Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 44
Filter
1.
Acta Pharmacol Sin ; 40(6): 850-858, 2019 Jun.
Article in English | MEDLINE | ID: mdl-30796354

ABSTRACT

Serine/threonine phosphatase (Stp1) is a member of the bacterial Mg2+- or Mn2+- dependent protein phosphatase/protein phosphatase 2C family, which is involved in the regulation of Staphylococcus aureus virulence. Aurintricarboxylic acid (ATA) is a known Stp1 inhibitor with an IC50 of 1.03 µM, but its inhibitory mechanism has not been elucidated in detail because the Stp1-ATA cocrystal structure has not been determined thus far. In this study, we performed 400 ns molecular dynamics (MD) simulations of the apo-Stp1 and Stp1-ATA complex models. During MD simulations, the flap subdomain of the Stp1-ATA complex experienced a clear conformational transition from an open state to a closed state, whereas the flap domain of apo-Stp1 changed from an open state to a semi-open state. In the Stp1-ATA complex model, the hydrogen bond (H-bond) between D137 and N142 disappeared, whereas critical H-bond interactions were formed between Q160 and H13, Q160/R161 and ATA, as well as N162 and D198. Finally, four residues (D137, N142, Q160, and R161) in Stp1 were mutated to alanine and the mutant enzymes were assessed using phosphate enzyme activity assays, which confirmed their important roles in maintaining Stp1 activity. This study indicated the inhibitory mechanism of ATA targeting Stp1 using MD simulations and sheds light on the future design of allosteric Stp1 inhibitors.


Subject(s)
Aurintricarboxylic Acid/metabolism , Bacterial Proteins/antagonists & inhibitors , Enzyme Inhibitors/metabolism , Phosphoprotein Phosphatases/antagonists & inhibitors , Staphylococcus aureus/enzymology , Amino Acid Sequence , Aurintricarboxylic Acid/chemistry , Bacterial Proteins/chemistry , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Catalytic Domain , Enzyme Inhibitors/chemistry , Hydrogen Bonding , Molecular Dynamics Simulation , Mutation , Phosphoprotein Phosphatases/chemistry , Phosphoprotein Phosphatases/genetics , Phosphoprotein Phosphatases/metabolism , Protein Binding , Protein Conformation , Sequence Alignment
2.
J Biol Chem ; 290(3): 1623-38, 2015 Jan 16.
Article in English | MEDLINE | ID: mdl-25477506

ABSTRACT

Pharmacological induction of proteotoxic stress is rapidly emerging as a promising strategy for cancer cell-directed chemotherapeutic intervention. Here, we describe the identification of a novel drug-like heat shock response inducer for the therapeutic induction of proteotoxic stress targeting malignant human melanoma cells. Screening a focused library of compounds containing redox-directed electrophilic pharmacophores employing the Stress & Toxicity PathwayFinder(TM) PCR Array technology as a discovery tool, a drug-like triphenylmethane-derivative (aurin; 4-[bis(p-hydroxyphenyl)methylene]-2,5-cyclohexadien-1-one) was identified as an experimental cell stress modulator that causes (i) heat shock factor transcriptional activation, (ii) up-regulation of heat shock response gene expression (HSPA6, HSPA1A, DNAJB4, HMOX1), (iii) early unfolded protein response signaling (phospho-PERK, phospho-eIF2α, CHOP (CCAAT/enhancer-binding protein homologous protein)), (iv) proteasome impairment with increased protein-ubiquitination, and (v) oxidative stress with glutathione depletion. Fluorescence polarization-based experiments revealed that aurin displays activity as a geldanamycin-competitive Hsp90α-antagonist, a finding further substantiated by molecular docking and ATPase inhibition analysis. Aurin exposure caused caspase-dependent cell death in a panel of human malignant melanoma cells (A375, G361, LOX-IMVI) but not in non-malignant human skin cells (Hs27 fibroblasts, HaCaT keratinocytes, primary melanocytes) undergoing the aurin-induced heat shock response without impairment of viability. Aurin-induced melanoma cell apoptosis depends on Noxa up-regulation as confirmed by siRNA rescue experiments demonstrating that siPMAIP1-based target down-regulation suppresses aurin-induced cell death. Taken together, our data suggest feasibility of apoptotic elimination of malignant melanoma cells using the quinone methide-derived heat shock response inducer aurin.


Subject(s)
Adaptor Proteins, Vesicular Transport/metabolism , Apoptosis , Aurintricarboxylic Acid/analogs & derivatives , Heat-Shock Proteins/metabolism , Melanoma/drug therapy , Skin Neoplasms/drug therapy , Adaptor Proteins, Signal Transducing , Aurintricarboxylic Acid/chemistry , Cell Line, Tumor , Cell Survival , Drug Screening Assays, Antitumor , Flow Cytometry , Glutathione/metabolism , Heat-Shock Response/genetics , Humans , Indolequinones/chemistry , Inhibitory Concentration 50 , Keratinocytes/drug effects , Melanocytes/drug effects , Membrane Potential, Mitochondrial , Models, Molecular , Oxidative Stress , Polymerase Chain Reaction , RNA, Small Interfering/metabolism , Up-Regulation
3.
World J Microbiol Biotechnol ; 32(10): 163, 2016 Oct.
Article in English | MEDLINE | ID: mdl-27562597

ABSTRACT

Yersinia sp. bacteria owe their viability and pathogenic virulence to the YopH factor, which is a highly active bacterial protein tyrosine phosphatase. Inhibition of YopH phosphatase results in the lack of Yersinia sp. pathogenicity. We have previously described that aurintricarboxylic acid inhibits the activity of YopH at nanomolar concentrations and represents a unique mechanism of YopH inactivation due to a redox process. This work is a continuation of our previous studies. Here we show that modifications of the structure of aurintricarboxylic acid reduce the ability to inactivate YopH and lead to higher cytotoxicity. In the present paper we examine the inhibitory properties of aurintricarboxylic acid analogues, such as eriochrome cyanine R (ECR) and pararosaniline. Computational docking studies we report here indicate that ATA analogues are not precluded to bind in the YopH active site and in all obtained binding conformations ECR and pararosaniline bind to YopH active site. The free binding energy calculations show that ECR has a stronger binding affinity to YopH than pararosaniline, which was confirmed by experimental YopH enzymatic activity studies. We found that ATA analogues can reversibly reduce the enzymatic activity of YopH, but possess weaker inhibitory properties than ATA. The ATA analogues induced inactivation of YopH is probably due to oxidative mechanism, as pretreatment with catalase prevents from inhibition. We also found that ATA analogues significantly decrease the viability of macrophage cells, especially pararosaniline, while ATA reveals only slight effect on cell viability.


Subject(s)
Aurintricarboxylic Acid/analogs & derivatives , Bacterial Outer Membrane Proteins/chemistry , Benzenesulfonates/chemistry , Protein Tyrosine Phosphatases/chemistry , Rosaniline Dyes/chemistry , Toluidines/chemistry , Yersinia/drug effects , Animals , Aurintricarboxylic Acid/chemistry , Aurintricarboxylic Acid/pharmacology , Bacterial Outer Membrane Proteins/antagonists & inhibitors , Benzenesulfonates/pharmacology , Catalytic Domain/drug effects , Cell Line , Cell Survival/drug effects , Mice , Models, Molecular , Molecular Docking Simulation , Molecular Dynamics Simulation , Molecular Structure , Protein Binding , Protein Tyrosine Phosphatases/antagonists & inhibitors , Rosaniline Dyes/pharmacology , Toluidines/pharmacology , Yersinia/enzymology
4.
Mol Biol Cell ; 17(11): 4632-44, 2006 Nov.
Article in English | MEDLINE | ID: mdl-16928960

ABSTRACT

A cornerstone of the antiviral interferon response is phosphorylation of eukaryotic initiation factor (eIF)2alpha. This limits the availability of eIF2.GTP.Met-tRNA(i)(Met) ternary complexes, reduces formation of 43S preinitiation complexes, and blocks viral (and most cellular) mRNA translation. However, many viruses have developed counterstrategies that circumvent this cellular response. Herein, we characterize a novel class of translation initiation inhibitors that block ternary complex formation and prevent the assembly of 43S preinitiation complexes. We find that translation driven by the HCV IRES is refractory to inhibition by these compounds at concentrations that effectively block cap-dependent translation in vitro and in vivo. Analysis of initiation complexes formed on the HCV IRES in the presence of inhibitor indicates that eIF2alpha and Met-tRNA(i)(Met) are present, defining a tactic used by HCV to evade part of the antiviral interferon response.


Subject(s)
Eukaryotic Initiation Factor-2/metabolism , Guanosine Triphosphate/metabolism , Hepacivirus/genetics , Protein Biosynthesis/genetics , RNA, Transfer, Met/metabolism , Animals , Aurintricarboxylic Acid/chemistry , Aurintricarboxylic Acid/pharmacology , Hepacivirus/drug effects , Mice , Models, Genetic , Protein Biosynthesis/drug effects , Protein Synthesis Inhibitors/chemistry , Protein Synthesis Inhibitors/pharmacology , Regulatory Sequences, Nucleic Acid/genetics , Ribosomes/drug effects , Ribosomes/metabolism
5.
Pharm Dev Technol ; 14(4): 341-2, 2009.
Article in English | MEDLINE | ID: mdl-19235046

ABSTRACT

Factors associated with the intravaginal release of an anti-HIV agent from an alginate complex were considered. Among these is citrate associated with prostatic fluid. This study demonstrates that citrate, at a physiologically appropriate concentration, facilitates the release of an anti-HIV polymer from a calcium alginate complex. The release of the agent can be modified by the concentration of the calcium and alginate in the complex. These results suggest that seminal and prostatic fluid can be considered in the design of an intravaginal system for HIV chemoprophylaxis.


Subject(s)
Alginates/chemistry , Anti-HIV Agents/chemistry , Aurintricarboxylic Acid/chemistry , Citrates/chemistry , Administration, Intravaginal , Anti-HIV Agents/administration & dosage , Aurintricarboxylic Acid/administration & dosage , Chemistry, Pharmaceutical , Drug Carriers/chemistry , Glucuronic Acid/chemistry , HIV Infections/prevention & control , Hexuronic Acids/chemistry , Sodium Citrate , Solubility
6.
J Med Chem ; 62(3): 1677-1683, 2019 02 14.
Article in English | MEDLINE | ID: mdl-30562026

ABSTRACT

We identify three submicromolar inhibitors with new chemical scaffolds for cystathionine γ-lyase (CSE) by a tandem-well-based high-throughput assay. NSC4056, the most potent inhibitor with an IC50 of 0.6 µM, which is also known as aurintricarboxylic acid, selectively binds to Arg and Tyr residues of CSE active site and preferably inhibits the CSE activity in cells rather than cystathionine ß-synthase (CBS), the other H2S-generating enzyme. Moreover, NSC4056 effectively rescues hypotension in hemorrhagic shock rats.


Subject(s)
Aurintricarboxylic Acid/pharmacology , Cystathionine gamma-Lyase/antagonists & inhibitors , Enzyme Inhibitors/pharmacology , Animals , Aurintricarboxylic Acid/chemistry , Aurintricarboxylic Acid/metabolism , Catalytic Domain/drug effects , Cystathionine gamma-Lyase/chemistry , Cystathionine gamma-Lyase/metabolism , Drug Discovery , Enzyme Inhibitors/metabolism , HEK293 Cells , Humans , Male , Mice , Molecular Docking Simulation , Molecular Structure , Nitroquinolines/pharmacology , Protein Binding , RAW 264.7 Cells , Rats, Sprague-Dawley , Structure-Activity Relationship
7.
SLAS Discov ; 23(10): 1083-1091, 2018 12.
Article in English | MEDLINE | ID: mdl-29958052

ABSTRACT

CC-chemokine receptor 7 (CCR7) is a G protein-coupled receptor expressed on a variety of immune cells. CCR7 plays a critical role in the migration of lymphocytes into secondary lymphoid tissues. CCR7 expression, however, has been linked to numerous disease states. Due to its therapeutic relevance and absence of available CCR7 inhibitors, we undertook a high-throughput screen (HTS) to identify small-molecule antagonists of the receptor. Here, we describe a robust HTS approach using a commercially available ß-galactosidase enzyme fragment complementation system and confirmatory transwell chemotaxis assays. This work resulted in the identification of several compounds with activity against CCR7. The most potent of these was subsequently determined to be cosalane, a cholesterol derivative previously designed as a therapeutic for human immunodeficiency virus. Cosalane inhibited both human and murine CCR7 in response to both CCL19 and CCL21 agonists at physiologic concentrations. Furthermore, cosalane produced durable inhibition of the receptor following a cellular incubation period with subsequent washout. Overall, our work describes the development of an HTS-compatible assay, completion of a large HTS campaign, and demonstration for the first time that cosalane is a validated CCR7 antagonist. These efforts could pave the way for new approaches to address CCR7-associated disease processes.


Subject(s)
Aurintricarboxylic Acid/analogs & derivatives , High-Throughput Screening Assays , Receptors, CCR7/antagonists & inhibitors , Signal Transduction/drug effects , Animals , Aurintricarboxylic Acid/chemistry , Aurintricarboxylic Acid/pharmacology , Cell Line , Chemotaxis/drug effects , Drug Design , Humans , Ligands , Mice , Molecular Structure , Receptors, CCR7/chemistry , Receptors, G-Protein-Coupled/antagonists & inhibitors , Receptors, G-Protein-Coupled/chemistry , Structure-Activity Relationship , T-Lymphocytes/drug effects , T-Lymphocytes/metabolism
8.
Anticancer Res ; 37(9): 4799-4806, 2017 09.
Article in English | MEDLINE | ID: mdl-28870898

ABSTRACT

BACKGROUND: Rapidly-dividing cancer cells have higher requirement for iron compared to non-transformed cells, making iron chelating a potential anticancer strategy. In the present study we compared the anticancer activity of uncommon iron chelator aurintricarboxylic acid (ATA) with the known deferoxamine (DFO). MATERIALS AND METHODS: We investigated the impact of ATA and DFO on the viability and proliferation of MCF-7 cancer cells. Moreover we performed enzymatic activity assays and computational analysis of the ATA and DFO effects on pro-oncogenic phosphatases PTP1B and SHP2. RESULTS: ATA and DFO decrease the viability and proliferation of breast cancer cells, but only ATA considerably reduces the activity of PTP1B and SHP2 phosphatases. Our studies indicated that ATA strongly inactivates and binds in the PTP1B and SHP2 active site, interacting with arginine residue essential for enzyme activity. CONCLUSION: We confirmed that iron chelating can be considered as a potential strategy for the adjunctive treatment of breast cancer.


Subject(s)
Aurintricarboxylic Acid/pharmacology , Breast Neoplasms/enzymology , Deferoxamine/pharmacology , Iron Chelating Agents/pharmacology , Protein Tyrosine Phosphatase, Non-Receptor Type 11/antagonists & inhibitors , Protein Tyrosine Phosphatase, Non-Receptor Type 1/antagonists & inhibitors , Aurintricarboxylic Acid/chemistry , Binding Sites , Breast Neoplasms/pathology , Catalase/metabolism , Cell Proliferation/drug effects , Cell Survival/drug effects , Deferoxamine/chemistry , Female , Humans , Inhibitory Concentration 50 , MCF-7 Cells , Molecular Docking Simulation , Protein Tyrosine Phosphatase, Non-Receptor Type 1/metabolism , Protein Tyrosine Phosphatase, Non-Receptor Type 11/metabolism
9.
Oncotarget ; 8(7): 12234-12246, 2017 Feb 14.
Article in English | MEDLINE | ID: mdl-28103571

ABSTRACT

The survival of patients diagnosed with glioblastoma (GBM), the most deadly form of brain cancer, is compromised by the proclivity for local invasion into the surrounding normal brain, which prevents complete surgical resection and contributes to therapeutic resistance. Tumor necrosis factor-like weak inducer of apoptosis (TWEAK), a member of the tumor necrosis factor (TNF) superfamily, can stimulate glioma cell invasion and survival via binding to fibroblast growth factor-inducible 14 (Fn14) and subsequent activation of the transcription factor NF-κB. To discover small molecule inhibitors that disrupt the TWEAK-Fn14 signaling axis, we utilized a cell-based drug-screening assay using HEK293 cells engineered to express both Fn14 and a NF-κB-driven firefly luciferase reporter protein. Focusing on the LOPAC1280 library of 1280 pharmacologically active compounds, we identified aurintricarboxylic acid (ATA) as an agent that suppressed TWEAK-Fn14-NF-κB dependent signaling, but not TNFα-TNFR-NF-κB driven signaling. We demonstrated that ATA repressed TWEAK-induced glioma cell chemotactic migration and invasion via inhibition of Rac1 activation but had no effect on cell viability or Fn14 expression. In addition, ATA treatment enhanced glioma cell sensitivity to both the chemotherapeutic agent temozolomide (TMZ) and radiation-induced cell death. In summary, this work reports a repurposed use of a small molecule inhibitor that targets the TWEAK-Fn14 signaling axis, which could potentially be developed as a new therapeutic agent for treatment of GBM patients.


Subject(s)
Aurintricarboxylic Acid/pharmacology , Brain Neoplasms/drug therapy , Glioblastoma/drug therapy , Receptors, Tumor Necrosis Factor/metabolism , Signal Transduction/drug effects , Tumor Necrosis Factors/metabolism , Animals , Antineoplastic Agents, Alkylating/pharmacology , Aurintricarboxylic Acid/chemistry , Brain Neoplasms/genetics , Brain Neoplasms/metabolism , Cell Line, Tumor , Cell Movement/drug effects , Cell Movement/genetics , Cell Survival/drug effects , Cell Survival/genetics , Cell Survival/radiation effects , Cytokine TWEAK , Dacarbazine/analogs & derivatives , Dacarbazine/pharmacology , Drug Synergism , Glioblastoma/genetics , Glioblastoma/metabolism , HEK293 Cells , Humans , Kaplan-Meier Estimate , Mice, Nude , Molecular Structure , RNA Interference , Receptors, Tumor Necrosis Factor/genetics , Signal Transduction/genetics , Small Molecule Libraries/chemistry , Small Molecule Libraries/pharmacology , TWEAK Receptor , Temozolomide , Tumor Necrosis Factors/genetics , Xenograft Model Antitumor Assays/methods
10.
J Med Chem ; 49(12): 3595-601, 2006 Jun 15.
Article in English | MEDLINE | ID: mdl-16759101

ABSTRACT

Previously, we have reported that aurintricarboxylic acid (ATA) is one of the most potent inhibitors of the DNA binding of transcription factor NF-kappaB. We now report the NF-kappaB-DNA binding inhibitory activity of ATA analogues. An electrophoretic mobility shift assay has shown that bromopyrogallol red (BPR) is the most effective inhibitor of NF-kappaB-DNA binding among the studied analogues. The molecular modeling studies showed that BPR makes a strong network of hydrogen bonds with the DNA-binding region of the p50 subunit of NF-kappaB and has electronegative potential on its peripheral surface. Because zinc has been reported to influence the DNA binding of NF-kappaB, the interaction of these analogues with zinc was studied. Chemical speciation and formation-constant studies showed that BPR forms the most stable 1:1 complex with zinc. BPR has also been found to be the most potent antioxidant among the studied analogues.


Subject(s)
Antiviral Agents/chemistry , Aurintricarboxylic Acid/analogs & derivatives , Aurintricarboxylic Acid/chemistry , Chelating Agents/chemistry , DNA/metabolism , Models, Molecular , NF-kappa B/metabolism , Organometallic Compounds/chemistry , Zinc , DNA/chemistry , Electricity , Electrophoretic Mobility Shift Assay , Hydrogen Bonding , NF-kappa B/chemistry , NF-kappa B p50 Subunit/chemistry , Protein Binding , Pyrogallol/chemistry , Structure-Activity Relationship , Zinc/chemistry
11.
Chem Commun (Camb) ; (1): 34-44, 2006 Jan 07.
Article in English | MEDLINE | ID: mdl-16353086

ABSTRACT

Dendritic molecules constitute one of the most exciting areas of modern nanochemistry, largely as a consequence of the unique properties associated with their branched architectures. This article describes how 'dendritic function' can also be achieved using small, synthetically accessible branched building blocks (individual dendrons) which simply self-assemble via non-covalent interactions to generate dendritic nanoscale architectures with novel behaviour. (a) Using non-covalent interactions at the focal point of a dendron allows the self-assembly of nanometre-sized supramolecular dendrimers around an appropriate template species. Such systems have potential applications in the controlled encapsulation and release of active ingredients. (b) Employing non-covalent intermolecular dendron-dendron interactions can give rise to the hierarchical assembly of nanostructured materials. Such assemblies of dendritic molecules ultimately express their molecular scale information on a macroscopic scale, and therefore have applications in materials science, for example as gels. (c) The multiple surface groups of dendrons are capable of forming multiple interactions with large surfaces, such as those found on biomolecules or in biological systems. Employing multivalent interactions between dendron surfaces and biological molecules opens up the potential application of dendritic systems as medicinal therapies. In summary, dendritic supermolecules offer a potentially cost-effective approach to the future application of dendritic systems to a range of real-world problems.


Subject(s)
Dendrimers/chemistry , Nanostructures/chemistry , Aurintricarboxylic Acid/chemistry , Gels , Lanthanum/chemistry , Molecular Structure
12.
Cell Chem Biol ; 23(8): 1002-13, 2016 08 18.
Article in English | MEDLINE | ID: mdl-27499528

ABSTRACT

The increasing threats of antibiotic resistance urge the need for developing new strategies against bacterial infections. Targeting eukaryotic-like Ser/Thr phosphatase Stp1-mediated virulence regulation represents a promising approach for combating staphylococcal infection yet to be explored. Here, we report the 2.32-Å resolution crystal structure of Stp1. Stp1 binds an unexpected fourth metal ion, which is important for Stp1's enzymatic activity as demonstrated by amino acid substitution studies. Inspired by the structural details of Stp1, we identified a potent and selective Stp1 inhibitor, aurintricarboxylic acid (ATA). Transcriptome analysis and biochemical studies supported Stp1 as the target of ATA inhibition within the pathogen, preventing upregulation of virulence genes. Notably, ATA did not affect in vitro growth of Staphylococcus aureus, while simultaneously attenuating staphylococcal virulence in mice. Our findings demonstrate that ATA is a potent anti-virulence compound against staphylococcal infection, laying the foundation for further developing new scaffolds for Stp1-targeted small molecules.


Subject(s)
Aurintricarboxylic Acid/pharmacology , Phosphoric Monoester Hydrolases/antagonists & inhibitors , Staphylococcal Infections/drug therapy , Staphylococcal Infections/microbiology , Staphylococcus aureus/drug effects , Staphylococcus aureus/pathogenicity , Animals , Aurintricarboxylic Acid/chemistry , Dose-Response Relationship, Drug , Female , Mice , Mice, Inbred BALB C , Models, Molecular , Molecular Structure , Phosphoric Monoester Hydrolases/metabolism , Staphylococcus aureus/enzymology , Structure-Activity Relationship , Virulence/drug effects
13.
Oncotarget ; 6(21): 18364-73, 2015 Jul 30.
Article in English | MEDLINE | ID: mdl-26286963

ABSTRACT

YopH is a bacterial protein tyrosine phosphatase, which is essential for the viability and pathogenic virulence of the plague-causing Yersinia sp. bacteria. Inactivation of YopH activity would lead to the loss of bacterial pathogenicity. We have studied the inhibitory properties of aurintricarboxylic acid (ATA) against YopH phosphatase and found that at nanomolar concentrations ATA reversibly decreases the activity of YopH. Computational docking studies indicated that in all binding poses ATA binds in the YopH active site. Molecular dynamics simulations showed that in the predicted binding pose, ATA binds to the essential Cys403 and Arg409 residues in the active site and has a stronger binding affinity than the natural substrate (pTyr). The cyclic voltammetry experiments suggest that ATA reacts remarkably strongly with molecular oxygen. Additionally, the electrochemical reduction of ATA in the presence of a negative potential from -2.0 to 2.5 V generates a current signal, which is observed for hydrogen peroxide. Here we showed that ATA indicates a unique mechanism of YopH inactivation due to a redox process. We proposed that the potent inhibitory properties of ATA are a result of its strong binding in the YopH active site and in situ generation of hydrogen peroxide near catalytic cysteine residue.


Subject(s)
Aurintricarboxylic Acid/chemistry , Bacterial Outer Membrane Proteins/chemistry , Protein Tyrosine Phosphatases/chemistry , Virulence Factors/chemistry , Algorithms , Aurintricarboxylic Acid/metabolism , Aurintricarboxylic Acid/pharmacology , Bacterial Outer Membrane Proteins/antagonists & inhibitors , Bacterial Outer Membrane Proteins/metabolism , Humans , Kinetics , Molecular Conformation , Molecular Dynamics Simulation , Molecular Structure , Oxidation-Reduction , Plague/microbiology , Protein Binding , Protein Structure, Tertiary , Protein Tyrosine Phosphatases/antagonists & inhibitors , Protein Tyrosine Phosphatases/metabolism , Virulence , Virulence Factors/antagonists & inhibitors , Virulence Factors/metabolism , Yersinia pestis/metabolism , Yersinia pestis/pathogenicity , Yersinia pestis/physiology
14.
J Med Chem ; 34(1): 337-42, 1991 Jan.
Article in English | MEDLINE | ID: mdl-1704066

ABSTRACT

Several compounds corresponding to fragments of the schematic representation of the polymeric structure of aurintricarboxylic acid (ATA) have been prepared and tested for prevention of the cytopathic effect of HIV-1 and HIV-2 in MT-4 cell culture and HIV-1 in CEM cell culture. Both the triphenylcarbinol 3 as well as the triphenylmethane 5 were found to afford protection against the cytopathogenicity of HIV-2 in MT-4 cells and HIV-1 in CEM cells, but they were inactive against HIV-1 in MT-4 cells. Both substances were also found to inhibit syncytium formation when MOLT-4 cells were cocultured with HIV-2-infected HUT-78 cells, but were inactive in this assay against HIV-1-infected cells. When observed, the activity is generally moderate in degree of protection and requires concentrations in the 10(-4) molar range. In contrast to ATA, both of these substances were inactive when tested for prevention of the binding of the OKT4A monoclonal antibody to the CD4 receptor and also for inhibition of HIV-1 reverse transcriptase. These substances therefore appear act by a mechanism that is distinct from that of polymeric ATA. Several active and inactive structural analogues of 3 and 5 were also synthesized. The anti-HIV activity in this series seems to depend on the presence of anionic carboxylate groups, since the methyl esters 4, 6, and 12 were uniformly inactive. The diphenylmethanes 8, 14, 18, and 19 also reproducibly inhibited the cytopathic effect of HIV-1 in CEM cell culture.


Subject(s)
Antiviral Agents/chemical synthesis , Aurintricarboxylic Acid/analogs & derivatives , Aurintricarboxylic Acid/chemical synthesis , HIV-1/drug effects , HIV-2/drug effects , Aurintricarboxylic Acid/chemistry , Aurintricarboxylic Acid/pharmacology , CD4 Antigens/metabolism , Cell Line , HIV-1/enzymology , HIV-2/enzymology , Humans , Indicators and Reagents , Molecular Structure , Molecular Weight , Reverse Transcriptase Inhibitors , Structure-Activity Relationship
15.
J Med Chem ; 44(5): 703-14, 2001 Mar 01.
Article in English | MEDLINE | ID: mdl-11262081

ABSTRACT

Cosalane and its synthetic derivatives inhibit the binding of gp120 to CD4 as well as the fusion of the viral envelope with the cell membrane. The binding of the cosalanes to CD4 is proposed to involve ionic interactions of the negatively charged carboxylates of the ligands with positively charged arginine and lysine amino acid side chains of the protein. To investigate the effect of anion spacing on anti-HIV activity in the cosalane system, a series of cosalane tetracarboxylates was synthesized in which the two proximal and two distal carboxylates are separated by 6--12 atoms. Maximum activity was observed when the proximal and distal carboxylates are separated by 8 atoms. In a series of cosalane amino acid derivatives containing glutamic acid, glycine, aspartic acid, beta-alanine, leucine, and phenylalanine residues, maximum activity was displayed by the di(glutamic acid) analogue. A hypothetical model has been devised for the binding of the cosalane di(glutamic acid) conjugate to CD4. In general, the compounds in this series are more potent against HIV-1(RF) in CEM-SS cells than they are vs HIV-1(IIIB) in MT-4 cells, and they are least potent vs HIV-2(ROD) in MT-4 cells.


Subject(s)
Anti-HIV Agents/chemical synthesis , Aurintricarboxylic Acid/chemical synthesis , HIV-1/drug effects , Anions/chemistry , Anti-HIV Agents/chemistry , Anti-HIV Agents/pharmacology , Aurintricarboxylic Acid/analogs & derivatives , Aurintricarboxylic Acid/chemistry , Aurintricarboxylic Acid/pharmacology , CD4 Antigens/chemistry , Cell Line , Humans , Models, Molecular , Structure-Activity Relationship
16.
J Med Chem ; 39(2): 508-14, 1996 Jan 19.
Article in English | MEDLINE | ID: mdl-8558520

ABSTRACT

In order to define the role of the cholestane moiety in the anti-HIV agent cosalane, a series of cosalane analogs was synthesized in which the cholestane ring system was replaced by normal alkenyl and phosphodiester substituents having varied chain lengths and lipophilicities. The compounds containing simple alkenyl substituents were found to be more potent as inhibitors of the cytopathic effect of HIV-1 in cell culture than the phosphodiesters. In addition, the potencies of the alkene congeners correlated positively with chain length and lipophilicity of the alkene. The results indicate that the cholestane moiety of cosalane functions as a lipophilic accessory appendage to escort the dichlorodisalicylmethane pharmacophore to a lipid environment.


Subject(s)
Antiviral Agents/pharmacology , Aurintricarboxylic Acid/analogs & derivatives , HIV-1/drug effects , Lipid Metabolism , Antiviral Agents/chemistry , Antiviral Agents/metabolism , Aurintricarboxylic Acid/chemistry , Aurintricarboxylic Acid/metabolism , Aurintricarboxylic Acid/pharmacology , Cells, Cultured , Cytopathogenic Effect, Viral/drug effects , HIV-1/pathogenicity , Humans , Spectrum Analysis
17.
J Med Chem ; 34(1): 329-37, 1991 Jan.
Article in English | MEDLINE | ID: mdl-1704065

ABSTRACT

Aurintricarboxylic acid (ATA) was fractionated by a combination of dialysis, ultrafiltration, and gel permeation chromatography. The number average and weight average molecular weights of the ATA fractions were determined by the universal calibration method. The sulfonic acid analogue of ATA was prepared and separated in high and low molecular weight fractions. The phosphonic acid analogue of ATA was also synthesized. All of the ATA fractions were tested for prevention of the cytopathic effect of HIV-1 and HIV-2 in MT-4 cell culture as well as against HIV-1 in CEM cell culture. The abilities of the fractions and analogues to inhibit syncytium formation between HIV-1- and HIV-2-infected HUT-78 cells and uninfected MOLT-4 cells were evaluated. In addition, the fractions and analogues were tested for cytotoxicity in mock-infected MT-4 cells, prevention of the binding of the OKT4A monoclonal antibody to the CD4 receptor, inhibition of the binding of anti-gp120 monoclonal antibody to gp120, inhibition of attachment of HIV-1 virions to MT-4 cells, and inhibition of HIV-1 reverse transcriptase. In all of these assays except cytotoxicity, there was a correlation of potency with molecular weight. The higher the molecular weight, the higher the activity. Several of the lower molecular weight fractions of ATA, which bound to gp120 but not to CD4, prevented HIV-1 and HIV-2 cytopathicity. A similar profile was observed for the phosphonic acid analogue of ATA and the lower molecular weight fraction of the sulfonic acid analogue. The results on the ATA fractions indicate that the binding of ATA to gp120 in the absence of CD4 binding is sufficient for anti-HIV activity. The active compounds bind more avidly to gp120 than to CD4. The anti-HIV activity of the ATA fractions is due to inhibition of virus binding due to an interference with the gp120-CD4 interaction.


Subject(s)
Antiviral Agents/chemical synthesis , Aurintricarboxylic Acid/analogs & derivatives , Aurintricarboxylic Acid/chemical synthesis , HIV-1/drug effects , HIV-2/drug effects , Aurintricarboxylic Acid/chemistry , Aurintricarboxylic Acid/pharmacology , CD4 Antigens/metabolism , Cell Line , Cell Survival/drug effects , HIV-1/enzymology , HIV-2/enzymology , Humans , Indicators and Reagents , Molecular Structure , Molecular Weight , Reverse Transcriptase Inhibitors , Structure-Activity Relationship
18.
J Med Chem ; 42(10): 1767-77, 1999 May 20.
Article in English | MEDLINE | ID: mdl-10346929

ABSTRACT

The anti-HIV agent cosalane inhibits both the binding of gp120 to CD4 as well as an undefined postattachment event prior to reverse transcription. Several cosalane analogues having an extended polyanionic "pharmacophore" were designed based on a hypothetical model of the binding of cosalane to CD4. The analogues were synthesized, and a number of them displayed anti-HIV activity. One of the new analogues was found to possess enhanced potency as an anti-HIV agent relative to cosalane itself. Although the new analogues inhibited both HIV-1 and HIV-2, they were more potent as inhibitors of HIV-1 than HIV-2. Mechanism of action studies indicated that the most potent of the new analogues inhibited fusion of the viral envelope with the cell membrane at lower concentrations than it inhibited attachment, suggesting inhibition of fusion as the primary mechanism of action.


Subject(s)
Anti-HIV Agents/chemical synthesis , Aurintricarboxylic Acid/analogs & derivatives , Benzoates/chemical synthesis , Cholestanes/chemical synthesis , Animals , Anti-HIV Agents/chemistry , Anti-HIV Agents/metabolism , Anti-HIV Agents/pharmacology , Aurintricarboxylic Acid/chemistry , Benzoates/chemistry , Benzoates/metabolism , Benzoates/pharmacology , CD4 Antigens/chemistry , CD4 Antigens/metabolism , Cell Line , Cholestanes/chemistry , Cholestanes/metabolism , Cholestanes/pharmacology , HIV-1/drug effects , HIV-2/drug effects , Humans , Models, Molecular , Protein Binding , Structure-Activity Relationship
19.
J Med Chem ; 38(3): 443-52, 1995 Feb 03.
Article in English | MEDLINE | ID: mdl-7853337

ABSTRACT

Several new analogues of the novel anti-HIV agent cosalane have been synthesized and evaluated as inhibitors of HIV-1 integrase and protease, HIV-1 replication, HIV-1 and HIV-2 cytopathicity, HIV-1- and HIV-2-mediated syncytium formation, and cytopathicity of a variety of human pathogenic viruses. The congeners displayed enhanced potencies relative to cosalane itself as inhibitors of HIV-1 integrase and protease. The two most potent analogues against HIV-1 integrase displayed IC50 values of 2.2 microM, while the three most potent compounds against HIV-1 protease had IC50 values in the 0.35-0.39 microM range. In addition to its activity against HIV-1 and HIV-2 cytopathicity, cosalane inhibited the cytopathic effects of herpes simplex virus-1, herpes simplex virus-2, and human cytomegalovirus at concentrations that were well below the cytotoxic concentrations. Potentially useful antiviral activities were also revealed for some of the new cosalane congeners against influenza virus, Junin virus, and Tacaribe virus.


Subject(s)
Antiviral Agents/pharmacology , Aurintricarboxylic Acid/analogs & derivatives , DNA Nucleotidyltransferases/antagonists & inhibitors , HIV Protease Inhibitors/pharmacology , HIV-1/drug effects , Amino Acid Sequence , Aurintricarboxylic Acid/chemistry , Aurintricarboxylic Acid/pharmacology , Cell Line , HIV-1/enzymology , HIV-1/physiology , Herpesviridae/drug effects , Humans , Integrases , Microbial Sensitivity Tests , Molecular Sequence Data , Virus Replication/drug effects
20.
Mol Cells ; 18(1): 46-52, 2004 Aug 31.
Article in English | MEDLINE | ID: mdl-15359123

ABSTRACT

Aurintricarboxylic acid (ATA) prevents apoptosis in a diverse range of cell types including PC12 cells. It is known to stimulate tyrosine phosphorylation of signaling proteins including Shc proteins, phosphatidylinositol 3-kinase, phospholipase C-g and mitogen-activated protein kinases (MAPKs). However, it has been unclear how ATA increases the phosphorylation of these proteins as it was believed to be membrane impermeable. We found that ATA translocates across the plasma membrane of PC12 cells and have confirmed that it is a potent inhibitor of protein tyrosine phosphatases (PTP ases). Other PTPase inhibitors also prevented apoptosis independent of ATA. These observations indicate that ATA exerts its anti-apoptotic effect on PC12 cells at least in part by inhibiting certain PTPase(s).


Subject(s)
Apoptosis/physiology , Aurintricarboxylic Acid/metabolism , Cell Membrane/metabolism , Protein Tyrosine Phosphatases/metabolism , Animals , Aurintricarboxylic Acid/chemistry , Biological Transport/physiology , DNA Fragmentation , Molecular Structure , PC12 Cells , Protein Tyrosine Phosphatases/antagonists & inhibitors , Rats
SELECTION OF CITATIONS
SEARCH DETAIL