Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 19 de 19
Filtrar
1.
J Virol ; 90(9): 4681-4695, 2016 May.
Artigo em Inglês | MEDLINE | ID: mdl-26912618

RESUMO

UNLABELLED: The multifunctional HIV-1 accessory protein Vif counters the antiviral activities of APOBEC3G (A3G) and APOBEC3F (A3F), and some Vifs counter stable alleles of APOBEC3H (A3H). Studies in humanized mice have shown that HIV-1 lacking Vif expression is not viable. Here, we look at the relative contributions of the three APOBEC3s to viral extinction. Inoculation of bone marrow/liver/thymus (BLT) mice with CCR5-tropic HIV-1JRCSF(JRCSF) expressing a vif gene inactive for A3G but not A3F degradation activity (JRCSFvifH42/43D) displayed either no or delayed replication. JRCSF expressing a vif gene mutated to inactivate A3F degradation but not A3G degradation (JRCSFvifW79S) always replicated to high viral loads with variable delays. JRCSF with vif mutated to lack both A3G and A3F degradation activities (JRCSFvifH42/43DW79S) failed to replicate, mimicking JRCSF without Vif expression (JRCSFΔvif). JRCSF and JRCSFvifH42/43D, but not JRCSFvifW79S or JRCSFvifH42/43DW79S, degraded APOBEC3D. With one exception, JRCSFs expressing mutant Vifs that replicated acquired enforced vif mutations. These mutations partially restored A3G or A3F degradation activity and fully replaced JRCSFvifH42/43D or JRCSFvifW79S by 10 weeks. Surprisingly, induced mutations temporally lagged behind high levels of virus in blood. In the exceptional case, JRCSFvifH42/43D replicated after a prolonged delay with no mutations in vif but instead a V27I mutation in the RNase H coding sequence. JRCSFvifH42/43D infections exhibited massive GG/AG mutations in pol viral DNA, but in viral RNA, there were no fixed mutations in the Gag or reverse transcriptase coding sequence. A3H did not contribute to viral extinction but, in combination with A3F, could delay JRCSF replication. A3H was also found to hypermutate viral DNA. IMPORTANCE: Vif degradation of A3G and A3F enhances viral fitness, as virus with even a partially restored capacity for degradation outgrows JRCSFvifH42/43D and JRCSFvifW79S. Unexpectedly, fixation of mutations that replaced H42/43D or W79S in viral RNA lagged behind the appearance of high viral loads. In one exceptional JRCSFvifH42/43D infection, vif was unchanged but replication proceeded after a long delay. These results suggest that Vif binds and inhibits the non-cytosine deaminase activities of intact A3G and intact A3F, allowing JRCSFvifH42/43D and JRCSFvifW79S to replicate with reduced fitness. Subsequently, enhanced Vif function is acquired by enforced mutations. In infected cells, JRCSFΔvif and JRCSFvifH42/43DW79S are exposed to active A3F and A3G and fail to replicate. JRCSFvifH42/43D Vif degrades A3F and, in some cases, overcomes A3G mutagenic activity to replicate. Vif may have evolved to inhibit A3F and A3G by stoichiometric binding and subsequently acquired the ability to target these proteins to proteasomes.


Assuntos
Citidina Desaminase/metabolismo , Citosina Desaminase/metabolismo , Infecções por HIV/metabolismo , Infecções por HIV/virologia , HIV-1/fisiologia , Replicação Viral , Desaminase APOBEC-3G , Alelos , Aminoidrolases/genética , Aminoidrolases/metabolismo , Animais , Sequência de Bases , Linhagem Celular , Citidina Desaminase/genética , Citosina Desaminase/genética , Análise Mutacional de DNA , DNA Viral , Modelos Animais de Doenças , Amplificação de Genes , Infecções por HIV/imunologia , Haplótipos , Humanos , Camundongos , Camundongos Transgênicos , Dados de Sequência Molecular , Mutação , Ligação Proteica , Proteólise , Alinhamento de Sequência , Carga Viral , Produtos do Gene vif do Vírus da Imunodeficiência Humana/química , Produtos do Gene vif do Vírus da Imunodeficiência Humana/genética , Produtos do Gene vif do Vírus da Imunodeficiência Humana/metabolismo
2.
Retrovirology ; 12: 61, 2015 Jul 14.
Artigo em Inglês | MEDLINE | ID: mdl-26169178

