Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 7 de 7
Filter
Add more filters










Database
Language
Publication year range
1.
EMBO Mol Med ; 13(8): e13901, 2021 08 09.
Article in English | MEDLINE | ID: mdl-34289240

ABSTRACT

HIV-1 infects lymphoid and myeloid cells, which can harbor a latent proviral reservoir responsible for maintaining lifelong infection. Glycolytic metabolism has been identified as a determinant of susceptibility to HIV-1 infection, but its role in the development and maintenance of HIV-1 latency has not been elucidated. By combining transcriptomic, proteomic, and metabolomic analyses, we here show that transition to latent HIV-1 infection downregulates glycolysis, while viral reactivation by conventional stimuli reverts this effect. Decreased glycolytic output in latently infected cells is associated with downregulation of NAD+ /NADH. Consequently, infected cells rely on the parallel pentose phosphate pathway and its main product, NADPH, fueling antioxidant pathways maintaining HIV-1 latency. Of note, blocking NADPH downstream effectors, thioredoxin and glutathione, favors HIV-1 reactivation from latency in lymphoid and myeloid cellular models. This provides a "shock and kill effect" decreasing proviral DNA in cells from people living with HIV/AIDS. Overall, our data show that downmodulation of glycolysis is a metabolic signature of HIV-1 latency that can be exploited to target latently infected cells with eradication strategies.


Subject(s)
HIV Infections , HIV-1 , CD4-Positive T-Lymphocytes , Down-Regulation , Glycolysis , Humans , Oxidative Stress , Proteomics , Virus Activation , Virus Latency
2.
EMBO Mol Med ; 13(5): e13314, 2021 05 07.
Article in English | MEDLINE | ID: mdl-33793064

ABSTRACT

Reactive oxygen species (ROS) regulates the replication of human immunodeficiency virus (HIV-1) during infection. However, the application of this knowledge to develop therapeutic strategies remained unsuccessful due to the harmful consequences of manipulating cellular antioxidant systems. Here, we show that vanadium pentoxide (V2 O5 ) nanosheets functionally mimic natural glutathione peroxidase activity to mitigate ROS associated with HIV-1 infection without adversely affecting cellular physiology. Using genetic reporters of glutathione redox potential and hydrogen peroxide, we showed that V2 O5 nanosheets catalyze ROS neutralization in HIV-1-infected cells and uniformly block viral reactivation and replication. Mechanistically, V2 O5 nanosheets suppressed HIV-1 by affecting the expression of pathways coordinating redox balance, virus transactivation (e.g., NF-κB), inflammation, and apoptosis. Importantly, a combination of V2 O5 nanosheets with a pharmacological inhibitor of NF-κB (BAY11-7082) abrogated reactivation of HIV-1. Lastly, V2 O5 nanosheets inhibit viral reactivation upon prostratin stimulation of latently infected CD4+ T cells from HIV-infected patients receiving suppressive antiretroviral therapy. Our data successfully revealed the usefulness of V2 O5 nanosheets against HIV and suggested nanozymes as future platforms to develop interventions against infectious diseases.


Subject(s)
HIV Infections , HIV-1 , Antioxidants , CD4-Positive T-Lymphocytes , HIV Infections/drug therapy , Humans , Oxidation-Reduction , Virus Latency
3.
Proc Natl Acad Sci U S A ; 117(10): 5486-5493, 2020 03 10.
Article in English | MEDLINE | ID: mdl-32094182

ABSTRACT

HIV-1 capsid core disassembly (uncoating) must occur before integration of viral genomic DNA into the host chromosomes, yet remarkably, the timing and cellular location of uncoating is unknown. Previous studies have proposed that intact viral cores are too large to fit through nuclear pores and uncoating occurs in the cytoplasm in coordination with reverse transcription or at the nuclear envelope during nuclear import. The capsid protein (CA) content of the infectious viral cores is not well defined because methods for directly labeling and quantifying the CA in viral cores have been unavailable. In addition, it has been difficult to identify the infectious virions because only one of ∼50 virions in infected cells leads to productive infection. Here, we developed methods to analyze HIV-1 uncoating by direct labeling of CA with GFP and to identify infectious virions by tracking viral cores in living infected cells through viral DNA integration and proviral DNA transcription. Astonishingly, our results show that intact (or nearly intact) viral cores enter the nucleus through a mechanism involving interactions with host protein cleavage and polyadenylation specificity factor 6 (CPSF6), complete reverse transcription in the nucleus before uncoating, and uncoat <1.5 h before integration near (<1.5 µm) their genomic integration sites. These results fundamentally change our current understanding of HIV-1 postentry replication events including mechanisms of nuclear import, uncoating, reverse transcription, integration, and evasion of innate immunity.


Subject(s)
Capsid Proteins/analysis , Cell Nucleus/virology , HIV Infections/virology , HIV-1/physiology , Virus Integration , Virus Uncoating , Active Transport, Cell Nucleus , Capsid Proteins/metabolism , Green Fluorescent Proteins/analysis , Humans , Nuclear Pore/metabolism , Proteolysis , Virus Replication , mRNA Cleavage and Polyadenylation Factors/metabolism
4.
Sci Transl Med ; 11(518)2019 11 13.
Article in English | MEDLINE | ID: mdl-31723039

ABSTRACT

The capacity of Mycobacterium tuberculosis (Mtb) to tolerate multiple antibiotics represents a major problem in tuberculosis (TB) management. Heterogeneity in Mtb populations is one of the factors that drives antibiotic tolerance during infection. However, the mechanisms underpinning this variation in bacterial population remain poorly understood. Here, we show that phagosomal acidification alters the redox physiology of Mtb to generate a population of replicating bacteria that display drug tolerance during infection. RNA sequencing of this redox-altered population revealed the involvement of iron-sulfur (Fe-S) cluster biogenesis, hydrogen sulfide (H2S) gas, and drug efflux pumps in antibiotic tolerance. The fraction of the pH- and redox-dependent tolerant population increased when Mtb infected macrophages with actively replicating HIV-1, suggesting that redox heterogeneity could contribute to high rates of TB therapy failure during HIV-TB coinfection. Pharmacological inhibition of phagosomal acidification by the antimalarial drug chloroquine (CQ) eradicated drug-tolerant Mtb, ameliorated lung pathology, and reduced postchemotherapeutic relapse in in vivo models. The pharmacological profile of CQ (C max and AUClast) exhibited no major drug-drug interaction when coadministered with first line anti-TB drugs in mice. Our data establish a link between phagosomal pH, redox metabolism, and drug tolerance in replicating Mtb and suggest repositioning of CQ to shorten TB therapy and achieve a relapse-free cure.


Subject(s)
Drug Resistance, Bacterial , Mycobacterium tuberculosis/growth & development , Acids , Animals , Antitubercular Agents/pharmacology , Antitubercular Agents/therapeutic use , Bacterial Proteins/metabolism , Chloroquine/pharmacology , Chloroquine/therapeutic use , Cysteine/metabolism , Drug Interactions , Drug Resistance, Bacterial/drug effects , Drug Resistance, Multiple, Bacterial/drug effects , Female , HIV Infections/microbiology , Macrophages/drug effects , Macrophages/microbiology , Macrophages/pathology , Mice, Inbred BALB C , Mycobacterium tuberculosis/drug effects , Mycobacterium tuberculosis/genetics , Oxidation-Reduction , Phagosomes/drug effects , Phagosomes/microbiology , RNA-Seq , Recurrence , Transcriptome/drug effects , Transcriptome/genetics , Tuberculosis/drug therapy , Tuberculosis/microbiology
5.
ACS Infect Dis ; 2(9): 592-607, 2016 09 09.
Article in English | MEDLINE | ID: mdl-27759382

ABSTRACT

The global mechanisms and associated molecular alterations that occur in drug-resistant mycobacteria are poorly understood. To address this, we obtain genomics data and then construct a genome-scale response network in isoniazid-resistant Mycobacterium smegmatis and apply a network-mining algorithm. Through this, we decipher global alterations in an unbiased manner and identify emergent vulnerabilities in resistant bacilli, of which redox response was prominent. Using phenotypic profiling, we find that resistant bacilli exhibit collateral sensitivity to several compounds that block antioxidant responses. We find that nanogram/milliliter concentrations of ebselen, vancomycin, and phenylarsine oxide, in combination with isoniazid, are highly effective against Mycobacterium tuberculosis H37Rv and three clinical drug-resistant strains. Dynamic measurements of cytoplasmic redox potential revealed a surprisingly diminished capacity of clinical drug-resistant strains to counteract oxidative stress, providing a mechanistic basis for efficient and synergistic mycobactericidal activity of the drug combinations. Ebselen and vancomycin appear to be promising repurposable drugs.


Subject(s)
Antitubercular Agents/pharmacology , Drug Resistance, Bacterial , Mycobacterium Infections, Nontuberculous/microbiology , Mycobacterium smegmatis/drug effects , Drug Synergism , Genome, Bacterial , Humans , Isoniazid/pharmacology , Microbial Sensitivity Tests , Mycobacterium smegmatis/genetics , Mycobacterium smegmatis/metabolism , Mycobacterium tuberculosis/drug effects , Mycobacterium tuberculosis/genetics , Mycobacterium tuberculosis/metabolism , Oxidation-Reduction , Tuberculosis/microbiology
6.
J Biol Chem ; 290(2): 1020-38, 2015 Jan 09.
Article in English | MEDLINE | ID: mdl-25406321

ABSTRACT

Redox signaling plays a crucial role in the pathogenesis of human immunodeficiency virus type-1 (HIV-1). The majority of HIV redox research relies on measuring redox stress using invasive technologies, which are unreliable and do not provide information about the contributions of subcellular compartments. A major technological leap emerges from the development of genetically encoded redox-sensitive green fluorescent proteins (roGFPs), which provide sensitive and compartment-specific insights into redox homeostasis. Here, we exploited a roGFP-based specific bioprobe of glutathione redox potential (E(GSH); Grx1-roGFP2) and measured subcellular changes in E(GSH) during various phases of HIV-1 infection using U1 monocytic cells (latently infected U937 cells with HIV-1). We show that although U937 and U1 cells demonstrate significantly reduced cytosolic and mitochondrial E(GSH) (approximately -310 mV), active viral replication induces substantial oxidative stress (E(GSH) more than -240 mV). Furthermore, exposure to a physiologically relevant oxidant, hydrogen peroxide (H2O2), induces significant deviations in subcellular E(GSH) between U937 and U1, which distinctly modulates susceptibility to apoptosis. Using Grx1-roGFP2, we demonstrate that a marginal increase of about ∼25 mV in E(GSH) is sufficient to switch HIV-1 from latency to reactivation, raising the possibility of purging HIV-1 by redox modulators without triggering detrimental changes in cellular physiology. Importantly, we show that bioactive lipids synthesized by clinical drug-resistant isolates of Mycobacterium tuberculosis reactivate HIV-1 through modulation of intracellular E(GSH). Finally, the expression analysis of U1 and patient peripheral blood mononuclear cells demonstrated a major recalibration of cellular redox homeostatic pathways during persistence and active replication of HIV.


Subject(s)
Glutathione/metabolism , HIV Infections/metabolism , HIV-1/metabolism , Oxidation-Reduction , Apoptosis/genetics , Glutathione/chemistry , Green Fluorescent Proteins/chemistry , HIV Infections/pathology , HIV-1/isolation & purification , HIV-1/pathogenicity , Humans , Macrophages/virology , Mycobacterium tuberculosis/metabolism , Mycobacterium tuberculosis/pathogenicity , Oxidative Stress/genetics , U937 Cells
7.
Mol Microbiol ; 85(6): 1148-65, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22780904

ABSTRACT

Host-generated oxidative stress is considered one of the main mechanisms constraining Mycobacterium tuberculosis (Mtb) growth. The redox-sensing mechanisms in Mtb are not completely understood. Here we show that WhiB4 responds to oxygen (O2) and nitric oxide (NO) via its 4Fe-4S cluster and controls the oxidative stress response in Mtb. The WhiB4 mutant (MtbΔwhiB4) displayed an altered redox balance and a reduced membrane potential. Microarray analysis demonstrated that MtbΔwhiB4 overexpresses the antioxidant systems including alkyl hydroperoxidase (ahpC-ahpD) and rubredoxins (rubA-rubB). DNA binding assays showed that WhiB4 [4Fe-4S] cluster is dispensable for DNA binding. However, oxidation of the apo-WhiB4 Cys thiols induced disulphide-linked oligomerization, DNA binding and transcriptional repression, whereas reduction reversed the effect. Furthermore, WhiB4 binds DNA with a preference for GC-rich sequences. Expression analysis showed that oxidative stress repressed whiB4 and induced antioxidants in Mtb, while their hyper-induction was observed in MtbΔwhiB4. MtbΔwhiB4 showed increased resistance to oxidative stress in vitro and enhanced survival inside the macrophages. Lastly, MtbΔwhiB4 displayed hypervirulence in the lungs of guinea pigs, but showed a defect in dissemination to their spleen. These findings suggest that WhiB4 systematically calibrates the activation of oxidative stress response in Mtb to maintain redox balance, and to modulate virulence.


Subject(s)
Bacterial Proteins/metabolism , Gene Expression Regulation, Bacterial , Microbial Viability , Mycobacterium tuberculosis/genetics , Mycobacterium tuberculosis/pathogenicity , Oxidative Stress , Stress, Physiological , Animals , Bacterial Proteins/genetics , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Gene Deletion , Gene Expression Profiling , Guinea Pigs , Lung/microbiology , Macrophages/immunology , Macrophages/microbiology , Microarray Analysis , Nitric Oxide/toxicity , Oxidants/toxicity , Oxygen/toxicity , Spleen/microbiology , Virulence , Virulence Factors/genetics , Virulence Factors/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...