Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 25
Filter
1.
Acta Crystallogr F Struct Biol Commun ; 78(Pt 7): 265-269, 2022 Jul 01.
Article in English | MEDLINE | ID: mdl-35787553

ABSTRACT

Receptor-type protein tyrosine phosphatases (RPTPs) receive extracellular stimuli and transfer them into cells. They regulate cell growth, differentiation and death via specific signals. They have also been implicated in cancer, diabetes and neurological diseases. RPTPH, a member of the type 3 RPTP (R3-PTP) family, is an important regulator of colorectal cancer and hepatic carcinoma. Despite its importance in drug development, the structure of RPTPH has not yet been resolved. Here, the crystal structure of the catalytic domain of RPTPH was determined at 1.56 Šresolution. Despite similarities to other R3-PTPs in its overall structure, RPTPH exhibited differences in its loop regions and side-chain conformations. Compared with other R3-PTPs, RPTPH has unique side chains near its active site that may confer specificity for inhibitor binding. Therefore, detailed information on the structure of RPTPH provides clues for the development of specific inhibitors.


Subject(s)
Protein Tyrosine Phosphatases , Catalytic Domain , Crystallography, X-Ray , Humans , Protein Tyrosine Phosphatases/chemistry , Receptor-Like Protein Tyrosine Phosphatases, Class 3/chemistry
2.
Protein Pept Lett ; 29(7): 567-573, 2022.
Article in English | MEDLINE | ID: mdl-35546749

ABSTRACT

Protein tyrosine phosphatase receptor-type Q (PTPRQ), a member of the type III tyrosine phosphatase receptor (R3 PTPR) family, is composed of three domains, including 18 extracellular fibronectin type III (FN3) repeats, a transmembrane helix, and a cytoplasmic phosphotyrosine phosphatase (PTP) domain. PTPRQ was initially identified as a transcript upregulated in glomerular mesangial cells in a rat model of glomerulonephritis. Subsequently, studies found that PTPRQ has phosphotyrosine phosphatase and phosphatidylinositol phosphatase activities and can regulate cell proliferation, apoptosis, differentiation, and survival. Further in vivo studies showed that PTPRQ is necessary for the maturation of cochlear hair bundles and is considered a potential gene for deafness. In the recent two decades, 21 mutations in PTPRQ have been linked to autosomal recessive hearing loss (DFNB84) and autosomal dominant hearing loss (DFNA73). Recent mutations, deletions, and amplifications of PTPRQ have been observed in many types of cancers, which indicate that PTPRQ might play an essential role in the development of many cancers. In this review, we briefly describe PTPRQ structure and enzyme activity and focus on the correlation between PTPRQ and human disease. A profound understanding of PTPRQ could be helpful in the identification of new therapeutic targets to treat associated diseases.


Subject(s)
Cochlea/metabolism , Hearing Loss , Receptor-Like Protein Tyrosine Phosphatases, Class 3/metabolism , Animals , Cochlea/growth & development , Fibronectins , Hearing Loss/genetics , Humans , Phosphatidylinositols , Protein Tyrosine Phosphatases/genetics , Protein Tyrosine Phosphatases/metabolism , Rats , Receptor-Like Protein Tyrosine Phosphatases, Class 3/chemistry
3.
Biomed Res Int ; 2021: 5584788, 2021.
Article in English | MEDLINE | ID: mdl-33997018

ABSTRACT

The inner ear is an essential part of a well-developed and well-coordinated hearing system. However, hearing loss can make communication and interaction more difficult. Inherited hearing loss (HL) can occur from pathogenic genetic variants that negatively alter the intricate inner ear sensory mechanism. Recessively inherited forms of HL are highly heterogeneous and account for a majority of prelingual deafness. The current study is designed to investigate genetic causes of HL in three consanguineous Pakistani families. After IRB approval, the clinical history and pure tone audiometric data was obtained for the clinical diagnosis of HL segregating in these three Pakistani families. We performed whole exome sequencing (WES) followed by Sanger sequencing in order to identify and validate the HL-associated pathogenic variants, respectively. The 3-D molecular modeling and the Ramachandran analysis of the identified missense variants were compiled to evaluate the impact of the variants on the encoded proteins. Clinical evaluation revealed prelingual severe to profound sensorineural HL segregating among the affected individuals in all three families. Genetic analysis revealed segregation of several novel variants associated with HL, including a canonical splice-site variant (c.55-2A>G) of PTPRQ in family GCFHL-01, a missense variant [c.1079G>A; p.(Arg360Gln)] of SERPINB6 in family LUHL-01, and an insertion variant (c.10208-10211insCCACCAGGCCCGTGCCTC) within MYO15A in family LUHL-011. All the identified variants had very low frequencies in the control databases. The molecular modeling of p.Arg360Gln missense variant also predicted impaired folding of SERPINB6 protein. This study reports the identification of novel disease-causing variants in three known deafness genes and further highlights the genetic heterogeneity of HL in Pakistani population.


