Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 26
Filter
1.
J Med Chem ; 59(19): 9094-9106, 2016 Oct 13.
Article in English | MEDLINE | ID: mdl-27676368

ABSTRACT

The low molecular weight protein tyrosine phosphatase (LMW-PTP) is a regulator of a number of signaling pathways and has been implicated as a potential target for oncology and diabetes/obesity. There is significant therapeutic interest in developing potent and selective inhibitors to control LMW-PTP activity. We report the discovery of a novel class of LMW-PTP inhibitors derived from sulfophenyl acetic amide (SPAA), some of which exhibit greater than 50-fold preference for LMW-PTP over a large panel of PTPs. X-ray crystallography reveals that binding of SPAA-based inhibitors induces a striking conformational change in the LMW-PTP active site, leading to the formation of a previously undisclosed hydrophobic pocket to accommodate the α-phenyl ring in the ligand. This induced-fit mechanism is likely a major contributor responsible for the exquisite inhibitor selectivity.

2.
Cancer Res ; 76(16): 4805-15, 2016 08 15.
Article in English | MEDLINE | ID: mdl-27325652

ABSTRACT

Phosphatase of regenerating liver (PRL) oncoproteins are phosphatases overexpressed in numerous types of human cancer. Elevated levels of PRL associate with metastasis and poor clinical outcomes. In principle, PRL phosphatases offer appealing therapeutic targets, but they remain underexplored due to the lack of specific chemical probes. In this study, we address this issue by exploiting a unique property of PRL phosphatases, namely, that they may function as homotrimers. Starting from a sequential structure-based virtual screening and medicinal chemistry strategy, we identified Cmpd-43 and several analogs that disrupt PRL1 trimerization. Biochemical and structural analyses demonstrate that Cmpd-43 and its close analogs directly bind the PRL1 trimer interface and obstruct PRL1 trimerization. Cmpd-43 also specifically blocks the PRL1-induced cell proliferation and migration through attenuation of both ERK1/2 and Akt activity. Importantly, Cmpd-43 exerted potent anticancer activity both in vitro and in vivo in a murine xenograft model of melanoma. Our results validate a trimerization-dependent signaling mechanism for PRL and offer proof of concept for trimerization inhibitors as candidate therapeutics to treat PRL-driven cancers. Cancer Res; 76(16); 4805-15. ©2016 AACR.


Subject(s)
Antineoplastic Agents/pharmacology , Drug Screening Assays, Antitumor , Melanoma, Experimental/drug therapy , Protein Tyrosine Phosphatases/antagonists & inhibitors , Animals , Cell Line, Tumor , Enzyme Inhibitors/pharmacology , Humans , Immunohistochemistry , Mice , Mice, Inbred NOD , Mice, SCID , Mice, Transgenic , Xenograft Model Antitumor Assays
3.
Biochemistry ; 53(25): 4136-51, 2014 Jul 01.
Article in English | MEDLINE | ID: mdl-24935154

ABSTRACT

The Src homology 2 (SH2) domain-containing protein tyrosine phosphatase 2 (SHP2) is a critical signal transducer downstream of growth factors that promotes the activation of the RAS-ERK1/2 cascade. In its basal state, SHP2 exists in an autoinhibited closed conformation because of an intramolecular interaction between its N-SH2 and protein tyrosine phosphatase (PTP) domains. Binding to pTyr ligands present on growth factor receptors and adaptor proteins with its N-SH2 domain localizes SHP2 to its substrates and frees the active site from allosteric inhibition. Germline mutations in SHP2 are known to cause both Noonan syndrome (NS) and LEOPARD syndrome (LS), two clinically similar autosomal dominant developmental disorders. NS-associated SHP2 mutants display elevated phosphatase activity, while LS-associated SHP2 mutants exhibit reduced catalytic activity. A conundrum in how clinically similar diseases result from mutations to SHP2 that have opposite effects on this enzyme's catalytic functionality exists. Here we report a comprehensive investigation of the kinetic, structural, dynamic, and biochemical signaling properties of the wild type as well as all reported LS-associated SHP2 mutants. The results reveal that LS-causing mutations not only affect SHP2 phosphatase activity but also induce a weakening of the intramolecular interaction between the N-SH2 and PTP domains, leading to mutants that are more readily activated by competing pTyr ligands. Our data also indicate that the residual phosphatase activity associated with the LS SHP2 mutant is required for enhanced ERK1/2 activation. Consequently, catalytically impaired SHP2 mutants could display gain-of-function properties because of their ability to localize to the vicinity of substrates for longer periods of time, thereby affording the opportunity for prolonged substrate turnover and sustained RAS-ERK1/2 activation.


