Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 21
Filtrar
1.
J Virol ; 97(7): e0065223, 2023 07 27.
Artigo em Inglês | MEDLINE | ID: mdl-37310263

RESUMO

HIV-1 (HIV) infects CD4+ T cells, the gradual depletion of which can lead to AIDS in the absence of antiretroviral therapy (ART). Some cells, however, survive HIV infection and persist as part of the latently infected reservoir that causes recurrent viremia after ART cessation. Improved understanding of the mechanisms of HIV-mediated cell death could lead to a way to clear the latent reservoir. Death induced by survival gene elimination (DISE), an RNA interference (RNAi)-based mechanism, kills cells through short RNAs (sRNAs) with toxic 6-mer seeds (positions 2 to 7 of sRNA). These toxic seeds target the 3' untranslated region (UTR) of mRNAs, decreasing the expression of hundreds of genes critical for cell survival. In most cells under normal conditions, highly expressed cell-encoded nontoxic microRNAs (miRNAs) block access of toxic sRNAs to the RNA-induced silencing complex (RISC) that mediates RNAi, promoting cell survival. HIV has been shown to inhibit the biogenesis of host miRNAs in multiple ways. We now report that HIV infection of cells deficient in miRNA expression or function results in enhanced RISC loading of an HIV-encoded miRNA HIV-miR-TAR-3p, which can kill cells by DISE through a noncanonical (positions 3 to 8) 6-mer seed. In addition, cellular RISC-bound sRNAs shift to lower seed viability. This also occurs after latent HIV provirus reactivation in J-Lat cells, suggesting independence of permissiveness of cells to viral infection. More precise targeting of the balance between protective and cytotoxic sRNAs could provide new avenues to explore novel cell death mechanisms that could be used to kill latent HIV. IMPORTANCE Several mechanisms by which initial HIV infection is cytotoxic to infected cells have been reported and involve various forms of cell death. Characterizing the mechanisms underlying the long-term survival of certain T cells that become persistent provirus reservoirs is critical to developing a cure. We recently discovered death induced by survival gene elimination (DISE), an RNAi-based mechanism of cell death whereby toxic short RNAs (sRNAs) containing 6-mer seed sequences (exerting 6-mer seed toxicity) targeting essential survival genes are loaded into RNA-induced silencing complex (RISC) complexes, resulting in inescapable cell death. We now report that HIV infection in cells with low miRNA expression causes a shift of mostly cellular RISC-bound sRNAs to more toxic seeds. This could prime cells to DISE and is further enhanced by the viral microRNA (miRNA) HIV-miR-TAR-3p, which carries a toxic noncanonical 6-mer seed. Our data provide multiple new avenues to explore novel cell death mechanisms that could be used to kill latent HIV.


Assuntos
Infecções por HIV , HIV-1 , MicroRNAs , Humanos , HIV-1/fisiologia , Latência Viral/genética , MicroRNAs/genética , MicroRNAs/metabolismo , Complexo de Inativação Induzido por RNA/metabolismo
2.
Commun Biol ; 5(1): 878, 2022 08 26.
Artigo em Inglês | MEDLINE | ID: mdl-36028574

RESUMO

Translocated lipopolysaccharide (LPS) activates monocytes via TLR4 and is hypothesized to increase cardiovascular disease risk in persons living with HIV. We tested whether mTOR activity supports LPS-stimulated monocyte production of pro-inflammatory cytokines and tissue factor (TF), as it propels the inflammatory response in several immune cell types besides monocytes. However, multi-omics analyses here demonstrate that mTOR activates a metabolic pathway that limits abundance of these gene products in monocytes. Treatment of primary human monocytes with catalytic mTOR inhibitors (mTORi) increased LPS-induced polyfunctional responses, including production of IL-1ß, IL-6, and the pro-coagulant, TF. NF-κB-driven transcriptional activity is enhanced with LPS stimulation after mTORi treatment to increase expression of F3 (TF). Moreover, intracellular NAD+ availability is restricted due to decreased salvage pathway synthesis. These results document mTOR-mediated restraint of the LPS-induced transcriptional response in monocytes and a metabolic mechanism informing strategies to reverse enhanced risk of coagulopathy in pro-inflammatory states.


