Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
1.
J Med Chem ; 64(1): 768-781, 2021 01 14.
Article in English | MEDLINE | ID: mdl-33440945

ABSTRACT

Berberine (BBR), a traditional Chinese medicine, has therapeutic effects on a variety of inflammation-related diseases, but its direct proteomic targets remain unknown. Using activity-based protein profiling, we first demonstrated that BBR directly targets the NEK7 protein via the hydrogen bond between the 2,3-methylenedioxy and 121-arginine (R121) residues. The fact that R121 is located precisely within the key domain involved in the NEK7-NLRP3 interaction allows BBR to specifically block the NEK7-NLRP3 interaction and successively inhibit IL-1ß release, independent of the NF-κB and TLR4 signaling pathways. Moreover, BBR displays in vivo anti-inflammatory efficacy in a NEK7-dependent manner. Therefore, we consider NEK7 to be a key target of BBR in the treatment of NLRP3-related inflammatory diseases, and the development of novel NEK7-NLRP3 interaction inhibitors might be easily achieved using NEK7 as a target.


Subject(s)
Anti-Inflammatory Agents/chemistry , Berberine/chemistry , NIMA-Related Kinases/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Animals , Anti-Inflammatory Agents/metabolism , Anti-Inflammatory Agents/pharmacology , Berberine/metabolism , Berberine/pharmacology , Binding Sites , Humans , Hydrogen Bonding , Interleukin-1beta/metabolism , Male , Mice , Mice, Inbred C57BL , Molecular Docking Simulation , Monocytes/cytology , Monocytes/drug effects , Monocytes/metabolism , NF-kappa B/metabolism , NIMA-Related Kinases/antagonists & inhibitors , NIMA-Related Kinases/genetics , NLR Family, Pyrin Domain-Containing 3 Protein/chemistry , Protein Interaction Domains and Motifs/drug effects , RNA Interference , RNA, Small Interfering/metabolism , Signal Transduction/drug effects , Structure-Activity Relationship
2.
SLAS Discov ; 26(5): 698-711, 2021 06.
Article in English | MEDLINE | ID: mdl-33345679

ABSTRACT

ETV6 is an ETS family transcriptional repressor for which head-to-tail polymerization of its PNT domain facilitates cooperative binding to DNA by its ETS domain. Chromosomal translocations frequently fuse the ETV6 PNT domain to one of several protein tyrosine kinases. The resulting chimeric oncoproteins undergo ligand-independent self-association, autophosphorylation, and aberrant stimulation of downstream signaling pathways, leading to a variety of cancers. Currently, no small-molecule inhibitors of ETV6 PNT domain polymerization are known and no assays targeting PNT domain polymerization have been described. In this study, we developed complementary experimental and computational approaches for identifying such inhibitory compounds. One mammalian cellular approach utilized a mutant PNT domain heterodimer system covalently attached to split Gaussia luciferase fragments. In this protein-fragment complementation assay, inhibition of PNT domain heterodimerization reduces luminescence. A yeast assay took advantage of activation of the reporter HIS3 gene upon heterodimerization of mutant PNT domains fused to DNA-binding and transactivation domains. In this two-hybrid screen, inhibition of PNT domain heterodimerization prevents cell growth in medium lacking histidine. The Bristol University Docking Engine (BUDE) was used to identify virtual ligands from the ZINC8 library predicted to bind the PNT domain polymerization interfaces. More than 75 hits from these three assays were tested by nuclear magnetic resonance spectroscopy for binding to the purified ETV6 PNT domain. Although none were found to bind, the lessons learned from this study may facilitate future approaches for developing therapeutics that act against ETV6 oncoproteins by disrupting PNT domain polymerization.


Subject(s)
Drug Discovery/methods , Drug Screening Assays, Antitumor/methods , Protein Interaction Domains and Motifs/drug effects , Protein Multimerization/drug effects , Proto-Oncogene Proteins c-ets/antagonists & inhibitors , Proto-Oncogene Proteins c-ets/chemistry , Repressor Proteins/antagonists & inhibitors , Repressor Proteins/chemistry , Biological Assay/methods , Genes, Reporter , Humans , Protein Binding , Structure-Activity Relationship , ETS Translocation Variant 6 Protein
3.
J Mol Graph Model ; 101: 107716, 2020 12.
Article in English | MEDLINE | ID: mdl-32866780