Subject(s)
LEOPARD Syndrome/enzymology , Protein Tyrosine Phosphatase, Non-Receptor Type 11/chemistry , Enzyme Activation , HEK293 Cells , Humans , Kinetics , LEOPARD Syndrome/genetics , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Models, Molecular , Mutation , Protein Conformation , Protein Tyrosine Phosphatase, Non-Receptor Type 11/genetics , Protein Tyrosine Phosphatase, Non-Receptor Type 11/metabolism , src Homology Domains
4.
J Med Chem ; 56(12): 4990-5008, 2013 Jun 27.
Article in English | MEDLINE | ID: mdl-23713581

ABSTRACT

Lymphoid-specific tyrosine phosphatase (LYP), a member of the protein tyrosine phosphatase (PTP) family of signaling enzymes, is associated with a broad spectrum of autoimmune diseases. Herein we describe our structure-based lead optimization efforts within a 6-hydroxy-benzofuran-5-carboxylic acid series culminating in the identification of compound 8b, a potent and selective inhibitor of LYP with a K(i) value of 110 nM and more than 9-fold selectivity over a large panel of PTPs. The structure of LYP in complex with 8b was obtained by X-ray crystallography, providing detailed information about the molecular recognition of small-molecule ligands binding LYP. Importantly, compound 8b possesses highly efficacious cellular activity in both T- and mast cells and is capable of blocking anaphylaxis in mice. Discovery of 8b establishes a starting point for the development of clinically useful LYP inhibitors for treating a wide range of autoimmune disorders.


Subject(s)
Autoimmune Diseases/drug therapy , Molecular Targeted Therapy , Protein Kinase Inhibitors/pharmacology , Protein-Tyrosine Kinases/antagonists & inhibitors , Small Molecule Libraries/pharmacology , Anaphylaxis/drug therapy , Animals , Carboxylic Acids/chemistry , Down-Regulation/drug effects , Female , Humans , Jurkat Cells , Lymphocyte Activation/drug effects , Mast Cells/drug effects , Mast Cells/immunology , Mice , Models, Molecular , Protein Conformation , Protein Kinase Inhibitors/chemistry , Protein Kinase Inhibitors/therapeutic use , Small Molecule Libraries/chemistry , Small Molecule Libraries/therapeutic use
5.
J Biol Chem ; 288(15): 10472-82, 2013 Apr 12.
Article in English | MEDLINE | ID: mdl-23457302

ABSTRACT

SHP2 is an allosteric phosphatase essential for growth factor-mediated Ras activation. Germ-line mutations in SHP2 cause clinically similar LEOPARD and Noonan syndromes, two of several autosomal-dominant conditions characterized by gain-of-function mutations in the Ras pathway. Interestingly, Noonan syndrome SHP2 mutants are constitutively active, whereas LEOPARD syndrome SHP2 mutants exhibit reduced phosphatase activity. How do catalytically impaired LEOPARD syndrome mutants engender gain-of-function phenotypes? Our study reveals that LEOPARD syndrome mutations weaken the intramolecular interaction between the N-SH2 and phosphatase domains, leading to a change in SHP2 molecular switching mechanism. Consequently, LEOPARD syndrome SHP2 mutants bind upstream activators preferentially and are hypersensitive to growth factor stimulation. They also stay longer with scaffolding adapters, thus prolonging substrate turnover, which compensates for the reduced phosphatase activity. The study provides a solid framework for understanding how individual SHP2 mutations cause diseases.


Subject(s)
LEOPARD Syndrome/enzymology , Mutation , Protein Tyrosine Phosphatase, Non-Receptor Type 11/chemistry , Crystallography, X-Ray , Humans , LEOPARD Syndrome/genetics , Protein Structure, Tertiary , Protein Tyrosine Phosphatase, Non-Receptor Type 11/genetics , Protein Tyrosine Phosphatase, Non-Receptor Type 11/metabolism , Structure-Activity Relationship
6.
J Am Chem Soc ; 134(43): 18116-24, 2012 Oct 31.
Article in English | MEDLINE | ID: mdl-23075115

ABSTRACT

Protein tyrosine phosphatases (PTPs) constitute a large family of signaling enzymes that control the cellular levels of protein tyrosine phosphorylation. A detailed understanding of PTP functions in normal physiology and in pathogenic conditions has been hampered by the absence of PTP-specific, cell-permeable small-molecule agents. We present a stepwise focused library approach that transforms a weak and general non-hydrolyzable pTyr mimetic (F(2)Pmp, phosphonodifluoromethyl phenylalanine) into a highly potent and selective inhibitor of PTP-MEG2, an antagonist of hepatic insulin signaling. The crystal structures of the PTP-MEG2-inhibitor complexes provide direct evidence that potent and selective PTP inhibitors can be obtained by introducing molecular diversity into the F(2)Pmp scaffold to engage both the active site and unique nearby peripheral binding pockets. Importantly, the PTP-MEG2 inhibitor possesses highly efficacious cellular activity and is capable of augmenting insulin signaling and improving insulin sensitivity and glucose homeostasis in diet-induced obese mice. The results indicate that F(2)Pmp can be converted into highly potent and selective PTP inhibitory agents with excellent in vivo efficacy. Given the general nature of the approach, this strategy should be applicable to other members of the PTP superfamily.


Subject(s)
Diabetes Mellitus, Type 2/drug therapy , Phenylalanine/analogs & derivatives , Protein Tyrosine Phosphatases, Non-Receptor/antagonists & inhibitors , Animals , Disease Models, Animal , Mice , Mice, Inbred C57BL , Mice, Obese , Models, Molecular , Molecular Structure , Phenylalanine/chemical synthesis , Phenylalanine/chemistry , Phenylalanine/pharmacology , Protein Tyrosine Phosphatases, Non-Receptor/chemistry , Protein Tyrosine Phosphatases, Non-Receptor/metabolism
7.
Bioorg Med Chem ; 20(6): 1940-6, 2012 Mar 15.
Article in English | MEDLINE | ID: mdl-22133902

ABSTRACT

Protein tyrosine phosphatases (PTPs) constitute a large and structurally diverse family of signaling enzymes that control the cellular levels of protein tyrosine phosphorylation. Malfunction of PTP activity has significant implications in many human diseases, and the PTP protein family provides an exciting array of validated diabetes/obesity (PTP1B), oncology (SHP2), autoimmunity (Lyp), and infectious disease (mPTPB) targets. However, despite the fact that PTPs have been garnering attention as novel therapeutic targets, they remain largely an untapped resource. The main challenges facing drug developers by the PTPs are inhibitor specificity and bioavailability. Work over the last ten years has demonstrated that it is feasible to develop potent and selective inhibitors for individual members of the PTP family by tethering together small ligands that can simultaneously occupy both the active site and unique nearby peripheral binding sites. Recent results with the bicyclic salicylic acid pharmacophores indicate that the new chemistry platform may provide a potential solution to overcome the bioavailability issue that has plagued the PTP drug discovery field for many years. Structural analysis of PTP-inhibitor complexes reveals molecular determinants important for the development of more potent and selective PTP inhibitors, thus offering hope in the medicinal chemistry of a largely unexploited protein class with a wealth of attractive drug targets.


Subject(s)
Benzofurans/chemistry , Benzofurans/pharmacology , Protein Kinase Inhibitors/chemistry , Protein Kinase Inhibitors/pharmacology , Protein Tyrosine Phosphatases/antagonists & inhibitors , Salicylates/chemistry , Salicylates/pharmacology , Animals , Bridged Bicyclo Compounds/chemistry , Bridged Bicyclo Compounds/pharmacology , Catalytic Domain , Combinatorial Chemistry Techniques , Drug Design , Humans , Indoles/chemistry , Indoles/pharmacology , Models, Molecular , Protein Tyrosine Phosphatases/chemistry , Protein Tyrosine Phosphatases/metabolism
8.
J Biol Chem ; 286(49): 42316-42324, 2011 Dec 09.
Article in English | MEDLINE | ID: mdl-22009749

ABSTRACT

Phosphatases of the regenerating liver (PRL) play oncogenic roles in cancer development and metastasis. Although previous studies indicate that PRL-1 promotes cell growth and migration by activating both the ERK1/2 and RhoA pathways, the mechanism by which it activates these signaling events remains unclear. We have identified a PRL-1-binding peptide (Peptide 1) that shares high sequence identity with a conserved motif in the Src homology 3 (SH3) domain of p115 Rho GTPase-activating protein (GAP). p115 RhoGAP directly binds PRL-1 in vitro and in cells via its SH3 domain. Structural analyses of the PRL-1·Peptide 1 complex revealed a novel protein-protein interaction whereby a sequence motif within the PxxP ligand-binding site of the p115 RhoGAP SH3 domain occupies a folded groove within PRL-1. This prevents the canonical interaction between the SH3 domain of p115 RhoGAP and MEKK1 and results in activation of ERK1/2. Furthermore, PRL-1 binding activates RhoA signaling by inhibiting the catalytic activity of p115 RhoGAP. The results demonstrate that PRL-1 binding to p115 RhoGAP provides a coordinated mechanism underlying ERK1/2 and RhoA activation.


Subject(s)
Cell Cycle Proteins/metabolism , Gene Expression Regulation, Enzymologic , Guanine Nucleotide Exchange Factors/chemistry , Immediate-Early Proteins/metabolism , Membrane Proteins/metabolism , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Protein Tyrosine Phosphatases/metabolism , src-Family Kinases/metabolism , Amino Acid Motifs , Animals , Fibroblasts/metabolism , HEK293 Cells , Humans , Ligands , Mice , Protein Binding , Rho Guanine Nucleotide Exchange Factors , Signal Transduction , rho GTP-Binding Proteins/metabolism
9.
J Biol Chem ; 286(35): 30526-30534, 2011 Sep 02.
Article in English | MEDLINE | ID: mdl-21719704

ABSTRACT

A missense single-nucleotide polymorphism in the gene encoding the lymphoid-specific tyrosine phosphatase (Lyp) has been identified as a causal factor in a wide spectrum of autoimmune diseases. Interestingly, the autoimmune-predisposing variant of Lyp appears to represent a gain-of-function mutation, implicating Lyp as an attractive target for the development of effective strategies for the treatment of many autoimmune disorders. Unfortunately, the precise biological functions of Lyp in signaling cascades and cellular physiology are poorly understood. Identification and characterization of Lyp substrates will help define the chain of molecular events coupling Lyp dysfunction to diseases. In the current study, we identified consensus sequence motifs for Lyp substrate recognition using an "inverse alanine scanning" combinatorial library approach. The intrinsic sequence specificity data led to the discovery and characterization of SKAP-HOM, a cytosolic adaptor protein required for proper activation of the immune system, as a bona fide Lyp substrate. To determine the molecular basis for Lyp substrate recognition, we solved crystal structures of Lyp in complex with the consensus peptide as well as the phosphopeptide derived from SKAP-HOM. Together with the biochemical data, the structures define the molecular determinants for Lyp substrate specificity and provide a solid foundation upon which novel therapeutics targeting Lyp can be developed for multiple autoimmune diseases.


Subject(s)
Intracellular Signaling Peptides and Proteins/physiology , Protein Tyrosine Phosphatase, Non-Receptor Type 22/physiology , src-Family Kinases/metabolism , Amino Acid Motifs , Autoimmunity , Cytosol/metabolism , Glutathione Transferase/metabolism , Humans , Intracellular Signaling Peptides and Proteins/chemistry , Kinetics , Models, Molecular , Molecular Conformation , Mutation, Missense , Peptides/chemistry , Phosphorylation , Protein Binding , Protein Tyrosine Phosphatase, Non-Receptor Type 22/chemistry , Substrate Specificity
10.
Bioorg Med Chem Lett ; 21(14): 4238-42, 2011 Jul 15.
Article in English | MEDLINE | ID: mdl-21669525

ABSTRACT

SHP2, encoded by PTPN11, is a non-receptor protein tyrosine phosphatase (PTP) containing two tandem Src homology-2 (SH2) domains. It is expressed ubiquitously and plays critical roles in growth factor mediated processes, primarily by promoting the activation of the RAS/ERK signaling pathway. Genetic and biochemical studies have identified SHP2 as the first bona fide oncoprotein in the PTP superfamily, and a promising target for anti-cancer and anti-leukemia therapy. Here, we report a structure-based approach to identify SHP2 inhibitors with a novel scaffold. Through sequential virtual screenings and in vitro inhibition assays, a reversible competitive SHP2 inhibitor (C21) was identified. C21 is structurally distinct from all known SHP2 inhibitors. Combining molecular dynamics simulation and binding free energy calculation, a most likely binding mode of C21 with SHP2 is proposed, and further validated by site-directed mutagenesis and structure-activity relationship studies. This binding mode is consistent with the observed potency and specificity of C21, and reveals the molecular determinants for further optimization based on the new scaffold.


Subject(s)
Benzenesulfonates/chemistry , Enzyme Inhibitors/chemistry , Protein Tyrosine Phosphatase, Non-Receptor Type 11/antagonists & inhibitors , Pyrazoles/chemistry , Small Molecule Libraries/chemistry , Amino Acid Sequence , Benzenesulfonates/pharmacology , Binding Sites , Drug Evaluation, Preclinical , Enzyme Inhibitors/pharmacology , Kinetics , Molecular Dynamics Simulation , Molecular Sequence Data , Mutagenesis, Site-Directed , Protein Tyrosine Phosphatase, Non-Receptor Type 11/genetics , Protein Tyrosine Phosphatase, Non-Receptor Type 11/metabolism , Pyrazoles/pharmacology , Sequence Alignment , Small Molecule Libraries/pharmacology , Structure-Activity Relationship , Thermodynamics , src Homology Domains
11.
Chem Biol ; 18(1): 101-10, 2011 Jan 28.
Article in English | MEDLINE | ID: mdl-21276943

ABSTRACT

SHP2 phosphatase is a positive transducer of growth factor and cytokine signaling. SHP2 is also a bona fide oncogene; gain-of-function SHP2 mutations leading to increased phosphatase activity cause Noonan syndrome, as well as multiple forms of leukemia and solid tumors. We report that tautomycetin (TTN), an immunosuppressor in organ transplantation, and its engineered analog TTN D-1 are potent SHP2 inhibitors. TTN and TTN D-1 block T cell receptor-mediated tyrosine phosphorylation and ERK activation and gain-of-function mutant SHP2-induced hematopoietic progenitor hyperproliferation and monocytic differentiation. Crystal structure of the SHP2⋅TTN D-1 complex reveals that TTN D-1 occupies the SHP2 active site in a manner similar to that of a peptide substrate. Collectively, the data support the notion that SHP2 is a cellular target for TTN and provide a potential mechanism for the immunosuppressive activity of TTN. Moreover, the structure furnishes molecular insights upon which therapeutics targeting SHP2 can be developed on the basis of the TTN scaffold.


Subject(s)
Enzyme Inhibitors/metabolism , Enzyme Inhibitors/pharmacology , Furans/metabolism , Furans/pharmacology , Immunosuppressive Agents/metabolism , Immunosuppressive Agents/pharmacology , Lipids/pharmacology , Protein Tyrosine Phosphatase, Non-Receptor Type 11/metabolism , Amino Acid Sequence , Cell Differentiation/drug effects , Cell Proliferation/drug effects , Enzyme Activation/drug effects , Enzyme Inhibitors/chemistry , Furans/chemistry , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/drug effects , Humans , Immunosuppressive Agents/chemistry , Jurkat Cells , Lipids/chemistry , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Models, Molecular , Molecular Sequence Data , Monocytes/cytology , Monocytes/drug effects , Phosphorylation/drug effects , Protein Conformation , Protein Tyrosine Phosphatase, Non-Receptor Type 11/antagonists & inhibitors , Protein Tyrosine Phosphatase, Non-Receptor Type 11/chemistry , Pyrans/chemistry , Pyrans/metabolism , Pyrans/pharmacology , Signal Transduction/drug effects , Spiro Compounds/chemistry , Spiro Compounds/metabolism , Spiro Compounds/pharmacology , Substrate Specificity , Tyrosine/metabolism
12.
J Med Chem ; 53(6): 2482-93, 2010 Mar 25.
Article in English | MEDLINE | ID: mdl-20170098

ABSTRACT

The Src homology-2 domain containing protein tyrosine phosphatase-2 (SHP2) plays a pivotal role in growth factor and cytokine signaling. Gain-of-function SHP2 mutations are associated with Noonan syndrome, various kinds of leukemias, and solid tumors. Thus, there is considerable interest in SHP2 as a potential target for anticancer and antileukemia therapy. We report a salicylic acid based combinatorial library approach aimed at binding both active site and unique nearby subpockets for enhanced affinity and selectivity. Screening of the library led to the identification of a SHP2 inhibitor II-B08 (compound 9) with highly efficacious cellular activity. Compound 9 blocks growth factor stimulated ERK1/2 activation and hematopoietic progenitor proliferation, providing supporting evidence that chemical inhibition of SHP2 may be therapeutically useful for anticancer and antileukemia treatment. X-ray crystallographic analysis of the structure of SHP2 in complex with 9 reveals molecular determinants that can be exploited for the acquisition of more potent and selective SHP2 inhibitors.


Subject(s)
Enzyme Inhibitors/chemistry , Enzyme Inhibitors/pharmacology , Indoles/chemistry , Indoles/pharmacology , Protein Tyrosine Phosphatase, Non-Receptor Type 11/antagonists & inhibitors , Salicylic Acid/chemistry , Triazoles/chemistry , Triazoles/pharmacology , Animals , Apoptosis/drug effects , Area Under Curve , Cell Line , Cell Line, Tumor , Cells, Cultured , Crystallography, X-Ray , Enzyme Inhibitors/pharmacokinetics , Female , Humans , Indoles/pharmacokinetics , Inhibitory Concentration 50 , Mice , Mice, Inbred C57BL , Mice, Nude , Models, Chemical , Models, Molecular , Molecular Structure , Mutation , Neoplasms/metabolism , Neoplasms/pathology , Neoplasms/prevention & control , Protein Binding , Protein Structure, Tertiary , Protein Tyrosine Phosphatase, Non-Receptor Type 11/chemistry , Protein Tyrosine Phosphatase, Non-Receptor Type 11/genetics , Small Molecule Libraries , Triazoles/pharmacokinetics , Xenograft Model Antitumor Assays
13.
J Am Chem Soc ; 130(50): 17075-84, 2008 Dec 17.
Article in English | MEDLINE | ID: mdl-19012396

ABSTRACT

There has been considerable interest in protein tyrosine phosphatase 1B (PTP1B) as a therapeutic target for diabetes, obesity, as well as cancer. Identifying inhibitory compounds with good bioavailability is a major challenge of drug discovery programs targeted toward PTPs. Most current PTP active site-directed pharmacophores are negatively charged pTyr mimetics which cannot readily enter the cell. This lack of cell permeability limits the utility of such compounds in signaling studies and further therapeutic development. We identify aryl diketoacids as novel pTyr surrogates and show that neutral amide-linked aryl diketoacid dimers also exhibit excellent PTP inhibitory activity. Kinetic studies establish that these aryl diketoacid derivatives act as noncompetitive inhibitors of PTP1B. Crystal structures of ligand-bound PTP1B reveal that both the aryl diketoacid and its dimeric derivative bind PTP1B at the active site, albeit with distinct modes of interaction, in the catalytically inactive, WPD loop open conformation. Furthermore, dimeric aryl diketoacids are cell permeable and enhance insulin signaling in hepatoma cells, suggesting that targeting the inactive conformation may provide a unique opportunity for creating active site-directed PTP1B inhibitors with improved pharmacological properties.


Subject(s)
Enzyme Inhibitors/chemical synthesis , Enzyme Inhibitors/pharmacology , Keto Acids/chemical synthesis , Keto Acids/pharmacology , Protein Tyrosine Phosphatase, Non-Receptor Type 1/antagonists & inhibitors , Amides/chemistry , Cell Line, Tumor , Crystallography, X-Ray , Dimerization , Drug Evaluation, Preclinical , Enzyme Activation/drug effects , Enzyme Inhibitors/chemistry , Humans , Keto Acids/chemistry , Models, Molecular , Molecular Structure , Protein Binding , Protein Tyrosine Phosphatase, Non-Receptor Type 1/chemistry , Protein Tyrosine Phosphatase, Non-Receptor Type 1/genetics , Protein Tyrosine Phosphatase, Non-Receptor Type 1/metabolism , Structure-Activity Relationship
14.
J Am Chem Soc ; 130(40): 13196-7, 2008 Oct 08.
Article in English | MEDLINE | ID: mdl-18781746

ABSTRACT

A homotyrosine based seleninic acid irreversibly inhibits protein tyrosine phosphatases by forming a covalent selenosulfide linkage with the active site cysteine sulfhydryl specifically. The details of the event are revealed by model synthetic studies and by kinetic, mass spectrometric, and crystallographic characterization.


Subject(s)
Phosphates/chemistry , Protein Tyrosine Phosphatases/antagonists & inhibitors , Selenium Compounds/chemistry , Selenium Compounds/pharmacology , Apraxia, Ideomotor , Crystallography, X-Ray , Models, Molecular , Molecular Structure , Protein Tyrosine Phosphatases/metabolism
15.
J Am Chem Soc ; 130(26): 8251-60, 2008 Jul 02.
Article in English | MEDLINE | ID: mdl-18528979

ABSTRACT

Protein tyrosine phosphatases (PTPs) play key roles in the regulation of normal and pathological processes ranging from cell proliferation, differentiation, metabolism, and survival to many human diseases including cancer and diabetes. Functional studies of PTP can be greatly facilitated by small molecule probes that covalently label the active site of a PTP through an activity-dependent chemical reaction. In this article, we characterize phenyl vinyl sulfonate (PVSN) and phenyl vinyl sulfone (PVS) as a new class of mechanism-based PTP probes. PVSN and PVS inactivate a broad range of PTPs in a time- and concentration-dependent fashion. The PVSN- and PVS-mediated PTP inactivation is active site-directed and irreversible, resulting from a Michael addition of the active-site Cys Sgamma onto the terminal carbon of the vinyl group. Structural and mechanistic analyses reveal the molecular basis for the preference of PVSN/PVS toward the PTPs, which lies in the ability of PVSN and PVS to engage the conserved structural and catalytic machinery of the PTP active site. In contrast to early alpha-bromobenzyl phosphonate-based probes, PVSN and PVS are resistant to solvolysis and are cell-permeable and thus hold promise for in vivo applications. Collectively, these properties bode well for the development of aryl vinyl sulfonate/sulfone-based PTP probes to interrogate PTP activity in complex proteomes.


Subject(s)
Arylsulfonates , Molecular Probe Techniques , Protein Tyrosine Phosphatases/analysis , Sulfones
16.
Proc Natl Acad Sci U S A ; 104(50): 19767-72, 2007 Dec 11.
Article in English | MEDLINE | ID: mdl-18056643

ABSTRACT

The lymphoid-specific tyrosine phosphatase (Lyp) has generated enormous interest because a single-nucleotide polymorphism in the gene (PTPN22) encoding Lyp produces a gain-of-function mutant phosphatase that is associated with several autoimmune diseases, including type I diabetes, rheumatoid arthritis, Graves disease, and systemic lupus erythematosus. Thus, Lyp represents a potential target for a broad spectrum of autoimmune disorders. Unfortunately, no Lyp inhibitor has been reported. In addition, little is known about the structure and biochemical mechanism that directly regulates Lyp function. Here, we report the identification of a bidentate salicylic acid-based Lyp inhibitor I-C11 with excellent cellular efficacy. Structural and mutational analyses indicate that the inhibitor binds both the active site and a nearby peripheral site unique to Lyp, thereby furnishing a solid foundation upon which inhibitors with therapeutic potency and selectivity can be developed. Moreover, a comparison of the apo- and inhibitor-bound Lyp structures reveals that the Lyp-specific region S(35)TKYKADK(42), which harbors a PKC phosphorylation site, could adopt either a loop or helical conformation. We show that Lyp is phosphorylated exclusively at Ser-35 by PKC both in vitro and in vivo. We provide evidence that the status of Ser-35 phosphorylation may dictate the conformational state of the insert region and thus Lyp substrate recognition. We demonstrate that Ser-35 phosphorylation impairs Lyp's ability to inactivate the Src family kinases and down-regulate T cell receptor signaling. Our data establish a mechanism by which PKC could attenuate the cellular function of Lyp, thereby augmenting T cell activation.


Subject(s)
Autoimmune Diseases/enzymology , Benzofurans/chemistry , Benzofurans/pharmacology , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/pharmacology , Protein Tyrosine Phosphatase, Non-Receptor Type 22/chemistry , Protein Tyrosine Phosphatase, Non-Receptor Type 22/metabolism , Triazoles/chemistry , Triazoles/pharmacology , Amino Acid Sequence , Autoimmune Diseases/genetics , Autoimmune Diseases/immunology , Crystallography, X-Ray , Humans , Inhibitory Concentration 50 , Jurkat Cells , Kinetics , Lymphocyte Activation , Models, Molecular , Molecular Structure , Mutation/genetics , Phosphorylation , Phosphoserine/metabolism , Protein Kinase C/metabolism , Protein Structure, Tertiary , Protein Tyrosine Phosphatase, Non-Receptor Type 22/antagonists & inhibitors , Protein Tyrosine Phosphatase, Non-Receptor Type 22/genetics , Receptors, Antigen, T-Cell/metabolism , Sequence Alignment , Signal Transduction , Substrate Specificity , T-Lymphocytes/enzymology , T-Lymphocytes/immunology
17.
Biochemistry ; 46(14): 4370-8, 2007 Apr 10.
Article in English | MEDLINE | ID: mdl-17352459

ABSTRACT

The Yersinia protein tyrosine phosphatase (YopH) contains a loop of ten amino acids (the WPD loop) that covers the entrance of the active site of the enzyme during substrate binding. In this work the substrate mimicking competitive inhibitor p-nitrocatechol sulfate (PNC) is used as a probe of the active site. The dynamics of the WPD loop was determined by subjecting an equilibrated system containing YopH, PNC, and YopH bound to PNC to a laser induced temperature jump, and subsequently following the change in equilibrium due to the perturbation. Using this methodology the dynamics associated with substrate binding in YopH have been determined. These results indicate that substrate binding is coupled to the WPD loop motion, and WPD loop dynamics occur in the sub-millisecond time scale. The significance of these dynamic results is interpreted in terms of the catalytic cycle of the enzyme.


Subject(s)
Protein Tyrosine Phosphatases/chemistry , Protein Tyrosine Phosphatases/metabolism , Yersinia/enzymology , Amino Acid Sequence , Aspartic Acid/chemistry , Binding Sites , Binding, Competitive , Catalysis , Catalytic Domain , Catechols/chemistry , Catechols/metabolism , Crystallization , Cysteine/chemistry , Hot Temperature , Kinetics , Lasers , Ligands , Models, Molecular , Molecular Structure , Protein Binding , Protein Conformation , Protein Structure, Secondary , Protein Structure, Tertiary , Protein Tyrosine Phosphatases/isolation & purification , Spectrometry, Fluorescence , Substrate Specificity , Thermodynamics , Tryptophan/chemistry
18.
J Biol Chem ; 281(50): 38834-44, 2006 Dec 15.
Article in English | MEDLINE | ID: mdl-17046812

ABSTRACT

ERK2, a prototypic member of the MAPK family, plays a central role in regulating cell growth and differentiation. MKP3, an ERK2-specific phosphatase, terminates ERK2 signaling. To understand the molecular basis of ERK2 recognition by MKP3, we carried out hydrogen/deuterium exchange mass spectrometry experiments to map the interaction surfaces between the two proteins. The results show that the exquisite specificity of MKP3 for ERK2 is governed by two distinctive protein-protein interactions. To increase the "effective concentration" of the interacting molecules, the kinase interaction motif in MKP3 ((64)RRLQKGNLPVR(74)) and an MKP3-specific segment ((101)NSSDWNE(107)) bind the common docking site in ERK2 defined by residues in L(16), L(5), beta(7)-beta(8), and alpha(d)-L(8)-alpha(e), located opposite the kinase active site. In addition to this "tethering" effect, additional interactions between the (364)FTAP(367) sequence in MKP3 and the ERK2 substrate-binding site, formed by residues in the activation lip and the P+1 site (beta(9)-alpha(f) loop), L(13) (alpha(f)-alpha(g) loop), and the MAPK insert (L(14)-alpha(1L14)-alpha(2L14)), are essential for allosteric activation of MKP3 and formation of a productive complex whereby the MKP3 catalytic site is correctly juxtaposed to carry out the dephosphorylation of phospho-Thr(183)/phospho-Tyr(185) in ERK2. This bipartite protein-protein interaction model may be applicable to the recognition of other MAPKs by their cognate regulators and substrates.


Subject(s)
Mass Spectrometry/methods , Mitogen-Activated Protein Kinase 1/metabolism , Protein Tyrosine Phosphatases/metabolism , Amino Acid Sequence , Binding Sites , Deuterium/chemistry , Dual-Specificity Phosphatases , Hydrogen/chemistry , Mitogen-Activated Protein Kinase 1/genetics , Mitogen-Activated Protein Kinase Phosphatases , Models, Molecular , Molecular Sequence Data , Mutagenesis, Site-Directed , Protein Conformation , Protein Tyrosine Phosphatases/chemistry , Protein Tyrosine Phosphatases/genetics
19.
Proc Natl Acad Sci U S A ; 103(14): 5326-31, 2006 Apr 04.
Article in English | MEDLINE | ID: mdl-16567630

ABSTRACT

Mitogen-activated protein (MAP) kinases are central components of signal transduction pathways for cell proliferation, stress responses, and differentiation. Signaling efficiency and specificity are modulated in large part by docking interactions between individual MAP kinase and the kinase interaction motif (KIM), (R/K)(2-3)-X(1-6)-Phi(A)-X-Phi(B), in its cognate kinases, phosphatases, scaffolding proteins, and substrates. We have determined the crystal structure of extracellular signal-regulated protein kinase 2 bound to the KIM peptide from MAP kinase phosphatase 3, an extracellular signal-regulated protein kinase 2-specific phosphatase. The structure reveals that the KIM docking site, situated in a noncatalytic region opposite of the kinase catalytic pocket, is comprised of a highly acidic patch and a hydrophobic groove, which engage the basic and Phi(A)-X-Phi(B) residues, respectively, in the KIM sequence. The specific docking interactions observed in the structure consolidate all known biochemical data. In addition, structural comparison indicates that the KIM docking site is conserved in all MAP kinases. The results establish a structural model for understanding how MAP kinases interact with their regulators and substrates and provide new insights into how MAP kinase docking specificity can be achieved.


Subject(s)
Mitogen-Activated Protein Kinase 1/metabolism , Protein Tyrosine Phosphatases/metabolism , Amino Acid Motifs , Amino Acid Sequence , Binding Sites , Dual Specificity Phosphatase 6 , Mitogen-Activated Protein Kinase 1/chemistry , Models, Molecular , Molecular Sequence Data , Protein Binding , Sequence Homology, Amino Acid
20.
Biochemistry ; 44(50): 16377-84, 2005 Dec 20.
Article in English | MEDLINE | ID: mdl-16342930

ABSTRACT

Expression of the PSR132 protein from Dianthus caryophyllus (carnation, clover pink) is induced in response to ethylene production associated with petal senescence, and thus the protein is named petal death protein (PDP). Recent work has established that despite the annotation of PDP in sequence databases as carboxyphosphoenolpyruvate mutase, the enzyme is actually a C-C bond cleaving lyase exhibiting a broad substrate profile. The crystal structure of PDP has been determined at 2.7 A resolution, revealing a dimer-of-dimers oligomeric association. Consistent with sequence homology, the overall alpha/beta barrel fold of PDP is the same as that of other isocitrate lyase/PEP mutase superfamily members, including a swapped eighth helix within a dimer. Moreover, Mg(2+) binds in the active site of PDP with a coordination pattern similar to that seen in other superfamily members. A compound, covalently bound to the catalytic residue, Cys144, was interpreted as a thiohemiacetal adduct resulting from the reaction of glutaraldehyde used to cross-link the crystals. The Cys144-carrying flexible loop that gates access to the active site is in the closed conformation. Models of bound substrates and comparison with the closed conformation of isocitrate lyase and 2-methylisocitrate lyase revealed the structural basis for the broad substrate profile of PDP.


Subject(s)
Dianthus/chemistry , Plant Proteins/chemistry , Amino Acid Sequence , Binding Sites , Catalysis , Crystallography, X-Ray , Dianthus/enzymology , Isocitrate Lyase/chemistry , Isocitrate Lyase/metabolism , Models, Molecular , Molecular Sequence Data , Plant Proteins/metabolism , Protein Conformation , Sequence Homology, Amino Acid , Substrate Specificity
SELECTION OF CITATIONS
SEARCH DETAIL
...