Assuntos
Lipopolissacarídeos , Monócitos , Serina-Treonina Quinases TOR , Citocinas , Humanos , Serina-Treonina Quinases TOR/metabolismo , Tromboplastina
3.
PLoS One ; 16(1): e0244855, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33507994

RESUMO

Severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2), the causative agent of the pandemic human respiratory illness COVID-19, is a global health emergency. While severe acute disease has been linked to an expansion of antibody-secreting plasmablasts, we sought to identify B cell responses that correlated with positive clinical outcomes in convalescent patients. We characterized the peripheral blood B cell immunophenotype and plasma antibody responses in 40 recovered non-hospitalized COVID-19 subjects that were enrolled as donors in a convalescent plasma treatment study. We observed a significant negative correlation between the frequency of peripheral blood memory B cells and the duration of symptoms for convalescent subjects. Memory B cell subsets in convalescent subjects were composed of classical CD24+ class-switched memory B cells, but also activated CD24-negative and natural unswitched CD27+ IgD+ IgM+ subsets. Memory B cell frequency was significantly correlated with both IgG1 and IgM responses to the SARS-CoV-2 spike protein receptor binding domain (RBD) in most seropositive subjects. IgM+ memory, but not switched memory, directly correlated with virus-specific antibody responses, and remained stable over 3 months. Our findings suggest that the frequency of memory B cells is a critical indicator of disease resolution, and that IgM+ memory B cells may play an important role in SARS-CoV-2 immunity.


Assuntos
Subpopulações de Linfócitos B/imunologia , COVID-19/imunologia , Adulto , Idoso , Anticorpos Neutralizantes/imunologia , Anticorpos Antivirais/imunologia , Formação de Anticorpos , Linfócitos B/imunologia , Convalescença , Progressão da Doença , Feminino , Humanos , Imunidade/imunologia , Imunoglobulina G/imunologia , Imunoglobulina M/metabolismo , Imunofenotipagem/métodos , Masculino , Pessoa de Meia-Idade , Recuperação de Função Fisiológica/imunologia , SARS-CoV-2/imunologia , SARS-CoV-2/isolamento & purificação
4.
Immunometabolism ; 2(1)2020.
Artigo em Inglês | MEDLINE | ID: mdl-33235814

RESUMO

HIV infection is characterized by elevated glycolytic metabolism in CD4 T cells. In their recent study, Valle-Casuso et al. demonstrated that both increased glucose utilization and glutamine metabolism are essential for HIV infectivity and replication in CD4 T cells. Here, we discuss the broader implications of immunometabolism in studies of HIV persistence and their potential to inform new treatment and curative strategies.

5.
medRxiv ; 2020 Sep 16.
Artigo em Inglês | MEDLINE | ID: mdl-32908991

RESUMO

Severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2), the causative agent of the pandemic human respiratory illness COVID-19, is a global health emergency. While severe acute disease has been linked to an expansion of antibody-secreting plasmablasts, we sought to identify B cell responses that correlated with positive clinical outcomes in convalescent patients. We characterized the peripheral blood B cell immunophenotype and plasma antibody responses in 40 recovered non-hospitalized COVID-19 subjects that were enrolled as donors in a convalescent plasma treatment study. We observed a significant negative correlation between the frequency of peripheral blood memory B cells and the duration of symptoms for convalescent subjects. Memory B cell subsets in convalescent subjects were composed of classical CD24+ class-switched memory B cells, but also activated CD24-negative and natural unswitched CD27+ IgD+ IgM+ subsets. Memory B cell frequency was significantly correlated with both IgG1 and IgM responses to the SARS-CoV-2 spike protein receptor binding domain (RBD). IgM+ memory, but not switched memory, directly correlated with virus-specific antibody responses, and remained stable over time. Our findings suggest that the frequency of memory B cells is a critical indicator of disease resolution, and that IgM+ memory B cells play an important role in SARS-CoV-2 immunity.

6.
Cell Rep ; 31(12): 107810, 2020 06 23.
Artigo em Inglês | MEDLINE | ID: mdl-32579936

RESUMO

Cellular metabolism governs the susceptibility of CD4 T cells to HIV-1 infection. Multiple early post-fusion steps of HIV-1 replication are restricted in resting peripheral blood CD4 T cells; however, molecular mechanisms that underlie metabolic control of these steps remain undefined. Here, we show that mTOR activity following T cell stimulatory signals overcomes metabolic restrictions in these cells by enabling the expansion of dNTPs to fuel HIV-1 reverse transcription (RT), as well as increasing acetyl-CoA to stabilize microtubules that transport RT products. We find that catalytic mTOR inhibition diminishes the expansion of pools of both of these metabolites by limiting glucose and glutamine utilization in several pathways, thereby suppressing HIV-1 infection. We demonstrate how mTOR-coordinated biosyntheses enable the early steps of HIV-1 replication, add metabolic mechanisms by which mTOR inhibitors block HIV-1, and identify some metabolic modules downstream of mTOR as druggable targets for HIV-1 inhibition.


Assuntos
HIV-1/genética , HIV-1/metabolismo , Espaço Intracelular/metabolismo , Transcrição Reversa/genética , Serina-Treonina Quinases TOR/metabolismo , Acetilcoenzima A/metabolismo , Acetilação , Transporte Biológico , Linfócitos T CD4-Positivos/virologia , Citocinas/metabolismo , Retroalimentação Fisiológica , Glicólise , HIV-1/imunologia , HIV-1/fisiologia , Humanos , Lisina/metabolismo , Alvo Mecanístico do Complexo 1 de Rapamicina/metabolismo , Alvo Mecanístico do Complexo 2 de Rapamicina/metabolismo , Microtúbulos/metabolismo , Modelos Biológicos , Receptores de Antígenos de Linfócitos T/metabolismo , Tubulina (Proteína)/metabolismo , Replicação Viral/genética
7.
AIDS ; 33(4): 757-759, 2019 03 15.
Artigo em Inglês | MEDLINE | ID: mdl-30531319

RESUMO

: Evidence suggests that systemic inflammation increases due to HIV infection. C-reactive protein (CRP), interleukin (IL)-6 and tumour necrosis factor (TNF)-α values were compared between HIV-positive and HIV-negative young MSM and transgender women. CRP values were more than 3 mg/l among 49.8% of participants. HIV status was not significantly associated with CRP nor IL-6. TNF-α was significantly higher among HIV-positive participants. These results suggest the need for further study of the causes and health consequences of elevated systemic inflammation among this population.


Assuntos
Infecções por HIV/complicações , Homossexualidade Masculina , Inflamação/patologia , Adolescente , Adulto , Proteína C-Reativa/análise , Humanos , Interleucina-6/sangue , Estudos Longitudinais , Masculino , Fator de Necrose Tumoral alfa/sangue , Adulto Jovem
8.
AIDS ; 32(18): 2847-2851, 2018 11 28.
Artigo em Inglês | MEDLINE | ID: mdl-30234610

RESUMO

: Chronic elevation of plasma cytokines is a key feature of HIV infection. The physiological consequences of this response to infection and its role in HIV persistence are not fully understood. Here, we show that common gamma chain (γc)-cytokines induce both proliferation and expression of T cell exhaustion markers in a mammalian target of rapamycin (mTOR)-dependent fashion, suggesting a possible therapeutic target that, if inhibited, could diminish HIV reservoir expansion, persistence, and resistance to immune surveillance.


Assuntos
Proliferação de Células , Citocinas/metabolismo , Infecções por HIV/patologia , Subunidade gama Comum de Receptores de Interleucina/metabolismo , Transdução de Sinais , Linfócitos T/imunologia , Serina-Treonina Quinases TOR/metabolismo , Adulto , Infecções por HIV/virologia , HIV-1/crescimento & desenvolvimento , HIV-1/imunologia , Humanos , Linfócitos T/fisiologia
9.
PLoS Pathog ; 11(5): e1004864, 2015 May.
Artigo em Inglês | MEDLINE | ID: mdl-26020637

RESUMO

Quiescent CD4+ T cells restrict human immunodeficiency virus type 1 (HIV-1) infection at early steps of virus replication. Low levels of both deoxyribonucleotide triphosphates (dNTPs) and the biosynthetic enzymes required for their de novo synthesis provide one barrier to infection. CD4+ T cell activation induces metabolic reprogramming that reverses this block and facilitates HIV-1 replication. Here, we show that phospholipase D1 (PLD1) links T cell activation signals to increased HIV-1 permissivity by triggering a c-Myc-dependent transcriptional program that coordinates glucose uptake and nucleotide biosynthesis. Decreasing PLD1 activity pharmacologically or by RNA interference diminished c-Myc-dependent expression during T cell activation at the RNA and protein levels. PLD1 inhibition of HIV-1 infection was partially rescued by adding exogenous deoxyribonucleosides that bypass the need for de novo dNTP synthesis. Moreover, the data indicate that low dNTP levels that impact HIV-1 restriction involve decreased synthesis, and not only increased catabolism of these nucleotides. These findings uncover a unique mechanism of action for PLD1 inhibitors and support their further development as part of a therapeutic combination for HIV-1 and other viral infections dependent on host nucleotide biosynthesis.


