Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 4 de 4
Filter
Add more filters










Database
Language
Publication year range
1.
Cell Host Microbe ; 24(5): 689-702.e7, 2018 11 14.
Article in English | MEDLINE | ID: mdl-30344003

ABSTRACT

Long-lived resting memory CD4+ T cells (TCM) are a major reservoir of latent HIV infection. We hypothesized that latent HIV-TCM cells are maintained by aberrant expression of cell survival factors, including XIAP, BIRC2/cIAP1, and beclin-1. DIABLO/SMAC mimetics are therapeutic agents that compromise cell survival by hijacking host apoptotic machinery. We found that DIABLO/SMAC mimetics (birinapant, GDC-0152, and embelin) selectively kill HIV-TCM without increasing virus production or targeting uninfected TCM. Treatment of HIV-TCM with DIABLO/SMAC mimetics promoted XIAP and BIRC2 degradation, which triggered autophagy and the formation of a cell death complex consisting of pro-apoptotic (FADD, RIPK1, RIPK3, and caspase 8) and autophagy (ATG5, ATG7, and SQSTM1) proteins. Genetic or pharmacological inhibition of autophagy induction, but not autophagy-mediated degradation, abrogated this interaction and subsequent cell death. Our findings identify a mechanism whereby DIABLO/SMAC mimetics exploit autophagy and apoptotic machinery to selectively induce killing of HIV-TCM without viral reactivation while sparing uninfected cells.


Subject(s)
Apoptosis/drug effects , Apoptosis/physiology , Autophagy/drug effects , Autophagy/physiology , CD4-Positive T-Lymphocytes/immunology , HIV Infections/immunology , HIV-1/drug effects , HIV-1/physiology , Apoptosis Regulatory Proteins , Autophagy-Related Protein 5/metabolism , Autophagy-Related Protein 7/metabolism , Baculoviral IAP Repeat-Containing 3 Protein/metabolism , Beclin-1/metabolism , Benzoquinones/pharmacology , Caspase 8/metabolism , Cell Death , Cell Line , Cyclohexanes/pharmacology , Dipeptides/pharmacology , Fas-Associated Death Domain Protein/metabolism , HIV-1/pathogenicity , Humans , Indoles/pharmacology , Inhibitor of Apoptosis Proteins/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Mitochondrial Proteins/metabolism , Pyrroles/pharmacology , Receptor-Interacting Protein Serine-Threonine Kinases/metabolism , Sequestosome-1 Protein/metabolism , Ubiquitin-Protein Ligases/metabolism , X-Linked Inhibitor of Apoptosis Protein/metabolism
2.
J Biol Chem ; 293(16): 5808-5820, 2018 04 20.
Article in English | MEDLINE | ID: mdl-29475942

ABSTRACT

In this study, we investigated the effects of the dual phosphatidylinositol 3-kinase/mechanistic target of rapamycin (PI3K/MTOR) inhibitor dactolisib (NVP-BEZ235), the PI3K/MTOR/bromodomain-containing protein 4 (BRD4) inhibitor SF2523, and the bromodomain and extra terminal domain inhibitor JQ1 on the productive infection of primary macrophages with human immunodeficiency type-1 (HIV). These inhibitors did not alter the initial susceptibility of macrophages to HIV infection. However, dactolisib, JQ1, and SF2523 all decreased HIV replication in macrophages in a dose-dependent manner via degradation of intracellular HIV through autophagy. Macrophages treated with dactolisib, JQ1, or SF2523 displayed an increase in LC3B lipidation combined with SQSTM1 degradation without inducing increased cell death. LC3B-II levels were further increased in the presence of pepstatin A suggesting that these inhibitors induce autophagic flux. RNA interference for ATG5 and ATG7 and pharmacological inhibitors of autophagosome-lysosome fusion and of lysosomal hydrolases all blocked the inhibition of HIV. Thus, we demonstrate that the mechanism of PI3K/MTOR and PI3K/MTOR/BRD4 inhibitor suppression of HIV requires the formation of autophagosomes, as well as their subsequent maturation into autolysosomes. These data provide further evidence in support of a role for autophagy in the control of HIV infection and open new avenues for the use of this class of drugs in HIV therapy.


Subject(s)
Anti-HIV Agents/pharmacology , Autophagy/drug effects , Azepines/pharmacology , HIV Infections/drug therapy , HIV-1/drug effects , Imidazoles/pharmacology , Protein Kinase Inhibitors/pharmacology , Quinolines/pharmacology , Triazoles/pharmacology , Virus Replication/drug effects , Cell Cycle Proteins , Cells, Cultured , HIV Infections/metabolism , HIV Infections/pathology , HIV Infections/virology , HIV-1/physiology , Humans , Macrophages/drug effects , Macrophages/metabolism , Macrophages/pathology , Macrophages/virology , Nuclear Proteins/antagonists & inhibitors , Nuclear Proteins/metabolism , Phosphatidylinositol 3-Kinase/metabolism , Phosphoinositide-3 Kinase Inhibitors , Signal Transduction/drug effects , TOR Serine-Threonine Kinases/antagonists & inhibitors , TOR Serine-Threonine Kinases/metabolism , Transcription Factors/antagonists & inhibitors , Transcription Factors/metabolism
3.
PLoS Pathog ; 11(6): e1005018, 2015 Jun.
Article in English | MEDLINE | ID: mdl-26115100

ABSTRACT

HIV Nef acts as an anti-autophagic maturation factor through interaction with beclin-1 (BECN1). We report that exposure of macrophages to infectious or non-infectious purified HIV induces toll-like receptor 8 (TLR8) and BECN1 dependent dephosphorylation and nuclear translocation of TFEB and that this correlates with an increase in autophagy markers. RNA interference for ATG13, TFEB, TLR8, or BECN1 inhibits this HIV-induced autophagy. However, once HIV establishes a productive infection, TFEB phosphorylation and cytoplasmic sequestration are increased resulting in decreased autophagy markers. Moreover, by 7 d post-infection, autophagy levels are similar to mock infected controls. Conversely, although Nef deleted HIV similarly induces TFEB dephosphorylation and nuclear localization, and increases autophagy, these levels remain elevated during continued productive infection. Thus, the interaction between HIV and TLR8 serves as a signal for autophagy induction that is dependent upon the dephosphorylation and nuclear translocation of TFEB. During permissive infection, Nef binds BECN1 resulting in mammalian target of rapamycin (MTOR) activation, TFEB phosphorylation and cytosolic sequestration, and the inhibition of autophagy. To our knowledge, this is the first report of a virus modulating TFEB localization and helps to explain how HIV modulates autophagy to promote its own replication and cell survival.


Subject(s)
Autophagy/immunology , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/metabolism , HIV-1/immunology , Macrophages/metabolism , nef Gene Products, Human Immunodeficiency Virus/immunology , Apoptosis Regulatory Proteins/metabolism , Beclin-1 , Humans , Membrane Proteins/metabolism , Microtubule-Associated Proteins/metabolism , TOR Serine-Threonine Kinases/metabolism , Toll-Like Receptor 8/metabolism , Virus Replication/immunology
4.
J Biol Chem ; 290(8): 5028-5040, 2015 Feb 20.
Article in English | MEDLINE | ID: mdl-25540204

ABSTRACT

Histone deacetylase inhibitors (HDACi) are being evaluated in a "shock-and-kill" therapeutic approach to reverse human immunodeficiency virus type-1 (HIV) latency from CD4(+) T cells. Using this approach, HDACi have induced HIV RNA synthesis in latently infected cells from some patients. The hope is that the increase in viral production will lead to killing of the infected cell either by the virus itself or by the patient's immune system, a "sterilizing cure." Although administered within the context of combination antiretroviral therapy, the infection of bystander cells remains a concern. In this study, we investigated the effect of HDACi (belinostat, givinostat, panobinostat, romidepsin, and vorinostat) on the productive infection of macrophages. We demonstrate that the HDACi tested do not alter the initial susceptibility of macrophages to HIV infection. However, we demonstrate that HDACi decrease HIV release from macrophages in a dose-dependent manner (belinostat < givinostat < vorinostat < panobinostat < romidepsin) via degradation of intracellular HIV through the canonical autophagy pathway. This mechanism involves unc-51-like autophagy-activating kinase 1 (ULK1) and the inhibition of the mammalian target of rapamycin and requires the formation of autophagosomes and their maturation into autolysosomes in the absence of increased cell death. These data provide further evidence in support of a role for autophagy in the control of HIV infection and suggest that careful consideration of off-target effects will be essential if HDACi are to be a component of a multipronged approach to eliminate latently infected cells.


Subject(s)
Autophagy/drug effects , HIV Infections , HIV-1/physiology , Histone Deacetylase Inhibitors/pharmacology , Macrophages , Virus Latency/drug effects , Autophagy-Related Protein-1 Homolog , Female , HIV Infections/drug therapy , HIV Infections/enzymology , HIV Infections/pathology , Humans , Intracellular Signaling Peptides and Proteins/metabolism , Lysosomes/pathology , Lysosomes/virology , Macrophages/enzymology , Macrophages/pathology , Macrophages/virology , Male , Protein Serine-Threonine Kinases/metabolism , TOR Serine-Threonine Kinases/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...