Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 24
Filter
1.
Cardiovasc Res ; 117(6): 1546-1556, 2021 05 25.
Article in English | MEDLINE | ID: mdl-32653904

ABSTRACT

AIMS: Receptor-type vascular endothelial protein tyrosine phosphatase (VE-PTP) dephosphorylates Tie-2 as well as CD31, VE-cadherin, and vascular endothelial growth factor receptor 2 (VEGFR2). The latter form a signal transduction complex that mediates the endothelial cell response to shear stress, including the activation of the endothelial nitric oxide (NO) synthase (eNOS). As VE-PTP expression is increased in diabetes, we investigated the consequences of VE-PTP inhibition (using AKB-9778) on blood pressure in diabetic patients and the role of VE-PTP in the regulation of eNOS activity and vascular reactivity. METHODS AND RESULTS: In diabetic patients AKB-9778 significantly lowered systolic and diastolic blood pressure. This could be linked to elevated NO production, as AKB increased NO generation by cultured endothelial cells and elicited the NOS inhibitor-sensitive relaxation of endothelium-intact rings of mouse aorta. At the molecular level, VE-PTP inhibition increased the phosphorylation of eNOS on Tyr81 and Ser1177 (human sequence). The PIEZO1 activator Yoda1, which was used to mimic the response to shear stress, also increased eNOS Tyr81 phosphorylation, an effect that was enhanced by VE-PTP inhibition. Two kinases, i.e. abelson-tyrosine protein kinase (ABL)1 and Src were identified as eNOS Tyr81 kinases as their inhibition and down-regulation significantly reduced the basal and Yoda1-induced tyrosine phosphorylation and activity of eNOS. VE-PTP, on the other hand, formed a complex with eNOS in endothelial cells and directly dephosphorylated eNOS Tyr81 in vitro. Finally, phosphorylation of eNOS on Tyr80 (murine sequence) was found to be reduced in diabetic mice and diabetes-induced endothelial dysfunction (isolated aortic rings) was blunted by VE-PTP inhibition. CONCLUSIONS: VE-PTP inhibition enhances eNOS activity to improve endothelial function and decrease blood pressure indirectly, through the activation of Tie-2 and the CD31/VE-cadherin/VEGFR2 complex, and directly by dephosphorylating eNOS Tyr81. VE-PTP inhibition, therefore, represents an attractive novel therapeutic option for diabetes-induced endothelial dysfunction and hypertension.


Subject(s)
Aniline Compounds/therapeutic use , Antihypertensive Agents/therapeutic use , Diabetes Mellitus/drug therapy , Endothelial Cells/drug effects , Endothelium, Vascular/drug effects , Enzyme Inhibitors/therapeutic use , Hypertension/drug therapy , Nitric Oxide Synthase Type III/metabolism , Receptor-Like Protein Tyrosine Phosphatases, Class 3/antagonists & inhibitors , Sulfonic Acids/therapeutic use , Animals , Blood Pressure/drug effects , Cells, Cultured , Diabetes Mellitus/enzymology , Diabetes Mellitus/genetics , Diabetes Mellitus/physiopathology , Disease Models, Animal , Endothelial Cells/enzymology , Endothelium, Vascular/enzymology , Endothelium, Vascular/physiopathology , Humans , Hypertension/enzymology , Hypertension/genetics , Hypertension/physiopathology , Mice, Inbred C57BL , Mice, Transgenic , Nitric Oxide/metabolism , Nitric Oxide Synthase Type III/genetics , Phosphorylation , Receptor-Like Protein Tyrosine Phosphatases, Class 3/genetics , Receptor-Like Protein Tyrosine Phosphatases, Class 3/metabolism , Signal Transduction , Treatment Outcome , United States
2.
Invest Ophthalmol Vis Sci ; 61(14): 12, 2020 12 01.
Article in English | MEDLINE | ID: mdl-33315051

ABSTRACT

Purpose: Tyrosine kinase with immunoglobulin-like and EGF-like domains 2 (Tie2) activation in Schlemm's canal (SC) endothelium is required for the maintenance of IOP, making the angiopoietin/Tie2 pathway a target for new and potentially disease modifying glaucoma therapies. The goal of the present study was to examine the effects of a Tie2 activator, AKB-9778, on IOP and outflow function. Methods: AKB-9778 effects on IOP was evaluated in humans, rabbits, and mice. Localization studies of vascular endothelial protein tyrosine phosphatase (VE-PTP), the target of AKB-9778 and a negative regulator of Tie2, were performed in human and mouse eyes. Mechanistic studies were carried out in mice, monitoring AKB-9778 effects on outflow facility, Tie2 phosphorylation, and filtration area of SC. Results: AKB-9778 lowered IOP in patients treated subcutaneously for diabetic eye disease. In addition to efficacious, dose-dependent IOP lowering in rabbit eyes, topical ocular AKB-9778 increased Tie2 activation in SC endothelium, reduced IOP, and increased outflow facility in mouse eyes. VE-PTP was localized to SC endothelial cells in human and mouse eyes. Mechanistically, AKB-9778 increased the filtration area of SC for aqueous humor efflux in both wild type and in Tie2+/- mice. Conclusions: This is the first report of IOP lowering in humans with a Tie2 activator and functional demonstration of its action in remodeling SC to increase outflow facility and lower IOP in fully developed mice. Based on these studies, a phase II clinical trial is in progress to advance topical ocular AKB-9778 as a first in class, Tie2 activator for treatment for ocular hypertension and glaucoma.


Subject(s)
Aniline Compounds/pharmacology , Intraocular Pressure/drug effects , Receptor, TIE-2/metabolism , Receptor-Like Protein Tyrosine Phosphatases, Class 3/antagonists & inhibitors , Sulfonic Acids/pharmacology , Trabecular Meshwork/drug effects , Animals , Diabetic Retinopathy/drug therapy , Double-Blind Method , Female , Fluorescent Antibody Technique , Glaucoma/drug therapy , Glaucoma/pathology , Humans , Male , Mice , Mice, Inbred C57BL , Middle Aged , Trabecular Meshwork/metabolism , Trabecular Meshwork/pathology
3.
Bioorg Med Chem ; 28(23): 115777, 2020 12 01.
Article in English | MEDLINE | ID: mdl-32992253

ABSTRACT

A series of novel triaryl-based sulfamic acid analogs was designed, synthesized and evaluated as inhibitors of human protein tyrosine phosphatase beta (HPTPß). A novel, easy and efficient synthetic method was developed for target compounds, and the activity determination results showed that most of compounds were good HPTPß inhibitors. Interestingly, the compounds G4 and G25 with simple structure not only showed potent inhibitory activity on HPTPß but also had good inhibitory selectivity over other PTPs (PTP1B, SHP2, LAR and TC-PTP). The molecular docking simulation of compounds with the protein HPTPß helped us understand the structure-activity relationship and clarify some confusing assay results. This research provides references for further drug design of HPTPß and other PTPs inhibitors.


Subject(s)
Enzyme Inhibitors/chemistry , Receptor-Like Protein Tyrosine Phosphatases, Class 3/antagonists & inhibitors , Sulfonic Acids/chemistry , Benzene Derivatives/chemistry , Binding Sites , Drug Design , Enzyme Inhibitors/metabolism , Humans , Molecular Docking Simulation , Protein Isoforms/antagonists & inhibitors , Protein Isoforms/metabolism , Protein Tyrosine Phosphatases/antagonists & inhibitors , Protein Tyrosine Phosphatases/metabolism , Receptor-Like Protein Tyrosine Phosphatases, Class 3/metabolism , Structure-Activity Relationship , Sulfonic Acids/metabolism
4.
EMBO Rep ; 20(7): e47046, 2019 07.
Article in English | MEDLINE | ID: mdl-31267715

ABSTRACT

Inhibition of VE-PTP, an endothelial receptor-type tyrosine phosphatase, triggers phosphorylation of the tyrosine kinase receptor Tie-2, which leads to the suppression of inflammation-induced vascular permeability. Analyzing the underlying mechanism, we show here that inhibition of VE-PTP and activation of Tie-2 induce tyrosine phosphorylation of FGD5, a GTPase exchange factor (GEF) for Cdc42, and stimulate its translocation to cell contacts. Interfering with the expression of FGD5 blocks the junction-stabilizing effect of VE-PTP inhibition in vitro and in vivo. Likewise, FGD5 is required for strengthening cortical actin bundles and inhibiting radial stress fiber formation, which are each stimulated by VE-PTP inhibition. We identify Y820 of FGD5 as the direct substrate for VE-PTP. The phosphorylation of FGD5-Y820 is required for the stabilization of endothelial junctions and for the activation of Cdc42 by VE-PTP inhibition but is dispensable for the recruitment of FGD5 to endothelial cell contacts. Thus, activation of FGD5 is a two-step process that comprises membrane recruitment and phosphorylation of Y820. These steps are necessary for the junction-stabilizing effect stimulated by VE-PTP inhibition and Tie-2 activation.