Assuntos
Linfócitos T CD4-Positivos/virologia , Desoxirribonucleotídeos/metabolismo , Infecções por HIV/virologia , HIV-1/fisiologia , Fosfolipase D/metabolismo , Proteínas Proto-Oncogênicas c-myc/metabolismo , Replicação Viral , Apoptose , Western Blotting , Linfócitos T CD4-Positivos/metabolismo , Proliferação de Células , Células Cultivadas , Replicação do DNA , Infecções por HIV/imunologia , Infecções por HIV/metabolismo , Humanos , Ativação Linfocitária , RNA Mensageiro/genética , Reação em Cadeia da Polimerase em Tempo Real , Reação em Cadeia da Polimerase Via Transcriptase Reversa
10.
J Virol ; 89(4): 2415-24, 2015 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-25505075

RESUMO

UNLABELLED: Members of the APOBEC3 family of cytidine deaminases vary in their proportions of a virion-incorporated enzyme that is localized to mature retrovirus cores. We reported previously that APOBEC3F (A3F) was highly localized into mature human immunodeficiency virus type 1 (HIV-1) cores and identified that L306 in the C-terminal cytidine deaminase (CD) domain contributed to its core localization (C. Song, L. Sutton, M. Johnson, R. D'Aquila, J. Donahue, J Biol Chem 287:16965-16974, 2012, http://dx.doi.org/10.1074/jbc.M111.310839). We have now determined an additional genetic determinant(s) for A3F localization to HIV-1 cores. We found that one pair of leucines in each of A3F's C-terminal and N-terminal CD domains jointly determined the degree of localization of A3F into HIV-1 virion cores. These are A3F L306/L368 (C-terminal domain) and A3F L122/L184 (N-terminal domain). Alterations to one of these specific leucine residues in either of the two A3F CD domains (A3F L368A, L122A, and L184A) decreased core localization and diminished HIV restriction without changing virion packaging. Furthermore, double mutants in these leucine residues in each of A3F's two CD domains (A3F L368A plus L184A or A3F L368A plus L122A) still were packaged into virions but completely lost core localization and anti-HIV activity. HIV virion core localization of A3F is genetically separable from its virion packaging, and anti-HIV activity requires some core localization. IMPORTANCE: Specific leucine-leucine interactions are identified as necessary for A3F's core localization and anti-HIV activity but not for its packaging into virions. Understanding these signals may lead to novel strategies to enhance core localization that may augment effects of A3F against HIV and perhaps of other A3s against retroviruses, parvoviruses, and hepatitis B virus.


Assuntos
Citosina Desaminase/análise , Citosina Desaminase/genética , HIV-1/fisiologia , Montagem de Vírus , Linhagem Celular , Citosina Desaminase/imunologia , Análise Mutacional de DNA , Genes Reporter , HIV-1/química , HIV-1/imunologia , Humanos , Luciferases/análise , Modelos Moleculares , Mutagênese Sítio-Dirigida , Mutação de Sentido Incorreto , Coloração e Rotulagem , beta-Galactosidase/análise
11.
PLoS One ; 9(7): e101367, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-25010677

RESUMO

The global AIDS pandemic continues to expand and in some regions of the world, such as southern Africa, the prevalence of HIV-1 infection exceeds 20%. The devastating spread of the virus in young women in these countries appears disproportional to overall risk of infection. Regions with high prevalence of HIV-1 are often also highly endemic for other pathogenic viruses including HSV, CMV and HTLV. We propose that acquisition by HIV-1 of the envelope glycoproteins of other viruses, in a process we call "natural pseudotyping," expands the cellular tropism of HIV-1, enabling it to infect female genital epithelial cells directly and thereby dramatically increasing risk of infection during sexual intercourse. In this proof-of-concept study, we demonstrate that when HIV-1 co-infects T cells along with the gammaretrovirus xenotropic murine leukemia virus-related virus (XMRV), progeny HIV-1 particles are produced capable of infecting primary vaginal, ectocervical and endocervical epithelial cells. These cell types are normally resistant to HIV-1 infection. Infection of primary genital cells was neutralized by antisera against the XMRV glycoprotein, confirming that infection was mediated by the XMRV glycoprotein acquired through pseudotyping of HIV. Inhibition by AZT showed that active replication of HIV-1 occurred in these cells and ruled out non-specific endocytic uptake of the virus. These results demonstrate that natural pseudotyping can expand the tropism of HIV-1 to include genital epithelial cells and have potential implications for sexual transmission of the virus.