ABSTRACT

BACKGROUND: The receptor binding domain (RBD) of spike protein S1 domain SARS-CoV-2 plays a key role in the interaction with ACE2, which leads to subsequent S2 domain mediated membrane fusion and incorporation of viral RNA into host cells. In this study we tend to repurpose already approved drugs as inhibitors of the interaction between S1-RBD and the ACE2 receptor. METHODS: 2456 approved drugs were screened against the RBD of S1 protein of SARS-CoV-2 (target PDB ID: 6M17). As the interacting surface between S1-RBD and ACE2 comprises of bigger region, the interacting surface was divided into 3 sites on the basis of interactions (site 1, 2 and 3) and a total of 5 grids were generated (site 1, site 2, site 3, site 1+site 2 and site 2+site 3). A virtual screening was performed using GLIDE implementing HTVS, SP and XP screening. The top hits (on the basis of docking score) were further screened for MM-GBSA. All the top hits were further evaluated in molecular dynamics studies. Performance of the virtual screening protocol was evaluated using enrichment studies. RESULT: and discussion: We performed 5 virtual screening against 5 grids generated. A total of 42 compounds were identified after virtual screening. These drugs were further assessed for their interaction dynamics in molecular dynamics simulation. On the basis of molecular dynamics studies, we come up with 10 molecules with favourable interaction profile, which also interacted with physiologically important residues (residues taking part in the interaction between S1-RBD and ACE2. These are antidiabetic (acarbose), vitamins (riboflavin and levomefolic acid), anti-platelet agents (cangrelor), aminoglycoside antibiotics (Kanamycin, amikacin) bronchodilator (fenoterol), immunomodulator (lamivudine), and anti-neoplastic agents (mitoxantrone and vidarabine). However, while considering the relative side chain fluctuations when compared to the S1-RBD: ACE2 complex riboflavin, fenoterol, cangrelor and vidarabine emerged out as molecules with prolonged relative stability. CONCLUSION: We identified 4 already approved drugs (riboflavin, fenoterol, cangrelor and vidarabine) as possible agents for repurposing as inhibitors of S1:ACE2 interaction. In-vitro validation of these findings are necessary for identification of a safe and effective inhibitor of S1: ACE2 mediated entry of SARS-CoV-2 into the host cell.


Subject(s)
Antiviral Agents/pharmacology , Drug Evaluation, Preclinical/methods , Peptidyl-Dipeptidase A/metabolism , Protein Interaction Domains and Motifs/drug effects , Spike Glycoprotein, Coronavirus/metabolism , Angiotensin-Converting Enzyme 2 , Antiviral Agents/chemistry , Computer Simulation , Databases, Pharmaceutical , Host-Pathogen Interactions/drug effects , Models, Molecular , Molecular Dynamics Simulation , Peptidyl-Dipeptidase A/chemistry , Reproducibility of Results , Spike Glycoprotein, Coronavirus/chemistry
4.
J Med Chem ; 62(15): 7250-7263, 2019 08 08.
Article in English | MEDLINE | ID: mdl-31298541

ABSTRACT

A series of C2-symmetric inhibitors was designed and evaluated for inhibitory activity against the programmed cell death-1/programmed death-ligand 1(PD-1/PD-L1) protein-protein interaction (PPI) in a homogenous time-resolved fluorescence (HTRF) assay and PD-1 signaling in cell-based coculture assays. C2-symmetric inhibitors 2a (LH1306) and 2b (LH1307) exhibited IC50 values of 25 and 3.0 nM, respectively, in the HTRF assay. While 2a was ∼3.8-fold more potent than previously reported inhibitor 1a, 2b could not be differentiated from 1b due to their high potency and the limit of our HTRF assay conditions. In one cell-based coculture PD-1 signaling assay, 2a and 2b were 8.2- and 2.8-fold more potent in inhibiting PD-1 signaling than 1a and 1b, respectively. NMR and X-ray cocrystal structural studies provided more structural insights into the interaction between 2b and PD-L1; 2b binds to PD-L1 at the PD-1 binding site and induces the formation of a more symmetrically arranged PD-L1 homodimer than that previously reported for other inhibitors.


