Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 6 de 6
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Cell Host Microbe ; 26(2): 203-216.e6, 2019 08 14.
Artigo em Inglês | MEDLINE | ID: mdl-31415753

RESUMO

The HIV-1 capsid is an ordered protein shell that houses the viral genome during early infection. Its expansive surface consists of an ordered and interfacing array of capsid protein hexamers and pentamers that are recognized by numerous cellular proteins. Many of these proteins recognize specific, assembled capsid interfaces not present in unassembled capsid subunits. We used protein-engineering tools to capture diverse capsid assembly intermediates. We built a repertoire of capsid assemblies (ranging from two to 42 capsid protein molecules) that recreate the various surfaces in infectious capsids. These assemblies reveal unique capsid-targeting mechanisms for each of the anti-HIV factors, TRIMCyp, MxB, and TRIM5α, linked to inhibition of virus uncoating and nuclear entry, as well as the HIV-1 cofactor FEZ1 that facilitates virus intracellular trafficking. This capsid assembly repertoire enables elucidation of capsid recognition modes by known capsid-interacting factors, identification of new capsid-interacting factors, and potentially, development of capsid-targeting therapeutics.


Assuntos
Proteínas do Capsídeo/química , Proteínas do Capsídeo/ultraestrutura , Capsídeo/química , Capsídeo/ultraestrutura , HIV-1/fisiologia , HIV-1/ultraestrutura , Animais , Fármacos Anti-HIV/farmacologia , Fatores de Restrição Antivirais , Capsídeo/metabolismo , Proteínas do Capsídeo/genética , Proteínas do Capsídeo/metabolismo , Proteínas de Transporte/metabolismo , HIV-1/genética , Humanos , Macaca fascicularis , Macaca mulatta , Proteínas de Resistência a Myxovirus , Ligação Proteica , Domínios Proteicos , Engenharia de Proteínas , Domínios e Motivos de Interação entre Proteínas , Proteínas Recombinantes de Fusão , Proteínas com Motivo Tripartido , Ubiquitina-Proteína Ligases
2.
Mol Immunol ; 67(2 Pt B): 193-205, 2015 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-26052070

RESUMO

The complement system plays a central role in a number of human inflammatory diseases, and there is a significant need for development of complement-directed therapies. The discovery of an arsenal of anti-complement proteins secreted by the pathogen Staphylococcus aureus brought with it the potential for harnessing the powerful inhibitory properties of these molecules. One such family of inhibitors, the SCINs, interact with a functional "hot-spot" on the surface of C3b. SCINs not only stabilize an inactive form of the alternative pathway (AP) C3 convertase (C3bBb), but also overlap the C3b binding site of complement factors B and H. Here we determined that a conserved Arg residue in SCINs is critical for function of full-length SCIN proteins. Despite this, we also found SCIN-specific differences in the contributions of other residues found at the C3b contact site, which suggested that a more diverse repertoire of residues might be able to recognize this region of C3b. To investigate this possibility, we conducted a phage display screen aimed at identifying SCIN-competitive 12-mer peptides. In total, seven unique sequences were identified and all exhibited direct C3b binding. A subset of these specifically inhibited the AP in assays of complement function. The mechanism of AP inhibition by these peptides was probed through surface plasmon resonance approaches, which revealed that six of the seven peptides disrupted C3bBb formation by interfering with factor B/C3b binding. To our knowledge this study has identified the first small molecules that retain inhibitory properties of larger staphylococcal immune evasion proteins.


Assuntos
Proteínas de Bactérias/metabolismo , Via Alternativa do Complemento/efeitos dos fármacos , Peptídeos/farmacologia , Staphylococcus aureus/metabolismo , Animais , Arginina/metabolismo , Ligação Competitiva/efeitos dos fármacos , C3 Convertase da Via Alternativa do Complemento , Complemento C3b/metabolismo , Fator B do Complemento/metabolismo , Sequência Conservada , Humanos , Proteínas Mutantes/química , Proteínas Mutantes/metabolismo , Biblioteca de Peptídeos , Peptídeos/química , Ligação Proteica/efeitos dos fármacos , Estrutura Secundária de Proteína , Coelhos , Soluções , Ressonância de Plasmônio de Superfície
3.
PLoS One ; 10(3): e0118911, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-25775123

RESUMO

Natural infection of Anopheles gambiae by malaria-causing Plasmodium parasites is significantly influenced by the APL1 genetic locus. The locus contains three closely related leucine-rich repeat (LRR) genes, APL1A, APL1B and APL1C. Multiple studies have reported the participation of APL1A-C in the immune response of A. gambiae to invasion by both rodent and human Plasmodium isolates. APL1C forms a heterodimer with the related LRR protein LRIM1 via a C-terminal coiled-coil domain that is also present in APL1A and APL1B. The LRIM1/APL1C heterodimer protects A. gambiae from infection by binding the complement-like protein TEP1 to form a stable and active immune complex. Here we report solution x-ray scatting data for the LRIM1/APL1C heterodimer, the oligomeric state of LRIM1/APL1 LRR domains in solution and the crystal structure of the APL1B LRR domain. The LRIM1/APL1C heterodimeric complex has a flexible and extended structure in solution. In contrast to the APL1A, APL1C and LRIM1 LRR domains, the APL1B LRR domain is a homodimer. The crystal structure of APL1B-LRR shows that the homodimer is formed by an N-terminal helix that complements for the absence of an N-terminal capping motif in APL1B, which is a unique distinction within the LRIM1/APL1 protein family. Full-length APL1A1 and APL1B form a stable complex with LRIM1. These results support a model in which APL1A1, APL1B and APL1C can all form an extended, flexible heterodimer with LRIM1, providing a repertoire of functional innate immune complexes to protect A. gambiae from a diverse array of pathogens.


Assuntos
Anopheles/química , Anopheles/imunologia , Proteínas de Insetos/metabolismo , Mapas de Interação de Proteínas , Proteínas/metabolismo , Sequência de Aminoácidos , Animais , Anopheles/metabolismo , Anopheles/parasitologia , Humanos , Proteínas de Insetos/química , Proteínas de Insetos/genética , Proteínas de Insetos/imunologia , Proteínas de Repetições Ricas em Leucina , Malária/transmissão , Modelos Moleculares , Dados de Sequência Molecular , Plasmodium/fisiologia , Conformação Proteica , Multimerização Proteica , Estrutura Terciária de Proteína , Proteínas/química , Proteínas/imunologia , Alinhamento de Sequência
4.
Cell Host Microbe ; 16(5): 627-638, 2014 Nov 12.
Artigo em Inglês | MEDLINE | ID: mdl-25312384

RESUMO

The myxovirus resistance (Mx) proteins are interferon-induced dynamin GTPases that can inhibit a variety of viruses. Recently, MxB, but not MxA, was shown to restrict HIV-1 by an unknown mechanism that likely occurs in close proximity to the host cell nucleus and involves the viral capsid. Here, we present the crystal structure of MxB and reveal determinants involved in HIV-1 restriction. MxB adopts an extended antiparallel dimer and dimerization, but not higher-ordered oligomerization, is critical for restriction. Although MxB is structurally similar to MxA, the orientation of individual domains differs between MxA and MxB, and their antiviral functions rely on separate determinants, indicating distinct mechanisms for virus inhibition. Additionally, MxB directly binds the HIV-1 capsid, and this interaction depends on dimerization and the N terminus of MxB as well as the assembled capsid lattice. These insights establish a framework for understanding the mechanism by which MxB restricts HIV-1.


Assuntos
HIV-1/fisiologia , Proteínas de Resistência a Myxovirus/química , Capsídeo/metabolismo , Capsídeo/virologia , Linhagem Celular Tumoral , Cristalografia por Raios X , Regulação da Expressão Gênica , Interações Hospedeiro-Patógeno , Humanos , Microscopia Confocal , Conformação Proteica , Multimerização Proteica , Montagem de Vírus
5.
J Biol Chem ; 288(4): 2870-81, 2013 Jan 25.
Artigo em Inglês | MEDLINE | ID: mdl-23233676

RESUMO

Complement is a network of interacting circulatory and cell surface proteins that recognizes, marks, and facilitates clearance of microbial invaders. To evade complement attack, the pathogenic organism Staphylococcus aureus expresses a number of secreted proteins that interfere with activation and regulation of the complement cascade. Staphylococcal complement inhibitors (SCINs) are one important class of these immunomodulators and consist of three active members (SCIN-A/-B/-C). SCINs inhibit a critical enzymatic complex, the alternative pathway C3 convertase, by targeting a functional "hot spot" on the central opsonin of complement, C3b. Although N-terminal truncation mutants of SCINs retain complement inhibitory properties, they are significantly weaker binders of C3b. To provide a structural basis for this observation, we undertook a series of crystallographic and NMR dynamics studies on full-length SCINs. This work reveals that N-terminal SCIN domains are characterized by a conformationally dynamic helical motif. C3b binding and functional experiments further demonstrate that this sequence-divergent N-terminal region of SCINs is both functionally important and context-dependent. Finally, surface plasmon resonance data provide evidence for the formation of inhibitor·enzyme·substrate complexes ((SCIN·C3bBb)·C3). Similar to the (SCIN·C3bBb)(2) pseudodimeric complexes, ((SCIN·C3bBb)·C3) interferes with the interaction of complement receptors and C3b. This activity provides an additional mechanism by which SCIN couples convertase inhibition to direct blocking of phagocytosis. Together, these data suggest that tethering multi-host protein complexes by small modular bacterial inhibitors may be a global strategy of immune evasion used by S. aureus. The work presented here provides detailed structure-activity relationships and improves our understanding of how S. aureus circumvents human innate immunity.


Assuntos
C3 Convertase da Via Alternativa do Complemento/química , Complemento C3b/química , Proteínas Inativadoras do Complemento/metabolismo , Staphylococcus aureus/metabolismo , Proteínas Inativadoras do Complemento/química , Reagentes de Ligações Cruzadas/química , Cristalografia por Raios X/métodos , Humanos , Sistema Imunitário , Imunidade Inata , Espectroscopia de Ressonância Magnética/métodos , Fagocitose , Ligação Proteica , Conformação Proteica , Mapeamento de Interação de Proteínas/métodos , Estrutura Terciária de Proteína , Ressonância de Plasmônio de Superfície
6.
J Biol Chem ; 287(1): 628-640, 2012 Jan 02.
Artigo em Inglês | MEDLINE | ID: mdl-22086928

RESUMO

To survive in immune-competent hosts, the pathogen Staphylococcus aureus expresses and secretes a sophisticated array of proteins that inhibit the complement system. Among these are the staphylococcal complement inhibitors (SCIN), which are composed of three active proteins (SCIN-A, -B, and -C) and one purportedly inactive member (SCIN-D or ORF-D). Because previous work has focused almost exclusively on SCIN-A, we sought to provide initial structure/function information on additional SCIN proteins. To this end we determined crystal structures of an active, N-terminal truncation mutant of SCIN-B (denoted SCIN-B18-85) both free and bound to the C3c fragment of complement component C3 at 1.5 and 3.4 Å resolution, respectively. Comparison of the C3c/SCIN-B18-85 structure with that of C3c/SCIN-A revealed that both proteins target the same functional hotspot on the C3b/C3c surface yet harbor diversity in both the type of residues and interactions formed at their C3b/C3c interfaces. Most importantly, these structures allowed identification of Arg44 and Tyr51 as residues key for SCIN-B binding to C3b and subsequent inhibition of the AP C3 convertase. In addition, we also solved several crystal structures of SCIN-D to 1.3 Å limiting resolution. This revealed an unexpected structural deviation in the N-terminal α helix relative to SCIN-A and SCIN-B. Comparative analysis of both electrostatic potentials and surface complementarity suggest a physical explanation for the inability of SCIN-D to bind C3b/C3c. Together, these studies provide a more thorough understanding of immune evasion by S. aureus and enhance potential use of SCIN proteins as templates for design of complement targeted therapeutics.


Assuntos
Proteínas de Bactérias/química , Proteínas de Bactérias/metabolismo , Convertases de Complemento C3-C5/metabolismo , Complemento C3b/metabolismo , Staphylococcus aureus/metabolismo , Animais , Proteínas de Bactérias/farmacologia , Convertases de Complemento C3-C5/antagonistas & inibidores , Complemento C3c/metabolismo , Cristalografia por Raios X , Humanos , Modelos Moleculares , Estrutura Terciária de Proteína
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...