Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 129
Filter
1.
Eye (Lond) ; 31(10): 1399-1408, 2017 Oct.
Article in English | MEDLINE | ID: mdl-28983094

ABSTRACT

PurposeThe aim of this study was to determine a sequence of structural changes in acute posterior multifocal placoid pigment epitheliopathy (APMPPE) using optical coherence tomography-angiography (OCT-A) and comparing with other imaging modalities.Patients and methodsPatients with a new diagnosis of acute-onset APMPPE referred to a regional specialist centre from October 2015 to October 2016 were included. Multimodal imaging employed on all patients from diagnosis included the following: fundus fluorescein angiography, indocyanine green angiography, fundus autofluorescence, spectral domain-OCT (SD-OCT), and OCT-A. All non-invasive imaging processes were repeated during follow-up.ResultsTen eyes of five patients were included in the study, three males and two females, with a mean age of 26.2 years (range: 21-32) and a mean follow-up of 6.4 months (range: 2.6-13.3). All patients presented with bilateral disease and macular involving lesions. OCT-A imaging of the choriocapillaris was supportive of hypoperfusion at the site of APMPPE lesions during the acute phase of this condition with normalisation of choroidal vasculature during follow-up. Multimodal imaging consistently highlighted four sequential phases from presentation to resolution of active disease.ConclusionsMultimodal imaging in patients with APMPPE in acute and long-term follow-up demonstrates a reversible choroidal hypoperfusion supporting the primary inciting pathology as a choriocapillaritis. The evolution shows resolution of the ischaemia through a defined sequence that results in persistent changes at the level of the retinal pigment epithelium and outer retina. OCT-A was able to detect preclinical changes and chart resolution at the level of the choriocapillaris.


Subject(s)
Choroiditis/diagnosis , Fluorescein Angiography/methods , Macula Lutea/pathology , Multimodal Imaging , Posterior Eye Segment/pathology , Tomography, Optical Coherence/methods , Acute Disease , Adult , Choroiditis/physiopathology , Disease Progression , Female , Follow-Up Studies , Fundus Oculi , Humans , Male , Multifocal Choroiditis , Reproducibility of Results , Retinal Pigment Epithelium/pathology , Retrospective Studies , Visual Acuity , Young Adult
3.
Curr Med Chem ; 19(33): 5726-34, 2012.
Article in English | MEDLINE | ID: mdl-22934808

ABSTRACT

Plague is an infectious disease with a high mortality rate that has repeatedly impacted human society. It remains a threat in many parts of the world today. Plague is caused by the bacterium, Yersinia pestis (Y. pestis), which has as one of its required virulence factors, the protein-tyrosine phosphatase, YopH. Therefore, YopH represents a potential target for the treatment of Y. pestis infection. Recent recognition of Y. pestis as a possible bioterrorism agent and the fact that it is still the cause of endemic disease around the world make it an important object of study and heighten the need for new anti-plague agents. The current review covers aspects of plague and its historical occurrence and summarizes approaches to developing YopH inhibitors.


Subject(s)
Anti-Bacterial Agents/chemistry , Anti-Bacterial Agents/pharmacology , Bacterial Outer Membrane Proteins/antagonists & inhibitors , Plague/drug therapy , Protein Tyrosine Phosphatases/antagonists & inhibitors , Virulence Factors/antagonists & inhibitors , Yersinia pestis/enzymology , Bacterial Outer Membrane Proteins/chemistry , Bacterial Outer Membrane Proteins/metabolism , Drug Discovery , History, 19th Century , History, 20th Century , History, 21st Century , History, Medieval , Humans , Models, Molecular , Plague/history , Plague/microbiology , Protein Tyrosine Phosphatases/chemistry , Protein Tyrosine Phosphatases/metabolism , Small Molecule Libraries/chemistry , Small Molecule Libraries/pharmacology , Virulence Factors/chemistry , Virulence Factors/metabolism , Yersinia pestis/chemistry , Yersinia pestis/drug effects
4.
Neurochem Res ; 35(6): 888-93, 2010 Jun.
Article in English | MEDLINE | ID: mdl-19937113

ABSTRACT

HIV-1 integrase (IN) catalyzes integration of viral DNA into cell DNA through 3'-processing of viral DNA and strand transfer reactions. To learn on binding of IN to DNAs and IN inhibition we applied spectroscopy (circular dichroism, fluorescence) in a simplified model consisting in a peptide analogue (K156) of alpha4 helix involved in recognition of viral and cell DNA; an oligonucleotide corresponding to the U5' LTR DNA end; and an inhibitor (TB11) of the diketo acid (DKA) family. Results extrapolated to IN show that: the enzyme binds viral DNA with high affinity and specificity, but cell DNA with low affinity and specificity; the affinity of TB11 for IN is high enough to impair the binding of IN to cell DNA, but not to viral DNA. This explains why TB11 is an inhibitor of strand transfer but not of 3'-processing. These results can help in the search of new IN inhibitors.


Subject(s)
DNA/chemistry , HIV Integrase Inhibitors/chemistry , HIV Integrase/chemistry , Circular Dichroism , DNA, Viral/chemistry , Dimerization , HIV-1/enzymology , HIV-1/genetics , Ketones/chemistry , Models, Molecular , Peptides/chemistry , Protein Binding , Protein Structure, Secondary , Spectrometry, Fluorescence , Virus Integration
5.
Antimicrob Agents Chemother ; 45(9): 2510-6, 2001 Sep.
Article in English | MEDLINE | ID: mdl-11502522

ABSTRACT

To study the effect of potential human immunodeficiency virus type 1 (HIV-1) integrase inhibitors during virus replication in cell culture, we used a modified nested Alu-PCR assay to quantify integrated HIV DNA in combination with the quantitative analysis of extrachromosomal HIV DNA. The two diketo acid integrase inhibitors (L-708,906 and L-731,988) blocked the accumulation of integrated HIV-1 DNA in T cells following infection but did not alter levels of newly synthesized extrachromosomal HIV DNA. In contrast, we demonstrated that L17 (a member of the bisaroyl hydrazine family of integrase inhibitors) and AR177 (an oligonucleotide inhibitor) blocked the HIV replication cycle at, or prior to, reverse transcription, although both drugs inhibited integrase activity in cell-free assays. Quercetin dihydrate (a flavone) was shown to not have any antiviral activity in our system despite reported anti-integration properties in cell-free assays. This refined Alu-PCR assay for HIV provirus is a useful tool for screening anti-integration compounds identified in biochemical assays for their ability to inhibit the accumulation of integrated HIV DNA in cell culture, and it may be useful for studying the effects of these inhibitors in clinical trials.


Subject(s)
DNA, Viral/drug effects , HIV Integrase Inhibitors/pharmacology , HIV Integrase/metabolism , HIV-1/drug effects , Acetoacetates/pharmacology , Cells, Cultured , DNA, Viral/physiology , HIV Integrase/drug effects , HIV-1/enzymology , HIV-1/physiology , Humans , Hydrazines/pharmacology , Microbial Sensitivity Tests , Oligonucleotides/pharmacology , Virus Replication
6.
J Med Chem ; 44(18): 2869-78, 2001 Aug 30.
Article in English | MEDLINE | ID: mdl-11520195

ABSTRACT

Examination of the PTP1B inhibitory potency of an extensive series of phosphotyrosyl (pTyr) mimetics (Xxx) expressed in the EGFr-derived hexapeptide platform Ac-Asp-Ala-Asp-Xxx-Leu-amide previously led to the finding of high inhibitory potency when Xxx = 4-(phosphonodifluoromethyl)phenylalanyl (F2Pmp) (K(i) = 0.2 microM) and when Xxx = 3-carboxy-4-carboxymethyloxyphenylalanyl (K(i) = 3.6 microM). In the first instance, further work led from the F2Pmp-containing peptide to monomeric inhibitor, 6-(phosphonodifluoromethyl)-2-naphthoic acid (K(i) = 22 microM), and to the pseudo-dipeptide mimetic, N-[6-(phosphonodifluoromethyl)-2-naphthoyl]-glutamic acid (K(i) = 12 microM). In the current study, a similar approach was applied to the 3-carboxy-4-carboxymethyloxyphenylalanyl-containing peptide, which led to the preparation of monomeric 5-carboxy-6-carboxymethyloxy-2-naphthoic acid (K(i) = 900 microM). However, contrary to expectations based on the aforementioned F2Pmp work, incorporation of this putative pTyr mimetic into the pseudo-dipeptide, N-[5-carboxy-6-carboxymethyloxy-2-naphthoyl]-glutamic acid, resulted in a substantial loss of binding affinity. A reevaluation of binding orientation for 5-carboxy-6-carboxymethyloxy-2-naphthoic acid was therefore undertaken, which indicated a 180 degrees reversal of the binding orientation within the PTP1B catalytic site. In the new orientation, the naphthyl 2-carboxyl group, and not the o-carboxy carboxymethyloxy groups, mimics a phosphoryl group. Indeed, when 5-carboxy-2-naphthoic acid itself was examined at neutral pH for inhibitory potency, it was found to have K(i) = 31 +/- 7 microM, which is lower than parent 5-carboxy-6-carboxymethyloxy-2-naphthoic acid. In this fashion, 5-carboxy-2-naphthoic acid (or more appropriately, 6-carboxy-1-naphthoic acid) has been identified as a novel PTP1B binding motif.


Subject(s)
Enzyme Inhibitors/chemistry , Naphthalenes/chemical synthesis , Oligopeptides/chemical synthesis , Protein Tyrosine Phosphatases/antagonists & inhibitors , Catalytic Domain , Humans , Models, Molecular , Molecular Conformation , Molecular Mimicry , Naphthalenes/chemistry , Oligopeptides/chemistry , Protein Binding , Protein Tyrosine Phosphatase, Non-Receptor Type 1 , Protein Tyrosine Phosphatases/chemistry , Recombinant Proteins/chemistry
7.
Bioorg Med Chem Lett ; 11(14): 1889-92, 2001 Jul 23.
Article in English | MEDLINE | ID: mdl-11459654

ABSTRACT

Grubbs' olefin metathesis reaction was utilized to prepare a macrocyclic variant of a linear Grb2 SH2 domain antagonist in an attempt to induce a beta-bend conformation known to be required for high affinity binding. In extracellular Grb2 SH2 domain binding assays, the macrocyclic analogue exhibited an approximate 100-fold enhancement in binding potency relative to its linear counterpart. The macrocycle was not as effective in whole cell binding assays as would be expected based on its extracellular binding potency.


Subject(s)
Adaptor Proteins, Signal Transducing , Peptides/chemistry , Proteins/antagonists & inhibitors , Proteins/metabolism , Receptors, Drug/chemistry , Tyrosine/metabolism , Tyrosine/pharmacology , src Homology Domains/drug effects , Breast Neoplasms/pathology , Cell Division/drug effects , Cyclization , Drug Design , Enzyme-Linked Immunosorbent Assay , GRB2 Adaptor Protein , Humans , Inhibitory Concentration 50 , Models, Molecular , Molecular Conformation , Peptides/metabolism , Protein Binding/physiology , Receptors, Drug/metabolism , Signal Transduction/physiology , Structure-Activity Relationship , Tumor Cells, Cultured/drug effects , Tumor Cells, Cultured/metabolism , Tyrosine/analogs & derivatives , src Homology Domains/physiology
8.
Bioorg Med Chem Lett ; 11(10): 1265-8, 2001 May 21.
Article in English | MEDLINE | ID: mdl-11392533

ABSTRACT

Azatyrosine (AzaTyr, 4) is a natural product isolated from Streptomyces chibanesis, whose structure is characterized by a nitrogen atom in the aryl ring of a tyrosyl residue. This seemingly minor modification to the tyrosyl residue results in profound physiological effects, as AzaTyr has been shown to promote permanent reversion of ras-dependent transformed cells to the normal phenotype in culture and to inhibit chemical induction of carcinogenesis in transgenic mice bearing oncogenic human ras. The mechanisms underlying these effects are not known, however ras-pathways involve an intricate balance between both protein-tyrosine kinases (PTKs) and protein-tyrosine phosphatases (PTPs). The present study was undertaken to examine the general utility of AzaTyr as a structural motif for PTP inhibitor design by examining the phospho-azatyrosine (pAzaTyr)-containing peptide Ac-Asp-Ala-Asp-Glu-pAzaTyr-Leu-amide (8) in a PTP1 enzyme system. Kinetic analysis indicated that 8 binds with a Km value of 210 microM and a catalytic turnover rate, kcat of 52 s(-1). This represents a greater than 50-fold reduction in binding affinity relative to the parent phosphotyrosine-containing peptide, indicating that the aryl nitrogen adversely affects binding affinity. The much lower PTP affinity of the pAzaTyr-containing peptide reduces the potential utility of the AzaTyr pharmacophore for PTP inhibitor design. These results are discussed from the point of view that incorporation of AzaTyr residues into proteins could result in perturbation of protein-tyrosine phosphorylation,dephosphorylation cascades that control signal transduction processes, including ras-dependent pathways.


Subject(s)
Alanine/chemical synthesis , Alanine/pharmacology , Anticarcinogenic Agents/chemical synthesis , Phosphotyrosine/analogs & derivatives , Protein Tyrosine Phosphatases/antagonists & inhibitors , Alanine/analogs & derivatives , Animals , Anticarcinogenic Agents/pharmacology , Catalytic Domain/genetics , Drug Design , Enzyme Inhibitors/chemical synthesis , Enzyme Inhibitors/pharmacology , Humans , Kinetics , Molecular Mimicry , Peptides/chemistry , Phosphotyrosine/pharmacology , Structure-Activity Relationship , Substrate Specificity
9.
Bioorg Med Chem ; 9(7): 1649-57, 2001 Jul.
Article in English | MEDLINE | ID: mdl-11425564

ABSTRACT

Integrase is an enzyme found in human immunodeficiency virus, which is required for the viral life cycle, yet has no human cellular homologue. For this reason, HIV integrase (IN) has become an important target for the development of new AIDS therapeutics. Irreversible affinity ligands have proven to be valuable tools for studying a number of enzyme and protein systems, yet to date there have been no reports of such affinity ligands for the study of IN. As an initial approach toward irreversible ligand design directed against IN, we appended isothiocyanate functionality onto caffeic acid phenethyl ester (CAPE), a known HIV integrase inhibitor. The choice of isothiocyanate as the reactive functionality, was based on its demonstrated utility in the preparation of affinity ligands directed against a number of other protein targets. Several isomeric CAPE isothiocyanates were prepared to explore the enzyme topography for reactive nitrogen and sulfur nucleophiles vicinal to the enzyme-bound CAPE. The preparation of these CAPE isothiocyanates, required development of new synthetic methodology which employed phenyl thiocarbamates as latent isothiocyanates which could be unmasked near the end of the synthetic sequence. When it was observed that beta-mercaptoethanol (beta-ME), which is required to maintain the catalytic activity of soluble IN (a F185KC280S mutant), reacted with CAPE isothiocyanate functionality to form the corresponding hydroxyethylthiocarbamate, a variety of mutant IN were examined which did not require the presence of beta-ME for catalytic activity. Although in these latter enzymes, CAPE isothiocyanate functionality was presumed to be present and available for acylation by IN nucleophiles, they were equally effective against Cys to Ser mutants. One conclusion of these studies, is that upon binding of CAPE to the integrase, nitrogen or sulfur nucleophiles may not be properly situated in the vicinity of the phenethyl aryl ring to allow reaction with and covalent modification of reactive functionality, such as isothiocyanate groups. The fact that introduction of the isothiocyanate group onto various positions of the phenethyl ring or replacement of the phenyl ring with naphthyl rings, failed to significantly affect inhibitory potency, indicates a degree of insensitivity of this region of the molecule toward structural modification. These findings may be useful in future studies concerned with the development and use of HIV-1 integrase affinity ligands.


Subject(s)
Caffeic Acids/metabolism , HIV Integrase/metabolism , HIV-1/enzymology , Isothiocyanates/chemistry , Amino Acid Substitution , Base Sequence , Caffeic Acids/chemistry , Cysteine/chemistry , Esters , HIV Integrase/chemistry , HIV Long Terminal Repeat , Ligands , Magnetic Resonance Spectroscopy , Oligodeoxyribonucleotides , Serine/chemistry , Spectrometry, Mass, Fast Atom Bombardment
10.
Org Lett ; 3(11): 1617-20, 2001 May 31.
Article in English | MEDLINE | ID: mdl-11405669

ABSTRACT

One drawback frequently associated with olefin metathesis-mediated peptide macrocyclization, the loss of side chain functionality at sites of ring closure, may be circumvented by incorporation of side chain functionality within the ring-closing olefin segments. This approach is demonstrated in the preparation of a macrocyclic Grb2 SH2 domain antagonist designed as a conformationally constrained beta-bend mimic.


Subject(s)
Adaptor Proteins, Signal Transducing , Alkenes/chemical synthesis , ErbB Receptors/chemistry , Proteins/chemistry , src Homology Domains/drug effects , Alkenes/pharmacology , Catalysis , Chromatography, High Pressure Liquid , Drug Design , GRB2 Adaptor Protein , Molecular Conformation , Ruthenium
11.
Bioorg Med Chem ; 9(6): 1439-45, 2001 Jun.
Article in English | MEDLINE | ID: mdl-11408162

ABSTRACT

High affinity binding of peptides to Src homology 2 (SH2) domains, often requires the presence of phosphotyrosyl (pTyr) or pTyr-mimicking moieties in the N-terminal position of the binding ligand. Several reports have shown that N(alpha)-acylation of the critical pTyr residue can result in increased SH2 domain binding potency. For Grb2 SH2 domains which recognize pTyr-Xxx-Asn-NH(2) motifs, significant potency enhancement can be incurred by N(alpha)-(3-amino)Z derivatization of tripeptides such as pTyr-Ile-Asn-NH(2). Using ligands based on the high affinity pY-Ac(6)c-Asn-(naphthylpropylamide) motif, (where Ac(6)c=1-aminocyclohexanecarboxylic acid), additional reports have shown moderate potentiating effects of N(alpha)-oxalyl derivatization. The current study examined variations of the N(alpha)-oxalyl theme in the context of a Xxx-Ac(6)c-Asn-(naphthylpropylamide) platform, where Xxx=the hydrolytically stable pTyr mimetics phosphonomethyl phenylalanine (Pmp) or carboxymethyl phenylalanine (Cmf). The effects of N(alpha)-(3-amino)Z derivatization were also investigated for this platform, to ascertain whether the large binding enhancement reported for tripeptides such as pTyr-Ile-Asn-NH(2) could be observed. In ELISA-based extracellular Grb2 SH2 domain binding assays, it was found for the Pmp-based series, that extending the oxalyl carboxyl out by one methylene unit or replacing carboxyl functionality with a tetrazole isostere, resulted in binding potency greater than the parent N(alpha)-acetyl-containing compound, with enhancement approximating that observed for the N(alpha)-oxalyl derivative. When Cmf was used as the pTyr mimetic, only modest differences in IC(50) values were observed for the series. Examination of the N(alpha)-(3-amino)Z derivatized Pmp-Ac(6)c-Asn-(naphthylpropylamide), showed that binding affinity was reduced relative to the parent N(alpha)-acetyl analogue, in contrast to the reported significant enhancement of affinity observed with other peptide ligands. Treatment of MDA-453 tumor cells, which are mitogenically driven through erbB-2 tyrosine kinase-dependent pathways, with Pmp-containing inhibitors resulted in growth inhibition, with the N(alpha)-oxalyl and N(alpha)-malonyl-containing compounds exhibiting IC(50) values (4.3 and 4.6 microM, respectively) approximately five-fold lower than the parent N(alpha)-acetyl-containing compound. Tetrazole and N(alpha)-(3-amino)Z-containing inhibitors were from two- to four-fold less potent than these latter analogues in the growth inhibition assays.


Subject(s)
Adaptor Proteins, Signal Transducing , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Naphthalenes/chemistry , Naphthalenes/pharmacology , Oligopeptides/chemistry , Oligopeptides/pharmacology , Proteins/drug effects , Proteins/metabolism , src Homology Domains/drug effects , Amides/chemistry , Antineoplastic Agents/metabolism , Binding Sites , Drug Screening Assays, Antitumor/methods , Enzyme-Linked Immunosorbent Assay , GRB2 Adaptor Protein , Humans , Inhibitory Concentration 50 , Ligands , Oligopeptides/metabolism , Structure-Activity Relationship , Tetrazoles/chemistry , Tumor Cells, Cultured
12.
Biopolymers ; 60(1): 32-44, 2001.
Article in English | MEDLINE | ID: mdl-11376431

ABSTRACT

The central roles played by protein-tyrosine kinase (PTK)-dependent signal transduction in normal cellular regulation and homeostasis have made inappropriate or aberrant functions of certain of these pathways contributing factors to a variety of diseases, including several cancers. For this reason, development of PTK signaling inhibitors has evolved into an important approach toward new therapeutics. Since in these pathways phosphotyrosyl (pTyr) residues provide unique and defining functions either by their creation under the catalysis of PTKs, their recognition and binding by protein modules such as SH2 and phosphotyrosyl binding (PTB) domains, or their destruction by protein-tyrosine phosphatases, pTyr mimetics provide useful general starting points for inhibitor design. Important considerations in the development of such pTyr mimetics include enzymatic stability (particularly toward PTPs), high affinity recognition by target pTyr binding proteins, and good cellular bioavailability. Although small molecule, nonpeptide inhibitors may be ultimate objectives of inhibitor development, peptides frequently serve as display platforms for pTyr mimetics, which afford useful and conceptually straightforward starting points in the development process. Reported herein is a limited overview of pTyr mimetic development as it relates to peptide-based agents. Of particular interest are recent findings that highlight potential limitations of peptides as display platforms for the identification of small molecule leads. One conclusion that results from this work is that while peptide-based approaches toward small molecule inhibitor design are often intellectually satisfying from a structure-based perspective, extrapolation of negative findings to small molecule, nonpeptide contexts should be undertaken with extreme caution.


Subject(s)
Molecular Mimicry , Peptides/pharmacology , Phosphotyrosine/chemistry , Phosphotyrosine/pharmacology , Protein Tyrosine Phosphatases/antagonists & inhibitors , Protein-Tyrosine Kinases/antagonists & inhibitors , Signal Transduction/drug effects , Binding Sites , Drug Design , Models, Molecular , Peptides/chemistry , Peptides/metabolism , Phosphotyrosine/metabolism , Protein Binding/drug effects , Protein Conformation , Protein Tyrosine Phosphatases/chemistry , Protein Tyrosine Phosphatases/metabolism , Protein-Tyrosine Kinases/chemistry , Protein-Tyrosine Kinases/metabolism , src Homology Domains
13.
J Biol Chem ; 276(17): 14308-14, 2001 Apr 27.
Article in English | MEDLINE | ID: mdl-11278639

ABSTRACT

Hepatocyte growth factor (HGF) stimulates mitogenesis, motogenesis, and morphogenesis in a wide range of cellular targets during development, homeostasis and tissue regeneration. Inappropriate HGF signaling occurs in several human cancers, and the ability of HGF to initiate a program of protease production, cell dissociation, and motility has been shown to promote cellular invasion and is strongly linked to tumor metastasis. Upon HGF binding, several tyrosines within the intracellular domain of its receptor, c-Met, become phosphorylated and mediate the binding of effector proteins, such as Grb2. Grb2 binding through its SH2 domain is thought to link c-Met with downstream mediators of cell proliferation, shape change, and motility. We analyzed the effects of Grb2 SH2 domain antagonists on HGF signaling and observed potent blockade of cell motility, matrix invasion, and branching morphogenesis, with ED(50) values of 30 nm or less, but only modest inhibition of mitogenesis. These compounds are 1000-10,000-fold more potent anti-motility agents than any previously characterized Grb2 SH2 domain antagonists. Our results suggest that SH2 domain-mediated c-Met-Grb2 interaction contributes primarily to the motogenic and morphogenic responses to HGF, and that these compounds may have therapeutic application as anti-metastatic agents for tumors where the HGF signaling pathway is active.


Subject(s)
Adaptor Proteins, Signal Transducing , Hepatocyte Growth Factor/metabolism , Proteins/metabolism , src Homology Domains , Animals , Cell Line , Cell Movement , Collagen/metabolism , DNA, Complementary/metabolism , Dogs , Dose-Response Relationship, Drug , Extracellular Matrix/metabolism , GRB2 Adaptor Protein , Hepatocyte Growth Factor/chemistry , Hepatocyte Growth Factor/genetics , Humans , Immunoblotting , Phosphorylation , Precipitin Tests , Protein Structure, Tertiary , Proteins/antagonists & inhibitors , Proteins/genetics , Proto-Oncogene Proteins c-met/metabolism , Signal Transduction , Time Factors , Transfection
16.
J Biol Chem ; 275(52): 40817-26, 2000 Dec 29.
Article in English | MEDLINE | ID: mdl-11007796

ABSTRACT

Glucose serves as both a nutrient and regulator of physiological and pathological processes. Presently, we found that glucose and certain sugars rapidly activated extracellular signal-regulated kinase (ERK) by a mechanism that was: (a) independent of glucose uptake/metabolism and protein kinase C but nevertheless cytochalasin B-inhibitable; (b) dependent upon proline-rich tyrosine kinase-2 (PYK2), GRB2, SOS, RAS, RAF, and MEK1; and (c) amplified by overexpression of the Glut1, but not Glut2, Glut3, or Glut4, glucose transporter. This amplifying effect was independent of glucose uptake but dependent on residues 463-468, IASGFR, in the Glut1 C terminus. Accordingly, glucose effects on ERK were amplified by expression of Glut4/Glut1 or Glut2/Glut1 chimeras containing IASGFR but not by Glut1/Glut4 or Glut1/Glut2 chimeras lacking these residues. Also, deletion of Glut1 residues 469-492 was without effect, but mutations involving serine 465 or arginine 468 yielded dominant-negative forms that inhibited glucose-dependent ERK activation. Glucose stimulated the phosphorylation of tyrosine residues 402 and 881 in PYK2 and binding of PYK2 to Myc-Glut1. Our findings suggest that: (a) glucose activates the GRB2/SOS/RAS/RAF/MEK1/ERK pathway by a mechanism that requires PYK2 and residues 463-468, IASGFR, in the Glut1 C terminus and (b) Glut1 serves as a sensor, transducer, and amplifier for glucose signaling to PYK2 and ERK.


Subject(s)
Glucose/pharmacology , Mitogen-Activated Protein Kinases/metabolism , Monosaccharide Transport Proteins/physiology , Protein-Tyrosine Kinases/physiology , 3T3 Cells , Adipocytes/metabolism , Animals , Deoxyglucose/metabolism , Disaccharides/pharmacology , Focal Adhesion Kinase 2 , Glucose Transporter Type 1 , Glucose Transporter Type 2 , Mice , Muscle, Smooth, Vascular/cytology , Muscle, Smooth, Vascular/metabolism , Rats
17.
J Biol Chem ; 275(44): 34205-12, 2000 Nov 03.
Article in English | MEDLINE | ID: mdl-10952978

ABSTRACT

The binding of several phosphonodifluoromethyl phenylalanine (F(2)Pmp)-containing peptides to protein-tyrosine phosphatase 1B (PTP1B) and its substrate-trapping mutants (C215S and D181A) has been studied using isothermal titration calorimetry. The binding of a high affinity ligand, Ac-Asp-Ala-Asp-Glu-F(2)Pmp-Leu-NH(2), to PTP1B (K(d) = 0.24 microm) is favored by both enthalpic and entropic contributions. Disruption of ionic interactions between the side chain of Arg-47 and the N-terminal acidic residues reduces the binding affinity primarily through the reduction of the TDeltaS term. The role of Arg-47 may be to maximize surface contact between PTP1B and the peptide, which contributes to high affinity binding. The active site Cys-215 --> Ser mutant PTP1B binds ligands with the same affinity as the wild-type enzyme. However, unlike wild-type PTP1B, peptide binding to C215S is predominantly driven by enthalpy change, which likely results from the elimination of the electrostatic repulsion between the thiolate anion and the phosphonate group. The increased enthalpic contribution is offset by reduction in the binding entropy, which may be the result of increased entropy of the unbound protein caused by this mutation. The general acid-deficient mutant D181A binds the peptide 5-fold tighter than the C215S mutant, consistent with the observation that the Asp to Ala mutant is a better "substrate-trapping" reagent than C215S. The increased binding affinity for D181A as compared with the wild-type PTP1B results primarily from an increase in the DeltaH of binding in the mutant, which may be related to decreased electrostatic repulsion between the phosphate moiety and PTP1B. These results have important implications for the design of high affinity PTP1B inhibitors.


Subject(s)
Protein Tyrosine Phosphatases/metabolism , Amino Acid Sequence , Calorimetry/methods , Hydrolysis , Ligands , Peptides/metabolism , Protein Binding , Protein Tyrosine Phosphatase, Non-Receptor Type 1 , Protein Tyrosine Phosphatases/genetics , Substrate Specificity , Temperature , Thermodynamics
18.
Mol Pharmacol ; 58(3): 641-8, 2000 Sep.
Article in English | MEDLINE | ID: mdl-10953059

ABSTRACT

The antiviral activity of L-chicoric acid against HIV-1 has been attributed previously to the inhibition of HIV-1 integration. This conclusion was based on the inhibition of integrase activity in enzymatic assays and the isolation of a resistant HIV strain with a mutation (G140S) in the integrase gene. Here we show that the primary antiviral target of L-CA and its analogs in cell culture is viral entry. L- and D-chicoric acid (L-CA and D-CA) and their respective tetra-acetyl esters inhibit the replication of HIV-1 (III(B) and NL4.3) and HIV-2 (ROD) in MT-4 cells at a 50% effective concentration (EC(50)) ranging from 1.7 to 70.6 microM. In a time-of-addition experiment, L-CA, D-CA, L-CATA, and D-CATA were found to interfere with an early event in the viral replication cycle. Moreover, L-CA, D-CA, and their analogs did not inhibit the replication of virus strains that were resistant toward polyanionic and polycationic compounds at subtoxic concentrations. Furthermore, HIV-1 strains resistant to L-CA and D-CA were selected in the presence of L-CA and D-CA, respectively. Mutations were found in the V2, V3, and V4 loop region of the envelope glycoprotein gp120 of the L-CA and D-CA-resistant NL4.3 strains that were not present in the wild-type NL4.3 strain. Recombination of the gp120 gene of the L-CA and D-CA resistant strain in a NL4.3 wild-type molecular clone fully rescued the phenotypic resistance toward L-CA and D-CA. No significant mutations were detected in the integrase gene of the drug-resistant virus strains. Although inhibition of HIV integrase activity by L-CA and its derivatives was confirmed in an oligonucleotide-driven assay, integrase carrying the G140S mutation was inhibited to the same extent as the wild-type integrase.


Subject(s)
Anti-HIV Agents/pharmacology , Caffeic Acids , HIV Envelope Protein gp120/metabolism , HIV-1/drug effects , Succinates/pharmacology , Anti-HIV Agents/chemistry , Cells, Cultured , Escherichia coli/metabolism , HIV Envelope Protein gp120/drug effects , HIV Envelope Protein gp120/genetics , HIV Integrase/drug effects , HIV Integrase/genetics , HIV Integrase/metabolism , HIV-1/enzymology , HIV-1/genetics , HIV-1/physiology , Humans , Integrase Inhibitors/chemistry , Integrase Inhibitors/pharmacology , Leukocytes, Mononuclear/drug effects , Leukocytes, Mononuclear/physiology , Leukocytes, Mononuclear/virology , Mutagenesis, Site-Directed , Recombinant Proteins/drug effects , Recombinant Proteins/metabolism , Succinates/chemistry , Time Factors , Virus Replication/drug effects
19.
Bioorg Med Chem Lett ; 10(9): 923-7, 2000 May 01.
Article in English | MEDLINE | ID: mdl-10853661

ABSTRACT

Inhibitory potencies were compared of several mono- and dicarboxy-based pTyr mimetics in Grb2 SH2 domain versus PTP1B assays. Although in both systems pTyr residues provide critical binding elements, significant differences in the manner of recognition exist between the two. This is reflected in the current study, where marked variation in relative potencies was observed between the two systems. Of particular note was the poor potency of all monocarboxy-based pTyr mimetics against PTP1B when incorporated into a hexapeptide platform. The recently reported high PTP1B inhibitory potency of similar phenylphosphate mimicking moieties displayed in small molecule, non-peptide structures, raises questions on the limitations of using peptides as platforms for pTyr mimetics in the discovery of small molecule inhibitors.


Subject(s)
Adaptor Proteins, Signal Transducing , Phosphotyrosine/metabolism , Protein Tyrosine Phosphatases/adverse effects , Proteins/metabolism , Catalysis , GRB2 Adaptor Protein , Humans , Molecular Mimicry , Phosphotyrosine/chemistry , Protein Tyrosine Phosphatase, Non-Receptor Type 1 , Structure-Activity Relationship
20.
J Med Chem ; 43(5): 911-20, 2000 Mar 09.
Article in English | MEDLINE | ID: mdl-10715157

ABSTRACT

Nonhydrolyzable phosphotyrosyl (pTyr) mimetics serve as important components of many competitive Grb2 SH2 domain inhibitors. To date, the most potent of these inhibitors have relied on phosphonate-based structures to replace the 4-phosphoryl group of the parent pTyr residue. Reported herein is the design and evaluation of a new pTyr mimetic, p-malonylphenylalanine (Pmf), which does not contain phosphorus yet, in Grb2 SH2 domain binding systems, approaches the potency of phosphonate-based pTyr mimetics. When incorporated into high affinity Grb2 SH2 domain-directed platforms, Pmf is 15-20 times more potent than the closely related previously reported pTyr mimetic, O-malonyltyrosine (OMT). Pmf-containing inhibitors show inhibition constants as low as 8 nM in extracellular Grb2 binding assays and in whole cell systems, effective blockade of both endogenous Grb2 binding to cognate erbB-2, and downstream MAP kinase activation. Evidence is provided that use of an N(alpha)()-oxalyl auxiliary enhances effectiveness of Pmf and other inhibitors in both extracellular and intracellular contexts. As one of the most potent Grb2 SH2 domain-directed pTyr mimetics yet disclosed, Pmf may potentially have utility in the design of new chemotherapeutics for the treatment of various proliferative diseases, including breast cancer.


Subject(s)
Adaptor Proteins, Signal Transducing , Malonates/chemical synthesis , Phenylalanine/analogs & derivatives , Phosphotyrosine/chemistry , Proteins/antagonists & inhibitors , src Homology Domains , Cell Line , Enzyme Activation , Enzyme Inhibitors/chemical synthesis , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/metabolism , Enzyme-Linked Immunosorbent Assay , GRB2 Adaptor Protein , Humans , Ligands , Malonates/chemistry , Malonates/metabolism , Mitogen-Activated Protein Kinases/antagonists & inhibitors , Models, Molecular , Molecular Mimicry , Phenylalanine/chemical synthesis , Phenylalanine/chemistry , Phenylalanine/metabolism , Protein Binding , Receptor, ErbB-2/metabolism , Structure-Activity Relationship
SELECTION OF CITATIONS
SEARCH DETAIL
...