Subject(s)
B7-H1 Antigen/antagonists & inhibitors , Drug Design , Programmed Cell Death 1 Receptor/antagonists & inhibitors , Protein Interaction Domains and Motifs/drug effects , B7-H1 Antigen/metabolism , Cell Survival/drug effects , Cell Survival/physiology , Coculture Techniques , Dose-Response Relationship, Drug , Drug Evaluation, Preclinical/methods , Humans , Jurkat Cells , Programmed Cell Death 1 Receptor/metabolism , Protein Interaction Domains and Motifs/physiology , Protein Structure, Secondary , X-Ray Diffraction/methods
5.
SLAS Discov ; 24(6): 682-692, 2019 07.
Article in English | MEDLINE | ID: mdl-30802416

ABSTRACT

Intramolecular CFP-YFP fluorescence resonance energy transfer (FRET) sensors expressed in cells are powerful research tools but have seen relatively little use in screening. We exploited the discovery that the expression of a CFP-YFP FRET diacylglycerol sensor (DAGR) increases over time when cells are incubated at room temperature to assess requirements for robust measurements using a Molecular Devices Spectramax i3x fluorescence plate reader. Expression levels resulting in YFP fluorescence >10-fold higher than untransfected cells and phorbol ester-stimulated FRET ratio changes of 60% or more were required to consistently give robust Z' > 0.5. As a means of confirming that these conditions are suitable for screening, we developed a novel multiple-read protocol to assay the NCI's Mechanistic Set III for agonists and antagonists of C1 domain activation. Sixteen compounds prevented C1 domain translocation. However, none blocked phorbol ester-stimulated protein kinase C (PKC) activity assessed using a phospho-specific antibody-six actually stimulated PKC activity. Cytometry, which produces higher Z' for a given FRET ratio change, might have been a better approach for discovering antagonists, as it would have allowed lower phorbol ester concentrations to be used. We conclude that CFP-YFP FRET measured in a Spectramax i3x plate reader can be used for screening under the conditions we defined. Our strategy of varying expression level and FRET ratio could be useful to others for determining conditions needed for robust cell-based intramolecular CFP-YFP FRET measurements on their instrumentation.


Subject(s)
Biosensing Techniques , Diglycerides/chemistry , Drug Discovery/methods , Luminescent Proteins/chemistry , Luminescent Proteins/genetics , Protein Interaction Domains and Motifs/drug effects , Biomarkers , Cell Line, Tumor , Drug Evaluation, Preclinical/methods , Flow Cytometry , Fluorescence Resonance Energy Transfer/methods , Humans , Immunohistochemistry , Microscopy, Fluorescence
6.
Sci Adv ; 4(11): eaav0824, 2018 11.
Article in English | MEDLINE | ID: mdl-30443601

ABSTRACT

A third of the global population relies heavily upon traditional or folk medicines, such as the African shrub Mallotus oppositifolius. Here, we used pharmacological screening and electrophysiological analysis in combination with in silico docking and site-directed mutagenesis to elucidate the effects of M. oppositifolius constituents on KCNQ1, a ubiquitous and influential cardiac and epithelial voltage-gated potassium (Kv) channel. Two components of the M. oppositifolius leaf extract, mallotoxin (MTX) and 3-ethyl-2-hydroxy-2-cyclopenten-1-one (CPT1), augmented KCNQ1 current by negative shifting its voltage dependence of activation. MTX was also highly effective at augmenting currents generated by KCNQ1 in complexes with native partners KCNE1 or SMIT1; conversely, MTX inhibited KCNQ1-KCNE3 channels. MTX and CPT1 activated KCNQ1 by hydrogen bonding to the foot of the voltage sensor, a previously unidentified drug site which we also find to be essential for MTX activation of the related KCNQ2/3 channel. The findings elucidate the molecular mechanistic basis for modulation by a widely used folk medicine of an important human Kv channel and uncover novel molecular approaches for therapeutic modulation of potassium channel activity.


Subject(s)
Acetophenones/pharmacology , Benzopyrans/pharmacology , Cyclopentanes/pharmacology , Ion Channel Gating/drug effects , KCNQ1 Potassium Channel/metabolism , Medicine, Traditional , Plant Extracts/pharmacology , Potassium Channels, Voltage-Gated/metabolism , Potassium/metabolism , Protein Interaction Domains and Motifs/physiology , Animals , Humans , Oocytes/drug effects , Oocytes/metabolism , Protein Interaction Domains and Motifs/drug effects , Xenopus laevis/metabolism
7.
Leukemia ; 32(9): 1899-1907, 2018 09.
Article in English | MEDLINE | ID: mdl-30076373

ABSTRACT

Despite advances in the treatment of multiple myeloma, the disease still remains incurable for the majority of patients. The overexpression of anti-apoptotic proteins (i.e., Bcl-2, Bcl-XL or Mcl-1) is a hallmark of cancer and favors tumor cell survival and resistance to therapy. The oral drug venetoclax is the first-in-class Bcl-2-specific BH3 mimetic. In myeloma, in vitro sensitivity to venetoclax is mainly observed in plasma cells harboring the t(11;14) translocation, a molecular subgroup associated with high Bcl-2 and low Mcl-1/Bcl-XL expression. In addition with Bcl-2 members expression profile, functional tests as BH3 profiling or in vitro BH3 mimetic drug testing also predict sensitivity to the drug. Phase 1 clinical trials recently confirmed the efficacy of venetoclax monotherapy in heavily pretreated myeloma patients, mostly in patients with t(11;14). In combination with the proteasome inhibitor bortezomib, venetoclax therapy was found to be feasible and allowed promising response rate in relapsed myeloma patients, independent of t(11;14) status. The present review summarizes the current knowledge, "from bench to bedside", about venetoclax for the treatment of multiple myeloma.


Subject(s)
Antineoplastic Agents/therapeutic use , Molecular Targeted Therapy , Multiple Myeloma/drug therapy , Multiple Myeloma/metabolism , Proto-Oncogene Proteins c-bcl-2/antagonists & inhibitors , Animals , Antineoplastic Agents/pharmacology , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Apoptosis/drug effects , Apoptosis/genetics , Biomarkers , Bridged Bicyclo Compounds, Heterocyclic/pharmacology , Bridged Bicyclo Compounds, Heterocyclic/therapeutic use , Clinical Trials, Phase I as Topic , Drug Evaluation, Preclinical , Drug Resistance, Neoplasm/genetics , Gene Expression Regulation, Neoplastic/drug effects , Humans , Molecular Targeted Therapy/methods , Multiple Myeloma/genetics , Mutation , Protein Binding , Protein Interaction Domains and Motifs/drug effects , Proto-Oncogene Proteins c-bcl-2/chemistry , Proto-Oncogene Proteins c-bcl-2/genetics , Proto-Oncogene Proteins c-bcl-2/metabolism , Signal Transduction/drug effects , Sulfonamides/pharmacology , Sulfonamides/therapeutic use , Treatment Outcome
8.
ACS Chem Biol ; 13(3): 733-741, 2018 03 16.
Article in English | MEDLINE | ID: mdl-29359908

ABSTRACT

In-cell NMR spectroscopy was used to screen for drugs that disrupt the interaction between prokaryotic ubiquitin like protein, Pup, and mycobacterial proteasome ATPase, Mpa. This interaction is critical for Mycobacterium tuberculosis resistance against nitric oxide (NO) stress; interruption of this process was proposed as a mechanism to control latent infection. Three compounds isolated from the NCI Diversity set III library rescued the physiological proteasome substrate from degradation suggesting that the proteasome degradation pathway was selectively targeted. Two of the compounds bind to Mpa with sub-micromolar to nanomolar affinity, and all three exhibit potency toward mycobacteria comparable to antibiotics currently available on the market, inhibiting growth in the low micromolar range.


Subject(s)
Drug Evaluation, Preclinical/methods , Magnetic Resonance Spectroscopy/methods , Mycobacterium tuberculosis/drug effects , Bacterial Proteins/metabolism , Mycobacterium tuberculosis/growth & development , Proteasome Endopeptidase Complex/drug effects , Proteasome Endopeptidase Complex/metabolism , Protein Binding/drug effects , Protein Interaction Domains and Motifs/drug effects , Ubiquitins/metabolism
9.
Chem Biol Drug Des ; 85(3): 290-5, 2015 Mar.
Article in English | MEDLINE | ID: mdl-24954548

ABSTRACT

Lovastatin was identified through virtual screening as a potential inhibitor of the LEDGF/p75-HIV-1 integrase interaction. In an AlphaScreen assay, lovastatin inhibited the purified recombinant protein-protein interaction (IC50 = 1.97 ± 0.45 µm) more effectively than seven other tested statins. None of the eight statins, however, yielded antiviral activity in vitro, while only pravastatin lactone yielded detectable inhibition of HIV-1 integrase strand transfer activity (31.65% at 100 µm). A correlation between lipophilicity and increased cellular toxicity of the statins was observed.


Subject(s)
HIV Integrase/chemistry , Hydroxymethylglutaryl-CoA Reductase Inhibitors/chemistry , Intercellular Signaling Peptides and Proteins/chemistry , Cells, Cultured , Drug Evaluation, Preclinical , HIV Integrase/genetics , HIV Integrase/metabolism , HIV Integrase Inhibitors/chemistry , HIV Integrase Inhibitors/pharmacology , HIV-1/enzymology , HIV-1/physiology , Humans , Hydroxymethylglutaryl-CoA Reductase Inhibitors/pharmacology , Intercellular Signaling Peptides and Proteins/genetics , Intercellular Signaling Peptides and Proteins/metabolism , Leukocytes, Mononuclear/cytology , Leukocytes, Mononuclear/drug effects , Leukocytes, Mononuclear/metabolism , Lovastatin/chemistry , Lovastatin/pharmacology , Protein Interaction Domains and Motifs/drug effects , Recombinant Proteins/biosynthesis , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Virus Replication/drug effects
10.
Cardiovasc Res ; 105(1): 118-28, 2015 Jan 01.
Article in English | MEDLINE | ID: mdl-25411383

ABSTRACT

AIMS: The ryanodine receptor (RyR2) is an intracellular Ca(2+) release channel essential for cardiac excitation-contraction coupling. Abnormal RyR2 channel function results in the generation of arrhythmias and sudden cardiac death. The present study was undertaken to investigate the mechanistic basis of RyR2 dysfunction in inherited arrhythmogenic cardiac disease. METHODS AND RESULTS: We present several lines of complementary evidence, indicating that the arrhythmia-associated L433P mutation disrupts RyR2 N-terminus self-association. A combination of yeast two-hybrid, co-immunoprecipitation, and chemical cross-linking assays collectively demonstrate that a RyR2 N-terminal fragment carrying the L433P mutation displays substantially reduced self-interaction compared with wild type. Moreover, sucrose density gradient centrifugation reveals that the L433P mutation impairs tetramerization of the full-length channel. [(3)H]Ryanodine-binding assays demonstrate that disrupted N-terminal intersubunit interactions within RyR2(L433P) confer an altered sensitivity to Ca(2+) activation. Calcium imaging of RyR2(L433P)-expressing cells reveals substantially prolonged Ca(2+) transients and reduced Ca(2+) store content indicating defective channel closure. Importantly, dantrolene treatment reverses the L433P mutation-induced impairment and restores channel function. CONCLUSION: The N-terminus domain constitutes an important structural determinant for the functional oligomerization of RyR2. Our findings are consistent with defective N-terminus self-association as a molecular mechanism underlying RyR2 channel deregulation in inherited arrhythmogenic cardiac disease. Significantly, the therapeutic action of dantrolene may occur via the restoration of normal RyR2 N-terminal intersubunit interactions.


Subject(s)
Arrhythmias, Cardiac/drug therapy , Dantrolene/pharmacology , Mutant Proteins/genetics , Mutant Proteins/metabolism , Ryanodine Receptor Calcium Release Channel/genetics , Ryanodine Receptor Calcium Release Channel/metabolism , Amino Acid Substitution , Anti-Arrhythmia Agents/pharmacology , Arrhythmias, Cardiac/genetics , Arrhythmias, Cardiac/metabolism , Arrhythmogenic Right Ventricular Dysplasia/etiology , Arrhythmogenic Right Ventricular Dysplasia/genetics , Arrhythmogenic Right Ventricular Dysplasia/metabolism , Calcium Signaling/drug effects , HEK293 Cells , Heart Failure/etiology , Heart Failure/genetics , Heart Failure/metabolism , Humans , Models, Cardiovascular , Mutagenesis, Site-Directed , Mutant Proteins/chemistry , Myocardium/metabolism , Protein Interaction Domains and Motifs/drug effects , Protein Multimerization/drug effects , Protein Stability/drug effects , Ryanodine Receptor Calcium Release Channel/chemistry , Tachycardia, Ventricular/etiology , Tachycardia, Ventricular/genetics , Tachycardia, Ventricular/metabolism
11.
Proc Natl Acad Sci U S A ; 111(34): 12556-61, 2014 Aug 26.
Article in English | MEDLINE | ID: mdl-25114221

ABSTRACT

In a fluorescence polarization screen for the MYC-MAX interaction, we have identified a novel small-molecule inhibitor of MYC, KJ-Pyr-9, from a Kröhnke pyridine library. The Kd of KJ-Pyr-9 for MYC in vitro is 6.5 ± 1.0 nM, as determined by backscattering interferometry; KJ-Pyr-9 also interferes with MYC-MAX complex formation in the cell, as shown in a protein fragment complementation assay. KJ-Pyr-9 specifically inhibits MYC-induced oncogenic transformation in cell culture; it has no or only weak effects on the oncogenic activity of several unrelated oncoproteins. KJ-Pyr-9 preferentially interferes with the proliferation of MYC-overexpressing human and avian cells and specifically reduces the MYC-driven transcriptional signature. In vivo, KJ-Pyr-9 effectively blocks the growth of a xenotransplant of MYC-amplified human cancer cells.


Subject(s)
Antineoplastic Agents/pharmacology , Proto-Oncogene Proteins c-myc/antagonists & inhibitors , Pyridines/pharmacology , Pyrimidines/pharmacology , Animals , Antineoplastic Agents/chemistry , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/antagonists & inhibitors , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/chemistry , Cell Proliferation/drug effects , Cell Transformation, Neoplastic/drug effects , Cell Transformation, Neoplastic/genetics , Cells, Cultured , Chick Embryo , Drug Evaluation, Preclinical , Female , Fluorescence Polarization , Genes, myc , Humans , Interferometry , Mice , Mice, Nude , Multiprotein Complexes/antagonists & inhibitors , Multiprotein Complexes/chemistry , Protein Interaction Domains and Motifs/drug effects , Proto-Oncogene Proteins c-myc/chemistry , Pyridines/chemistry , Pyrimidines/chemistry , Xenograft Model Antitumor Assays
12.
Bioorg Med Chem Lett ; 23(20): 5575-7, 2013 Oct 15.
Article in English | MEDLINE | ID: mdl-24012120

ABSTRACT

The identification of a novel hit compound inhibitor of the protein-protein interaction between the influenza RNA-polymerase PA and PB1 subunits has been accomplished by means of high-throughput screening. A small family of structurally related molecules has been synthesized and biologically evaluated with most of the compounds showing micromolar potency of inhibition against viral replication.


Subject(s)
Antiviral Agents/toxicity , Benzoxazoles/chemistry , DNA-Directed RNA Polymerases/metabolism , Enzyme Inhibitors/chemical synthesis , Influenza A virus/drug effects , Animals , Antiviral Agents/chemical synthesis , Antiviral Agents/chemistry , Benzoxazoles/chemical synthesis , Benzoxazoles/toxicity , DNA-Directed RNA Polymerases/chemistry , Dogs , Drug Evaluation, Preclinical , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/toxicity , Influenza A virus/enzymology , Madin Darby Canine Kidney Cells , Protein Interaction Domains and Motifs/drug effects , Protein Subunits/chemistry , Protein Subunits/metabolism , Structure-Activity Relationship
13.
Bioorg Med Chem Lett ; 23(5): 1253-6, 2013 Mar 01.
Article in English | MEDLINE | ID: mdl-23375228

ABSTRACT

The neonatal Fc receptor, FcRn, prolongs the half-life of IgG in the serum and represents a potential therapeutic target for the treatment of autoimmune disease. Small molecules that block the protein-protein interactions of human IgG-human FcRn may lower pathogenic autoantibodies and provide effective treatment. A novel class of quinoxalines has been discovered as antagonists of the IgG:FcRn protein-protein interaction through optimization of a hit derived from a virtual ligand-based screen.


Subject(s)
Histocompatibility Antigens Class I/metabolism , Immunoglobulin G/metabolism , Quinoxalines/pharmacology , Receptors, Fc/antagonists & inhibitors , Receptors, Fc/metabolism , Drug Evaluation, Preclinical , Histocompatibility Antigens Class I/immunology , Humans , Immunoglobulin G/immunology , Ligands , Protein Interaction Domains and Motifs/drug effects , Receptors, Fc/immunology , Structure-Activity Relationship
14.
Proc Natl Acad Sci U S A ; 110(4): 1261-6, 2013 Jan 22.
Article in English | MEDLINE | ID: mdl-23284167

ABSTRACT

Signaling through the Rho family of small GTPases has been intensely investigated for its crucial roles in a wide variety of human diseases. Although RhoA and Rac1 signaling pathways are frequently exploited with the aid of effective small molecule modulators, studies of the Cdc42 subclass have lagged because of a lack of such means. We have applied high-throughput in silico screening and identified compounds that are able to fit into the surface groove of Cdc42, which is critical for guanine nucleotide exchange factor binding. Based on the interaction between Cdc42 and intersectin (ITSN), a specific Cdc42 guanine nucleotide exchange factor, we discovered compounds that rendered ITSN-like interactions in the binding pocket. By using in vitro binding and imaging as well as biochemical and cell-based assays, we demonstrated that ZCL278 has emerged as a selective Cdc42 small molecule modulator that directly binds to Cdc42 and inhibits its functions. In Swiss 3T3 fibroblast cultures, ZCL278 abolished microspike formation and disrupted GM130-docked Golgi structures, two of the most prominent Cdc42-mediated subcellular events. ZCL278 reduces the perinuclear accumulation of active Cdc42 in contrast to NSC23766, a selective Rac inhibitor. ZCL278 suppresses Cdc42-mediated neuronal branching and growth cone dynamics as well as actin-based motility and migration in a metastatic prostate cancer cell line (i.e., PC-3) without disrupting cell viability. Thus, ZCL278 is a small molecule that specifically targets Cdc42-ITSN interaction and inhibits Cdc42-mediated cellular processes, thus providing a powerful tool for research of Cdc42 subclass of Rho GTPases in human pathogenesis, such as those of cancer and neurological disorders.


Subject(s)
Adaptor Proteins, Vesicular Transport/antagonists & inhibitors , Adaptor Proteins, Vesicular Transport/chemistry , cdc42 GTP-Binding Protein/antagonists & inhibitors , cdc42 GTP-Binding Protein/chemistry , Adaptor Proteins, Vesicular Transport/physiology , Amino Acid Sequence , Animals , Binding Sites , Cell Movement/drug effects , Cell Movement/physiology , Cell Survival/drug effects , Cells, Cultured , Drug Evaluation, Preclinical , Golgi Apparatus/drug effects , Golgi Apparatus/physiology , Humans , Mice , Models, Molecular , Molecular Sequence Data , Neurons/drug effects , Neurons/ultrastructure , Protein Interaction Domains and Motifs/drug effects , Sequence Homology, Amino Acid , Signal Transduction/drug effects , Swiss 3T3 Cells , User-Computer Interface , Wound Healing/drug effects , cdc42 GTP-Binding Protein/genetics , cdc42 GTP-Binding Protein/physiology
15.
Curr Pharm Biotechnol ; 14(5): 501-12, 2013.
Article in English | MEDLINE | ID: mdl-22429133

ABSTRACT

Peptides are ideally suited to mimic natural ligands and thereby function in an antagonistic or agonistic way. Furthermore they are able to physiologically disrupt functional complexes due to their small size and specific binding properties. Proteins form homo- or heteromeric (macro)molecular complexes and intricate networks by interacting with small molecules, peptides, nucleic acids or other proteins. On average, five interaction partners have been estimated for any given cellular protein, illustrating the complexity of the formed 'interactomes' and the impact of their investigation. Many protein-protein interactions are mediated by hot-spots, which comprise only a small part of the large binding interface but account for 80% of the binding energy. Thus, these hot spots provide an 'Achilles heel' for pharmaceutical interventions aiming at the disruption of functional protein-protein complexes. Methods to select peptides for defined target structures include display technologies on phages, ribosomes or yeast, and the yeast-two-hybrid system. Once selected, these peptides can be optimized for their binding affinity using peptide arrays. Stabilization of biologically unstable peptides is achieved by the introduction of non-natural amino acids to form so-called peptidomimetics that are resistant to cellular proteases. Moreover, lipocalins and peptide aptamers represent scaffolded binding structures with unique binding characteristics and enhanced stability. In case of extracellular targets, like cell surface receptors or pathogens in patients` plasma, peptide inhibitors have direct access. Addressing intracellular targets with peptides is more difficult since short hydrophilic peptides generally do not cross plasma membranes on their own. However, intracellular uptake of peptides can be achieved by coupling to carrier systems like liposomes or nanoparticles or upon fusion to a protein transduction domain. Alternatively, peptides may be expressed within cells after transduction with viral vectors in a gene therapy setting. This review will summarize the broad potential of peptides as drugs, with special emphasis on peptides which inhibit protein-protein interactions.


Subject(s)
Peptides/pharmacology , Peptides/therapeutic use , Protein Interaction Domains and Motifs/drug effects , Animals , Drug Evaluation, Preclinical/methods , Humans , Protein Binding/drug effects
16.
J Biomol Screen ; 17(4): 435-47, 2012 Apr.
Article in English | MEDLINE | ID: mdl-22156223

ABSTRACT

Activation of the antioxidant response element (ARE) upregulates enzymes involved in detoxification of electrophiles and reactive oxygen species. The induction of ARE genes is regulated by the interaction between redox sensor protein Keap1 and the transcription factor Nrf2. Fluorescently labeled Nrf2 peptides containing the ETGE motif were synthesized and optimized as tracers in the development of a fluorescence polarization (FP) assay to identify small-molecule inhibitors of the Keap1-Nrf2 interaction. The tracers were optimized to increase the dynamic range of the assay and their binding affinities to the Keap1 Kelch domain. The binding affinities of Nrf2 peptide inhibitors obtained in our FP assay using FITC-9mer Nrf2 peptide amide as the probe were in good agreement with those obtained previously by a surface plasmon resonance assay. The FP assay exhibits considerable tolerance toward DMSO and produced a Z' factor greater than 0.6 in a 384-well format. Further optimization of the probe led to cyanine-labeled 9mer Nrf2 peptide amide, which can be used along with the FITC-9mer Nrf2 peptide amide in a high-throughput screening assay to discover small-molecule inhibitors of Keap1-Nrf2 interaction.


Subject(s)
Fluorescence Polarization Immunoassay/methods , Fluorescent Dyes/chemistry , Intracellular Signaling Peptides and Proteins/metabolism , NF-E2-Related Factor 2/metabolism , Peptides/chemistry , Binding, Competitive , Drug Evaluation, Preclinical , Intracellular Signaling Peptides and Proteins/chemistry , Kelch-Like ECH-Associated Protein 1 , NF-E2-Related Factor 2/chemistry , Protein Binding/drug effects , Protein Interaction Domains and Motifs/drug effects , Staining and Labeling
17.
J Biomol Screen ; 17(4): 530-4, 2012 Apr.
Article in English | MEDLINE | ID: mdl-22127421

ABSTRACT

Aberrant activation of the Wnt/ß-catenin signaling pathway is associated with a wide range of human cancers. The interaction of ß-catenin with T cell factor (Tcf) is a key step in activation of proliferative genes in this pathway. Interruption of this interaction would be a valuable strategy as a tumor therapy. In this study, we developed a novel fluorescein isothiocyanate (FITC)-labeled Tcf4-derived probe for identification of inhibitors of the ß-catenin/Tcf4 interaction using a fluorescence polarization assay. This assay shows high potential for use in high-throughput screening for the discovery of inhibitors of the ß-catenin/Tcf4 interaction.


Subject(s)
Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/metabolism , Fluorescence Polarization/methods , Transcription Factors/metabolism , beta Catenin/metabolism , Binding, Competitive/drug effects , Drug Evaluation, Preclinical/methods , High-Throughput Screening Assays , Humans , Protein Binding/drug effects , Protein Interaction Domains and Motifs/drug effects , Transcription Factor 4
18.
Bioorg Med Chem ; 19(8): 2589-95, 2011 Apr 15.
Article in English | MEDLINE | ID: mdl-21459001

ABSTRACT

The pharmacological suppression of the DNA damage response and DNA repair can increase the therapeutic indices of conventional chemotherapeutics. Replication Protein A (RPA), the major single-stranded DNA binding protein in eukaryotes, is required for DNA replication, DNA repair, DNA recombination, and DNA damage response signaling. Through the use of high-throughput screening of 1500 compounds, we have identified a small molecule inhibitor, 15-carboxy-13-isopropylatis-13-ene-17,18-dioic acid (NSC15520), that inhibited both the binding of Rad9-GST and p53-GST fusion proteins to the RPA N-terminal DNA binding domain (DBD), interactions that are essential for robust DNA damage signaling. NSC15520 competitively inhibited the binding of p53-GST peptide with an IC(50) of 10 µM. NSC15520 also inhibited helix destabilization of a duplex DNA (dsDNA) oligonucleotide, an activity dependent on the N-terminal domain of RPA70. NSC15520 did not inhibit RPA from binding single-stranded oligonucleotides, suggesting that the action of this inhibitor is specific for the N-terminal DBD of RPA, and does not bind to DBDs essential for single-strand DNA binding. Computer modeling implicates direct competition between NSC15520 and Rad9 for the same binding surface on RPA. Inhibitors of protein-protein interactions within the N-terminus of RPA are predicted to act synergistically with DNA damaging agents and inhibitors of DNA repair. Novel compounds such as NSC15520 have the potential to serve as chemosensitizing agents.


Subject(s)
Protein Interaction Domains and Motifs/drug effects , Replication Protein A/antagonists & inhibitors , Binding Sites , Computer Simulation , DNA Repair , DNA, Single-Stranded , Drug Evaluation, Preclinical/methods , High-Throughput Screening Assays , Humans , Oligonucleotides
SELECTION OF CITATIONS
SEARCH DETAIL