Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
Add more filters










Publication year range
1.
Biochemistry ; 59(20): 1896-1908, 2020 05 26.
Article in English | MEDLINE | ID: mdl-32348128

ABSTRACT

Dynamics and conformational motions are important to the activity of enzymes, including protein tyrosine phosphatases. These motions often extend to regions outside the active site, called allosteric regions. In the tyrosine phosphatase Vaccinia H1-related (VHR) enzyme, we demonstrate the importance of the allosteric interaction between the variable insert region and the active-site loops in VHR. These studies include solution nuclear magnetic resonance, computation, steady-state, and rapid kinetic measurements. Overall, the data indicate concerted millisecond motions exist between the variable insert and the catalytic acid loop in wild-type (WT) VHR. The 150 ns computation studies show a flexible acid loop in WT VHR that opens during the simulation from its initial closed structure. Mutation of the variable insert residue, asparagine 74, to alanine results in a rigidification of the acid loop as observed by molecular dynamics simulations and a disruption of crucial active-site hydrogen bonds. Moreover, enzyme kinetic analysis shows a weakening of substrate affinity in the N74A mutant and a >2-fold decrease in substrate cleavage and hydrolysis rates. These data show that despite being nearly 20 Å from the active site, the variable insert region is linked to the acid loop by coupled millisecond motions, and that disruption of the communication between the variable insert and active site alters the normal catalytic function of VHR and perturbs the active-site environment.


Subject(s)
Dual Specificity Phosphatase 3/metabolism , Allosteric Regulation , Biocatalysis , Dual Specificity Phosphatase 3/chemistry , Dual Specificity Phosphatase 3/isolation & purification , Humans , Hydrolysis , Kinetics , Models, Molecular , Mutagenesis, Site-Directed , Protein Conformation
2.
J Mol Biol ; 429(15): 2360-2372, 2017 07 21.
Article in English | MEDLINE | ID: mdl-28625849

ABSTRACT

Drug-like molecules targeting allosteric sites in proteins are of great therapeutic interest; however, identification of potential sites is not trivial. A straightforward approach to identify hidden allosteric sites is demonstrated in protein tyrosine phosphatases (PTP) by creation of single alanine mutations in the catalytic acid loop of PTP1B and VHR. This approach relies on the reciprocal interactions between an allosteric site and its coupled orthosteric site. The resulting NMR chemical shift perturbations (CSPs) of each mutant reveal clusters of distal residues affected by acid loop mutation. In PTP1B and VHR, two new allosteric clusters were identified in each enzyme. Mutations in these allosteric clusters altered phosphatase activity with changes in kcat/KM ranging from 30% to nearly 100-fold. This work outlines a simple method for identification of new allosteric sites in PTP, and given the basis of this method in thermodynamics, it is expected to be generally useful in other systems.


Subject(s)
Allosteric Site , Dual Specificity Phosphatase 3/chemistry , Protein Tyrosine Phosphatase, Non-Receptor Type 1/chemistry , Amino Acid Substitution , DNA Mutational Analysis , Dual Specificity Phosphatase 3/genetics , Dual Specificity Phosphatase 3/metabolism , Kinetics , Magnetic Resonance Spectroscopy , Models, Molecular , Mutagenesis, Site-Directed , Mutant Proteins/chemistry , Mutant Proteins/genetics , Protein Conformation , Protein Tyrosine Phosphatase, Non-Receptor Type 1/genetics , Protein Tyrosine Phosphatase, Non-Receptor Type 1/metabolism
3.
Integr Biol (Camb) ; 7(1): 73-89, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25375676

