Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 44
Filter
1.
Int J Mol Sci ; 22(9)2021 Apr 23.
Article in English | MEDLINE | ID: mdl-33922601

ABSTRACT

Many human diseases are the result of abnormal expression or activation of protein tyrosine kinases (PTKs) and protein tyrosine phosphatases (PTPs). Not surprisingly, more than 30 tyrosine kinase inhibitors (TKIs) are currently in clinical use and provide unique treatment options for many patients. PTPs on the other hand have long been regarded as "undruggable" and only recently have gained increased attention in drug discovery. Striatal-enriched tyrosine phosphatase (STEP) is a neuron-specific PTP that is overactive in Alzheimer's disease (AD) and other neurodegenerative and neuropsychiatric disorders, including Parkinson's disease, schizophrenia, and fragile X syndrome. An emergent model suggests that the increase in STEP activity interferes with synaptic function and contributes to the characteristic cognitive and behavioral deficits present in these diseases. Prior efforts to generate STEP inhibitors with properties that warrant clinical development have largely failed. To identify novel STEP inhibitor scaffolds, we developed a biophysical, label-free high-throughput screening (HTS) platform based on the protein thermal shift (PTS) technology. In contrast to conventional HTS using STEP enzymatic assays, we found the PTS platform highly robust and capable of identifying true hits with confirmed STEP inhibitory activity and selectivity. This new platform promises to greatly advance STEP drug discovery and should be applicable to other PTP targets.


Subject(s)
Drug Discovery , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/pharmacology , High-Throughput Screening Assays/methods , Protein Tyrosine Phosphatases, Non-Receptor/antagonists & inhibitors , Protein Tyrosine Phosphatases, Non-Receptor/metabolism , Humans , Molecular Structure
2.
Molecules ; 26(6)2021 Mar 14.
Article in English | MEDLINE | ID: mdl-33799458

ABSTRACT

Inhibition of the megakaryocyte protein tyrosine phosphatase 2 (PTP-MEG2, also named PTPN9) activity has been shown to be a potential therapeutic strategy for the treatment of type 2 diabetes. Previously, we reported that PTP-MEG2 knockdown enhances adenosine monophosphate activated protein kinase (AMPK) phosphorylation, suggesting that PTP-MEG2 may be a potential antidiabetic target. In this study, we found that phloridzin, isolated from Ulmus davidiana var. japonica, inhibits the catalytic activity of PTP-MEG2 (half-inhibitory concentration, IC50 = 32 ± 1.06 µM) in vitro, indicating that it could be a potential antidiabetic drug candidate. Importantly, phloridzin stimulated glucose uptake by differentiated 3T3-L1 adipocytes and C2C12 muscle cells compared to that by the control cells. Moreover, phloridzin led to the enhanced phosphorylation of AMPK and Akt relevant to increased insulin sensitivity. Importantly, phloridzin attenuated palmitate-induced insulin resistance in C2C12 muscle cells. We also found that phloridzin did not accelerate adipocyte differentiation, suggesting that phloridzin improves insulin sensitivity without significant lipid accumulation. Taken together, our results demonstrate that phloridzin, an inhibitor of PTP-MEG2, stimulates glucose uptake through the activation of both AMPK and Akt signaling pathways. These results strongly suggest that phloridzin could be used as a potential therapeutic candidate for the treatment of type 2 diabetes.


Subject(s)
Insulin Resistance/physiology , Phlorhizin/pharmacology , Protein Tyrosine Phosphatases, Non-Receptor/antagonists & inhibitors , Protein Tyrosine Phosphatases/antagonists & inhibitors , 3T3 Cells , Adipocytes/drug effects , Adipocytes/metabolism , Animals , Cell Differentiation/drug effects , Cell Line , Diabetes Mellitus, Type 2/drug therapy , Diabetes Mellitus, Type 2/metabolism , Glucose/metabolism , Hypoglycemic Agents/pharmacology , Insulin/metabolism , Mice , Palmitates/pharmacology , Phosphorylation/drug effects , Signal Transduction/drug effects
3.
J Biol Chem ; 294(45): 16684-16697, 2019 11 08.
Article in English | MEDLINE | ID: mdl-31543504

ABSTRACT

Macrophage phagocytosis is required for effective clearance of invading bacteria and other microbes. Coordinated phosphoinositide signaling is critical both for phagocytic particle engulfment and subsequent phagosomal maturation to a degradative organelle. Phosphatidylinositol 3-phosphate (PtdIns(3)P) is a phosphoinositide that is rapidly synthesized and degraded on phagosomal membranes, where it recruits FYVE domain- and PX motif-containing proteins that promote phagosomal maturation. However, the molecular mechanisms that regulate PtdIns(3)P removal from the phagosome have remained unclear. We report here that a myotubularin PtdIns(3)P 3-phosphatase, myotubularin-related protein-4 (MTMR4), regulates macrophage phagocytosis. MTMR4 overexpression reduced and siRNA-mediated Mtmr4 silencing increased levels of cell-surface immunoglobulin receptors (i.e. Fcγ receptors (FcγRs)) on RAW 264.7 macrophages, associated with altered pseudopodal F-actin. Furthermore, MTMR4 negatively regulated the phagocytosis of IgG-opsonized particles, indicating that MTMR4 inhibits FcγR-mediated phagocytosis, and was dynamically recruited to phagosomes of macrophages during phagocytosis. MTMR4 overexpression decreased and Mtmr4-specific siRNA expression increased the duration of PtdIns(3)P on phagosomal membranes. Macrophages treated with Mtmr4-specific siRNA were more resistant to Mycobacterium marinum-induced phagosome arrest, associated with increased maturation of mycobacterial phagosomes, indicating that extended PtdIns(3)P signaling on phagosomes in the Mtmr4-knockdown cells permitted trafficking of phagosomes to acidic late endosomal and lysosomal compartments. In conclusion, our findings indicate that MTMR4 regulates PtdIns(3)P degradation in macrophages and thereby controls phagocytosis and phagosomal maturation.