RESUMO

BACKGROUND: Nef is a multifunctional HIV-1 protein critical for progression to AIDS. Humans infected with nef(-) HIV-1 have greatly delayed or no disease consequences. We have contrasted nef(-) and nef(+) infection of BLT humanized mice to better characterize Nef's pathogenic effects. RESULTS: Mice were inoculated with CCR5-tropic HIV-1JRCSF (JRCSF) or JRCSF with an irreversibly inactivated nef (JRCSFNefdd). In peripheral blood (PB), JRCSF exhibited high levels of viral RNA (peak viral loads of 4.71 × 10(6) ± 1.23 × 10(6) copies/ml) and a progressive, 75% loss of CD4(+) T cells over 17 weeks. Similar losses were observed in CD4(+) T cells from bone marrow, spleen, lymph node, lung and liver but thymocytes were not significantly decreased. JRCSFNefdd also had high peak viral loads (2.31 × 10(6) ± 1.67 × 10(6)) but induced no loss of PB CD4(+) T cells. In organs, JRCSFNefdd produced small, but significant, reductions in CD4(+) T cell levels and did not affect the level of thymocytes. Uninfected mice have low levels of HLA-DR(+)CD38(+)CD8(+) T cells in blood (1-2%). Six weeks post inoculation, JRCSF infection resulted in significantly elevated levels of activated CD8(+) T cells (6.37 ± 1.07%). T cell activation coincided with PB CD4(+) T cell loss which suggests a common Nef-dependent mechanism. At 12 weeks, in JRCSF infected animals PB T cell activation sharply increased to 19.7 ± 2.9% then subsided to 5.4 ± 1.4% at 14 weeks. HLA-DR(+)CD38(+)CD8(+) T cell levels in JRCSFNefdd infected mice did not rise above 1-2% despite sustained high levels of viremia. Interestingly, we also noted that in mice engrafted with human tissue expressing a putative protective HLA-B allele (B42:01), JRCSFNefdd exhibited a substantial (200-fold) reduced viral load compared to JRCSF. CONCLUSIONS: Nef expression was necessary for both systemic T cell activation and substantial CD4(+) T cell loss from blood and tissues. JRCSFNefdd infection did not activate CD8(+) T cells or reduce the level of CD4(+) T cells in blood but did result in a small Nef-independent decrease in CD4(+) T cells in organs. These observations strongly support the conclusion that viral pathogenicity is mostly driven by Nef. We also observed for the first time substantial host-specific suppression of HIV-1 replication in a small animal infection model.


Assuntos
Linfócitos T CD4-Positivos/imunologia , Infecções por HIV/imunologia , HIV-1/fisiologia , Interações Hospedeiro-Patógeno , Ativação Linfocitária , Replicação Viral , Produtos do Gene nef do Vírus da Imunodeficiência Humana/metabolismo , Estruturas Animais/imunologia , Animais , Sangue/imunologia , Linfócitos T CD4-Positivos/virologia , Linfócitos T CD8-Positivos/imunologia , Infecções por HIV/virologia , Humanos , Camundongos , Camundongos SCID
3.
Retrovirology ; 10: 125, 2013 Oct 30.
Artigo em Inglês | MEDLINE | ID: mdl-24172637

RESUMO

BACKGROUND: The HIV-1 accessory protein, Nef, is decisive for progression to AIDS. In vitro characterization of the protein has described many Nef activities of unknown in vivo significance including CD4 downregulation and a number of activities that depend on Nef interacting with host SH3 domain proteins. Here, we use the BLT humanized mouse model of HIV-1 infection to assess their impact on viral replication and pathogenesis and the selection pressure to restore these activities using enforced in vivo evolution. RESULTS: We followed the evolution of HIV-1LAI (LAI) with a frame-shifted nef (LAINeffs) during infection of BLT mice. LAINeffs was rapidly replaced in blood by virus with short deletions in nef that restored the open reading frame (LAINeffs∆-1 and LAINeffs∆-13). Subsequently, LAINeffs∆-1 was often replaced by wild type LAI. Unexpectedly, LAINeffs∆-1 and LAINeffs∆-13 Nefs were specifically defective for CD4 downregulation activity. Viruses with these mutant nefs were used to infect BLT mice. LAINeffs∆-1 and LAINeffs∆-13 exhibited three-fold reduced viral replication (compared to LAI) and a 50% reduction of systemic CD4+ T cells (>90% for LAI) demonstrating the importance of CD4 downregulation. These results also demonstrate that functions other than CD4 downregulation enhanced viral replication and pathogenesis of LAINeffs∆-1 and LAINeffs∆-13 compared to LAINeffs. To gain insight into the nature of these activities, we constructed the double mutant P72A/P75A. Multiple Nef activities can be negated by mutating the SH3 domain binding site (P72Q73V74P75L76R77) to P72A/P75A and this mutation does not affect CD4 downregulation. Virus with nef mutated to P72A/P75A closely resembled the wild-type virus in vivo as viral replication and pathogenesis was not significantly altered. Unlike LAINeffs described above, the P72A/P75A mutation had a very weak tendency to revert to wild type sequence. CONCLUSIONS: The in vivo phenotype of Nef is significantly dependent on CD4 downregulation but minimally on the numerous Nef activities that require an intact SH3 domain binding motif. These results suggest that CD4 downregulation plus one or more unknown Nef activities contribute to enhanced viral replication and pathogenesis and are suitable targets for anti-HIV therapy. Enforced evolution studies in BLT mice will greatly facilitate identification of these critical activities.


