Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 205
Filter
1.
Biomolecules ; 11(10)2021 09 26.
Article in English | MEDLINE | ID: mdl-34680042

ABSTRACT

Green fluorescent protein (GFP) chromophore and its congeners draw significant attention mostly for bioimaging purposes. In this work we probed these compounds as antiviral agents. We have chosen LTR-III DNA G4, the major G-quadruplex (G4) present in the long terminal repeat (LTR) promoter region of the human immunodeficiency virus-1 (HIV-1), as the target for primary screening and designing antiviral drug candidates. The stabilization of this G4 was previously shown to suppress viral gene expression and replication. FRET-based high-throughput screening (HTS) of 449 GFP chromophore-like compounds revealed a number of hits, sharing some general structural features. Structure-activity relationships (SAR) for the most effective stabilizers allowed us to establish structural fragments, important for G4 binding. Synthetic compounds, developed on the basis of SAR analysis, exhibited high LTR-III G4 stabilization level. NMR spectroscopy and molecular modeling revealed the possible formation of LTR-III G4-ligand complex with one of the lead selective derivative ZS260.1 positioned within the cavity, thus supporting the LTR-III G4 attractiveness for drug targeting. Selected compounds showed moderate activity against HIV-I (EC50 1.78-7.7 µM) in vitro, but the activity was accompanied by pronounced cytotoxicity.


Subject(s)
G-Quadruplexes , Green Fluorescent Proteins/chemistry , HIV Infections/drug therapy , HIV-1/drug effects , Anti-HIV Agents/chemistry , Green Fluorescent Proteins/pharmacology , HIV Infections/virology , HIV Long Terminal Repeat/drug effects , HIV Long Terminal Repeat/genetics , HIV-1/genetics , HIV-1/pathogenicity , Humans , Magnetic Resonance Spectroscopy , Models, Molecular , Structure-Activity Relationship
2.
Viruses ; 13(10)2021 10 06.
Article in English | MEDLINE | ID: mdl-34696435

ABSTRACT

The HIV-1 Tat protein interacts with TAR RNA and recruits CDK9/cyclin T1 and other host factors to induce HIV-1 transcription. Thus, Tat-TAR RNA interaction, which is unique for HIV-1, represents an attractive target for anti-HIV-1 therapeutics. To target Tat-TAR RNA interaction, we used a crystal structure of acetylpromazine bound to the bulge of TAR RNA, to dock compounds from the Enamine database containing over two million individual compounds. The docking procedure identified 173 compounds that were further analyzed for the inhibition of HIV-1 infection. The top ten inhibitory compounds with IC50 ≤ 6 µM were selected and the three least toxic compounds, T6780107 (IC50 = 2.97 µM), T0516-4834 (IC50 = 0.2 µM) and T5628834 (IC50 = 3.46 µM), were further tested for HIV-1 transcription inhibition. Only the T0516-4834 compound showed selective inhibition of Tat-induced HIV-1 transcription, whereas the T6780107 compound inhibited equally basal and Tat-induced transcription and the T5628834 compound only inhibited basal HIV-1 transcription. The compounds were tested for the inhibition of translation and showed minimal (<25%) effect. The T0516-4834 compound also showed the strongest inhibition of HIV-1 RNA expression and p24 production in CEM T cells and peripheral blood mononuclear cells infected with HIV-1 IIIB. Of the three compounds, only the T0516-4834 compound significantly disrupted Tat-TAR RNA interaction. Additionally, of the three tested compounds, T5628834 and, to a lesser extent, T0516-4834 disrupted Tat-CDK9/cyclin T1 interaction. None of the three compounds showed significant inhibition of the cellular CDK9 and cyclin T1 levels. In silico modelling showed that the T0516-4834 compound interacted with TAR RNA by binding to the bulge formed by U23, U25, C39, G26,C39 and U40 residues. Taken together, our study identified a novel benzoxazole compound that disrupted Tat-TAR RNA interaction and inhibited Tat-induced transcription and HIV-1 infection, suggesting that this compound might serve as a new lead for anti-HIV-1 therapeutics.


Subject(s)
HIV Infections/prevention & control , HIV Long Terminal Repeat/genetics , tat Gene Products, Human Immunodeficiency Virus/genetics , Cyclin T/metabolism , Cyclin-Dependent Kinase 9/metabolism , Gene Expression/genetics , Gene Expression Regulation, Viral/genetics , HEK293 Cells , HIV Infections/genetics , HIV Long Terminal Repeat/drug effects , HIV Long Terminal Repeat/physiology , HIV-1/genetics , HIV-1/metabolism , HIV-1/pathogenicity , Humans , Leukocytes, Mononuclear/metabolism , Molecular Docking Simulation , Phosphorylation , Protein Binding/drug effects , RNA, Viral/genetics , Small Molecule Libraries/pharmacology , Virus Replication/drug effects , tat Gene Products, Human Immunodeficiency Virus/drug effects , tat Gene Products, Human Immunodeficiency Virus/metabolism
3.
RNA ; 27(1): 12-26, 2021 01.
Article in English | MEDLINE | ID: mdl-33028652

ABSTRACT

Identifying small molecules that selectively bind an RNA target while discriminating against all other cellular RNAs is an important challenge in RNA-targeted drug discovery. Much effort has been directed toward identifying drug-like small molecules that minimize electrostatic and stacking interactions that lead to nonspecific binding of aminoglycosides and intercalators to many stem-loop RNAs. Many such compounds have been reported to bind RNAs and inhibit their cellular activities. However, target engagement and cellular selectivity assays are not routinely performed, and it is often unclear whether functional activity directly results from specific binding to the target RNA. Here, we examined the propensities of three drug-like compounds, previously shown to bind and inhibit the cellular activities of distinct stem-loop RNAs, to bind and inhibit the cellular activities of two unrelated HIV-1 stem-loop RNAs: the transactivation response element (TAR) and the rev response element stem IIB (RREIIB). All compounds bound TAR and RREIIB in vitro, and two inhibited TAR-dependent transactivation and RRE-dependent viral export in cell-based assays while also exhibiting off-target interactions consistent with nonspecific activity. A survey of X-ray and NMR structures of RNA-small molecule complexes revealed that aminoglycosides and drug-like molecules form hydrogen bonds with functional groups commonly accessible in canonical stem-loop RNA motifs, in contrast to ligands that specifically bind riboswitches. Our results demonstrate that drug-like molecules can nonspecifically bind stem-loop RNAs most likely through hydrogen bonding and electrostatic interactions and reinforce the importance of assaying for off-target interactions and RNA selectivity in vitro and in cells when assessing novel RNA-binders.


Subject(s)
Aminoglycosides/pharmacology , Genes, env/drug effects , HIV Long Terminal Repeat/drug effects , RNA, Viral/antagonists & inhibitors , Small Molecule Libraries/pharmacology , Aminoglycosides/chemistry , Aminoglycosides/metabolism , Base Pairing , Base Sequence , Binding Sites , Biological Assay , Drug Discovery , HIV-1/drug effects , HIV-1/genetics , HIV-1/metabolism , Humans , Hydrogen Bonding , Isoquinolines/chemistry , Isoquinolines/metabolism , Isoquinolines/pharmacology , Nucleic Acid Conformation , Pentamidine/chemistry , Pentamidine/metabolism , Pentamidine/pharmacology , RNA, Viral/genetics , RNA, Viral/metabolism , Small Molecule Libraries/chemistry , Small Molecule Libraries/metabolism , Static Electricity , Transcriptional Activation/drug effects , Yohimbine/chemistry , Yohimbine/metabolism , Yohimbine/pharmacology
4.
Sci Rep ; 10(1): 13271, 2020 08 06.
Article in English | MEDLINE | ID: mdl-32764708

ABSTRACT

Human immunodeficiency virus 1 (HIV-1) is a life-threatening pathogen that still lacks a curative therapy or vaccine. Despite the reduction in AIDS-related deaths achieved by current antiretroviral therapies, drawbacks including drug resistance and the failure to eradicate infection highlight the need to identify new pathways to target the infection. Circadian rhythms are endogenous 24-h oscillations which regulate physiological processes including immune responses to infection, and there is an emerging role for the circadian components in regulating viral replication. The molecular clock consists of transcriptional/translational feedback loops that generate rhythms. In mammals, BMAL1 and CLOCK activate rhythmic transcription of genes including the nuclear receptor REV-ERBα, which represses BMAL1 and plays an essential role in sustaining a functional clock. We investigated whether REV-ERB activity regulates HIV-1 replication and found REV-ERB agonists inhibited HIV-1 promoter activity in cell lines, primary human CD4 T cells and macrophages, whilst antagonism or genetic disruption of REV-ERB increased promoter activity. The REV-ERB agonist SR9009 inhibited promoter activity of diverse HIV-subtypes and HIV-1 replication in primary T cells. This study shows a role for REV-ERB synthetic agonists to inhibit HIV-1 LTR promoter activity and viral replication, supporting a role for circadian clock components in regulating HIV-1 replication.


Subject(s)
Antiviral Agents/pharmacology , HIV Long Terminal Repeat/drug effects , HIV-1/physiology , Pyrrolidines/pharmacology , Thiophenes/pharmacology , CD4-Positive T-Lymphocytes/cytology , CD4-Positive T-Lymphocytes/drug effects , CD4-Positive T-Lymphocytes/metabolism , CD4-Positive T-Lymphocytes/virology , Cell Line , Circadian Clocks/drug effects , HIV-1/drug effects , Humans , Jurkat Cells , Macrophages/cytology , Macrophages/drug effects , Macrophages/metabolism , Macrophages/virology , Promoter Regions, Genetic/drug effects , Receptors, Thyroid Hormone/metabolism , Virus Replication/drug effects , rev Gene Products, Human Immunodeficiency Virus/metabolism
5.
Methods Enzymol ; 623: 339-372, 2019.
Article in English | MEDLINE | ID: mdl-31239053

ABSTRACT

RNA structures play a pivotal role in many biological processes and the progression of human disease, making them an attractive target for therapeutic development. Often RNA structures operate through the formation of complexes with RNA-binding proteins, however, much like protein-protein interactions, RNA-protein interactions span large surface areas and often lack traditional druggable properties, making it challenging to target them with small molecules. Peptides provide much greater surface areas and therefore greater potential for forming specific and high affinity interactions with RNA. In this chapter, we discuss our approach for engineering peptides that bind to structured RNAs by highlighting methods and design strategies from previous successful projects aimed at inhibiting the HIV Tat-TAR interaction and the biogenesis of oncogenic microRNAs.


Subject(s)
Drug Design , Macrocyclic Compounds/pharmacology , Peptides/pharmacology , Peptidomimetics/pharmacology , RNA/metabolism , Animals , Drug Discovery/methods , HIV Infections/drug therapy , HIV Long Terminal Repeat/drug effects , HIV-1/drug effects , Humans , Macrocyclic Compounds/chemistry , Models, Molecular , Nuclear Magnetic Resonance, Biomolecular/methods , Peptides/chemistry , Peptidomimetics/chemistry , RNA/chemistry
6.
J Biol Chem ; 294(24): 9326-9341, 2019 06 14.
Article in English | MEDLINE | ID: mdl-31080171

ABSTRACT

Small molecules and short peptides that potently and selectively bind RNA are rare, making the molecular structures of these complexes highly exceptional. Accordingly, several recent investigations have provided unprecedented structural insights into how peptides and proteins recognize the HIV-1 transactivation response (TAR) element, a 59-nucleotide-long, noncoding RNA segment in the 5' long terminal repeat region of viral transcripts. Here, we offer an integrated perspective on these advances by describing earlier progress on TAR binding to small molecules, and by drawing parallels to recent successes in the identification of compounds that target the hepatitis C virus internal ribosome entry site (IRES) and the flavin-mononucleotide riboswitch. We relate this work to recent progress that pinpoints specific determinants of TAR recognition by: (i) viral Tat proteins, (ii) an innovative lab-evolved TAR-binding protein, and (iii) an ultrahigh-affinity cyclic peptide. New structural details are used to model the TAR-Tat-super-elongation complex (SEC) that is essential for efficient viral transcription and represents a focal point for antiviral drug design. A key prediction is that the Tat transactivation domain makes modest contacts with the TAR apical loop, whereas its arginine-rich motif spans the entire length of the TAR major groove. This expansive interface has significant implications for drug discovery and design, and it further suggests that future lab-evolved proteins could be deployed to discover steric restriction points that block Tat-mediated recruitment of the host SEC to HIV-1 TAR.


Subject(s)
Antiviral Agents/chemistry , Drug Discovery , HIV Infections/genetics , HIV Long Terminal Repeat/genetics , HIV-1/genetics , RNA, Viral/genetics , tat Gene Products, Human Immunodeficiency Virus/metabolism , Antiviral Agents/pharmacology , HIV Infections/drug therapy , HIV Infections/virology , HIV Long Terminal Repeat/drug effects , HIV-1/drug effects , HIV-1/pathogenicity , Humans , Transcription, Genetic , tat Gene Products, Human Immunodeficiency Virus/genetics
7.
Nucleic Acids Res ; 47(3): 1523-1531, 2019 02 20.
Article in English | MEDLINE | ID: mdl-30481318

ABSTRACT

The HIV-1 trans-activator protein Tat binds the trans-activation response element (TAR) to facilitate recruitment of the super elongation complex (SEC) to enhance transcription of the integrated pro-viral genome. The Tat-TAR interaction is critical for viral replication and the emergence of the virus from the latent state, therefore, inhibiting this interaction has long been pursued to discover new anti-viral or latency reversal agents. However, discovering active compounds that directly target RNA with high affinity and selectivity remains a significant challenge; limiting pre-clinical development. Here, we report the rational design of a macrocyclic peptide mimic of the arginine rich motif of Tat, which binds to TAR with low pM affinity and 100-fold selectivity against closely homologous RNAs. Despite these unprecedented binding properties, the new ligand (JB181) only moderately inhibits Tat-dependent reactivation in cells and recruitment of positive transcription elongation factor (P-TEFb) to TAR. The NMR structure of the JB181-TAR complex revealed that the ligand induces a structure in the TAR loop that closely mimics the P-TEFb/Tat1:57/AFF4/TAR complex. These results strongly suggest that high-affinity ligands which bind the UCU bulge are not likely to inhibit recruitment of the SEC and suggest that targeting of the TAR loop will be an essential feature of effective Tat inhibitors.


Subject(s)
HIV Infections/genetics , HIV Long Terminal Repeat/genetics , HIV-1/genetics , tat Gene Products, Human Immunodeficiency Virus/genetics , Antiviral Agents/chemistry , Antiviral Agents/pharmacology , HIV Infections/drug therapy , HIV Infections/virology , HIV Long Terminal Repeat/drug effects , HIV-1/drug effects , HIV-1/pathogenicity , Humans , Ligands , Multiprotein Complexes/drug effects , Multiprotein Complexes/genetics , Positive Transcriptional Elongation Factor B/chemistry , Positive Transcriptional Elongation Factor B/genetics , Protein Binding , RNA, Viral/genetics , Transcription, Genetic/drug effects , tat Gene Products, Human Immunodeficiency Virus/chemistry
8.
PLoS One ; 13(11): e0206700, 2018.
Article in English | MEDLINE | ID: mdl-30408070

ABSTRACT

Clinical trials are necessary in order to develop treatments for diseases; however, they can often be costly, time consuming, and demanding to the patients. This paper summarizes several common methods used for optimal design that can be used to address these issues. In addition, we introduce a novel method for optimizing experiment designs applied to HIV 2-LTR clinical trials. Our method employs Bayesian techniques to optimize the experiment outcome by maximizing the Expected Kullback-Leibler Divergence (EKLD) between the a priori knowledge of system parameters before the experiment and the a posteriori knowledge of the system parameters after the experiment. We show that our method is robust and performs equally well if not better than traditional optimal experiment design techniques.


Subject(s)
HIV Long Terminal Repeat/drug effects , HIV Long Terminal Repeat/genetics , HIV/drug effects , HIV/genetics , Algorithms , Bayes Theorem , Clinical Trials as Topic/methods , Clinical Trials as Topic/statistics & numerical data , Computer Simulation , HIV/physiology , HIV Infections/therapy , HIV Infections/virology , HIV Long Terminal Repeat/physiology , Humans , Immunotherapy, Adoptive , Markov Chains , Models, Biological , Models, Statistical , Monte Carlo Method , RNA, Viral/biosynthesis , RNA, Viral/genetics , Research Design , Virus Replication/drug effects , Virus Replication/genetics
9.
Sci Rep ; 8(1): 14702, 2018 10 02.
Article in English | MEDLINE | ID: mdl-30279437

ABSTRACT

Persistence of latent HIV-1 in macrophages (MACs) and T-helper lymphocytes (THLs) remain a major therapeutic challenge. Currently available latency reversing agents (LRAs) are not very effective in vivo. Therefore, understanding of physiologic mechanisms that dictate HIV-1 latency/reactivation in reservoirs is clearly needed. Mesenchymal stromal/stem cells (MSCs) regulate the function of immune cells; however, their role in regulating virus production from latently-infected MACs & THLs is not known. We documented that exposure to MSCs or their conditioned media (MSC-CM) rapidly increased HIV-1 p24 production from the latently-infected U1 (MAC) & ACH2 (THL) cell lines. Exposure to MSCs also increased HIV-1 long terminal repeat (LTR) directed gene expression in the MAC and THL reporter lines, U937-VRX and J-Lat (9.2), respectively. MSCs exposed to CM from U1 cells (U1-CM) showed enhanced migratory ability towards latently-infected cells and retained their latency-reactivation potential. Molecular studies showed that MSC-mediated latency-reactivation was dependent upon both the phosphatidyl inositol-3-kinase (PI3K) and nuclear factor-κB (NFκB) signaling pathways. The pre-clinically tested inhibitors of PI3K (PX-866) and NFκB (CDDO-Me) suppressed MSC-mediated HIV-1 reactivation. Furthermore, coexposure to MSC-CM enhanced the latency-reactivation efficacy of the approved LRAs, vorinostat and panobinostat. Our findings on MSC-mediated latency-reactivation may provide novel strategies against persistent HIV-1 reservoirs.


Subject(s)
Anti-HIV Agents/pharmacology , HIV-1/physiology , Mesenchymal Stem Cells/metabolism , Virus Activation/drug effects , Anti-HIV Agents/therapeutic use , Cell Line , Culture Media, Conditioned/pharmacology , Drug Evaluation, Preclinical , Gene Expression Regulation, Viral/drug effects , Gonanes/pharmacology , HIV Infections/virology , HIV Long Terminal Repeat/drug effects , HIV-1/drug effects , Humans , Mesenchymal Stem Cells/drug effects , NF-kappa B/metabolism , Oleanolic Acid/analogs & derivatives , Oleanolic Acid/pharmacology , Panobinostat/pharmacology , Panobinostat/therapeutic use , Phosphatidylinositol 3-Kinases/metabolism , Signal Transduction/drug effects , Virus Latency/drug effects , Vorinostat/pharmacology , Vorinostat/therapeutic use
10.
Biosci Rep ; 38(5)2018 10 31.
Article in English | MEDLINE | ID: mdl-30068696

ABSTRACT

Lentiviral vectors have emerged as the most efficient system to stably transfer and insert genes into cells. By adding a tetracycline (Tet)-inducible promoter, transgene expression delivered by a lentiviral vector can be expressed whenever needed and halted when necessary. Here we have constructed a doxycycline (Dox)-inducible lentiviral vector which efficiently introduces a designed zinc finger protein, 2-long terminal repeat zinc-finger protein (2LTRZFP), into hematopoietic cell lines and evaluated its expression in pluripotent stem cells. As a result this lentiviral inducible system can regulate 2LTRZFP expression in the SupT1 T-cell line and in pluripotent stem cells. Using this vector, no basal expression was detected in the T-cell line and its induction was achieved with low Dox concentrations. Remarkably, the intracellular regulatory expression of 2LTRZFP significantly inhibited HIV-1 integration and replication in HIV-inoculated SupT1 cells. This approach could provide a potential tool for gene therapy applications, which efficiently control and reduce the side effect of therapeutic genes expression.


Subject(s)
Genetic Therapy/methods , Genetic Vectors , HIV Long Terminal Repeat/genetics , HIV-1/genetics , Virus Integration/physiology , Dose-Response Relationship, Drug , Doxycycline/administration & dosage , Doxycycline/pharmacology , Gene Expression Regulation/drug effects , HEK293 Cells , HIV Infections/genetics , HIV Long Terminal Repeat/drug effects , HIV-1/pathogenicity , Humans , Lentivirus/genetics , Pluripotent Stem Cells/virology , Tetracycline/pharmacology , Transgenes , Virus Integration/drug effects , Virus Integration/genetics , Zinc Fingers
11.
Molecules ; 23(8)2018 Jul 27.
Article in English | MEDLINE | ID: mdl-30060461

ABSTRACT

G-quadruplex (G4) nucleic acid structures have been reported to be involved in several human pathologies, including cancer, neurodegenerative disorders and infectious diseases; however, G4 targeting compounds still need implementation in terms of drug-like properties and selectivity in order to reach the clinical use. So far, G4 ligands have been mainly identified through high-throughput screening methods or design of molecules with pre-set features. Here, we describe the development of new heterocyclic ligands through a fragment-based drug discovery (FBDD) approach. The ligands were designed against the major G4 present in the long terminal repeat (LTR) promoter region of the human immunodeficiency virus-1 (HIV-1), the stabilization of which has been shown to suppress viral gene expression and replication. Our method is based on the generation of molecular fragment small libraries, screened against the target to further elaborate them into lead compounds. We screened 150 small molecules, composed by structurally and chemically different fragments, selected from commercially available and in-house compounds; synthetic elaboration yielded several G4 ligands and two final G4 binders, both embedding an amidoxime moiety; one of these two compounds showed preferential binding for the HIV-1 LTR G4. This work presents the discovery of a novel potential pharmacophore and highlights the possibility to apply a fragment-based approach to develop G4 ligands with unexpected chemical features.


Subject(s)
HIV Long Terminal Repeat/drug effects , Heterocyclic Compounds/chemical synthesis , Oximes/chemistry , Small Molecule Libraries/pharmacology , Drug Design , Drug Evaluation, Preclinical , G-Quadruplexes , Heterocyclic Compounds/chemistry , Ligands , Molecular Structure , Small Molecule Libraries/chemistry
12.
Antiviral Res ; 152: 94-103, 2018 04.
Article in English | MEDLINE | ID: mdl-29476895

ABSTRACT

HIV-1 inhibitors that act by mechanisms distinct from existing antiretrovirals can provide novel insights into viral replication and potentially inform development of new therapeutics. Using a multi-cycle HIV-1 replication assay, we screened 252 pure compounds derived from marine invertebrates and microorganisms and identified 6 (actinomycin Z2, bastadin 6, bengamide A, haliclonacyclamine A + B, keramamine C, neopetrosiamide B) that inhibited HIV-1 with 50% effective concentrations (EC50s) of 3.8 µM or less. The most potent inhibitor, bengamide A, blocked HIV-1 in a T cell line with an EC50 of 0.015 µM and in peripheral blood mononuclear cells with an EC50 of 0.032 µM. Bengamide A was previously described to inhibit NF-κB signaling. Consistent with this mechanism, bengamide A suppressed reporter expression from an NF-κB-driven minimal promoter and an HIV-1 long terminal repeat (LTR) with conserved NF-κB response elements, but lacked activity against an LTR construct with mutation of these elements. In single-cycle HIV-1 infection assays, bengamide A also suppressed viral protein expression when viruses encoded an intact LTR but exhibited minimal activity against those with mutated NF-κB elements. Finally, bengamide A did not inhibit viral DNA accumulation, indicating that it likely acts downstream of this step in HIV-1 replication. Our study identifies multiple new antiviral compounds including an unusually potent inhibitor of HIV-1 gene expression.


Subject(s)
Anti-HIV Agents/pharmacology , Biological Products/pharmacology , HIV Infections/metabolism , HIV-1/physiology , NF-kappa B/metabolism , Virus Replication/drug effects , Anti-HIV Agents/chemistry , Aquatic Organisms/chemistry , Biological Products/chemistry , Drug Evaluation, Preclinical , Gene Expression Regulation, Viral/drug effects , HIV Infections/genetics , HIV Infections/virology , HIV Long Terminal Repeat/drug effects , HIV-1/genetics , Humans , Leukocytes, Mononuclear/virology , NF-kappa B/genetics
13.
J Biol Chem ; 293(1): 296-311, 2018 01 05.
Article in English | MEDLINE | ID: mdl-29158267

ABSTRACT

HIV-1 infection and methamphetamine (METH) abuse frequently occur simultaneously and may have synergistic pathological effects. Although HIV-positive/active METH users have been shown to have higher HIV viral loads and experience more severe neurological complications than non-users, the direct impact of METH on HIV infection and its link to the development of neurocognitive alternations are still poorly understood. In the present study, we hypothesized that METH impacts HIV infection of neural progenitor cells (NPCs) by a mechanism encompassing NFκB/SP1-mediated HIV LTR activation. Mouse and human NPCs were infected with EcoHIV (modified HIV virus infectious to mice) and HIV, respectively, in the presence or absence of METH (50 or 100 µm). Pretreatment with METH, but not simultaneous exposure, significantly increased HIV production in both mouse and human NPCs. To determine the mechanisms underlying these effects, cells were transfected with different variants of HIV LTR promoters and then exposed to METH. METH treatment induced transcriptional activity of the HIV LTR promotor, an effect that required both NFκB and SP1 signaling. Pretreatment with METH also decreased neuronal differentiation of HIV-infected NPCs in both in vitro and in vivo settings. Importantly, NPC-derived daughter cells appeared to be latently infected with HIV. This study indicates that METH increases HIV infectivity of NPCs, through the NFκB/SP1-dependent activation of the HIV LTR and with the subsequent alterations of NPC neurogenesis. Such events may underlie METH- exacerbated neurocognitive dysfunction in HIV-infected patients.


Subject(s)
HIV Infections/virology , HIV-1/drug effects , Methamphetamine/pharmacology , Animals , Cell Line , HIV Long Terminal Repeat/drug effects , Humans , Male , Methamphetamine/adverse effects , Mice , Mice, Inbred C57BL , NF-kappa B/drug effects , NF-kappa B/metabolism , Neural Stem Cells/drug effects , Neurons/drug effects , Signal Transduction/drug effects , Sp1 Transcription Factor/drug effects , Stem Cells/drug effects , Viral Load
14.
Antiviral Res ; 146: 76-85, 2017 Oct.
Article in English | MEDLINE | ID: mdl-28842263

ABSTRACT

Combination antiretroviral therapy (cART) has been proven to efficiently inhibit ongoing replication of human immunodeficiency virus type 1 (HIV-1), and significantly improve the health outcome in patients of acquired immune deficiency syndrome (AIDS). However, cART is unable to cure HIV-1/AIDS. Even in presence of cART there exists a residual viremia, contributed from the viral reservoirs of latently infected HIV-1 proviruses; this constitutes a major hurdle. Currently, there are multiple strategies aimed at eliminating or permanently silence these HIV-1 latent reservoirs being intensely explored. One such strategy, a recently emerged "block and lock" approach is appealing. For this approach, so-called HIV-1 latency-promoting agents (LPAs) are used to reinforce viral latency and to prevent the low-level or sporadic transcription of integrated HIV-1 proviruses. Although several LPAs have been reported, there is still a question of their suitability to be further developed as a safe and valid therapeutic agent for the clinical use. In this study, we aimed to identify new potential LPAs through the screening an FDA-approved compound library. A new and promising anti-HIV-1 inhibitor, levosimendan, was identified from these screens. Levosimendan is currently used to treat heart failure in clinics, but it demonstrates strong inhibition of TNFα-induced HIV-1 reactivation in multiple cell lines of HIV-1 latency through affecting the HIV-1 Tat-LTR transcriptional axis. Furthermore, we confirmed that in primary CD4+ T cells levosimendan inhibits both the acute HIV-1 replication and the reactivation of latent HIV-1 proviruses. As a summary, our studies successfully identify levosimendan as a novel and promising anti-HIV-1 inhibitor, which should be immediately investigated in vivo given that it is already an FDA-approved drug.


Subject(s)
Anti-HIV Agents/pharmacology , Drug Discovery/methods , HIV-1/drug effects , Hydrazones/pharmacology , Pyridazines/pharmacology , Transcription, Genetic/drug effects , Anti-HIV Agents/chemistry , Anti-HIV Agents/isolation & purification , Cell Line , Drug Approval , Gene Expression Regulation, Viral/drug effects , HIV Infections/drug therapy , HIV Infections/virology , HIV Long Terminal Repeat/drug effects , HIV-1/physiology , Humans , Hydrazones/isolation & purification , Pyridazines/isolation & purification , Simendan , Small Molecule Libraries , Virus Activation/drug effects , Virus Latency/drug effects
15.
J Med Chem ; 59(24): 11148-11160, 2016 12 22.
Article in English | MEDLINE | ID: mdl-28002966

ABSTRACT

Small molecules that bind to RNA potently and specifically are relatively rare. The study of molecules that bind to the HIV-1 transactivation response (TAR) hairpin, a cis-acting HIV genomic element, has long been an important model system for the chemistry of targeting RNA. Here we report the synthesis, biochemical, and structural evaluation of a series of molecules that bind to HIV-1 TAR RNA. A promising analogue, 15, retained the TAR binding affinity of the initial hit and displaced a Tat-derived peptide with an IC50 of 40 µM. NMR characterization of a soluble analogue, 2, revealed a noncanonical binding mode for this class of compounds. Finally, evaluation of 2 and 15 by selective 2'-hydroxyl acylation analyzed by primer extension (SHAPE) indicates specificity in binding to TAR within the context of an in vitro-synthesized 365-nt HIV-1 5'-untranslated region (UTR). Thus, these compounds exhibit a novel and specific mode of interaction with TAR, providing important suggestions for RNA ligand design.


Subject(s)
HIV Long Terminal Repeat/drug effects , RNA, Viral/drug effects , Small Molecule Libraries/pharmacology , Binding Sites/drug effects , Dose-Response Relationship, Drug , HIV Long Terminal Repeat/genetics , Molecular Structure , RNA, Viral/genetics , Small Molecule Libraries/chemical synthesis , Small Molecule Libraries/chemistry , Structure-Activity Relationship , Transcriptional Activation/drug effects , Transcriptional Activation/genetics
16.
Org Biomol Chem ; 14(6): 2052-6, 2016 Feb 14.
Article in English | MEDLINE | ID: mdl-26765486

ABSTRACT

Recognition of RNA by high-affinity binding small molecules is crucial for expanding existing approaches in RNA recognition, and for the development of novel RNA binding drugs. A novel neomycin dimer benzimidazole conjugate 5 (DPA 83) was synthesized by conjugating a neomycin-dimer with a benzimidazole alkyne using click chemistry to target multiple binding sites on HIV TAR RNA. Ligand 5 significantly enhances the thermal stability of HIV TAR RNA and interacts stoichiometrically with HIV TAR RNA with a low nanomolar affinity. 5 displayed enhanced binding compared to its individual building blocks including the neomycin dimer azide and benzimidazole alkyne. In essence, a high affinity multivalent ligand was designed and synthesized to target HIV TAR RNA.


Subject(s)
Aminoglycosides/pharmacology , Benzimidazoles/pharmacology , HIV Long Terminal Repeat/drug effects , RNA, Viral/antagonists & inhibitors , Aminoglycosides/chemical synthesis , Aminoglycosides/chemistry , Benzimidazoles/chemical synthesis , Benzimidazoles/chemistry , Binding Sites/drug effects , Click Chemistry , Dose-Response Relationship, Drug , Ligands , Molecular Conformation , Neomycin/chemistry , Neomycin/pharmacology , Structure-Activity Relationship
17.
Food Funct ; 6(11): 3412-9, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26404185

ABSTRACT

No safe and effective cure currently exists for human immunodeficiency virus (HIV). However, antiretroviral therapy can prolong the lives of HIV patients and lowers the secondary infections. Natural compounds, which are considered to be pleiotropic molecules, could be useful against HIV. Curcumin, a yellow pigment present in the spice turmeric (Curcuma longa), can be used for the treatment of several diseases including HIV-AIDS because of its antioxidant, anti-inflammatory, anticancer, antiviral, and antibacterial nature. In this review we have summarized that how curcumin and its analogues inhibit the infection and replication of viral genes and prevent multiplicity of HIV. They are inhibitors of HIV protease and integrase. Curcumin also inhibits Tat transactivation of the HIV1-LTR genome, inflammatory molecules (interleukins, TNF-α, NF-κB, COX-2) and HIV associated various kinases including tyrosine kinase, PAK1, MAPK, PKC, cdk and others. In addition, curcumin enhances the effect of conventional therapeutic drugs and minimizes their side effects.


Subject(s)
Antiviral Agents/pharmacology , Curcuma/chemistry , Curcumin/analogs & derivatives , Curcumin/pharmacology , HIV Infections/drug therapy , Phytotherapy , Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Antioxidants/pharmacology , Enzyme Inhibitors/pharmacology , HIV Long Terminal Repeat/drug effects , HIV-1/drug effects , Humans
18.
Org Biomol Chem ; 13(45): 11096-104, 2015 Dec 07.
Article in English | MEDLINE | ID: mdl-26399751

ABSTRACT

RNA is a drug target involved in diverse cellular functions and viral processes. Molecules that inhibit the HIV TAR RNA-Tat protein interaction may attenuate Tat/TAR-dependent protein expression and potentially serve as anti-HIV therapeutics. By incorporating positively charged residues with mixed side chain lengths, we designed peptides that bind TAR RNA with enhanced intracellular activity. Tat-derived peptides that were individually substituted with positively charged residues with varying side chain lengths were evaluated for TAR RNA binding. Positively charged residues with different side chain lengths were incorporated at each Arg and Lys position in the Tat-derived peptide to enhance TAR RNA binding. The resulting peptides showed enhanced TAR RNA binding affinity, cellular uptake, nuclear localization, proteolytic resistance, and inhibition of intracellular Tat/TAR-dependent protein expression compared to the parent Tat-derived peptide with no cytotoxicity. Apparently, the enhanced inhibition of protein expression by these peptides was not determined by RNA binding affinity, but by proteolytic resistance. Despite the high TAR binding affinity, a higher binding specificity would be necessary for practical purposes. Importantly, altering the positively charged residue side chain length should be a viable strategy to generate potentially useful RNA-targeting bioactive molecules.


Subject(s)
Anti-HIV Agents/pharmacology , Gene Expression Regulation, Viral/drug effects , Gene Products, tat/pharmacology , HIV Long Terminal Repeat , HIV/genetics , Peptides/pharmacology , RNA, Viral/genetics , Amino Acid Sequence , Anti-HIV Agents/chemistry , Anti-HIV Agents/pharmacokinetics , Cell Line , Gene Products, tat/chemistry , Gene Products, tat/pharmacokinetics , HIV/drug effects , HIV/metabolism , HIV Infections/drug therapy , HIV Long Terminal Repeat/drug effects , Humans , Peptides/chemistry , Peptides/pharmacokinetics , RNA, Viral/metabolism
19.
Sci Rep ; 5: 12442, 2015 Jul 22.
Article in English | MEDLINE | ID: mdl-26199173

ABSTRACT

Multiple studies have shown that HIV-1 patients may develop virus reservoirs that impede eradication; these reservoirs include the central nervous system (CNS). Despite an undetectable viral load in patients treated with potent antiretrovirals, current therapy is unable to purge the virus from these latent reservoirs. To broaden the inhibitory range and effectiveness of current antiretrovirals, the potential of bryostatin was investigated as a latent HIV-1 activator. We used primary astrocytes, NHA cells, and astrocytoma cells U-87. Infected cells with HIV-1(NL4.3) were treated with bryostatin alone or in combination with different inhibitors. HIV-1 production was quantified by using ELISA. Transcriptional activity was measured using luciferase reporter gene assays by using lipofectin. We performed cotransfection experiments of the LTR promoter with the active NF-κB member p65/relA. To confirm the NF-κB role, Western blot and confocal microscopy were performed. Bryostatin reactivates latent viral infection in the NHA and U87 cells via activation of protein kinase C (PKC)-alpha and -delta, because the PKC inhibitors rottlerin and GF109203X abrogated the bryostatin effect. No alteration in cell proliferation was found. Moreover, bryostatin strongly stimulated LTR transcription by activating the transcription factor NF-κB. Bryostatin could be a beneficial adjunct to the treatment of HIV-1 brain infection.


Subject(s)
Astrocytes/metabolism , Astrocytes/virology , Bryostatins/pharmacology , HIV-1/drug effects , NF-kappa B/metabolism , Protein Kinase C/metabolism , Virus Latency/drug effects , Cell Line, Tumor , Gene Expression Regulation, Viral/drug effects , Gene Expression Regulation, Viral/genetics , HIV Infections/metabolism , HIV Infections/virology , HIV Long Terminal Repeat/drug effects , HIV Long Terminal Repeat/genetics , HIV-1/genetics , Humans , Promoter Regions, Genetic/drug effects , Promoter Regions, Genetic/genetics , Signal Transduction/drug effects , Signal Transduction/genetics , Transcriptional Activation/drug effects , Transcriptional Activation/genetics , Virus Latency/genetics
20.
Bioorg Med Chem ; 23(9): 2139-47, 2015 May 01.
Article in English | MEDLINE | ID: mdl-25819332

ABSTRACT

Nucleobase modified neamines with a lysine as the linker (NbK-neamines) were synthesized and their binding toward hairpin RNAs derived from HIV-1 activator region were studied. NbK-neamines were bind those RNAs with micro molar level of binding affinities and compete with corresponding activator peptide for TAR RNA, but not for RRE RNA. GbK-neamine denotes the highest binding affinity with TAR RNA, three to five times higher than other three NbK-neamines. GbK-neamine could be a candidate of potential inhibitor for TAR-Tat.


Subject(s)
Framycetin/chemistry , Framycetin/pharmacology , HIV Long Terminal Repeat/drug effects , HIV-1/chemistry , HIV-1/genetics , Lysine/chemistry , tat Gene Products, Human Immunodeficiency Virus/antagonists & inhibitors , Adenine/chemistry , Cytosine/chemistry , Framycetin/chemical synthesis , Guanine/chemistry , Molecular Structure , Structure-Activity Relationship , Thymine/chemistry
SELECTION OF CITATIONS
SEARCH DETAIL