Subject(s)
Phagocytosis , Phagosomes/metabolism , Phosphatidylinositol Phosphates/metabolism , Protein Tyrosine Phosphatases, Non-Receptor/metabolism , Actins/metabolism , Animals , Endosomes/metabolism , Humans , Immunoglobulin G/immunology , Lysosomes/metabolism , Macrophages/cytology , Macrophages/metabolism , Mice , Mycobacterium marinum/pathogenicity , Protein Tyrosine Phosphatases, Non-Receptor/antagonists & inhibitors , Protein Tyrosine Phosphatases, Non-Receptor/genetics , RAW 264.7 Cells , RNA Interference , RNA, Small Interfering/metabolism , Receptors, IgG/metabolism , Signal Transduction
4.
Int J Mol Sci ; 21(15)2020 Jul 22.
Article in English | MEDLINE | ID: mdl-32707818

ABSTRACT

Tyrosine phosphatase STEP (striatal-enriched tyrosine protein phosphatase) is a brain-specific protein phosphatase and is involved in the pathogenesis of many neurodegenerative diseases. Here, we examined the impact of STEP on the development of age-related macular degeneration (AMD)-like pathology in senescence-accelerated OXYS rats. Using OXYS and Wistar rats (control), we for the first time demonstrated age-dependent changes in Ptpn5 mRNA expression, STEP46 and STEP61 protein levels, and their phosphatase activity in the retina. The increases in STEP protein levels and the decrease of total and STEP phosphatase activities in the retina (as compared with Wistar rats) preceded the manifestation of clinical signs of AMD in OXYS rats (age 20 days). There were no differences in these retinal parameters between 13-month-old Wistar rats and OXYS rats with pronounced signs of AMD. Inhibition of STEP with TC-2153 during progressive AMD-like retinopathy (from 9 to 13 months of age) reduced the thickness of the retinal inner nuclear layer, as evidenced by a decreased amount of parvalbumin-positive amacrine neurons. Prolonged treatment with TC-2153 had no effect on Ptpn5 mRNA expression, STEP46 and STEP61 protein levels, and their phosphatase activity in the OXYS retina. Thus, TC-2153 may negatively affect the retina through mechanisms unrelated to STEP.


Subject(s)
Aging/metabolism , Gene Expression Regulation/genetics , Macular Degeneration/metabolism , Protein Tyrosine Phosphatases, Non-Receptor/metabolism , Retina/metabolism , Retinal Diseases/metabolism , Aging/genetics , Animals , Apoptosis/drug effects , Apoptosis/genetics , Benzothiepins/pharmacology , Brain-Derived Neurotrophic Factor/metabolism , Cellular Senescence/genetics , Gene Expression Regulation/drug effects , Macular Degeneration/pathology , Male , Nerve Growth Factor/metabolism , Neuroglia/drug effects , Neuroglia/metabolism , Neurons/drug effects , Neurons/metabolism , Protein Tyrosine Phosphatases, Non-Receptor/antagonists & inhibitors , Protein Tyrosine Phosphatases, Non-Receptor/genetics , Rats , Rats, Wistar , Retinal Diseases/enzymology , Retinal Diseases/genetics
5.
Neurochem Res ; 44(12): 2832-2842, 2019 Dec.
Article in English | MEDLINE | ID: mdl-31691882

ABSTRACT

Sepsis-associated encephalopathy (SAE) is a potentially irreversible acute cognitive dysfunction with unclear mechanism. Striatal-enriched protein tyrosine phosphatase (STEP) is a brain-specific phosphatase which normally opposes synaptic strengthening by regulating key signaling molecules involved in synaptic plasticity and neuronal function. Thus, we hypothesized that abnormal STEP signaling pathway was involved in sepsis-induced cognitive impairment evoked by lipopolysaccharides (LPS) injection. The levels of STEP, phosphorylation of GluN2B (pGluN2B), the kinases extracellular signal-regulated kinase 1/2 (pERK), cAMP-response element binding protein (CREB), synaptophysin, brain derived neurotrophic factor (BDNF), and post-synaptic density protein 95 (PSD95) in the hippocampus, prefrontal cortex, and striatum were determined at the indicated time points. In the present study, we found that STEP levels were significantly increased in the hippocampus, prefrontal cortex, and striatum following LPS injection, which might resulted from the disruption of the ubiquitin-proteasome system. Notably, a STEP inhibitor TC-2153 treatment alleviated sepsis-induced memory impairment by increasing phosphorylation of GluN2B and ERK1/2, CREB/BDNF, and PSD95. In summary, our results support the key role of STEP in sepsis-induced memory impairment in a mouse model of SAE, whereas inhibition of STEP may provide a novel therapeutic approach for this disorder and possible other neurodegenerative diseases.


Subject(s)
Memory Disorders/physiopathology , Protein Tyrosine Phosphatases, Non-Receptor/metabolism , Sepsis-Associated Encephalopathy/physiopathology , Signal Transduction/physiology , Animals , Benzothiepins/pharmacology , Brain-Derived Neurotrophic Factor/chemistry , Brain-Derived Neurotrophic Factor/metabolism , Corpus Striatum/metabolism , Cyclic AMP Response Element-Binding Protein/chemistry , Cyclic AMP Response Element-Binding Protein/metabolism , Disks Large Homolog 4 Protein/chemistry , Disks Large Homolog 4 Protein/metabolism , Hippocampus/metabolism , Lipopolysaccharides , Male , Memory/drug effects , Memory/physiology , Memory Disorders/chemically induced , Mice, Inbred C57BL , Mitogen-Activated Protein Kinase 1/chemistry , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/chemistry , Mitogen-Activated Protein Kinase 3/metabolism , Phosphorylation/drug effects , Prefrontal Cortex/metabolism , Protein Tyrosine Phosphatases, Non-Receptor/antagonists & inhibitors , Receptors, N-Methyl-D-Aspartate/chemistry , Receptors, N-Methyl-D-Aspartate/metabolism , Sepsis-Associated Encephalopathy/chemically induced , Signal Transduction/drug effects
6.
Bioorg Chem ; 90: 103087, 2019 09.
Article in English | MEDLINE | ID: mdl-31284101

ABSTRACT

Natural products as antidiabetic agents have been shown to stimulate insulin signaling via the inhibition of the protein tyrosine phosphatases relevant to insulin resistance. Previously, we have identified PTPN9 and DUSP9 as potential antidiabetic targets and a multi-targeting natural product thereof. In this study, knockdown of PTPN11 increased AMPK phosphorylation in differentiated C2C12 muscle cells by 3.8 fold, indicating that PTPN11 could be an antidiabetic target. Screening of a library of 658 natural products against PTPN9, DUSP9, or PTPN11 identified chebulinic acid (CA) as a strong allosteric inhibitor with a slow cooperative binding to PTPN9 (IC50 = 34 nM) and PTPN11 (IC50 = 37 nM), suggesting that it would be a potential antidiabetic candidate. Furthermore, CA stimulated glucose uptake and resulted in increased AMP-activated protein kinase (AMPK) phosphorylation. Taken together, we demonstrated that CA increased glucose uptake as a dual inhibitor of PTPN9 and PTPN11 through activation of the AMPK signaling pathway. These results strongly suggest that CA could be used as a potential therapeutic candidate for the treatment of type 2 diabetes.


Subject(s)
Diabetes Mellitus, Type 2/drug therapy , Hydrolyzable Tannins/pharmacology , Hypoglycemic Agents/pharmacology , Protein Tyrosine Phosphatase, Non-Receptor Type 11/antagonists & inhibitors , Protein Tyrosine Phosphatases, Non-Receptor/antagonists & inhibitors , Glucose/metabolism , Humans , Insulin/metabolism , Insulin Resistance , Phosphorylation , Signal Transduction
7.
Exp Cell Res ; 371(1): 287-296, 2018 10 01.
Article in English | MEDLINE | ID: mdl-30149002

ABSTRACT

The use of imatinib mesylate has greatly improved the clinical outcome for gastrointestinal stromal tumor (GIST) patients. However, imatinib resistance is still a major clinical challenge, and the molecular mechanisms are not fully understood. We have previously shown that miR-125a-5p and its mRNA target PTPN18 modulate imatinib response in GIST cells. Herein, we evaluated phosphorylated FAK (pFAK) as a candidate downstream target of PTPN18 and the possible association of this regulation with imatinib resistance in GIST. FAK and pFAK expressions were evaluated in GIST882 cells transfected with short hairpin RNA or short interfering RNA targeting PTPN18 or miR-125a-5p mimic, imatinib-resistant GIST882R subclones and clinical samples using Western blot analyses. FAK phosphorylation was blocked using the FAK inhibitor 14 (FAKi) and the effects on cell viability and apoptosis were evaluated using WST-1 assay and cleaved PARP expression. Clinical associations of FAK and pFAK expression with imatinib resistance, KIT mutation and patient outcome were assessed by Fisher's exact test or log-rank test. Over-expression of miR-125a-5p and silencing of PTPN18 increased pFAK, but not FAK, expression in GIST cells. Higher pFAK expression was observed in the GIST882R subclones with acquired imatinib resistance compared to their imatinib-sensitive parental cells. Treatment with FAKi in imatinib-resistant GIST882R cells reduced cell viability and increased apoptosis upon imatinib treatment. Additionally, FAKi could rescue the imatinib resistance effect mediated by miR-125a-5p over-expression. In clinical samples, high FAK and pFAK expressions were associated with KIT mutation status, and high FAK expression was also associated with metastasis in GIST. Higher pFAK was found in cases with shorter overall survival. Our findings highlight an important role for miR-125a-5p regulation and its downstream target pFAK for imatinib resistance in GIST. pFAK and FAK may have prognostic values in GIST.


Subject(s)
Drug Resistance, Neoplasm/genetics , Focal Adhesion Kinase 1/genetics , Gastrointestinal Neoplasms/genetics , Gastrointestinal Stromal Tumors/genetics , Gene Expression Regulation, Neoplastic , Imatinib Mesylate/pharmacology , MicroRNAs/genetics , Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Drug Resistance, Neoplasm/drug effects , Focal Adhesion Kinase 1/metabolism , Gastrointestinal Neoplasms/diagnosis , Gastrointestinal Neoplasms/drug therapy , Gastrointestinal Neoplasms/mortality , Gastrointestinal Stromal Tumors/diagnosis , Gastrointestinal Stromal Tumors/drug therapy , Gastrointestinal Stromal Tumors/mortality , Humans , MicroRNAs/metabolism , Oligoribonucleotides/genetics , Oligoribonucleotides/metabolism , Phosphorylation , Prognosis , Protein Kinase Inhibitors/pharmacology , Protein Tyrosine Phosphatases, Non-Receptor/antagonists & inhibitors , Protein Tyrosine Phosphatases, Non-Receptor/genetics , Protein Tyrosine Phosphatases, Non-Receptor/metabolism , RNA, Messenger/antagonists & inhibitors , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , STAT3 Transcription Factor/genetics , STAT3 Transcription Factor/metabolism , Signal Transduction , Survival Analysis
8.
Bioorg Med Chem ; 26(9): 2610-2620, 2018 05 15.
Article in English | MEDLINE | ID: mdl-29673715

ABSTRACT

Protein tyrosine phosphatases (PTPs), which catalyze the dephosphorylation of phosphotyrosine in protein substrates, are important cell-signaling regulators, as well as potential drug targets for a range of human diseases. Chemical tools for selectively targeting the activities of individual PTPs would help to elucidate PTP signaling roles and potentially expedite the validation of PTPs as therapeutic targets. We have recently reported a novel strategy for the design of non-natural allosteric-inhibition sites in PTPs, in which a tricysteine moiety is engineered within the PTP catalytic domain at a conserved location outside of the active site. Introduction of the tricysteine motif, which does not exist in any wild-type PTP, serves to sensitize target PTPs to inhibition by a biarsenical compound, providing a generalizable strategy for the generation of allosterically sensitized (as) PTPs. Here we show that the potency, selectivity, and kinetics of asPTP inhibition can be significantly improved by exploring the inhibitory action of a range of biarsenical compounds that differ in interarsenical distance, steric bulk, and electronic structure. By investigating the inhibitor sensitivities of five asPTPs from four different subfamilies, we have found that asPTP catalytic domains can be broadly divided into two groups: one that is most potently inhibited by biarsenical compounds with large interarsenical distances, such as AsCy3-EDT2, and one that is most potently inhibited by compounds with relatively small interarsenical distances, such as FlAsH-EDT2. Moreover, we show that a tetrachlorinated derivative of FlAsH-EDT2, Cl4FlAsH-EDT2, targets asPTPs significantly more potently than the parent compound, both in vitro and in asPTP-expressing cells. Our results show that biarsenicals with altered interarsenical distances and electronic properties are important tools for optimizing the control of asPTP activity and, more broadly, suggest that diversification of biarsenical libraries can serve to increase the efficacy of these compounds in targeted control of protein function.


Subject(s)
Arsenicals/pharmacology , Enzyme Inhibitors/pharmacology , Protein Tyrosine Phosphatases, Non-Receptor/antagonists & inhibitors , Allosteric Site/genetics , Amino Acid Sequence , Arsenicals/chemistry , Catalytic Domain/genetics , Enzyme Inhibitors/chemistry , Escherichia coli/metabolism , Kinetics , Molecular Structure , Mutagenesis, Site-Directed , Mutation , Phosphopeptides/chemistry , Phosphopeptides/metabolism , Protein Engineering , Protein Tyrosine Phosphatases, Non-Receptor/genetics , Protein Tyrosine Phosphatases, Non-Receptor/metabolism
9.
Bioorg Chem ; 81: 264-269, 2018 12.
Article in English | MEDLINE | ID: mdl-30153591

ABSTRACT

Several protein tyrosine phosphatases (PTPs) that disrupt the insulin-signaling pathway were investigated by siRNAs to identify potential antidiabetic targets. Individual knockdown of PTPN9 and DUSP9 in 3T3-L1 preadipocytes increased AMPK phosphorylation, respectively, and furthermore, concurrent knockdown of both PTPN9 and DUSP9 synergistically increased AMPK phosphorylation. Next, 658 natural products were screened to identify dual inhibitors of both PTPN9 and DUSP9. Based on the selectivity and inhibition potency of the compounds, ginkgolic acid (GA) was selected for further study as a potential antidiabetic drug candidate. GA inhibited the enzymatic activity of PTPN9 (Ki = 53 µM) and DUSP9 (Ki = 2.5 µM) in vitro and resulted in a significant increase of glucose-uptake in differentiated C2C12 muscle cells and 3T3-L1 adipocytes. In addition, GA increased phosphorylation of AMPK in 3T3L1 adipocytes. In this study, GA as a dual targeting inhibitor of PTPN9 and DUSP9 increased glucose uptake in 3T3L1 and C2C12 cells by activating the AMPK signaling pathway. These results strongly suggest GA could be used as a therapeutic candidate for type 2 diabetes.


