Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 26
Filter
1.
PLoS One ; 18(2): e0281876, 2023.
Article in English | MEDLINE | ID: mdl-36809290

ABSTRACT

Neurofibromatosis Type 2 is an inherited disease characterized by Schwann cell tumors of cranial and peripheral nerves. The NF2 gene encodes Merlin, a member of the ERM family consisting of an N-terminal FERM domain, a central α-helical region, and a C-terminal domain. Changes in the intermolecular FERM-CTD interaction allow Merlin to transition between an open, FERM accessible conformation and a closed, FERM-inaccessible conformation, modulating Merlin activity. Merlin has been shown to dimerize, but the regulation and function Merlin dimerization is not clear. We used a nanobody based binding assay to show that Merlin dimerizes via a FERM-FERM interaction, orientated with each C-terminus close to each other. Patient derived and structural mutants show that dimerization controls interactions with specific binding partners, including HIPPO pathway components, and correlates with tumor suppressor activity. Gel filtration experiments showed that dimerization occurs after a PIP2 mediated transition from closed to open conformation monomers. This process requires the first 18 amino acids of the FERM domain and is inhibited by phosphorylation at serine 518. The discovery that active, open conformation Merlin is a dimer represents a new paradigm for Merlin function with implications for the development of therapies designed to compensate for Merlin loss.


Subject(s)
Genes, Neurofibromatosis 2 , Neurofibromin 2 , Humans , Dimerization , Genes, Tumor Suppressor , Neurofibromin 2/genetics , Protein Structure, Tertiary , Protein Multimerization
2.
Leukemia ; 36(3): 637-647, 2022 03.
Article in English | MEDLINE | ID: mdl-34711926

ABSTRACT

Aberrant RHO guanine nucleotide exchange factor (RhoGEF) activation is chief mechanism driving abnormal activation of their GTPase targets in transformation and tumorigenesis. Consequently, a small-molecule inhibitor of RhoGEF can make an anti-cancer drug. We used cellular, mouse, and humanized models of RAC-dependent BCR-ABL1-driven and Ph-like acute lymphoblastic leukemia to identify VAV3, a tyrosine phosphorylation-dependent RacGEF, as the target of the small molecule IODVA1. We show that through binding to VAV3, IODVA1 inhibits RAC activation and signaling and increases pro-apoptotic activity in BCR-ABL1-transformed cells. Consistent with this mechanism of action, cellular and animal models of BCR-ABL1-induced leukemia in Vav3-null background do not respond to IODVA1. By durably decreasing in vivo RAC signaling, IODVA1 eradicates leukemic propagating activity of TKI-resistant BCR-ABL1(T315I) B-ALL cells after treatment withdrawal. Importantly, IODVA1 suppresses the leukemic burden in the treatment refractory pediatric Ph+ and TKI-resistant Ph+ B-ALL patient-derived xenograft models better than standard-of-care dasatinib or ponatinib and provides a more durable response after treatment withdrawal. Pediatric leukemia samples with diverse genetic lesions show high sensitivity to IODVA1 ex vivo and this sensitivity is VAV3 dependent. IODVA1 thus spearheads a novel class of drugs that inhibits a RacGEF and holds promise as an anti-tumor therapy.


Subject(s)
Antineoplastic Agents/pharmacology , Drug Resistance, Neoplasm/drug effects , Precursor Cell Lymphoblastic Leukemia-Lymphoma/drug therapy , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins c-vav/antagonists & inhibitors , Signal Transduction/drug effects , Animals , Antineoplastic Agents/therapeutic use , Female , Humans , Male , Mice, Inbred C57BL , Mice, SCID , Precursor Cell Lymphoblastic Leukemia-Lymphoma/metabolism , Protein Kinase Inhibitors/therapeutic use , Proto-Oncogene Proteins c-vav/metabolism , Small Molecule Libraries/pharmacology , Small Molecule Libraries/therapeutic use , Tumor Cells, Cultured
3.
Am J Hum Genet ; 107(6): 1149-1156, 2020 12 03.
Article in English | MEDLINE | ID: mdl-33186543

ABSTRACT

The Congenital Dyserythropoietic Anemia (CDA) Registry was established with the goal to facilitate investigations of natural history, biology, and molecular pathogenetic mechanisms of CDA. Three unrelated individuals enrolled in the registry had a syndrome characterized by CDA and severe neurodevelopmental delay. They were found to have missense mutations in VPS4A, a gene coding for an ATPase that regulates the ESCRT-III machinery in a variety of cellular processes including cell division, endosomal vesicle trafficking, and viral budding. Bone marrow studies showed binucleated erythroblasts and erythroblasts with cytoplasmic bridges indicating abnormal cytokinesis and abscission. Circulating red blood cells were found to retain transferrin receptor (CD71) in their membrane, demonstrating that VPS4A is critical for normal reticulocyte maturation. Using proband-derived induced pluripotent stem cells (iPSCs), we have successfully modeled the hematologic aspects of this syndrome in vitro, recapitulating their dyserythropoietic phenotype. Our findings demonstrate that VPS4A mutations cause cytokinesis and trafficking defects leading to a human disease with detrimental effects to erythropoiesis and neurodevelopment.


Subject(s)
ATPases Associated with Diverse Cellular Activities/genetics , Anemia, Dyserythropoietic, Congenital/genetics , Endosomal Sorting Complexes Required for Transport/genetics , Vacuolar Proton-Translocating ATPases/genetics , Adenosine Triphosphatases/metabolism , Anemia, Dyserythropoietic, Congenital/pathology , Bone Marrow/pathology , Bone Marrow Cells/metabolism , Child , Child, Preschool , Cytokinesis , Endosomes/metabolism , Erythroblasts/metabolism , Erythrocytes/cytology , Erythropoiesis , Female , Humans , Induced Pluripotent Stem Cells/cytology , Male , Neurodevelopmental Disorders/metabolism , Phenotype , Protein Transport , Reticulocytes/cytology
4.
PLoS One ; 15(3): e0229801, 2020.
Article in English | MEDLINE | ID: mdl-32163428

ABSTRACT

We report the synthesis and preliminary characterization of IODVA1, a potent small molecule that is active in xenograft mouse models of Ras-driven lung and breast cancers. In an effort to inhibit oncogenic Ras signaling, we combined in silico screening with inhibition of proliferation and colony formation of Ras-driven cells. NSC124205 fulfilled all criteria. HPLC analysis revealed that NSC124205 was a mixture of at least three compounds, from which IODVA1 was determined to be the active component. IODVA1 decreased 2D and 3D cell proliferation, cell spreading and ruffle and lamellipodia formation through downregulation of Rac activity. IODVA1 significantly impaired xenograft tumor growth of Ras-driven cancer cells with no observable toxicity. Immuno-histochemistry analysis of tumor sections suggests that cell death occurs by increased apoptosis. Our data suggest that IODVA1 targets Rac signaling to induce death of Ras-transformed cells. Therefore, IODVA1 holds promise as an anti-tumor therapeutic agent.


Subject(s)
Antineoplastic Agents/pharmacology , Benzimidazoles/pharmacology , Breast Neoplasms/drug therapy , Lung Neoplasms/drug therapy , ras Proteins/antagonists & inhibitors , Animals , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/therapeutic use , Benzimidazoles/chemical synthesis , Benzimidazoles/therapeutic use , Cell Proliferation/drug effects , Female , HEK293 Cells , Humans , MCF-7 Cells , Mice , Mice, Nude , NIH 3T3 Cells , Tumor Stem Cell Assay , Xenograft Model Antitumor Assays
5.
Leukemia ; 33(3): 749-761, 2019 03.
Article in English | MEDLINE | ID: mdl-30254339

ABSTRACT

Mobilization of hematopoietic stem cells (HSCs) from bone marrow (BM) to peripheral blood (PB) by cytokine granulocyte colony-stimulating factor (G-CSF) or the chemical antagonist of CXCR4, AMD3100, is important in the treatment of blood diseases. Due to clinical conditions of each application, there is a need for continued improvement of HSC mobilization regimens. Previous studies have shown that genetic ablation of the Rho GTPase Cdc42 in HSCs results in their mobilization without affecting survival. Here we rationally identified a Cdc42 activity-specific inhibitor (CASIN) that can bind to Cdc42 with submicromolar affinity and competitively interfere with guanine nucleotide exchange activity. CASIN inhibits intracellular Cdc42 activity specifically and transiently to induce murine hematopoietic stem/progenitor cell egress from the BM by suppressing actin polymerization, adhesion, and directional migration of stem/progenitor cells, conferring Cdc42 knockout phenotypes. We further show that, although, CASIN administration to mice mobilizes similar number of phenotypic HSCs as AMD3100, it produces HSCs with better long-term reconstitution potential than that by AMD3100. Our work validates a specific small molecule inhibitor for Cdc42, and demonstrates that signaling molecules downstream of cytokines and chemokines, such as Cdc42, constitute a useful target for long-term stem cell mobilization.


Subject(s)
Hematopoietic Stem Cells/drug effects , Small Molecule Libraries/pharmacology , cdc42 GTP-Binding Protein/antagonists & inhibitors , Animals , Benzylamines , Bone Marrow/drug effects , Bone Marrow/metabolism , Cell Movement/drug effects , Cyclams , Cytokines/metabolism , Granulocyte Colony-Stimulating Factor/metabolism , Hematopoietic Stem Cell Mobilization/methods , Hematopoietic Stem Cell Transplantation/methods , Hematopoietic Stem Cells/metabolism , Heterocyclic Compounds/pharmacology , Mice , Mice, Inbred C57BL , Stem Cells/drug effects , Stem Cells/metabolism
6.
JCI Insight ; 2(3): e89473, 2017 02 09.
Article in English | MEDLINE | ID: mdl-28194436

ABSTRACT

Many patients with histiocytic disorders such as Langerhans cell histiocytosis (LCH) or Erdheim-Chester disease (ECD) have treatment-refractory disease or suffer recurrences. Recent findings of gene mutations in histiocytoses have generated options for targeted therapies. We sought to determine the utility of prospective sequencing of select genes to further characterize mutations and identify targeted therapies for patients with histiocytoses. Biopsies of 72 patients with a variety of histiocytoses underwent comprehensive genomic profiling with targeted DNA and RNA sequencing. Fifteen patients (21%) carried the known BRAF V600E mutation, and 11 patients (15%) carried various mutations in MAP2K1, which we confirm induce constitutive activation of extracellular signal-regulated kinase (ERK) and were sensitive to inhibitors of mitogen-activated protein kinase kinase (MEK, the product of MAP2K1). We also identified recurring ALK rearrangements, and 4 LCH patients with an uncommon in-frame deletion in BRAF (N486_P490del or N486_T491>K), resulting in constitutive activation of ERK with resistance to V600E-specific inhibitors. We subsequently describe clinical cases where patients with aggressive multisystem LCH experience dramatic and sustained responses to monotherapy with either dabrafenib or trametinib. These findings support our conclusion that comprehensive genomic profiling should be regularly applied to these disorders at diagnosis, and can positively impact clinical care.


Subject(s)
Anaplastic Lymphoma Kinase/genetics , Histiocytosis/drug therapy , MAP Kinase Kinase 1/genetics , Protein Kinase Inhibitors/administration & dosage , Proto-Oncogene Proteins B-raf/genetics , Sequence Analysis, DNA/methods , Sequence Analysis, RNA/methods , 3T3 Cells , Adult , Animals , Drug Resistance, Neoplasm , Female , Histiocytosis/genetics , Histiocytosis/pathology , Humans , Imidazoles/administration & dosage , Imidazoles/therapeutic use , Infant , Male , Mice , Mutation , Oximes/administration & dosage , Oximes/therapeutic use , Prospective Studies , Protein Kinase Inhibitors/therapeutic use , Pyridones/administration & dosage , Pyridones/therapeutic use , Pyrimidinones/administration & dosage , Pyrimidinones/therapeutic use , Treatment Outcome , Young Adult
7.
Biochem J ; 453(1): 27-35, 2013 Jul 01.
Article in English | MEDLINE | ID: mdl-23565972

ABSTRACT

Sts (suppressor of T-cell receptor signalling)-1 and Sts-2 are HPs (histidine phosphatases) that negatively regulate TCR (T-cell receptor) signalling pathways, including those involved in cytokine production. HPs play key roles in such varied biological processes as metabolism, development and intracellular signalling. They differ considerably in their primary sequence and substrate specificity, but possess a catalytic core formed by an invariant quartet of active-site residues. Two histidine and two arginine residues cluster together within the HP active site and are thought to participate in a two-step dephosphorylation reaction. To date there has been little insight into any additional residues that might play an important functional role. In the present study, we identify and characterize an additional residue within the Sts phosphatases (Sts-1 Arg383 or Sts-2 Arg369) that is critical for catalytic activity and intracellular function. Mutation of Sts-1 Arg383 to an alanine residue compromises the enzyme's activity and renders Sts-1 unable to suppress TCR-induced cytokine induction. Of the multiple amino acids substituted for Arg383, only lysine partially rescues the catalytic activity of Sts-1. Although Sts-1 Arg383 is conserved in all Sts homologues, it is only conserved in one of the two sub-branches of HPs. The results of the present study highlight an essential role for Sts-1 phosphatase activity in regulating T-cell activation and add a new dimension of complexity to our understanding of HP catalytic activity.


Subject(s)
Arginine/chemistry , Catalytic Domain , Phosphoric Monoester Hydrolases/metabolism , Protein Tyrosine Phosphatases/metabolism , Arginine/metabolism , HEK293 Cells , Humans
8.
Cell Cycle ; 12(1): 51-66, 2013 Jan 01.
Article in English | MEDLINE | ID: mdl-23255114

ABSTRACT

Stem cells are essential for development and tissue maintenance and display molecular markers and functions distinct from those of differentiated cell types in a given tissue. Malignant cells that exhibit stem cell-like activities have been detected in many types of cancers and have been implicated in cancer recurrence and drug resistance. Normal stem cells and cancer stem cells have striking commonalities, including shared cell surface markers and signal transduction pathways responsible for regulating quiescence vs. proliferation, self-renewal, pluripotency and differentiation. As the search continues for markers that distinguish between stem cells, progenitor cells and cancer stem cells, growing evidence suggests that a unique chromatin-associated protein called DEK may confer stem cell-like qualities. Here, we briefly describe current knowledge regarding stem and progenitor cells. We then focus on new findings that implicate DEK as a regulator of stem and progenitor cell qualities, potentially through its unusual functions in the regulation of local or global chromatin organization.


Subject(s)
Chromatin/metabolism , Chromosomal Proteins, Non-Histone/metabolism , Neoplastic Stem Cells/metabolism , Oncogene Proteins/metabolism , Chromatin/chemistry , DNA Repair , Humans , Poly-ADP-Ribose Binding Proteins , Tumor Suppressor Protein p53/metabolism
9.
J Biol Chem ; 286(18): 15943-54, 2011 May 06.
Article in English | MEDLINE | ID: mdl-21393235

ABSTRACT

T cell activity is controlled in large part by the T cell receptor (TCR). The TCR detects the presence of foreign pathogens and activates the T cell-mediated immune reaction. Numerous intracellular signaling pathways downstream of the TCR are involved in the process of T cell activation. Negative regulation of these pathways helps prevent excessive and deleterious T cell responses. Two homologous proteins, Sts-1 and Sts-2, have been shown to function as critical negative regulators of TCR signaling. The phosphoglycerate mutase-like domain of Sts-1 (Sts-1(PGM)) has a potent phosphatase activity that contributes to the suppression of TCR signaling. The function of Sts-2(PGM) as a phosphatase has been less clear, principally because its intrinsic enzyme activity has been difficult to detect. Here, we demonstrate that Sts-2 regulates the level of tyrosine phosphorylation on targets within T cells, among them the critical T cell tyrosine kinase Zap-70. Utilizing new phosphorylated substrates, we demonstrate that Sts-2(PGM) has clear, albeit weak, phosphatase activity. We further pinpoint Sts-2 residues Glu-481, Ser-552, and Ser-582 as specificity determinants, in that an Sts-2(PGM) triple mutant in which these three amino acids are altered to their counterparts in Sts-1(PGM) has substantially increased activity. Our results suggest that the phosphatase activities of both suppressor of TCR signaling homologues cooperate in a similar but independent fashion to help set the threshold for TCR-induced T cell activation.


Subject(s)
Phosphoprotein Phosphatases/metabolism , Receptors, Antigen, T-Cell/metabolism , Signal Transduction/physiology , T-Lymphocytes/enzymology , ZAP-70 Protein-Tyrosine Kinase/metabolism , Animals , Cell Line , Enzyme Activation/physiology , Lymphocyte Activation/physiology , Mice , Mice, Knockout , Phosphoprotein Phosphatases/genetics , Phosphoprotein Phosphatases/immunology , Phosphorylation/physiology , Protein Tyrosine Phosphatases , Receptors, Antigen, T-Cell/genetics , Receptors, Antigen, T-Cell/immunology , Substrate Specificity/physiology , T-Lymphocytes/immunology , ZAP-70 Protein-Tyrosine Kinase/genetics , ZAP-70 Protein-Tyrosine Kinase/immunology
10.
Article in English | MEDLINE | ID: mdl-20516590

ABSTRACT

The suppressor of T-cell signaling (Sts) proteins are multidomain proteins that negatively regulate the signaling of membrane-bound receptors, including the T-cell receptor (TCR) and the epidermal growth-factor receptor (EGFR). They contain at their C-terminus a 2H-phosphatase homology (PGM) domain that is responsible for their protein tyrosine phosphatase activity. Here, the crystal structure of the phosphatase domain of Sts-1, Sts-1(PGM), was determined at pH 4.6. The asymmetric unit contains two independent molecules and each active site is occupied by a sulfate ion. Each sulfate is located at the phosphate-binding site and makes similar interactions with the catalytic residues. The structure suggests an explanation for the lower Michaelis-Menten constants at acidic pH.


Subject(s)
Phosphoric Monoester Hydrolases/chemistry , Receptors, Antigen, T-Cell/chemistry , Sulfates/chemistry , Animals , Crystallography, X-Ray , Hydrogen-Ion Concentration , Mice , Models, Molecular , Phosphoric Monoester Hydrolases/metabolism , Protein Binding , Protein Structure, Tertiary , Protein Tyrosine Phosphatases , Receptors, Antigen, T-Cell/metabolism , Sulfates/metabolism
11.
Biochemistry ; 49(9): 1970-4, 2010 Mar 09.
Article in English | MEDLINE | ID: mdl-20131908

ABSTRACT

The use of the dominant negative mutant of Ras has been crucial in elucidating the cellular signaling of Ras in response to the activation of various membrane-bound receptors. Although several point mutants of Ras exhibit a dominant negative effect, the asparagine to serine mutation at position 17 (S17N) remains the most popular and the most effective at inhibiting the activation of endogenous Ras. It is now widely accepted that the dominant negative effect is due to the ability of the mutant to sequester upstream activators and its inability to activate downstream effectors. Here, we present the crystal structure of RasS17N in the GDP-bound form. In the three molecules that populate the asymmetric unit, the Mg(2+) ion that normally coordinates the beta-phosphate is absent because of steric hindrance from the Asn17 side chain. Instead, a Ca(2+) ion is coordinating the alpha-phosphate. Also absent from one molecule is electron density for Phe28, a conserved residue that normally stabilizes the nucleotide's guanine base. Except for Phe28, the nucleotide makes conserved interactions with Ras. Combined, the inability of Phe28 to stabilize the guanine base and the absence of a Mg(2+) ion to neutralize the negative charges on the phosphates explain the weaker affinity of GDP for Ras. Our data suggest that the absence of the Mg(2+) should also dramatically affect GTP binding to Ras and the proper positioning of Thr35 necessary for the activation of switch 1 and the binding to downstream effectors, a prerequisite for the triggering of signaling pathways.


Subject(s)
Amino Acid Substitution/genetics , Asparagine/genetics , Serine/genetics , ras Proteins/chemistry , ras Proteins/genetics , Asparagine/chemistry , Crystallography, X-Ray , Guanosine Diphosphate/chemistry , Guanosine Diphosphate/genetics , Guanosine Diphosphate/metabolism , Guanosine Triphosphate/chemistry , Guanosine Triphosphate/metabolism , Humans , Magnesium/chemistry , Magnesium/metabolism , Models, Molecular , Protein Binding/genetics , Serine/chemistry , Signal Transduction/genetics , Structure-Activity Relationship , ras Proteins/metabolism
12.
Biochemistry ; 48(48): 11449-57, 2009 Dec 08.
Article in English | MEDLINE | ID: mdl-19883123

ABSTRACT

We previously characterized the G60A mutant of Ras and showed that the switch regions of the GTP-bound but not the GDP-bound form of this mutant adopt an "open conformation" similar to that seen in nucleotide-free Ras. Here, we mutate Lys147 of the conserved (145)SAK(147) motif in the G60A background and characterize the resulting double mutant (DM). We show that RasDM is the first structure of a Ras protein with identical GDP- and GTP-bound structures. Both structures adopt the open conformation of the active form of RasG60A. The increase in the accessible surface area of the nucleotide is consistent with a 4-fold increase in its dissociation rate. Stopped-flow experiments show no major difference in the two-step kinetics of association of GDP or GTP with the wild type, G60A, or RasDM. Addition of Sos fails to accelerate nucleotide exchange. Overexpression of the G60A or double mutant of Ras in COS-1 cells fails to activate Erk and shows a strong dominant negative effect. Our data suggest that flexibility at position 60 is required for proper Sos-catalyzed nucleotide exchange and that structural information is somehow shared among the switch regions and the different nucleotide binding motifs.


Subject(s)
Guanosine Diphosphate/metabolism , Guanosine Triphosphate/metabolism , Mutant Proteins/chemistry , Mutant Proteins/metabolism , Mutation , ras Proteins/chemistry , ras Proteins/metabolism , Animals , COS Cells , Catalysis , Chlorocebus aethiops , Crystallography, X-Ray , Kinetics , Lysine/chemistry , Lysine/genetics , Protein Conformation , Substrate Specificity , Thermodynamics , ras Proteins/genetics
13.
Mol Immunol ; 46(16): 3224-31, 2009 Oct.
Article in English | MEDLINE | ID: mdl-19733910

ABSTRACT

The T cell receptor (TCR) detects the presence of infectious pathogens and activates numerous intracellular signaling pathways. Protein tyrosine phosphorylation and ubiquitination serve as key regulatory mechanisms downstream of the TCR. Negative regulation of TCR signaling pathways is important in controlling the immune response, and the Suppressor of TCR Signaling proteins (Sts-1 and Sts-2) have been shown to function as critical negative regulators of TCR signaling. Although their mechanism of action has yet to be fully uncovered, it is known that the Sts proteins possess intrinsic phosphatase activity. Here, we demonstrate that Sts-1 and Sts-2 are instrumental in down-modulating proteins that are dually modified by both protein tyrosine phosphorylation and ubiquitination. Specifically, both naïve and activated T cells derived from genetically engineered mice that lack the Sts proteins display strikingly elevated levels of tyrosine phosphorylated, ubiquitinated proteins following TCR stimulation. The accumulation of the dually modified proteins is transient, and in activated T cells but not naïve T cells is significantly enhanced by co-receptor engagement. Our observations hint at a novel regulatory mechanism downstream of the T cell receptor.


Subject(s)
Lymphocyte Activation/immunology , Receptors, Antigen, T-Cell/immunology , Signal Transduction/immunology , T-Lymphocytes/immunology , Ubiquitination/immunology , Animals , Lymphocyte Activation/genetics , Mice , Mice, Knockout , Phosphorylation/genetics , Phosphorylation/immunology , Protein Tyrosine Phosphatases , Receptors, Antigen, T-Cell/genetics , Signal Transduction/genetics , Tyrosine/genetics , Tyrosine/immunology , Ubiquitination/genetics
14.
Biochemistry ; 48(34): 8129-35, 2009 Sep 01.
Article in English | MEDLINE | ID: mdl-19627098

ABSTRACT

The C-terminal domain of the suppressor of T cell receptor (TCR) signaling 1 and 2 (Sts-1 and -2) proteins has homology to the 2H-phosphatase family of enzymes. The phosphatase activity of the correspondent Sts-1 domain, Sts-1(PGM), is key for its ability to negatively regulate the signaling of membrane-bound receptors including TCR and the epidermal growth factor receptor (EGFR). A nucleophilic histidine, which is transiently phosphorylated during the phosphatase reaction, is essential for the activity. Here, we present the crystal structure of Sts-2(PGM) in the phosphorylated active form and bound to VO(3), which represent structures of an intermediate and of a transition state analogue along the path of the dephosphorylation reaction. In the former structure, the proposed nucleophilic His366 is the only phoshorylated residue and is stabilized by several interactions with conserved basic residues within the active site. In the latter structure, the vanadium atom sits in the middle of a trigonal bipyramid formed by the three oxygen atoms of the VO(3) molecule, atom NE2 of His366, and an apical water molecule W(a). The V-NE2 bond length (2.25 A) suggests that VO(3) is not covalently attached to His366 and that the reaction mechanism is partially associative. The two structures also suggest a role for Glu476 in activating a uniquely positioned water molecule. In both structures, the conformation of the active site is remarkably similar to the one seen in apo-Sts-2(PGM) suggesting that the spatial arrangement of the catalytic residues does not change during the dephosphorylation reaction.


Subject(s)
Oxides/metabolism , Phosphoric Monoester Hydrolases/metabolism , Receptors, Antigen, T-Cell/chemistry , Receptors, Antigen, T-Cell/metabolism , Vanadium Compounds/metabolism , Animals , Catalytic Domain , Crystallography, X-Ray , Histidine/analogs & derivatives , Histidine/chemistry , Histidine/metabolism , Humans , Mice , Models, Molecular , Phosphoric Monoester Hydrolases/chemistry , Phosphorylation , Protein Structure, Tertiary
15.
Biochemistry ; 48(8): 1681-90, 2009 Mar 03.
Article in English | MEDLINE | ID: mdl-19196006

ABSTRACT

The suppressors of T cell receptor (TCR) signaling 1 and 2 (Sts-1 and -2, respectively) are multidomain proteins that negatively regulate the signaling of membrane-bound receptors, including TCR and the epidermal growth factor receptor (EGFR). Sts-1 was recently shown to be a new type of protein tyrosine phosphatase (PTP), with the phosphatase activity located within its C-terminal phosphoglycerate mutase (PGM) homology domain and key for the regulation of TCR signaling in T cells. The activity of the related Sts-2 enzyme is significantly less than that of Sts-1. Here we investigate the phosphatase activity of the PGM domain of Sts-2, Sts-2(PGM). The crystal structure of Sts-2(PGM) is remarkably similar to Sts-1(PGM), including conservation of all catalytic residues. Insight into mechanistic details is provided by the structures of the apo, tungstate-bound, and phosphate-bound enzyme. The active site shows stringent specificity, with the k(cat) optimum at pH 5.0 suggesting that Sts-2 might function as an acid-dependent phosphatase. Mutation of active site residues Gln372, Ala446, Glu481, Ser552, and Ser582 to their equivalents in Sts-1 increases the phosphatase activity of Sts-2(PGM) toward model substrates. Overall, our data demonstrate that Sts-2(PGM) adopts the conformation of an active phosphatase whose activity is fundamentally different from that of Sts-1 despite the strong structural homology. They also demonstrate that nonconserved active site residues are responsible for the difference in activity between the two isoforms. These differences reflect possible distinct physiological substrates.


Subject(s)
Phosphoric Monoester Hydrolases/metabolism , Receptors, Antigen, T-Cell/chemistry , Receptors, Antigen, T-Cell/metabolism , Animals , Biocatalysis/drug effects , Catalytic Domain , Crystallography, X-Ray , Enzyme Inhibitors/pharmacology , Inhibitory Concentration 50 , Kinetics , Mice , Models, Molecular , Mutant Proteins/chemistry , Mutant Proteins/metabolism , Phosphoric Monoester Hydrolases/antagonists & inhibitors , Protein Structure, Secondary , Protein Structure, Tertiary , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism
16.
Biochemistry ; 47(46): 12135-45, 2008 Nov 18.
Article in English | MEDLINE | ID: mdl-18937503

ABSTRACT

Here, we present the crystal structure of the ecdysone phosphate phosphatase (EPPase) phosphoglycerate mutase (PGM) homology domain, the first structure of a steroid phosphate phosphatase. The structure reveals an alpha/beta-fold common to members of the two histidine (2H)-phosphatase superfamily with strong homology to the Suppressor of T-cell receptor signaling-1 (Sts-1 PGM) protein. The putative EPPase PGM active site contains signature residues shared by 2H-phosphatase enzymes, including a conserved histidine (His80) that acts as a nucleophile during catalysis. The physiological substrate ecdysone 22-phosphate was modeled in a hydrophobic cavity close to the phosphate-binding site. EPPase PGM shows limited substrate specificity with an ability to hydrolyze steroid phosphates, the phospho-tyrosine (pTyr) substrate analogue para-nitrophenylphosphate ( pNPP) and pTyr-containing peptides and proteins. Altogether, our data demonstrate a new protein tyrosine phosphatase (PTP) activity for EPPase. They suggest that EPPase and its closest homologues can be grouped into a distinct subfamily in the large 2H-phosphatase superfamily of proteins.


Subject(s)
Bombyx/enzymology , Insect Proteins/chemistry , Phosphoric Monoester Hydrolases/chemistry , Protein Tyrosine Phosphatases/chemistry , Animals , Carrier Proteins/chemistry , Catalysis , Catalytic Domain/physiology , Humans , Protein Structure, Tertiary , Structural Homology, Protein
17.
Mol Cell ; 27(3): 486-97, 2007 Aug 03.
Article in English | MEDLINE | ID: mdl-17679096

ABSTRACT

Precise signaling by the T cell receptor (TCR) is crucial for a proper immune response. To ensure that T cells respond appropriately to antigenic stimuli, TCR signaling pathways are subject to multiple levels of regulation. Sts-1 negatively regulates signaling pathways downstream of the TCR by an unknown mechanism(s). Here, we demonstrate that Sts-1 is a phosphatase that can target the tyrosine kinase Zap-70 among other proteins. The X-ray structure of the Sts-1 C terminus reveals that it has homology to members of the phosphoglycerate mutase/acid phosphatase (PGM/AcP) family of enzymes, with residues known to be important for PGM/AcP catalytic activity conserved in nature and position in Sts-1. Point mutations that impair Sts-1 phosphatase activity in vitro also impair the ability of Sts-1 to regulate TCR signaling in T cells. These observations reveal a PGM/AcP-like enzyme activity involved in the control of antigen receptor signaling.


Subject(s)
Protein Tyrosine Phosphatases/metabolism , Receptors, Antigen, T-Cell/metabolism , Signal Transduction/physiology , Animals , Blotting, Western , Cell Differentiation/physiology , Cells, Cultured , Down-Regulation/physiology , Humans , Immunoprecipitation , Mice , Mice, Knockout , Point Mutation , Receptors, Antigen, T-Cell/antagonists & inhibitors , Receptors, Antigen, T-Cell/physiology , T-Lymphocytes/physiology , ZAP-70 Protein-Tyrosine Kinase/genetics , ZAP-70 Protein-Tyrosine Kinase/metabolism
18.
Curr Top Med Chem ; 6(11): 1109-16, 2006.
Article in English | MEDLINE | ID: mdl-16842149

ABSTRACT

Rho GTPases of the Ras superfamily are involved in the regulation of multiple cell functions and have been implicated in the pathology of various human diseases including cancer. They are attractive drug targets in future targeted therapy. A wealth of structure-function information made available by high resolution structures and mutagenesis studies has laid out the foundation for the derivation of a mechanism-based targeting strategy. Here we describe the rational design and characterizations of a first generation Rac-specific small molecule inhibitor. Based on the structure-function information of Rac interaction with GEFs, in a computer based Virtual Screening we have identified NSC23766, a highly soluble and membrane permeable compound, as a specific inhibitor of a subset of GEF binding to Rac and therefore Rac activation. In fibroblast cells NSC23766 inhibited Rac1 GTP-loading without affecting Cdc42 or RhoA activity and suppressed the Rac-GEF, Tiam1, and oncogenic Ras induced cell growth and transformation. NSC23766 also potently inhibited the prostate PC-3 cancer cell proliferation and invasion induced by Rac hyperactivation. Intraperitoneal administration of NSC23766 to laboratory mice resulted in effective Rac GTPase suppression and hematopoietic stem cell mobilization from the bone marrow to the peripheral blood, similar to the effects of genetically targeted disruption of Rac GTPases in the animals. A co-crystal structure of NSC23766 bound to Rac1 provided further insight for future medicinal chemistry modification and improvement of this lead Rac-specific inhibitor. Thus, structure-function based rational design may represent a new avenue for generating lead small molecule inhibitors of Ras superfamily GTPases that are useful for modulating pathological conditions in which the small GTPase deregulation may play a role.


Subject(s)
Drug Design , Enzyme Inhibitors/pharmacology , rho GTP-Binding Proteins/antagonists & inhibitors , Animals , Enzyme Inhibitors/chemistry , Humans , Models, Molecular , Molecular Structure , Structure-Activity Relationship
19.
J Mol Biol ; 359(1): 35-46, 2006 May 26.
Article in English | MEDLINE | ID: mdl-16616186

ABSTRACT

The synaptic localization of ion channel receptors is essential for efficient synaptic transmission and the precise regulation of diverse neuronal functions. In the central nervous system, ion channel receptors reside in the postsynaptic membrane where they are juxtaposed to presynaptic terminals. For proper function, these ion channels have to be anchored to the cytoskeleton, and in the case of the inhibitory glycine and gamma-amino-butyric acid type A (GABA(A)) receptors this interaction is mediated by a gephyrin centered scaffold. Highlighting its central role in this receptor anchoring scaffold, gephyrin interacts with a number of proteins, including the neurospecific guanine nucleotide exchange factor collybistin. Collybistin belongs to the Dbl family of guanine nucleotide exchange factors, occurs in multiple splice variants, and is specific for Cdc42, a small GTPase belonging to the Rho family. The 2.3 Angstroms resolution crystal structure of the Cdc42-collybistin II complex reveals a novel conformation of the switch I region of Cdc42. It also provides the first direct observation of structural changes in the relative orientation of the Dbl-homology domain and the pleckstrin-homology domain in the same Dbl family protein. Biochemical data indicate that gephyrin negatively regulates collybistin activity.


Subject(s)
Carrier Proteins/metabolism , Guanine Nucleotide Exchange Factors/chemistry , Membrane Proteins/metabolism , Protein Structure, Quaternary , cdc42 GTP-Binding Protein/chemistry , Animals , Carrier Proteins/chemistry , Crystallography, X-Ray , Guanine Nucleotide Exchange Factors/metabolism , Membrane Proteins/chemistry , Mice , Models, Molecular , Rats , Two-Hybrid System Techniques , cdc42 GTP-Binding Protein/metabolism
20.
Structure ; 14(3): 427-36, 2006 Mar.
Article in English | MEDLINE | ID: mdl-16531227

ABSTRACT

The flexibility of the conserved 57DTAGQ61 motif is essential for Ras proper cycling in response to growth factors. Here, we increase the flexibility of the 57DTAGQ61 motif by mutating Gln61 to Gly. The crystal structure of the RasQ61G mutant reveals a new conformation of switch 2 that bears remarkable structural homology to an intermediate for GTP hydrolysis revealed by targeted molecular dynamics simulations. The mutation increased retention of GTP and inhibited Ras binding to the catalytic site, but not to the distal site of Sos. Most importantly, the thermodynamics of RafRBD binding to Ras are altered even though the structure of switch 1 is not affected by the mutation. Our results suggest that interplay and transmission of structural information between the switch regions are important factors for Ras function. They propose that initiation of GTP hydrolysis sets off the separation of the Ras/effector complex even before the GDP conformation is reached.


Subject(s)
Glucosephosphate Dehydrogenase/metabolism , Guanosine Triphosphate/chemistry , Oncogene Protein p21(ras)/genetics , Crystallography, X-Ray , Glucosephosphate Dehydrogenase/chemistry , Guanosine Triphosphate/metabolism , Hydrolysis , Magnesium/metabolism , Models, Molecular , Mutagenesis, Site-Directed , Mutant Proteins/chemistry , Nucleotides/chemistry , Oncogene Protein p21(ras)/chemistry , Oncogene Protein p21(ras)/metabolism , Protein Binding , Protein Conformation , SOS Response, Genetics/genetics , Signal Transduction , raf Kinases/chemistry , raf Kinases/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...