Subject(s)
Genetic Predisposition to Disease , Hearing Loss/genetics , Mutation/genetics , Myosins/genetics , Receptor-Like Protein Tyrosine Phosphatases, Class 3/genetics , Serpins/genetics , Alleles , DNA Mutational Analysis , Family , Female , Humans , Male , Models, Molecular , Myosins/chemistry , Pakistan , Pedigree , Phenotype , Receptor-Like Protein Tyrosine Phosphatases, Class 3/chemistry , Serpins/chemistry
4.
J Transl Med ; 17(1): 351, 2019 10 26.
Article in English | MEDLINE | ID: mdl-31655630

ABSTRACT

BACKGROUND: Biallelic PTPRQ pathogenic variants have been previously reported as causative for autosomal recessive non-syndromic hearing loss. In 2018 the first heterozygous PTPRQ variant has been implicated in the development of autosomal dominant non-syndromic hearing loss (ADNSHL) in a German family. The study presented the only, so far known, PTPRQ pathogenic variant (c.6881G>A) in ADNSHL. It is located in the last PTPRQ coding exon and introduces a premature stop codon (p.Trp2294*). METHODS: A five-generation Polish family with ADNSHL was recruited for the study (n = 14). Thorough audiological, neurotological and imaging studies were carried out to precisely define the phenotype. Genomic DNA was isolated from peripheral blood samples or buccal swabs of available family members. Clinical exome sequencing was conducted for the proband. Family segregation analysis of the identified variants was performed using Sanger sequencing. Single nucleotide polymorphism array on DNA samples from the Polish and the original German family was used for genome-wide linkage analysis. RESULTS: Combining clinical exome sequencing and family segregation analysis, we have identified the same (NM_001145026.2:c.6881G>A, NP_001138498.1:p.Trp2294*) PTPRQ alteration in the Polish ADNSHL family. Using genome-wide linkage analysis, we found that the studied family and the original German family derive from a common ancestor. Deep phenotyping of the affected individuals showed that in contrast to the recessive form, the PTPRQ-related ADNSHL is not associated with vestibular dysfunction. In both families ADNSHL was progressive, affected mainly high frequencies and had a variable age of onset. CONCLUSION: Our data provide the first confirmation of PTPRQ involvement in ADNSHL. The finding strongly reinforces the inclusion of PTPRQ to the small set of genes leading to both autosomal recessive and dominant hearing loss.


Subject(s)
Hearing Loss, Sensorineural/genetics , Receptor-Like Protein Tyrosine Phosphatases, Class 3/genetics , Adolescent , Adult , Age of Onset , Child , Female , Genes, Dominant , Hearing Loss, Sensorineural/physiopathology , Heterozygote , Humans , Male , Middle Aged , Mutant Proteins/chemistry , Mutant Proteins/genetics , Mutant Proteins/physiology , Mutation , Pedigree , Peptide Chain Termination, Translational/genetics , Phenotype , Poland , Polymorphism, Single Nucleotide , Receptor-Like Protein Tyrosine Phosphatases, Class 3/chemistry , Receptor-Like Protein Tyrosine Phosphatases, Class 3/physiology , Translational Research, Biomedical , Young Adult
5.
Mol Cell Proteomics ; 18(10): 2058-2077, 2019 10.
Article in English | MEDLINE | ID: mdl-31427368

ABSTRACT

Vascular endothelial protein tyrosine phosphatase (VE-PTP, PTPRB) is a receptor type phosphatase that is crucial for the regulation of endothelial junctions and blood vessel development. We and others have shown recently that VE-PTP regulates vascular integrity by dephosphorylating substrates that are key players in endothelial junction stability, such as the angiopoietin receptor TIE2, the endothelial adherens junction protein VE-cadherin and the vascular endothelial growth factor receptor VEGFR2. Here, we have systematically searched for novel substrates of VE-PTP in endothelial cells by utilizing two approaches. First, we studied changes in the endothelial phosphoproteome on exposing cells to a highly VE-PTP-specific phosphatase inhibitor followed by affinity isolation and mass-spectrometric analysis of phosphorylated proteins by phosphotyrosine-specific antibodies. Second, we used a substrate trapping mutant of VE-PTP to pull down phosphorylated substrates in combination with SILAC-based quantitative mass spectrometry measurements. We identified a set of substrate candidates of VE-PTP, of which a remarkably large fraction (29%) is related to cell junctions. Several of those were found in both screens and displayed very high connectivity in predicted functional interaction networks. The receptor protein tyrosine kinase EPHB4 was the most prominently phosphorylated protein on VE-PTP inhibition among those VE-PTP targets that were identified by both proteomic approaches. Further analysis revealed that EPHB4 forms a ternary complex with VE-PTP and TIE2 in endothelial cells. VE-PTP controls the phosphorylation of each of these two tyrosine kinase receptors. Despite their simultaneous presence in a ternary complex, stimulating each of the receptors with their own specific ligand did not cross-activate the respective partner receptor. Our systematic approach has led to the identification of novel substrates of VE-PTP, of which many are relevant for the control of cellular junctions further promoting the importance of VE-PTP as a key player of junctional signaling.


Subject(s)
Proteomics/methods , Receptor, EphB4/metabolism , Receptor, TIE-2/metabolism , Receptor-Like Protein Tyrosine Phosphatases, Class 3/genetics , Receptor-Like Protein Tyrosine Phosphatases, Class 3/metabolism , Aniline Compounds/pharmacology , Chromatography, Liquid , Endothelial Cells , Human Umbilical Vein Endothelial Cells , Humans , Intercellular Junctions , Mutation , Phosphorylation/drug effects , Protein Multimerization , Protein Structure, Quaternary , Receptor, EphB4/chemistry , Receptor, TIE-2/chemistry , Receptor-Like Protein Tyrosine Phosphatases, Class 3/chemistry , Substrate Specificity , Sulfonic Acids/pharmacology , Tandem Mass Spectrometry
6.
J Cell Biochem ; 120(10): 17015-17029, 2019 10.
Article in English | MEDLINE | ID: mdl-31125141

ABSTRACT

Diabetic macular edema, also known as diabetic eye disease, is mainly caused by the overexpression of vascular endothelial protein tyrosine phosphatase (VE-PTP) at hypoxia/ischemic. AKB-9778 is a known VE-PTP inhibitor that can effectively interact with the active site of VE-PTP to inhibit the activity of VE-PTP. However, the binding pattern of VE-PTP with AKB-9778 and the dynamic implications of AKB-9778 on VE-PTP system at the molecular level are poorly understood. Through molecular docking, it was found that the AKB-9778 was docked well in the binding pocket of VE-PTP by the interactions of hydrogen bond and Van der Waals. Furthermore, after molecular dynamic simulations on VE-PTP system and VE-PTP AKB-9778 system, a series of postdynamic analyses found that the flexibility and conformation of the active site undergone an obvious transition after VE-PTP binding with AKB-9778. Moreover, by constructing the RIN, it was found that the different interactions in the active site were the detailed reasons for the conformational differences between these two systems. Thus, the finding here might provide a deeper understanding of AKB-9778 as VE-PTP Inhibitor.


Subject(s)
Aniline Compounds/chemistry , Enzyme Inhibitors/chemistry , Hypoglycemic Agents/chemistry , Molecular Docking Simulation , Receptor-Like Protein Tyrosine Phosphatases, Class 3/chemistry , Sulfonic Acids/chemistry , Amino Acid Motifs , Aniline Compounds/metabolism , Catalytic Domain , Enzyme Inhibitors/metabolism , Humans , Hydrogen Bonding , Hypoglycemic Agents/metabolism , Kinetics , Molecular Dynamics Simulation , Protein Binding , Protein Conformation, alpha-Helical , Protein Conformation, beta-Strand , Protein Interaction Domains and Motifs , Receptor-Like Protein Tyrosine Phosphatases, Class 3/antagonists & inhibitors , Receptor-Like Protein Tyrosine Phosphatases, Class 3/metabolism , Sulfonic Acids/metabolism , Thermodynamics
7.
J Chem Inf Model ; 58(11): 2331-2342, 2018 11 26.
Article in English | MEDLINE | ID: mdl-30299094

ABSTRACT

Accurate protein structure in the ligand-bound state is a prerequisite for successful structure-based virtual screening (SBVS). Therefore, applications of SBVS against targets for which only an apo structure is available may be severely limited. To address this constraint, we developed a computational strategy to explore the ligand-bound state of a target protein, by combined use of molecular dynamics simulation, MM/GBSA binding energy calculation, and fragment-centric topographical mapping. Our computational strategy is validated against low-molecular weight protein tyrosine phosphatase (LMW-PTP) and then successfully employed in the SBVS against protein tyrosine phosphatase receptor type O (PTPRO), a potential therapeutic target for various diseases. The most potent hit compound GP03 showed an IC50 value of 2.89 µM for PTPRO and possessed a certain degree of selectivity toward other protein phosphatases. Importantly, we also found that neglecting the ligand energy penalty upon binding partially accounts for the false positive SBVS hits. The preliminary structure-activity relationships of GP03 analogs are also reported.


Subject(s)
Computer-Aided Design , Drug Design , Receptor-Like Protein Tyrosine Phosphatases, Class 3/antagonists & inhibitors , Receptor-Like Protein Tyrosine Phosphatases, Class 3/metabolism , Small Molecule Libraries/chemistry , Small Molecule Libraries/pharmacology , Humans , Ligands , Molecular Docking Simulation , Molecular Dynamics Simulation , Protein Binding , Receptor-Like Protein Tyrosine Phosphatases, Class 3/chemistry , Thermodynamics
8.
Bioorg Chem ; 81: 270-277, 2018 12.
Article in English | MEDLINE | ID: mdl-30165257

ABSTRACT

A series of novel (3'-amino-[1,1'-biphenyl]-4-yl) sulfamic acid derivatives were designed as nonphosphonate-based phosphotyrosy (pTyr) mimetics, synthesized and screened for use as HPTPß inhibitors. Compounds C22 and C2 showed favorable HPTPß inhibitory activity and better selectivity for HPTPß than for PTP1B and SHP2. Docking results suggested that compounds C2 and C22 could not only efficiently fit into the catalytic site of the HPTPß enzyme but also interact with the Lys1807, Arg1809 and Lys1811 residues of the secondary binding site, which was next to the catalytic center of the enzyme. The mode of interaction of the synthesized compound with the protein was different from the one found in a complex crystal of small molecules with HPTPß (2I4H), in which the inhibitory molecule formed hydrogen bonds with the Gln1948 and Asn1735 residues of the secondary binding site.


Subject(s)
Biphenyl Compounds/chemistry , Receptor-Like Protein Tyrosine Phosphatases, Class 3/antagonists & inhibitors , Sulfonic Acids/chemistry , Biphenyl Compounds/chemical synthesis , Catalytic Domain , Drug Design , Humans , Molecular Docking Simulation , Molecular Structure , Receptor-Like Protein Tyrosine Phosphatases, Class 3/chemistry , Structure-Activity Relationship , Sulfonic Acids/chemical synthesis
9.
Neural Plast ; 2018: 9425725, 2018.
Article in English | MEDLINE | ID: mdl-29849575

ABSTRACT

PTPRQ gene, encoding protein tyrosine phosphatase receptor Q, is essential for the normal maturation and function of hair bundle in the cochlea. Its mutations can cause the defects of stereocilia in hair cell, which lead to nonsyndromic sensorineural hearing loss. Using next-generation sequencing and Sanger sequencing method, we identified a novel compound heterozygous missense mutation, c.4472C>T p.T1491M (maternal allele) and c.1973T>C p.V658A (paternal allele), in PTPRQ gene. The two mutations are the first reported to be the cause of recessively inherited sensorineural hearing loss. Hearing loss levels and progression involved by PTPRQ mutations among the existing cases seem to be varied, and the relationship between genotypes and phenotypes is unclear. Our data here further prove the important role of PTPRQ in auditory function and provide more information for the further mechanism research of PTPRQ-related hearing loss.


Subject(s)
Genetic Predisposition to Disease , Hearing Loss, Sensorineural/genetics , Mutation, Missense , Receptor-Like Protein Tyrosine Phosphatases, Class 3/genetics , Asian People/genetics , Child, Preschool , China , Female , Genes, Recessive , Hearing Loss, Sensorineural/congenital , Heterozygote , Humans , Mutant Proteins/chemistry , Pedigree , Protein Structure, Tertiary , Receptor-Like Protein Tyrosine Phosphatases, Class 3/chemistry
10.
Molecules ; 23(3)2018 Mar 02.
Article in English | MEDLINE | ID: mdl-29498714

ABSTRACT

Protein tyrosine phosphatases (PTPs), of the receptor and non-receptor classes, are key signaling molecules that play critical roles in cellular regulation underlying diverse physiological events. Aberrant signaling as a result of genetic mutation or altered expression levels has been associated with several diseases and treatment via pharmacological intervention at the level of PTPs has been widely explored; however, the challenges associated with development of small molecule phosphatase inhibitors targeting the intracellular phosphatase domain (the "inside-out" approach) have been well documented and as yet there are no clinically approved drugs targeting these enzymes. The alternative approach of targeting receptor PTPs with biotherapeutic agents (such as monoclonal antibodies or engineered fusion proteins; the "outside-in" approach) that interact with the extracellular ectodomain offers many advantages, and there have been a number of exciting recent developments in this field. Here we provide a brief overview of the receptor PTP family and an update on the emerging area of receptor PTP-targeted biotherapeutics for CD148, vascular endothelial-protein tyrosine phosphatase (VE-PTP), receptor-type PTPs σ, γ, ζ (RPTPσ, RPTPγ, RPTPζ) and CD45, and discussion of future potential in this area.


Subject(s)
Antibodies, Neutralizing/pharmacology , Enzyme Inhibitors/pharmacology , Immunoconjugates/pharmacology , Receptor-Like Protein Tyrosine Phosphatases, Class 3/antagonists & inhibitors , Receptor-Like Protein Tyrosine Phosphatases/antagonists & inhibitors , Animals , Arthritis, Rheumatoid/drug therapy , Arthritis, Rheumatoid/enzymology , Arthritis, Rheumatoid/genetics , Arthritis, Rheumatoid/pathology , Asthma/drug therapy , Asthma/enzymology , Asthma/genetics , Asthma/pathology , Enzyme Inhibitors/chemical synthesis , Gene Expression Regulation , Humans , Immunoconjugates/chemistry , Immunotoxins/chemistry , Molecular Targeted Therapy , Neoplasms/drug therapy , Neoplasms/enzymology , Neoplasms/genetics , Neoplasms/pathology , Protein Domains , Receptor-Like Protein Tyrosine Phosphatases/chemistry , Receptor-Like Protein Tyrosine Phosphatases/genetics , Receptor-Like Protein Tyrosine Phosphatases/metabolism , Receptor-Like Protein Tyrosine Phosphatases, Class 3/chemistry , Receptor-Like Protein Tyrosine Phosphatases, Class 3/genetics , Receptor-Like Protein Tyrosine Phosphatases, Class 3/metabolism , Ribosome Inactivating Proteins, Type 1/chemistry , Saporins , Signal Transduction
11.
PLoS One ; 12(9): e0184574, 2017.
Article in English | MEDLINE | ID: mdl-28926625

ABSTRACT

Receptor-type protein tyrosine phosphatases (RPTPs) of the R3 subgroup play key roles in the immune, vascular and nervous systems. They are characterised by a large ectodomain comprising multiple FNIII-like repeats, a transmembrane domain, and a single intracellular phosphatase domain. The functional role of the extracellular region has not been clearly defined and potential roles in ligand interaction, dimerization, and regulation of cell-cell contacts have been reported. Here bimolecular fluorescence complementation (BiFC) in live cells was used to examine the molecular basis for the interaction of VE-PTP with VE-cadherin, two proteins involved in endothelial cell contact and maintenance of vascular integrity. The potential of other R3-PTPs to interact with VE-cadherin was also explored using this method. Quantitative BiFC analysis, using a VE-PTP construct expressing only the ectodomain and transmembrane domain, revealed a specific interaction with VE-cadherin, when compared with controls. Controls were sialophorin, an unrelated membrane protein with a large ectodomain, and a membrane anchored C-terminal Venus-YFP fragment, lacking both ectodomain and transmembrane domains. Truncation of the first 16 FNIII-like repeats from the ectodomain of VE-PTP indicated that removal of this region is not sufficient to disrupt the interaction with VE-cadherin, although it occurs predominantly in an intracellular location. A construct with a deletion of only the 17th domain of VE-PTP was, in contrast to previous studies, still able to interact with VE-cadherin, although this also was predominantly intracellular. Other members of the R3-PTP family (DEP-1, GLEPP1 and SAP-1) also exhibited the potential to interact with VE-cadherin. The direct interaction of DEP-1 with VE-cadherin is likely to be of physiological relevance since both proteins are expressed in endothelial cells. Together the data presented in the study suggest a role for both the ectodomain and transmembrane domain of R3-PTPs in interaction with VE-cadherin.


Subject(s)
Antigens, CD/metabolism , Cadherins/metabolism , Receptor-Like Protein Tyrosine Phosphatases, Class 3/metabolism , Antigens, CD/chemistry , Antigens, CD/genetics , Cadherins/chemistry , Cadherins/genetics , Genetic Vectors/metabolism , HEK293 Cells , Humans , Microscopy, Confocal , Mutagenesis , Receptor-Like Protein Tyrosine Phosphatases, Class 3/chemistry , Receptor-Like Protein Tyrosine Phosphatases, Class 3/genetics , Recombinant Fusion Proteins/biosynthesis , Recombinant Fusion Proteins/chemistry , Transfection
12.
J Biol Chem ; 292(35): 14371-14380, 2017 09 01.
Article in English | MEDLINE | ID: mdl-28684416

ABSTRACT

Regulation of growth factor signaling involves reversible inactivation of protein tyrosine phosphatases (PTPs) through the oxidation and reduction of their active site cysteine. However, there is limited mechanistic understanding of these redox events and their co-ordination in the presence of cellular antioxidant networks. Here we investigated interactions between PTP1B and the peroxiredoxin 2 (Prx2)/thioredoxin 1 (Trx1)/thioredoxin reductase 1 (TrxR1) network. We found that Prx2 becomes oxidized in PDGF-treated fibroblasts, but only when TrxR1 has first been inhibited. Using purified proteins, we also found that PTP1B is relatively insensitive to inactivation by H2O2 but found no evidence for a relay mechanism in which Prx2 or Trx1 facilitates PTP1B oxidation. Instead, these proteins prevented PTP1B inactivation by H2O2 Intriguingly, we discovered that TrxR1/NADPH directly protects PTP1B from inactivation when present during the H2O2 exposure. This protection was dependent on the concentration of TrxR1 and independent of Trx1 and Prx2. The protection was blocked by auranofin and required an intact selenocysteine residue in TrxR1. This activity likely involves reduction of the sulfenic acid intermediate form of PTP1B by TrxR1 and is therefore distinct from the previously described reactivation of end-point oxidized PTP1B, which requires both Trx1 and TrxR1. The ability of TrxR1 to directly reduce an oxidized phosphatase is a novel activity that can help explain previously observed increases in PTP1B oxidation and PDGF receptor phosphorylation in TrxR1 knockout cells. The activity of TrxR1 is therefore of potential relevance for understanding the mechanisms of redox regulation of growth factor signaling pathways.


Subject(s)
NADP/metabolism , Protein Tyrosine Phosphatase, Non-Receptor Type 1/metabolism , Receptor-Like Protein Tyrosine Phosphatases, Class 3/metabolism , Thioredoxin Reductase 1/metabolism , Animals , Auranofin/pharmacology , Catalytic Domain , Cells, Cultured , Dimerization , Embryo, Mammalian/cytology , Homeodomain Proteins/chemistry , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Humans , Hydrogen Peroxide/pharmacology , Mice , Oxidants/pharmacology , Oxidation-Reduction , Peptide Fragments/chemistry , Peptide Fragments/genetics , Peptide Fragments/metabolism , Protein Interaction Domains and Motifs , Protein Tyrosine Phosphatase, Non-Receptor Type 1/antagonists & inhibitors , Protein Tyrosine Phosphatase, Non-Receptor Type 1/genetics , Rats , Receptor-Like Protein Tyrosine Phosphatases, Class 3/chemistry , Receptor-Like Protein Tyrosine Phosphatases, Class 3/genetics , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/metabolism , Selenocysteine/chemistry , Selenocysteine/metabolism , Thioredoxin Reductase 1/antagonists & inhibitors , Thioredoxin Reductase 1/chemistry , Thioredoxin Reductase 1/genetics , Thioredoxins/chemistry , Thioredoxins/genetics , Thioredoxins/metabolism
13.
PLoS One ; 10(4): e0124757, 2015.
Article in English | MEDLINE | ID: mdl-25919374

ABSTRACT

Mutations in PTPRQ are associated with deafness in humans due to defects of stereocilia in hair cells. Using whole exome sequencing, we identified responsible gene of family 1572 with autosomal recessively non-syndromic hearing loss (ARNSHL). We also used DNA from 74 familial patients with ARNSHL and 656 ethnically matched control chromosomes to perform extended variant analysis. We identified two novel compound heterozygous missense mutations, c. 3125 A>G p.D1042G (maternal allele) and c.5981 A>G p.E1994G (paternal allele), in the PTPRQ gene, as the cause of recessively inherited sensorineural hearing loss in family 1572. Both variants co-segregated with hearing loss phenotype in family 1572, but were absent in 74 familial patients. Heterozygosity for c. 3125 A>G was identified in two samples from unaffected Chinese individuals (656 chromosomes). Therefore, the hearing loss in this family was caused by two novel compound heterozygous mutations in PTPRQ.


Subject(s)
Asian People/genetics , Genes, Recessive , Genetic Association Studies , Genetic Predisposition to Disease , Hearing Loss/genetics , Mutation/genetics , Receptor-Like Protein Tyrosine Phosphatases, Class 3/genetics , Amino Acid Sequence , Base Sequence , Child , DNA Mutational Analysis , Exome/genetics , Family , Female , Heterozygote , Humans , Male , Models, Molecular , Molecular Sequence Data , Mutant Proteins/chemistry , Pedigree , Receptor-Like Protein Tyrosine Phosphatases, Class 3/chemistry , Young Adult
14.
Theor Biol Med Model ; 10: 49, 2013 Aug 28.
Article in English | MEDLINE | ID: mdl-23981594

ABSTRACT

Protein tyrosine phosphatase receptor type Q (PTPRQ) is an unusual PTP that has intrinsic dephosphorylating activity for various phosphatidyl inositides instead of phospho-tyrosine substrates. Although PTPRQ was known to be involved in the pathogenesis of obesity, no small-molecule inhibitor has been reported so far. Here we report six novel PTPRQ inhibitors identified with computer-aided drug design protocol involving the virtual screening with docking simulations and enzyme inhibition assay. These inhibitors exhibit moderate potencies against PTPRQ with the associated IC50 values ranging from 29 to 86 µM. Because the newly discovered inhibitors were also computationally screened for having desirable physicochemical properties as a drug candidate, they deserve consideration for further development by structure-activity relationship studies to optimize the antiobestic activities. Structural features relevant to the stabilization of the inhibitors in the active site of PTPRQ are addressed in detail.


Subject(s)
Drug Evaluation, Preclinical , Enzyme Inhibitors/analysis , Enzyme Inhibitors/pharmacology , Molecular Docking Simulation , Receptor-Like Protein Tyrosine Phosphatases, Class 3/antagonists & inhibitors , User-Computer Interface , Catalytic Domain , Enzyme Inhibitors/chemistry , Humans , Hydrolysis/drug effects , Inhibitory Concentration 50 , Inositol Phosphates/metabolism , Receptor-Like Protein Tyrosine Phosphatases, Class 3/chemistry , Receptor-Like Protein Tyrosine Phosphatases, Class 3/metabolism
15.
J Biol Chem ; 288(32): 23421-31, 2013 Aug 09.
Article in English | MEDLINE | ID: mdl-23814054

ABSTRACT

Receptor-like protein-tyrosine phosphatases (RPTPs) are involved in various aspects of cellular functions, such as proliferation, differentiation, survival, migration, and metabolism. A small number of RPTPs have been reported to regulate activities of some cellular proteins including receptor protein-tyrosine kinases (RPTKs). However, our understanding about the roles of individual RPTPs in the regulation of RPTKs is still limited. The R3 RPTP subfamily reportedly plays pivotal roles in the development of several tissues including the vascular and nervous systems. Here, we examined enzyme-substrate relationships between the four R3 RPTP subfamily members and 21 RPTK members selected from 14 RPTK subfamilies by using a mammalian two-hybrid system with substrate-trapping RPTP mutants. Among the 84 RPTP-RPTK combinations conceivable, we detected 30 positive interactions: 25 of the enzyme-substrate relationships were novel. We randomly chose several RPTKs assumed to be substrates for R3 RPTPs, and validated the results of this screen by in vitro dephosphorylation assays, and by cell-based assays involving overexpression and knock-down experiments. Because their functional relationships were verified without exception, it is probable that the RPTKs identified as potential substrates are actually physiological substrates for the R3 RPTPs. Interestingly, some RPTKs were recognized as substrates by all R3 members, but others were recognized by only one or a few members. The enzyme-substrate relationships identified in the present study will shed light on physiological roles of the R3 RPTP subfamily.


Subject(s)
Receptor Protein-Tyrosine Kinases/metabolism , Receptor-Like Protein Tyrosine Phosphatases, Class 3/metabolism , Animals , COS Cells , Chlorocebus aethiops , Humans , Mice , Phosphorylation/physiology , Receptor Protein-Tyrosine Kinases/chemistry , Receptor Protein-Tyrosine Kinases/genetics , Receptor-Like Protein Tyrosine Phosphatases, Class 3/chemistry , Receptor-Like Protein Tyrosine Phosphatases, Class 3/genetics , Substrate Specificity/physiology , Two-Hybrid System Techniques
16.
Acta Crystallogr D Biol Crystallogr ; 69(Pt 8): 1522-9, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23897475

ABSTRACT

Unlike other classical protein tyrosine phosphatases (PTPs), PTPRQ (PTP receptor type Q) has dephosphorylating activity towards phosphatidylinositide (PI) substrates. Here, the structure of the catalytic domain of PTPRQ was solved at 1.56 Šresolution. Overall, PTPRQ adopts a tertiary fold typical of other classical PTPs. However, the disordered M6 loop of PTPRQ surrounding the catalytic core and the concomitant absence of interactions of this loop with residues in the PTP loop results in a flat active-site pocket. On the basis of structural and biochemical analyses, it is proposed that this structural feature might facilitate the accommodation of large substrates, making it suitable for the dephosphorylation of PI substrates. Moreover, subsequent kinetic experiments showed that PTPRQ has a strong preferences for PI(3,4,5)P3 over other PI substrates, suggesting that its regulation of cell survival and proliferation reflects downregulation of Akt signalling.


Subject(s)
Receptor-Like Protein Tyrosine Phosphatases, Class 3/chemistry , Receptor-Like Protein Tyrosine Phosphatases, Class 3/metabolism , Catalytic Domain , Crystallography, X-Ray , Humans , Kinetics , Models, Molecular , Mutation , Phosphatidylinositol Phosphates/chemistry , Phosphatidylinositol Phosphates/metabolism , Phosphorylation , Protein Conformation , Receptor-Like Protein Tyrosine Phosphatases, Class 3/genetics , Substrate Specificity
17.
ACS Chem Biol ; 8(7): 1497-506, 2013 Jul 19.
Article in English | MEDLINE | ID: mdl-23627474

ABSTRACT

PTPRJ is a receptor protein tyrosine phosphatase involved in both physiological and oncogenic pathways. We previously reported that its expression is strongly reduced in the majority of explored cancer cell lines and tumor samples; moreover, its restoration blocks in vitro cancer cell proliferation and in vivo tumor formation. By means of a phage display library screening, we recently identified two peptides able to bind and activate PTPRJ, resulting in cell growth inhibition and apoptosis of both cancer and endothelial cells. Here, on a previously discovered PTPRJ agonist peptide, PTPRJ-pep19, we synthesized and assayed a panel of nonapeptide analogues with the aim to identify specific amino acid residues responsible for peptide activity. These second-generation nonapeptides were tested on both cancer and primary endothelial cells (HeLa and HUVEC, respectively); interestingly, one of them (PTPRJ-19.4) was able to both dramatically reduce cell proliferation and effectively trigger apoptosis of both HeLa and HUVECs compared to its first-generation counterpart. Moreover, PTPRJ-pep19.4 significantly inhibited in vitro tube formation on Matrigel. Intriguingly, while ERK1/2 phosphorylation and cell proliferation were both inhibited by PTPRJ-pep19.4 in breast cancer cells (MCF-7 and SKBr3), no effects were observed on primary normal human mammary endothelial cells (HMEC). We further characterized these peptides by molecular modeling and NMR experiments reporting, for the most active peptide, the possibility of self-aggregation states and highlighting new hints of structure-activity relationship. Thus, our results indicate that this nonapeptide might represent a great potential lead for the development of novel targeted anticancer drugs.


Subject(s)
Oligopeptides/chemistry , Oligopeptides/pharmacology , Peptides/chemistry , Peptides/pharmacology , Amino Acid Sequence , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Apoptosis , Breast Neoplasms/drug therapy , Cell Line , Cell Proliferation/drug effects , Female , HeLa Cells , Humans , Magnetic Resonance Spectroscopy , Models, Molecular , Monte Carlo Method , Protein Folding/drug effects , Receptor-Like Protein Tyrosine Phosphatases, Class 3/chemistry
18.
Blood ; 121(21): 4295-302, 2013 May 23.
Article in English | MEDLINE | ID: mdl-23580664

ABSTRACT

T-cell receptor (TCR) triggering results in a cascade of intracellular tyrosine phosphorylation events that ultimately leads to T-cell activation. It is dependent on changes in the relative activities of membrane-associated tyrosine kinases and phosphatases near the engaged TCR. CD45 and CD148 are transmembrane tyrosine phosphatases with large ectodomains that have activatory and inhibitory effects on TCR triggering. This study investigates whether and how the ectodomains of CD45 and CD148 modulate their inhibitory effect on TCR signaling. Expression in T cells of forms of these phosphatases with truncated ectodomains inhibited TCR triggering. In contrast, when these phosphatases were expressed with large ectodomains, they had no inhibitory effect. Imaging studies revealed that truncation of the ectodomains enhanced colocalization of these phosphatases with ligated TCR at the immunological synapse. Our results suggest that the large ectodomains of CD45 and CD148 modulate their inhibitory effect by enabling their passive, size-based segregation from ligated TCR, supporting the kinetic-segregation model of TCR triggering.


Subject(s)
CD4-Positive T-Lymphocytes/metabolism , CD8-Positive T-Lymphocytes/metabolism , Leukocyte Common Antigens/metabolism , Lymphocyte Activation/immunology , Receptors, Antigen, T-Cell/metabolism , Animals , CD4-Positive T-Lymphocytes/cytology , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/cytology , CD8-Positive T-Lymphocytes/immunology , Cell Line , Interleukin-2/metabolism , Leukocyte Common Antigens/chemistry , Leukocyte Common Antigens/genetics , Lymphocyte Activation/genetics , Mice , Models, Chemical , Mutagenesis/immunology , Protein Binding/immunology , Protein Structure, Tertiary/physiology , Receptor-Like Protein Tyrosine Phosphatases, Class 3/chemistry , Receptor-Like Protein Tyrosine Phosphatases, Class 3/genetics , Receptor-Like Protein Tyrosine Phosphatases, Class 3/metabolism , Receptors, Antigen, T-Cell/immunology , Signal Transduction/genetics , Signal Transduction/immunology , Structure-Activity Relationship
19.
Mol Cell Biol ; 32(19): 3913-24, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22851698

ABSTRACT

Identification of genes that are upregulated during mammary epithelial cell morphogenesis may reveal novel regulators of tumorigenesis. We have demonstrated that gene expression programs in mammary epithelial cells grown in monolayer cultures differ significantly from those in three-dimensional (3D) cultures. We identify a protein tyrosine phosphate, PTPRO, that was upregulated in mature MCF-10A mammary epithelial 3D structures but had low to undetectable levels in monolayer cultures. Downregulation of PTPRO by RNA interference inhibited proliferation arrest during morphogenesis. Low levels of PTPRO expression correlated with reduced survival for breast cancer patients, suggesting a tumor suppressor function. Furthermore, we showed that the receptor tyrosine kinase ErbB2/HER2 is a direct substrate of PTPRO and that loss of PTPRO increased ErbB2-induced cell proliferation and transformation, together with tyrosine phosphorylation of ErbB2. Moreover, in patients with ErbB2-positive breast tumors, low PTPRO expression correlated with poor clinical prognosis compared to ErbB2-positive patients with high levels of PTPRO. Thus, PTPRO is a novel regulator of ErbB2 signaling, a potential tumor suppressor, and a novel prognostic marker for patients with ErbB2-positive breast cancers. We have identified the protein tyrosine phosphatase PTPRO as a regulator of three-dimensional epithelial morphogenesis of mammary epithelial cells and as a regulator of ErbB2-mediated transformation. In addition, we demonstrated that ErbB2 is a direct substrate of PTPRO and that decreased expression of PTPRO predicts poor prognosis for ErbB2-positive breast cancer patients. Thus, our results identify PTPRO as a novel regulator of mammary epithelial transformation, a potential tumor suppressor, and a predictive biomarker for breast cancer.


Subject(s)
Breast Neoplasms/genetics , Mammary Glands, Human/cytology , Receptor, ErbB-2/metabolism , Receptor-Like Protein Tyrosine Phosphatases, Class 3/genetics , Receptor-Like Protein Tyrosine Phosphatases, Class 3/metabolism , Breast/metabolism , Breast/pathology , Breast Neoplasms/diagnosis , Breast Neoplasms/metabolism , Cell Death , Cell Line, Tumor , Cell Proliferation , Down-Regulation , Female , Gene Expression Regulation, Developmental , Gene Expression Regulation, Neoplastic , Humans , Mammary Glands, Human/growth & development , Mammary Glands, Human/metabolism , Prognosis , Protein Structure, Tertiary , Receptor, ErbB-2/genetics , Receptor-Like Protein Tyrosine Phosphatases, Class 3/chemistry , Transcriptome , Up-Regulation
20.
Proc Natl Acad Sci U S A ; 109(6): 1985-90, 2012 Feb 07.
Article in English | MEDLINE | ID: mdl-22308318

ABSTRACT

CD148 is a receptor-type protein tyrosine phosphatase that is expressed in several cell types, including vascular endothelial cells and duct epithelial cells. Growing evidence demonstrates a prominent role for CD148 in negative regulation of growth factor signals, suppressing cell proliferation and transformation. However, its extracellular ligand(s) remain unknown. To identify the ligand(s) of CD148, we introduced HA-tagged CD148 into cultured endothelial cells and then isolated its interacting extracellular protein(s) by biotin surface labeling and subsequent affinity purifications. The binding proteins were identified by mass spectrometry. Here we report that soluble thrombospondin-1 (TSP1) binds to the extracellular part of CD148 with high affinity and specificity, and its binding increases CD148 catalytic activity, leading to dephosphorylation of the substrate proteins. Consistent with these findings, introduction of CD148 conferred TSP1-mediated inhibition of cell growth to cells which lack CD148 and TSP1 inhibition of growth. Further, we demonstrate that TSP1-mediated inhibition of endothelial cell growth is antagonized by soluble CD148 ectodomain as well as by CD148 gene silencing. These findings provide evidence that CD148 functions as a receptor for TSP1 and mediates its inhibition of cell growth.


Subject(s)
Thrombospondin 1/metabolism , Cell Proliferation , Extracellular Space/metabolism , Gene Knockdown Techniques , Human Umbilical Vein Endothelial Cells/cytology , Human Umbilical Vein Endothelial Cells/enzymology , Humans , Kidney/cytology , Ligands , Microvessels/cytology , Protein Binding , Protein Structure, Tertiary , Receptor-Like Protein Tyrosine Phosphatases, Class 3/chemistry , Receptor-Like Protein Tyrosine Phosphatases, Class 3/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...