Subject(s)
Dual-Specificity Phosphatases/antagonists & inhibitors , Enzyme Inhibitors/pharmacology , Hypoglycemic Agents/pharmacology , Mitogen-Activated Protein Kinase Phosphatases/antagonists & inhibitors , Protein Tyrosine Phosphatases, Non-Receptor/antagonists & inhibitors , Salicylates/pharmacology , AMP-Activated Protein Kinases/metabolism , Animals , Cell Line , Dual-Specificity Phosphatases/genetics , Gene Knockdown Techniques , Glucose/metabolism , Mice , Mitogen-Activated Protein Kinase Phosphatases/genetics , Phosphorylation/drug effects , Phosphorylation/genetics , Protein Tyrosine Phosphatases, Non-Receptor/genetics
10.
Addict Biol ; 23(1): 219-229, 2018 01.
Article in English | MEDLINE | ID: mdl-28349660

ABSTRACT

Cocaine self-administration in rats results in dysfunctional neuroadaptations in the prelimbic (PrL) cortex during early abstinence. Central to these adaptations is decreased phospho-extracellular signal-regulated kinase (p-ERK), which plays a key role in cocaine seeking. Normalizing ERK phosphorylation in the PrL cortex immediately after cocaine self-administration decreases subsequent cocaine seeking. The disturbance in ERK phosphorylation is accompanied by decreased phosphorylation of striatal-enriched protein tyrosine phosphatase (STEP), indicating increased STEP activity. STEP is a well-recognized ERK phosphatase but whether STEP activation during early abstinence mediates the decrease in p-ERK and is involved in relapse is unknown. Here, we show that a single intra-PrL cortical microinfusion of the selective STEP inhibitor, TC-2153, immediately after self-administration suppressed post-abstinence context-induced relapse under extinction conditions and cue-induced reinstatement, but not cocaine prime-induced drug seeking or sucrose seeking. Moreover, an intra-PrL cortical TC-2153 microinfusion immediately after self-administration prevented the cocaine-induced decrease in p-ERK within the PrL cortex during early abstinence. Interestingly, a systemic TC-2153 injection at the same timepoint failed to suppress post-abstinence context-induced relapse or cue-induced reinstatement, but did suppress cocaine prime-induced reinstatement. These data indicate that the STEP-induced ERK dephosphorylation in the PrL cortex during early abstinence is a critical neuroadaptation that promotes relapse to cocaine seeking and that systemic versus intra-PrL cortical inhibition of STEP during early abstinence differentially suppresses cocaine seeking.


Subject(s)
Benzothiepins/pharmacology , Cocaine/administration & dosage , Dopamine Uptake Inhibitors/administration & dosage , Drug-Seeking Behavior/drug effects , Protein Tyrosine Phosphatases, Non-Receptor/antagonists & inhibitors , Animals , Extracellular Signal-Regulated MAP Kinases , Male , Phosphoproteins , Prefrontal Cortex , Rats , Rats, Sprague-Dawley , Self Administration
11.
PLoS Biol ; 12(8): e1001923, 2014 Aug.
Article in English | MEDLINE | ID: mdl-25093460

ABSTRACT

STEP (STriatal-Enriched protein tyrosine Phosphatase) is a neuron-specific phosphatase that regulates N-methyl-D-aspartate receptor (NMDAR) and α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid receptor (AMPAR) trafficking, as well as ERK1/2, p38, Fyn, and Pyk2 activity. STEP is overactive in several neuropsychiatric and neurodegenerative disorders, including Alzheimer's disease (AD). The increase in STEP activity likely disrupts synaptic function and contributes to the cognitive deficits in AD. AD mice lacking STEP have restored levels of glutamate receptors on synaptosomal membranes and improved cognitive function, results that suggest STEP as a novel therapeutic target for AD. Here we describe the first large-scale effort to identify and characterize small-molecule STEP inhibitors. We identified the benzopentathiepin 8-(trifluoromethyl)-1,2,3,4,5-benzopentathiepin-6-amine hydrochloride (known as TC-2153) as an inhibitor of STEP with an IC50 of 24.6 nM. TC-2153 represents a novel class of PTP inhibitors based upon a cyclic polysulfide pharmacophore that forms a reversible covalent bond with the catalytic cysteine in STEP. In cell-based secondary assays, TC-2153 increased tyrosine phosphorylation of STEP substrates ERK1/2, Pyk2, and GluN2B, and exhibited no toxicity in cortical cultures. Validation and specificity experiments performed in wild-type (WT) and STEP knockout (KO) cortical cells and in vivo in WT and STEP KO mice suggest specificity of inhibitors towards STEP compared to highly homologous tyrosine phosphatases. Furthermore, TC-2153 improved cognitive function in several cognitive tasks in 6- and 12-mo-old triple transgenic AD (3xTg-AD) mice, with no change in beta amyloid and phospho-tau levels.


Subject(s)
Alzheimer Disease/drug therapy , Alzheimer Disease/enzymology , Cognition Disorders/drug therapy , Cognition Disorders/enzymology , Enzyme Inhibitors/therapeutic use , Protein Tyrosine Phosphatases, Non-Receptor/antagonists & inhibitors , Alzheimer Disease/complications , Alzheimer Disease/pathology , Amino Acid Sequence , Animals , Benzothiepins/pharmacology , Benzothiepins/therapeutic use , Catalytic Domain , Cell Death/drug effects , Cerebral Cortex/pathology , Cognition Disorders/complications , Cognition Disorders/pathology , Cysteine/metabolism , Disease Models, Animal , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/pharmacology , High-Throughput Screening Assays , Humans , Male , Mice, Inbred C57BL , Mice, Knockout , Molecular Sequence Data , Neurons/drug effects , Neurons/pathology , Phosphorylation/drug effects , Phosphotyrosine/metabolism , Protein Tyrosine Phosphatases, Non-Receptor/chemistry , Protein Tyrosine Phosphatases, Non-Receptor/metabolism , Substrate Specificity/drug effects
12.
J Biol Chem ; 290(15): 9646-59, 2015 Apr 10.
Article in English | MEDLINE | ID: mdl-25681440

ABSTRACT

We used a loss-of-function screen to investigate the role of classical protein-tyrosine phosphatases (PTPs) in three-dimensional mammary epithelial cell morphogenesis and ERBB2 signaling. The study revealed a novel role for PTPD2 as a positive regulator of ERBB2 signaling. Suppression of PTPD2 attenuated the ERBB2-induced multiacinar phenotype in three-dimensional cultures specifically by inhibiting ERBB2-mediated loss of polarity and lumen filling. In contrast, overexpression of PTPD2 enhanced the ERBB2 phenotype. We also found that a lipid second messenger, phosphatidic acid, bound PTPD2 in vitro and enhanced its catalytic activity. Small molecule inhibitors of phospholipase D (PLD), an enzyme that produces phosphatidic acid in cells, also attenuated the ERBB2 phenotype. Exogenously added phosphatidic acid rescued the PLD-inhibition phenotype, but only when PTPD2 was present. These findings illustrate a novel pathway involving PTPD2 and the lipid second messenger phosphatidic acid that promotes ERBB2 function.


Subject(s)
Epithelial Cells/metabolism , Phosphatidic Acids/metabolism , Protein Tyrosine Phosphatases, Non-Receptor/metabolism , Receptor, ErbB-2/metabolism , Signal Transduction , Apoptosis/drug effects , Apoptosis/genetics , Cell Culture Techniques , Cell Line , Collagen , Drug Combinations , Epithelial Cells/drug effects , Humans , Immunoblotting , Indoles/pharmacology , Laminin , Mammary Glands, Human/cytology , Microscopy, Fluorescence , Microscopy, Phase-Contrast , Phospholipase D/antagonists & inhibitors , Phospholipase D/metabolism , Protein Binding , Protein Tyrosine Phosphatases, Non-Receptor/antagonists & inhibitors , Protein Tyrosine Phosphatases, Non-Receptor/genetics , Proteoglycans , RNA Interference , Receptor, ErbB-2/genetics , Sulfonamides/pharmacology , Tacrolimus/analogs & derivatives , Tacrolimus/pharmacology , src-Family Kinases/antagonists & inhibitors , src-Family Kinases/metabolism
13.
EMBO Rep ; 14(1): 57-64, 2013 Jan.
Article in English | MEDLINE | ID: mdl-23154468

ABSTRACT

Although phosphatidylinositol 5-phosphate (PtdIns5P) is present in many cell types and its biogenesis is increased by diverse stimuli, its precise cellular function remains elusive. Here we show that PtdIns5P levels increase when cells are stimulated to move and we find PtdIns5P to promote cell migration in tissue culture and in a Drosophila in vivo model. First, class III phosphatidylinositol 3-kinase, which produces PtdIns3P, was shown to be involved in migration of fibroblasts. In a cell migration screen for proteins containing PtdIns3P-binding motifs, we identified the phosphoinositide 5-kinase PIKfyve and the phosphoinositide 3-phosphatase MTMR3, which together constitute a phosphoinositide loop that produces PtdIns5P via PtdIns(3,5)P(2). The ability of PtdIns5P to stimulate cell migration was demonstrated directly with exogenous PtdIns5P and a PtdIns5P-producing bacterial enzyme. Thus, the identified phosphoinositide loop defines a new role for PtdIns5P in cell migration.


Subject(s)
Cell Movement/physiology , Drosophila melanogaster/metabolism , Fibroblasts/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Phosphatidylinositol Phosphates/biosynthesis , Protein Tyrosine Phosphatases, Non-Receptor/metabolism , Animals , Binding Sites , Cell Line , Class III Phosphatidylinositol 3-Kinases/genetics , Class III Phosphatidylinositol 3-Kinases/metabolism , Drosophila melanogaster/genetics , Fibroblasts/cytology , Gene Expression Regulation , Humans , Phosphatidylinositol 3-Kinases/genetics , Phosphatidylinositol Phosphates/metabolism , Protein Binding , Protein Interaction Domains and Motifs , Protein Tyrosine Phosphatases, Non-Receptor/antagonists & inhibitors , Protein Tyrosine Phosphatases, Non-Receptor/genetics , RNA, Small Interfering/genetics , Signal Transduction
14.
Biochem J ; 461(3): 383-90, 2014 Aug 01.
Article in English | MEDLINE | ID: mdl-24840251

ABSTRACT

Previously, we have shown that the phosphoinositide metabolizing enzymes PIKfyve (phosphoinositide 5-kinase, FYVE finger containing) and MTMR3 (myotubularin-related protein 3), together with their lipid product PtdIns5P, are important for migration of normal human fibroblasts. As these proteins are a kinase and a phosphatase respectively, and thereby considered druggable, we wanted to test their involvement in cancer cell migration and invasion. First, we showed that PIKfyve and MTMR3 are expressed in most cancer cells. Next, we demonstrated that depletion of PIKfyve or MTMR3 resulted in decreased velocity in three different cancer cell lines by using new software for cell tracking. Inhibition of the enzymatic activity of PIKfyve by the inhibitor YM201636 also led to a strong reduction in cell velocity. Mechanistically, we show that PIKfyve and MTMR3 regulate the activation of the Rho family GTPase Rac1. Further experiments also implicated PtdIns5P in the activation of Rac1. The results suggest a model for the activation of Rac1 in cell migration where PIKfyve and MTMR3 produce PtdIns5P on cellular membranes which may then serve to recruit effectors to activate Rac1. Finally, in an invasion assay, we demonstrate that both PIKfyve and MTMR3 are implicated in invasive behaviour of cancer cells. Thus PIKfyve and MTMR3 could represent novel therapeutic targets in metastatic cancer.


Subject(s)
Carcinoma/metabolism , Neoplasm Proteins/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Phosphatidylinositol Phosphates/metabolism , Protein Tyrosine Phosphatases, Non-Receptor/metabolism , Sarcoma/metabolism , rac1 GTP-Binding Protein/agonists , Carcinoma/drug therapy , Carcinoma/pathology , Cell Line , Cell Line, Tumor , Cell Movement/drug effects , Cell Polarity , Computational Biology , Databases, Genetic , Enzyme Activation/drug effects , Enzyme Inhibitors/pharmacology , Expert Systems , Gene Expression Regulation, Neoplastic , Humans , Neoplasm Invasiveness , Neoplasm Proteins/agonists , Neoplasm Proteins/antagonists & inhibitors , Neoplasm Proteins/genetics , Phosphatidylinositol 3-Kinases/genetics , Phosphoinositide-3 Kinase Inhibitors , Protein Tyrosine Phosphatases, Non-Receptor/antagonists & inhibitors , Protein Tyrosine Phosphatases, Non-Receptor/genetics , RNA Interference , Sarcoma/drug therapy , Sarcoma/pathology , Software , rac1 GTP-Binding Protein/metabolism
15.
Z Naturforsch C J Biosci ; 70(3-4): 93-6, 2015.
Article in English | MEDLINE | ID: mdl-26023990

ABSTRACT

A novel C25 sterol peroxide, phomasterol A (1), together with two known compounds (2-3), was isolated from the endophytic fungus Phoma sp. EA-122. The structure of phomasterol A (1) was elucidated by MS, 1D, and 2D NMR data analyses. Phomasterol A (1) was evaluated for its inhibitory activities against protein-tyrosine phosphatases MEG2 and PTP1Bc, showing moderate activities with identical IC50 values of 25 µM.


Subject(s)
Ascomycota/chemistry , Peroxides/chemistry , Sterols/chemistry , Magnetic Resonance Spectroscopy , Molecular Structure , Peroxides/isolation & purification , Protein Tyrosine Phosphatase, Non-Receptor Type 1/antagonists & inhibitors , Protein Tyrosine Phosphatases, Non-Receptor/antagonists & inhibitors , Sterols/isolation & purification
16.
Oral Dis ; 20(3): e65-75, 2014 Apr.
Article in English | MEDLINE | ID: mdl-23731011

ABSTRACT

OBJECTIVE: We aimed at studying the role of the most deregulated miR-99a, identifying its downstream targets, and exploring the clinical potential of miR-99a and its target(s) in oral cancer. SUBJECTS AND METHODS: Following confirmation of miR-99a deregulation in nine oral lines and 26 pairwise clinical specimens, miR-99a-manipulated oral cancer cells were subjected to cell proliferation, migration, invasion, and in vivo murine metastasis assays. We characterized putative miR-99a target(s) using luciferase reporter assays and genetic manipulation. The inverse relation of miR-99a and its target(s) was examined in clinical specimens using real-time PCR and Western blot analysis. RESULTS: MiR-99a down-regulation was confirmed both in tested oral cancer cell lines and clinical specimens. Ectopic miR-99a expression inhibited oral cancer cell migration and invasion. Anti-miR-99a, silencing miR-99a functions, had the opposite effect. Myotubularin-related protein 3 (MTMR3) with one evolutionarily conserved seed region in the 3'-untranslated region was a novel miR-99a target. Depleting MTMR3 expression significantly reduced cell proliferation, migration, or invasion. There was an inverse expression of miR-99a and MTMR3 protein in oral cancer lines and clinical specimens. CONCLUSION: miR-99a repressed oral cancer cell migration and invasion partly through decreasing MTMR3 expression. MTMR3 may serve as a therapeutic target for oral cancer treatment.


Subject(s)
MicroRNAs/physiology , Mouth Neoplasms/genetics , Mouth Neoplasms/pathology , Protein Tyrosine Phosphatases, Non-Receptor/antagonists & inhibitors , Protein Tyrosine Phosphatases, Non-Receptor/biosynthesis , Gene Expression Regulation, Neoplastic , Humans , Neoplasm Metastasis , Tumor Cells, Cultured
17.
J Med Chem ; 67(11): 8817-8835, 2024 Jun 13.
Article in English | MEDLINE | ID: mdl-38768084

ABSTRACT

Together with protein tyrosine kinases, protein tyrosine phosphatases (PTPs) control protein tyrosine phosphorylation and regulate numerous cellular functions. Dysregulated PTP activity is associated with the onset of multiple human diseases. Nevertheless, understanding of the physiological function and disease biology of most PTPs remains limited, largely due to the lack of PTP-specific chemical probes. In this study, starting from a well-known nonhydrolyzable phosphotyrosine (pTyr) mimetic, phosphonodifluoromethyl phenylalanine (F2Pmp), we synthesized 7 novel phosphonodifluoromethyl-containing bicyclic/tricyclic aryl derivatives with improved cell permeability and potency toward various PTPs. Furthermore, with fragment- and structure-based design strategies, we advanced compound 9 to compound 15, a first-in-class, potent, selective, and bioavailable inhibitor of human CDC14A and B phosphatases. This study demonstrates the applicability of the fragment-based design strategy in creating potent, selective, and bioavailable PTP inhibitors and provides a valuable probe for interrogating the biological roles of hCDC14 phosphatases and assessing their potential for therapeutic interventions.


Subject(s)
Enzyme Inhibitors , Phosphotyrosine , Humans , Phosphotyrosine/metabolism , Phosphotyrosine/chemistry , Enzyme Inhibitors/pharmacology , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/chemical synthesis , Structure-Activity Relationship , Protein Tyrosine Phosphatases, Non-Receptor/antagonists & inhibitors , Protein Tyrosine Phosphatases, Non-Receptor/metabolism , Protein Tyrosine Phosphatases, Non-Receptor/chemistry , Protein Tyrosine Phosphatases/antagonists & inhibitors , Protein Tyrosine Phosphatases/metabolism , Molecular Structure , Biological Availability
18.
Proc Natl Acad Sci U S A ; 107(44): 19014-9, 2010 Nov 02.
Article in English | MEDLINE | ID: mdl-20956308

ABSTRACT

Alzheimer's disease (AD) is a progressive and incurable neurodegenerative disorder. Early in the pathophysiology of AD, synaptic function is disrupted by soluble Aß oligomers, possibly through Aß-mediated internalization of NMDA receptors. Striatal-enriched phosphatase (STEP) is a tyrosine phosphatase that regulates the internalization of NMDA receptors. Recent work shows that STEP is elevated in the prefrontal cortex of human AD patients and in animal models of AD. Here, we use genetic manipulations to reduce STEP activity in a triple transgenic AD mouse model and show that a decrease in STEP levels reverses cognitive and cellular deficits observed in these mice. Our results suggest that STEP inhibitors may prove therapeutic for this devastating disorder.


Subject(s)
Alzheimer Disease/enzymology , Cerebral Cortex/enzymology , Protein Tyrosine Phosphatases, Non-Receptor/metabolism , Alzheimer Disease/drug therapy , Alzheimer Disease/genetics , Alzheimer Disease/pathology , Amyloid beta-Protein Precursor/genetics , Amyloid beta-Protein Precursor/metabolism , Animals , Cerebral Cortex/pathology , Disease Models, Animal , Enzyme Inhibitors/therapeutic use , Humans , Mice , Mice, Transgenic , Protein Tyrosine Phosphatases, Non-Receptor/antagonists & inhibitors , Receptors, N-Methyl-D-Aspartate/genetics , Receptors, N-Methyl-D-Aspartate/metabolism
19.
BMC Cell Biol ; 13: 28, 2012 Oct 31.
Article in English | MEDLINE | ID: mdl-23114011

ABSTRACT

BACKGROUND: Autophagy is a ubiquitous cellular process responsible for the bulk degradation of cytoplasmic components through the autophagosomal-lysosomal pathway. In skeletal muscle, autophagy has been regarded as a key regulator for muscle mass maintenance, and its imbalance leads to sarcopenia. However, the underlying mechanism is poorly understood. RESULTS: In this study, we demonstrate that ceMTM3, a FYVE-domain containing myotubalarin family phosphatase, is required for the maintenance of muscle fibers by preventing excessive autophagy in Caenorhabditis elegans. Knockdown of ceMTM3 by using feeding-based RNA interference caused loss of muscle fibers accompanied by shortening of muscle cell and body size in aged C. elegans worms. This was preceded by the occurrence of excessive autophagy in the muscle and other tissues, which subsequently resulted in increased lysosomal activity and necrotic cell death. However, knockdown of ceMTM3 did not aggravate the abnormalities of muscle wasting in autophagy-deficient atg-18 mutant worms. CONCLUSIONS: Our data suggest an important role of ceMTM3 in regulating autophagy and maintaining muscle fibers. This study may have clinical implications for prevention and treatment of sarcopenia.


Subject(s)
Autophagy , Caenorhabditis elegans Proteins/metabolism , Muscles/metabolism , Protein Tyrosine Phosphatases, Non-Receptor/metabolism , Animals , Body Size , Caenorhabditis elegans/metabolism , Caenorhabditis elegans Proteins/antagonists & inhibitors , Caenorhabditis elegans Proteins/genetics , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mutation , Protein Tyrosine Phosphatases, Non-Receptor/antagonists & inhibitors , Protein Tyrosine Phosphatases, Non-Receptor/genetics , RNA Interference , RNA, Small Interfering/metabolism
20.
J Am Chem Soc ; 134(43): 18116-24, 2012 Oct 31.
Article in English | MEDLINE | ID: mdl-23075115

ABSTRACT

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


Subject(s)
Diabetes Mellitus, Type 2/drug therapy , Phenylalanine/analogs & derivatives , Protein Tyrosine Phosphatases, Non-Receptor/antagonists & inhibitors , Animals , Disease Models, Animal , Mice , Mice, Inbred C57BL , Mice, Obese , Models, Molecular , Molecular Structure , Phenylalanine/chemical synthesis , Phenylalanine/chemistry , Phenylalanine/pharmacology , Protein Tyrosine Phosphatases, Non-Receptor/chemistry , Protein Tyrosine Phosphatases, Non-Receptor/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL