Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 21
Filter
1.
J Biol Chem ; 296: 100263, 2021.
Article in English | MEDLINE | ID: mdl-33837744

ABSTRACT

The development of a targeted therapy would significantly improve the treatment of periodontitis and its associated diseases including Alzheimer's disease, rheumatoid arthritis, and cardiovascular diseases. Glutaminyl cyclases (QCs) from the oral pathogens Porphyromonas gingivalis, Tannerella forsythia, and Prevotella intermedia represent attractive target enzymes for small-molecule inhibitor development, as their action is likely to stabilize essential periplasmic and outer membrane proteins by N-terminal pyroglutamination. In contrast to other microbial QCs that utilize the so-called type I enzymes, these oral pathogens possess sequences corresponding to type II QCs, observed hitherto only in animals. However, whether differences between these bacteroidal QCs and animal QCs are sufficient to enable development of selective inhibitors is not clear. To learn more, we recombinantly expressed all three QCs. They exhibit comparable catalytic efficiencies and are inhibited by metal chelators. Crystal structures of the enzymes from P. gingivalis (PgQC) and T. forsythia (TfQC) reveal a tertiary structure composed of an eight-stranded ß-sheet surrounded by seven α-helices, typical of animal type II QCs. In each case, an active site Zn ion is tetrahedrally coordinated by conserved residues. Nevertheless, significant differences to mammalian enzymes are found around the active site of the bacteroidal enzymes. Application of a PgQC-selective inhibitor described here for the first time results in growth inhibition of two P. gingivalis clinical isolates in a dose-dependent manner. The insights gained by these studies will assist in the development of highly specific small-molecule bacteroidal QC inhibitors, paving the way for alternative therapies against periodontitis and associated diseases.


Subject(s)
Aminoacyltransferases/chemistry , Periodontitis/microbiology , Porphyromonas gingivalis/enzymology , Prevotella intermedia/enzymology , Aminoacyltransferases/antagonists & inhibitors , Aminoacyltransferases/genetics , Aminoacyltransferases/ultrastructure , Catalytic Domain/drug effects , Crystallography, X-Ray , Humans , Periodontitis/drug therapy , Periodontitis/genetics , Porphyromonas gingivalis/pathogenicity , Prevotella intermedia/pathogenicity , Protein Structure, Tertiary/drug effects , Pyrrolidonecarboxylic Acid/chemistry , Pyrrolidonecarboxylic Acid/metabolism , Tannerella forsythia/enzymology , Tannerella forsythia/pathogenicity
2.
Int J Biol Macromol ; 174: 309-318, 2021 Mar 31.
Article in English | MEDLINE | ID: mdl-33524481

ABSTRACT

Human immunodeficiency virus type 1 reverse transcriptase (HIV-1 RT) is the key enzyme for the virus gene replication and the most important target for antiviral therapy. Toxicity, drug resistance and side effects have led to search for new antiviral agents. Farnesiferol C (FC) is a well-known biologically active sesquiterpene coumarin derivative from genus Ferula. The current study was designed to examine the impacts of FC on the structure and function of HIV-1 RT, using some theoretical and experimental methods. FC inhibited HIV-1RT activity via mixed inhibition mechanism (IC50 = 30 µM). Spectroscopic data showed some conformational changes in the secondary as well as tertiary structure of HIV-1RT following the interaction with FC. Results showed that FC could quench the intrinsic fluorescence emission of HIV-1RT through static quenching mechanism. Thermodynamic parameters revealed that hydrogen bondings and van der Waals forces are the major forces in the binding reaction and the low equilibrium constants (KD) value obtained from surface plasmon resonance data, confirmed the high affinity of FC for HIV-1RT. Molecular docking studies indicated that FC interacts with enzyme through hydrophobic pocket. Taken together, the outcomes of this research revealed that, sesquiterpene coumarines can be used to design natural remedies as anti-HIV agents.


Subject(s)
Coumarins/pharmacology , Ferula/chemistry , HIV Reverse Transcriptase/metabolism , HIV-1/enzymology , Reverse Transcriptase Inhibitors/pharmacology , Coumarins/chemistry , HIV Reverse Transcriptase/chemistry , HIV-1/drug effects , Hydrogen Bonding , Models, Molecular , Molecular Docking Simulation , Molecular Structure , Phytochemicals/chemistry , Phytochemicals/pharmacology , Protein Structure, Secondary/drug effects , Protein Structure, Tertiary/drug effects , Reverse Transcriptase Inhibitors/chemistry , Surface Plasmon Resonance
3.
Nat Commun ; 7: 12579, 2016 08 30.
Article in English | MEDLINE | ID: mdl-27573496

ABSTRACT

Protein disulfide isomerase (PDI) is an oxidoreductase essential for folding proteins in the endoplasmic reticulum. The domain structure of PDI is a-b-b'-x-a', wherein the thioredoxin-like a and a' domains mediate disulfide bond shuffling and b and b' domains are substrate binding. The b' and a' domains are connected via the x-linker, a 19-amino-acid flexible peptide. Here we identify a class of compounds, termed bepristats, that target the substrate-binding pocket of b'. Bepristats reversibly block substrate binding and inhibit platelet aggregation and thrombus formation in vivo. Ligation of the substrate-binding pocket by bepristats paradoxically enhances catalytic activity of a and a' by displacing the x-linker, which acts as an allosteric switch to augment reductase activity in the catalytic domains. This substrate-driven allosteric switch is also activated by peptides and proteins and is present in other thiol isomerases. Our results demonstrate a mechanism whereby binding of a substrate to thiol isomerases enhances catalytic activity of remote domains.


Subject(s)
Endoplasmic Reticulum/metabolism , Enzyme Inhibitors/pharmacology , Platelet Aggregation/drug effects , Protein Disulfide-Isomerases/metabolism , Protein Folding , Allosteric Regulation/drug effects , Animals , Blood Platelets/drug effects , Blood Platelets/physiology , Catalytic Domain/drug effects , Disease Models, Animal , Drug Evaluation, Preclinical , Enzyme Inhibitors/therapeutic use , Healthy Volunteers , Humans , Hydrophobic and Hydrophilic Interactions/drug effects , Insulin/metabolism , Male , Mice , Mice, Inbred C57BL , Protein Binding/drug effects , Protein Disulfide-Isomerases/antagonists & inhibitors , Protein Disulfide-Isomerases/chemistry , Protein Structure, Tertiary/drug effects , Recombinant Proteins/chemistry , Recombinant Proteins/isolation & purification , Recombinant Proteins/metabolism , Thrombosis/blood , Thrombosis/drug therapy , Thrombosis/pathology
4.
Proc Natl Acad Sci U S A ; 112(34): 10768-73, 2015 Aug 25.
Article in English | MEDLINE | ID: mdl-26261308

ABSTRACT

Th17 responses are critical to a variety of human autoimmune diseases, and therapeutic targeting with monoclonal antibodies against IL-17 and IL-23 has shown considerable promise. Here, we report data to support selective bromodomain blockade of the transcriptional coactivators CBP (CREB binding protein) and p300 as an alternative approach to inhibit human Th17 responses. We show that CBP30 has marked molecular specificity for the bromodomains of CBP and p300, compared with 43 other bromodomains. In unbiased cellular testing on a diverse panel of cultured primary human cells, CBP30 reduced immune cell production of IL-17A and other proinflammatory cytokines. CBP30 also inhibited IL-17A secretion by Th17 cells from healthy donors and patients with ankylosing spondylitis and psoriatic arthritis. Transcriptional profiling of human T cells after CBP30 treatment showed a much more restricted effect on gene expression than that observed with the pan-BET (bromo and extraterminal domain protein family) bromodomain inhibitor JQ1. This selective targeting of the CBP/p300 bromodomain by CBP30 will potentially lead to fewer side effects than with the broadly acting epigenetic inhibitors currently in clinical trials.


Subject(s)
Benzimidazoles/pharmacology , Immunosuppressive Agents/pharmacology , Interleukin-17/metabolism , Isoxazoles/pharmacology , Th17 Cells/drug effects , p300-CBP Transcription Factors/antagonists & inhibitors , Adult , Aged , Arthritis, Psoriatic/metabolism , Arthritis, Psoriatic/pathology , Azepines/pharmacology , Benzimidazoles/chemistry , CD4-Positive T-Lymphocytes/drug effects , CD4-Positive T-Lymphocytes/metabolism , Calorimetry , Cells, Cultured , Crystallography, X-Ray , Drug Evaluation, Preclinical , Female , Gene Expression Regulation/drug effects , Humans , Immunosuppressive Agents/chemistry , Interleukin-17/biosynthesis , Interleukin-17/genetics , Isoxazoles/chemistry , Kinetics , Male , Middle Aged , Models, Molecular , Molecular Structure , Protein Conformation , Protein Structure, Tertiary/drug effects , Recombinant Proteins/metabolism , Spondylitis, Ankylosing/metabolism , Spondylitis, Ankylosing/pathology , Structure-Activity Relationship , Th17 Cells/immunology , Triazoles/pharmacology
5.
Eur J Med Chem ; 75: 301-7, 2014 Mar 21.
Article in English | MEDLINE | ID: mdl-24556145

ABSTRACT

Our previous study reported that a mixture of cerebrosides from traditional Chinese medicine Baifuzi could activate BKCa channel. It is curious to know the effect of each single cerebroside on the channel. Here we isolated 5 pure cerebrosides from the mixture and determined their chemical structures. The most potent one increased the single channel open probability 6 folds with EC50 value of 1.0 µM. The structure-activity relationship revealed that acyl chain length of cerebrosides has potent effect, while configuration of double bond at C8-C9 on their long chain base has weak effect on the channel activity. Thus, this research provides a guide for design and synthesis of a lead cerebroside with more potent effect on the BKCa channel.


Subject(s)
Cerebrosides/chemistry , Cerebrosides/pharmacology , Drugs, Chinese Herbal/chemistry , Drugs, Chinese Herbal/pharmacology , Large-Conductance Calcium-Activated Potassium Channels/agonists , Large-Conductance Calcium-Activated Potassium Channels/metabolism , Animals , CHO Cells , Cerebrosides/isolation & purification , Cricetulus , Large-Conductance Calcium-Activated Potassium Channels/chemistry , Patch-Clamp Techniques , Protein Structure, Tertiary/drug effects , Structure-Activity Relationship
6.
Int J Mol Sci ; 13(2): 2459-2471, 2012.
Article in English | MEDLINE | ID: mdl-22408464

ABSTRACT

Circular dichroism (CD) and deconvolution were used to study the structural integrity of a "plugged" and an "open" FhuA transmembrane channel protein in the presence of varied concentrations of tetrahydrofuran (THF), ethanol (EtOH) and chloroform/methanol (C/M). FhuA is an Escherichia coli outer membrane protein (78.9 kDa) consisting of 22 ß-sheets and an internal globular cork domain which acts as an iron transporter. FhuA and the deletion variant FhuA Δ1-159 showed comparable and remarkable resistance in the presence of THF (≤40 vol%) and EtOH (≤10 vol%). In C/M, significant differences in structural resistance were observed (FhuA stable ≤10 vol%; FhuA Δ1-159 ≤1 vol%). Deconvolution of CD-spectra for FhuA and FhuA Δ1-159 yielded ß-sheet contents of 61 % (FhuA) and 58% (FhuA Δ1-159). Interestingly, FhuA and FhuA Δ1-159 had comparable ß-sheet contents in the presence and absence of all three organic cosolvents. Additionally, precipitated FhuA and FhuA Δ1-159 (in 40 vol% C/M or 65 vol% THF) redissolved by supplementing the detergent n-octyl-oligo-oxyethylene (oPOE).


Subject(s)
Bacterial Outer Membrane Proteins/drug effects , Escherichia coli Proteins/drug effects , Organic Chemicals/pharmacology , Solvents/pharmacology , Bacterial Outer Membrane Proteins/chemistry , Bacterial Outer Membrane Proteins/genetics , Chloroform/pharmacology , Circular Dichroism , Escherichia coli , Escherichia coli Proteins/chemistry , Escherichia coli Proteins/genetics , Ethanol/pharmacology , Furans/pharmacology , Gene Deletion , Methanol/pharmacology , Mutant Proteins/chemistry , Mutant Proteins/drug effects , Protein Structure, Secondary/drug effects , Protein Structure, Tertiary/drug effects
7.
Bioorg Med Chem ; 20(6): 1887-92, 2012 Mar 15.
Article in English | MEDLINE | ID: mdl-22316554

ABSTRACT

The combination of histone posttranslational modifications occurring in nucleosomal histones determines the epigenetic code. Histone modifications such as acetylation are dynamically controlled in response to a variety of signals during the cell cycle and differentiation, but they are paradoxically maintained through cell division to impart tissue specific gene expression patterns to progeny. The dynamics of histone modifications in living cells are poorly understood, because of the lack of experimental tools to monitor them in a real-time fashion. Recently, FRET-based imaging probes for histone H4 acetylation have been developed, which enabled monitoring of changes in histone acetylation during the cell cycle and drug treatment. Further development of this type of fluorescent probes for other modifications will make it possible to visualize complicated epigenetic regulation in living cells.


Subject(s)
Fluorescence Resonance Energy Transfer/methods , Fluorescent Dyes/chemistry , Histones/metabolism , Protein Processing, Post-Translational , Acetylation/drug effects , Animals , COS Cells , Chlorocebus aethiops , Drug Evaluation, Preclinical/methods , Epigenesis, Genetic , Histone Deacetylase Inhibitors/pharmacology , Histones/chemistry , Histones/genetics , Models, Molecular , Protein Structure, Tertiary/drug effects
8.
Neuro Endocrinol Lett ; 33 Suppl 3: 25-32, 2012.
Article in English | MEDLINE | ID: mdl-23353840

ABSTRACT

OBJECTIVES: The herbal drug aristolochic acid (AA) derived from Aristolochia species has been shown to be the cause of aristolochic acid nephropathy (AAN), Balkan endemic nephropathy (BEN) and their urothelial malignancies. One of the common features of AAN and BEN is that not all individuals exposed to AA suffer from nephropathy and tumor development. One cause for these different responses may be individual differences in the activities of the enzymes catalyzing the biotransformation of AA. Thus, the identification of enzymes principally involved in the metabolism of AAI, the major toxic component of AA, and detailed knowledge of their catalytic specificities is of major importance. Human cytochrome P450 (CYP) 1A1 and 1A2 enzymes were found to be responsible for the AAI reductive activation to form AAI-DNA adducts, while its structurally related analogue, CYP1B1 is almost without such activity. However, knowledge of the differences in mechanistic details of CYP1A1-, 1A2-, and 1B1- mediated reduction is still lacking. Therefore, this feature is the aim of the present study. METHODS: Molecular modeling capable of evaluating interactions of AAI with the active site of human CYP1A1, 1A2 and 1B1 under the reductive conditions was used. In silico docking, employing soft-soft (flexible) docking procedure was used to study the interactions of AAI with the active sites of these human enzymes. RESULTS: The predicted binding free energies and distances between an AAI ligand and a heme cofactor are similar for all CYPs evaluated. AAI also binds to the active sites of CYP1A1, 1A2 and 1B1 in similar orientations. The carboxylic group of AAI is in the binding position situated directly above heme iron. This ligand orientation is in CYP1A1/1A2 further stabilized by two hydrogen bonds; one between an oxygen atom of the AAI nitro-group and the hydroxyl group of Ser122/Thr124; and the second bond between an oxygen atom of dioxolane ring of AAI and the hydroxyl group of Thr497/Thr498. For the CYP1B1:AAI complex, however, any hydrogen bonding of the nitro-group of AAI is prevented as Ser122/Thr124 residues are in CYP1B1 protein replaced by hydrophobic residue Ala133. CONCLUSION: The experimental observations indicate that CYP1B1 is more than 10× less efficient in reductive activation of AAI than CYP1A2. The docking simulation however predicts the binding pose and binding energy of AAI in the CYP1B1 pocket to be analogous to that found in CYP1A1/2. We believe that the hydroxyl group of S122/T124 residue, with its polar hydrogen placed close to the nitro group of the substrate (AAI), is mechanistically important, for example it could provide a proton required for the stepwise reduction process. The absence of a suitable proton donor in the AAI-CYP1B1 binary complex could be the key difference, as the nitro group is in this complex surrounded only by the hydrophobic residues with potential hydrogen donors not closer than 5 Å.


Subject(s)
Aristolochic Acids/adverse effects , Aristolochic Acids/pharmacokinetics , Aryl Hydrocarbon Hydroxylases/metabolism , Cytochrome P-450 CYP1A1/metabolism , Cytochrome P-450 CYP1A2/metabolism , Amino Acid Sequence , Aristolochia/chemistry , Aristolochic Acids/chemistry , Aryl Hydrocarbon Hydroxylases/chemistry , Aryl Hydrocarbon Hydroxylases/genetics , Catalytic Domain/drug effects , Computer Simulation , Cytochrome P-450 CYP1A1/chemistry , Cytochrome P-450 CYP1A1/genetics , Cytochrome P-450 CYP1A2/chemistry , Cytochrome P-450 CYP1A2/genetics , Cytochrome P-450 CYP1B1 , DNA Adducts/chemistry , DNA Adducts/metabolism , Drugs, Chinese Herbal/adverse effects , Drugs, Chinese Herbal/chemistry , Drugs, Chinese Herbal/pharmacokinetics , Humans , Hydrogen Bonding/drug effects , Hydrophobic and Hydrophilic Interactions/drug effects , Kidney Diseases/chemically induced , Models, Chemical , Molecular Sequence Data , Nitroreductases/adverse effects , Nitroreductases/chemistry , Nitroreductases/pharmacokinetics , Protein Structure, Tertiary/drug effects
9.
Dis Model Mech ; 4(6): 822-31, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21810907

ABSTRACT

Recent reports point to small soluble oligomers, rather than insoluble fibrils, of amyloid ß (Aß), as the primary toxic species in Alzheimer's disease. Previously, we developed a low-throughput assay in yeast that is capable of detecting small Aß(42) oligomer formation. Specifically, Aß(42) fused to the functional release factor domain of yeast translational termination factor, Sup35p, formed sodium dodecyl sulfate (SDS)-stable low-n oligomers in living yeast, which impaired release factor activity. As a result, the assay for oligomer formation uses yeast growth to indicate restored release factor activity and presumably reduced oligomer formation. We now describe our translation of this assay into a high-throughput screen (HTS) for anti-oligomeric compounds. By doing so, we also identified two presumptive anti-oligomeric compounds from a sub-library of 12,800 drug-like small molecules. Subsequent biochemical analysis confirmed their anti-oligomeric activity, suggesting that this form of HTS is an efficient, sensitive and cost-effective approach to identify new inhibitors of Aß(42) oligomerization.


Subject(s)
Amyloid beta-Peptides/antagonists & inhibitors , Amyloid beta-Peptides/metabolism , Drug Evaluation, Preclinical/methods , High-Throughput Screening Assays/methods , Peptide Fragments/antagonists & inhibitors , Peptide Fragments/metabolism , Polymerization , Protein Structure, Quaternary/drug effects , Saccharomyces cerevisiae/metabolism , Amyloid beta-Peptides/chemistry , Electrophoresis, Polyacrylamide Gel , Models, Biological , Peptide Fragments/chemistry , Pilot Projects , Protein Structure, Tertiary/drug effects , Reproducibility of Results
10.
Biol Pharm Bull ; 34(4): 523-7, 2011.
Article in English | MEDLINE | ID: mdl-21467640

ABSTRACT

Resveratrol, which is found in grapes, red wine, and berries, has many beneficial health effects, such as anti-cancer, neuro-protective, anti-inflammatory, and life-prolonging effects. However, the cellular mechanisms by which resveratrol acts are relatively unknown, especially in terms of possible regulation of receptors involved in synaptic transmission. 5-Hydroxytryptamine type 3A (5-HT(3A)) receptor is one of several ligand-gated ion channels involved in fast synaptic transmission. In the present study, we investigated the effect of resveratrol on mouse 5-HT(3A) receptor channel activity. 5-HT(3A) receptor was expressed in Xenopus oocytes, and the current was measured using a two-electrode voltage clamp technique. Treatment of resveratrol itself had no effect on the oocytes injected with H(2)O as well as on the oocytes injected with 5-HT(3A) receptor cRNA. In the oocytes injected with 5-HT(3A) receptor cRNA, co- or pre-treatment of resveratrol with 5-HT potentiated 5-HT-induced inward peak current (I(5-HT)) with concentration-, reversible, and voltage-independent manners. The EC(50) of resveratrol was 28.0±2.4 µM. The presence of resveratrol caused a leftward shift of 5-HT concentration-response curve. Protein kinase C (PKC) activator or inhibitor had no effect on resveratrol action on I(5-HT). Site-directed mutations of pre-transmembrane domain 1 (pre-TM1) such as R222A, R222D, R222E, R222K, and R222T abolished or attenuated resveratrol-induced enhancement of I(5-HT), indicating that resveratrol might interact with pre-TM1 of 5-HT(3A) receptor. These results indicate that resveratrol might regulate 5-HT(3A) receptor channel activity via interaction with the N-terminal domain and these results further show that resveratrol-mediated regulation of 5-HT(3A) receptor channel activity might be one of cellular mechanisms of resveratrol action.


Subject(s)
Arginine/metabolism , Ion Channel Gating/drug effects , Plant Extracts/pharmacology , Receptors, Serotonin, 5-HT3/metabolism , Serotonin/metabolism , Stilbenes/pharmacology , Synaptic Transmission/drug effects , Animals , Fruit , Ion Transport/drug effects , Mice , Mutagenesis, Site-Directed , Mutation , Neurotransmitter Agents/pharmacology , Oocytes/drug effects , Oocytes/metabolism , Patch-Clamp Techniques , Protein Structure, Tertiary/drug effects , RNA, Complementary/metabolism , Receptors, Serotonin, 5-HT3/genetics , Resveratrol , Synaptic Membranes/drug effects , Synaptic Membranes/metabolism , Vitis/chemistry , Wine , Xenopus
11.
J Pharmacol Exp Ther ; 335(3): 614-21, 2010 Dec.
Article in English | MEDLINE | ID: mdl-20858708

ABSTRACT

The N-methyl-D-aspartate (NMDA) receptor family regulates various central nervous system functions, such as synaptic plasticity. However, hypo- or hyperactivation of NMDA receptors is critically involved in many neurological and psychiatric conditions, such as pain, stroke, epilepsy, neurodegeneration, schizophrenia, and depression. Consequently, subtype-selective positive and negative modulators of NMDA receptor function have many potential therapeutic applications not addressed by currently available compounds. We have identified allosteric modulators with several novel patterns of NMDA receptor subtype selectivity that have a novel mechanism of action. In a series of carboxylated naphthalene and phenanthrene derivatives, compounds were identified that selectively potentiate responses at GluN1/GluN2A [e.g., 9-iodophenanthrene-3-carboxylic acid (UBP512)]; GluN1/GluN2A and GluN1/GluN2B [9-cyclopropylphenanthrene-3-carboxylic acid (UBP710)]; GluN1/GluN2D [3,5-dihydroxynaphthalene-2-carboxylic acid (UBP551)]; or GluN1/GluN2C and GluN1/GluN2D receptors [6-, 7-, 8-, and 9-nitro isomers of naphth[1,2-c][1,2,5]oxadiazole-5-sulfonic acid (NSC339614)] and have no effect or inhibit responses at the other NMDA receptors. Selective inhibition was also observed; UBP512 inhibits only GluN1/GluN2C and GluN1/GluN2D receptors, whereas 6-bromo-2-oxo-2H-chromene-3-carboxylic acid (UBP608) inhibits GluN1/GluN2A receptors with a 23-fold selectivity compared with GluN1/GluN2D receptors. The actions of these compounds were not competitive with the agonists L-glutamate or glycine and were not voltage-dependent. Whereas the N-terminal regulatory domain was not necessary for activity of either potentiators or inhibitors, segment 2 of the agonist ligand-binding domain was important for potentiating activity, whereas subtype-specific inhibitory activity was dependent upon segment 1. In terms of chemical structure, activity profile, and mechanism of action, these modulators represent a new class of pharmacological agents for the study of NMDA receptor subtype function and provide novel lead compounds for a variety of neurological disorders.


Subject(s)
Membrane Transport Modulators/pharmacology , Receptors, N-Methyl-D-Aspartate/antagonists & inhibitors , Receptors, N-Methyl-D-Aspartate/drug effects , Allosteric Regulation/drug effects , Animals , Binding Sites/physiology , Binding, Competitive , Excitatory Postsynaptic Potentials/drug effects , Excitatory Postsynaptic Potentials/physiology , Glutamic Acid/pharmacology , Glycine/pharmacology , Humans , Membrane Transport Modulators/metabolism , Oocytes/drug effects , Oocytes/physiology , Patch-Clamp Techniques , Protein Structure, Tertiary/drug effects , Protein Structure, Tertiary/genetics , RNA, Complementary/administration & dosage , RNA, Complementary/genetics , Receptors, N-Methyl-D-Aspartate/genetics , Receptors, N-Methyl-D-Aspartate/metabolism , Recombinant Fusion Proteins/drug effects , Recombinant Fusion Proteins/genetics , Sequence Deletion/physiology , Xenopus laevis
12.
Bioorg Med Chem ; 16(10): 5815-25, 2008 May 15.
Article in English | MEDLINE | ID: mdl-18396405

ABSTRACT

Podophyllotoxin (PT), a strong cytotoxic agent from berberidaceae, has been known to inhibit tubulin polymerization. Although PT has been used for developing anticancer drugs as one of seed compounds, clinical treatment by itself has been unsuccessful because of the side effects, except one example in the treatments of warts. In this study, we screened peptides binding to PT with T7 phage display clonings in order to obtain more information about molecular mechanism of the action. A selected phage clone has a specific amino acid sequence to be SVPSRRRPDGRTHRSSRG. A homology search by protein database BLAST showed that this sequence had a similarity to a hinge domain (HD) of E2 protein in human papillomavirus (HPV) type 1a which is known to cause plantar warts. Surface plasmon resonance (SPR) analysis showed that PT bound to a recombinant HPV 1a E2 protein giving a K(D)=24.1microM which has compared with those of other domains of E2 protein. Also we demonstrated whether PT inhibited HD interaction or not. E7 protein of HPV has been known to be an oncoprotein and was reported to interact with HD of E2 protein. We demonstrated that an E2/E7 interaction was inhibited by the addition of PT in this report. And we showed the bindings of PT to other types of HPV. Our results suggest that PT is potential as a tool for clarifying the molecular mechanism of HPV.


Subject(s)
DNA-Binding Proteins/chemistry , DNA-Binding Proteins/drug effects , Oncogene Proteins, Viral/chemistry , Oncogene Proteins, Viral/drug effects , Podophyllotoxin/pharmacology , Animals , Binding Sites , Cell Line, Tumor , Cell Proliferation/drug effects , DNA-Binding Proteins/genetics , Dose-Response Relationship, Drug , Drug Evaluation, Preclinical , Mice , Molecular Conformation , Oncogene Proteins, Viral/genetics , Papillomavirus E7 Proteins , Peptide Library , Podophyllotoxin/chemical synthesis , Podophyllotoxin/chemistry , Protein Binding/drug effects , Protein Structure, Tertiary/drug effects , Stereoisomerism , Structure-Activity Relationship , Time Factors
13.
Org Biomol Chem ; 6(6): 979-81, 2008 Mar 21.
Article in English | MEDLINE | ID: mdl-18327319

ABSTRACT

N-Benzylanilines were designed and synthesized as vascular endothelial growth factor (VEGF)-2 inhibitors using de novo drug design systems based on the X-ray structure of VEGFR-2 kinase domain. Among compounds synthesized, compound showed the most potent inhibitory activity toward VEGFR-2 (KDR) tyrosine kinase and its IC(50) value was 0.57 microM.


Subject(s)
Aniline Compounds/chemical synthesis , Drug Design , Vascular Endothelial Growth Factor Receptor-2/antagonists & inhibitors , Aniline Compounds/chemistry , Aniline Compounds/pharmacology , Cell Line , Dose-Response Relationship, Drug , Drug Evaluation, Preclinical , Endothelium, Vascular/drug effects , Endothelium, Vascular/enzymology , Humans , Models, Biological , Molecular Structure , Phosphorylation/drug effects , Protein Structure, Tertiary/drug effects
14.
Biochemistry ; 46(35): 10016-23, 2007 Sep 04.
Article in English | MEDLINE | ID: mdl-17685560

ABSTRACT

One of the key pathological features of Alzheimer's disease is the aggregation of tau protein. We are therefore searching for compounds capable of inhibiting this reaction. On the basis of an initial screen of 200000 compounds [Pickhardt, M., Gazova, Z., von Bergen, M., Khlistunova, I., Wang, Y., Hascher, A., Mandelkow, E. M., Biernat, J., and Mandelkow, E. (2005) Anthraquinones inhibit tau aggregation and dissolve Alzheimer's paired helical filaments in vitro and in cells, J. Biol. Chem. 280, 3628-3635], we performed an in silico screen and predicted a new phenylthiazolyl-hydrazide (PTH) compound as a possible hit [Larbig, G., Pickhardt, M., Lloyd, D. G., Schmidt, B., and Mandelkow, E. (2007) Screening for inhibitors of tau protein aggregation into Alzheimer paired helical filaments: A ligand based approach results in successful scaffold hopping. Curr. Alzheimer Res. 4 (3), 315-323.]. Synthesis of this compound showed that it was indeed active in terms of inhibiting de novo tau aggregation and disassembling preformed aggregates (IC50 = 7.7 microM and DC50 = 10.8 microM). We have now synthesized 49 similar structures and identified the core of the PTHs to be crucial for activity, thus representing a lead structure. Analysis of the binding epitope by saturation transfer difference NMR shows strong interactions between the tau protein and the ligand in the aromatic regions of the inhibitor. By chemical variation of the core, we improved the inhibitory potency five-fold. The compounds showed a low toxicity as judged by an N2A cell model of tau aggregation and lend themselves for further development.


Subject(s)
Hydrazines/chemistry , Hydrazines/pharmacology , Neurofibrillary Tangles/drug effects , Thiazoles/chemistry , Thiazoles/pharmacology , tau Proteins/chemistry , tau Proteins/metabolism , Animals , Benzothiazoles , Binding Sites/drug effects , Cell Line, Tumor , Cell Survival/drug effects , Drug Evaluation, Preclinical/methods , Humans , Hydrazines/metabolism , Hydrazines/toxicity , Hydro-Lyases/metabolism , Ligands , Models, Biological , Models, Chemical , Protein Binding/drug effects , Protein Denaturation/drug effects , Protein Structure, Tertiary/drug effects , Tauopathies/drug therapy , Tauopathies/prevention & control , Thiazoles/metabolism , Thiazoles/toxicity
15.
Plant Cell ; 18(12): 3519-34, 2006 Dec.
Article in English | MEDLINE | ID: mdl-17172355

ABSTRACT

Phosphatidyl inositol 4,5-bisphosphate (PI 4,5-P2) accumulates in a Rac/Rop-dependent manner in the pollen tube tip plasma membrane, where it may control actin organization and membrane traffic. PI 4,5-P2 is hydrolyzed by phospholipase C (PLC) activity to the signaling molecules inositol 1,4,5-trisphosphate and diacyl glycerol (DAG). To investigate PLC activity during tip growth, we cloned Nt PLC3, specifically expressed in tobacco (Nicotiana tabacum) pollen tubes. Recombinant Nt PLC3 displayed Ca2+-dependent PI 4,5-P2-hydrolyzing activity sensitive to U-73122 and to mutations in the active site. Nt PLC3 overexpression, but not that of inactive mutants, inhibited pollen tube growth. Yellow fluorescent protein (YFP) fused to Nt PLC3, or to its EF and C2 domains, accumulated laterally at the pollen tube tip plasma membrane in a pattern complementary to the distribution of PI 4,5-P2. The DAG marker Cys1:YFP displayed a similar intracellular localization as PI 4,5-P2. Blocking endocytic membrane recycling affected the intracellular distribution of DAG but not of PI 4,5-P2. U-73122 at low micromolar concentrations inhibited and partially depolarized pollen tube growth, caused PI 4,5-P2 spreading at the apex, and abolished DAG membrane accumulation. We show that Nt PLC3 is targeted by its EF and C2 domains to the plasma membrane laterally at the pollen tube tip and that it maintains, together with endocytic membrane recycling, an apical domain enriched in PI 4,5-P2 and DAG required for polar cell growth.


Subject(s)
Cell Membrane/metabolism , Endocytosis , Nicotiana/enzymology , Nicotiana/growth & development , Plant Proteins/metabolism , Pollen Tube/growth & development , Type C Phospholipases/metabolism , Amino Acid Sequence , Animals , Binding Sites/drug effects , Calcium/metabolism , Cell Membrane/drug effects , Cell Membrane/enzymology , Diglycerides/metabolism , Endocytosis/drug effects , Estrenes/pharmacology , Gene Expression Profiling , Gene Expression Regulation, Plant/drug effects , Isoenzymes/chemistry , Isoenzymes/genetics , Isoenzymes/metabolism , Molecular Sequence Data , Mutation/genetics , Phosphatidylinositol 4,5-Diphosphate/metabolism , Plant Proteins/chemistry , Plant Proteins/genetics , Pollen Tube/cytology , Pollen Tube/drug effects , Pollen Tube/enzymology , Protein Structure, Tertiary/drug effects , Protein Transport/drug effects , Pyrrolidinones/pharmacology , RNA, Messenger/genetics , RNA, Messenger/metabolism , Recombinant Proteins/metabolism , Sequence Homology , Substrate Specificity/drug effects , Nicotiana/drug effects , Nicotiana/genetics , Type C Phospholipases/chemistry , Type C Phospholipases/genetics
16.
Brain Res ; 1106(1): 91-98, 2006 Aug 23.
Article in English | MEDLINE | ID: mdl-16836988

ABSTRACT

Ginseng, the root of Panax ginseng C.A. Meyer (Araliaceae), has been extensively used in traditional oriental medicine for the prevention and treatment of aging-related disorders for over 2000 years. Accumulating evidence suggests that ginsenosides such as Rg1 and Rb1, which are the pharmacologically active ingredients of ginseng, modulate neurotransmission. Synapsins are abundant phosphoproteins essential for regulating neurotransmitter release. All synapsins contain a short amino-terminal domain A that is highly conserved and phosphorylated by cAMP-dependent protein kinase (PKA), which plays a key role in regulating neurotransmitter release. In the present study, we demonstrated that both Rg1 and Rb1 increased neurotransmitter release in undifferentiated and differentiated PC12 cells. However, in the presence of the PKA inhibitor H89, Rg1, but not Rb1, still induced neurotransmitter release. Moreover, Rb1, but not Rg1, enhanced the phosphorylation of synapsins via PKA pathway. In summary, Rb1 promotes neurotransmitter release by increasing the phosphorylation of synapsins through the PKA pathway, whereas the similar effects observed with Rg1 are independent of the phosphorylation of synapsins.


Subject(s)
Cyclic AMP-Dependent Protein Kinases/drug effects , Ginsenosides/pharmacology , Neurotransmitter Agents/metabolism , Presynaptic Terminals/drug effects , Synapsins/drug effects , Synaptic Transmission/drug effects , Animals , Animals, Newborn , Brain/drug effects , Brain/metabolism , Cyclic AMP/metabolism , Cyclic AMP-Dependent Protein Kinases/metabolism , Drugs, Chinese Herbal/pharmacology , Enzyme Inhibitors/pharmacology , Male , Nootropic Agents/pharmacology , PC12 Cells , Phosphorylation/drug effects , Presynaptic Terminals/metabolism , Protein Structure, Tertiary/drug effects , Protein Structure, Tertiary/physiology , Rats , Rats, Sprague-Dawley , Signal Transduction/drug effects , Signal Transduction/physiology , Synapsins/metabolism , Synaptic Transmission/physiology , Synaptosomes , Up-Regulation/drug effects , Up-Regulation/physiology
17.
Mol Cell Endocrinol ; 217(1-2): 221-7, 2004 Mar 31.
Article in English | MEDLINE | ID: mdl-15134821

ABSTRACT

The mineralocorticoid receptor (MR) plays a role in congestive heart failure; however, the molecular mechanism(s) remains undefined. We hypothesized that interaction of the MR with a cardiac protein modulates the transcriptional activation function of the MR within the heart. We used the yeast two-hybrid technique to screen a human heart library and found an aldosterone-dependent interaction between the hMR and the cardiac myosin binding protein (cMBP-c). The EC(50) of the hMR-MBP-c interaction was approximately 80nM, and the cMBP-c did not interact with the glucocorticoid receptor (GR). The GST pull-down technique was used to confirm an interaction between the MR and the cMBP-c as well as the lack of interaction with the GR. Spironolactone partially blocked this interaction, further suggesting MR specificity. We also determined the cMBP-c binding site lies within the C-terminus of the MR. We propose that interaction of the MR with cMBP-c may play a role in cardiac remodeling.


Subject(s)
Aldosterone/pharmacology , Carrier Proteins/metabolism , Myocardium/metabolism , Receptors, Mineralocorticoid/metabolism , Carrier Proteins/genetics , Cloning, Molecular , DNA, Complementary/genetics , Gene Library , Humans , Protein Binding/drug effects , Protein Binding/genetics , Protein Structure, Tertiary/drug effects , Protein Structure, Tertiary/genetics , Receptors, Mineralocorticoid/genetics , Two-Hybrid System Techniques , Yeasts
18.
J Mol Neurosci ; 20(3): 323-6, 2003.
Article in English | MEDLINE | ID: mdl-14501015

ABSTRACT

Factors limiting the therapeutic application of neurotrophins to neurodegenerative diseases include poor stability and CNS penetration. Moreover, certain neurotrophin effects, such as promotion of neuronal death via interaction with the p75NTR receptor, might further limit their application. We have proposed that development of small molecule mimetics of neurotrophins might serve to overcome these limitations. In previous work, our laboratory established the proof-of-principle that mimetics of specific nerve growth factor (NGF) domains could prevent neuronal death. Peptidomimetics of the loop 1 domain prevent death via p75NTR-dependent signaling and peptidomimetics of the loop 4 domain prevent death via Trk-related signaling. In current work we are designing pharmacophore queries corresponding to loop domains 1 or 4 that incorporate features of the NGF crystal structure along with features derived from peptidomimetic structure-activity-relationships. Screening of in silico databases containing non-peptide, small molecules has identified a number of candidate NGF domain mimetics. Preliminary assessment of these compounds using neurotrophin bioassays indicates that several are capable of preventing neuronal death. Ongoing studies will determine whether these compounds act via p75NTR or Trk receptors.


Subject(s)
Alzheimer Disease/drug therapy , Molecular Mimicry/physiology , Nerve Growth Factors/pharmacology , Receptor, trkA/drug effects , Receptors, Nerve Growth Factor/drug effects , Animals , Cells, Cultured , Chick Embryo , Drug Design , Drug Evaluation, Preclinical , Mice , Molecular Conformation , Molecular Structure , Nerve Growth Factors/chemistry , Neuroprotective Agents/chemistry , Neuroprotective Agents/pharmacology , Protein Structure, Tertiary/drug effects , Protein Structure, Tertiary/physiology , Receptor, Nerve Growth Factor , Receptor, trkA/chemistry , Receptors, Nerve Growth Factor/chemistry
19.
Mol Cell ; 12(1): 27-37, 2003 Jul.
Article in English | MEDLINE | ID: mdl-12887890

ABSTRACT

Bcr-Abl is a dysregulated tyrosine kinase whose mechanism of activation is unclear. Here, we demonstrate that, like c-Abl, Bcr-Abl is negatively regulated through its SH3 domain. Kinase activity, transformation, and leukemogenesis by Bcr-Abl are greatly impaired by mutations of the Bcr coiled-coil domain that disrupt oligomerization, but restored by an SH3 point mutation that blocks ligand binding or a complementary mutation at the intramolecular SH3 binding site defined in c-Abl. Phosphorylation of tyrosines in the activation loop of the catalytic domain and the linker between the SH2 and catalytic domains (SH2-CD linker) is dependent on oligomerization and required for leukemogenesis. These results suggest that Bcr-Abl has a monomeric, unphosphorylated state with the SH3 domain engaged intramolecularly to Pro1124 in the SH2-CD linker, the form that is sensitive to the inhibitor imatinib (STI-571). The sole function of the coiled-coil domain is to disrupt the autoinhibited conformation through oligomerization and intermolecular autophosphorylation.


Subject(s)
Cell Transformation, Neoplastic/metabolism , Eukaryotic Cells/enzymology , Feedback, Physiological/genetics , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/enzymology , Protein-Tyrosine Kinases/metabolism , 3T3 Cells , Alanine/genetics , Alanine/metabolism , Amino Acid Sequence/genetics , Animals , Binding Sites/genetics , Catalytic Domain/drug effects , Catalytic Domain/genetics , Cell Transformation, Neoplastic/genetics , Enzyme Inhibitors/pharmacology , Feedback, Physiological/drug effects , Fusion Proteins, bcr-abl , Humans , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/genetics , Mice , Mice, Inbred BALB C , Models, Molecular , Mutation/drug effects , Mutation/genetics , Phosphorylation/drug effects , Proline/genetics , Proline/metabolism , Protein Binding/drug effects , Protein Binding/genetics , Protein Structure, Tertiary/drug effects , Protein Structure, Tertiary/genetics , Protein-Tyrosine Kinases/antagonists & inhibitors , Protein-Tyrosine Kinases/genetics , Tyrosine/metabolism
20.
Neuroscience ; 104(2): 599-607, 2001.
Article in English | MEDLINE | ID: mdl-11377859

ABSTRACT

Syntaxin 1A has a pronounced inhibitory effect on the activation kinetics and current amplitude of voltage-gated Ca(2+) channels. This study explores the molecular basis of syntaxin interaction with N- and Lc-type Ca(2+) channels by way of functional assays of channel gating in a Xenopus oocytes expression system. A chimera of syntaxin 1A and syntaxin 2 in which the transmembrane domain of syntaxin 2 replaced the transmembrane of syntaxin 1A (Sx1-2), significantly reduced the rate of activation of N- and Lc-channels. This shows a similar effect to that demonstrated by syntaxin 1A, though the current was not inhibited. The major sequence differences at the transmembrane of the syntaxin isoforms are that the two highly conserved cysteines Cys 271 and Cys 272 in syntaxin 1A correspond to the valines Val 272 and Val 273 in syntaxin 2 transmembrane. Mutating either cysteines in Sx1-1 (syntaxin 1A) to valines, did not affect modulation of the channel while a double mutant C271/272V was unable to regulate inward current. Transfer of these two cysteines to the transmembrane of syntaxin 2 by mutating Val 272 and Val 273 to Cys 272 and Cys 273 led to channel inhibition. When cleaved by botulinum toxin, the syntaxin 1A fragments, amino acids 1-253 and 254-288, which includes the transmembrane domain, were both unable to inhibit current amplitude but retained the ability to modify the activation kinetics of the channel. A full-length syntaxin 1A and the integrity of the two cysteines within the transmembrane are crucial for coordinating Ca(2+) entry through the N- and Lc-channels. These results suggest that upon membrane depolarization, the voltage-gated N- and Lc-type Ca(2+)-channels signal the exocytotic machinery by interacting with syntaxin 1A at the transmembrane and the cytosolic domains. Cleavage with botulinum toxin disrupts the coupling of the N- and Lc-type channels with syntaxin 1A and abolishes exocytosis, supporting the hypothesis that these channels actively participate in Ca(2+) regulated secretion.


Subject(s)
Antigens, Surface/metabolism , Calcium Channels, L-Type/metabolism , Calcium Channels, N-Type/metabolism , Cell Membrane/metabolism , Membrane Potentials/genetics , Mutation/physiology , Nerve Tissue Proteins/metabolism , Recombinant Fusion Proteins/metabolism , Amino Acid Sequence/genetics , Animals , Antigens, Surface/genetics , Calcium Channels, L-Type/drug effects , Calcium Channels, N-Type/drug effects , Cell Membrane/drug effects , Cysteine/genetics , Cysteine/metabolism , Female , Gene Expression Regulation/physiology , Kinetics , Membrane Potentials/drug effects , Nerve Tissue Proteins/genetics , Oocytes/cytology , Oocytes/drug effects , Oocytes/metabolism , Protein Structure, Tertiary/drug effects , Protein Structure, Tertiary/physiology , RNA, Complementary/pharmacology , Recombinant Fusion Proteins/genetics , Syntaxin 1 , Xenopus laevis
SELECTION OF CITATIONS
SEARCH DETAIL