Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 101
Filtrar
1.
Gene ; 792: 145735, 2021 Aug 05.
Artigo em Inglês | MEDLINE | ID: mdl-34048875

RESUMO

Human immunodeficiency virus (HIV) infection causes acquired immunodeficiency syndrome (AIDS), one of the most devastating diseases affecting humankind. Here, we have proposed a framework to examine the differences among microarray gene expression data of uninfected and three different HIV-1 infection stages using module preservation statistics. We leverage the advantage of gene co-expression networks (GCN) constructed for each infection stages to detect the topological and structural changes of a group of differentially expressed genes. We examine the relationship among a set of co-expression modules by constructing a module eigengene network considering the overall similarity/dissimilarity among the genes within the modules. We have utilized different module preservation statistics with two composite statistics: "Zsummary" and "MedianRank" to examine the changes in co-expression patterns between modules. We have found several interesting results on the preservation characteristics of gene modules across different stages. Some genes are identified to be preserved in a pair of stages while altering their characteristics across other stages. We further validated the obtained results using permutation test and classification techniques. The biological significances of the obtained modules have also been examined using gene ontology and pathway-based analysis. Additionally, we have identified a set of key immune regulatory hub genes in the associated protein-protein interaction networks (PPINs) of the differentially expressed (DE) genes, which interacts with HIV-1 proteins and are likely to act as potential biomarkers in HIV-1 progression.


Assuntos
Antígenos CD/genética , Quimiocinas/genética , Infecções por HIV/genética , HIV-1/patogenicidade , Interações Hospedeiro-Patógeno/genética , Proteínas do Vírus da Imunodeficiência Humana/genética , Doença Aguda , Antígenos CD/classificação , Antígenos CD/imunologia , Quimiocinas/classificação , Quimiocinas/imunologia , Doença Crônica , Conjuntos de Dados como Assunto , Progressão da Doença , Perfilação da Expressão Gênica , Regulação da Expressão Gênica , Ontologia Genética , Redes Reguladoras de Genes , Infecções por HIV/imunologia , Infecções por HIV/patologia , Infecções por HIV/virologia , HIV-1/crescimento & desenvolvimento , Interações Hospedeiro-Patógeno/imunologia , Proteínas do Vírus da Imunodeficiência Humana/classificação , Proteínas do Vírus da Imunodeficiência Humana/imunologia , Humanos , Análise em Microsséries , Anotação de Sequência Molecular , Ligação Proteica , Transdução de Sinais
2.
Viruses ; 13(3)2021 03 05.
Artigo em Inglês | MEDLINE | ID: mdl-33807824

RESUMO

The Human Immunodeficiency Virus type 1 (HIV-1) virion contains a conical shell, termed capsid, encasing the viral RNA genome. After cellular entry of the virion, the capsid is released and ensures the protection and delivery of the HIV-1 genome to the host nucleus for integration. The capsid relies on many virus-host factor interactions which are regulated spatiotemporally throughout the course of infection. In this paper, we will review the current understanding of the highly dynamic HIV-1 capsid-host interplay during the early stages of viral replication, namely intracellular capsid trafficking after viral fusion, nuclear import, uncoating, and integration of the viral genome into host chromatin. Conventional anti-retroviral therapies primarily target HIV-1 enzymes. Insights of capsid structure have resulted in a first-in-class, long-acting capsid-targeting inhibitor, GS-6207 (Lenacapavir). This inhibitor binds at the interface between capsid protein subunits, a site known to bind host factors, interferes with capsid nuclear import, HIV particle assembly, and ordered assembly. Our review will highlight capsid structure, the host factors that interact with capsid, and high-throughput screening techniques, specifically genomic and proteomic approaches, that have been and can be used to identify host factors that interact with capsid. Better structural and mechanistic insights into the capsid-host factor interactions will significantly inform the understanding of HIV-1 pathogenesis and the development of capsid-centric antiretroviral therapeutics.


Assuntos
Proteínas do Capsídeo/imunologia , Infecções por HIV/virologia , HIV-1/fisiologia , Interações entre Hospedeiro e Microrganismos/imunologia , Proteínas do Vírus da Imunodeficiência Humana/imunologia , Vírion/imunologia , Humanos , Desenvelopamento do Vírus
3.
Protein Pept Lett ; 28(6): 687-698, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33390106

RESUMO

BACKGROUND: The combination antiretroviral therapy (cART) could increase the number of circulating naive CD4 T lymphocytes, but was not able to eradicate human immunodeficiency virus-1 (HIV-1) infection. OBJECTIVE: Thus, induction of strong immune responses is important for control of HIV-1 infection. Furthermore, a simple and perfect serological method is required to detect virus in untreated-, treated- and drug resistant- HIV-1 infected individuals. METHODS: This study was conducted to assess and compare immunogenic properties of Nef, Vif, Vpr and Vpu accessory proteins as an antigen candidate in mice and their diagnostic importance in human as a biomarker. RESULTS: Our data showed that in mice, all heterologous prime/ boost regimens were more potent than homologous prime/ boost regimens in eliciting Th1 response and Granzyme B secretion as CTL activity. Moreover, the Nef, Vpu and Vif proteins could significantly increase Th1 immune response. In contrast, the Vpr protein could considerably induce Th2 immune response. On the other hand, among four accessory proteins, HIV-1 Vpu could significantly detect treated group from untreated group as a possible biomarker in human. CONCLUSION: Generally, among accessory proteins, Nef, Vpu and Vif antigens were potentially more suitable vaccine antigen candidates than Vpr antigen. Human antibodies against all these proteins were higher in HIV-1 different groups than healthy group. Among them, Vpu was known as a potent antigen in diagnosis of treated from untreated individuals. The potency of accessory proteins as an antigen candidate in an animal model and a human cohort study are underway.


Assuntos
Antígenos HIV/imunologia , Infecções por HIV , HIV-1 , Proteínas do Vírus da Imunodeficiência Humana/imunologia , Vacinas contra a AIDS/química , Vacinas contra a AIDS/imunologia , Animais , Biomarcadores/sangue , Anticorpos Anti-HIV/imunologia , Infecções por HIV/diagnóstico , Infecções por HIV/imunologia , Infecções por HIV/virologia , HIV-1/química , HIV-1/imunologia , Humanos , Camundongos
4.
Proc Natl Acad Sci U S A ; 117(45): 28232-28238, 2020 11 10.
Artigo em Inglês | MEDLINE | ID: mdl-33097667

RESUMO

Human leukocyte antigen (HLA) class I allotypes vary in their ability to present peptides in the absence of tapasin, an essential component of the peptide loading complex. We quantified tapasin dependence of all allotypes that are common in European and African Americans (n = 97), which revealed a broad continuum of values. Ex vivo examination of cytotoxic T cell responses to the entire HIV-1 proteome from infected subjects indicates that tapasin-dependent allotypes present a more limited set of distinct peptides than do tapasin-independent allotypes, data supported by computational predictions. This suggests that variation in tapasin dependence may impact the strength of the immune responses by altering peptide repertoire size. In support of this model, we observed that individuals carrying HLA class I genotypes characterized by greater tapasin independence progress more slowly to AIDS and maintain lower viral loads, presumably due to increased breadth of peptide presentation. Thus, tapasin dependence level, like HLA zygosity, may serve as a means to restrict or expand breadth of the HLA-I peptide repertoire across humans, ultimately influencing immune responses to pathogens and vaccines.


Assuntos
Apresentação de Antígeno/genética , Infecções por HIV , Antígenos de Histocompatibilidade Classe I , Proteínas de Membrana Transportadoras , Infecções por HIV/genética , Infecções por HIV/imunologia , HIV-1/imunologia , Antígenos de Histocompatibilidade Classe I/genética , Antígenos de Histocompatibilidade Classe I/imunologia , Antígenos de Histocompatibilidade Classe I/metabolismo , Proteínas do Vírus da Imunodeficiência Humana/imunologia , Humanos , Proteínas de Membrana Transportadoras/genética , Proteínas de Membrana Transportadoras/imunologia , Proteínas de Membrana Transportadoras/metabolismo , Peptídeos/imunologia , Peptídeos/metabolismo , Linfócitos T Citotóxicos/imunologia , Carga Viral/genética , Carga Viral/imunologia
5.
Biotechnol Lett ; 42(10): 1847-1863, 2020 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-32449070

RESUMO

OBJECTIVES: Enhancement of the potential ability of biomacromolecules to cross cell membranes is a critical step for development of effective therapeutic vaccine especially DNA vaccine against human immunodeficiency virus-1 (HIV-1) infection. The supercharged proteins were known as powerful weapons for delivery of different types of cargoes such as DNA and protein. Hence, we applied B1 protein with + 43 net charges obtained from a single frameshift in the gene encoding enhanced green fluorescent protein (eGFP) for delivery of two multi-epitope DNA constructs (nef-vpu-gp160-p24 and nef-vif-gp160-p24) in vitro and in vivo for the first time. For this purpose, B1 protein was generated in bacterial expression system under native conditions, and used to interact with both DNA constructs. RESULTS: Our data indicated that B1 protein (~ 27 kDa) was able to form a stable nanoparticle (~ 80-110 nm) with both DNA constructs at nitrogen: phosphate (N: P) ratio of 1:1. Moreover, the transfection efficiency of B1 protein for DNA delivery into HEK-293T cell line indicated that the cellular uptake of nef-vif-gp160-p24 DNA/ B1 and nef-vpu-gp160-p24 DNA/ B1 nanoparticles was about 32-35% with lower intensity as compared to TurboFect commercial reagent. On the other hand, immunization of BALB/c mice with different modalities demonstrated that B1 protein could enhance the levels of antibody, IFN-gamma and Granzyme B eliciting potent and strong Th1-directed cellular immunity. CONCLUSION: Generally, our findings showed the potency of B1 protein as a promising gene delivery system to improve an effective therapeutic vaccine against HIV-1 infection.


Assuntos
Vacinas contra a AIDS , Peptídeos Penetradores de Células , Técnicas de Transferência de Genes , Proteínas do Vírus da Imunodeficiência Humana , Vacinas de DNA , Animais , Peptídeos Penetradores de Células/genética , Peptídeos Penetradores de Células/metabolismo , Clonagem Molecular , Feminino , Proteínas de Fluorescência Verde/genética , Proteínas de Fluorescência Verde/metabolismo , Células HEK293 , HIV-1 , Proteínas do Vírus da Imunodeficiência Humana/genética , Proteínas do Vírus da Imunodeficiência Humana/imunologia , Humanos , Camundongos , Camundongos Endogâmicos BALB C , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/imunologia , Proteínas Recombinantes de Fusão/metabolismo
6.
PLoS Med ; 17(2): e1003038, 2020 02.
Artigo em Inglês | MEDLINE | ID: mdl-32092060