Assuntos
HIV-1/fisiologia , Replicação Viral , Produtos do Gene nef do Vírus da Imunodeficiência Humana/metabolismo , Animais , Antígenos CD4/biossíntese , Antígenos CD4/imunologia , Regulação para Baixo , Mutação da Fase de Leitura , HIV-1/patogenicidade , Camundongos , Camundongos SCID , Mutação de Sentido Incorreto , Deleção de Sequência , Produtos do Gene nef do Vírus da Imunodeficiência Humana/genética
4.
PLoS Pathog ; 9(3): e1003242, 2013 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-23555255

RESUMO

Innate immune restriction factors represent important specialized barriers to zoonotic transmission of viruses. Significant consideration has been given to their possible use for therapeutic benefit. The apolipoprotein B mRNA editing enzyme catalytic polypeptide 3 (APOBEC3) family of cytidine deaminases are potent immune defense molecules capable of efficiently restricting endogenous retroelements as well as a broad range of viruses including Human Immunodeficiency virus (HIV), Hepatitis B virus (HBV), Human Papilloma virus (HPV), and Human T Cell Leukemia virus (HTLV). The best characterized members of this family are APOBEC3G (A3G) and APOBEC3F (A3F) and their restriction of HIV. HIV has evolved to counteract these powerful restriction factors by encoding an accessory gene designated viral infectivity factor (vif). Here we demonstrate that APOBEC3 efficiently restricts CCR5-tropic HIV in the absence of Vif. However, our results also show that CXCR4-tropic HIV can escape from APOBEC3 restriction and replicate in vivo independent of Vif. Molecular analysis identified thymocytes as cells with reduced A3G and A3F expression. Direct injection of vif-defective HIV into the thymus resulted in viral replication and dissemination detected by plasma viral load analysis; however, vif-defective viruses remained sensitive to APOBEC3 restriction as extensive G to A mutation was observed in proviral DNA recovered from other organs. Remarkably, HIV replication persisted despite the inability of HIV to develop resistance to APOBEC3 in the absence of Vif. Our results provide novel insight into a highly specific subset of cells that potentially circumvent the action of APOBEC3; however our results also demonstrate the massive inactivation of CCR5-tropic HIV in the absence of Vif.


Assuntos
Citosina Desaminase/genética , Infecções por HIV/imunologia , HIV-1/imunologia , Replicação Viral/genética , Desaminases APOBEC , Animais , Sequência de Bases , Citidina Desaminase , Citosina Desaminase/metabolismo , DNA Viral , Modelos Animais de Doenças , Infecções por HIV/virologia , Interações Hospedeiro-Patógeno , Humanos , Camundongos , Camundongos Transgênicos , Dados de Sequência Molecular , Organismos Livres de Patógenos Específicos , Timócitos/metabolismo , Timócitos/virologia , Produtos do Gene vif do Vírus da Imunodeficiência Humana/genética
5.
Retrovirology ; 9: 47, 2012 May 31.
Artigo em Inglês | MEDLINE | ID: mdl-22651890

RESUMO

BACKGROUND: HIV-1 Nef is a multifunctional protein required for full pathogenicity of the virus. As Nef has no known enzymatic activity, it necessarily functions through protein-protein interaction interfaces. A critical Nef protein interaction interface is centered on its polyproline segment (P69VRPQVPLRP78) which contains the helical SH3 domain binding protein motif, PXXPXR. We hypothesized that any Nef-SH3 domain interactions would be lost upon mutation of the prolines or arginine of PXXPXR. Further, mutation of the non-motif "X" residues, (Q73, V74, and L75) would give altered patterns of inhibition for different Nef/SH3 domain protein interactions. RESULTS: We found that mutations of either of the prolines or the arginine of PXXPXR are defective for Nef-Hck binding, Nef/activated PAK2 complex formation and enhancement of virion infectivity (EVI). Mutation of the non-motif "X" residues (Q, V and L) gave similar patterns of inhibition for Nef/activated PAK2 complex formation and EVI which were distinct from the pattern for Hck binding. These results implicate an SH3 domain containing protein other than Hck for Nef/activated PAK2 complex formation and EVI. We have also mutated Nef residues at the N-and C-terminal ends of the polyproline segment to explore interactions outside of PXXPXR. We discovered a new locus GFP/F (G67, F68, P69 and F90) that is required for Nef/activated PAK2 complex formation and EVI.MHC Class I (MHCI) downregulation was only partially inhibited by mutating the PXXPXR motif residues, but was fully inhibited by mutating the C-terminal P78. Further, we observed that MHCI downregulation strictly requires G67 and F68. Our mutational analysis confirms the recently reported structure of the complex between Nef, AP-1 µ1 and the cytoplasmic tail of MHCI, but does not support involvement of an SH3 domain protein in MHCI downregulation. CONCLUSION: Nef has evolved to be dependent on interactions with multiple SH3 domain proteins. To the N- and C- terminal sides of the polyproline helix are multifunctional protein interaction sites. The polyproline segment is also adapted to downregulate MHCI with a non-canonical binding surface. Our results demonstrate that Nef polyproline helix is highly adapted to directly interact with multiple host cell proteins.


Assuntos
HIV-1/química , Peptídeos/química , Produtos do Gene nef do Vírus da Imunodeficiência Humana/química , Adaptação Biológica , Motivos de Aminoácidos , Arginina/química , Western Blotting , Genes MHC Classe I , Células HEK293 , Infecções por HIV/virologia , HIV-1/genética , HIV-1/patogenicidade , Células HeLa , Humanos , Mutação , Prolina/química , Ligação Proteica , Mapeamento de Interação de Proteínas , Transfecção , Domínios de Homologia de src
6.
Retrovirology ; 9: 44, 2012 May 28.
Artigo em Inglês | MEDLINE | ID: mdl-22640559

RESUMO

BACKGROUND: The outcome of untreated HIV-1 infection is progression to AIDS and death in nearly all cases. Some important exceptions are the small number of patients infected with HIV-1 deleted for the accessory gene, nef. With these infections, disease progression is entirely suppressed or greatly delayed. Whether Nef is critical for high levels of replication or is directly cytotoxic remains controversial. The major problem in determining the role of Nef in HIV/AIDS has been the lack of tractable in vivo models where Nef's complex pathogenic phenotype can be recapitulated. RESULTS: Intravenous inoculation (3000 to 600,000 TCIU) of BLT humanized mice with HIV-1LAI reproducibly establishes a systemic infection. HIV-1LAI (LAI) replicates to high levels (peak viral load in blood 8,200,000 ± 1,800,000 copies of viral RNA/ml, range 3,600,000 to 20,400,000; n = 9) and exhaustively depletes CD4+ T cells in blood and tissues. CD4+CD8+ thymocytes were also efficiently depleted but CD4+CD8- thymocytes were partially resistant to cell killing by LAI. Infection with a nef-deleted LAI (LAINefdd) gave lower peak viral loads (1,220,000 ± 330,000, range 27,000 to 4,240,000; n = 17). For fourteen of seventeen LAINefdd-infected mice, there was little to no loss of either CD4+ T cells or thymocytes. Both LAI- and LAINefdd-infected mice had about 8% of total peripheral blood CD8+ T cells that were CD38+HLA-DR+ compared <1% for uninfected mice. Three exceptional LAINefdd-infected mice that lost CD4+ T cells received 600,000 TCIU. All three exhibited peak viral loads over 3,000,000 copies of LAINefdd RNA/ml. Over an extended time course, substantial systemic CD4+ T cell loss was observed for the three mice, but there was no loss of CD4+CD8+ or CD4+CD8- thymocytes. CONCLUSION: We conclude Nef is necessary for elevated viral replication and as a result indirectly contributes to CD4+ T cell killing. Further, Nef was not necessary for the activation of peripheral blood CD8+ T cells following infection. However, CD4+CD8+ thymocyte killing was dependent on Nef even in cases of elevated LAINefdd replication and T cell loss. This depletion of thymic T cell precursors may be a significant factor in the elevated pathogenicity of CXCR4 trophic HIV-1.


Assuntos
Antígenos CD4/metabolismo , Antígenos CD8/metabolismo , HIV-1/fisiologia , Timócitos/virologia , Replicação Viral , Produtos do Gene nef do Vírus da Imunodeficiência Humana/metabolismo , Síndrome da Imunodeficiência Adquirida/virologia , Sequência de Aminoácidos , Animais , Antígenos CD34/metabolismo , Linfócitos T CD4-Positivos/metabolismo , Linfócitos T CD4-Positivos/virologia , Linfócitos T CD8-Positivos/metabolismo , Linfócitos T CD8-Positivos/virologia , Células HEK293 , HIV-1/genética , HIV-1/patogenicidade , Humanos , Ativação Linfocitária , Camundongos , Camundongos Endogâmicos NOD , Camundongos SCID , Dados de Sequência Molecular , RNA Viral/metabolismo , Receptores CXCR4/metabolismo , Timócitos/metabolismo , Fatores de Tempo , Carga Viral , Produtos do Gene nef do Vírus da Imunodeficiência Humana/genética
7.
J Neuroimmune Pharmacol ; 6(2): 230-46, 2011 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-21336563

RESUMO

Advances in the last several years have enhanced mechanistic understanding of Nef-induced CD4 and MHCI downregulation and have suggested a new paradigm for analyzing Nef function. In both of these cases, Nef acts by forming ternary complexes with significant contributions to stability imparted by non-canonical interactions. The mutational analyses and binding assays that have led to these conclusions are discussed. The recent progress has been dependent on conservative mutations and multi-protein binding assays. The poorly understood Nef functions of p21 activated protein kinase (PAK2) activation, enhancement of virion infectivity, and inhibition of immunoglobulin class switching are also likely to involve ternary complexes and non-canonical interactions. Hence, investigation of these latter Nef functions should benefit from a similar approach. Six historically used alanine substitutions for determining structure-function relationships of Nef are discussed. These are M20A, E62A/E63A/E64A/E65A (AAAA), P72A/P75A (AXXA), R106A, L164A/L165A, and D174A/D175A. Investigations of less-disruptive mutations in place of AAAA and AXXA have led to different interpretations of mechanism. Two recent examples of this alternate approach, F191I for studying PAK2 activation and D123E for the critical residue D123 are discussed. The implications of the new findings and the resulting new paradigm for Nef structure-function are discussed with respect to creating a map of Nef functions on the protein surface. We report the results of a PPI-Pred analysis for protein-protein interfaces. There are three predicted patches produced by the analysis which describe regions consistent with the currently known mutational analyses of Nef function.


Assuntos
HIV-1/imunologia , Líquido Intracelular/imunologia , Transdução de Sinais/imunologia , Produtos do Gene nef do Vírus da Imunodeficiência Humana/fisiologia , Animais , Antígenos CD4/fisiologia , Regulação para Baixo/imunologia , Antígenos de Histocompatibilidade Classe I/fisiologia , Humanos , Produtos do Gene nef do Vírus da Imunodeficiência Humana/metabolismo
8.
Retrovirology ; 7: 77, 2010 Sep 23.
Artigo em Inglês | MEDLINE | ID: mdl-20863404

RESUMO

BACKGROUND: The HIV-1 pathogenic factor, Nef, is a multifunctional protein present in the cytosol and on membranes of infected cells. It has been proposed that a spatial and temporal regulation of the conformation of Nef sequentially matches Nef's multiple functions to the process of virion production. Further, it has been suggested that dimerization is required for multiple Nef activities. A dimerization interface has been proposed based on intermolecular contacts between Nefs within hexagonal Nef/FynSH3 crystals. The proposed dimerization interface consists of the hydrophobic B-helix and flanking salt bridges between R105 and D123. Here, we test whether Nef self-association is mediated by this interface and address the overall significance of oligomerization. RESULTS: By co-immunoprecipitation assays, we demonstrated that HIV-1Nef exists as monomers and oligomers with about half of the Nef protomers oligomerized. Nef oligomers were found to be present in the cytosol and on membranes. Removal of the myristate did not enhance the oligomerization of soluble Nef. Also, SIVNef oligomerizes despite lacking a dimerization interface functionally homologous to that proposed for HIV-1Nef. Moreover, HIV-1Nef and SIVNef form hetero-oligomers demonstrating the existence of homologous oligomerization interfaces that are distinct from that previously proposed (R105-D123). Intracellular cross-linking by formaldehyde confirmed that SF2Nef dimers are present in intact cells, but surprisingly self-association was dependent on R105, but not D123. SIV(MAC239)Nef can be cross-linked at its only cysteine, C55, and SF2Nef is also cross-linked, but at C206 instead of C55, suggesting that Nefs exhibit multiple dimeric structures. ClusPro dimerization analysis of HIV-1Nef homodimers and HIV-1Nef/SIVNef heterodimers identified a new potential dimerization interface, including a dibasic motif at R105-R106 and a six amino acid hydrophobic surface. CONCLUSIONS: We have demonstrated significant levels of intracellular Nef oligomers by immunoprecipitation from cellular extracts. However, our results are contrary to the identification of salt bridges between R105 and D123 as necessary for self-association. Importantly, binding between HIV-1Nef and SIVNef demonstrates evolutionary conservation and therefore significant function(s) for oligomerization. Based on modeling studies of Nef self-association, we propose a new dimerization interface. Finally, our findings support a stochastic model of Nef function with a dispersed intracellular distribution of Nef oligomers.


Assuntos
HIV-1/metabolismo , Produtos do Gene nef do Vírus da Imunodeficiência Humana/química , Linhagem Celular , Linhagem Celular Tumoral , Membrana Celular/metabolismo , Citosol/metabolismo , Humanos , Imunoprecipitação , Modelos Moleculares , Conformação Proteica , Multimerização Proteica , Vírus da Imunodeficiência Símia/metabolismo , Produtos do Gene nef do Vírus da Imunodeficiência Humana/metabolismo
9.
Retrovirology ; 5: 84, 2008 Sep 22.
Artigo em Inglês | MEDLINE | ID: mdl-18808677

RESUMO

The development of anti-virals has blunted the AIDS epidemic in the Western world but globally the epidemic has not been curtailed. Standard vaccines have not worked, and attenuated vaccines are not being developed because of safety concerns. Interest in attenuated vaccines has centered on isolated cases of patients infected with HIV-1 containing a deleted nef gene. Nef is a multifunctional accessory protein that is necessary for full HIV-1 virulence. Unfortunately, some patients infected with the nef-deleted virus eventually lose their CD4+ T cells to levels indicating progression to AIDS. This renders the possibility of an attenuated HIV-1 based solely on a deleted nef remote. In this review we discuss the knowledge gained both from the study of these patients and from in vitro investigations of Nef function to assess the possibility of developing new anti-HIV-1 drugs based on Nef. Specifically, we consider CD4 downregulation, major histocompatibility complex I downregulation, Pak2 activation, and enhancement of virion infectivity. We also consider the recent proposal that simian immunodeficiency viruses are non-pathogenic in their hosts because they have Nefs that downregulate CD3, but HIV-1 is pathogenic because its Nef fails to downregulate CD3. The possibility of incorporating the CD3 downregulation function into HIV-1 Nef as a therapeutic option is also considered. Finally, we conclude that inhibiting the CD4 downregulation function is the most promising Nef-targeted approach for developing a new anti-viral as a contribution to combating AIDS.


Assuntos
HIV-1/fisiologia , Fatores de Virulência , Produtos do Gene nef do Vírus da Imunodeficiência Humana/fisiologia , Síndrome da Imunodeficiência Adquirida/virologia , Humanos
10.
J Virol ; 82(19): 9657-67, 2008 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-18653452

RESUMO

The human immunodeficiency virus type 1 (HIV-1) accessory protein Nef downregulates major histocompatibility complex class I (MHC-I) from the cell surface. It has been proposed that the direct interaction of the acidic cluster (AC) of Nef, (62)EEEE(65), with the furin binding region (fbr) of PACS-1 is crucial for this Nef function. Contrary to this proposal, evidence is presented here that the four glutamates in Nef do not functionally engage the PACS-1 fbr. (i) The binding of Nef to the PACS-1 fbr in vitro is much weaker than the binding of the canonical furin AC to the PACS-1 fbr. (ii) The mutation of two of the four glutamates in Nef's AC to alanines does not alter Nef's ability to downregulate MHC-I, and triply mutated Nefs exhibit 50% activity. (iii) The introduction of lysine into the AC has little effect on Nef function. (iv) The mutation of all four glutamates to alanine does debilitate Nef MHC-I downregulation, but this quadruple mutation also impairs the ability of Nef to regulate p21-activated protein kinase and enhance viral particle infectivity. (v) The replacement of the Nef AC with the bona fide AC from furin results in the loss of the expected regulatory properties of the furin AC. (vi) The insertion of the conformation-disrupting amino acid proline into the Nef AC does not disrupt MHC-I downregulation. Our results are consistent with an alternative model in which (62)EEEE(65) plays a stabilizing role in the formation of a ternary complex between Nef, the MHC-I cytoplasmic domain, and AP-1.


Assuntos
Regulação Viral da Expressão Gênica , Produtos do Gene nef/genética , HIV-1/metabolismo , Antígenos de Histocompatibilidade Classe I/química , Fator de Transcrição AP-1/química , Sequência de Aminoácidos , Produtos do Gene nef/metabolismo , Células HeLa , Humanos , Modelos Moleculares , Conformação Molecular , Dados de Sequência Molecular , Mutagênese Sítio-Dirigida , Mutação , Ligação Proteica , Estrutura Terciária de Proteína
11.
Int J Pharm ; 360(1-2): 231-5, 2008 Aug 06.
Artigo em Inglês | MEDLINE | ID: mdl-18550304

RESUMO

In this study, Pluronic F127 hydrogels were characterised as an injectable system for the controlled release of drugs with variable molecular weights (FITC-Dextran at 70 and 40 kDa). In addition, the polymer-solvent interaction parameter (chi) was successfully estimated. Pluronic hydrogels (10-25 wt.%) were redox cured and their swelling behaviour investigated in PBS (pH 7.45) at 37 degrees C. After swelling to equilibrium, the hydrogels were compressed and the rubber-elasticity theory was applied to evaluate chi. Tensile tests proved the hydrogels were elastic and their chi values ranged between 0.50 and 0.53. The full drug load could be delivered over a period of approximately 15 h suggesting that redox cured Pluronic F127 hydrogels can function as injectable systems for controlled and sustained release of macromolecules.


Assuntos
Acrilatos/química , Sistemas de Liberação de Medicamentos , Poloxâmero/química , Algoritmos , Química Farmacêutica , Dextranos , Difusão , Ésteres/química , Fluoresceína-5-Isotiocianato/análogos & derivados , Hidrogéis , Polímeros , Solventes , Termodinâmica
12.
Virology ; 368(1): 7-16, 2007 Nov 10.
Artigo em Inglês | MEDLINE | ID: mdl-17632197

RESUMO

HIV-1 Nef is a multifunctional protein that exerts its activities through interactions with multiple cellular partners. Nef uses different domains and mechanisms to exert its functions including cell surface down-modulation of CD4 and MHC-I receptors and activation of the serine/threonine kinase PAK-2. We inserted tags at the C-terminus and proximal to the N-terminus of Nef and the effects on Nef's structure/function relationships were examined. We discovered significant defects in MHC-I down-modulation with the insertion of HA/FLAG tags at either region. We also found impaired PAK-2 activation with a C-terminal fusion with GFP. Interestingly, Nef-GFP and Nef-GH(7) induced MHC-I down-modulation, suggesting that the negative charge of the HA/FLAG tag could contribute to the observed defect. Together, these observations highlight elements of Nef's functional complexity and demonstrate previously unsuspected structural requirements for PAK-2 activation and MHC-1 down-modulation in Nef's flexible N- and C-terminal regions.


Assuntos
Antígenos CD4/metabolismo , HIV-1/fisiologia , Antígenos de Histocompatibilidade Classe I/metabolismo , Proteína Quinase C/metabolismo , Produtos do Gene nef do Vírus da Imunodeficiência Humana/fisiologia , Linhagem Celular , Regulação para Baixo , HIV-1/genética , Humanos , Estrutura Secundária de Proteína , Produtos do Gene nef do Vírus da Imunodeficiência Humana/química , Produtos do Gene nef do Vírus da Imunodeficiência Humana/genética
14.
Cell ; 125(6): 1034-5, 2006 Jun 16.
Artigo em Inglês | MEDLINE | ID: mdl-16777594

RESUMO

In this issue of Cell, Schindler et al. (2006) show that the Nef protein from nonpathogenic strains of simian immunodeficiency virus (SIV) induces the downregulation of host T cell receptor/CD3, whereas Nef from human immunodeficiency virus (HIV-1) does not. This loss of function in the Nef of HIV-1 may partly explain the pathogenicity of this virus.


Assuntos
Produtos do Gene nef/imunologia , HIV-1/fisiologia , HIV-2/imunologia , Lentivirus de Primatas/fisiologia , Linfócitos T/imunologia , Linfócitos T/virologia , Animais , Regulação para Baixo , HIV-1/imunologia , HIV-1/patogenicidade , Humanos , Lentivirus de Primatas/imunologia , Ativação Linfocitária , Complexo Receptor-CD3 de Antígeno de Linfócitos T/imunologia , Vírus da Imunodeficiência Símia/imunologia , Vírus da Imunodeficiência Símia/fisiologia , Produtos do Gene nef do Vírus da Imunodeficiência Humana
15.
J Virol ; 80(3): 1311-20, 2006 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-16415008

RESUMO

The human immunodeficiency virus type 1 (HIV-1) early gene product Nef is a multifunctional protein that alters numerous pathways of T-cell function, including endocytosis, signal transduction, vesicular trafficking, and immune modulation, and is a major determinant of pathogenesis. Individual Nef functions include PAK-2 activation, CD4 downregulation, major histocompatibility complex (MHC) class I downregulation, and enhancement of viral particle infectivity. How Nef accomplishes its multiple tasks presents a difficult problem of mechanistic analysis because of the complications associated with multiple, overlapping functional domains in the context of significant sequence variability. To address these issues we determined the conservation of each Nef residue based on 1,643 subtype B Nef sequences. Mutational analysis based on conservative substitutions and Nef sequence data allowed us to search for amino acids on the surface of Nef that are specifically required for PAK-2 activation. We found residues 85, 89, and 191 to be highly significant determinants for Nef's PAK-2 activation function but functionally unlinked to CD4 and MHC class I downregulation or enhancement of infectivity. These residues are not conserved across HIV-1 subtypes but are confined to separate sets of surface elements within a subtype. Thus, L85/H89/F191 and F85/F89/R191 are dominant in subtype B and subtype E or C, respectively. Our results provide support for developing subtype-specific interventions in HIV-1 disease.


Assuntos
Evolução Molecular , Produtos do Gene nef/genética , HIV-1/genética , Sequência de Aminoácidos , Substituição de Aminoácidos , Linhagem Celular , Sequência Conservada , Bases de Dados de Proteínas , Ativação Enzimática , Produtos do Gene nef/química , Produtos do Gene nef/fisiologia , HIV-1/classificação , HIV-1/patogenicidade , HIV-1/fisiologia , Células HeLa , Humanos , Técnicas In Vitro , Mutagênese Sítio-Dirigida , Proteínas Serina-Treonina Quinases/metabolismo , Estrutura Terciária de Proteína , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Virulência/genética , Virulência/fisiologia , Produtos do Gene nef do Vírus da Imunodeficiência Humana , Quinases Ativadas por p21
16.
J Virol ; 79(23): 14976-80, 2005 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-16282498

RESUMO

The accessory human immunodeficiency virus type 1 (HIV-1) protein Nef activates the autophosphorylation activity of p21-activated kinase 2 (PAK2). Merlin, a cellular substrate of PAK2, is homologous to the ezrin-radixin-moesin family and plays a critical role in Rac signaling. To assess the possible impact on host cell metabolism of Nef-induced PAK2 activation, we investigated the phosphorylation of merlin in Nef expressing cells. Here we report that Nef induces merlin phosphorylation in multiple cell lines independently of protein kinase A. This intracellular phosphorylation of merlin directly correlates with in vitro assay of the autophosphorylation activity of Nef-activated PAK2. Importantly, merlin phosphorylation induced by Nef was also observed in human primary T cells. The finding that Nef induces phosphorylation of the key signaling molecule merlin suggests several possible roles for PAK2 activation in HIV pathogenesis.


Assuntos
Produtos do Gene nef/fisiologia , HIV-1/metabolismo , Neurofibromina 2/metabolismo , Proteínas Serina-Treonina Quinases/metabolismo , Linhagem Celular , Ativação Enzimática , Produtos do Gene nef/genética , Humanos , Fosforilação , Produtos do Gene nef do Vírus da Imunodeficiência Humana , Quinases Ativadas por p21
17.
J Virol ; 79(20): 12732-41, 2005 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-16188976

RESUMO

Human immunodeficiency virus type 1 (HIV-1) Nef activation of p21-activated kinase 2 (PAK-2) was recapitulated in a cell-free system consisting of in vitro-transcribed RNA, rabbit reticulocyte lysate, and microsomal membranes on the basis of the following observations: (i) Nef associated with a kinase endogenous to the rabbit reticulocyte lysate that was identified as PAK-2, (ii) Nef-associated kinase activity was detected with Nefs from HIV-1(SF2), HIV-1(YU2), and SIV(mac239), (iii) kinase activation was not detected with a myristoylation-defective Nef (HIV-1(SF2)NefG2A) or with a Nef defective in PAK-2 activation but fully competent in other Nef functions (HIV-1(SF2)NefF195I), and (iv) Nef-associated kinase activation required activated endogenous p21 GTPases (Rac1 or Cdc42). The cell-free system was used to analyze the mechanism of Nef activation of PAK-2. First, studies suggest that the p21 GTPases may act transiently to enhance Nef activation of PAK-2 in vitro. Second, addition of wortmannin to the cell-free system demonstrated that Nef activation of PAK-2 does not require PI 3-kinase activity. Third, ultracentrifugation analysis revealed that whereas the majority of Nef and PAK-2 partitioned to the supernatant, Nef-associated PAK-2 activity partitioned to the membrane-containing pellet as a low-abundance complex. Lastly, Nef activation of PAK-2 in vitro requires addition of microsomal membranes either during or after translation of the Nef RNA. These results are consistent with a model in which activation of PAK-2 by Nef occurs by recruiting PAK-2 to membranes. As demonstrated herein, the cell-free system is a new and important tool in the investigation of the mechanism of PAK-2 activation by Nef.


Assuntos
Produtos do Gene nef/metabolismo , Infecções por HIV/virologia , HIV-1/enzimologia , Proteínas Serina-Treonina Quinases/genética , Membrana Celular/metabolismo , Sistema Livre de Células , Ativação Enzimática , HIV-1/patogenicidade , Humanos , Proteínas Serina-Treonina Quinases/metabolismo , Virulência , Produtos do Gene nef do Vírus da Imunodeficiência Humana , Quinases Ativadas por p21 , Proteínas rac1 de Ligação ao GTP/metabolismo
18.
J Virol ; 79(9): 5705-12, 2005 May.
Artigo em Inglês | MEDLINE | ID: mdl-15827185

RESUMO

We previously reported that inhibition of endosomal/lysosomal function can dramatically enhance human immunodeficiency virus type 1 (HIV-1) infectivity, suggesting that under these conditions productive HIV-1 infection can occur via the endocytic pathway. Here we further examined this effect with bafilomycin A1 (BFLA-1) and show that this enhancement of infectivity extends to all HIV-1 isolates tested regardless of coreceptor usage. However, isolate-specific differences were observed in the magnitude of the effect. This was particularly evident in the case of the weakly infectious HIV-1(SF2), for which we observed the greatest enhancement. Using reciprocal chimeric viruses, we were able to determine that both the disproportionate increase in the infectivity of HIV-1(SF2) in response to BFLA-1 and its weak infectivity in the absence of BFLA-1 mapped to its envelope gene. Further, we found HIV-1(SF2) to have lower fusion activity and to be 12-fold more sensitive to the fusion inhibitor T-20 than HIV-1(NL4-3). Proteasomal inhibitors also enhance HIV-1 infectivity, and we report that the combination of a lysosomal and a proteasomal inhibitor greatly enhanced infectivity of all isolates tested. Again, HIV-1(SF2) was unique in exhibiting a synergistic 400-fold increase in infectivity. We also determined that inhibition of proteasomal function increased the infectivity of HIV-1 pseudotyped with vesicular stomatitis virus G protein. The evidence presented here highlights the important role of the lysosomes/proteasomes in the destruction of infectious HIV-1(SF2) and could have implications for the development of novel antiviral agents that might take advantage of these innate defenses.


Assuntos
Infecções por HIV/virologia , HIV-1/fisiologia , Lisossomos/metabolismo , Inibidores de Proteassoma , Linhagem Celular , Inibidores Enzimáticos/farmacologia , Infecções por HIV/genética , Humanos , Lisossomos/efeitos dos fármacos , Macrolídeos/farmacologia , Complexo de Endopeptidases do Proteassoma/metabolismo , Recombinação Genética , Especificidade da Espécie , Replicação Viral
19.
Curr HIV Res ; 1(1): 41-50, 2003 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-15043211

RESUMO

The nef gene is present in all primate lentiviruses (HIV-1, HIV-2, and SIVs). In vivo, Nef has been shown to be a major determinant of virus pathogenicity. In vitro, many different Nef activities have been reported, including CD4 and MHC I downregulation, enhanced virion infectivity, and T-cell activation. These four different activities have been extensively investigated and appear to increase the pathogenicity of the virus. However, the contribution that these activities (individually or together) make to the in vivo phenotype has not been elucidated. The mechanism(s) by which Nef modulates distinct host cell properties has provided great insights into the intricate interaction between virus and host. In this manuscript, we review the different model systems that have been used to study Nef function in vivo and the information that they have provided regarding the best characterized in vitro Nef activities. The knowledge that has been accumulated has provided clues to our understanding of Nef function but they have also left us with many unanswered questions that should be the focus of future in vivo analysis of Nef function.


Assuntos
Produtos do Gene nef/fisiologia , HIV-1/fisiologia , HIV-2/fisiologia , Vírus da Imunodeficiência Símia/fisiologia , Animais , Modelos Animais de Doenças , Produtos do Gene nef/imunologia , Produtos do Gene nef/metabolismo , HIV-1/genética , HIV-1/crescimento & desenvolvimento , HIV-1/imunologia , HIV-2/crescimento & desenvolvimento , HIV-2/imunologia , Humanos , Macaca mulatta , Camundongos , Camundongos SCID , Camundongos Transgênicos , Vírus da Imunodeficiência Símia/crescimento & desenvolvimento , Vírus da Imunodeficiência Símia/imunologia , Produtos do Gene nef do Vírus da Imunodeficiência Humana
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA