directly steal iron from them. The most populous form of iron in humans is haem: a functionally significant coordination complex that is central to oxygen transport and predominantly bound by haemoglobin. Haemoglobin is therefore the largest source of iron in humans and, as a result, bacterial pathogens in critical need of iron have evolved complex and creative ways to acquire haem from haemoglobin. Bacteria of all cell wall types have the ability to bind haemoglobin at their cell surface, to accept the haem from it and transport this to the cytoplasm for downstream uses. This review describes the systems employed by various pathogenic bacteria to utilise haemoglobin as an iron source within human hosts and discusses their contribution to virulence.

15.
Org Lett ; 24(43): 7924-7927, 2022 11 04.
Artigo em Inglês | MEDLINE | ID: mdl-36265082

RESUMO

A library of thiazoles and selenothiazoles were synthesized via Ir-catalyzed ylide insertion chemistry. This process is a functional group, particularly heterocycle-substituent tolerant. This was applied to the synthesis of fanetizole, an anti-inflammatory drug, and a thiazole-containing drug fragment that binds to the peptidyl-tRNA hydrolase (Pth) in Neisseria gonorrheae bacteria.


Assuntos
Irídio , Tiazóis , Bactérias , Catálise , Enxofre
16.
Trials ; 23(1): 50, 2022 Jan 17.
Artigo em Inglês | MEDLINE | ID: mdl-35039057

RESUMO

BACKGROUND: It is the investigator's responsibility to communicate the relevant information about a clinical trial to participants before they provide informed consent to take part. Systematic reviews indicate that participants often have a poor understanding of the concepts which are key to ensuring valid informed consent, such as randomisation and risks/discomforts. Paper-based participant information leaflets and informed consent forms (PIL/ICFs) are becoming longer and are often too complex for many participants. Multimedia interventions and enhanced PIL/ICFs have been trialled in an attempt to improve participants' understanding of various aspects of research studies. However, there is insufficient empirical evidence to determine how effective such interventions are. This protocol describes a study to evaluate whether an enhanced PIL/ICF and website help research participants to understand important information about a human immunodeficiency virus (HIV) randomised clinical trial. METHODS: This Study Within A Trial (SWAT) is a prospective, multi-centre, randomised, controlled, parallel-group study embedded in a host clinical trial. The host trial (the SWIFT trial; EudraCT: 2019-002314-39) is a prospective, multi-centre, randomised, open-label, controlled trial investigating if semaglutide along with dietary advice assists individuals with HIV and obesity to lose weight, compared to dietary advice alone. For the SWAT, participants will be randomised in a 1:1 ratio to either the control (standard PIL/ICF) or the intervention (an enhanced PIL/ICF and a website which includes animations). The enhanced PIL/ICF and website were developed in line with the guidance from organisations which promote plain English and accessible public-facing materials in conjunction with HIV Ireland, a HIV advocacy organisation, and our previous work on consent documents. The primary outcome of the SWAT is the quality of informed consent, assessed by a validated comprehension test-the modified Deaconess Informed Consent Comprehension Test (DICCT). The DICCT will be administered within 48 h of consent to the host trial. The secondary is recall, measured by the modified DICCT questionnaire scores 2 weeks post-consent to the host trial. DISCUSSION: The results of this SWAT will add to the methodological evidence base on the use of multimedia to improve the quality of informed consent to randomised clinical trials. TRIAL REGISTRATION: ClinicalTrials.gov NCT04174755 . EudraCT 2019-002314-39. SWAT 160, Northern Ireland Hub for Trials Methodology Research SWAT repository (Clarke M, et al., Trials. 16:P209, 2015).


Assuntos
Infecções por HIV , Multimídia , Infecções por HIV/diagnóstico , Humanos , Consentimento Livre e Esclarecido , Estudos Multicêntricos como Assunto , Obesidade/diagnóstico , Obesidade/terapia , Estudos Prospectivos , Ensaios Clínicos Controlados Aleatórios como Assunto
17.
Mol Pharmacol ; 80(4): 565-72, 2011 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-21719464

RESUMO

Raltegravir (RAL) and related HIV-1 integrase (IN) strand transfer inhibitors (INSTIs) efficiently block viral replication in vitro and suppress viremia in patients. These small molecules bind to the IN active site, causing it to disengage from the deoxyadenosine at the 3' end of viral DNA. The emergence of viral strains that are highly resistant to RAL underscores the pressing need to develop INSTIs with improved resistance profiles. Herein, we show that the candidate second-generation drug dolutegravir (DTG, S/GSK1349572) effectively inhibits a panel of HIV-1 IN variants resistant to first-generation INSTIs. To elucidate the structural basis for the increased potency of DTG against RAL-resistant INs, we determined crystal structures of wild-type and mutant prototype foamy virus intasomes bound to this compound. The overall IN binding mode of DTG is strikingly similar to that of the tricyclic hydroxypyrrole MK-2048. Both second-generation INSTIs occupy almost the same physical space within the IN active site and make contacts with the ß4-α2 loop of the catalytic core domain. The extended linker region connecting the metal chelating core and the halobenzyl group of DTG allows it to enter farther into the pocket vacated by the displaced viral DNA base and to make more intimate contacts with viral DNA, compared with those made by RAL and other INSTIs. In addition, our structures suggest that DTG has the ability to subtly readjust its position and conformation in response to structural changes in the active sites of RAL-resistant INs.