RESUMO

BACKGROUND: HVTN 100 evaluated the safety and immunogenicity of an HIV subtype C pox-protein vaccine regimen, investigating a 12-month booster to extend vaccine-induced immune responses. METHODS AND FINDINGS: A phase 1-2 randomized double-blind placebo-controlled trial enrolled 252 participants (210 vaccine/42 placebo; median age 23 years; 43% female) between 9 February 2015 and 26 May 2015. Vaccine recipients received ALVAC-HIV (vCP2438) alone at months 0 and 1 and with bivalent subtype C gp120/MF59 at months 3, 6, and 12. Antibody (IgG, IgG3 binding, and neutralizing) and CD4+ T-cell (expressing interferon-gamma, interleukin-2, and CD40 ligand) responses were evaluated at month 6.5 for all participants and at months 12, 12.5, and 18 for a randomly selected subset. The primary analysis compared IgG binding antibody (bAb) responses and CD4+ T-cell responses to 3 vaccine-matched antigens at peak (month 6.5 versus 12.5) and durability (month 12 versus 18) timepoints; IgG responses to CaseA2_gp70_V1V2.B, a primary correlate of risk in RV144, were also compared at these same timepoints. Secondary and exploratory analyses compared IgG3 bAb responses, IgG bAb breadth scores, neutralizing antibody (nAb) responses, antibody-dependent cellular phagocytosis, CD4+ polyfunctionality responses, and CD4+ memory sub-population responses at the same timepoints. Vaccines were generally safe and well tolerated. During the study, there were 2 deaths (both in the vaccine group and both unrelated to study products). Ten participants became HIV-infected during the trial, 7% (3/42) of placebo recipients and 3% (7/210) of vaccine recipients. All 8 serious adverse events were unrelated to study products. Less waning of immune responses was seen after the fifth vaccination than after the fourth, with higher antibody and cellular response rates at month 18 than at month 12: IgG bAb response rates to 1086.C V1V2, 21.0% versus 9.7% (difference = 11.3%, 95% CI = 0.6%-22.0%, P = 0.039), and ZM96.C V1V2, 21.0% versus 6.5% (difference = 14.5%, 95% CI = 4.1%-24.9%, P = 0.004). IgG bAb response rates to all 4 primary V1V2 antigens were higher 2 weeks after the fifth vaccination than 2 weeks after the fourth vaccination: 87.7% versus 75.4% (difference = 12.3%, 95% CI = 1.7%-22.9%, P = 0.022) for 1086.C V1V2, 86.0% versus 63.2% (difference = 22.8%, 95% CI = 9.1%-36.5%, P = 0.001) for TV1c8.2.C V1V2, 67.7% versus 44.6% (difference = 23.1%, 95% CI = 10.4%-35.7%, P < 0.001) for ZM96.C V1V2, and 81.5% versus 60.0% (difference = 21.5%, 95% CI = 7.6%-35.5%, P = 0.002) for CaseA2_gp70_V1V2.B. IgG bAb response rates to the 3 primary vaccine-matched gp120 antigens were all above 90% at both peak timepoints, with no significant differences seen, except a higher response rate to ZM96.C gp120 at month 18 versus month 12: 64.5% versus 1.6% (difference = 62.9%, 95% CI = 49.3%-76.5%, P < 0.001). CD4+ T-cell response rates were higher at month 18 than month 12 for all 3 primary vaccine-matched antigens: 47.3% versus 29.1% (difference = 18.2%, 95% CI = 2.9%-33.4%, P = 0.021) for 1086.C, 61.8% versus 38.2% (difference = 23.6%, 95% CI = 9.5%-37.8%, P = 0.001) for TV1.C, and 63.6% versus 41.8% (difference = 21.8%, 95% CI = 5.1%-38.5%, P = 0.007) for ZM96.C, with no significant differences seen at the peak timepoints. Limitations were that higher doses of gp120 were not evaluated, this study was not designed to investigate HIV prevention efficacy, and the clinical significance of the observed immunological effects is uncertain. CONCLUSIONS: In this study, a 12-month booster of subtype C pox-protein vaccines restored immune responses, and slowed response decay compared to the 6-month vaccination. TRIAL REGISTRATION: ClinicalTrials.gov NCT02404311. South African National Clinical Trials Registry (SANCTR number: DOH--27-0215-4796).


Assuntos
Vacinas contra a AIDS/uso terapêutico , Anticorpos Neutralizantes/imunologia , Linfócitos T CD4-Positivos/imunologia , Infecções por HIV/prevenção & controle , Proteínas do Vírus da Imunodeficiência Humana/imunologia , Imunização Secundária , Imunoglobulina G/imunologia , Vacinas contra a AIDS/imunologia , Adulto , Artralgia/induzido quimicamente , Método Duplo-Cego , Feminino , Cefaleia/induzido quimicamente , Humanos , Imunogenicidade da Vacina , Reação no Local da Injeção , Injeções Intramusculares , Masculino , África do Sul , Adulto Jovem
7.
Mol Immunol ; 119: 106-122, 2020 03.
Artigo em Inglês | MEDLINE | ID: mdl-32007753

RESUMO

A licensed vaccine against human immunodeficiency virus-1 (HIV-1) infection has not become available up to now. Hence, it is more rational to use immune-informatics tools for prediction of T cell epitopes (in silico study) and development of an effective epitope-driven vaccine against hypervariable pathogens. Multiepitope vaccines were considered as the next generation of an effective vaccine against HIV-1 infection. In the current study, we developed two different constructs encoding T cell epitopes derived from Nef, Vif, Vpu, Gp160 and P24 proteins in BALB/c mice. To overcome their poor immunogenicity, four different cell penetrating peptides (MPG and HR9 for DNA delivery, and CyLoP-1 and LDP-NLS for protein delivery), Montanide adjuvant, and heterologous prime-boost immunization strategy were utilized. The generation of cytokines, Granzyme B, and total IgG and its subclasses was determined using ELISA. Our data indicated that the levels of IFN-γ and Granzyme B in mice injected with Nef-Vif-Gp160-P24 multiepitope constructs were higher than those immunized with Nef-Vpu-Gp160-P24 multiepitope constructs. Moreover, the heterologous DNA priming/ multiepitope peptide boosting in both Nef-Vif-Gp160-P24 and Nef-Vpu-Gp160-P24 regimens induced significantly high antigen-specific IgG2a and IgG2b responses in comparison with other groups. There was no significant difference between MPG and HR9 as well as CyLoP-1 and LDP-NLS as a delivery system for enhancement of immune responses. Generally, the heterologous DNA prime/ multiepitope peptide boost modalities for both constructs could significantly enhance the levels of IgG2a, IgG2b, IFN-γ, and Granzyme B directed toward Th1 immune responses as compared to homologous prime/ boost with DNA or polypeptide constructs.


Assuntos
Vacinas contra a AIDS/imunologia , Epitopos de Linfócito T/imunologia , HIV-1/imunologia , Proteínas do Vírus da Imunodeficiência Humana/imunologia , Animais , Citocinas/sangue , Epitopos de Linfócito B/imunologia , Feminino , Granzimas/metabolismo , Anticorpos Anti-HIV/sangue , Anticorpos Anti-HIV/imunologia , Imunogenicidade da Vacina , Camundongos Endogâmicos BALB C , Modelos Moleculares
8.
PLoS One ; 14(10): e0223844, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31671105

RESUMO

To develop an effective therapeutic vaccine against HIV-1, prediction of the most conserved epitopes derived from major proteins using bioinformatics tools is an alternative achievement. The epitope-driven vaccines against variable pathogens represented successful results. Hence, to overcome this hyper-variable virus, we designed the highly conserved and immunodominant peptide epitopes. Two servers were used to predict peptide-MHC-I binding affinity including NetMHCpan4.0 and Syfpeithi servers. The NetMHCIIpan3.2 server was utilized for MHC-II binding affinity. Then, we determined immunogenicity scores and allergenicity by the IEDB immunogenicity predictor and Algpred, respectively. Next, for estimation of toxicity and population coverage, ToxinPred server and IEDB population coverage tool were applied. After that, the MHC-peptide binding was investigated by GalexyPepDock peptide-protein flexible docking server. Finally, two different DNA and peptide constructs containing Nef-Vif-Gp160-P24 and Nef-Vpu-Gp160-P24 were prepared and complexed with four various cell penetrating peptides (CPPs) for delivery into mammalian cells (MPG and HR9 CPPs for DNA delivery, and CyLoP-1 and LDP-NLS CPPs for protein delivery). Our results indicated that the designed DNA and peptide constructs could form non-covalent stable nanoparticles at certain ratios as observed by scanning electron microscope (SEM) and Zetasizer. The flow cytometry results obtained from in vitro transfection of the nanoparticles into HEK-293T cell lines showed that the percentage of GFP expressing cells was about 38.38 ± 1.34%, 25.36% ± 0.30, 54.95% ± 0.84, and 25.11% ± 0.36 for MPG/pEGFP-nef-vif-gp160-p24, MPG/pEGFP-nef-vpu-gp160-p24, HR9/pEGFP-nef-vif-gp160-p24 and HR9/pEGFP-nef-vpu-gp160-p24, respectively. Thus, these data showed that the DNA construct harboring nef-vif-gp160-p24 multi-epitope gene had higher efficiency than the DNA construct harboring nef-vpu-gp160-p24 multi-epitope gene to penetrate into the cells. Moreover, delivery of the recombinant Nef-Vif-Gp160-P24 and Nef-Vpu-Gp160-P24 polyepitope peptides in HEK-293T cells was confirmed as a single band about 32 kDa using western blot analysis. Although, both DNA and peptide constructs could be successfully transported by a variety of CPPs into the cells, but the difference between them in transfection rate will influence the levels of immune responses for development of therapeutic vaccines.


Assuntos
Peptídeos Penetradores de Células/metabolismo , Epitopos de Linfócito T/metabolismo , HIV-1/imunologia , Proteínas do Vírus da Imunodeficiência Humana/imunologia , Proteínas Virais Reguladoras e Acessórias/metabolismo , Produtos do Gene vif do Vírus da Imunodeficiência Humana/metabolismo , Sequência de Aminoácidos , Epitopos de Linfócito T/química , Epitopos de Linfócito T/imunologia , Células HEK293 , Proteína do Núcleo p24 do HIV/imunologia , Proteína gp160 do Envelope de HIV/imunologia , Proteínas do Vírus da Imunodeficiência Humana/metabolismo , Humanos , Produtos do Gene nef do Vírus da Imunodeficiência Humana/imunologia
9.
Curr Drug Deliv ; 16(9): 818-828, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31549593

RESUMO

OBJECTIVE: Novel vaccination approaches are required to control human immunodeficiency virus (HIV) infections. The membrane proximal external region (MPER) of Env gp41 subunit and the V3/glycans of Env gp120 subunit were known as potential antigenic targets for anti-HIV-1 vaccines. In this study, we prepared the modified dendritic cells (DCs) and mesenchymal stem cells (MSCs) with HIV-1 MPER-V3 gene using mechanical and chemical approaches. METHODS: At first, MPER-V3 fusion DNA delivery was optimized in dendritic cells (DCs) and mesenchymal stem cells (MSCs) using three mechanical (i.e., uniaxial cyclic stretch, equiaxial cyclic stretch and shear stress bioreactors), and two chemical (i.e., TurboFect or Lipofectamine) methods. Next, the modified DCs and MSCs with MPER-V3 antigen were compared to induce immune responses in vivo. RESULTS: Our data showed that the combination of equiaxial cyclic stretch loading and lipofectamine twice with 48 h intervals increased the efficiency of transfection about 60.21 ± 1.05 % and 65.06 ± 0.09 % for MSCs and DCs, respectively. Moreover, DCs and MSCs transfected with MPER-V3 DNA in heterologous DC or MSC prime/ peptide boost immunizations induced high levels of IgG2a, IgG2b, IFN-γ and IL-10 directed toward Th1 responses as well as an increased level of Granzyme B. Indeed, the modified MSCs and DCs with MPER-V3 DNA could significantly enhance the MPER/V3-specific T-cell responses compared to MPER/V3 peptide immunization. CONCLUSIONS: These findings showed that the modified MSC-based immunization could elicit effective immune responses against HIV antigen similar to the modified DC-based immunization.


Assuntos
Vacinas contra a AIDS/administração & dosagem , Células Dendríticas , Técnicas de Transferência de Genes , Células-Tronco Mesenquimais , Animais , Anticorpos Antivirais/sangue , Citocinas/imunologia , DNA/administração & dosagem , Feminino , Granzimas/imunologia , Proteínas do Vírus da Imunodeficiência Humana/genética , Proteínas do Vírus da Imunodeficiência Humana/imunologia , Imunoglobulina G/sangue , Lipídeos/administração & dosagem , Masculino , Fenômenos Mecânicos , Camundongos Endogâmicos BALB C
10.
Cells ; 8(8)2019 08 17.
Artigo em Inglês | MEDLINE | ID: mdl-31426525

RESUMO

Restriction factors are antiviral components of intrinsic immunity which constitute a first line of defense by blocking different steps of the human immunodeficiency virus (HIV) replication cycle. In immune cells, HIV infection is also sensed by several pattern recognition receptors (PRRs), leading to type I interferon (IFN-I) and inflammatory cytokines production that upregulate antiviral interferon-stimulated genes (ISGs). Several studies suggest a link between these two types of immunity. Indeed, restriction factors, that are generally interferon-inducible, are able to modulate immune responses. This review highlights recent knowledge of the interplay between restriction factors and immunity inducing antiviral defenses. Counteraction of this intrinsic and innate immunity by HIV viral proteins will also be discussed.


Assuntos
Infecções por HIV/imunologia , HIV-1 , Interações entre Hospedeiro e Microrganismos/fisiologia , Fatores Reguladores de Interferon/imunologia , Interferon Tipo I/imunologia , Receptores de Reconhecimento de Padrão/imunologia , Animais , Linhagem Celular , HIV-1/imunologia , HIV-1/patogenicidade , Proteínas do Vírus da Imunodeficiência Humana/imunologia , Humanos , Evasão da Resposta Imune/imunologia , Camundongos , Replicação Viral/imunologia
12.
mBio ; 10(3)2019 06 18.
Artigo em Inglês | MEDLINE | ID: mdl-31213558

RESUMO

The HIV-1 accessory protein Vpu enhances viral release by counteracting the restriction factor BST-2. Furthermore, Vpu promotes NK cell evasion by downmodulating cell surface NTB-A and PVR, known ligands of the NK cell receptors NTB-A and DNAM-1, respectively. While it has been established that Vpu's transmembrane domain (TMD) is required for the interaction and intracellular sequestration of BST-2, NTB-A, and PVR, it remains unclear how Vpu manages to target these proteins simultaneously. In this study, we show that upon upregulation, BST-2 is preferentially downregulated by Vpu over its other TMD substrates. We found that type I interferon (IFN)-mediated BST-2 upregulation greatly impairs the ability of Vpu to downregulate NTB-A and PVR. Our results suggest that occupation of Vpu by BST-2 affects its ability to downregulate other TMD substrates. Accordingly, knockdown of BST-2 increases Vpu's potency to downmodulate NTB-A and PVR in the presence of type I IFN treatment. Moreover, we show that expression of human BST-2, but not that of the macaque orthologue, decreases Vpu's capacity to downregulate NTB-A. Importantly, we show that type I IFNs efficiently sensitize HIV-1-infected cells to NTB-A- and DNAM-1-mediated direct and antibody-dependent NK cell responses. Altogether, our results reveal that type I IFNs decrease Vpu's polyfunctionality, thus reducing its capacity to protect HIV-1-infected cells from NK cell responses.IMPORTANCE The restriction factor BST-2 and the NK cell ligands NTB-A and PVR are among a growing list of membrane proteins found to be downregulated by HIV-1 Vpu. BST-2 antagonism enhances viral release, while NTB-A and PVR downmodulation contributes to NK cell evasion. However, it remains unclear how Vpu can target multiple cellular factors simultaneously. Here we provide evidence that under physiological conditions, BST-2 is preferentially targeted by Vpu over NTB-A and PVR. Specifically, we show that type I IFNs decrease Vpu's polyfunctionality by upregulating BST-2, thus reducing its capacity to protect HIV-1-infected cells from NK cell responses. This indicates that there is a hierarchy of Vpu substrates upon IFN treatment, revealing that for the virus, targeting BST-2 as part of its resistance to IFN takes precedence over evading NK cell responses. This reveals a potential weakness in HIV-1's immunoevasion mechanisms that may be exploited therapeutically to harness NK cell responses against HIV-1.


Assuntos
Antígenos CD/genética , Linfócitos T CD4-Positivos/imunologia , Infecções por HIV/imunologia , Proteínas do Vírus da Imunodeficiência Humana/genética , Interferon Tipo I/farmacologia , Células Matadoras Naturais/imunologia , Proteínas Virais Reguladoras e Acessórias/genética , Linfócitos T CD4-Positivos/virologia , Regulação para Baixo , Proteínas Ligadas por GPI/genética , Células HEK293 , HIV-1 , Proteínas do Vírus da Imunodeficiência Humana/imunologia , Humanos , Evasão da Resposta Imune , Receptores Virais/genética , Receptores Virais/imunologia , Família de Moléculas de Sinalização da Ativação Linfocitária/genética , Família de Moléculas de Sinalização da Ativação Linfocitária/imunologia , Ativação Transcricional , Regulação para Cima , Proteínas Virais Reguladoras e Acessórias/imunologia
13.
PLoS One ; 14(3): e0212888, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-30889178

RESUMO

TRIM5α is an interferon inducible restriction factor which contributes to intrinsic defense against HIV infection by targeting the HIV capsid protein CA. Although human TRIM5α (huTRIM5α) does not potently inhibit HIV-1 infection, the ability of huTRIM5α to exhibit some control of HIV-1 infection is evidenced by a single nucleotide polymorphism in huTRIM5α which substitutes aspartic acid to glycine at position 249 (G249D) in the L2 region and is associated with higher susceptibility to HIV-1 infection. To understand the mechanistic basis for the reduced antiviral activity, we employed biophysical and cell biological methods coupled with molecular dynamics simulations to compare WT and the G249D polymorphism of huTRIM5α. We investigated the differences in conformational dynamics of rhesus and huTRIM5α Coiled Coil-Linker 2 (CC-L2) dimers utilizing circular dichroism and single molecule-Fluorescence Energy Transfer (sm-FRET). These methods revealed that the G249D dimer exhibits secondary structure and conformational dynamics similar to WT huTRIM5α. Homology modelling revealed that G249 was present on the hairpin of the antiparallel dimer, in a position which may act to stabilize the adjacent BBox2 domain which mediates the inter-dimeric contacts required for the formation of TRIM5 assemblies. We therefore asked if the G249D mutant forms assemblies in cells with the same efficiency as WT protein by expressing these proteins as YFP fusions and quantifying the number of assemblies in cells. In cells expressing comparable amounts of protein, the G249D mutant formed fewer assemblies than WT protein, in agreement with our homology modeling predictions and molecular dynamics simulations of dimers and higher oligomers of TRIM5α, providing a mechanistic explanation of the reduced antiviral activity of the G249D polymorphism.


Assuntos
Proteínas de Transporte/genética , Infecções por HIV/genética , HIV-1/imunologia , Animais , Fatores de Restrição Antivirais , Proteínas do Capsídeo/imunologia , Proteínas do Capsídeo/metabolismo , Proteínas de Transporte/imunologia , Proteínas de Transporte/metabolismo , Gatos , Predisposição Genética para Doença , Células HEK293 , Infecções por HIV/imunologia , Infecções por HIV/virologia , HIV-1/metabolismo , Proteínas do Vírus da Imunodeficiência Humana/imunologia , Proteínas do Vírus da Imunodeficiência Humana/metabolismo , Humanos , Simulação de Dinâmica Molecular , Polimorfismo de Nucleotídeo Único , Conformação Proteica em alfa-Hélice/genética , Domínios Proteicos/genética , Estrutura Quaternária de Proteína/genética , Proteínas Recombinantes/genética , Proteínas Recombinantes/imunologia , Proteínas Recombinantes/metabolismo , Proteínas com Motivo Tripartido , Ubiquitina-Proteína Ligases
14.
PLoS Pathog ; 14(9): e1007257, 2018 09.
Artigo em Inglês | MEDLINE | ID: mdl-30180214

RESUMO

HIV-1 can downregulate HLA-C on infected cells, using the viral protein Vpu, and the magnitude of this downregulation varies widely between primary HIV-1 variants. The selection pressures that result in viral downregulation of HLA-C in some individuals, but preservation of surface HLA-C in others are not clear. To better understand viral immune evasion targeting HLA-C, we have characterized HLA-C downregulation by a range of primary HIV-1 viruses. 128 replication competent viral isolates from 19 individuals with effective anti-retroviral therapy, show that a substantial minority of individuals harbor latent reservoir virus which strongly downregulates HLA-C. Untreated infections display no change in HLA-C downregulation during the first 6 months of infection, but variation between viral quasispecies can be detected in chronic infection. Vpu molecules cloned from plasma of 195 treatment naïve individuals in chronic infection demonstrate that downregulation of HLA-C adapts to host HLA genotype. HLA-C alleles differ in the pressure they exert for downregulation, and individuals with higher levels of HLA-C expression favor greater viral downregulation of HLA-C. Studies of primary and mutant molecules identify 5 residues in the transmembrane region of Vpu, and 4 residues in the transmembrane domain of HLA-C, which determine interactions between Vpu and HLA. The observed adaptation of Vpu-mediated downregulation to host genotype indicates that HLA-C alleles differ in likelihood of mediating a CTL response that is subverted by viral downregulation, and that preservation of HLA-C expression is favored in the absence of these responses. Finding that latent reservoir viruses can downregulate HLA-C could have implications for HIV-1 cure therapy approaches in some individuals.


Assuntos
Infecções por HIV/genética , Infecções por HIV/imunologia , HIV-1/patogenicidade , Antígenos HLA-C/genética , Sequência de Aminoácidos , Reservatórios de Doenças/virologia , Regulação para Baixo , Variação Genética , Genótipo , Infecções por HIV/virologia , HIV-1/imunologia , Interações Hospedeiro-Patógeno/genética , Interações Hospedeiro-Patógeno/imunologia , Proteínas do Vírus da Imunodeficiência Humana/química , Proteínas do Vírus da Imunodeficiência Humana/genética , Proteínas do Vírus da Imunodeficiência Humana/imunologia , Humanos , Evasão da Resposta Imune , Proteínas Virais Reguladoras e Acessórias/química , Proteínas Virais Reguladoras e Acessórias/genética , Proteínas Virais Reguladoras e Acessórias/imunologia
15.
J Clin Invest ; 128(10): 4387-4396, 2018 10 01.
Artigo em Inglês | MEDLINE | ID: mdl-30148455

RESUMO

Activation of HIV-1 reservoirs and induction of anti-HIV-1 T cells are critical to control HIV-1 rebound after combined antiretroviral therapy (cART). Here we evaluated in humanized mice (hu-mice) with persistent HIV-1 infection the therapeutic effect of TLR3 agonist and a CD40-targeting HIV-1 vaccine, which consists of a string of 5 highly conserved CD4+ and CD8+ T cell epitope-rich regions of HIV-1 Gag, Nef, and Pol fused to the C-terminus of a recombinant anti-human CD40 antibody (αCD40.HIV5pep). We show that αCD40.HIV5pep vaccination coadministered with poly(I:C) adjuvant induced HIV-1-specific human CD8+ and CD4+ T cell responses in hu-mice. Interestingly, poly(I:C) treatment also reactivated HIV-1 reservoirs. When administrated in therapeutic settings in HIV-1-infected hu-mice under effective cART, αCD40.HIV5pep with poly(I:C) vaccination induced HIV-1-specific CD8+ T cells and reduced the level of cell-associated HIV-1 DNA (or HIV-1 reservoirs) in lymphoid tissues. Most strikingly, the vaccination significantly delayed HIV-1 rebound after cART cessation. In summary, the αCD40.HIV5pep with poly(I:C) vaccination approach both activates replication of HIV-1 reservoirs and enhances the anti-HIV-1 T cell response, leading to a reduced level of cell-associated HIV-1 DNA or reservoirs. Our proof-of-concept study has significant implication for the development of CD40-targeting HIV-1 vaccine to enhance anti-HIV-1 immunity and reduce HIV-1 reservoirs in patients with suppressive cART.


Assuntos
Vacinas contra a AIDS , Antígenos CD40/imunologia , Epitopos de Linfócito T , HIV-1/imunologia , Proteínas do Vírus da Imunodeficiência Humana , Poli I-C/farmacologia , Receptor 3 Toll-Like/agonistas , Vacinas contra a AIDS/imunologia , Vacinas contra a AIDS/farmacologia , Animais , Epitopos de Linfócito T/imunologia , Epitopos de Linfócito T/farmacologia , Proteínas do Vírus da Imunodeficiência Humana/imunologia , Proteínas do Vírus da Imunodeficiência Humana/farmacologia , Humanos , Imunidade Celular/efeitos dos fármacos , Camundongos , Camundongos Knockout , Receptor 3 Toll-Like/imunologia
16.
Nat Commun ; 9(1): 1371, 2018 04 10.
Artigo em Inglês | MEDLINE | ID: mdl-29636452

RESUMO

HIV-1 causes chronic inflammation and AIDS in humans, whereas related simian immunodeficiency viruses (SIVs) replicate efficiently in their natural hosts without causing disease. It is currently unknown to what extent virus-specific properties are responsible for these different clinical outcomes. Here, we incorporate two putative HIV-1 virulence determinants, i.e., a Vpu protein that antagonizes tetherin and blocks NF-κB activation and a Nef protein that fails to suppress T cell activation via downmodulation of CD3, into a non-pathogenic SIVagm strain and test their impact on viral replication and pathogenicity in African green monkeys. Despite sustained high-level viremia over more than 4 years, moderately increased immune activation and transcriptional signatures of inflammation, the HIV-1-like SIVagm does not cause immunodeficiency or any other disease. These data indicate that species-specific host factors rather than intrinsic viral virulence factors determine the pathogenicity of primate lentiviruses.


Assuntos
HIV-1/patogenicidade , Especificidade de Hospedeiro , Proteínas do Vírus da Imunodeficiência Humana/imunologia , Lentivirus de Primatas/crescimento & desenvolvimento , Vírus da Imunodeficiência Símia/patogenicidade , Proteínas Virais Reguladoras e Acessórias/imunologia , Produtos do Gene nef do Vírus da Imunodeficiência Humana/imunologia , Sequência de Aminoácidos , Animais , Antígeno 2 do Estroma da Médula Óssea/genética , Antígeno 2 do Estroma da Médula Óssea/imunologia , Complexo CD3/genética , Complexo CD3/imunologia , Chlorocebus aethiops , Feminino , Regulação da Expressão Gênica , HIV-1/crescimento & desenvolvimento , Proteínas do Vírus da Imunodeficiência Humana/genética , Humanos , Lentivirus de Primatas/patogenicidade , Ativação Linfocitária , NF-kappa B/genética , NF-kappa B/imunologia , Alinhamento de Sequência , Transdução de Sinais , Vírus da Imunodeficiência Símia/crescimento & desenvolvimento , Transcrição Gênica , Carga Viral , Proteínas Virais Reguladoras e Acessórias/genética , Virulência , Replicação Viral , Produtos do Gene nef do Vírus da Imunodeficiência Humana/genética
17.
Front Immunol ; 9: 3032, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30687306

RESUMO

APOBEC3G (A3G) is a host enzyme that mutates the genomes of retroviruses like HIV. Since A3G is expressed pre-infection, it has classically been considered an agent of innate immunity. We and others previously showed that the impact of A3G-induced mutations on the HIV genome extends to adaptive immunity also, by generating cytotoxic T cell (CTL) escape mutations. Accordingly, HIV genomic sequences encoding CTL epitopes often contain A3G-mutable "hotspot" sequence motifs, presumably to channel A3G action toward CTL escape. Here, we studied the depths and consequences of this apparent viral genome co-evolution with A3G. We identified all potential CTL epitopes in Gag, Pol, Env, and Nef restricted to several HLA class I alleles. We simulated A3G-induced mutations within CTL epitope-encoding sequences, and flanking regions. From the immune recognition perspective, we analyzed how A3G-driven mutations are predicted to impact CTL-epitope generation through modulating proteasomal processing and HLA class I binding. We found that A3G mutations were most often predicted to result in diminishing/abolishing HLA-binding affinity of peptide epitopes. From the viral genome evolution perspective, we evaluated enrichment of A3G hotspots at sequences encoding CTL epitopes and included control sequences in which the HIV genome was randomly shuffled. We found that sequences encoding immunogenic epitopes exhibited a selective enrichment of A3G hotspots, which were strongly biased to translate to non-synonymous amino acid substitutions. When superimposed on the known mutational gradient across the entire length of the HIV genome, we observed a gradient of A3G hotspot enrichment, and an HLA-specific pattern of the potential of A3G hotspots to lead to CTL escape mutations. These data illuminate the depths and extent of the co-evolution of the viral genome to subvert the host mutator A3G.


Assuntos
Desaminase APOBEC-3G/metabolismo , Imunidade Adaptativa/genética , Infecções por HIV/imunologia , HIV-1/genética , Interações Hospedeiro-Patógeno/genética , Evasão da Resposta Imune/genética , Coevolução Biológica/genética , Coevolução Biológica/imunologia , Epitopos de Linfócito T/genética , Epitopos de Linfócito T/imunologia , Genoma Viral/genética , Genoma Viral/imunologia , Infecções por HIV/virologia , HIV-1/imunologia , Antígenos de Histocompatibilidade Classe I/imunologia , Antígenos de Histocompatibilidade Classe I/metabolismo , Interações Hospedeiro-Patógeno/imunologia , Proteínas do Vírus da Imunodeficiência Humana/genética , Proteínas do Vírus da Imunodeficiência Humana/imunologia , Humanos , Mutação , Linfócitos T Citotóxicos/imunologia , Linfócitos T Citotóxicos/metabolismo
18.
Int J Mol Sci ; 18(10)2017 Oct 03.
Artigo em Inglês | MEDLINE | ID: mdl-28972547

RESUMO

Human immunodeficiency virus (HIV) hides from the immune system in part by mimicking host antigens, including human leukocyte antigens. It is demonstrated here that HIV also mimics the V-ß-D-J-ß of approximately seventy percent of about 600 randomly selected human T cell receptors (TCR). This degree of mimicry is greater than any other human pathogen, commensal or symbiotic organism studied. These data suggest that HIV may be evolving into a commensal organism just as simian immunodeficiency virus has done in some types of monkeys. The gp120 envelope protein, Nef protein and Pol protein are particularly similar to host TCR, camouflaging HIV from the immune system and creating serious barriers to the development of safe HIV vaccines. One consequence of HIV mimicry of host TCR is that antibodies against HIV proteins have a significant probability of recognizing the corresponding TCR as antigenic targets, explaining the widespread observation of lymphocytotoxic autoantibodies in acquired immunodeficiency syndrome (AIDS). Quantitative enzyme-linked immunoadsorption assays (ELISA) demonstrated that every HIV antibody tested recognized at least one of twelve TCR, and as many as seven, with a binding constant in the 10-8 to 10-9 m range. HIV immunity also affects microbiome tolerance in ways that correlate with susceptibility to specific opportunistic infections.


Assuntos
Síndrome da Imunodeficiência Adquirida/imunologia , HIV/imunologia , Proteínas do Vírus da Imunodeficiência Humana/imunologia , Receptores de Antígenos de Linfócitos T/imunologia , Síndrome da Imunodeficiência Adquirida/virologia , Animais , Autoimunidade , Reações Cruzadas , HIV/química , Proteínas do Vírus da Imunodeficiência Humana/química , Humanos , Microbiota , Mimetismo Molecular , Proteômica , Receptores de Antígenos de Linfócitos T/química
19.
Pathog Dis ; 75(8)2017 11 30.
Artigo em Inglês | MEDLINE | ID: mdl-28911033

RESUMO

Human immunodeficiency virus-1 (HIV-1) continues to be a major global public health issue and priority. Despite the variety of antiretroviral therapies, it seems that an effective vaccine against HIV-1 is still very necessary. An ideal HIV-1 vaccine should be able to elicit both humoral and cellular immunities. In this respect, polyepitope vaccines, incorporated from several conserved regions of HIV-1 proteins, have received much attention recently. Herein, the immunogenicity of the HIV-1 polyepitope protein-based candidate vaccines was evaluated in BALB/c mice. Following the plasmid (pET23a-HIV-1-tat/pol/gag/env) preparation and transformation, the recombinant protein expression was optimized in Escherichia coli BL21 (DE3) host cells. After the HIV-1-top4 protein purification, chitosan and alum adjuvants were added to the vaccines formulations to reinforce the immunogenicity of the candidate vaccines. Mice were subcutaneously immunized three times at 2-week intervals with the candidate vaccines and the elicitation of both humoral and cellular immune responses were investigated. Taken together, the results showed that chitosan adjuvanted candidate vaccine conferred a stronger immunogenicity and elicited higher cellular responses than other candidate vaccines (P < 0.05). Thereby, it seems that co-utilizing of potent adjuvants with the HIV-1 polyepitope protein vaccines can help to open new avenues for strategies for HIV/AIDS vaccine design.


Assuntos
Vacinas contra a AIDS/imunologia , Quitosana/imunologia , Anticorpos Anti-HIV/sangue , HIV-1/imunologia , Proteínas do Vírus da Imunodeficiência Humana/imunologia , Vacinas contra a AIDS/administração & dosagem , Adjuvantes Imunológicos/administração & dosagem , Animais , Proliferação de Células , Quitosana/administração & dosagem , Feminino , Infecções por HIV/prevenção & controle , Linfócitos/fisiologia , Camundongos , Camundongos Endogâmicos BALB C , Baço/citologia
20.
AIDS Res Ther ; 14(1): 42, 2017 Sep 12.
Artigo em Inglês | MEDLINE | ID: mdl-28893275

RESUMO

Collective evidence supporting a role of Antibody-Dependent Cell-Mediated Cytotoxicity (ADCC) in controlling HIV-1 transmission and disease progression emerged in the last few years. Non-neutralizing antibodies (nnAbs) recognizing conserved CD4-induced epitopes on Env and able to mediate potent ADCC against HIV-1-infected cells exposing Env in its CD4-bound conformation have been shown to be present in some RV144 vaccinees and most HIV-1-infected individuals. HIV-1 evolved sophisticated strategies to decrease exposure of this Env conformation by downregulating CD4 and by limiting the overall amount of cell-surface Env. In this review, we will summarize our contribution to this rapidly evolving field, discuss how structural properties of HIV-1 Env might have contributed to the modest efficacy of the RV144 trial and how we recently used this knowledge to develop new strategies aimed at sensitizing HIV-1-infected cells to ADCC mediated by easy to elicit nnAbs.


Assuntos
Citotoxicidade Celular Dependente de Anticorpos , Anticorpos Anti-HIV/imunologia , Infecções por HIV/imunologia , Produtos do Gene env do Vírus da Imunodeficiência Humana/imunologia , Antígenos CD4/química , Antígenos CD4/imunologia , Linfócitos T CD4-Positivos/imunologia , Ensaios Clínicos como Assunto , Produtos do Gene nef/imunologia , Proteína gp120 do Envelope de HIV/imunologia , Infecções por HIV/transmissão , HIV-1/imunologia , Proteínas do Vírus da Imunodeficiência Humana/imunologia , Humanos , Proteínas Virais Reguladoras e Acessórias/imunologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...