Assuntos
Colo do Útero/citologia , Coito , Células Epiteliais/virologia , Infecções por HIV/transmissão , HIV-1/fisiologia , Vagina/citologia , Anticorpos Neutralizantes/imunologia , Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD4-Positivos/virologia , Linhagem Celular , Feminino , HIV-1/imunologia , Humanos , Tropismo Viral , Replicação Viral , Vírus Relacionado ao Vírus Xenotrópico da Leucemia Murina/fisiologia
12.
J Biol Chem ; 289(2): 600-16, 2014 Jan 10.
Artigo em Inglês | MEDLINE | ID: mdl-24257753

RESUMO

The lack of innovative drug targets for glioblastoma multiforme (GBM) limits patient survival to approximately 1 year following diagnosis. The pro-survival kinase Akt provides an ideal target for the treatment of GBM as Akt signaling is frequently activated in this cancer type. However, the central role of Akt in physiological processes limits its potential as a therapeutic target. In this report, we show that the lipid-metabolizing enzyme phospholipaseD(PLD) is a novel regulator of Akt inGBM.Studies using a combination of small molecule PLD inhibitors and siRNA knockdowns establish phosphatidic acid, the product of the PLD reaction, as an essential component for the membrane recruitment and activation of Akt. Inhibition of PLD enzymatic activity and subsequent Akt activation decreases GBM cell viability by specifically inhibiting autophagic flux. We propose a mechanism whereby phosphorylation of beclin1 by Akt prevents binding of Rubicon (RUN domain cysteine-rich domain containing beclin1-interacting protein), an interaction known to inhibit autophagic flux. These findings provide a novel framework through which Akt inhibition can be achieved without directly targeting the kinase.


Assuntos
Autofagia/fisiologia , Fosfolipase D/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Transdução de Sinais/fisiologia , Animais , Proteínas Reguladoras de Apoptose/metabolismo , Autofagia/efeitos dos fármacos , Autofagia/genética , Proteínas Relacionadas à Autofagia , Proteína Beclina-1 , Linhagem Celular , Linhagem Celular Tumoral , Sobrevivência Celular/genética , Sobrevivência Celular/fisiologia , Meios de Cultura Livres de Soro/farmacologia , Inibidores Enzimáticos/farmacologia , Glioblastoma/genética , Glioblastoma/metabolismo , Glioblastoma/patologia , Células HEK293 , Compostos Heterocíclicos com 3 Anéis/farmacologia , Humanos , Immunoblotting , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Proteínas de Membrana/metabolismo , Microscopia Confocal , Ácidos Fosfatídicos/metabolismo , Fosfolipase D/antagonistas & inibidores , Fosfolipase D/genética , Fosforilação/efeitos dos fármacos , Proteínas Proto-Oncogênicas c-akt/antagonistas & inibidores , Proteínas Proto-Oncogênicas c-akt/genética , Interferência de RNA , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/genética
13.
J Virol ; 88(1): 592-603, 2014 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-24173214

RESUMO

Apolipoprotein L1 (APOL1) is a major component of the human innate immune response against African trypanosomes. Although the mechanism of the trypanolytic activity of circulating APOL1 has been recently clarified, the intracellular function(s) of APOL1 in human cells remains poorly defined. Like that of many genes linked to host immunity, APOL1 expression is induced by proinflammatory cytokines gamma interferon (IFN-γ) and tumor necrosis factor alpha (TNF-α). Additionally, IFN-γ-polarized macrophages that potently restrict HIV-1 replication express APOL1, which suggests that APOL1 may contribute to HIV-1 suppression. Here, we report that APOL1 inhibits HIV-1 replication by multiple mechanisms. We found that APOL1 protein targeted HIV-1 Gag for degradation by the endolysosomal pathway. Interestingly, we found that APOL1 stimulated both endocytosis and lysosomal biogenesis by promoting nuclear localization of transcription factor EB (TFEB) and expression of TFEB target genes. Moreover, we demonstrated that APOL1 depletes cellular viral accessory protein Vif, which counteracts the host restriction factor APOBEC3G, via a pathway involving degradation of Vif in lysosomes and by secretion of Vif in microvesicles. As a result of Vif depletion by APOL1, APOBEC3G was not degraded and reduced infectivity of progeny virions. In support of this model, we also showed that endogenous expression of APOL1 in differentiated U937 monocytic cells stimulated with IFN-γ resulted in a reduced production of virus particles. This finding supports the hypothesis that induction of APOL1 contributes to HIV-1 suppression in differentiated monocytes. Deciphering the precise mechanism of APOL1-mediated HIV-1 restriction may facilitate the design of unique therapeutics to target HIV-1 replication.


Assuntos
Apolipoproteínas/fisiologia , Infecções por HIV/imunologia , HIV-1/isolamento & purificação , Imunidade Inata , Lipoproteínas HDL/fisiologia , Apolipoproteína L1 , Diferenciação Celular , Linhagem Celular , Endocitose , Citometria de Fluxo , Produtos do Gene gag/imunologia , HIV-1/fisiologia , Humanos , Interferons/imunologia , Macrófagos/imunologia , Replicação Viral
14.
Virology ; 432(1): 110-9, 2012 Oct 10.
Artigo em Inglês | MEDLINE | ID: mdl-22748181

RESUMO

Caveolin-1 is an integral membrane protein primarily responsible for the formation of membrane structures known as caveolae. Caveolae are specialized lipid rafts involved in protein trafficking, cholesterol homeostasis, and a number of signaling functions. It has been demonstrated that caveolin-1 suppresses HIV-1 protein expression. We found that co-transfecting cells with HIV-1 and caveolin-1 constructs, results in a marked decrease in the level of HIV-1 transcription relative to cells transfected with HIV-1 DNA alone. Correspondingly, reduction of endogenous caveolin-1 expression by siRNA-mediated silencing resulted in an enhancement of HIV-1 replication. Further, we observed a loss of caveolin-mediated suppression of HIV-1 transcription in promoter studies with reporters containing mutations in the NF-κB binding site. Our analysis of the posttranslational modification status of the p65 subunit of NF-κB demonstrates hypoacetylation of p65 in the presence of caveolin-1. Since hypoacetylated p65 has been shown to inhibit transcription, we conclude that caveolin-1 inhibits HIV-1 transcription through a NF-κB-dependent mechanism.


Assuntos
Caveolina 1/metabolismo , HIV-1/imunologia , HIV-1/fisiologia , Interações Hospedeiro-Patógeno , NF-kappa B/metabolismo , Replicação Viral , Acetilação , Linhagem Celular , Humanos
15.
J Biol Chem ; 286(32): 28498-510, 2011 Aug 12.
Artigo em Inglês | MEDLINE | ID: mdl-21705339

RESUMO

HIV-1 Gag precursor directs virus particle assembly and release. In a search for Gag-interacting proteins that are involved in late stages of the HIV-1 replication cycle, we performed yeast two-hybrid screening against a human cDNA library and identified the non-muscle actin filament cross-linking protein filamin A as a novel Gag binding partner. The 280-kDa filamin A regulates cortical actin network dynamics and participates in the anchoring of membrane proteins to the actin cytoskeleton. Recent studies have shown that filamin A facilitates HIV-1 cell-to-cell transmission by binding to HIV receptors and coreceptors and regulating their clustering on the target cell surface. Here we report a novel role for filamin A in HIV-1 Gag intracellular trafficking. We demonstrate that filamin A interacts with the capsid domain of HIV-1 Gag and that this interaction is involved in particle release in a productive manner. Disruption of this interaction eliminated Gag localization at the plasma membrane and induced Gag accumulation within internal compartments. Moreover, blocking clathrin-dependent endocytic pathways did not relieve the restriction to particle release induced by filamin A depletion. These results suggest that filamin A is involved in the distinct step of the Gag trafficking pathway. The discovery of the Gag-filamin A interaction may provide a new therapeutic target for the treatment of HIV infection.


Assuntos
Proteínas Contráteis/metabolismo , Infecções por HIV/mortalidade , HIV-1/fisiologia , Proteínas dos Microfilamentos/metabolismo , Montagem de Vírus/fisiologia , Clatrina/genética , Clatrina/metabolismo , Proteínas Contráteis/genética , Endocitose/genética , Filaminas , Biblioteca Gênica , Infecções por HIV/genética , Infecções por HIV/transmissão , HIV-1/patogenicidade , Células HeLa , Humanos , Proteínas dos Microfilamentos/genética , Transporte Proteico/genética , Saccharomyces cerevisiae , Técnicas do Sistema de Duplo-Híbrido , Montagem de Vírus/efeitos dos fármacos , Produtos do Gene gag do Vírus da Imunodeficiência Humana
16.
J Virol ; 85(15): 7699-709, 2011 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-21613400

RESUMO

Cholesterol plays an essential role in the life cycle of several enveloped viruses. Many of these viruses manipulate host cholesterol metabolism to facilitate their replication. HIV-1 infection of CD4(+) T cells activates the sterol regulatory element-binding protein 2 (SREBP2) transcriptional program, which includes genes involved in cholesterol homeostasis. However, the role of SREBP2-dependent transcription in HIV-1 biology has not been fully examined. Here, we identify TFII-I, a gene critical for HIV-1 transcription in activated T cells, as a novel SREBP2 target gene. We found TFII-I expression increased after HIV-1 infection or activation of human primary CD4(+) T cells. We show that inhibition of SREBP2 activity reduced TFII-I induction in response to these stimuli. More importantly, small interfering RNA (siRNA)-mediated gene silencing of either SREBP2 or TFII-I significantly reduced HIV-1 production in CD4(+) T cells. We also found that TFII-I potentiates Tat-dependent viral gene expression, consistent with a role at the level of HIV-1 transcription. Collectively, our results demonstrate for the first time that HIV-1 transcription in T cells is linked to cholesterol homeostasis through control of TFII-I expression by SREBP2.


Assuntos
Colesterol/metabolismo , HIV-1/genética , Homeostase/fisiologia , Ativação Linfocitária , Proteína de Ligação a Elemento Regulador de Esterol 2/fisiologia , Linfócitos T/imunologia , Transcrição Gênica/fisiologia , Sequência de Bases , Linhagem Celular , Primers do DNA , Citometria de Fluxo , Humanos , Ligação Proteica , Proteína de Ligação a Elemento Regulador de Esterol 2/metabolismo , Fatores de Transcrição TFII/genética
17.
Virology ; 408(1): 119-27, 2010 Dec 05.
Artigo em Inglês | MEDLINE | ID: mdl-20937511

RESUMO

Podocyte damage induced by HIV-1 is critical to the pathogenesis of HIV-1 associated nephropathy (HIVAN) and is believed to result from productive replication of the virus. Here we demonstrate that HIV-1 readily enters human podocytes by a dynamin-mediated endocytosis but does not establish productive infection. We provide evidence suggesting that viral nucleic acids and proteins detected in podocytes are delivered by viral particles internalized by the cells. Endocytosed HIV-1 is only transiently harbored by podocytes and is subsequently released to the extracellular milieu as fully infectious virus. Similarly, primary podocytes established from normal human urine do not support productive infection by HIV-1 but sustain replication of VSV-G pseudotyped virus that bypasses HIV-1 entry receptors. Moreover, transfected podocytes expressing CD4 and CXCR4 receptors support productive replication of HIV-1. This further confirms that lack of HIV-1 entry receptors is the major barrier preventing productive infection of podocytes in vitro.


Assuntos
HIV-1/fisiologia , Podócitos/virologia , Receptores de HIV/biossíntese , Internalização do Vírus , Replicação Viral , Linhagem Celular , Endocitose , HIV-1/crescimento & desenvolvimento , HIV-1/patogenicidade , Humanos , Receptores de HIV/genética , Liberação de Vírus
18.
Virology ; 400(1): 68-75, 2010 Apr 25.
Artigo em Inglês | MEDLINE | ID: mdl-20153011

RESUMO

Human cytidine deaminases, including APOBEC3G (A3G) and A3F, are part of a cellular defense system against retroviruses and retroelements including non-LTR retrotransposons LINE-1 (L1) and Alu. Expression of cellular A3 proteins is sufficient for inhibition of L1 and Alu retrotransposition, but the effect of A3 proteins transferred in exosomes on retroelement mobilization is unknown. Here, we demonstrate for the first time that exosomes secreted by CD4(+)H9 T cells and mature monocyte-derived dendritic cells encapsidate A3G and A3F and inhibit L1 and Alu retrotransposition. A3G is the major contributor to the inhibitory activity of exosomes, however, the contribution of A3F in H9 exosomes cannot be excluded. Additionally, we show that exosomes encapsidate mRNAs coding for A3 proteins. A3G mRNA, and less so A3F, was enriched in exosomes secreted by H9 cells. Exosomal A3G mRNA was functional in vitro. Whether exosomes inhibit retrotransposons in vivo requires further investigation.


Assuntos
Elementos Alu , Citidina Desaminase/metabolismo , Citosina Desaminase/metabolismo , Exossomos/enzimologia , Exossomos/genética , Elementos Nucleotídeos Longos e Dispersos , Desaminase APOBEC-3G , Linfócitos T CD4-Positivos/metabolismo , Linhagem Celular , Células Dendríticas/metabolismo , Humanos , Plasmídeos/genética , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Retroviridae/genética
19.
J Virol ; 83(2): 512-21, 2009 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-18987139

RESUMO

The human cytidine deaminase APOBEC3G (A3G) is a part of a cellular defense system against human immunodeficiency virus type 1 (HIV-1) and other retroviruses. Antiretroviral activity of A3G can be severely blunted in the presence of the HIV-1 protein Vif. However, in some cells expressing the enzymatically active low-molecular-mass form of A3G, HIV-1 replication is restricted at preintegration steps, before accumulation of Vif. Here, we show that A3G can be secreted by cells in exosomes that confer resistance to both vif-defective and wild-type HIV-1 in exosome recipient cells. Our results also suggest that A3G is the major exosomal component responsible for the anti-HIV-1 activity of exosomes. However, enzymatic activity of encapsidated A3G does not correlate with the observed limited cytidine deamination in HIV-1 DNA, suggesting that A3G-laden exosomes restrict HIV-1 through a nonenzymatic mechanism. Real-time PCR quantitation demonstrated that A3G exosomes reduce accumulation of HIV-1 reverse transcription products and steady-state levels of HIV-1 Gag and Vif proteins. Our findings suggest that A3G exosomes could be developed into a novel class of anti-HIV-1 therapeutics.


Assuntos
Citidina Desaminase/imunologia , Exossomos/imunologia , HIV-1/imunologia , Desaminase APOBEC-3G , Linhagem Celular , DNA Viral/genética , DNA Viral/metabolismo , Humanos , Reação em Cadeia da Polimerase , Proteínas Virais/biossíntese , Replicação Viral/imunologia
20.
PLoS Biol ; 4(7): e229, 2006 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-16774454

RESUMO

Activation of the insect innate immune system is dependent on a limited number of pattern recognition receptors (PRRs) capable of interacting with pathogen-associated molecular pattern. Here we report a novel role of an alternatively spliced hypervariable immunoglobulin domain-encoding gene, Dscam, in generating a broad range of PRRs implicated in immune defense in the malaria vector Anopheles gambiae. The mosquito Down syndrome cell adhesion molecule gene, AgDscam, has a complex genome organization with 101 exons that can produce over 31,000 potential alternative splice forms with different combinations of adhesive domains and interaction specificities. AgDscam responds to infection by producing pathogen challenge-specific splice form repertoires. Transient silencing of AgDscam compromises the mosquito's resistance to infections with bacteria and the malaria parasite Plasmodium. AgDscam is mediating phagocytosis of bacteria with which it can associate and defend against in a splice form-specific manner. AgDscam is a hypervariable PRR of the A. gambiae innate immune system.


Assuntos
Anopheles/genética , Anopheles/imunologia , Proteínas de Insetos/genética , Receptores de Reconhecimento de Padrão/genética , Processamento Alternativo , Animais , Anopheles/microbiologia , Anopheles/parasitologia , Bactérias/imunologia , Éxons , Feminino , Genoma de Inseto , Imunidade Inata , Imunoglobulinas , Proteínas de Insetos/química , Dados de Sequência Molecular , Fagocitose , Plasmodium/imunologia , Estrutura Terciária de Proteína , Interferência de RNA , Receptores de Reconhecimento de Padrão/química
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA