Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 107
Filter
1.
EMBO Rep ; 24(8): e57499, 2023 08 03.
Article in English | MEDLINE | ID: mdl-37401859

ABSTRACT

Abnormal tau protein impairs mitochondrial function, including transport, dynamics, and bioenergetics. Mitochondria interact with the endoplasmic reticulum (ER) via mitochondria-associated ER membranes (MAMs), which coordinate and modulate many cellular functions, including mitochondrial cholesterol metabolism. Here, we show that abnormal tau loosens the association between theĀ ER and mitochondria in vivo and in vitro. Especially, ER-mitochondria interactions via vesicle-associated membrane protein-associated protein (VAPB)-protein tyrosine phosphatase-interacting protein 51 (PTPIP51) are decreased in the presence of abnormal tau. Disruption of MAMs in cells with abnormal tau alters the levels of mitochondrial cholesterol and pregnenolone, indicating that conversion of cholesterol into pregnenolone is impaired. Opposite effects are observed in the absence of tau. Besides, targeted metabolomics reveals overall alterations in cholesterol-related metabolites by tau. The inhibition of GSK3Ɵ decreases abnormal tau hyperphosphorylation and increases VAPB-PTPIP51 interactions, restoring mitochondrial cholesterol and pregnenolone levels. This study is the first to highlight a link between tau-induced impairments in the ER-mitochondria interaction and cholesterol metabolism.


Subject(s)
Mitochondria , tau Proteins , tau Proteins/metabolism , Mitochondria/metabolism , Endoplasmic Reticulum/metabolism , Protein Tyrosine Phosphatases/metabolism , Protein Tyrosine Phosphatases/pharmacology , Cholesterol/metabolism
2.
Arch Pharm (Weinheim) ; 356(3): e2200459, 2023 Mar.
Article in English | MEDLINE | ID: mdl-36417559

ABSTRACT

A series of 1H-1,2,3-triazole-4H-chromene-D-glucose hybrid compounds 7a-w were synthesized using click chemistry of 2-amino-7-propargyloxy-4H-chromene-3-carbonitriles 5a-w. CuNPs@montmorillonite was used as a catalyst in the presence of DIPEA as an additive for this chemistry. All synthesized 1H-1,2,3-triazoles were examined for in vitro inhibition against Mycobacterium tuberculosis protein tyrosine phosphatase B (MtbPtpB). Nine 1H-1,2,3-triazoles, including 7c-e, 7h, 7i, and 7r-t, displayed remarkable inhibitory activity against MtbPtpB with IC50 < 10 ĀµM; compound 7t exhibited the most potent inhibition in vitro with an IC50 value of 0.61 ĀµM. Kinetic studies of the three most active compounds, 7c,h,t, showed their competitive inhibition toward the MtbPtpB enzyme. Induced-fit docking and MM-GBSA studies on the enzyme (PDB: 2OZ5) revealed that the most active compound 7t was more effective against MtbPtpB. Residues Arg64, Arg136, Ash165, Arg166, and Arg63 in the binding pocket were identified as potential ligand-binding hot-spot residues for ligand 7t. The binding free energy calculation by the MM-GBSA approach for ligand 7t indicated that Coulomb, lipophilic, and van der Waals energy terms are major contributors to the inhibitor binding. Furthermore, the stability of the ligand-protein complex and the structural insights into the mode of binding were confirmed by 300-ns molecular dynamics simulation of 7t/2OZ5.


Subject(s)
Mycobacterium tuberculosis , Glucose , Structure-Activity Relationship , Triazoles/pharmacology , Triazoles/chemistry , Benzopyrans/pharmacology , Benzopyrans/chemistry , Kinetics , Ligands , Protein Tyrosine Phosphatases/metabolism , Protein Tyrosine Phosphatases/pharmacology , Molecular Docking Simulation
3.
Microsurgery ; 37(6): 624-631, 2017 Sep.
Article in English | MEDLINE | ID: mdl-27859622

ABSTRACT

INTRODUCTION: Inhibition of protein tyrosine phosphatases (PTP) enhances endothelial receptor tyrosine kinases activation and may have beneficial effects on vessel growth and improve blood flow to ischemic tissue. The purpose of this study is to determine influence of hPTPƟ inhibitors on ischemia-reperfusion injury in muscle flap. MATERIALS AND METHODS: Following cremaster muscle dissection, 60 rats divided into 10 experimental groups (placebo and treatment groups following 0, 1, 2, 3, and 4 h of ischemia). Following group-specific treatment (placebo/hPTPƟ inhibitor, 15 mg/kg), 2 h of reperfusion is initiated. Observations are performed at 4 h after completion of reperfusion and microcirculatory hemodynamics and leukocyte-endothelial activation were recorded. RESULTS: Administration of hPTPƟ inhibitor showed preservation of capillary perfusion in group subjected to 2 h of ischemia when compared with placebo (P < .05). The effect of hPTPƟ inhibitor on mean venule diameter was found to be altered by duration of ischemia and this effect was statistically significant (P < .05). Treated ischemic groups (1 h, 2 h, and 3 h) showed decreased activation of rolling, sticking, and transmigrating leukocytes compared to respective placebo groups at all time points. The differences were significant for transmigrating leukocytes after 2 h and 3 h of ischemia (P < .05). Endothelial edema index was also significantly reduced in 2 h ischemia group (P < .05). CONCLUSION: Administration of hPTP inhibitors after submission of tissue to subcritical ischemia (1-2 h) improved functional capillary perfusion and decreased leukocyte-endothelial activation after 4 h after reperfusion. These results indicate that hPTP inhibitor has a potential postischemic therapeutic effect applied after tissue ischemia just before the reperfusion injury.


Subject(s)
Abdominal Muscles/surgery , Microcirculation/drug effects , Protein Tyrosine Phosphatases/antagonists & inhibitors , Reperfusion Injury/drug therapy , Surgical Flaps/blood supply , Abdominal Muscles/transplantation , Analysis of Variance , Animals , Blood Flow Velocity/drug effects , Disease Models, Animal , Male , Protein Tyrosine Phosphatases/pharmacology , Random Allocation , Rats , Rats, Inbred Lew , Reference Values , Statistics, Nonparametric
4.
Toxicol Appl Pharmacol ; 276(1): 47-54, 2014 Apr 01.
Article in English | MEDLINE | ID: mdl-24440445

ABSTRACT

Network and protein-protein interaction analyses of proteins undergoing HgĀ²Ć¢ĀĀŗ-induced phosphorylation and dephosphorylation in HgĀ²Ć¢ĀĀŗ-intoxicated mouse WEHI-231 B cells identified Lyn as the most interconnected node. Lyn is a Src family protein tyrosine kinase known to be intimately involved in the B cell receptor (BCR) signaling pathway. Under normal signaling conditions the tyrosine kinase activity of Lyn is controlled by phosphorylation, primarily of two well known canonical regulatory tyrosine sites, Y-397 and Y-508. However, Lyn has several tyrosine residues that have not yet been determined to play a major role under normal signaling conditions, but are potentially important sites for phosphorylation following mercury exposure. In order to determine how HgĀ²Ć¢ĀĀŗ exposure modulates the phosphorylation of additional residues in Lyn, a targeted MS assay was developed. Initial mass spectrometric surveys of purified Lyn identified 7 phosphorylated tyrosine residues. A quantitative assay was developed from these results using the multiple reaction monitoring (MRM) strategy. WEHI-231 cells were treated with HgĀ²Ć¢ĀĀŗ, pervanadate (a phosphatase inhibitor), or anti-Ig antibody (to stimulate the BCR). Results from these studies showed that the phosphoproteomic profile of Lyn after exposure of the WEHI-231 cells to a low concentration of HgĀ²Ć¢ĀĀŗ closely resembled that of anti-Ig antibody stimulation, whereas exposure to higher concentrations of HgĀ²Ć¢ĀĀŗ led to increases in the phosphorylation of Y-193/Y-194, Y-501 and Y-508 residues. These data indicate that mercury can disrupt a key regulatory signal transduction pathway in B cells and point to phospho-Lyn as a potential biomarker for mercury exposure.


Subject(s)
B-Lymphocytes/drug effects , Mercury/toxicity , Signal Transduction/drug effects , src-Family Kinases/metabolism , Animals , Autoimmune Diseases/chemically induced , Autoimmune Diseases/immunology , Autoimmune Diseases/metabolism , B-Lymphocytes/immunology , B-Lymphocytes/metabolism , Biomarkers/metabolism , Cell Line , Chromatography, High Pressure Liquid , Mercury Poisoning/enzymology , Mercury Poisoning/immunology , Mercury Poisoning/metabolism , Mice , Osmolar Concentration , Peptide Fragments/agonists , Peptide Fragments/antagonists & inhibitors , Peptide Fragments/chemistry , Peptide Fragments/metabolism , Phosphorylation/drug effects , Protein Processing, Post-Translational/drug effects , Protein Tyrosine Phosphatases/pharmacology , Receptors, Antigen, B-Cell/agonists , Receptors, Antigen, B-Cell/metabolism , Tandem Mass Spectrometry , Tyrosine/metabolism , Vanadates/pharmacology , src-Family Kinases/antagonists & inhibitors , src-Family Kinases/chemistry
5.
Nat Rev Cancer ; 4(9): 688-94, 2004 Sep.
Article in English | MEDLINE | ID: mdl-15343275

ABSTRACT

Infection with strains of Helicobacter pylori that carry the cytotoxin-associated antigen A (cagA) gene is associated with gastric carcinoma. Recent studies have shed light on the mechanism through which the cagA gene product, CagA, elicits pathophysiological actions. CagA is delivered into gastric epithelial cells by the bacterial type IV secretion system, where it deregulates the SHP2 oncoprotein. Intriguingly, CagA is noted for its variation, particularly at the SHP2-binding site, which could affect the potential of different strains of H. pylori to promote gastric carcinogenesis.


Subject(s)
Antigens, Bacterial/pharmacology , Bacterial Proteins/pharmacology , Carcinoma/etiology , Carcinoma/microbiology , Cell Transformation, Neoplastic , Helicobacter Infections/complications , Helicobacter pylori/pathogenicity , Protein Tyrosine Phosphatases/metabolism , Stomach Neoplasms/etiology , Stomach Neoplasms/microbiology , Antigens, Bacterial/metabolism , Bacterial Proteins/metabolism , Bacterial Proteins/pharmacokinetics , Down-Regulation , Humans , Intracellular Signaling Peptides and Proteins , Phosphorylation , Protein Tyrosine Phosphatase, Non-Receptor Type 11 , Protein Tyrosine Phosphatases/pharmacology , SH2 Domain-Containing Protein Tyrosine Phosphatases , src Homology Domains
6.
CNS Neurosci Ther ; 29(1): 365-377, 2023 01.
Article in English | MEDLINE | ID: mdl-36419337

ABSTRACT

INTRODUCTION: Mitochondrial-associated ER membranes (MAMs) control many cellular functions, including calcium and lipid exchange, intracellular trafficking, and mitochondrial biogenesis. The disruption of these functions contributes to neurocognitive disorders, such as spatial memory impairment and premature brain aging. Using neuronal cells, we demonstrated that HIV-1 Tat protein deregulates the mitochondria. METHODS& RESULTS: To determine the mechanisms, we used a neuronal cell line and showed that Tat-induced changes in expression and interactions of both MAM-associated proteins and MAM tethering proteins. The addition of HIV-1 Tat protein alters expression levels of PTPIP51 and VAPB proteins in the MAM fraction but not the whole cell. Phosphorylation of PTPIP51 protein regulates its subcellular localization and function. We demonstrated that the Tat protein promotes PTPIP51 phosphorylation on tyrosine residues and prevents its binding to VAPB. Treatment of the cells with a kinase inhibitor restores the PTPIP51-VAPB interaction and overcomes the effect of Tat. CONCLUSION: These results suggest that Tat disrupts the MAM, through the induction of PTPIP51 phosphorylation, leading to ROS accumulation, mitochondrial stress, and altered movement. Hence, we concluded that interfering in the MAM-associated cellular pathways contributes to spatial memory impairment and premature brain aging often observed in HIV-1-infected patients.


Subject(s)
HIV-1 , Humans , Brain/metabolism , Gene Products, tat/metabolism , Gene Products, tat/pharmacology , HIV-1/metabolism , Mitochondria/metabolism , Protein Tyrosine Phosphatases/metabolism , Protein Tyrosine Phosphatases/pharmacology , Endoplasmic Reticulum/metabolism
7.
Curr Pharm Des ; 29(8): 562-575, 2023.
Article in English | MEDLINE | ID: mdl-36892024

ABSTRACT

Tuberculosis (TB) is a chronic, air-borne infectious disease caused by Mycobacterium tuberculosis (Mtb), which prominently affects the lungs and usually manifests in other organs. TB is preventable and curable but what makes it challenging is the emergence of resistance to the available treatment options. MDR-continued TB's expansion is one of the world's most pressing and difficult problems. Mtb revives via the reciprocity between Mycobacterium and host signalling pathways. Mtb secretes a virulence component called Mycobacterium tuberculosis protein tyrosine phosphatase (MptpB), which helps to survive against host macrophages. It indicates that targeting secreted virulence factors offers more benefits to circumvent the emergence of resistance. Many effective inhibitors of MptpA and MptpB have been discovered, providing a solid foundation for future research and development. Aside from possessing a structurally unique binding site in the Mtb enzyme, MptpB's minimal resemblance to other human phosphatases provides a broad platform for improving selectivity over host PTPs. We believe that addressing several parts of infection processes in the host and bacteria with combination therapy is the greatest way to reduce treatment burden and medication resistance. We have discussed the recent potent, selective, and efficacious MptpB inhibitors, such as natural and marine-based, isoxazole- linked carboxylic acid-based, oxamic acid-based, and lactone-based inhibitors, as potential strategies for treating TB.


Subject(s)
Mycobacterium tuberculosis , Tuberculosis , Humans , Antitubercular Agents/pharmacology , Antitubercular Agents/therapeutic use , Antitubercular Agents/chemistry , Tuberculosis/drug therapy , Signal Transduction , Protein Tyrosine Phosphatases/metabolism , Protein Tyrosine Phosphatases/pharmacology
8.
Bioengineered ; 13(3): 7697-7708, 2022 03.
Article in English | MEDLINE | ID: mdl-35290144

ABSTRACT

Myocardial ischemia-reperfusion injury in diabetic patients leads to an increased incidence of complications and mortality. Secreted frizzled-related protein 4 (SFRP4) plays a critical role in diabetic myocardial ischemia-reperfusion. This paper aims to uncover the underlying mechanisms of SFRP4 in hypoxia/reoxygenation (H/R) injury of diabetic myocardial cells. An in vitro ischemia/reperfusion (I/R) injury model was established using high glucose-induced H9c2 cardiomyocytes. Expression of SFRP4 was detected by real-time reverse transcriptase-polymerase chain reaction and Western blotting. After transfection of SFRP4, the binding of SFRP4 to protein tyrosine phosphatase nonreceptor type 12 (PTPN12) was predicted by database and verified by co-immunoprecipitation assay. P13Ā K/AKT protein levels were examined by Western blotting. PTPN12 levels were tested by RT-qPCR and Western blotting, cell viability by Cell Counting Kit-8, lactose dehydrogenase kit, terminal dUTP nick-end labeling assay, and cell inflammation and oxidative stress by Western blotting and enzyme linked immunosorbent assay. After overexpression of PTPN12, the experiments for cell viability, inflammation and oxidative stress were repeated once more. SFRP4 expression was upregulated in a high-glucose-stimulated H/R cardiomyocyte model. The interference of SFRP4 promoted cell viability, inhibited the inflammatory and oxidative stress response of H/R cardiomyocytes induced by high glucose. SFRP4 interacted with PTPN12 and inhibited the PI3K/AKT signaling pathway. PTPN12 overexpression reversed the inhibitory effect of sh-SFRP4 on H/R cardiomyocyte damage induced by high glucose. Downregulation of SFRP4 inhibited H/R cell damage in diabetic cardiomyocytes by binding to PTPN12.


Subject(s)
Diabetes Mellitus , Myocytes, Cardiac , Apoptosis/genetics , Down-Regulation , Glucose/metabolism , Glucose/toxicity , Humans , Hypoxia/metabolism , Inflammation/metabolism , Intracellular Signaling Peptides and Proteins , Myocytes, Cardiac/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Protein Tyrosine Phosphatase, Non-Receptor Type 12/metabolism , Protein Tyrosine Phosphatases/metabolism , Protein Tyrosine Phosphatases/pharmacology , Proto-Oncogene Proteins c-akt/metabolism
9.
Curr Pharm Biotechnol ; 23(7): 920-931, 2022.
Article in English | MEDLINE | ID: mdl-34375185

ABSTRACT

Protein Tyrosine Phosphatase (PTP) superfamily is a key enzyme involved in the regulation of growth-related cell signaling cascades, such as the RAS/MAPK pathway, that directly affect cancer cell growth and metastasis. Several studies have indicated that the drug resistance observed in several late-stage tumors might also be affected by the levels of PTP in the cell. Hence, these phosphatases have been in the limelight for the past few decades as potential drug targets and several promising drug candidates have been developed, even though none of these drugs have reached the market yet. In this review, we explore the potential of PTP as a viable anti-cancer drug target by studying PTPs, their regulation of several key cancer cell signaling pathways, and how their levels affect various types of cancer. Furthermore, we present the current scenario of PTP as a molecular target and the various challenges faced in the development of PTP-targeting anti-cancer drugs.


Subject(s)
Antineoplastic Agents , Neoplasms , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Drug Delivery Systems , Humans , Neoplasms/drug therapy , Protein Tyrosine Phosphatases/metabolism , Protein Tyrosine Phosphatases/pharmacology , Protein Tyrosine Phosphatases/therapeutic use , Signal Transduction
10.
FEBS Lett ; 596(12): 1503-1515, 2022 06.
Article in English | MEDLINE | ID: mdl-35397176

ABSTRACT

The Mycobacterium tuberculosis tyrosine-specific phosphatase MptpA and its cognate kinase PtkA are prospective targets for anti-tuberculosis drugs as they interact with the host defense response within the macrophages. Although both are structurally well-characterized, the functional mechanism regulating their activity remains poorly understood. Here, we investigate the effect of post-translational oxidation in regulating the function of MptpA. Treatment of MptpA with H2 O2 /NaHCO3 , mimicking cellular oxidative stress conditions, leads to oxidation of the catalytic cysteine (C11) and to a conformational rearrangement of the phosphorylation loop (D-loop) by repositioning the conserved tyrosine 128 (Y128) and generating a temporarily inactive preclosed state of the phosphatase. Thus, the catalytic cysteine in the P-loop acts as a redox switch and regulates the phosphatase activity of MptpA.


Subject(s)
Bacterial Proteins , Mycobacterium tuberculosis , Protein Tyrosine Phosphatases , Virulence Factors , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Cysteine/metabolism , Mycobacterium tuberculosis/genetics , Mycobacterium tuberculosis/metabolism , Mycobacterium tuberculosis/pathogenicity , Oxidation-Reduction , Prospective Studies , Protein Tyrosine Phosphatases/genetics , Protein Tyrosine Phosphatases/metabolism , Protein Tyrosine Phosphatases/pharmacology , Tyrosine/metabolism , Virulence Factors/genetics , Virulence Factors/metabolism
11.
Plant Sci ; 323: 111396, 2022 Oct.
Article in English | MEDLINE | ID: mdl-35878696

ABSTRACT

Serotonin (5-hydroxytryptamine) acts as a neurotransmitter in mammals and is widely distributed in the plant kingdom, where it influences root growth and defense. Mitogen-Activated Protein Kinases (MAPKs) and MAPK phosphatases (MKPs) play critical functions in decoding hormonal signalling, but their possible roles in mediating serotonin responses await investigation. In this report, we unveiled positive roles for the MITOGEN-ACTIVATED PROTEIN KINASE PHOSPHATASE1 (MKP1) in the inhibition of the primary root growth, cell division, meristem structure, and differentiation events in Arabidopsis seedlings. mkp1 mutants were less sensitive to jasmonic acid applications that halted primary root growth in wild-type (WT) plants, and consistently, the neurotransmitter activated the expression of the JASMONATE ZIM-domain (JAZ) proteins JAZ1 and JAZ10, two critical proteins orchestrating jasmonic acid signalling. This effect correlated with exacerbated production of endogenous reactive oxygen species (ROS) in the WT, a process constitutively manifested in mkp1 mutants. These data help to clarify the relationship between serotonin and growth/defense trade-offs, and reveal the importance of the MAPK pathway in root development through ROS production.


Subject(s)
Arabidopsis Proteins , Arabidopsis , Arabidopsis/metabolism , Arabidopsis Proteins/genetics , Arabidopsis Proteins/metabolism , Cyclopentanes , Gene Expression Regulation, Plant , Mitogen-Activated Protein Kinases/metabolism , Oxylipins , Protein Tyrosine Phosphatases/genetics , Protein Tyrosine Phosphatases/metabolism , Protein Tyrosine Phosphatases/pharmacology , Reactive Oxygen Species/metabolism , Serotonin/metabolism , Serotonin/pharmacology
12.
J Neurosci ; 30(31): 10282-93, 2010 Aug 04.
Article in English | MEDLINE | ID: mdl-20685973

ABSTRACT

Prostatic acid phosphatase (PAP) is expressed in nociceptive dorsal root ganglion (DRG) neurons, functions as an ectonucleotidase, and generates adenosine extracellularly. Here, we found that PAP inhibits noxious thermal sensitivity and sensitization that is associated with chronic pain through sustained activation of the adenosine A(1) receptor (A(1)R) and phospholipase C-mediated depletion of phosphatidylinositol 4,5-bisphosphate (PIP(2)). In mice, intrathecal injection of PAP reduced PIP(2) levels in DRGs, inhibited thermosensation through TRPV1, and enduringly reduced thermal hyperalgesia and mechanical allodynia caused by inflammation, nerve injury, and pronociceptive receptor activation. This included inhibitory effects on lysophosphatidic acid, purinergic (ATP), bradykinin, and protease-activated (thrombin) receptors. Conversely, PIP(2) levels were significantly elevated in DRGs from Pap(-/-) mice, and this correlated with enhanced thermal hyperalgesia and mechanical allodynia in Pap(-/-) mice. To directly test the importance of PIP(2) in nociception, we intrathecally injected PIP(2) into mice. This transiently (2 h) elevated PIP(2) levels in lumbar DRGs and transiently (2 h) enhanced thermosensation. Additionally, thermal hyperalgesia and mechanical allodynia were enduringly enhanced when PIP(2) levels were elevated coincident with injury/pronociceptive receptor stimulation. Nociceptive sensitization was not affected if PIP(2) levels were elevated in the absence of ongoing pronociceptive receptor stimulation. Together, our data suggest that PIP(2) levels in DRGs directly influence thermosensation and the magnitude of nociceptive sensitization. Moreover, our data suggest there is an underlying "phosphoinositide tone" that can be manipulated by an adenosine-generating ectonucleotidase. This tone regulates how effectively acute nociceptive insults promote the transition to chronic pain.


Subject(s)
Ganglia, Spinal/drug effects , Hyperalgesia/metabolism , Nociceptors/drug effects , Pain Threshold/drug effects , Phosphatidylinositol 4,5-Diphosphate/metabolism , Protein Tyrosine Phosphatases/pharmacology , Acid Phosphatase , Animals , Calcium/metabolism , Cell Line , Electrophysiology , Ganglia, Spinal/metabolism , Hot Temperature , Humans , Male , Mice , Mice, Transgenic , Nociceptors/metabolism , Pain Measurement , Pain Threshold/physiology , Receptor, Adenosine A1/metabolism , TRPV Cation Channels/genetics , TRPV Cation Channels/metabolism , Type C Phospholipases/metabolism
13.
Nat Cell Biol ; 6(1): 38-44, 2004 Jan.
Article in English | MEDLINE | ID: mdl-14688794

ABSTRACT

Fibroblast growth factors (FGFs) signal through high-affinity tyrosine kinase receptors to regulate a diverse range of cellular processes, including cell growth, differentiation and migration, as well as cell death. Here we identify XFLRT3, a member of a leucine-rich-repeat transmembrane protein family, as a novel modulator of FGF signalling. XFLRT3 is co-expressed with FGFs, and its expression is both induced after activation and downregulated after inhibition of FGF signalling. In gain- and loss-of function experiments, FLRT3 and FLRT2 phenocopy FGF signalling in Xenopus laevis. XFLRT3 signalling results in phosphorylation of ERK and is blocked by MAPK phosphatase 1, but not by expression of a dominant-negative phosphatidyl inositol 3-OH kinase (PI(3)K) mutant. XFLRT3 interacts with FGF receptors (FGFRs) in co-immunoprecipitation experiments in vitro and in bioluminescence resonance energy transfer assays in vivo. The results indicate that XFLRT3 is a transmembrane modulator of FGF-MAP kinase signalling in vertebrates.


Subject(s)
Fibroblast Growth Factors/metabolism , Membrane Proteins/metabolism , Receptors, Fibroblast Growth Factor/metabolism , Xenopus Proteins/metabolism , Xenopus laevis/embryology , Animals , Base Sequence/genetics , Cell Differentiation/genetics , Cell Membrane/metabolism , Cells, Cultured , DNA, Complementary/analysis , DNA, Complementary/genetics , Down-Regulation/genetics , Dual Specificity Phosphatase 1 , Embryo, Nonmammalian/cytology , Embryo, Nonmammalian/embryology , Embryo, Nonmammalian/metabolism , Embryonic Induction/genetics , Feedback, Physiological/genetics , Humans , MAP Kinase Signaling System/physiology , Membrane Proteins/genetics , Membrane Proteins/isolation & purification , Mitogen-Activated Protein Kinases/metabolism , Molecular Sequence Data , Phosphatidylinositol 3-Kinases/genetics , Phosphatidylinositol 3-Kinases/metabolism , Protein Phosphatase 1 , Protein Tyrosine Phosphatases/metabolism , Protein Tyrosine Phosphatases/pharmacology , Xenopus Proteins/genetics , Xenopus Proteins/isolation & purification , Xenopus laevis/metabolism
14.
Commun Biol ; 4(1): 923, 2021 07 29.
Article in English | MEDLINE | ID: mdl-34326464

ABSTRACT

PRL3, a unique oncotarget, is specifically overexpressed in 80.6% of cancers. In 2003, we reported that PRL3 promotes cell migration, invasion, and metastasis. Herein, firstly, we show that PRL3 induces Polyploid Giant Cancer Cells (PGCCs) formation. PGCCs constitute stem cell-like pools to facilitate cell survival, chemo-resistance, and tumor relapse. The correlations between PRL3 overexpression and PGCCs attributes raised possibilities that PRL3 could be involved in PGCCs formation. Secondly, we show that PRL3+ PGCCs co-express the embryonic stem cell markers SOX2 and OCT4 and arise mainly due to incomplete cytokinesis despite extensive DNA damage. Thirdly, we reveal that PRL3+ PGCCs tolerate prolonged chemotherapy-induced genotoxic stress via suppression of the pro-apoptotic ATM DNA damage-signaling pathway. Fourthly, we demonstrated PRL3-zumab, a First-in-Class humanized antibody drug against PRL3 oncotarget, could reduce tumor relapse in 'tumor removal' animal model. Finally, we confirmed that PGCCs were enriched in relapse tumors versus primary tumors. PRL3-zumab has been approved for Phase 2 clinical trials in Singapore, US, and China to block all solid tumors. This study further showed PRL3-zumab could potentially serve an 'Adjuvant Immunotherapy' after tumor removal surgery to eliminate PRL3+ PGCC stem-like cells, preventing metastasis and relapse.


Subject(s)
Giant Cells/pathology , Immediate-Early Proteins/genetics , Neoplasms/prevention & control , Polyploidy , Protein Tyrosine Phosphatases/genetics , Secondary Prevention/methods , Animals , Antineoplastic Agents/pharmacology , Immediate-Early Proteins/pharmacology , Mice , Neoplasms/pathology , Protein Tyrosine Phosphatases/pharmacology
15.
Mol Biochem Parasitol ; 242: 111360, 2021 03.
Article in English | MEDLINE | ID: mdl-33428948

ABSTRACT

The Entamoeba histolytica parasite is the causative agent of amebiasis, infecting approximately 1% of the world population and causing 100,000 deaths per year. It binds to Fibronectin (FN), activating signaling pathways regulated by kinases and phosphatases. EhLMW-PTPs genes from E. histolytica encode for Low Molecular Weight Tyrosine Phosphatases expressed in trophozoites and amoebic cysts. The role of these phosphatases in the virulence of the parasite has not yet been well characterized. Our results showed a differential expression of the EhLMW-PTPs, at the mRNA and protein levels, in an asynchronous trophozoites culture. Furthermore, we observed that trophozoites transfected that overexpressed EhLMW-PTP2 phagocytized fewer erythrocytes, possibly due to decreased phagocytic cups, and showed deficiencies in adherence to FN and less cytopathic effect. These analyzes suggest that the parasite's EhLMW-PTPs have an essential role in the mechanisms of proliferation, adhesion, and phagocytosis, regulating its pathogenicity.


Subject(s)
Entamoeba histolytica/pathogenicity , Protein Tyrosine Phosphatases/genetics , Protozoan Proteins/genetics , Trophozoites/pathogenicity , Virulence Factors/genetics , Animals , Caco-2 Cells , Cell Adhesion , Cell Proliferation , Cloning, Molecular , Coculture Techniques , Entamoeba histolytica/enzymology , Entamoeba histolytica/genetics , Erythrocytes/cytology , Escherichia coli/genetics , Escherichia coli/metabolism , Female , Fibronectins/chemistry , Fibronectins/metabolism , Gene Expression , Genetic Vectors/chemistry , Genetic Vectors/metabolism , Humans , Mice , Mice, Inbred BALB C , Molecular Weight , Phagocytosis/physiology , Protein Tyrosine Phosphatases/metabolism , Protein Tyrosine Phosphatases/pharmacology , Protozoan Proteins/metabolism , Protozoan Proteins/pharmacology , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Recombinant Proteins/pharmacology , Trophozoites/enzymology , Trophozoites/genetics , Virulence , Virulence Factors/metabolism , Virulence Factors/pharmacology
16.
J Exp Med ; 190(9): 1343-50, 1999 Nov 01.
Article in English | MEDLINE | ID: mdl-10544205

ABSTRACT

The acquired immune responses are crucial to the survival of Yersinia-infected animals. Mice lacking T cells are sensitive to Yersinia infection, and a humoral response to Yersinia can be protective. Diverse mechanisms for Yersinia to impair and evade the host innate immune defense have been suggested, but the effects of Yersinia on lymphocytes are not known. Here, we demonstrate that after a transient exposure to Y. pseudotuberculosis, T and B cells are impaired in their ability to be activated through their antigen receptors. T cells are inhibited in their ability to produce cytokines, and B cells are unable to upregulate surface expression of the costimulatory molecule, B7.2, in response to antigenic stimulation. The block of lymphocyte activation results from the inhibition of early phosphorylation events of the antigen receptor signaling complex. Through the use of Y. pseudotuberculosis mutants, we show that the inhibitory effect in both T cells and B cells is dependent on the production of Yersinia outermembrane protein (Yop) H, a tyrosine phosphatase. Our results suggest a mechanism by which the pathogenic bacteria may modulate a wide range of T and B cell-mediated immune responses.


Subject(s)
B-Lymphocytes/microbiology , Bacterial Outer Membrane Proteins/pharmacology , Lymphocyte Activation/immunology , Protein Tyrosine Phosphatases/pharmacology , T-Lymphocytes/microbiology , Yersinia pseudotuberculosis Infections/immunology , Animals , Antigens, CD/immunology , B7-2 Antigen , Calcium/metabolism , Cell Line , Interleukin-2/immunology , Interleukin-2/metabolism , Ionomycin/pharmacology , Membrane Glycoproteins/immunology , Mice , Mice, Transgenic , Phosphorylation , Phosphotyrosine/analysis , Receptors, Antigen, T-Cell/metabolism , Signal Transduction , Tetradecanoylphorbol Acetate/pharmacology , Up-Regulation
17.
J Gene Med ; 12(2): 137-46, 2010 Feb.
Article in English | MEDLINE | ID: mdl-20052742

ABSTRACT

BACKGROUND: An improvement of retroviral infection has been postulated using a naturally occurring fragment of the abundant semen marker prostatic acidic phosphatase. This peptide, termed semen-derived enhancer of virus infection (SEVI), promotes HIV attachment to the target cells. METHODS: In the present study, we examined whether SEVI would also enhance the infectivity of other viruses with different envelope proteins. We focused on retroviruses pseudotyped with envelopes that are commonly used for the genetic modification of cells, in particular, T cells and hematopoietic progenitor cells. Because the effect of SEVI is considered to be a result of its cationic properties, we compared SEVI with other cationic agents such as protamine sulfate and Polybrene. RESULTS: We found that SEVI increases the efficiency of gene transfer for lentiviral and gammaretroviral vector constructs pseudotyped with VSV-G, GALV, RD114 or foamy viral envelopes on hematopoietic and nonhematopoietic cell lines. On T cells, the transduction efficiency of GALV and RD114 pseudotyped vectors was significantly increased by SEVI. A significant increase of the gene transfer rate was also detected for foamy virally pseudotyped lentivirus on murine hematopoietic progenitor cells. No toxic effect of SEVI treatment was detected on any cell type tested, including human and murine hematopoietic stem/progenitor cells. When directly comparing the effect of SEVI with Polybrene or protamine sulfate, we show that the semen-derived protein is more efficient in increasing the gene transfer rate. CONCLUSIONS: SEVI is a promising agent for promoting and improving gene transfer and may also be useful for clinical gene therapy studies.


Subject(s)
Gene Transfer Techniques , Peptide Fragments/pharmacology , Protein Tyrosine Phosphatases/pharmacology , Retroviridae/genetics , Acid Phosphatase , Animals , Antigens, CD34/metabolism , Cations , Cell Adhesion/drug effects , Cell Death/drug effects , Flow Cytometry , HeLa Cells , Hematopoietic System/cytology , Hematopoietic System/drug effects , Humans , Male , Mice , NIH 3T3 Cells , Peptide Fragments/chemistry , Protein Tyrosine Phosphatases/chemistry , Retroviridae/drug effects , Transduction, Genetic , Viral Envelope Proteins/metabolism
18.
J Cell Biol ; 144(6): 1323-36, 1999 Mar 22.
Article in English | MEDLINE | ID: mdl-10087273

ABSTRACT

Cell adhesion is critical to the establishment of proper connections in the nervous system. Some receptor-type protein tyrosine phosphatases (RPTPs) have adhesion molecule-like extracellular segments with intracellular tyrosine phosphatase domains that may transduce signals in response to adhesion. PTPmu is a RPTP that mediates cell aggregation and is expressed at high levels in the nervous system. In this study, we demonstrate that PTPmu promotes neurite outgrowth of retinal ganglion cells when used as a culture substrate. In addition, PTPmu was found in a complex with N-cadherin in retinal cells. To determine the physiological significance of the association between PTPmu and N-cadherin, the expression level and enzymatic activity of PTPmu were perturbed in retinal explant cultures. Downregulation of PTPmu expression through antisense techniques resulted in a significant decrease in neurite outgrowth on an N-cadherin substrate, whereas there was no effect on laminin or L1-dependent neurite outgrowth. The overexpression of a catalytically inactive form of PTPmu significantly decreased neurite outgrowth on N-cadherin. These data indicate that PTPmu specifically regulates signals required for neurites to extend on an N-cadherin substrate, implicating reversible tyrosine phosphorylation in the control of N-cadherin function. Together, these results suggest that PTPmu plays a dual role in the regulation of neurite outgrowth.


Subject(s)
Cadherins/physiology , Neurites/physiology , Protein Tyrosine Phosphatases/physiology , Animals , Axons/physiology , Chick Embryo , Culture Techniques , Gene Expression , Neurites/drug effects , Neurites/ultrastructure , Phosphorylation , Protein Tyrosine Phosphatases/genetics , Protein Tyrosine Phosphatases/pharmacology , Rats , Receptor-Like Protein Tyrosine Phosphatases, Class 2 , Receptor-Like Protein Tyrosine Phosphatases, Class 8 , Retina/embryology , Retina/physiology , Retinal Ganglion Cells/drug effects , Retinal Ganglion Cells/physiology , Retinal Ganglion Cells/ultrastructure , Tyrosine/physiology
19.
J Cell Biol ; 137(2): 433-43, 1997 Apr 21.
Article in English | MEDLINE | ID: mdl-9128253

ABSTRACT

The spindle assembly checkpoint prevents cells whose spindles are defective or chromosomes are misaligned from initiating anaphase and leaving mitosis. Studies of Xenopus egg extracts have implicated the Erk2 mitogen-activated protein kinase (MAP kinase) in this checkpoint. Other studies have suggested that MAP kinases might be important for normal mitotic progression. Here we have investigated whether MAP kinase function is required for mitotic progression or the spindle assembly checkpoint in vivo in Xenopus tadpole cells (XTC). We determined that Erk1 and/or Erk2 are present in the mitotic spindle during prometaphase and metaphase, consistent with the idea that MAP kinase might regulate or monitor the status of the spindle. Next, we microinjected purified recombinant XCL100, a Xenopus MAP kinase phosphatase, into XTC cells in various stages of mitosis to interfere with MAP kinase activation. We found that mitotic progression was unaffected by the phosphatase. However, XCL100 rendered the cells unable to remain arrested in mitosis after treatment with nocodazole. Cells injected with phosphatase at prometaphase or metaphase exited mitosis in the presence of nocodazole-the chromosomes decondensed and the nuclear envelope re-formed-whereas cells injected with buffer or a catalytically inactive XCL100 mutant protein remained arrested in mitosis. Coinjection of constitutively active MAP kinase kinase-1, which opposes XCL100's effects on MAP kinase, antagonized the effects of XCL100. Since the only known targets of MAP kinase kinase-1 are Erk1 and Erk2, these findings argue that MAP kinase function is required for the spindle assembly checkpoint in XTC cells.


Subject(s)
Calcium-Calmodulin-Dependent Protein Kinases/physiology , Mitogen-Activated Protein Kinase Kinases , Mitosis/physiology , Spindle Apparatus/chemistry , Xenopus Proteins , Animals , Calcium-Calmodulin-Dependent Protein Kinases/analysis , Calcium-Calmodulin-Dependent Protein Kinases/metabolism , Cell Line , Enzyme Activation , Immediate-Early Proteins/pharmacology , MAP Kinase Kinase 1 , Metaphase , Microinjections , Microtubules/drug effects , Mitogen-Activated Protein Kinase Phosphatases , Mitosis/drug effects , Nocodazole/pharmacology , Protein Serine-Threonine Kinases/pharmacology , Protein Tyrosine Phosphatases/pharmacology , Protein-Tyrosine Kinases/pharmacology , Recombinant Fusion Proteins , Xenopus
20.
J Cell Biol ; 154(6): 1259-73, 2001 Sep 17.
Article in English | MEDLINE | ID: mdl-11564762

ABSTRACT

The structurally related cell adhesion molecules L1 and Nr-CAM have overlapping expression patterns in cerebellar granule cells. Here we analyzed their involvement in granule cell development using mutant mice. Nr-CAM-deficient cerebellar granule cells failed to extend neurites in vitro on contactin, a known ligand for Nr-CAM expressed in the cerebellum, confirming that these mice are functionally null for Nr-CAM. In vivo, Nr-CAM-null cerebella did not exhibit obvious histological defects, although a mild size reduction of several lobes was observed, most notably lobes IV and V in the vermis. Mice deficient for both L1 and Nr-CAM exhibited severe cerebellar folial defects and a reduction in the thickness of the inner granule cell layer. Additionally, anti-L1 antibodies specifically disrupted survival and maintenance of Nr-CAM-deficient granule cells in cerebellar cultures treated with antibodies. The combined results indicate that Nr-CAM and L1 play a role in cerebellar granule cell development, and suggest that closely related molecules in the L1 family have overlapping functions.


Subject(s)
Cell Adhesion Molecules/pharmacology , Cerebellar Cortex/drug effects , Membrane Glycoproteins/pharmacology , Neural Cell Adhesion Molecules/pharmacology , Animals , Brain/abnormalities , Brain/drug effects , Brain/pathology , Cell Adhesion Molecules/genetics , Cell Adhesion Molecules/physiology , Cell Adhesion Molecules, Neuronal/pharmacology , Cerebellar Cortex/cytology , Cerebellar Cortex/growth & development , Contactins , Female , Leukocyte L1 Antigen Complex , Male , Membrane Glycoproteins/physiology , Mice , Mice, Knockout , Nerve Tissue Proteins/pharmacology , Neural Cell Adhesion Molecules/physiology , Neurites/drug effects , Neurites/ultrastructure , Protein Tyrosine Phosphatases/pharmacology , Purkinje Cells/metabolism , Receptor-Like Protein Tyrosine Phosphatases, Class 5
SELECTION OF CITATIONS
SEARCH DETAIL