ABSTRACT

The atypical dual-specificity phosphatases (aDUSPs) are a group of protein tyrosine phosphatases (PTPs) that have been increasingly studied recently, but little is known about their substrates or their roles and regulation. aDUSPs are typically low-molecular-weight enzymes that are distinct from the mitogen-activated protein kinase phosphatases (MKPs) but that still function in the regulation of the MAPK signalling cascade. aDUSPs may also have non-MAPK substrates, based on homologies observed in the sequences flanking potential phosphotyrosine target sites of other proteins and the cell type-specific characteristics of certain aDUSPs. Here, we combined experimental and computational tools to identify new substrates and protein partners of VHR (DUSP3) phosphatase in HeLa cells exposed to genotoxic stress. Experimental approaches confirmed the good stability of VHR and its nuclear co-localisation with classical MAPK substrates. The bioinformatics analysis of 4539 human nuclear proteins to identify a subset with functions related to DNA damage response and repair or to checkpoints and cell cycle control, that contain the phosphorylatable Thr-X-Tyr motif of MAPK with a high probability of dual phosphorylation, and that have structural homology to the MAPK activation loop resulted in a list of 57 putative VHR substrates. Fluorescence confocal microscopy and pull-down experiments followed by immunoblots revealed that VHR co-localised and interacted with components of the MRN complex and pH2AX, a DNA double-strand break sensor. Our platform, which combines experimental data from structure-function and bioinformatics analyses based on MAPK substrate similarities, provides a low-cost and rapid approach for the identification of novel aDUSP-interacting proteins with unknown roles in genotoxic stress response and genome stability maintenance.


Subject(s)
Cell Nucleus/metabolism , DNA Damage/physiology , DNA Repair/physiology , Dual Specificity Phosphatase 3/metabolism , Nuclear Proteins/metabolism , Binding Sites , Computational Biology/methods , Dual Specificity Phosphatase 3/chemistry , HeLa Cells , Humans , MAP Kinase Signaling System/physiology , Nuclear Proteins/chemistry , Protein Binding , Protein Interaction Mapping/methods , Sequence Analysis, Protein/methods
4.
ACS Chem Biol ; 9(7): 1451-9, 2014 Jul 18.
Article in English | MEDLINE | ID: mdl-24798147

ABSTRACT

Vaccinia H1-related (VHR) phosphatase is a dual specificity phosphatase that is required for cell-cycle progression and plays a role in cell growth of certain cancers. Therefore, it represents a potential drug target. VHR is structurally and biochemically well characterized, yet its regulatory principles are still poorly understood. Understanding its regulation is important, not only to comprehend VHR's biological mechanisms and roles but also to determine its potential and druggability as a target in cancer. Here, we investigated the functional role of the unique "variable insert" region in VHR by selectively introducing the photo-cross-linkable amino acid para-benzoylphenylalanine (pBPA) using the amber suppression method. This approach led to the discovery of VHR dimerization, which was further confirmed using traditional chemical cross-linkers. Phe68 in VHR was discovered as a residue involved in the dimerization. We demonstrate that VHR can dimerize inside cells, and that VHR catalytic activity is reduced upon dimerization. Our results suggest that dimerization could occlude the active site of VHR, thereby blocking its accessibility to substrates. These findings indicate that the previously unknown transient self-association of VHR acts as a means for the negative regulation of its catalytic activity.


Subject(s)
Benzophenones/metabolism , Dual Specificity Phosphatase 3/metabolism , Phenylalanine/analogs & derivatives , Protein Multimerization , Animals , Benzophenones/chemistry , COS Cells , Catalytic Domain , Chlorocebus aethiops , Cross-Linking Reagents/chemistry , Dual Specificity Phosphatase 3/chemistry , Dual Specificity Phosphatase 3/genetics , Enzyme Activation , Humans , Models, Molecular , Mutagenesis , Mutation , Phenylalanine/chemistry , Phenylalanine/genetics , Phenylalanine/metabolism , Photochemical Processes
6.
J Biol Chem ; 288(9): 6498-510, 2013 Mar 01.
Article in English | MEDLINE | ID: mdl-23322772

ABSTRACT

Vaccinia VH1-related (VHR) is a dual specificity phosphatase that consists of only a single catalytic domain. Although several protein substrates have been identified for VHR, the elements that control the in vivo substrate specificity of this enzyme remain unclear. In this work, the in vitro substrate specificity of VHR was systematically profiled by screening combinatorial peptide libraries. VHR exhibits more stringent substrate specificity than classical protein-tyrosine phosphatases and recognizes two distinct classes of Tyr(P) peptides. The class I substrates are similar to the Tyr(P) motifs derived from the VHR protein substrates, having sequences of (D/E/ϕ)(D/S/N/T/E)(P/I/M/S/A/V)pY(G/A/S/Q) or (D/E/ϕ)(T/S)(D/E)pY(G/A/S/Q) (where ϕ is a hydrophobic amino acid and pY is phosphotyrosine). The class II substrates have the consensus sequence of (V/A)P(I/L/M/V/F)X1-6pY (where X is any amino acid) with V/A preferably at the N terminus of the peptide. Site-directed mutagenesis and molecular modeling studies suggest that the class II peptides bind to VHR in an opposite orientation relative to the canonical binding mode of the class I substrates. In this alternative binding mode, the Tyr(P) side chain binds to the active site pocket, but the N terminus of the peptide interacts with the carboxylate side chain of Asp(164), which normally interacts with the Tyr(P) + 3 residue of a class I substrate. Proteins containing the class II motifs are efficient VHR substrates in vitro, suggesting that VHR may act on a novel class of yet unidentified Tyr(P) proteins in vivo.


Subject(s)
Dual Specificity Phosphatase 3/chemistry , Amino Acid Motifs , Dual Specificity Phosphatase 3/genetics , Dual Specificity Phosphatase 3/metabolism , Humans , Mutagenesis, Site-Directed , Peptide Library , Protein Binding , Substrate Specificity/physiology
7.
J Biol Chem ; 286(16): 14373-82, 2011 Apr 22.
Article in English | MEDLINE | ID: mdl-21362620

ABSTRACT

The gene product of Vaccinia virus gene H1, VH1, is the first identified dual specificity phosphatase (DSP). The human genome encodes 38 different VH1-like DSPs, which include major regulators of signaling pathways, highly dysregulated in disease states. VH1 down-regulates cellular antiviral response by dephosphorylating activated STAT1 in the IFN-γ/STAT1 signaling pathway. In this report, we have investigated the molecular basis for VH1 catalytic activity. Using small-angle x-ray scattering (SAXS), we determined that VH1 exists in solution as a boomerang-shaped dimer. Targeted alanine mutations in the dimerization domain (aa 1-27) decrease phosphatase activity while leaving the dimer intact. Deletion of the N-terminal dimer swapped helix (aa 1-20) completely abolishes dimerization and severely reduces phosphatase activity. An engineered chimera of VH1 that contains only one active site retains wild-type levels of catalytic activity. Thus, a dimeric quaternary structure, as opposed to two cooperative active sites within the same dimer is essential for VH1 catalytic activity. Together with laforin, VH1 is the second DSP reported in literature for which dimerization via an N-terminal dimerization domain is necessary for optimal catalytic activity. We propose that dimerization may represent a common mechanism to regulate the activity and substrate recognition of DSPs, often assumed to function as monomers.


Subject(s)
Dual Specificity Phosphatase 3/chemistry , STAT1 Transcription Factor/chemistry , Tyrosine/chemistry , Vaccinia virus/metabolism , Catalytic Domain , Dimerization , Gene Deletion , Humans , Kinetics , Phosphorylation , Protein Structure, Quaternary , Protein Structure, Tertiary , Protein Tyrosine Phosphatases, Non-Receptor/chemistry , Recombinant Fusion Proteins/chemistry , Signal Transduction
8.
J Med Chem ; 52(21): 6716-23, 2009 Nov 12.
Article in English | MEDLINE | ID: mdl-19888758

ABSTRACT

Loss of VHR phosphatase causes cell cycle arrest in HeLa carcinoma cells, suggesting that VHR inhibition may be a useful approach to halt the growth of cancer cells. We recently reported that VHR is upregulated in several cervix cancer cell lines as well as in carcinomas of the uterine cervix. Here we report the development of multidentate small-molecule inhibitors of VHR that inhibit its enzymatic activity at nanomolar concentrations and exhibit antiproliferative effects on cervix cancer cells. Chemical library screening was used to identify hit compounds, which were further prioritized in profiling and kinetic experiments. SAR analysis was applied in the search for analogs with improved potency and selectivity, resulting in the discovery of novel inhibitors that are able to interact with both the phosphate-binding pocket and several distinct hydrophobic regions within VHR's active site. This multidentate binding mode was confirmed by X-ray crystallography. The inhibitors decreased the proliferation of cervix cancer cells, while growth of primary normal keratinocytes was not affected. These compounds may be a starting point to develop drugs for the treatment of cervical cancer.


Subject(s)
Antineoplastic Agents/chemical synthesis , Dual Specificity Phosphatase 3/antagonists & inhibitors , Thiazolidines/chemical synthesis , Anthracenes/chemical synthesis , Anthracenes/chemistry , Anthracenes/pharmacology , Anthraquinones/chemical synthesis , Anthraquinones/chemistry , Anthraquinones/pharmacology , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Catalytic Domain , Cell Line, Tumor , Crystallography, X-Ray , Databases, Factual , Drug Screening Assays, Antitumor , Dual Specificity Phosphatase 3/chemistry , Female , Humans , Keratinocytes/drug effects , Kinetics , Models, Molecular , Protein Binding , Pyrazoles/chemical synthesis , Pyrazoles/chemistry , Pyrazoles/pharmacology , Stereoisomerism , Structure-Activity Relationship , Sulfonic Acids , Thiazolidines/chemistry , Thiazolidines/isolation & purification , Uterine Cervical Neoplasms
9.
J Biol Chem ; 284(15): 10129-37, 2009 Apr 10.
Article in English | MEDLINE | ID: mdl-19211553

ABSTRACT

The Vaccinia virus H1 gene product, VH1, is a dual specificity phosphatase that down-regulates the cellular antiviral response by dephosphorylating STAT1. The crystal structure of VH1, determined at 1.32 A resolution, reveals a novel dimeric quaternary structure, which exposes two active sites spaced approximately 39 A away from each other. VH1 forms a stable dimer via an extensive domain swap of the N-terminal helix (residues 1-20). In vitro, VH1 can dephosphorylate activated STAT1, in a reaction that is competed by the nuclear transport adapter importin alpha5. Interestingly, VH1 is inactive with respect to STAT1 bound to DNA, suggesting that the viral phosphatase acts predominantly on the cytoplasmic pool of activated STAT1. We propose that the dimeric quaternary structure of VH1 is essential for specific recognition of activated STAT1, which prevents its nuclear translocation, thus blocking interferon-gamma signal transduction and antiviral response.


Subject(s)
Dual Specificity Phosphatase 3/chemistry , Dual Specificity Phosphatase 3/physiology , STAT1 Transcription Factor/metabolism , Vaccinia virus/enzymology , Active Transport, Cell Nucleus , Catalytic Domain , Circular Dichroism , DNA/chemistry , Dimerization , Humans , Interferon-gamma/metabolism , Models, Molecular , Protein Conformation , Protein Structure, Quaternary , Protein Structure, Tertiary , Signal Transduction
11.
Chembiochem ; 8(17): 2092-9, 2007 Nov 23.
Article in English | MEDLINE | ID: mdl-17933004

ABSTRACT

Human vaccinia H1-related phosphatase (VHR) is a dual-specific phosphatase (DSPs) that plays an important role in the mitogen-activated protein (MAP) kinase cascade regulation. It is also a potential drug target for diseases that are related to immune response. By combining a virtual and NMR-based ligand-screening strategy, we successfully identified four VHR inhibitors, of which GATPT ((glucosamine-aminoethoxy)triphenyltin) can bind to VHR with a K(i) value of 2.54 muM. The putative binding mode of GATPT was constructed by a molecular docking simulation to provide structural insights into the ligand-binding mechanism. Furthermore, we found that this compound can significantly inhibit the dephosphorylation of the extracellular regulated kinases (ERKs), and c-Jun N-terminal kinases (JNKs) and block the G(1)-S phase transition in the cell cycle. Therefore, GATPT is a promising lead structure for designing more effective inhibitors of VHR.


Subject(s)
Dual Specificity Phosphatase 3/antagonists & inhibitors , Protease Inhibitors/chemistry , Protease Inhibitors/pharmacology , Cell Cycle/drug effects , Computer Simulation , Dual Specificity Phosphatase 3/chemistry , Dual Specificity Phosphatase 3/genetics , Dual Specificity Phosphatase 3/metabolism , Enzyme Activation , HeLa Cells , Humans , Ligands , Magnetic Resonance Spectroscopy , Mitogen-Activated Protein Kinases/antagonists & inhibitors , Mitogen-Activated Protein Kinases/metabolism , Models, Molecular , Molecular Structure , Phosphorylation , Protein Kinase Inhibitors/chemistry , Protein Kinase Inhibitors/pharmacology , RNA, Small Interfering/genetics , Structure-Activity Relationship
SELECTION OF CITATIONS
SEARCH DETAIL
...