Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 37
Filter
1.
Biochemistry ; 60(24): 1896-1908, 2021 06 22.
Article in English | MEDLINE | ID: mdl-34096272

ABSTRACT

Feline immunodeficiency virus (FIV) is a veterinary infective agent for which there is currently no efficient drug available. Drugs targeting the lentivirus capsid are currently under development for the treatment of human immunodeficiency virus 1 (HIV-1). Here we describe a lead compound that interacts with the FIV capsid. This compound, 696, modulates the in vitro assembly of and stabilizes the assembled capsid protein. To decipher the mechanism of binding of this compound to the protein, we performed the first nuclear magnetic resonance (NMR) assignment of the FIV p24 capsid protein. Experimental NMR chemical shift perturbations (CSPs) observed after the addition of 696 enabled the characterization of a specific binding site for 696 on p24. This site was further analyzed by molecular modeling of the protein:compound interaction, demonstrating a strong similarity with the binding sites of existing drugs targeting the HIV-1 capsid protein. Taken together, we characterized a promising capsid-interacting compound with a low cost of synthesis, for which derivatives could lead to the development of efficient treatments for FIV infection. More generally, our strategy combining the NMR assignment of FIV p24 with NMR CSPs and molecular modeling will be useful for the analysis of future compounds targeting p24 in the quest to identify an efficient treatment for FIV.


Subject(s)
Antiviral Agents/pharmacology , Benzimidazoles/pharmacology , Gene Products, gag/antagonists & inhibitors , Immunodeficiency Virus, Feline/drug effects , Animals , Binding Sites , Capsid/metabolism , Capsid Proteins/antagonists & inhibitors , Capsid Proteins/metabolism , Cats , Gene Products, gag/metabolism , Immunodeficiency Virus, Feline/metabolism , Lead/pharmacology , Protein Domains
2.
Mar Drugs ; 19(5)2021 Apr 28.
Article in English | MEDLINE | ID: mdl-33925208

ABSTRACT

Walleye dermal sarcoma virus (WDSV) is a type of retrovirus, which affects most of the adult walleye fishes during the spawning time. The virus causes multiple epithelial tumors on the fish's skin and fins that are liable for more than 50% of the mortality rate of fish around the world. Till now, no effective antiviral drug or vaccine candidates have been developed that can block the progression of the disease caused by the pathogen. It was found that the 582-amino-acid (aa) residues long internal structural gag polyprotein of the virus plays an important role in virus budding and virion maturation outside of the cell. Inhibition of the protein can block the budding and virion maturation process and can be developed as an antiviral drug candidate against the virus. Therefore, the study aimed to identify potential natural antiviral drug candidates from the tropical mangrove marine plant Avicennia alba, which will be able to block the budding and virion maturation process by inhibiting the activity of the gag protein of the virus. Initially, a homology modeling approach was applied to identify the 3D structure, followed by refinement and validation of the protein. The refined protein structures were then utilized for molecular docking simulation. Eleven phytochemical compounds have been isolated from the marine plant and docked against the virus gag polyprotein. Three compounds, namely Friedlein (CID244297), Phytosterols (CID12303662), and 1-Triacontanol (CID68972) have been selected based on their docking score -8.5 kcal/mol, -8.0 kcal/mol and -7.9 kcal/mol, respectively, and were evaluated through ADME (Absorption, Distribution, Metabolism and Excretion), and toxicity properties. Finally, molecular dynamics (MD) simulation was applied to confirm the binding stability of the protein-ligands complex structure. The ADME and toxicity analysis reveal the efficacy and non-toxic properties of the compounds, where MD simulation confirmed the binding stability of the selected three compounds with the targeted protein. This computational study revealed the virtuous value of the selected three compounds against the targeted gag polyprotein and will be effective and promising antiviral candidates against the pathogen in a significant and worthwhile manner. Although in vitro and in vivo study is required for further evaluation of the compounds against the targeted protein.


Subject(s)
Antiviral Agents/pharmacology , Avicennia/chemistry , Epsilonretrovirus/drug effects , Fish Diseases/prevention & control , Plant Extracts/pharmacology , Retroviridae Infections/veterinary , Tumor Virus Infections/veterinary , Animals , Antiviral Agents/isolation & purification , Epsilonretrovirus/metabolism , Epsilonretrovirus/pathogenicity , Fish Diseases/virology , Gene Products, gag/antagonists & inhibitors , Gene Products, gag/metabolism , Ligands , Molecular Docking Simulation , Molecular Dynamics Simulation , Plant Extracts/isolation & purification , Protein Conformation , Retroviridae Infections/prevention & control , Retroviridae Infections/virology , Structure-Activity Relationship , Tumor Virus Infections/prevention & control , Tumor Virus Infections/virology , Virus Release/drug effects
3.
Curr Top Microbiol Immunol ; 389: 171-201, 2015.
Article in English | MEDLINE | ID: mdl-25731773

ABSTRACT

The advances made in the treatment of HIV-1 infection represent a major success of modern biomedical research, prolonging healthy life and reducing virus transmission. There remain, however, many challenges relating primarily to side effects of long-term therapy and the ever-present danger of the emergence of drug-resistant strains. To counter these threats, there is a continuing need for new and better drugs, ideally targeting multiple independent steps in the HIV-1 replication cycle. The most successful current drugs target the viral enzymes: protease (PR), reverse transcriptase (RT), and integrase (IN). In this review, we outline the advances made in targeting the Gag protein and its mature products, particularly capsid and nucleocapsid, and highlight possible targets for future pharmacological intervention.


Subject(s)
Acquired Immunodeficiency Syndrome/drug therapy , Anti-HIV Agents/pharmacology , Gene Products, gag/antagonists & inhibitors , HIV-1 , Capsid Proteins/antagonists & inhibitors , Humans , Nucleocapsid/antagonists & inhibitors , Viral Matrix Proteins/antagonists & inhibitors , Virus Replication/drug effects
4.
Nat Med ; 13(5): 631-5, 2007 May.
Article in English | MEDLINE | ID: mdl-17435772

ABSTRACT

Mammalian cells have developed diverse strategies to restrict retroviral infection. Retroviruses have therefore evolved to counteract such restriction factors, in order to colonize their hosts. Tripartite motif-containing 5 isoform-alpha (TRIM5alpha) protein from rhesus monkey (TRIM5alpharh) restricts human immunodeficiency virus type 1 (HIV-1) infection at a postentry, preintegration stage in the viral life cycle, by recognizing the incoming capsid and promoting its premature disassembly. TRIM5alpha comprises an RBCC (RING, B-box 2 and coiled-coil motifs) domain and a B30.2(SPRY) domain. Sequences in the B30.2(SPRY) domain dictate the potency and specificity of the restriction. As TRIM5alpharh targets incoming mature HIV-1 capsid, but not precursor Gag, it was assumed that TRIM5alpharh did not affect HIV-1 production. Here we provide evidence that TRIM5alpharh, but not its human ortholog (TRIM5alphahu), blocks HIV-1 production through rapid degradation of HIV-1 Gag polyproteins. The specificity for this restriction is determined by sequences in the RBCC domain. Our observations suggest that TRIM5alpharh interacts with HIV-1 Gag during or before Gag assembly through a mechanism distinct from the well-characterized postentry restriction. This finding demonstrates a cellular factor blocking HIV-1 production by actively degrading a viral protein. Further understanding of this previously unknown restriction mechanism may reveal new targets for future anti-HIV-1 therapy.


Subject(s)
Gene Products, gag/antagonists & inhibitors , HIV Infections/prevention & control , Proteins/physiology , Virus Replication , Animals , Cell Line , Cells, Cultured , Gene Products, gag/metabolism , HIV-1/physiology , Humans , Kidney , Macaca mulatta , Proteins/genetics , Transfection , Ubiquitin-Protein Ligases
5.
J Virol ; 86(12): 6643-55, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22496222

ABSTRACT

The emergence of resistance to existing classes of antiretroviral drugs necessitates finding new HIV-1 targets for drug discovery. The viral capsid (CA) protein represents one such potential new target. CA is sufficient to form mature HIV-1 capsids in vitro, and extensive structure-function and mutational analyses of CA have shown that the proper assembly, morphology, and stability of the mature capsid core are essential for the infectivity of HIV-1 virions. Here we describe the development of an in vitro capsid assembly assay based on the association of CA-NC subunits on immobilized oligonucleotides. This assay was used to screen a compound library, yielding several different families of compounds that inhibited capsid assembly. Optimization of two chemical series, termed the benzodiazepines (BD) and the benzimidazoles (BM), resulted in compounds with potent antiviral activity against wild-type and drug-resistant HIV-1. Nuclear magnetic resonance (NMR) spectroscopic and X-ray crystallographic analyses showed that both series of inhibitors bound to the N-terminal domain of CA. These inhibitors induce the formation of a pocket that overlaps with the binding site for the previously reported CAP inhibitors but is expanded significantly by these new, more potent CA inhibitors. Virus release and electron microscopic (EM) studies showed that the BD compounds prevented virion release, whereas the BM compounds inhibited the formation of the mature capsid. Passage of virus in the presence of the inhibitors selected for resistance mutations that mapped to highly conserved residues surrounding the inhibitor binding pocket, but also to the C-terminal domain of CA. The resistance mutations selected by the two series differed, consistent with differences in their interactions within the pocket, and most also impaired virus replicative capacity. Resistance mutations had two modes of action, either directly impacting inhibitor binding affinity or apparently increasing the overall stability of the viral capsid without affecting inhibitor binding. These studies demonstrate that CA is a viable antiviral target and demonstrate that inhibitors that bind within the same site on CA can have distinct binding modes and mechanisms of action.


Subject(s)
Anti-HIV Agents/pharmacology , Capsid/drug effects , Gene Products, gag/antagonists & inhibitors , HIV Infections/virology , HIV-1/drug effects , Benzimidazoles/pharmacology , Benzodiazepines/pharmacology , Capsid/metabolism , Cell Line , Gene Products, gag/chemistry , Gene Products, gag/genetics , Gene Products, gag/metabolism , HIV Infections/drug therapy , HIV-1/chemistry , HIV-1/genetics , HIV-1/physiology , Humans , Protein Structure, Tertiary , Virus Assembly/drug effects
6.
Biochemistry ; 49(19): 4006-17, 2010 May 18.
Article in English | MEDLINE | ID: mdl-20387899

ABSTRACT

Assembly of retrovirus particles is promoted by interaction of the Gag polyprotein with RNA. Nonspecific RNA association with the nucleocapsid domain (NC) of Gag induces the dimerization of Gag through protein-protein contacts in the capsid domain (CA), followed by higher order assembly to form the immature virus particle. NMR relaxation studies were conducted to investigate the initial steps of Rous sarcoma virus (RSV) assembly by examining the association with nucleic acid of a fragment of Gag comprising the C-terminal domain of CA (CTD) postulated to mediate Gag dimerization, the spacer region between CA and NC (SP), and NC. This fragment, CTD-SP-NC (residues 394-577), spans the critical SP region and allows assessment of this key Gag-nucleic acid interaction in the context of the Gag polyprotein rather than the isolated domains. Main-chain amide relaxation of CTD-SP-NC was measured in the absence and presence of (GT)(4), an 8-mer DNA oligonucleotide that binds tightly to the polyprotein but is too short to promote Gag dimerization. The results show that the CTD and NC domains tumble independently. In contrast, the two zinc finger domains within NC are rotationally coupled in both the unbound and bound states, even though only the first zinc finger appears to make direct contact with (GT)(4). In addition, the NMR data indicate that SP and flanking residues undergo a conformational exchange process that is slowed in the presence of (GT)(4). This region around SP where relaxation is strongly affected by (GT)(4) binding is nearly identical to the assembly domain defined previously by mutagenesis studies. Other changes in relaxation induced by (GT)(4) implicate conformational perturbations of helices 1 and 4 in CTD. On the basis of the combined data, we propose a model for the promotion of Gag dimerization by RNA association in which NC-RNA binding disrupts an assembly inhibitory, intramolecular interaction involving SP and CTD. Disruption of this intramolecular interaction is proposed to enhance the accessibility of the Gag dimer contact surface and release the assembly domain to promote intermolecular oligomerization.


Subject(s)
Gene Products, gag/chemistry , Gene Products, gag/metabolism , RNA, Viral/chemistry , Rous sarcoma virus/metabolism , Base Sequence , Binding Sites , Gene Products, gag/antagonists & inhibitors , Molecular Sequence Data , Nuclear Magnetic Resonance, Biomolecular , Nucleocapsid/chemistry , Nucleocapsid/metabolism , Protein Structure, Secondary , RNA, Viral/metabolism
7.
Science ; 270(5239): 1194-7, 1995 Nov 17.
Article in English | MEDLINE | ID: mdl-7502043

ABSTRACT

Strategies for the treatment of human immunodeficiency virus-type 1 (HIV-1) infection must contend with the obstacle of drug resistance. HIV-1 nucleocapsid protein zinc fingers are prime antiviral targets because they are mutationally intolerant and are required both for acute infection and virion assembly. Nontoxic disulfide-substituted benzamides were identified that attack the zinc fingers, inactivate cell-free virions, inhibit acute and chronic infections, and exhibit broad antiretroviral activity. The compounds were highly synergistic with other antiviral agents, and resistant mutants have not been detected. Zinc finger-reactive compounds may offer an anti-HIV strategy that restricts drug-resistance development.


Subject(s)
Antiviral Agents/pharmacology , Benzamides/pharmacology , Capsid Proteins , Capsid/metabolism , Disulfides/pharmacology , Gene Products, gag/antagonists & inhibitors , HIV-1/drug effects , Viral Proteins , Zinc Fingers/drug effects , Amino Acid Sequence , Animals , Antiviral Agents/chemistry , Antiviral Agents/pharmacokinetics , Benzamides/chemistry , Benzamides/pharmacokinetics , Biological Availability , Capsid/chemistry , Cell Line , Disulfides/chemistry , Disulfides/pharmacokinetics , Drug Resistance, Microbial , Drug Synergism , Gene Products, gag/chemistry , HIV-1/physiology , Humans , Male , Mice , Molecular Sequence Data , gag Gene Products, Human Immunodeficiency Virus
8.
Curr HIV Res ; 4(3): 379-86, 2006 Jul.
Article in English | MEDLINE | ID: mdl-16842089

ABSTRACT

There is a continued need to develop inexpensive and effective drugs specific for novel targets of human immunodeficiency virus type 1 (HIV-1). The HIV-1 nucleocapsid p7 (NCp7) protein plays a critical role in early and late stages of the virus life cycle and possesses two highly conserved retroviral zinc fingers that are essential for its function. We have previously shown that zinc finger inhibitors (ZFI) based on the S-acyl 2-mercaptobenzamide thioester (SAMT) chemotype specifically target HIV NCp7 and are effective at reducing levels of infectious virus in an HIV-1-transgenic mouse model. Here, we did an initial proof-of-concept study to test the potential of a lead SAMT compound to reduce virus infectivity in the simian immunodeficiency virus (SIV) nonhuman primate model. SAMT-19 had potent antiviral and virucidal effects against the primary pathogenic isolate SIV/DeltaB670 and was non-cytotoxic in vitro. Cynomolgus macaques were infected intrarectally with SIV/DeltaB670 and treated with a low dose of SAMT-19 by continuous infusion from day 8 to day 28 post infection. Monkeys in the treatment group had significantly lower levels of infectious virus in peripheral blood mononuclear cells during the course of therapy as compared to monkeys in the control group, although therapy had no demonstrable effect on virus load. SAMT-19 therapy did not alter liver, kidney or immunologic function and was well tolerated by all treated monkeys. These data demonstrate that SAMT-19 is safe and virucidal in the nonhuman primate model. Further studies directed at optimizing SAMT bioavailability and pharmacokinetics likely will result in enhanced therapeutic efficacy of this promising HIV therapeutic.


Subject(s)
Anti-HIV Agents/therapeutic use , Capsid Proteins/antagonists & inhibitors , Gene Products, gag/antagonists & inhibitors , Simian Acquired Immunodeficiency Syndrome/drug therapy , Sulfhydryl Compounds/therapeutic use , Viral Proteins/antagonists & inhibitors , Zinc Fingers/drug effects , Animals , Leukocytes, Mononuclear/virology , Macaca fascicularis , Male , Simian Acquired Immunodeficiency Syndrome/immunology , Simian Acquired Immunodeficiency Syndrome/virology , T-Lymphocytes/immunology , gag Gene Products, Human Immunodeficiency Virus
9.
Curr Opin Investig Drugs ; 7(8): 759-65, 2006 Aug.
Article in English | MEDLINE | ID: mdl-16955688

ABSTRACT

Panacos Pharmaceuticals Inc is developing the HIV Gag protein and viral maturation inhibitor bevirimat for the potential oral treatment of HIV infection. Phase II clinical trials are underway and phase III trials expected to commence in 2007.


Subject(s)
Gene Products, gag/antagonists & inhibitors , HIV Infections/drug therapy , HIV-1/metabolism , Succinates/pharmacology , Triterpenes/pharmacology , Animals , Clinical Trials, Phase II as Topic , Drug Evaluation, Preclinical , HIV Infections/metabolism , Humans , Succinates/adverse effects , Succinates/pharmacokinetics , Succinates/therapeutic use , Triterpenes/adverse effects , Triterpenes/pharmacokinetics , Triterpenes/therapeutic use
10.
J Med Chem ; 48(8): 2847-58, 2005 Apr 21.
Article in English | MEDLINE | ID: mdl-15828823

ABSTRACT

The HIV-1 nucleocapsid protein (NCp7) is a small basic protein with two CysCysHisCys zinc-binding domains that specifically recognizes the Psi-site of the viral RNA. NCp7 plays a number of crucial roles in the viral lifecycle, including reverse transcription and RNA encapsidation. Several classes of potential anti-HIV compounds have been designed to inactivate NCp7 through zinc ejection, including a special class of thioester compounds. We have investigated the mechanism of action of two N-substituted-S-acyl-2-mercaptobenzamide compounds (compounds 1 and 2) that target NCp7. UV/Visible spectroscopy studies demonstrated that both thioesters were able to eject metal from NCp7. NMR and mass spectroscopy studies showed that the thioester compounds specifically ejected zinc from the carboxyl-terminal zinc-binding domain of NCp7 by covalent modification of Cys(39). Exposure of NCp7 to compounds 1 and 2 destroyed its ability to specifically bind RNA, whereas NCp7 already bound to RNA was protected from zinc ejection by the thioesters. The thiol component of the thioesters (compound 3, 2-mercaptobenzoyl-beta-alaninamide) did not eject zinc from NCp7, but when compound 3 was incubated with acetyl CoA prior to incubation with NCp7, we observed extensive metal ejection. Thus, the thiol released by the reaction of compounds 1 and 2 could be re-acylated in vivo by acyl CoA to form a new thioester compound that is able to react with NCp7. These studies provide a better understanding of the mechanism of action of thioester compounds, which is important for future design of anti-HIV-1 compounds that target NCp7.


Subject(s)
Anti-HIV Agents/chemistry , Benzamides/chemistry , Capsid Proteins/antagonists & inhibitors , Capsid Proteins/chemistry , Gene Products, gag/antagonists & inhibitors , Gene Products, gag/chemistry , Viral Proteins/antagonists & inhibitors , Viral Proteins/chemistry , beta-Alanine/analogs & derivatives , beta-Alanine/chemistry , Amino Acid Sequence , Anti-HIV Agents/chemical synthesis , Benzamides/chemical synthesis , Cysteine/chemistry , Electrophoretic Mobility Shift Assay , Magnetic Resonance Spectroscopy , Mass Spectrometry , Molecular Sequence Data , Spectrophotometry/methods , Sulfhydryl Compounds/chemistry , Zinc/chemistry , beta-Alanine/chemical synthesis , gag Gene Products, Human Immunodeficiency Virus
11.
J Neuroimmunol ; 159(1-2): 196-202, 2005 Feb.
Article in English | MEDLINE | ID: mdl-15652420

ABSTRACT

We analyzed the relationship between the expression of interferon (IFN)-gamma and HTLV-I p19 antigen and activation of p38 mitogen-activated protein kinase (p38 MAPK) in two HTLV-I-infected T cell lines derived from two patients (HCT-1 and HCT-4) with HTLV-I-associated myelopathy/tropical spastic paraparesis (HAM/TSP), and three HTLV-I-infected T cell lines derived from three patients with adult T cell leukemia (ATL). Expression of phosphorylated (activated)-p38 MAPK was markedly increased concomitant with high levels of both IFN-gamma and HTLV-I p19 antigen expression in both HCT-1 and HCT-4 compared with cell lines derived from ATL patients. Treatment with SB203580, a specific inhibitor of p38 MAPK, suppressed IFN-gamma and HTLV-I p19 antigen expression levels in HCT-1, HCT-4 and peripheral blood CD4(+) T cells of HAM/TSP patients. These findings strongly suggest that activation of p38 MAPK signaling pathway is involved in the up-regulation of IFN-gamma expression with high HTLV-I proviral load in HAM/TSP patients.


Subject(s)
Gene Products, gag/biosynthesis , HTLV-I Antigens/biosynthesis , Human T-lymphotropic virus 1/immunology , Interferon-gamma/biosynthesis , MAP Kinase Signaling System/immunology , Paraparesis, Tropical Spastic/immunology , Retroviridae Proteins, Oncogenic/biosynthesis , p38 Mitogen-Activated Protein Kinases/physiology , Adult , Aged , Antiviral Agents/pharmacology , CD4-Positive T-Lymphocytes/drug effects , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/metabolism , CD4-Positive T-Lymphocytes/virology , Cell Line , Female , Gene Products, gag/antagonists & inhibitors , Humans , Imidazoles/pharmacology , Interferon-gamma/antagonists & inhibitors , Male , Middle Aged , Paraparesis, Tropical Spastic/enzymology , Paraparesis, Tropical Spastic/virology , Phosphorylation , Protein Kinase Inhibitors/pharmacology , Proviruses/immunology , Pyridines/pharmacology , Retroviridae Proteins, Oncogenic/antagonists & inhibitors , T-Lymphocytes/drug effects , T-Lymphocytes/enzymology , T-Lymphocytes/immunology , T-Lymphocytes/virology , gag Gene Products, Human Immunodeficiency Virus , p38 Mitogen-Activated Protein Kinases/biosynthesis , p38 Mitogen-Activated Protein Kinases/metabolism
12.
Protein Sci ; 10(7): 1434-45, 2001 Jul.
Article in English | MEDLINE | ID: mdl-11420445

ABSTRACT

A diverse set of electrophilic compounds that react with cysteine thiolates in retroviral nucleocapsid (NC) proteins and abolish virus infectivity has been identified. Although different in chemical composition, these compounds are all oxidizing agents that lead to the ejection of Zn(II) ions bound to conserved structural motifs (zinc fingers) present in retroviral NC proteins. The reactivity of a congeneric series of aromatic disulfides toward the NC protein of the human immunodeficiency virus type 1 (HIV-1), NCp7, has been characterized by HPLC separation of starting reagents from reaction products. We calculated the absolute redox potentials of these compounds in the gas phase and in aqueous solvent, using a density functional theory method and a continuum solvation model. Pulsed polarography experiments were performed and showed a direct correlation between calculated and experimentally determined redox propensities. A dependence between protein reactivity and redox potential for a specific compound was shown: Reaction with NCp7 did not take place below a threshold value of redox potential. This relationship permits the distinction between active and nonactive compounds targeted against NCp7, and provides a theoretical basis for a scale of reactivity with retroviral zinc fingers. Our results indicate that electrophilic agents with adequate thiophilicity to react with retroviral NC fingers can now be designed using known or calculated electrochemical properties. This may assist in the design of antiretroviral compounds with greater specificity for NC protein. Such electrophilic agents can be used in retrovirus inactivation with the intent of preparing a whole-killed virus vaccine formulation that exhibits unaffected surface antigenic properties.


Subject(s)
Anti-HIV Agents/chemistry , Capsid Proteins , Retroviridae Proteins/antagonists & inhibitors , Viral Proteins , Zinc Fingers/drug effects , Anti-HIV Agents/pharmacology , Capsid/antagonists & inhibitors , Capsid/chemistry , Capsid/metabolism , Disulfides/chemistry , Disulfides/pharmacology , Electrochemistry , Gene Products, gag/antagonists & inhibitors , Gene Products, gag/chemistry , Gene Products, gag/metabolism , Humans , Kinetics , Nucleocapsid Proteins/antagonists & inhibitors , Nucleocapsid Proteins/chemistry , Nucleocapsid Proteins/metabolism , Oxidation-Reduction , Quantitative Structure-Activity Relationship , Retroviridae Proteins/chemistry , Retroviridae Proteins/metabolism , gag Gene Products, Human Immunodeficiency Virus
13.
Biochimie ; 79(11): 673-80, 1997 Nov.
Article in English | MEDLINE | ID: mdl-9479450

ABSTRACT

The Gag-encoded nucleocapsid protein NCp7 (72 amino acids) from HIV-1, the regulatory protein, Vpr (96 amino acids) and numerous derivatives have been synthesized by solid phase method and their structures determined by 2D NMR. In NCp7, the two highly folded zinc fingers of the Cx2Cx4Hx4C type are in close spacial proximity and the replacement of H by C in the first zinc finger or P by L in the short interdigital domain led to structural modifications evidenced by NMR. In vivo, these point mutations induced a complete loss of viral infectivity by interrupting critical step(s) of the retroviral life cycle. To account for these findings, a model of the complex between NCp7 and d (ACGCC) has been proposed from NMR data, showing the intercalation of Trp37 in the oligonucleotide. This model could also explain the role of NCp7 in the formation of viral particles and agrees with the modifications in morphology of the virions containing mutations in the NCp7 zinc fingers. Vpr is essentially constituted by two long helical domains at its N- and C-terminals and the side chains of Leu60 and Leu67 participate in a leucine-zipper mode of intramolecular interaction. The results obtained have been used to try to develop new antiviral agents inhibiting NCp7 functions and thus possibly devoid of the resistance effects found with inhibitors of HIV enzymes (reverse transcriptase and protease).


Subject(s)
Capsid Proteins , Capsid/chemistry , Capsid/metabolism , Gene Products, gag/chemistry , Gene Products, gag/metabolism , Gene Products, vpr/chemistry , Gene Products, vpr/metabolism , HIV-1/chemistry , Viral Proteins , Amino Acid Sequence , Animals , Capsid/antagonists & inhibitors , Gene Products, gag/antagonists & inhibitors , Gene Products, vpr/antagonists & inhibitors , Humans , Models, Molecular , Molecular Sequence Data , Structure-Activity Relationship , gag Gene Products, Human Immunodeficiency Virus , vpr Gene Products, Human Immunodeficiency Virus
14.
J Med Chem ; 45(24): 5249-59, 2002 Nov 21.
Article in English | MEDLINE | ID: mdl-12431052

ABSTRACT

The interaction of a series gag peptide analogues with human cyclophilin A (hCypA) have been studied employing molecular docking and 3D-QSAR approaches. The Lamarckian Genetic Algorithm (LGA) and divide-and-conquer methods were applied to locate the binding orientations and conformations of the inhibitors interacting with hCypA. Good correlations between the calculated interaction free energies and experimental inhibitory activities suggest that the binding conformations of these inhibitors are reasonable. A novel interaction model was identified for inhibitors 11, 15, and 17 whose N-termini were modified by addition of the deaminovaline (Dav) group and the C-termini of 15 and 17 were modified by addition of a benzyl group. Accordingly, two new binding sites (sites A and D in Figure 1) were revealed, which show a strong correlation with inhibitor potency and thus can be used as a starting point for new inhibitor design. In addition, two predictive 3D-QSAR models were obtained by CoMFA and CoMSIA analyses based on the binding conformations derived from the molecular docking calculations. The reasonable r(cross)(2) (cross-validated) values 0.738 and 0.762 were obtained for CoMFA and CoMSIA models, respectively. The predictive ability of these models was validated by four peptide analogues test set. The CoMFA and CoMSIA field distributions are in general agreement with the structural characteristics of the binding groove of hCypA. This indicates the reasonableness of the binding model of the inhibitors with hCypA. Considering all these results together with the valuable clues of binding from references published recently, reasonable pharmacophore elements have been suggested, demonstrating that the 3D-QSAR models about peptide analogue inhibitors are expected to be further employed in predicting activities of the novel compounds for inhibiting hCypA.


Subject(s)
Cyclophilin A/chemistry , Gene Products, gag/chemistry , Oligopeptides/chemistry , Binding Sites , Gene Products, gag/antagonists & inhibitors , Humans , Molecular Conformation , Oligopeptides/antagonists & inhibitors , Quantitative Structure-Activity Relationship , Thermodynamics
15.
J Med Chem ; 42(1): 67-86, 1999 Jan 14.
Article in English | MEDLINE | ID: mdl-9888834

ABSTRACT

Nucleocapsid p7 protein (NCp7) zinc finger domains of the human immunodeficiency virus type 1 (HIV-1) are being developed as antiviral targets due to their key roles in viral replication and their mutationally nonpermissive nature. On the basis of our experience with symmetrical disulfide benzamides (DIBAs; Rice et al. Science 1995, 270, 1194-1197), we synthesized and evaluated variants of these dimers, including sets of 4,4'- and 3,3'-disubstituted diphenyl sulfones and their monomeric benzisothiazolone derivatives (BITA). BITAs generally exhibited diminished antiviral potency when compared to their disulfide precursors. Novel, monomeric structures were created by linking haloalkanoyl groups to the benzamide ring through -NH-C(=O)- (amide) or -S-C(=O)- (thiolester) bridges. Amide-linked compounds generally lacked antiviral activity, while haloalkanoyl thiolesters and non-halogen-bearing analogues frequently exhibited acceptable antiviral potency, thus establishing thiolester benzamides per se as a new anti-HIV chemotype. Pyridinioalkanoyl thiolesters (PATEs) exhibited superior anti-HIV-1 activity with minimal cellular toxicity and appreciable water solubility. PATEs were shown to preferentially target the NCp7 Zn finger when tested against other molecular targets, thus identifying thiolester benzamides, and PATEs in particular, as novel NCp7 Zn finger inhibitors for in vivo studies.


Subject(s)
Anti-HIV Agents/chemical synthesis , Capsid Proteins , Capsid/antagonists & inhibitors , Gene Products, gag/antagonists & inhibitors , HIV-1/drug effects , Pyridinium Compounds/chemical synthesis , Sulfonamides/chemical synthesis , Sulfones/chemical synthesis , Viral Proteins , Zinc Fingers , Animals , Anti-HIV Agents/chemistry , Anti-HIV Agents/pharmacology , Cell Line , HIV-1/metabolism , Ligands , Mice , Models, Molecular , Pyridinium Compounds/chemistry , Pyridinium Compounds/pharmacology , Structure-Activity Relationship , Sulfonamides/chemistry , Sulfonamides/pharmacology , Sulfones/chemistry , Sulfones/pharmacology , Virus Replication/drug effects , gag Gene Products, Human Immunodeficiency Virus
16.
J Med Chem ; 41(9): 1371-81, 1998 Apr 23.
Article in English | MEDLINE | ID: mdl-9554870

ABSTRACT

Agents that target the two highly conserved Zn fingers of the human immunodeficiency virus (HIV) nucleocapsid p7 (NCp7) protein are under development as antivirals. These agents covalently modify Zn-coordinating cysteine thiolates of the fingers, causing Zn ejection, loss of native protein structure and nucleic acid binding capacity, and disruption of virus replication. Concentrations of three antiviral agents that promoted in vitro Zn ejection from NCp7 and inhibited HIV replication did not impact the functions of cellular Zn finger proteins, including poly(ADP-ribose) polymerase and the Sp1 and GATA-1 transcription factors, nor did the compounds inhibit HeLa nuclear extract mediated transcription. Selectivity of interactions of these agents with NCp7 was supported by molecular modeling analysis which (1) identified a common saddle-shaped nucleophilic region on the surfaces of both NCp7 Zn fingers, (2) indicated a strong correspondence between computationally docked positions for the agents tested and overlap of frontier orbitals within the nucleophilic loci of the NCp7 Zn fingers, and (3) revealed selective steric exclusion of the agents from the core of the GATA-1 Zn finger. Further modeling analysis suggests that the thiolate of Cys49 in the carboxy-terminal finger is the site most susceptible to electrophilic attack. These data provide the first experimental evidence and rationale for antiviral agents that selectively target retroviral nucleocapsid protein Zn fingers.


Subject(s)
Anti-HIV Agents/pharmacology , Capsid Proteins , Capsid/metabolism , Gene Products, gag/metabolism , HIV-1/drug effects , Viral Proteins , Zinc Fingers , Animals , Anti-HIV Agents/metabolism , Azo Compounds/metabolism , Azo Compounds/pharmacology , Benzamides/metabolism , Benzamides/pharmacology , Binding Sites , Capsid/antagonists & inhibitors , Capsid/chemistry , Cell Line , DNA-Binding Proteins/chemistry , DNA-Binding Proteins/metabolism , Erythroid-Specific DNA-Binding Factors , GATA1 Transcription Factor , Gene Products, gag/antagonists & inhibitors , Gene Products, gag/chemistry , HIV-1/metabolism , HIV-1/physiology , Host Cell Factor C1 , Humans , Ligands , Mice , Models, Molecular , Nitroso Compounds/metabolism , Nitroso Compounds/pharmacology , Octamer Transcription Factor-1 , Poly(ADP-ribose) Polymerases/chemistry , Poly(ADP-ribose) Polymerases/metabolism , Sp1 Transcription Factor/chemistry , Sp1 Transcription Factor/metabolism , Sulfones/metabolism , Sulfones/pharmacology , Transcription Factors/chemistry , Transcription Factors/metabolism , Transcription, Genetic/drug effects , Virus Replication/drug effects , gag Gene Products, Human Immunodeficiency Virus
17.
AIDS Res Hum Retroviruses ; 19(2): 91-101, 2003 Feb.
Article in English | MEDLINE | ID: mdl-12639244

ABSTRACT

Control of human immunodeficiency virus through the use of inexpensive chemotherapeutics, with minimal side effects and decreased potential for engendering resistant virus, is a long-term therapeutic goal. In principle, this goal can be accomplished if viral replication in reservoirs of chronically and latently infected cells is addressed. As a first step, we have developed novel antiviral compounds based on a 2-mercaptobenzamide thioester chemotype, including the pyridinioalkanoyl thioesters, which specifically target the zinc fingers of the human immunodeficiency virus nucleocapsid protein (NCp7). Using these compounds in a murine transgenic model, in which infectious human immunodeficiency virus is induced from an integrated provirus, we show inhibition of transgenic spleen cell p24 expression with potencies comparable to acute infection assays using human peripheral blood lymphocytes. More importantly, transgenic mice treated in vivo with two 2-mercaptobenzamide thioesters expressed significantly lower plasma p24, and splenocytes from these animals produced fewer infectious virions. Thus, these thioesters may provide an effective means for inhibiting the expression of human immunodeficiency virus from integrated viral reservoirs.


Subject(s)
Anti-HIV Agents/pharmacology , Benzamides/pharmacology , Capsid Proteins , Esters/pharmacology , Gene Products, gag/antagonists & inhibitors , HIV-1/drug effects , Sulfhydryl Compounds/pharmacology , Viral Proteins , Zinc Fingers/drug effects , Animals , Benzamides/chemistry , Capsid/chemistry , Disease Models, Animal , Esters/chemistry , Gene Products, gag/chemistry , HIV Infections/drug therapy , HIV Infections/virology , HIV-1/genetics , Humans , Mice , Microbial Sensitivity Tests/methods , Pyridinium Compounds/chemistry , Pyridinium Compounds/pharmacology , Sulfhydryl Compounds/chemistry , gag Gene Products, Human Immunodeficiency Virus
18.
Expert Rev Anti Infect Ther ; 1(1): 97-128, 2003 Jun.
Article in English | MEDLINE | ID: mdl-15482105

ABSTRACT

There are presently 42 million people worldwide living with HIV/AIDS, the majority of which have limited access to antiretrovirals. Even if worldwide penetration was possible, our current chemotherapeutic strategies still suffer from issues of cost, patient compliance, deleterious acute and chronic side effects, emerging single and multidrug resistance, and generalized treatment and economic issues. Even our best antiretroviral therapeutic strategy, highly active antiretroviral therapy (HAART), falls short of completely suppressing HIV replication. Therefore, expansion of current therapeutic options by discovering new antiretrovirals and targets will be critical in the coming years. This review addresses the current status of reverse transcriptase and protease inhibitor development, and summarizes the progress in emerging classes of HIV inhibitors, including entry (T-20, T-1249), coreceptor (SCH-C, SCH-D), integrase (beta-Diketos) and p7 nucleocapsid Zn finger inhibitors (thioesters and PATEs). In addition, the processes of virus entry, PIC transport to the nucleus, HIV interaction with nuclear pores, Tat function, Rev function and virus budding (Tsg101 and ubiquitination) are examined, and proof of concept inhibitors and potential antiviral targets discussed.


Subject(s)
Acquired Immunodeficiency Syndrome/drug therapy , Anti-HIV Agents/pharmacology , Anti-HIV Agents/therapeutic use , Antiretroviral Therapy, Highly Active , Capsid Proteins/antagonists & inhibitors , Gene Products, gag/antagonists & inhibitors , HIV Integrase Inhibitors/pharmacology , Humans , Membrane Fusion/drug effects , Receptors, HIV/antagonists & inhibitors , Reverse Transcriptase Inhibitors/pharmacology , Viral Proteins/antagonists & inhibitors , Virus Assembly/drug effects , gag Gene Products, Human Immunodeficiency Virus
19.
Clin Perinatol ; 21(1): 39-73, 1994 Mar.
Article in English | MEDLINE | ID: mdl-8013186

ABSTRACT

In the past 10 years, a large number of investigators have produced an enormous amount of information concerning the molecular biology of HIV. These studies at the most basic biological level have provided essential insights into the pathogenesis of the disease. They have supplied the information necessary for the creation of the antiviral therapies now available and have indicated the direction for the development of new therapies now in clinical trials and under investigation. Although the relatively ineffective therapies currently available serve as a constant source of disappointment for those practitioners who care for HIV-infected patients, there is some comfort to be gained from the rapid pace of investigation into the basic biology of the virus and the certainty that any more effective therapy must build upon the basic biological knowledge already obtained. A detailed study of some of the unique features observed during pediatric and perinatal HIV infection, particularly the relatively shortened time from infection to symptoms and the relative importance of CNS disease, may suggest new therapeutic approaches that will benefit both adult and pediatric patients. Finally, a comprehensive knowledge of HIV biology is an essential requirement for therapeutic maneuvers designed to interrupt the transmission of HIV from mother to child.


Subject(s)
DNA, Viral , Gene Products, env , Gene Products, gag , Gene Products, pol , HIV , Viral Regulatory and Accessory Proteins , Female , Gene Expression Regulation, Viral , Gene Products, env/antagonists & inhibitors , Gene Products, env/chemistry , Gene Products, env/drug effects , Gene Products, env/genetics , Gene Products, env/ultrastructure , Gene Products, gag/antagonists & inhibitors , Gene Products, gag/chemistry , Gene Products, gag/drug effects , Gene Products, gag/genetics , Gene Products, gag/ultrastructure , Gene Products, pol/antagonists & inhibitors , Gene Products, pol/chemistry , Gene Products, pol/drug effects , Gene Products, pol/genetics , Gene Products, pol/ultrastructure , HIV/chemistry , HIV/genetics , HIV/growth & development , HIV/physiology , HIV/ultrastructure , HIV Infections/congenital , HIV Infections/microbiology , HIV Infections/prevention & control , HIV Infections/transmission , Humans , Infant, Newborn , Molecular Biology , Pregnancy , Pregnancy Complications, Infectious/microbiology , Pregnancy Complications, Infectious/prevention & control , Regulatory Sequences, Nucleic Acid , Time Factors , Trans-Activators/chemistry , Trans-Activators/genetics , Trans-Activators/ultrastructure , Transcription, Genetic , Viral Regulatory and Accessory Proteins/chemistry , Viral Regulatory and Accessory Proteins/drug effects , Viral Regulatory and Accessory Proteins/genetics , Viral Regulatory and Accessory Proteins/ultrastructure , Virion/chemistry , Virion/genetics , Virion/growth & development , Virion/physiology , Virion/ultrastructure , Virus Integration , Virus Replication
20.
J Virol Methods ; 173(1): 92-8, 2011 Apr.
Article in English | MEDLINE | ID: mdl-21277903

ABSTRACT

Since the discovery of RNAi technology, several functional genomic and disease therapy studies have been conducted using this technique in the field of oncology and virology. RNAi-based antiviral therapies are being studied for the treatment of retroviruses such as HIV-1. These studies include the silencing of regulatory, infectivity and structural genes. The HTLV-1 structural genes are responsible for the synthesis of proteins involved in the entry, assembly and release of particles during viral infection. To examine the possibility of silencing HTLV-1 genes gag and env by RNA interference technology, these genes were cloned into reporter plasmids. These vectors expressed the target mRNAs fused to EGFP reporter genes. Three small interference RNAs (siRNAs) corresponding to gag and three corresponding to env were designed to analyze the effect of silencing by RNAi technology. The plasmids and siRNAs were co-transfected into HEK 293 cells. The results demonstrated that the expression of the HTLV-1 gag and env genes decreased significantly in vitro. Thus, siRNAs can be used to inhibit HTLV-1 structural genes in transformed cells, which could provide a tool for clarifying the roles of HTLV-1 structural genes, as well as a therapy for this infection.


Subject(s)
Gene Products, env/antagonists & inhibitors , Gene Products, gag/antagonists & inhibitors , Gene Silencing , Human T-lymphotropic virus 1/genetics , RNA, Small Interfering/genetics , Artificial Gene Fusion , Gene Products, env/genetics , Gene Products, gag/genetics , Genes, Reporter , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , HEK293 Cells , Humans
SELECTION OF CITATIONS
SEARCH DETAIL