Assuntos
Inibidores de Integrase de HIV/química , Inibidores de Integrase de HIV/metabolismo , Integrase de HIV/metabolismo , Compostos Heterocíclicos com 3 Anéis/química , Compostos Heterocíclicos com 3 Anéis/metabolismo , Sítios de Ligação/fisiologia , Cristalografia por Raios X , Variação Genética/genética , Células HEK293 , Integrase de HIV/química , Inibidores de Integrase de HIV/farmacologia , Compostos Heterocíclicos com 3 Anéis/farmacologia , Humanos , Oxazinas , Piperazinas , Piridonas , Relação Estrutura-Atividade , Células Tumorais Cultivadas
18.
EMBO J ; 26(23): 4926-34, 2007 Nov 28.
Artigo em Inglês | MEDLINE | ID: mdl-17972918

RESUMO

Helicobacter pylori is one of the world's most successful human pathogens causing gastric ulcers and cancers. A key virulence factor of H. pylori is the Cag pathogenicity island, which encodes a type IV secretion system. HP0525 is an essential component of the Cag system and acts as an inner membrane associated ATPase. HP0525 forms double hexameric ring structures, with the C-terminal domains (CTDs) forming a closed ring and the N-terminal domains (NTDs) forming a dynamic, open ring. Here, the crystal structure of HP0525 in complex with a fragment of HP1451, a protein of previously unknown function, is reported. The HP1451 construct consists of two domains similar to nucleic acid-binding domains. Two HP1451 molecules bind to the HP0525 NTDs on opposite sides of the hexamer, locking it in the closed form and forming a partial lid over the HP0525 chamber. From the structure, it is suggested that HP1451 acts as an inhibitory factor of HP0525 to regulate Cag-mediated secretion, a suggestion confirmed by results of in vitro ATPase assay and in vivo pull-down experiments.


Assuntos
Adenosina Trifosfatases/química , Adenosina Trifosfatases/metabolismo , Antígenos de Bactérias/metabolismo , Proteínas de Bactérias/metabolismo , Regulação Bacteriana da Expressão Gênica , Helicobacter pylori/enzimologia , Difosfato de Adenosina/química , Adenosina Trifosfatases/isolamento & purificação , Proteínas de Bactérias/química , Cristalografia por Raios X , Modelos Moleculares , Conformação Molecular , Ligação Proteica , Conformação Proteica , Estrutura Terciária de Proteína
19.
PLoS Pathog ; 5(1): e1000259, 2009 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-19132083

RESUMO

Lens epithelium derived growth factor (LEDGF), also known as PC4 and SFRS1 interacting protein 1 (PSIP1) and transcriptional co-activator p75, is the cellular binding partner of lentiviral integrase (IN) proteins. LEDGF accounts for the characteristic propensity of Lentivirus to integrate within active transcription units and is required for efficient viral replication. We now present a crystal structure containing the N-terminal and catalytic core domains (NTD and CCD) of HIV-2 IN in complex with the IN binding domain (IBD) of LEDGF. The structure extends the known IN-LEDGF interface, elucidating primarily charge-charge interactions between the NTD of IN and the IBD. A constellation of acidic residues on the NTD is characteristic of lentiviral INs, and mutations of the positively charged residues on the IBD severely affect interaction with all lentiviral INs tested. We show that the novel NTD-IBD contacts are critical for stimulation of concerted lentiviral DNA integration by LEDGF in vitro and for its function during the early steps of HIV-1 replication. Furthermore, the new structural details enabled us to engineer a mutant of HIV-1 IN that primarily functions only when presented with a complementary LEDGF mutant. These findings provide structural basis for the high affinity lentiviral IN-LEDGF interaction and pave the way for development of LEDGF-based targeting technologies for gene therapy.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/química , Integrase de HIV/química , Integrase de HIV/genética , HIV-2/enzimologia , Fatores de Transcrição/química , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Cristalografia por Raios X , Integrase de HIV/metabolismo , Proteínas do Vírus da Imunodeficiência Humana , Humanos , Lentivirus , Mutação , Ligação Proteica , Fatores de Transcrição/metabolismo , Replicação Viral
20.
PLoS Pathog ; 5(7): e1000515, 2009 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-19609359

RESUMO

Experimental evidence suggests that a tetramer of integrase (IN) is the protagonist of the concerted strand transfer reaction, whereby both ends of retroviral DNA are inserted into a host cell chromosome. Herein we present two crystal structures containing the N-terminal and the catalytic core domains of maedi-visna virus IN in complex with the IN binding domain of the common lentiviral integration co-factor LEDGF. The structures reveal that the dimer-of-dimers architecture of the IN tetramer is stabilized by swapping N-terminal domains between the inner pair of monomers poised to execute catalytic function. Comparison of four independent IN tetramers in our crystal structures elucidate the basis for the closure of the highly flexible dimer-dimer interface, allowing us to model how a pair of active sites become situated for concerted integration. Using a range of complementary approaches, we demonstrate that the dimer-dimer interface is essential for HIV-1 IN tetramerization, concerted integration in vitro, and virus infectivity. Our structures moreover highlight adaptable changes at the interfaces of individual IN dimers that allow divergent lentiviruses to utilize a highly-conserved, common integration co-factor.


Assuntos
Integrases/química , Proteínas Virais/química , Vírus Visna-Maedi/enzimologia , Cristalografia por Raios X , Integrase de HIV/química , Integrase de HIV/genética , Integrase de HIV/metabolismo , HIV-1/enzimologia , HIV-1/genética , HIV-1/patogenicidade , Integrases/genética , Integrases/metabolismo , Peptídeos e Proteínas de Sinalização Intercelular/química , Modelos Moleculares , Mutação , Domínios e Motivos de Interação entre Proteínas , Multimerização Proteica , Proteínas Virais/genética , Proteínas Virais/metabolismo , Vírus Visna-Maedi/genética , Produtos do Gene vpr do Vírus da Imunodeficiência Humana/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA