Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 27
Filter
1.
Viruses ; 14(4)2022 04 15.
Article in English | MEDLINE | ID: mdl-35458546

ABSTRACT

HIV-1 Vpu targets the host cell proteins CD4 and BST-2/Tetherin for degradation, ultimately resulting in enhanced virus spread and host immune evasion. The discovery and characterization of small molecules that antagonize Vpu would further elucidate the contribution of Vpu to pathogenesis and lay the foundation for the study of a new class of novel HIV-1 therapeutics. To identify novel compounds that block Vpu activity, we have developed a cell-based 'gain of function' assay that produces a positive signal in response to Vpu inhibition. To develop this assay, we took advantage of the viral glycoprotein, GaLV Env. In the presence of Vpu, GaLV Env is not incorporated into viral particles, resulting in non-infectious virions. Vpu inhibition restores infectious particle production. Using this assay, a high throughput screen of >650,000 compounds was performed to identify inhibitors that block the biological activity of Vpu. From this screen, we identified several positive hits but focused on two compounds from one structural family, SRI-41897 and SRI-42371. We developed independent counter-screens for off target interactions of the compounds and found no off target interactions. Additionally, these compounds block Vpu-mediated modulation of CD4, BST-2/Tetherin and antibody dependent cell-mediated toxicity (ADCC). Unfortunately, both SRI-41897 and SRI-42371 were shown to be specific to the N-terminal region of NL4-3 Vpu and did not function against other, more clinically relevant, strains of Vpu; however, this assay may be slightly modified to include more significant Vpu strains in the future.


Subject(s)
Anti-HIV Agents , HIV-1 , Human Immunodeficiency Virus Proteins , Viral Regulatory and Accessory Proteins , Viroporin Proteins , Anti-HIV Agents/chemistry , Bone Marrow Stromal Antigen 2/metabolism , GPI-Linked Proteins/metabolism , HIV-1/metabolism , Human Immunodeficiency Virus Proteins/antagonists & inhibitors , Human Immunodeficiency Virus Proteins/metabolism , Leukemia Virus, Gibbon Ape/metabolism , Small Molecule Libraries , Viral Regulatory and Accessory Proteins/antagonists & inhibitors , Viral Regulatory and Accessory Proteins/metabolism , Viroporin Proteins/antagonists & inhibitors
2.
Int J Mol Sci ; 22(17)2021 Aug 27.
Article in English | MEDLINE | ID: mdl-34502213

ABSTRACT

Bone marrow stromal cell antigen 2 (BST-2), also known as CD317 or tetherin, has been identified as a host restriction factor that suppresses the release of enveloped viruses from host cells by physically tethering viral particles to the cell surface; however, this host defense can be subverted by multiple viruses. For example, human immunodeficiency virus (HIV)-1 encodes a specific accessory protein, viral protein U (Vpu), to counteract BST-2 by binding to it and directing its lysosomal degradation. Thus, blocking the interaction between Vpu and BST-2 will provide a promising strategy for anti-HIV therapy. Here, we report a NanoLuc Binary Technology (NanoBiT)-based high-throughput screening assay to detect inhibitors that disrupt the Vpu-BST-2 interaction. Out of more than 1000 compounds screened, four inhibitors were identified with strong activity at nontoxic concentrations. In subsequent cell-based BST-2 degradation assays, inhibitor Y-39983 HCl restored the cell-surface and total cellular level of BST-2 in the presence of Vpu. Furthermore, the Vpu-mediated enhancement of pesudotyped viral particle production was inhibited by Y-39983 HCl. Our findings indicate that our newly developed assay can be used for the discovery of potential antiviral molecules with novel mechanisms of action.


Subject(s)
Anti-HIV Agents/pharmacology , HIV Infections/drug therapy , HIV-1/drug effects , High-Throughput Screening Assays/methods , Human Immunodeficiency Virus Proteins/antagonists & inhibitors , Protein Interaction Domains and Motifs/drug effects , Viral Regulatory and Accessory Proteins/antagonists & inhibitors , Antigens, CD/metabolism , GPI-Linked Proteins/antagonists & inhibitors , GPI-Linked Proteins/metabolism , HIV Infections/metabolism , HIV Infections/virology , HeLa Cells , Human Immunodeficiency Virus Proteins/metabolism , Humans , Nanotechnology/methods , Viral Regulatory and Accessory Proteins/metabolism , Virus Replication
3.
Biotechnol Appl Biochem ; 68(4): 918-926, 2021 Aug.
Article in English | MEDLINE | ID: mdl-32860447

ABSTRACT

The importance of new effective treatment methodologies for human immunodeficiency virus (HIV) is undeniable for the medical society. Viral protein U (Vpu), one of the disparaged accessory proteins of HIV, is responsible for the dissemination of viral particles, and HIV mutants lacking Vpu protein have remarkably reduced pathogenicity. Here, we explored the marine natural products to find the leading structures which can potentially inhibit the activity of Vpu in silico. To fulfill this goal, we set up a virtual screening based on molecular docking to evaluate the binding capacity of different marine products to Vpu. For validation, we used molecular dynamics simulation and monitored the root mean square deviation value and binding interactions. The results were intriguing when we realized that the hit compounds (phlorotannins) had previously been identified as reverse transcriptase and HIV protease inhibitors. This research inaugurates a new road to combat HIV by multifaceted mode of action of these marine natural products without putting the normal cells in jeopardy (with their safe toxicological profile).


Subject(s)
Anti-Retroviral Agents/chemistry , Aquatic Organisms/chemistry , Biological Products/chemistry , HIV-1/chemistry , Human Immunodeficiency Virus Proteins , Molecular Docking Simulation , Viral Regulatory and Accessory Proteins , Human Immunodeficiency Virus Proteins/antagonists & inhibitors , Human Immunodeficiency Virus Proteins/chemistry , Humans , Viral Regulatory and Accessory Proteins/antagonists & inhibitors , Viral Regulatory and Accessory Proteins/chemistry
4.
J Infect Dis ; 223(11): 1914-1922, 2021 06 04.
Article in English | MEDLINE | ID: mdl-33038249

ABSTRACT

BIT225 is a first-in-class inhibitor of human immunodeficiency virus (HIV) type 1 Vpu. A phase II trial enrolled 36 HIV-1-infected, treatment-naive participants in Thailand to receive standard-of-care antiretroviral therapy (ART), tenofovir disoproxil fumarate/emtricitabine/efavirenz (Atripla), with 100 or 200 mg of BIT225 or placebo (daily) for 12 weeks. Combined treatment with BIT225 and ART was found to be generally safe and well tolerated, with antiviral efficacy comparable to that of ART alone. The secondary end point-soluble CD163, a marker of monocyte/macrophage inflammation-was noted to be significantly decreased in the BIT225 arm. Plasma-derived activated CD4+ and CD8+ T cells, natural killer cells, and interleukin 21 were increased in those treated with BIT225. These findings are consistent with inhibition of the known effects of HIV Vpu and may reflect clinically important modulation of inflammatory and immune function. Further clinical study is planned to both confirm and extend these important findings in treatment-naive, and treatment-experienced individuals. Clinical Trials Registration. Australian New Zealand Clinical Trials Registry (Universal Trial Number U1111-1191-2194).


Subject(s)
Anti-HIV Agents , Efavirenz, Emtricitabine, Tenofovir Disoproxil Fumarate Drug Combination , Guanidines/therapeutic use , HIV Infections , Pyrazoles/therapeutic use , Anti-HIV Agents/therapeutic use , Australia , CD4-Positive T-Lymphocytes , CD8-Positive T-Lymphocytes , Drug Therapy, Combination , Efavirenz, Emtricitabine, Tenofovir Disoproxil Fumarate Drug Combination/therapeutic use , HIV Infections/drug therapy , HIV Infections/immunology , HIV-1 , Human Immunodeficiency Virus Proteins/antagonists & inhibitors , Humans , Inflammation/drug therapy , Thailand , Viral Regulatory and Accessory Proteins/antagonists & inhibitors , Viroporin Proteins/antagonists & inhibitors
5.
Eur J Med Chem ; 190: 112111, 2020 Mar 15.
Article in English | MEDLINE | ID: mdl-32058240

ABSTRACT

Novel ethyl 2-(5-aryl-1H-imidazol-1-yl)-acetates 17 and propionates 18, together with their acetic acid 19 and acetohydrazide 20 derivatives, were designed and synthesized using TosMIC chemistry. Biological evaluation of these newly synthesized scaffolds in the HIV-1 Vpu- Host BST-2 ELISA assay identified seven hits (17a, 17b, 17c, 17g, 18a, 20f and 20g) with greater than 50% inhibitory activity. These hits were validated in the HIV-1 Vpu- Host BST-2 AlphaScreen™ and six of the seven compounds were found to have comparable percentage inhibitory activities to those of the ELISA assay. Compounds 17b and 20g, with consistent percentage inhibitory activities across the two assays, had IC50 values of 11.6 ± 1.1 µM and 17.6 ± 0.9 µM in a dose response AlphaScreen™ assay. In a cell-based HIV-1 antiviral assay, compound 17b exhibited an EC50 = 6.3 ± 0.7 µM at non-toxic concentrations (CC50 = 184.5 ± 0.8 µM), whereas compound 20g displayed antiviral activity roughly equivalent to its toxicity (CC50 = 159.5 ± 0.9 µM). This data suggests that compound 17b, active in both cell-based and biochemical assays, provides a good starting point for the design of possible lead compounds for prevention of HIV-1 Vpu and host BST-2 protein binding in new anti-HIV therapeutics.


Subject(s)
Anti-HIV Agents/pharmacology , HIV-1/drug effects , Human Immunodeficiency Virus Proteins/antagonists & inhibitors , Imidazoles/pharmacology , Protein Multimerization/drug effects , Viral Regulatory and Accessory Proteins/antagonists & inhibitors , Anti-HIV Agents/chemical synthesis , Antigens, CD , Cell Line , Drug Design , GPI-Linked Proteins/antagonists & inhibitors , HIV-1/chemistry , Humans , Imidazoles/chemical synthesis , Microbial Sensitivity Tests , Virus Replication/drug effects
6.
Adv Protein Chem Struct Biol ; 111: 197-222, 2018.
Article in English | MEDLINE | ID: mdl-29459032

ABSTRACT

Protein-protein interaction (PPI) inhibitors are a rapidly expanding class of therapeutics. Recent advances in our understanding of PPIs and success of early examples of PPI inhibitors demonstrate the feasibility of targeting PPIs. This review summarizes the techniques used for the discovery and optimization of a diverse set PPI inhibitors, focusing on the development of PPI inhibitors as new antibacterial and antiviral agents. We close with a summary of the advances responsible for making PPI inhibitors realistic targets for therapeutic intervention and brief outlook of the field.


Subject(s)
Anti-Bacterial Agents/pharmacology , Antiviral Agents/pharmacology , Communicable Diseases/drug therapy , HIV Fusion Inhibitors/pharmacology , HIV Integrase Inhibitors/pharmacology , Human Immunodeficiency Virus Proteins/antagonists & inhibitors , Anti-Bacterial Agents/chemistry , Antiviral Agents/chemistry , Bacteria/drug effects , Communicable Diseases/microbiology , Communicable Diseases/virology , HIV Fusion Inhibitors/chemistry , HIV Integrase Inhibitors/chemistry , Host Microbial Interactions/drug effects , Human Immunodeficiency Virus Proteins/metabolism , Humans
7.
Biochem J ; 474(10): 1559-1577, 2017 04 26.
Article in English | MEDLINE | ID: mdl-28446620

ABSTRACT

Infections by the human immunodeficiency virus type 1 (HIV-1), the causative agent of the acquired immunodeficiency syndrome (AIDS), are still totaling an appalling 36.7 millions worldwide, with 1.1 million AIDS deaths/year and a similar number of yearly new infections. All this, in spite of the discovery of HIV-1 as the AIDS etiological agent more than 30 years ago and the introduction of an effective combinatorial antiretroviral therapy (cART), able to control disease progression, more than 20 years ago. Although very effective, current cART is plagued by the emergence of drug-resistant viral variants and most of the efforts in the development of novel direct-acting antiviral agents (DAAs) against HIV-1 have been devoted toward the fighting of resistance. In this review, rather than providing a detailed listing of all the drugs and the corresponding resistance mutations, we aim, through relevant examples, at presenting to the general reader the conceptual shift in the approaches that are being taken to overcome the viral resistance hurdle. From the classic 'running faster' strategy, based on the development of novel DAAs active against the mutant viruses selected by the previous drugs and/or presenting to the virus a high genetic barrier toward the development of resilience, to a 'jumping higher' approach, which looks at the cell, rather than the virus, as a source of valuable drug targets, in order to make the cellular environment non-permissive toward the replication of both wild-type and mutated viruses.


Subject(s)
Anti-HIV Agents/therapeutic use , Drug Design , Drug Resistance, Multiple, Viral , Drug Therapy, Combination , HIV Infections/drug therapy , HIV-1/drug effects , Models, Biological , Animals , Anti-HIV Agents/adverse effects , Anti-HIV Agents/chemistry , Anti-HIV Agents/pharmacology , Antiretroviral Therapy, Highly Active/adverse effects , CCR5 Receptor Antagonists/chemistry , CCR5 Receptor Antagonists/pharmacology , CCR5 Receptor Antagonists/therapeutic use , DEAD-box RNA Helicases/antagonists & inhibitors , DEAD-box RNA Helicases/chemistry , DEAD-box RNA Helicases/genetics , DEAD-box RNA Helicases/metabolism , Drug Therapy, Combination/adverse effects , HIV Infections/metabolism , HIV Infections/virology , HIV Protease Inhibitors/adverse effects , HIV Protease Inhibitors/chemistry , HIV Protease Inhibitors/pharmacology , HIV Protease Inhibitors/therapeutic use , HIV-1/genetics , HIV-1/growth & development , HIV-1/physiology , Host-Pathogen Interactions/drug effects , Human Immunodeficiency Virus Proteins/antagonists & inhibitors , Human Immunodeficiency Virus Proteins/chemistry , Human Immunodeficiency Virus Proteins/genetics , Human Immunodeficiency Virus Proteins/metabolism , Humans , Molecular Structure , Molecular Targeted Therapy , Mutation , Protein Conformation , Reverse Transcriptase Inhibitors/chemistry , Reverse Transcriptase Inhibitors/pharmacology , Reverse Transcriptase Inhibitors/therapeutic use , Virus Physiological Phenomena/drug effects , Virus Replication/drug effects
8.
Future Med Chem ; 7(8): 1055-77, 2015.
Article in English | MEDLINE | ID: mdl-26062401

ABSTRACT

Protein-protein interactions (PPI) are essential in every step of the HIV replication cycle. Mapping the interactions between viral and host proteins is a fundamental target for the design and development of new therapeutics. In this review, we focus on rational development of anti-HIV-1 peptides based on mapping viral-host and viral-viral protein interactions all across the HIV-1 replication cycle. We also discuss the mechanism of action, specificity and stability of these peptides, which are designed to inhibit PPI. Some of these peptides are excellent tools to study the mechanisms of PPI in HIV-1 replication cycle and for the development of anti-HIV-1 drug leads that modulate PPI.


Subject(s)
Anti-HIV Agents/pharmacology , Human Immunodeficiency Virus Proteins/antagonists & inhibitors , Peptides/metabolism , Anti-HIV Agents/chemical synthesis , Anti-HIV Agents/chemistry , Human Immunodeficiency Virus Proteins/metabolism , Humans , Protein Binding/drug effects , Virus Replication/drug effects
9.
PLoS One ; 10(4): e0121099, 2015.
Article in English | MEDLINE | ID: mdl-25830320

ABSTRACT

The continued burden of HIV in resource-limited regions such as parts of sub-Saharan Africa, combined with adverse effects and potential risks of resistance to existing antiretroviral therapies, emphasize the need to identify new HIV inhibitors. Here we performed a virtual screen of molecules from the pan-African Natural Product Library, the largest collection of medicinal plant-derived pure compounds on the African continent. We identified eight molecules with structural similarity to reported interactors of Vpu, an HIV-1 accessory protein with reported ion channel activity. Using in vitro HIV-1 replication assays with a CD4+ T cell line and peripheral blood mononuclear cells, we confirmed antiviral activity and minimal cytotoxicity for two compounds, ixoratannin A-2 and boldine. Notably, ixoratannin A-2 retained inhibitory activity against recombinant HIV-1 strains encoding patient-derived mutations that confer resistance to protease, non-nucleoside reverse transcriptase, or integrase inhibitors. Moreover, ixoratannin A-2 was less effective at inhibiting replication of HIV-1 lacking Vpu, supporting this protein as a possible direct or indirect target. In contrast, boldine was less effective against a protease inhibitor-resistant HIV-1 strain. Both ixoratannin A-2 and boldine also inhibited in vitro replication of hepatitis C virus (HCV). However, BIT-225, a previously-reported Vpu inhibitor, demonstrated antiviral activity but also cytotoxicity in HIV-1 and HCV replication assays. Our work identifies pure compounds derived from African plants with potential novel activities against viruses that disproportionately afflict resource-limited regions of the world.


Subject(s)
Aporphines/pharmacology , Biological Products/chemistry , HIV-1/drug effects , Proanthocyanidins/pharmacology , Aporphines/chemistry , CD4-Positive T-Lymphocytes/cytology , CD4-Positive T-Lymphocytes/virology , Cell Line , Drug Resistance, Viral , Guanidines/pharmacology , HIV-1/physiology , Hepacivirus/drug effects , Hepacivirus/physiology , Human Immunodeficiency Virus Proteins/antagonists & inhibitors , Human Immunodeficiency Virus Proteins/metabolism , Humans , Leukocytes, Mononuclear/cytology , Leukocytes, Mononuclear/virology , Molecular Docking Simulation , Proanthocyanidins/chemistry , Pyrazoles/pharmacology , Viral Regulatory and Accessory Proteins/antagonists & inhibitors , Viral Regulatory and Accessory Proteins/metabolism , Virus Replication/drug effects
10.
Proteins ; 82(10): 2770-82, 2014 Oct.
Article in English | MEDLINE | ID: mdl-25043744

ABSTRACT

Rational design of proteins with novel binding specificities and increased affinity is one of the major goals of computational protein design. Epitope-scaffolds are a new class of antigens engineered by transplanting viral epitopes of predefined structure to protein scaffolds, or by building protein scaffolds around such epitopes. Epitope-scaffolds are of interest as vaccine components to attempt to elicit neutralizing antibodies targeting the specified epitope. In this study we developed a new computational protocol, MultiGraft Interface, that transplants epitopes but also designs additional scaffold features outside the epitope to enhance antibody-binding specificity and potentially influence the specificity of elicited antibodies. We employed MultiGraft Interface to engineer novel epitope-scaffolds that display the known epitope of human immunodeficiency virus 1 (HIV-1) neutralizing antibody 2F5 and that also interact with the functionally important CDR H3 antibody loop. MultiGraft Interface generated an epitope-scaffold that bound 2F5 with subnanomolar affinity (K(D) = 400 pM) and that interacted with the antibody CDR H3 loop through computationally designed contacts. Substantial structural modifications were necessary to engineer this antigen, with the 2F5 epitope replacing a helix in the native scaffold and with 15% of the native scaffold sequence being modified in the design stage. This epitope-scaffold represents a successful example of rational protein backbone engineering and protein-protein interface design and could prove useful in the field of HIV vaccine design. MultiGraft Interface can be generally applied to engineer novel binding partners with altered specificity and optimized affinity.


Subject(s)
Antibodies, Monoclonal/metabolism , Antibodies, Neutralizing/metabolism , Antibodies, Viral/metabolism , Antigens/chemistry , Complementarity Determining Regions/chemistry , Human Immunodeficiency Virus Proteins/antagonists & inhibitors , Models, Molecular , Amino Acid Sequence , Animals , Antibodies, Monoclonal/chemistry , Antibodies, Monoclonal/genetics , Antibodies, Neutralizing/chemistry , Antibodies, Neutralizing/genetics , Antibodies, Viral/chemistry , Antibodies, Viral/genetics , Antibody Affinity , Antibody Specificity , Antigens/genetics , Antigens/metabolism , Broadly Neutralizing Antibodies , Complementarity Determining Regions/genetics , Complementarity Determining Regions/metabolism , Computational Biology , Epitopes/chemistry , Epitopes/metabolism , HIV Antibodies , Human Immunodeficiency Virus Proteins/chemistry , Human Immunodeficiency Virus Proteins/genetics , Human Immunodeficiency Virus Proteins/metabolism , Internet , Molecular Sequence Data , Peptide Library , Protein Conformation , Protein Engineering , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism , Software
11.
Planta Med ; 80(8-9): 682-7, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24963618

ABSTRACT

Various drugs found in Chinese herbs are well known for their antiviral potency. We have tested several flavonoids with respect to their potency to block the viral protein U of the human immunodeficiency type 1 virus, which is believed to form a cation-permeable ion channel in the infected cell. We used Xenopus oocytes with heterologously expressed viral protein U as model system to test the efficacy of the drugs in voltage-clamp experiments. This method had been demonstrated in the past as a useful tool to screen drugs for their potency in inhibition of ion channel activity. The viral protein U-mediated current could be inhibited by Ba(2+) with a K1/2 value of 1.6 mM. Therefore, we determined viral protein U-mediated current as current component blocked by 10 mM Ba(2+). We screened several flavonoids with respect to their effects on this current. The flavonols quercetin and kaempferol, and the flavanols (-)epigallochatechin and (-)epichatechin were ineffective. The flavanone naringenin showed at 20 µM slight (about 10%) inhibition. The most potent drug was the isoflavon genistein which exhibited at 20 µM significant inhibition of about 40% with a K1/2 value of 81 ± 4 µM. We suggest that viral ion channels, in general, may be a good target for development of antiviral agents, and that, in particular, isoflavons may be candidates for development of drugs targeting viral protein U.


Subject(s)
Antiviral Agents/pharmacology , Flavanones/pharmacology , Flavonoids/pharmacology , Genistein/pharmacology , HIV Infections/drug therapy , HIV-1/drug effects , Animals , Antiviral Agents/chemistry , Female , Flavanones/chemistry , Flavonoids/chemistry , Gene Expression , Genistein/chemistry , Human Immunodeficiency Virus Proteins/antagonists & inhibitors , Humans , Ion Channels/drug effects , Patch-Clamp Techniques , RNA, Complementary/genetics , Transgenes , Viral Regulatory and Accessory Proteins/antagonists & inhibitors , Virus Release/drug effects , Xenopus laevis
12.
Virol Sin ; 27(6): 339-44, 2012 Dec.
Article in English | MEDLINE | ID: mdl-23188559

ABSTRACT

The cellular protein tetherin tethers the HIV-1 viral particles on the cellular membrane to inhibit the replication of HIV-1. However, the HIV-1 accessory protein Vpu counteracts the antiviral function of tetherin. In this study, two retroviral vector plasmids were constructed. One inhibited the vpu gene expression; the other one over-expressed the tetherin. Both retroviral vector plasmids could be packaged in the packaging cell line PT67 to obtain the corresponding retroviruses. The retroviral vector plasmids' functions of tetherin over-expression or vpu-RNAi were detected at the cell level. Retroviral vector plasmids were transfected to PT67 cells at different ratios from 0T3V to 3T0V, and then mixed retroviruses were harvested. The antiviral functions of mixed retroviruses were detected in HIV-1 infected TZM-bl cells. The results showed that packaged mixed retroviruses could repress the replication of HIV-1 in TZM-bl cells.


Subject(s)
HIV-1/physiology , Virus Assembly , Virus Replication , Antigens, CD/biosynthesis , Antigens, CD/genetics , Cell Line , Epithelial Cells/virology , GPI-Linked Proteins/biosynthesis , GPI-Linked Proteins/genetics , Gene Expression , Human Immunodeficiency Virus Proteins/antagonists & inhibitors , Human Immunodeficiency Virus Proteins/genetics , Humans , Plasmids , Viral Regulatory and Accessory Proteins/antagonists & inhibitors , Viral Regulatory and Accessory Proteins/genetics
13.
Biochemistry ; 51(6): 1288-96, 2012 Feb 14.
Article in English | MEDLINE | ID: mdl-22264038

ABSTRACT

Bone marrow stromal cell antigen 2 (BST-2) inhibits the release of enveloped viruses from the cell surface. Various viral counter measures have been discovered, which allow viruses to escape BST-2 restriction. Human immunodeficiency virus type 1 (HIV-1) encodes viral protein U (Vpu) that interacts with BST-2 through their transmembrane domains and causes the downregulation of cell surface BST-2. In this study, we used a computer modeling method to establish a molecular model to investigate the binding interface of the transmembrane domains of BST-2 and Vpu. The model predicts that the interface is composed of Vpu residues I6, A10, A14, A18, V25, and W22 and BST-2 residues L23, I26, V30, I34, V35, L41, I42, and T45. Introduction of mutations that have been previously reported to disrupt the Vpu-BST-2 interaction led to a calculated higher binding free energy (MMGBSA), which supports our molecular model. A pharmacophore was also generated on the basis of this model. Our results provide a precise model that predicts the detailed interaction occurring between the transmembrane domains of Vpu and BST-2 and should facilitate the design of anti-HIV agents that are able to disrupt this interaction.


Subject(s)
Antigens, CD/chemistry , Bone Marrow Cells/chemistry , Combinatorial Chemistry Techniques/methods , HIV-1/chemistry , Human Immunodeficiency Virus Proteins/chemistry , Models, Molecular , Multiprotein Complexes/chemistry , Viral Regulatory and Accessory Proteins/chemistry , Antigens, CD/genetics , Cell Membrane/chemistry , Cell Membrane/genetics , GPI-Linked Proteins/antagonists & inhibitors , GPI-Linked Proteins/chemistry , GPI-Linked Proteins/genetics , Human Immunodeficiency Virus Proteins/antagonists & inhibitors , Human Immunodeficiency Virus Proteins/genetics , Humans , Molecular Dynamics Simulation , Multiprotein Complexes/genetics , Predictive Value of Tests , Protein Binding/genetics , Protein Interaction Mapping , Protein Multimerization/genetics , Protein Structure, Secondary/genetics , Stromal Cells/chemistry , Viral Regulatory and Accessory Proteins/antagonists & inhibitors , Viral Regulatory and Accessory Proteins/genetics
14.
Virology ; 422(2): 265-77, 2012 Jan 20.
Article in English | MEDLINE | ID: mdl-22104209

ABSTRACT

We examined the antiviral activity of ADAR1 against HIV-1. Our results indicated that ADAR1 in a transfection system inhibited production of viral proteins and infectious HIV-1 in various cell lines including 293T, HeLa, Jurkat T and primary CD4+ T cells, and was active against a number of X4 and R5 HIV-1 of different clades. Further analysis showed that ADAR1 inhibited viral protein synthesis without any effect on viral RNA synthesis. Mutational analysis showed that ADAR1 introduced most of the A-to-G mutations in the rev RNA, in the region of RNA encoding for Rev Response Element (RRE) binding domain and in env RNA. These mutations inhibited the binding of rev to the RRE and inhibited transport of primary transcripts like gag, pol and env from nucleus to cytoplasm resulting in inhibition of viral protein synthesis without any effect on viral RNA synthesis. Furthermore, ADAR1 induced mutations in the env gene inhibited viral infectivity.


Subject(s)
Adenosine Deaminase/pharmacology , Anti-HIV Agents/pharmacology , HIV-1/drug effects , RNA-Binding Proteins/metabolism , Amino Acid Motifs , Amino Acid Sequence , Base Sequence , Cells, Cultured , Gene Expression Regulation, Viral/drug effects , Genes, env/drug effects , Genes, env/genetics , HIV-1/genetics , HIV-1/physiology , Human Immunodeficiency Virus Proteins/antagonists & inhibitors , Human Immunodeficiency Virus Proteins/biosynthesis , Human Immunodeficiency Virus Proteins/drug effects , Humans , Molecular Sequence Data , Mutation , Protein Structure, Tertiary , RNA, Viral/biosynthesis , RNA, Viral/drug effects , RNA, Viral/genetics , RNA-Binding Proteins/genetics
15.
PLoS Biol ; 9(11): e1001206, 2011 Nov.
Article in English | MEDLINE | ID: mdl-22140357

ABSTRACT

Persistence of a reservoir of latently infected memory T cells provides a barrier to HIV eradication in treated patients. Several reports have implicated the involvement of SWI/SNF chromatin remodeling complexes in restricting early steps in HIV infection, in coupling the processes of integration and remodeling, and in promoter/LTR transcription activation and repression. However, the mechanism behind the seemingly contradictory involvement of SWI/SNF in the HIV life cycle remains unclear. Here we addressed the role of SWI/SNF in regulation of the latent HIV LTR before and after transcriptional activation. We determined the predicted nucleosome affinity of the LTR sequence and found a striking reverse correlation when compared to the strictly positioned in vivo LTR nucleosomal structure; sequences encompassing the DNase hypersensitive regions displayed the highest nucleosome affinity, while the strictly positioned nucleosomes displayed lower affinity for nucleosome formation. To examine the mechanism behind this reverse correlation, we used a combinatorial approach to determine DNA accessibility, histone occupancy, and the unique recruitment and requirement of BAF and PBAF, two functionally distinct subclasses of SWI/SNF at the LTR of HIV-infected cells before and after activation. We find that establishment and maintenance of HIV latency requires BAF, which removes a preferred nucleosome from DHS1 to position the repressive nucleosome-1 over energetically sub-optimal sequences. Depletion of BAF resulted in de-repression of HIV latency concomitant with a dramatic alteration in the LTR nucleosome profile as determined by high resolution MNase nucleosomal mapping. Upon activation, BAF was lost from the HIV promoter, while PBAF was selectively recruited by acetylated Tat to facilitate LTR transcription. Thus BAF and PBAF, recruited during different stages of the HIV life cycle, display opposing function on the HIV promoter. Our data point to the ATP-dependent BRG1 component of BAF as a putative therapeutic target to deplete the latent reservoir in patients.


Subject(s)
Chromosomal Proteins, Non-Histone/physiology , HIV Long Terminal Repeat/genetics , HIV-1/physiology , Human Immunodeficiency Virus Proteins/physiology , Nucleosomes/physiology , Virus Latency , Acetylation , Chromatin Assembly and Disassembly , Chromosomal Proteins, Non-Histone/antagonists & inhibitors , Chromosomal Proteins, Non-Histone/genetics , Gene Expression Regulation, Viral , HIV-1/genetics , Human Immunodeficiency Virus Proteins/antagonists & inhibitors , Human Immunodeficiency Virus Proteins/genetics , Humans , Jurkat Cells , Models, Genetic , Promoter Regions, Genetic , T-Lymphocytes/virology , Transcriptional Activation , Virus Activation/genetics , tat Gene Products, Human Immunodeficiency Virus/metabolism , tat Gene Products, Human Immunodeficiency Virus/physiology
16.
PLoS One ; 6(11): e27660, 2011.
Article in English | MEDLINE | ID: mdl-22110710

ABSTRACT

Among its many roles, the HIV-1 accessory protein Vpu performs a viroporin function and also antagonizes the host cell restriction factor tetherin through its transmembrane domain. BIT225 is a small molecule inhibitor that specifically targets the Vpu viroporin function, which, in macrophages, resulted in late stage inhibition of virus release and decreased infectivity of released virus, a phenotype similar to tetherin-mediated restriction. Here, we investigated whether BIT225 might mediate its antiviral function, at least in part, via inhibition of Vpu-mediated tetherin antagonism. Using T-cell lines inducible for tetherin expression, we found that BIT225 does not exert its antiviral function by inhibiting Vpu-mediated tetherin downmodulation from the cell surface, the main site of action of tetherin activity. In addition, results from a bioluminescence resonance energy transfer (BRET) assay showed that the Vpu-tetherin interaction was not affected by BIT225. Our data provide support for the concept that tetherin antagonism and viroporin function are separable on the Vpu transmembrane and that viroporin function might be cell-type dependent. Further, this work contributes to the characterization of BIT225 as an inhibitor that specifically targets the viroporin function of Vpu.


Subject(s)
Anti-HIV Agents/pharmacology , Guanidine/pharmacology , Guanidines/pharmacology , HIV-1/physiology , Human Immunodeficiency Virus Proteins/antagonists & inhibitors , Human Immunodeficiency Virus Proteins/pharmacology , Pyrazoles/pharmacology , Viral Regulatory and Accessory Proteins/antagonists & inhibitors , Viral Regulatory and Accessory Proteins/pharmacology , Antigens, CD/metabolism , Cell Line , Cell Membrane/drug effects , Cell Membrane/metabolism , GPI-Linked Proteins/antagonists & inhibitors , GPI-Linked Proteins/metabolism , Gene Expression Regulation/drug effects , HIV-1/drug effects , HIV-1/metabolism , Human Immunodeficiency Virus Proteins/chemistry , Humans , Protein Structure, Tertiary , Viral Regulatory and Accessory Proteins/chemistry , Virus Release/drug effects
17.
Antiviral Res ; 91(3): 321-9, 2011 Sep.
Article in English | MEDLINE | ID: mdl-21777622

ABSTRACT

Bone marrow stromal cell antigen 2 (BST-2, also known as Tetherin) inhibits HIV-1 release and thereby severely impairs viral replication. HIV-1 accessory protein Vpu induces the down-regulation of cell surface BST-2, and counteracts the antiviral function of BST-2. Blocking Vpu-mediated down-regulation of cell surface BST-2 is viewed as a new opportunity for developing anti-HIV drugs. In this study, we have developed a high-throughput cell-based ELISA to identify small molecules that antagonize HIV-1 Vpu function and consequently inhibit HIV-1 replication through rescuing the antiviral activity of host BST-2. This cell-ELISA shows an excellent correlation with results obtained by flow cytometry (FACS). Under optimal conditions, a Z' factor of 0.605 was achieved in a 96-well format. Together, these results demonstrate that this assay can be used to quantify the cell surface level of BST-2 and be adapted to a high-throughput screening for novel anti-HIV compounds.


Subject(s)
Anti-HIV Agents/pharmacology , Antigens, CD/metabolism , HIV-1/drug effects , High-Throughput Screening Assays , Human Immunodeficiency Virus Proteins , Protein Binding/drug effects , RNA, Small Interfering/pharmacology , Stromal Cells/drug effects , Viral Regulatory and Accessory Proteins , Cell Membrane/drug effects , Cell Membrane/metabolism , Cell Membrane/virology , Enzyme-Linked Immunosorbent Assay , Flow Cytometry , GPI-Linked Proteins/agonists , GPI-Linked Proteins/metabolism , Gene Silencing/drug effects , HIV Infections/drug therapy , HIV Infections/metabolism , HIV Infections/pathology , HIV Infections/virology , HIV-1/genetics , HIV-1/metabolism , HeLa Cells , Human Immunodeficiency Virus Proteins/antagonists & inhibitors , Human Immunodeficiency Virus Proteins/genetics , Human Immunodeficiency Virus Proteins/metabolism , Humans , Plasmids , Real-Time Polymerase Chain Reaction , Stromal Cells/cytology , Stromal Cells/metabolism , Transfection , Viral Regulatory and Accessory Proteins/antagonists & inhibitors , Viral Regulatory and Accessory Proteins/genetics , Viral Regulatory and Accessory Proteins/metabolism , Virus Attachment/drug effects , Virus Replication/drug effects
18.
Clin Pharmacokinet ; 50(4): 267-80, 2011 Apr.
Article in English | MEDLINE | ID: mdl-21348539

ABSTRACT

BACKGROUND: Vicriviroc is a next-generation antiretroviral compound that blocks HIV from entering uninfected cells by binding to the virus's cellular co-receptor chemokine receptor 5 (CCR5). A potent inhibitor of HIV infection of human cells both in vitro and in vivo, vicriviroc is in development for use in treatment-naïve HIV-1-infected individuals. These patients often receive antiretroviral therapy regimens that include a ritonavir-enhanced protease inhibitor. Such regimens have a high potential for drug-drug interactions because many of the antiretroviral agents inhibit or induce elements of drug elimination pathways, such as the hepatic cytochromes, which may alter drug concentrations and affect both safety and efficacy. The aim of this set of studies was to determine what, if any, dose adjustments or monitoring would be required to use vicriviroc in regimens containing the most common antiretroviral agents. METHODS: Drug-drug interactions between vicriviroc and 11 other antiretroviral compounds were investigated in fixed-sequence or parallel-group clinical trials lasting 12-35 days. Fixed-sequence studies were conducted with the protease inhibitors atazanavir, darunavir, fosamprenavir, indinavir, nelfinavir, saquinavir and tipranavir. In these studies vicriviroc was administered with ritonavir for a fixed duration, followed by administration of vicriviroc with ritonavir plus the protease inhibitor. Parallel-group studies conducted with lopinavir, zidovudine/lamivudine and tenofovir disoproxil fumarate randomized subjects to receive vicriviroc with or without the study drug. All subjects enrolled in the studies were healthy male and female adults. STATISTICAL METHODS: The log-transformed data for vicriviroc primary pharmacokinetic parameters on appropriate days were statistically analysed using a one-way analysis of variance (ANOVA) model extracting the effects due to treatment. Steady state was evaluated by an ANOVA model on trough concentrations using day and subject as class variables. RESULTS: Vicriviroc exposure was not affected by the concurrently administered antiretroviral drugs in any clinically relevant manner, nor did vicriviroc have a clinically relevant effect on the exposure of other drugs. The drug combinations studied were safe and well tolerated, with most adverse events reported as mild to moderate. Aside from the known toxicities of the other antiretroviral drugs, no clinically relevant changes in blood chemistry, haematological parameters, ECGs or vital signs were associated with either vicriviroc or combination treatment. CONCLUSIONS: No dose modification or monitoring of vicriviroc concentrations is necessary when vicriviroc is co-administered with any of the antiretroviral agents reviewed here. The lack of drug-drug interactions suggests that it will be possible to add vicriviroc at the single clinically prescribed dose level to various background regimens that include a boosted protease inhibitor, with all other drugs also prescribed at their standard doses.


Subject(s)
Anti-Retroviral Agents/pharmacokinetics , CCR5 Receptor Antagonists , HIV Infections/drug therapy , Human Immunodeficiency Virus Proteins/antagonists & inhibitors , Piperazines/pharmacokinetics , Pyrimidines/pharmacokinetics , Adolescent , Adult , Anti-Retroviral Agents/adverse effects , Anti-Retroviral Agents/blood , Anti-Retroviral Agents/therapeutic use , Biological Availability , Drug Interactions , Drug Therapy, Combination/adverse effects , Female , HIV Protease Inhibitors/adverse effects , HIV Protease Inhibitors/blood , HIV Protease Inhibitors/pharmacokinetics , HIV Protease Inhibitors/therapeutic use , Half-Life , Humans , Male , Middle Aged , Piperazines/adverse effects , Piperazines/blood , Piperazines/therapeutic use , Pyrimidines/adverse effects , Pyrimidines/blood , Pyrimidines/therapeutic use , Ritonavir/pharmacology , Young Adult
19.
Chem Soc Rev ; 40(3): 1337-46, 2011 Mar.
Article in English | MEDLINE | ID: mdl-21152581

ABSTRACT

The human immunodeficiency virus (HIV), the causative agent of acquired immunodeficiency syndrome (AIDS), relies heavily on protein-protein interactions in almost every step of its lifecycle. Targeting these interactions, especially those between virus and host proteins, is increasingly viewed as an ideal avenue for the design and development of new therapeutics. In this tutorial review, we outline the lifecycle of HIV and describe some of the protein-protein interactions that control and regulate each step of this process, also detailing efforts to develop therapies that target these interactions.


Subject(s)
HIV/metabolism , Human Immunodeficiency Virus Proteins/metabolism , HIV/enzymology , HIV Envelope Protein gp120/chemistry , HIV Integrase/chemistry , HIV Protease/chemistry , HIV Protease/genetics , HIV Reverse Transcriptase/chemistry , Human Immunodeficiency Virus Proteins/antagonists & inhibitors , Humans , Protein Binding , Virus Replication
20.
Virol Sin ; 25(4): 267-80, 2010 Aug.
Article in English | MEDLINE | ID: mdl-20960300

ABSTRACT

Ion channels are membrane proteins that are found in a number of viruses and which are of crucial physiological importance in the viral life cycle. They have one common feature in that their action mode involves a change of electrochemical or proton gradient across the bilayer lipid membrane which modulates viral or cellular activity. We will discuss a group of viral channel proteins that belong to the viroproin family, and which participate in a number of viral functions including promoting the release of viral particles from cells. Blocking these channel-forming proteins may be "lethal", which can be a suitable and potential therapeutic strategy. In this review we discuss seven ion channels of viruses which can lead serious infections in human beings: M2 of influenza A, NB and BM2 of influenza B, CM2 of influenza C, Vpu of HIV-1, p7 of HCV and 2B of picornaviruses.


Subject(s)
Antiviral Agents/pharmacology , Ion Channels/antagonists & inhibitors , Viral Proteins/antagonists & inhibitors , Human Immunodeficiency Virus Proteins/antagonists & inhibitors , Human Immunodeficiency Virus Proteins/physiology , Humans , Ion Channels/physiology , Viral Matrix Proteins/antagonists & inhibitors , Viral Matrix Proteins/physiology , Viral Nonstructural Proteins/antagonists & inhibitors , Viral Nonstructural Proteins/physiology , Viral Proteins/physiology , Viral Regulatory and Accessory Proteins/antagonists & inhibitors , Viral Regulatory and Accessory Proteins/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...