Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 8 de 8
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Nature ; 536(7616): 349-53, 2016 08 18.
Artigo em Inglês | MEDLINE | ID: mdl-27509857

RESUMO

During the early stages of infection, the HIV-1 capsid protects viral components from cytosolic sensors and nucleases such as cGAS and TREX, respectively, while allowing access to nucleotides for efficient reverse transcription. Here we show that each capsid hexamer has a size-selective pore bound by a ring of six arginine residues and a 'molecular iris' formed by the amino-terminal ß-hairpin. The arginine ring creates a strongly positively charged channel that recruits the four nucleotides with on-rates that approach diffusion limits. Progressive removal of pore arginines results in a dose-dependent and concomitant decrease in nucleotide affinity, reverse transcription and infectivity. This positively charged channel is universally conserved in lentiviral capsids despite the fact that it is strongly destabilizing without nucleotides to counteract charge repulsion. We also describe a channel inhibitor, hexacarboxybenzene, which competes for nucleotide binding and efficiently blocks encapsidated reverse transcription, demonstrating the tractability of the pore as a novel drug target.


Assuntos
Capsídeo/metabolismo , Replicação do DNA , DNA Viral/biossíntese , HIV-1/metabolismo , Nucleotídeos/metabolismo , Arginina/metabolismo , Benzoatos/farmacologia , Ligação Competitiva/efeitos dos fármacos , Transporte Biológico Ativo/efeitos dos fármacos , Capsídeo/química , Capsídeo/efeitos dos fármacos , Replicação do DNA/efeitos dos fármacos , Difusão , Células HEK293 , HIV-1/efeitos dos fármacos , HIV-1/genética , HIV-1/crescimento & desenvolvimento , Células HeLa , Humanos , Cinética , Modelos Moleculares , Porosidade/efeitos dos fármacos , Transcrição Reversa/efeitos dos fármacos
2.
Nature ; 503(7476): 402-405, 2013 Nov 21.
Artigo em Inglês | MEDLINE | ID: mdl-24196705

RESUMO

Human immunodeficiency virus (HIV)-1 is able to replicate in primary human macrophages without stimulating innate immunity despite reverse transcription of genomic RNA into double-stranded DNA, an activity that might be expected to trigger innate pattern recognition receptors. We reasoned that if correctly orchestrated HIV-1 uncoating and nuclear entry is important for evasion of innate sensors then manipulation of specific interactions between HIV-1 capsid and host factors that putatively regulate these processes should trigger pattern recognition receptors and stimulate type 1 interferon (IFN) secretion. Here we show that HIV-1 capsid mutants N74D and P90A, which are impaired for interaction with cofactors cleavage and polyadenylation specificity factor subunit 6 (CPSF6) and cyclophilins (Nup358 and CypA), respectively, cannot replicate in primary human monocyte-derived macrophages because they trigger innate sensors leading to nuclear translocation of NF-κB and IRF3, the production of soluble type 1 IFN and induction of an antiviral state. Depletion of CPSF6 with short hairpin RNA expression allows wild-type virus to trigger innate sensors and IFN production. In each case, suppressed replication is rescued by IFN-receptor blockade, demonstrating a role for IFN in restriction. IFN production is dependent on viral reverse transcription but not integration, indicating that a viral reverse transcription product comprises the HIV-1 pathogen-associated molecular pattern. Finally, we show that we can pharmacologically induce wild-type HIV-1 infection to stimulate IFN secretion and an antiviral state using a non-immunosuppressive cyclosporine analogue. We conclude that HIV-1 has evolved to use CPSF6 and cyclophilins to cloak its replication, allowing evasion of innate immune sensors and induction of a cell-autonomous innate immune response in primary human macrophages.


Assuntos
HIV-1/imunologia , Evasão da Resposta Imune , Imunidade Inata/imunologia , Macrófagos/imunologia , Macrófagos/virologia , Proteínas do Capsídeo/genética , Proteínas do Capsídeo/metabolismo , Ciclofilinas/metabolismo , Ciclosporina/metabolismo , Infecções por HIV/imunologia , Infecções por HIV/metabolismo , Infecções por HIV/patologia , Infecções por HIV/virologia , HIV-1/metabolismo , Humanos , Fator Regulador 3 de Interferon/metabolismo , Interferon Tipo I/imunologia , Interferon Tipo I/metabolismo , Macrófagos/citologia , Macrófagos/patologia , Chaperonas Moleculares/metabolismo , Monócitos/citologia , NF-kappa B/metabolismo , Complexo de Proteínas Formadoras de Poros Nucleares/metabolismo , Receptores de Reconhecimento de Padrão , Internalização do Vírus , Replicação Viral/imunologia , Fatores de Poliadenilação e Clivagem de mRNA/deficiência , Fatores de Poliadenilação e Clivagem de mRNA/genética , Fatores de Poliadenilação e Clivagem de mRNA/metabolismo
3.
J Biol Chem ; 291(9): 4356-73, 2016 Feb 26.
Artigo em Inglês | MEDLINE | ID: mdl-26679998

RESUMO

The mitochondrial permeability transition pore is a recognized drug target for neurodegenerative conditions such as multiple sclerosis and for ischemia-reperfusion injury in the brain and heart. The peptidylprolyl isomerase, cyclophilin D (CypD, PPIF), is a positive regulator of the pore, and genetic down-regulation or knock-out improves outcomes in disease models. Current inhibitors of peptidylprolyl isomerases show no selectivity between the tightly conserved cyclophilin paralogs and exhibit significant off-target effects, immunosuppression, and toxicity. We therefore designed and synthesized a new mitochondrially targeted CypD inhibitor, JW47, using a quinolinium cation tethered to cyclosporine. X-ray analysis was used to validate the design concept, and biological evaluation revealed selective cellular inhibition of CypD and the permeability transition pore with reduced cellular toxicity compared with cyclosporine. In an experimental autoimmune encephalomyelitis disease model of neurodegeneration in multiple sclerosis, JW47 demonstrated significant protection of axons and improved motor assessments with minimal immunosuppression. These findings suggest that selective CypD inhibition may represent a viable therapeutic strategy for MS and identify quinolinium as a mitochondrial targeting group for in vivo use.


Assuntos
Córtex Cerebral/efeitos dos fármacos , Ciclofilinas/antagonistas & inibidores , Proteínas de Transporte da Membrana Mitocondrial/antagonistas & inibidores , Esclerose Múltipla/prevenção & controle , Neurônios/efeitos dos fármacos , Fármacos Neuroprotetores/uso terapêutico , Compostos de Quinolínio/uso terapêutico , Substituição de Aminoácidos , Animais , Proliferação de Células/efeitos dos fármacos , Células Cultivadas , Córtex Cerebral/imunologia , Córtex Cerebral/metabolismo , Córtex Cerebral/patologia , Peptidil-Prolil Isomerase F , Ciclofilinas/genética , Ciclofilinas/metabolismo , Ciclosporinas/efeitos adversos , Ciclosporinas/síntese química , Ciclosporinas/farmacologia , Ciclosporinas/uso terapêutico , Células Hep G2 , Humanos , Fígado/efeitos dos fármacos , Fígado/metabolismo , Masculino , Camundongos Endogâmicos , Camundongos Knockout , Proteínas de Transporte da Membrana Mitocondrial/genética , Proteínas de Transporte da Membrana Mitocondrial/metabolismo , Poro de Transição de Permeabilidade Mitocondrial , Esclerose Múltipla/imunologia , Esclerose Múltipla/metabolismo , Esclerose Múltipla/patologia , Mutação , Neurônios/imunologia , Neurônios/metabolismo , Neurônios/patologia , Fármacos Neuroprotetores/efeitos adversos , Fármacos Neuroprotetores/farmacologia , Peptídeos Cíclicos/efeitos adversos , Peptídeos Cíclicos/síntese química , Peptídeos Cíclicos/farmacologia , Peptídeos Cíclicos/uso terapêutico , Compostos de Quinolínio/efeitos adversos , Compostos de Quinolínio/síntese química , Compostos de Quinolínio/farmacologia , Distribuição Aleatória , Proteínas Recombinantes/química , Proteínas Recombinantes/metabolismo , Linfócitos T/efeitos dos fármacos , Linfócitos T/patologia
4.
Proc Natl Acad Sci U S A ; 111(26): 9609-14, 2014 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-24979782

RESUMO

Restriction factors (RFs) form important components of host defenses to retroviral infection. The Fv1, Trim5α, and TrimCyp RFs contain N-terminal dimerization and C-terminal specificity domains that target assembled retroviral capsid (CA) proteins enclosing the viral core. However, the molecular detail of the interaction between RFs and their CA targets is unknown. Therefore, we have determined the crystal structure of the B-box and coiled-coil (BCC) region from Trim5α and used small-angle X-ray scattering to examine the solution structure of Trim5α BCC, the dimerization domain of Fv1 (Fv1Ntd), and the hybrid restriction factor Fv1Cyp comprising Fv1NtD fused to the HIV-1 binding protein Cyclophilin A (CypA). These data reveal that coiled-coil regions of Fv1 and Trim5α form extended antiparallel dimers. In Fv1Cyp, two CypA moieties are located at opposing ends, creating a molecule with a dumbbell appearance. In Trim5α, the B-boxes are located at either end of the coiled-coil, held in place by interactions with a helical motif from the L2 region of the opposing monomer. A comparative analysis of Fv1Cyp and CypA binding to a preformed HIV-1 CA lattice reveals how RF dimerization enhances the affinity of interaction through avidity effects. We conclude that the antiparallel organization of the NtD regions of Fv1 and Trim5α dimers correctly positions C-terminal specificity and N-terminal effector domains and facilitates stable binding to adjacent CA hexamers in viral cores.


Assuntos
Capsídeo/metabolismo , HIV-1/metabolismo , Modelos Moleculares , Muramidase/química , Proteínas/química , Internalização do Vírus , Sequência de Aminoácidos , Animais , Bacteriófago T4/enzimologia , Sequência de Bases , Cromatografia em Gel , Cristalização , Dimerização , Escherichia coli , Modelos Lineares , Macaca mulatta , Microscopia Eletrônica , Dados de Sequência Molecular , Conformação Proteica , Proteínas/genética , Proteínas/metabolismo , Proteínas Recombinantes de Fusão/genética , Espalhamento a Baixo Ângulo , Análise de Sequência de DNA , Ressonância de Plasmônio de Superfície , Ubiquitina-Proteína Ligases , Difração de Raios X
5.
Proc Natl Acad Sci U S A ; 108(14): 5771-6, 2011 Apr 05.
Artigo em Inglês | MEDLINE | ID: mdl-21436027

RESUMO

The restriction factor Fv1 confers resistance to murine leukemia virus (MLV), blocking progression of the viral life cycle after reverse transcription, but before integration into the host chromosome. It is known that the specificity of restriction is determined by both the restriction factor and the viral capsid (CA), but a direct interaction between Fv1 and MLV CA has not yet been demonstrated. With the development of a previously unexplored method for in vitro polymerization of MLV CA, it has now been possible to display a binding interaction between Fv1 and MLV CA. C-terminally His-tagged CA molecules were assembled on Ni-chelating lipid nanotubes, and analysis by electron microscopy revealed the formation of a regular lattice. Comparison of binding data with existing restriction data confirmed the specificity of the binding interaction, with multiple positions of both Fv1 and CA shown to influence binding specificity.


Assuntos
Proteínas do Capsídeo/metabolismo , Vírus da Leucemia Murina/metabolismo , Nanotubos/virologia , Ligação Proteica , Proteínas/metabolismo , Animais , Proteínas do Capsídeo/genética , Primers do DNA/genética , Processamento de Imagem Assistida por Computador , Metabolismo dos Lipídeos , Camundongos , Microscopia Eletrônica , Mutagênese , Nanotubos/ultraestrutura , Plasmídeos/genética
6.
Nat Microbiol ; 7(11): 1762-1776, 2022 11.
Artigo em Inglês | MEDLINE | ID: mdl-36289397

RESUMO

Of the 13 known independent zoonoses of simian immunodeficiency viruses to humans, only one, leading to human immunodeficiency virus (HIV) type 1(M) has become pandemic, causing over 80 million human infections. To understand the specific features associated with pandemic human-to-human HIV spread, we compared replication of HIV-1(M) with non-pandemic HIV-(O) and HIV-2 strains in myeloid cell models. We found that non-pandemic HIV lineages replicate less well than HIV-1(M) owing to activation of cGAS and TRIM5-mediated antiviral responses. We applied phylogenetic and X-ray crystallography structural analyses to identify differences between pandemic and non-pandemic HIV capsids. We found that genetic reversal of two specific amino acid adaptations in HIV-1(M) enables activation of TRIM5, cGAS and innate immune responses. We propose a model in which the parental lineage of pandemic HIV-1(M) evolved a capsid that prevents cGAS and TRIM5 triggering, thereby allowing silent replication in myeloid cells. We hypothesize that this capsid adaptation promotes human-to-human spread through avoidance of innate immune response activation.


Assuntos
Infecções por HIV , HIV-1 , Vírus da Imunodeficiência Símia , Animais , Humanos , Filogenia , Vírus da Imunodeficiência Símia/metabolismo , Capsídeo/metabolismo , HIV-1/genética , Nucleotidiltransferases/genética , Nucleotidiltransferases/metabolismo , Infecções por HIV/epidemiologia , Infecções por HIV/metabolismo , Proteínas com Motivo Tripartido/genética , Proteínas com Motivo Tripartido/metabolismo , Ubiquitina-Proteína Ligases/genética , Ubiquitina-Proteína Ligases/metabolismo
7.
Elife ; 92020 06 17.
Artigo em Inglês | MEDLINE | ID: mdl-32553106

RESUMO

The type one interferon induced restriction factor Myxovirus resistance B (MxB) restricts HIV-1 nuclear entry evidenced by inhibition of 2-LTR but not linear forms of viral DNA. The HIV-1 capsid is the key determinant of MxB sensitivity and cofactor binding defective HIV-1 capsid mutants P90A (defective for cyclophilin A and Nup358 recruitment) and N74D (defective for CPSF6 recruitment) have reduced dependency on nuclear transport associated cofactors, altered integration targeting preferences and are not restricted by MxB expression. This has suggested that nuclear import mechanism may determine MxB sensitivity. Here we have use genetics to separate HIV-1 nuclear import cofactor dependence from MxB sensitivity. We provide evidence that MxB sensitivity depends on HIV-1 capsid conformation, rather than cofactor recruitment. We show that depleting CPSF6 to change nuclear import pathway does not impact MxB sensitivity, but mutants that recapitulate the effect of Cyclophilin A binding on capsid conformation and dynamics strongly impact MxB sensitivity. We demonstrate that HIV-1 primary isolates have different MxB sensitivities due to cytotoxic T lymphocyte (CTL) selected differences in Gag sequence but similar cofactor dependencies. Overall our work demonstrates a complex relationship between cyclophilin dependence and MxB sensitivity likely driven by CTL escape. We propose that cyclophilin binding provides conformational flexibility to HIV-1 capsid facilitating simultaneous evasion of capsid-targeting restriction factors including TRIM5 as well as MxB.


Assuntos
Capsídeo/química , HIV-1/fisiologia , Proteínas de Resistência a Myxovirus/química , Transporte Ativo do Núcleo Celular , HIV-1/química , Humanos
8.
Curr Opin Virol ; 4: 32-6, 2014 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-24525292

RESUMO

Lentiviruses have evolved to infect and replicate in a variety of cell types in vivo whilst avoiding the powerful inhibitory activities of restriction factors or cell autonomous innate immune responses. In this review we offer our opinions on how HIV-1 uses a series of host proteins as cofactors for infection. We present a model that may explain how the capsid protein has a fundamental role in the early part of the viral lifecycle by utilising cyclophilin A (CypA), cleavage and polyadenylation specificity factor-6 (CPSF6), Nup358 and TNPO3 to orchestrate a coordinated process of DNA synthesis, capsid uncoating and integration targeting that evades innate responses and promotes integration into preferred areas of chromatin.


Assuntos
Transporte Ativo do Núcleo Celular , HIV-1/fisiologia , Interações Hospedeiro-Patógeno , Transcrição Reversa , Ciclofilina A/metabolismo , Proteína do Núcleo p24 do HIV/metabolismo , Humanos , Chaperonas Moleculares/metabolismo , Complexo de Proteínas Formadoras de Poros Nucleares/metabolismo , beta Carioferinas/metabolismo , Fatores de Poliadenilação e Clivagem de mRNA/metabolismo
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa