Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 866
Filter
1.
PLoS Pathog ; 20(5): e1011821, 2024 May.
Article in English | MEDLINE | ID: mdl-38781120

ABSTRACT

The human immunodeficiency virus (HIV) integrates into the host genome forming latent cellular reservoirs that are an obstacle for cure or remission strategies. Viral transcription is the first step in the control of latency and depends upon the hijacking of the host cell RNA polymerase II (Pol II) machinery by the 5' HIV LTR. Consequently, "block and lock" or "shock and kill" strategies for an HIV cure depend upon a full understanding of HIV transcriptional control. The HIV trans-activating protein, Tat, controls HIV latency as part of a positive feed-forward loop that strongly activates HIV transcription. The recognition of the TATA box and adjacent sequences of HIV essential for Tat trans-activation (TASHET) of the core promoter by host cell pre-initiation complexes of HIV (PICH) has been shown to be necessary for Tat trans-activation, yet the protein composition of PICH has remained obscure. Here, DNA-affinity chromatography was employed to identify the mitotic deacetylase complex (MiDAC) as selectively recognizing TASHET. Using biophysical techniques, we show that the MiDAC subunit DNTTIP1 binds directly to TASHET, in part via its CTGC DNA motifs. Using co-immunoprecipitation assays, we show that DNTTIP1 interacts with MiDAC subunits MIDEAS and HDAC1/2. The Tat-interacting protein, NAT10, is also present in HIV-bound MiDAC. Gene silencing revealed a functional role for DNTTIP1, MIDEAS, and NAT10 in HIV expression in cellulo. Furthermore, point mutations in TASHET that prevent DNTTIP1 binding block the reactivation of HIV by latency reversing agents (LRA) that act via the P-TEFb/7SK axis. Our data reveal a key role for MiDAC subunits DNTTIP1, MIDEAS, as well as NAT10, in Tat-activated HIV transcription and latency. DNTTIP1, MIDEAS and NAT10 emerge as cell cycle-regulated host cell transcription factors that can control activated HIV gene expression, and as new drug targets for HIV cure strategies.


Subject(s)
Gene Expression Regulation, Viral , HIV Infections , HIV-1 , Promoter Regions, Genetic , Virus Latency , Humans , HIV-1/genetics , HIV-1/physiology , HIV Infections/virology , HIV Infections/metabolism , HIV Infections/genetics , tat Gene Products, Human Immunodeficiency Virus/metabolism , tat Gene Products, Human Immunodeficiency Virus/genetics , Viral Transcription
2.
Methods Mol Biol ; 2807: 245-258, 2024.
Article in English | MEDLINE | ID: mdl-38743233

ABSTRACT

The study of host-pathogen interaction often requires interrogating the protein-protein interactions and examining post-translational modifications of the proteins. Traditional protein detection strategies are limited in their sensitivity, specificity, and multiplexing capabilities. The Proximity Ligation Assay (PLA), a versatile and powerful molecular technique, can overcome these limitations. PLA blends the specificity of antibodies, two antibodies detecting two different epitopes on the same or two different proteins, with the amplification efficiency of a polymerase to allow highly specific and sensitive detection of low-abundant proteins, protein-protein interactions, or protein modifications. In this protocol, we describe the application of PLA to detect the proximity between HIV-1 Tat with one of its cellular partners, p65, in an infected host cell. The protocol could be applied to any other context with slight modifications. Of note, PLA can only confirm the physical proximity between two epitopes or proteins; however, the proximity need not necessarily allude to the functional interaction between the two proteins.


Subject(s)
HIV-1 , Host-Pathogen Interactions , Humans , HIV-1/immunology , Protein Interaction Mapping/methods , tat Gene Products, Human Immunodeficiency Virus/metabolism , HIV Infections/virology , Protein Binding
3.
Biochim Biophys Acta Gene Regul Mech ; 1867(2): 195030, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38670485

ABSTRACT

Antiretroviral therapy-naive people living with HIV possess less fat than people without HIV. Previously, we found that HIV-1 transactivator of transcription (TAT) decreases fat in ob/ob mice. The TAT38 (a.a. 20-57) is important in the inhibition of adipogenesis and contains three functional domains: Cys-ZF domain (a.a. 20-35 TACTNCYCAKCCFQVC), core-domain (a.a. 36-46, FITKALGISYG), and protein transduction domain (PTD)(a.a. 47-57, RAKRRQRRR). Interestingly, the TAT38 region interacts with the Cyclin T1 of the P-TEFb complex, of which expression increases during adipogenesis. The X-ray crystallographic structure of the complex showed that the Cys-ZF and the core domain bind to the Cyclin T1 via hydrophobic interactions. To prepare TAT38 mimics with structural and functional similarities to TAT38, we replaced the core domain with a hydrophobic aliphatic amino acid (from carbon numbers 5 to 8). The TAT38 mimics with 6-hexanoic amino acid (TAT38 Ahx (C6)) and 7-heptanoic amino acid (TAT38 Ahp (C7)) inhibited adipogenesis of 3T3-L1 potently, reduced cellular triglyceride content, and decreased body weight of diet-induced obese (DIO) mice by 10.4-11 % in two weeks. The TAT38 and the TAT38 mimics potently repressed the adipogenic transcription factors genes, C/EBPα, PPARγ, and SREBP1. Also, they inhibit the phosphorylation of PPARγ. The TAT peptides may be promising candidates for development into a drug against obesity or diabetes.


Subject(s)
Adipogenesis , PPAR gamma , Sterol Regulatory Element Binding Protein 1 , tat Gene Products, Human Immunodeficiency Virus , Animals , PPAR gamma/metabolism , Adipogenesis/drug effects , Mice , Sterol Regulatory Element Binding Protein 1/metabolism , Sterol Regulatory Element Binding Protein 1/genetics , tat Gene Products, Human Immunodeficiency Virus/metabolism , tat Gene Products, Human Immunodeficiency Virus/genetics , CCAAT-Enhancer-Binding Protein-alpha/metabolism , CCAAT-Enhancer-Binding Protein-alpha/genetics , 3T3-L1 Cells , Humans , Gene Expression Regulation , Mice, Obese , Male , Cyclin T/metabolism , Obesity/metabolism , Adipocytes/metabolism , Mice, Inbred C57BL , CCAAT-Enhancer-Binding Proteins
4.
J Extracell Vesicles ; 13(4): e12439, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38647111

ABSTRACT

Our previous findings demonstrated that astrocytic HIF-1α plays a major role in HIV-1 Tat-mediated amyloidosis which can lead to Alzheimer's-like pathology-a comorbidity of HIV-Associated Neurocognitive Disorders (HAND). These amyloids can be shuttled in extracellular vesicles, and we sought to assess whether HIV-1 Tat stimulated astrocyte-derived EVs (ADEVs) containing the toxic amyloids could result in neuronal injury in vitro and in vivo. We thus hypothesized that blocking HIF-1α could likely mitigate HIV-1 Tat-ADEV-mediated neuronal injury. Rat hippocampal neurons when exposed to HIV-1 Tat-ADEVs carrying the toxic amyloids exhibited amyloid accumulation and synaptodendritic injury, leading to functional loss as evidenced by alterations in miniature excitatory post synaptic currents. The silencing of astrocytic HIF-1α not only reduced the biogenesis of ADEVs, as well as amyloid cargos, but also ameliorated neuronal synaptodegeneration. Next, we determined the effect of HIV-1 Tat-ADEVs carrying amyloids in the hippocampus of naive mice brains. Naive mice receiving the HIV-1 Tat-ADEVs, exhibited behavioural changes, and Alzheimer's 's-like pathology accompanied by synaptodegeneration. This impairment(s) was not observed in mice injected with HIF-1α silenced ADEVs. This is the first report demonstrating the role of amyloid-carrying ADEVs in mediating synaptodegeneration leading to behavioural changes associated with HAND and highlights the protective role of HIF-1α.


Subject(s)
Astrocytes , Extracellular Vesicles , HIV-1 , Hippocampus , Hypoxia-Inducible Factor 1, alpha Subunit , Neurons , Extracellular Vesicles/metabolism , Animals , Astrocytes/metabolism , Mice , Rats , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , HIV-1/metabolism , Hippocampus/metabolism , Neurons/metabolism , tat Gene Products, Human Immunodeficiency Virus/metabolism , Humans , Neurocognitive Disorders/metabolism , Neurocognitive Disorders/etiology , HIV Infections/metabolism , HIV Infections/complications , Male , AIDS Dementia Complex/metabolism
5.
J Neurosci ; 44(21)2024 May 22.
Article in English | MEDLINE | ID: mdl-38664011

ABSTRACT

Fragile X syndrome (FXS) arises from the loss of fragile X messenger ribonucleoprotein (FMRP) needed for normal neuronal excitability and circuit functions. Recent work revealed that FMRP contributes to mossy fiber long-term potentiation by adjusting the Kv4 A-type current availability through interactions with a Cav3-Kv4 ion channel complex, yet the mechanism has not yet been defined. In this study using wild-type and Fmr1 knock-out (KO) tsA-201 cells and cerebellar sections from male Fmr1 KO mice, we show that FMRP associates with all subunits of the Cav3.1-Kv4.3-KChIP3 complex and is critical to enabling calcium-dependent shifts in Kv4.3 inactivation to modulate the A-type current. Specifically, upon depolarization Cav3 calcium influx activates dual-specific phosphatase 1/6 (DUSP1/6) to deactivate ERK1/2 (ERK) and lower phosphorylation of Kv4.3, a signaling pathway that does not function in Fmr1 KO cells. In Fmr1 KO mouse tissue slices, cerebellar granule cells exhibit a hyperexcitable response to membrane depolarizations. Either incubating Fmr1 KO cells or in vivo administration of a tat-conjugated FMRP N-terminus fragment (FMRP-N-tat) rescued Cav3-Kv4 function and granule cell excitability, with a decrease in the level of DUSP6. Together these data reveal a Cav3-activated DUSP signaling pathway critical to the function of a FMRP-Cav3-Kv4 complex that is misregulated in Fmr1 KO conditions. Moreover, FMRP-N-tat restores function of this complex to rescue calcium-dependent control of neuronal excitability as a potential therapeutic approach to alleviating the symptoms of FXS.


Subject(s)
Calcium , Fragile X Mental Retardation Protein , Fragile X Syndrome , Mice, Knockout , Neurons , Animals , Fragile X Mental Retardation Protein/genetics , Fragile X Mental Retardation Protein/metabolism , Mice , Male , Fragile X Syndrome/metabolism , Fragile X Syndrome/genetics , Fragile X Syndrome/physiopathology , Neurons/metabolism , Calcium/metabolism , Mice, Inbred C57BL , Shal Potassium Channels/metabolism , Shal Potassium Channels/genetics , tat Gene Products, Human Immunodeficiency Virus/genetics , tat Gene Products, Human Immunodeficiency Virus/metabolism
6.
Int J Mol Sci ; 25(6)2024 Mar 16.
Article in English | MEDLINE | ID: mdl-38542351

ABSTRACT

Viruses provide vital insights into gene expression control. Viral transactivators, with other viral and cellular proteins, regulate expression of self, other viruses, and host genes with profound effects on infected cells, underlying inflammation, control of immune responses, and pathogenesis. The multifunctional Tat proteins of lentiviruses (HIV-1, HIV-2, and SIV) transactivate gene expression by recruiting host proteins and binding to transacting responsive regions (TARs) in viral and host RNAs. SARS-CoV-2 nucleocapsid participates in early viral transcription, recruits similar cellular proteins, and shares intracellular, surface, and extracellular distribution with Tat. SARS-CoV-2 nucleocapsid interacting with the replication-transcription complex might, therefore, transactivate viral and cellular RNAs in the transcription and reactivation of self and other viruses, acute and chronic pathogenesis, immune evasion, and viral evolution. Here, we show, by using primary and secondary structural comparisons, that the leaders of SARS-CoV-2 and other coronaviruses contain TAR-like sequences in stem-loops 2 and 3. The coronaviral nucleocapsid C-terminal domains harbor a region of similarity to TAR-binding regions of lentiviral Tat proteins, and coronaviral nonstructural protein 12 has a cysteine-rich metal binding, dimerization domain, as do lentiviral Tat proteins. Although SARS-CoV-1 nucleocapsid transactivated gene expression in a replicon-based study, further experimental evidence for coronaviral transactivation and its possible implications is warranted.


Subject(s)
COVID-19 , HIV-1 , Humans , HIV-1/physiology , SARS-CoV-2/genetics , SARS-CoV-2/metabolism , Transcriptional Activation , HIV Long Terminal Repeat , COVID-19/genetics , Gene Products, tat/genetics , Lentivirus/genetics , Gene Expression , tat Gene Products, Human Immunodeficiency Virus/genetics , tat Gene Products, Human Immunodeficiency Virus/metabolism , RNA, Viral/metabolism
7.
J Neurochem ; 168(3): 185-204, 2024 03.
Article in English | MEDLINE | ID: mdl-38308495

ABSTRACT

Despite the advent of combination anti-retroviral therapy (cART), nearly half of people infected with HIV treated with cART still exhibit HIV-associated neurocognitive disorders (HAND). HAND can be worsened by co-morbid opioid use disorder. The basal ganglia are particularly vulnerable to HIV-1 and exhibit higher viral loads and more severe pathology, which can be exacerbated by co-exposure to opioids. Evidence suggests that dopaminergic neurotransmission is disrupted by HIV exposure, however, little is known about whether co-exposure to opioids may alter neurotransmitter levels in the striatum and if this in turn influences behavior. Therefore, we assayed motor, anxiety-like, novelty-seeking, exploratory, and social behaviors, and levels of monoamines and their metabolites following 2 weeks and 2 months of Tat and/or morphine exposure in transgenic mice. Morphine decreased dopamine levels, but significantly elevated norepinephrine, the dopamine metabolites dihydroxyphenylacetic acid (DOPAC) and homovanillic acid (HVA), and the serotonin metabolite 5-hydroxyindoleacetic acid, which typically correlated with increased locomotor behavior. The combination of Tat and morphine altered dopamine, DOPAC, and HVA concentrations differently depending on the neurotransmitter/metabolite and duration of exposure but did not affect the numbers of tyrosine hydroxylase-positive neurons in the mesencephalon. Tat exposure increased the latency to interact with novel conspecifics, but not other novel objects, suggesting the viral protein inhibits exploratory behavior initiation in a context-dependent manner. By contrast, and consistent with prior findings that opioid misuse can increase novelty-seeking behavior, morphine exposure increased the time spent exploring a novel environment. Finally, Tat and morphine interacted to affect locomotor activity in a time-dependent manner, while grip strength and rotarod performance were unaffected. Together, our results provide novel insight into the unique effects of HIV-1 Tat and morphine on monoamine neurochemistry that may underlie their divergent effects on motor and exploratory behavior.


Subject(s)
HIV Infections , HIV-1 , Humans , Mice , Animals , Morphine/pharmacology , Exploratory Behavior , HIV-1/metabolism , Dopamine/metabolism , 3,4-Dihydroxyphenylacetic Acid/metabolism , Mice, Transgenic , Analgesics, Opioid/pharmacology , Homovanillic Acid , Neurotransmitter Agents , tat Gene Products, Human Immunodeficiency Virus/metabolism
8.
Int J Mol Sci ; 25(3)2024 Jan 30.
Article in English | MEDLINE | ID: mdl-38338977

ABSTRACT

Each time the virus starts a new round of expression/replication, even under effective antiretroviral therapy (ART), the transactivator of viral transcription Tat is one of the first HIV-1 protein to be produced, as it is strictly required for HIV replication and spreading. At this stage, most of the Tat protein exits infected cells, accumulates in the extracellular matrix and exerts profound effects on both the virus and neighbor cells, mostly of the innate and adaptive immune systems. Through these effects, extracellular Tat contributes to the acquisition of infection, spreading and progression to AIDS in untreated patients, or to non-AIDS co-morbidities in ART-treated individuals, who experience inflammation and immune activation despite virus suppression. Here, we review the role of extracellular Tat in both the virus life cycle and on cells of the innate and adaptive immune system, and we provide epidemiological and experimental evidence of the importance of targeting Tat to block residual HIV expression and replication. Finally, we briefly review vaccine studies showing that a therapeutic Tat vaccine intensifies ART, while its inclusion in a preventative vaccine may blunt escape from neutralizing antibodies and block early events in HIV acquisition.


Subject(s)
HIV Infections , HIV-1 , Vaccines , Humans , HIV-1/metabolism , tat Gene Products, Human Immunodeficiency Virus/metabolism , Antibodies, Neutralizing , Vaccines/therapeutic use
9.
J Neurovirol ; 30(1): 1-21, 2024 Feb.
Article in English | MEDLINE | ID: mdl-38280928

ABSTRACT

Opioid overdose deaths have dramatically increased by 781% from 1999 to 2021. In the setting of HIV, opioid drug abuse exacerbates neurotoxic effects of HIV in the brain, as opioids enhance viral replication, promote neuronal dysfunction and injury, and dysregulate an already compromised inflammatory response. Despite the rise in fentanyl abuse and the close association between opioid abuse and HIV infection, the interactive comorbidity between fentanyl abuse and HIV has yet to be examined in vivo. The HIV-1 Tat-transgenic mouse model was used to understand the interactive effects between fentanyl and HIV. Tat is an essential protein produced during HIV that drives the transcription of new virions and exerts neurotoxic effects within the brain. The Tat-transgenic mouse model uses a glial fibrillary acidic protein (GFAP)-driven tetracycline promoter which limits Tat production to the brain and this model is well used for examining mechanisms related to neuroHIV. After 7 days of fentanyl exposure, brains were harvested. Tight junction proteins, the vascular cell adhesion molecule, and platelet-derived growth factor receptor-ß were measured to examine the integrity of the blood brain barrier. The immune response was assessed using a mouse-specific multiplex chemokine assay. For the first time in vivo, we demonstrate that fentanyl by itself can severely disrupt the blood-brain barrier and dysregulate the immune response. In addition, we reveal associations between inflammatory markers and tight junction proteins at the blood-brain barrier.


Subject(s)
Blood-Brain Barrier , Fentanyl , HIV-1 , Mice, Transgenic , Neuroinflammatory Diseases , tat Gene Products, Human Immunodeficiency Virus , Animals , Blood-Brain Barrier/drug effects , Blood-Brain Barrier/metabolism , Blood-Brain Barrier/pathology , Blood-Brain Barrier/virology , Mice , Fentanyl/pharmacology , HIV-1/drug effects , HIV-1/genetics , tat Gene Products, Human Immunodeficiency Virus/genetics , tat Gene Products, Human Immunodeficiency Virus/metabolism , Neuroinflammatory Diseases/genetics , Neuroinflammatory Diseases/pathology , Neuroinflammatory Diseases/virology , HIV Infections/virology , HIV Infections/genetics , HIV Infections/pathology , HIV Infections/drug therapy , Disease Models, Animal , Analgesics, Opioid/pharmacology , Analgesics, Opioid/adverse effects , Glial Fibrillary Acidic Protein/genetics , Glial Fibrillary Acidic Protein/metabolism , Tight Junction Proteins/metabolism , Tight Junction Proteins/genetics , Humans , Brain/drug effects , Brain/virology , Brain/metabolism , Brain/pathology , Opioid-Related Disorders/genetics , Opioid-Related Disorders/pathology , Opioid-Related Disorders/metabolism
10.
Int J Mol Sci ; 24(24)2023 Dec 17.
Article in English | MEDLINE | ID: mdl-38139395

ABSTRACT

During the antiretroviral era, individuals living with HIV continue to experience milder forms of HIV-associated neurocognitive disorder (HAND). Viral proteins, including Tat, play a pivotal role in the observed alterations within the central nervous system (CNS), with mitochondrial dysfunction emerging as a prominent hallmark. As a result, our objective was to examine the expression of genes associated with mitophagy and mitochondrial biogenesis in the brain exposed to the HIV-1 Tat protein. We achieved this by performing bilateral stereotaxic injections of 100 ng of HIV-1 Tat into the hippocampus of Sprague-Dawley rats, followed by immunoneuromagnetic cell isolation. Subsequently, we assessed the gene expression of Ppargc1a, Pink1, and Sirt1-3 in neurons using RT-qPCR. Additionally, to understand the role of Tert in telomeric dysfunction, we quantified the activity and expression of Tert. Our results revealed that only Ppargc1a, Pink1, and mitochondrial Sirt3 were downregulated in response to the presence of HIV-1 Tat in hippocampal neurons. Interestingly, we observed a reduction in the activity of Tert in the experimental group, while mRNA levels remained relatively stable. These findings support the compelling evidence of dysregulation in both mitophagy and mitochondrial biogenesis in neurons exposed to HIV-1 Tat, which in turn induces telomeric dysfunction.


Subject(s)
HIV Infections , HIV-1 , Neurocognitive Disorders , Sirtuin 3 , tat Gene Products, Human Immunodeficiency Virus , Animals , Rats , Gene Products, tat/metabolism , HIV Infections/metabolism , HIV-1/metabolism , Neurocognitive Disorders/metabolism , Neurocognitive Disorders/virology , Neurons/metabolism , Organelle Biogenesis , Protein Kinases/metabolism , Rats, Sprague-Dawley , Sirtuin 3/genetics , Sirtuin 3/metabolism , tat Gene Products, Human Immunodeficiency Virus/genetics , tat Gene Products, Human Immunodeficiency Virus/metabolism , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha
11.
Nat Commun ; 14(1): 7274, 2023 11 10.
Article in English | MEDLINE | ID: mdl-37949879

ABSTRACT

The HIV-1 Tat protein hijacks the Super Elongation Complex (SEC) to stimulate viral transcription and replication. However, the mechanisms underlying Tat activation and inactivation, which mediate HIV-1 productive and latent infection, respectively, remain incompletely understood. Here, through a targeted complementary DNA (cDNA) expression screening, we identify PRMT2 as a key suppressor of Tat activation, thus contributing to proviral latency in multiple cell line latency models and in HIV-1-infected patient CD4+ T cells. Our data reveal that the transcriptional activity of Tat is oppositely regulated by NPM1-mediated nucleolar retention and AFF4-induced phase separation in the nucleoplasm. PRMT2 preferentially methylates Tat arginine 52 (R52) to reinforce its nucleolar sequestration while simultaneously counteracting its incorporation into the SEC droplets, thereby leading to its functional inactivation to promote proviral latency. Thus, our studies unveil a central and unappreciated role for Tat methylation by PRMT2 in connecting its subnuclear distribution, liquid droplet formation, and transactivating function, which could be therapeutically targeted to eradicate latent viral reservoirs.


Subject(s)
HIV Infections , HIV-1 , Humans , HIV-1/physiology , Transcriptional Elongation Factors/metabolism , Cell Line , Proviruses/genetics , T-Lymphocytes/metabolism , tat Gene Products, Human Immunodeficiency Virus/genetics , tat Gene Products, Human Immunodeficiency Virus/metabolism , Virus Latency/genetics , HIV Infections/genetics , Protein-Arginine N-Methyltransferases/genetics , Protein-Arginine N-Methyltransferases/metabolism , Intracellular Signaling Peptides and Proteins/metabolism
12.
J Virol ; 97(12): e0187022, 2023 Dec 21.
Article in English | MEDLINE | ID: mdl-37991365

ABSTRACT

IMPORTANCE: Twenty-five years after the first report that HIV-2 infection can reduce HIV-1-associated pathogenesis in dual-infected patients, the mechanisms are still not well understood. We explored these mechanisms in cell culture and showed first that these viruses can co-infect individual cells. Under specific conditions, HIV-2 inhibits HIV-1 through two distinct mechanisms, a broad-spectrum interferon response and an HIV-1-specific inhibition conferred by the HIV-2 TAR. The former could play a prominent role in dually infected individuals, whereas the latter targets HIV-1 promoter activity through competition for HIV-1 Tat binding when the same target cell is dually infected. That mechanism suppresses HIV-1 transcription by stalling RNA polymerase II complexes at the promoter through a minimal inhibitory region within the HIV-2 TAR. This work delineates the sequence of appearance and the modus operandi of each mechanism.


Subject(s)
Coinfection , Gene Expression Regulation, Viral , HIV Long Terminal Repeat , HIV-1 , HIV-2 , Interferons , RNA, Viral , tat Gene Products, Human Immunodeficiency Virus , Humans , Coinfection/immunology , Coinfection/virology , HIV Long Terminal Repeat/genetics , HIV-1/genetics , HIV-1/immunology , HIV-2/genetics , HIV-2/immunology , HIV-2/metabolism , RNA, Viral/genetics , tat Gene Products, Human Immunodeficiency Virus/metabolism , Interferons/immunology , Promoter Regions, Genetic/genetics , Binding, Competitive , RNA Polymerase II/metabolism , Transcription, Genetic
13.
STAR Protoc ; 4(4): 102687, 2023 Dec 15.
Article in English | MEDLINE | ID: mdl-37979180

ABSTRACT

A critical virus-encoded regulator of HIV-1 transcription is the Tat protein, which is required to potently activate transcription. Tat is regulated by a wide variety of post-translational modifications. This protocol describes an in vitro assay to study Tat methylation. We describe steps for incorporation of radioactive methyl groups into Tat protein, visualization by gel analysis, Coomassie blue stain, gel drying, and detection by autoradiography. This protocol can also be used to assess methylation in other proteins such as histones. For complete details on the use and execution of this protocol, please refer to Boehm et al. (2023).1.


Subject(s)
HIV-1 , HIV-1/metabolism , tat Gene Products, Human Immunodeficiency Virus/genetics , tat Gene Products, Human Immunodeficiency Virus/metabolism , Methylation , Protein Processing, Post-Translational , Histones/metabolism
14.
PLoS One ; 18(10): e0292681, 2023.
Article in English | MEDLINE | ID: mdl-37819924

ABSTRACT

The peptide domain extending from residues 49 to 57 of the HIV-1 Tat protein (TAT) has been widely shown to facilitate cell entry of and blood-brain barrier (BBB) permeability to covalently bound macromolecules; therefore, TAT-linked therapeutic peptides trafficked through peripheral routes have been used to treat brain diseases in preclinical and clinical studies. Although the mechanisms underlying cell entry by similar peptides have been established to be temperature-dependent and cell-type specific and to involve receptor-mediated endocytosis, how these peptides cross the BBB remains unclear. Here, using an in vitro model, we studied the permeability of TAT, which was covalently bound to the fluorescent probe fluorescein isothiocyanate (FITC), and evaluated whether it crossed the "in vitro BBB", a monolayer of brain endothelial cells, and whether the mechanisms were similar to those involved in TAT entry into cells. Our results show that although TAT crossed the monolayer of brain endothelial cells in a temperature-dependent manner, in contrast to the reported mechanism of cell entry, it did not require receptor-mediated endocytosis. Furthermore, we revisited the hypothesis that TAT facilitates brain delivery of covalently bound macromolecules by causing BBB disruption. Our results demonstrated that the dose of TAT commonly used in preclinical and clinical studies did not exert an effect on BBB permeability in vitro or in vivo; however, an extremely high TAT concentration caused BBB disruption in vitro. In conclusion, the BBB permeability to TAT is temperature-dependent, but at treatment-level concentrations, it does not involve receptor-mediated endocytosis or BBB disruption.


Subject(s)
Endothelial Cells , tat Gene Products, Human Immunodeficiency Virus , Endothelial Cells/metabolism , tat Gene Products, Human Immunodeficiency Virus/metabolism , Brain/metabolism , Blood-Brain Barrier/metabolism , Peptides/metabolism , Endocytosis
15.
J Virol ; 97(11): e0104423, 2023 Nov 30.
Article in English | MEDLINE | ID: mdl-37905837

ABSTRACT

IMPORTANCE: HIV-infected host cells impose varied degrees of regulation on viral replication, from very high to abortive. Proliferation of HIV in astrocytes is limited when compared to immune cells, such as CD4+ T lymphocytes. Understanding such differential regulation is one of the key questions in the field as these cells permit HIV persistence and rebound viremia, challenging HIV treatment and clinical cure. This study focuses on understanding the molecular mechanism behind such cell-specific disparities. We show that one of the key mechanisms is the regulation of heterogenous nuclear ribonucleoprotein A2, a host factor involved in alternative splicing and RNA processing, by HIV-1 Tat in CD4+ T lymphocytes, not observed in astrocytes. This regulation causes an increase in the levels of unspliced/partially spliced viral RNA and nuclear export of Rev-RNA complexes which results in high viral propagation in CD4+ T lymphocytes. The study reveals a new mechanism imposed by HIV on host cells that determines the fate of infection.


Subject(s)
Active Transport, Cell Nucleus , HIV Infections , HIV-1 , Heterogeneous-Nuclear Ribonucleoprotein Group A-B , tat Gene Products, Human Immunodeficiency Virus , Humans , Alternative Splicing , Cell Nucleus/metabolism , Gene Products, rev/genetics , HIV-1/physiology , rev Gene Products, Human Immunodeficiency Virus/genetics , rev Gene Products, Human Immunodeficiency Virus/metabolism , RNA Splicing , RNA, Viral/genetics , RNA, Viral/metabolism , tat Gene Products, Human Immunodeficiency Virus/genetics , tat Gene Products, Human Immunodeficiency Virus/metabolism , Heterogeneous-Nuclear Ribonucleoprotein Group A-B/metabolism
16.
Antimicrob Agents Chemother ; 67(11): e0041723, 2023 11 15.
Article in English | MEDLINE | ID: mdl-37874295

ABSTRACT

A major barrier to HIV-1 cure is caused by the pool of latently infected CD4 T-cells that persist under combination antiretroviral therapy (cART). This latent reservoir is capable of producing replication-competent infectious viruses once prolonged suppressive cART is withdrawn. Inducing the reactivation of HIV-1 gene expression in T-cells harboring a latent provirus in people living with HIV-1 under cART may result in depletion of this latent reservoir due to cytopathic effects or immune clearance. Studies have investigated molecules that reactivate HIV-1 gene expression, but to date, no latency reversal agent has been identified to eliminate latently infected cells harboring replication-competent HIV in cART-treated individuals. Stochastic fluctuations in HIV-1 tat gene expression have been described and hypothesized to allow the progression into proviral latency. We hypothesized that exposing latently infected CD4+ T-cells to Tat would result in effective latency reversal. Our results indicate the capacity of a truncated Tat protein and mRNA to reactivate HIV-1 in latently infected T-cells ex vivo to a similar degree as the protein kinase C agonist: phorbol 12-myristate 13-acetate, without T-cell activation or any significant transcriptome perturbation.


Subject(s)
HIV Infections , HIV-1 , Virus Activation , tat Gene Products, Human Immunodeficiency Virus , Humans , CD4-Positive T-Lymphocytes , HIV Infections/genetics , HIV Infections/metabolism , Proviruses/genetics , Virus Latency , Virus Replication , tat Gene Products, Human Immunodeficiency Virus/genetics , tat Gene Products, Human Immunodeficiency Virus/metabolism , HIV-1/genetics , HIV-1/metabolism
17.
J Neurovirol ; 29(6): 658-668, 2023 12.
Article in English | MEDLINE | ID: mdl-37899420

ABSTRACT

Although the widespread use of antiretroviral therapy (ART) has prolonged the life span of people living with HIV (PLWH), the incidence of HIV-associated neurocognitive disorders (HAND) in PLWH is also gradually increasing, seriously affecting the quality of life for PLWH. However, the pathogenesis of HAND has not been elucidated, which leaves HAND without effective treatment. HIV protein transactivator of transcription (Tat), as an important regulatory protein, is crucial in the pathogenesis of HAND, and its mechanism of HAND has received widespread attention. The blood-brain barrier (BBB) and its cellular component brain microvascular endothelial cells (BMVECs) play a necessary role in protecting the central nervous system (CNS), and their damage associated with Tat is a potential therapeutic target of HAND. In this review, we will study the Tat-mediated damage mechanism of the BBB and present multiple lines of evidence related to BMVEC damage caused by Tat.


Subject(s)
HIV Infections , HIV-1 , Humans , Blood-Brain Barrier/metabolism , Endothelial Cells/metabolism , Tight Junction Proteins/genetics , Tight Junction Proteins/metabolism , Trans-Activators/metabolism , Quality of Life , tat Gene Products, Human Immunodeficiency Virus/metabolism , HIV-1/metabolism , Brain/metabolism , HIV Infections/pathology
18.
Biomolecules ; 13(6)2023 05 24.
Article in English | MEDLINE | ID: mdl-37371461

ABSTRACT

HIV-1 infection leads to a gradual loss of T helper cells, chronic immune activation, and eventual immune system breakdown. HIV-1 causes deregulation of the expression of IL-2, a cytokine important for T helper cell growth and survival, which is downregulated in HIV-1 patients. The present study addresses the regulation of IL2 expression via HIV-1 Tat transcriptional activator. We used J-LAT cells, a T cell line that serves as a latency model for studies of HIV-1 expression in T cells, and as controls a T cell line lacking HIV-1 elements and a T cell line with a stably integrated copy of the HIV-1-LTR promoter. We show that endogenously expressed Tat inhibits IL2 transcription in J-Lat cells via its presence in the ARRE-1/2 elements of the IL2 promoter and that the inhibition of IL2 expression is mediated by Tat inhibiting Pol II activity at the IL2 promoter, which is mediated by preventing the presence of Pol II at the ARRE-1/2 elements. Overall, Tat is present at the IL2 promoter, apart from its cognate HIV-1 LTR target. This supports our current knowledge of how HIV-1 affects the host transcriptional machinery and reflects the potential of Tat to disrupt transcriptional regulation of host genes to manipulate cell responses.


Subject(s)
HIV Infections , HIV-1 , Interleukin-2 , RNA Polymerase II , tat Gene Products, Human Immunodeficiency Virus , Humans , Gene Expression Regulation, Viral , HIV Infections/genetics , HIV Infections/immunology , HIV Infections/virology , HIV-1/genetics , HIV-1/metabolism , Interleukin-2/genetics , Interleukin-2/metabolism , RNA Polymerase II/genetics , RNA Polymerase II/metabolism , tat Gene Products, Human Immunodeficiency Virus/genetics , tat Gene Products, Human Immunodeficiency Virus/metabolism , Transcription Factors/metabolism , Transcription, Genetic , Transcriptional Activation
19.
J Neurovirol ; 29(4): 479-491, 2023 08.
Article in English | MEDLINE | ID: mdl-37358698

ABSTRACT

NOTCH receptors are relevant to multiple neurodegenerative diseases. However, the roles and mechanisms of NOTCH receptors in HIV-associated neurocognitive disorder (HAND) remain largely unclear. Transactivator of transcription (Tat) induces oxidative stress and inflammatory response in astrocytes, thereby leading to neuronal apoptosis in the central nervous system. We determined that NOTCH3 expression was upregulated during subtype B or C Tat expression in HEB astroglial cells. Moreover, bioinformatics analysis of the Gene Expression Omnibus (GEO) dataset revealed that NOTCH3 mRNA expression in the frontal cortex tissues of HIV encephalitis patients was higher than that of HIV control patients. Of note, subtype B Tat, rather than subtype C Tat, interacted with the extracellular domain of the NOTCH3 receptor, thus activating NOTCH3 signaling. Downregulation of NOTCH3 attenuated subtype B Tat-induced oxidative stress and reactive oxygen species generation. In addition, we demonstrated that NOTCH3 signaling facilitated subtype B Tat-activated NF-κB signaling pathway, thereby mediating pro-inflammatory cytokines IL-6 and TNF-α production. Furthermore, downregulation of NOTCH3 in HEB astroglial cells protected SH-SY5Y neuronal cells from astrocyte-mediated subtype B Tat neurotoxicity. Taken together, our study clarifies the potential role of NOTCH3 in subtype B Tat-induced oxidative stress and inflammatory response in astrocytes, which could be a novel therapeutic target for the relief of HAND.


Subject(s)
HIV Infections , HIV-1 , Neuroblastoma , Humans , Astrocytes/metabolism , HIV-1/genetics , HIV-1/metabolism , Trans-Activators/metabolism , Receptor, Notch3/genetics , Receptor, Notch3/metabolism , tat Gene Products, Human Immunodeficiency Virus/genetics , tat Gene Products, Human Immunodeficiency Virus/metabolism , Neuroblastoma/metabolism , Signal Transduction , NF-kappa B/genetics , NF-kappa B/metabolism , HIV Infections/genetics , HIV Infections/metabolism , Oxidative Stress , Apoptosis , Basic Helix-Loop-Helix Transcription Factors/metabolism
20.
ACS Nano ; 17(13): 12247-12265, 2023 07 11.
Article in English | MEDLINE | ID: mdl-37350353

ABSTRACT

Cells penetrating molecules in living systems hold promise of capturing and eliminating threats and damage that can plan intracellular fate promptly. However, it remains challenging to construct cell penetration systems that are physiologically stable with predictable self-assembly behavior and well-defined mechanisms. In this study, we develop a core-shell nanoparticle using a hyaluronic acid (HA)-coated protein transduction domain (PTD) derived from the human immunodeficiency virus (HIV). This nanoparticle can encapsulate pathogens, transporting the PTD into macrophages via lipid rafts. PTD forms hydrogen bonds with the components of the membrane through TAT, which has a high density of positive charges and reduces the degree of membrane order through Tryptophan (Trp)-zipper binding to the acyl tails of phospholipid molecules. HA-encapsulated PTD increases the resistance to trypsin and proteinase K, thereby penetrating macrophages and eliminating intracellular infections. Interestingly, the nonagglutination mechanism of PTD against pathogens ensures the safe operation of the cellular system. Importantly, PTD can activate the critical pathway of antiferroptosis in macrophages against pathogen infection. The nanoparticles developed in this study demonstrate safety and efficacy against Gram-negative and Gram-positive pathogens in three animal models. Overall, this work highlights the effectiveness of the PTD nanoparticle in encapsulating pathogens and provides a paradigm for transduction systems-anti-intracellular infection therapy.


Subject(s)
Ferroptosis , tat Gene Products, Human Immunodeficiency Virus , Animals , Humans , tat Gene Products, Human Immunodeficiency Virus/chemistry , tat Gene Products, Human Immunodeficiency Virus/genetics , tat Gene Products, Human Immunodeficiency Virus/metabolism , Tryptophan , Biological Transport , Macrophages/metabolism , Transduction, Genetic , Recombinant Fusion Proteins/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...