Subject(s)
Guanine Nucleotide Exchange Factors/metabolism , Human Umbilical Vein Endothelial Cells/metabolism , Intercellular Junctions/metabolism , Receptor-Like Protein Tyrosine Phosphatases, Class 3/metabolism , Actin Cytoskeleton/metabolism , Amino Acid Motifs , Animals , Female , Guanine Nucleotide Exchange Factors/chemistry , Guanine Nucleotide Exchange Factors/genetics , HEK293 Cells , Humans , Mice , Mutation , Phosphorylation , Receptor-Like Protein Tyrosine Phosphatases, Class 3/antagonists & inhibitors , cdc42 GTP-Binding Protein/metabolism , rac1 GTP-Binding Protein/metabolism
5.
J Immunother ; 42(7): 237-243, 2019 09.
Article in English | MEDLINE | ID: mdl-31348125

ABSTRACT

Administration of interleukin (IL)-2 has led to a durable response in patients with advanced renal cancer and melanoma but is restricted for clinical application because of adverse effects, including the vascular leak syndrome (VLS). VLS is associated with increased circulating levels of the Tie2 antagonist ligand, angiopoietin 2, and decreased Tie2 receptor phosphorylation and downstream signaling in endothelial cells (ECs). Given that vascular endothelial protein tyrosine phosphatase (VE-PTP) is a specific membrane phosphatase in ECs that dephosphorylates Tie2, the effects of targeting VE-PTP by a selective inhibitor AKB-9778 (AKB) in terms of VLS and antitumor efficacy were examined in this study. The authors found, by targeting VE-PTP, that the antitumor effects induced by IL-2 were augmented [tumor-free 44% (IL-2 alone) vs. 87.5% (IL-2+AKB)], associated with enhanced immune cell infiltrate (90% increase for CD8 T cells and natural killer cells). In addition, the side effects of IL-2 therapy were lessened, as demonstrated by diminished lung weight (less vascular leakage) as well as reduced cytokine levels (serum HMGB1 from 137.04±2.69 to 43.86±3.65 pg/mL; interferon-γ from 590.52±90.52 to 31.37±1.14 pg/mL). The authors further sought to determine the potential mechanism of the action of AKB-9778. The findings suggest that AKB-9778 may function through reducing serum angiopoietin 2 level and regulating EC viability. These findings provide insights into the targeting VE-PTP to improve tolerance and efficacy of IL-2 therapy and highlight the clinical potential of AKB-9778 for treating patients with VLS and cancer.


Subject(s)
Aniline Compounds/pharmacology , Antineoplastic Agents/pharmacology , Interleukin-2/administration & dosage , Receptor-Like Protein Tyrosine Phosphatases, Class 3/antagonists & inhibitors , Sulfonic Acids/pharmacology , Aniline Compounds/therapeutic use , Animals , Antineoplastic Agents/therapeutic use , Cell Line, Tumor , Cytokines/blood , Cytokines/metabolism , Disease Models, Animal , Drug Synergism , Female , Gene Expression , Genes, Reporter , Humans , Inflammation Mediators , Interleukin-2/adverse effects , Liver Neoplasms/drug therapy , Liver Neoplasms/pathology , Liver Neoplasms/secondary , Mice , Sulfonic Acids/therapeutic use , Xenograft Model Antitumor Assays
6.
Cell Death Dis ; 10(5): 352, 2019 04 30.
Article in English | MEDLINE | ID: mdl-31040266

ABSTRACT

Dysregulation of protein tyrosine phosphatase, receptor type B (PTPRB) correlates with the development of a variety of tumors. Here we show that PTPRB promotes metastasis of colorectal cancer (CRC) cells via inducing epithelial-mesenchymal transition (EMT). We find that PTPRB is expressed at significantly higher levels in CRC tissues compared to adjacent nontumor tissues and in CRC cell lines with high invasion. PTPRB knockdown decreased the number of invasive CRC cells in an in vitro wound healing model, and also reduced tumor metastasis in vivo. Conversely, PTPRB overexpression promoted CRC cell invasion in vitro and metastasis in vivo. PTPRB overexpression decreased vimentin expression and promoted E-cadherin expression, consistent with promotion of EMT, while PTPRB knockdown had the opposite effect. Hypoxic conditions induced EMT and promoted invasion in CRC cells, but these effects were eliminated by PTPRB knockdown. EMT blockade via TWIST1 knockdown inhibited the migration and invasiveness of CRC cells, and even increased PTPRB expression could not reverse this effect. Altogether, these data support the conclusion that PTPRB promotes invasion and metastasis of CRC cells via inducing EMT, and that PTPRB would be a novel therapeutic target for the treatment of CRC.


Subject(s)
Colorectal Neoplasms/pathology , Epithelial-Mesenchymal Transition , Receptor-Like Protein Tyrosine Phosphatases, Class 3/metabolism , Animals , Cadherins/metabolism , Cell Line, Tumor , Cell Movement , Colorectal Neoplasms/drug therapy , Colorectal Neoplasms/metabolism , Humans , Male , Mice , Mice, Nude , Neoplasm Invasiveness , Neoplasm Metastasis , Nuclear Proteins/antagonists & inhibitors , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , RNA Interference , RNA, Small Interfering/metabolism , RNA, Small Interfering/therapeutic use , Receptor-Like Protein Tyrosine Phosphatases, Class 3/antagonists & inhibitors , Receptor-Like Protein Tyrosine Phosphatases, Class 3/genetics , Transplantation, Heterologous , Twist-Related Protein 1/antagonists & inhibitors , Twist-Related Protein 1/genetics , Twist-Related Protein 1/metabolism
7.
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
8.
J Microbiol Biotechnol ; 29(4): 645-650, 2019 Apr 28.
Article in English | MEDLINE | ID: mdl-30845793

ABSTRACT

Brown adipocytes have an important role in the regulation of energy balance through uncoupling protein-1 (UCP-1)-mediated nonshivering thermogenesis. Although brown adipocytes have been highlighted as a new therapeutic target for the treatment of metabolic diseases, such as obesity and type II diabetes in adult humans, the molecular mechanism underlying brown adipogenesis is not fully understood. We recently found that protein tyrosine phosphatase receptor type B (PTPRB) expression dramatically decreased during brown adipogenic differentiation. In this study, we investigated the functional roles of PTPRB and its regulatory mechanism during brown adipocyte differentiation. Ectopic expression of PTPRB led to a reduced brown adipocyte differentiation by suppressing the tyrosine phosphorylation of VEGFR2, whereas a catalytic inactive PTPRB mutant showed no effects on differentiation and phosphorylation. Consistently, the expression of brown adipocyte-related genes, such as UCP-1, PGC-1α, PRDM16, PPAR-γ, and CIDEA, were significantly inhibited by PTPRB overexpression. Overall, these results suggest that PTPRB functions as a negative regulator of brown adipocyte differentiation through its phosphatase activity-dependent mechanism and may be used as a target protein for the regulation of obesity and type II diabetes.


Subject(s)
Adipocytes, Brown/drug effects , Adipogenesis/physiology , Cell Differentiation/drug effects , Receptor-Like Protein Tyrosine Phosphatases, Class 3/antagonists & inhibitors , Vascular Endothelial Growth Factor Receptor-2/metabolism , Cell Line , Diabetes Mellitus, Type 2 , Gene Expression Regulation , Humans , NADH Dehydrogenase , Obesity , Phosphorylation , Receptor-Like Protein Tyrosine Phosphatases, Class 3/genetics , Tyrosine/metabolism , Uncoupling Protein 1/metabolism
9.
Blood ; 133(12): 1346-1357, 2019 03 21.
Article in English | MEDLINE | ID: mdl-30591527

ABSTRACT

Inherited thrombocytopenias (ITs) are a heterogeneous group of disorders characterized by low platelet count that may result in bleeding tendency. Despite progress being made in defining the genetic causes of ITs, nearly 50% of patients with familial thrombocytopenia are affected with forms of unknown origin. Here, through exome sequencing of 2 siblings with autosomal-recessive thrombocytopenia, we identified biallelic loss-of-function variants in PTPRJ . This gene encodes for a receptor-like PTP, PTPRJ (or CD148), which is expressed abundantly in platelets and megakaryocytes. Consistent with the predicted effects of the variants, both probands have an almost complete loss of PTPRJ at the messenger RNA and protein levels. To investigate the pathogenic role of PTPRJ deficiency in hematopoiesis in vivo, we carried out CRISPR/Cas9-mediated ablation of ptprja (the ortholog of human PTPRJ) in zebrafish, which induced a significantly decreased number of CD41+ thrombocytes in vivo. Moreover, megakaryocytes of our patients showed impaired maturation and profound defects in SDF1-driven migration and formation of proplatelets in vitro. Silencing of PTPRJ in a human megakaryocytic cell line reproduced the functional defects observed in patients' megakaryocytes. The disorder caused by PTPRJ mutations presented as a nonsyndromic thrombocytopenia characterized by spontaneous bleeding, small-sized platelets, and impaired platelet responses to the GPVI agonists collagen and convulxin. These platelet functional defects could be attributed to reduced activation of Src family kinases. Taken together, our data identify a new form of IT and highlight a hitherto unknown fundamental role for PTPRJ in platelet biogenesis.


Subject(s)
Blood Platelets/pathology , Genetic Predisposition to Disease , Megakaryocytes/pathology , Mutation , Thrombocytopenia/pathology , Adolescent , Adult , Animals , Blood Platelets/metabolism , CRISPR-Cas Systems , Child , Female , Follow-Up Studies , Hematopoiesis , Humans , Male , Megakaryocytes/metabolism , Middle Aged , Pedigree , Prognosis , Receptor-Like Protein Tyrosine Phosphatases, Class 3/antagonists & inhibitors , Receptor-Like Protein Tyrosine Phosphatases, Class 3/genetics , Thrombocytopenia/etiology , Thrombocytopenia/genetics , Zebrafish
10.
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
11.
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
12.
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
13.
J Antibiot (Tokyo) ; 71(6): 557-563, 2018 06.
Article in English | MEDLINE | ID: mdl-29463888

ABSTRACT

Two new meroterpenoid-type fungal metabolites, furanoaustinol (1) and 7-acetoxydehydroaustinol (2), were isolated from the ethyl acetate extract of a marine-derived fungal strain Penicillium sp. SF-5497, along with eight (3-10) known meroterpenoids. Their structures were elucidated mainly based on the analysis of their NMR (1D and 2D) and MS data. Particularly, the novel meroterpenoid, furanoaustinol (1), belonging to the austin group, was identified to possess an unprecedented hexacyclic ring system. Biological evaluation of these compounds revealed that furanoaustinol (1) weakly inhibited the activity of protein tyrosine phosphatase 1B in a dose-dependent manner with an IC50 value of 77.2 µM. In addition, 7-acetoxydehydroaustinol (2) and four other known meroterpenoids (5, 7, 9, and 10) weakly suppressed the overproduction of nitric oxide in lipopolysaccharide-challenged BV2 microglial cells with IC50 values of 61.0, 30.1, 58.3, 37.6, and 40.2 µM, respectively.


Subject(s)
Penicillium/chemistry , Terpenes/isolation & purification , Dose-Response Relationship, Drug , Fermentation , Humans , Magnetic Resonance Spectroscopy , Mass Spectrometry , Microglia/drug effects , Microglia/metabolism , Molecular Structure , Nitric Oxide/biosynthesis , Receptor-Like Protein Tyrosine Phosphatases, Class 3/antagonists & inhibitors
14.
Ophthalmology ; 123(8): 1722-1730, 2016 08.
Article in English | MEDLINE | ID: mdl-27236272

ABSTRACT

PURPOSE: To assess the effect of AKB-9778 alone or in combination with ranibizumab in subjects with diabetic macular edema (DME). DESIGN: A phase IIa, randomized, placebo- and sham injection-controlled, double-masked clinical trial. PARTICIPANTS: Subjects (n = 144) with decreased vision from DME and central subfield thickness (CST) ≥325 µm measured by spectral-domain optical coherence tomography (SD OCT) enrolled at 36 sites. METHODS: Subjects were randomized to (1) AKB-9778 monotherapy: subcutaneous AKB-9778 15 mg twice per day (BID) + monthly sham intraocular injections; (2) combination therapy: subcutaneous AKB-9778 15 mg BID + monthly 0.3 mg ranibizumab; or (3) ranibizumab monotherapy: subcutaneous placebo injections BID + monthly 0.3 mg ranibizumab. Best-corrected visual acuity (BCVA) and CST were measured at baseline and every 4 weeks. MAIN OUTCOME MEASURES: Primary outcome measure was mean change from baseline CST at week 12. Other outcomes included BCVA, safety assessments, and Diabetic Retinopathy Severity Score (DRSS). RESULTS: At week 12, mean change from baseline CST was significantly greater in the combination group (-164.4±24.2 µm) compared with the ranibizumab monotherapy group (-110.4±17.2 µm; P = 0.008) and was 6.2±13.0 µm in the AKB-9778 monotherapy group. Mean CST at week 12 and percentage of eyes with resolved edema was 340.0±11.2 µm and 29.2%, respectively, in the combination group versus 392.1±17.1 µm and 17.0%, respectively, in the ranibizumab monotherapy group. Mean change from baseline BCVA (letters) was 6.3±1.3 in the combination group, 5.7±1.2 in the ranibizumab monotherapy group, and 1.5±1.2 in the AKB-9778 monotherapy group. The percentage of study eyes that gained ≥10 or ≥15 letters was 8.7% and 4.3%, respectively, in the AKB-9778 monotherapy group, 29.8% and 17.0%, respectively, in the ranibizumab monotherapy group, and 35.4% and 20.8%, respectively, in the combination group. Improvements in DRSS in study eyes were similar across groups, and the percentage of qualified fellow eyes with a ≥2-step change was 11.4% in all AKB-9778-treated subjects compared with 4.2% in the ranibizumab monotherapy group. AKB-9778 was well tolerated, with no clear by-treatment differences in adverse events. CONCLUSIONS: Activation of Tie2 by subcutaneous injections of AKB-9778 combined with suppression of vascular endothelial growth factor (VEGF) causes a significantly greater reduction in DME than that seen with suppression of VEGF alone.


Subject(s)
Angiogenesis Inhibitors/therapeutic use , Aniline Compounds/therapeutic use , Diabetic Retinopathy/drug therapy , Macular Edema/drug therapy , Ranibizumab/therapeutic use , Receptor, TIE-2/metabolism , Sulfonic Acids/therapeutic use , Vascular Endothelial Growth Factor A/antagonists & inhibitors , Diabetes Mellitus, Type 1/complications , Diabetes Mellitus, Type 2/complications , Diabetic Retinopathy/diagnosis , Diabetic Retinopathy/metabolism , Double-Blind Method , Drug Therapy, Combination , Female , Fluorescein Angiography , Humans , Injections, Subcutaneous , Intravitreal Injections , Macular Edema/diagnosis , Macular Edema/metabolism , Male , Middle Aged , Receptor-Like Protein Tyrosine Phosphatases, Class 3/antagonists & inhibitors , Tomography, Optical Coherence , Visual Acuity
15.
Oncol Rep ; 33(4): 1737-44, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25634668

ABSTRACT

Gene therapy is a promising therapeutic approach for chemoresistant cervical cancers. Therapeutic interventions targeting the key factors contributing to the initiation and progression of cervical cancer may be a more effective treatment strategy. In the present study, we firstly determined the expression of protein tyrosine phosphatase receptor J (PTPRJ) in 8-paired human cervical tumor and non-tumor tissues. We observed a striking downregulation of PTPRJ in the human cervical tumor tissues. Next, we investigated the roles and the function mechanism of PTPRJ in the human cervical carcinoma cell line C33A by loss- and gain-of-function experiments. Our study indicated that C33A cells with loss of PTPRJ expression showed a significantly increased cell viability, rising growth and migration rate, as well as a G1-S transition. We obtained the opposite results when we overexpressed PTPRJ in C33A cells. Our further study indicated that PTPRJ levels were highly correlated with cell survival when the C33A cells were treated with 5-fluorouracil (5-FU), an important chemotherapeutic agent for cervical cancer. In addition, the signaling pathway screening assay showed an obvious alteration of the Janus kinase 1/signal transducer and activator of transcription 3 (JAK1/STAT3) pathway. PTPRJ negatively regulated the activation of the JAK1/STAT3 pathway by decreasing the phosphorylation levels of JAK1 and STAT3. In addition, PTPRJ also regulated the expression of the downstream factors of STAT3, such as cyclin D, Bax, VEGF and MMP2. Our results suggest that PTPRJ may be a promising gene therapy target and its therapeutic potential can be fulfilled when used alone, or in combination with other anticancer agents.


Subject(s)
Antimetabolites, Antineoplastic/pharmacology , Carcinoma/pathology , Drug Resistance, Neoplasm/physiology , Fluorouracil/pharmacology , Neoplasm Proteins/antagonists & inhibitors , Protein Processing, Post-Translational , Signal Transduction/physiology , Tumor Suppressor Proteins/physiology , Uterine Cervical Neoplasms/pathology , Carcinoma/enzymology , Cell Division , Cell Line, Tumor , Cell Movement , Enzyme Activation , Female , G1 Phase Cell Cycle Checkpoints , Gene Expression Regulation, Enzymologic , Gene Expression Regulation, Neoplastic , Genes, Tumor Suppressor , HEK293 Cells , Humans , Janus Kinase 1 , Neoplasm Proteins/biosynthesis , Neoplasm Proteins/genetics , Neoplasm Proteins/physiology , Phosphorylation , RNA, Messenger/biosynthesis , RNA, Neoplasm/biosynthesis , Receptor-Like Protein Tyrosine Phosphatases, Class 3/antagonists & inhibitors , Receptor-Like Protein Tyrosine Phosphatases, Class 3/biosynthesis , Receptor-Like Protein Tyrosine Phosphatases, Class 3/genetics , Receptor-Like Protein Tyrosine Phosphatases, Class 3/physiology , Recombinant Fusion Proteins/metabolism , STAT3 Transcription Factor , Tumor Suppressor Proteins/antagonists & inhibitors , Tumor Suppressor Proteins/biosynthesis , Uterine Cervical Neoplasms/enzymology
16.
Ophthalmology ; 122(3): 545-54, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25439435

ABSTRACT

PURPOSE: AKB-9778 is a small-molecule competitive inhibitor of vascular endothelial-protein tyrosine phosphatase (VE-PTP) that promotes Tie2 activation and reduces vascular leakage and neovascularization in mouse models. The purpose of this study was to test the safety, tolerability, pharmacokinetics, and biological activity of AKB-9778 in patients with diabetic macular edema (DME). DESIGN: Open-label, dose-escalation clinical trial. PARTICIPANTS: Four dose cohorts of 6 patients with DME self-administered subcutaneous injections of 5 mg, 15 mg, 22.5 mg, or 30 mg AKB-9778 twice daily for 4 weeks. METHODS: Patients were seen weekly during a 4-week treatment period for safety assessments, best-corrected visual acuity (BCVA) assessment by Early Treatment Diabetic Retinopathy Study protocol, and measurement of central subfield thickness (CST) by spectral-domain optical coherence tomography. Additional safety assessments were performed at 6, 8, and 12 weeks. MAIN OUTCOME MEASURES: Safety assessments, change from baseline BCVA, and change from baseline CST. RESULTS: All doses were well tolerated. A modest, transient reduction in blood pressure and adverse events consistent with vasodilatory activity of AKB-9778 emerged at doses of 22.5 mg or more twice daily. At the week 4 primary end point, BCVA improved 5 letters or more from baseline in 13 of the 18 patients receiving 15 mg or more twice daily; 1 patient improved by 10 to 15 letters, and 2 patients improved by more than 15 letters. Among 18 patients receiving 15 mg or more twice daily, CST decreased by more than 100 µm in 5 patients and by 50 to 100 µm in 2 patients. There was a significant correlation between reduction in CST and improvement in BCVA. CONCLUSIONS: No safety concerns were identified after systemic administration of AKB-9778 for 4 weeks in patients with DME, and doses of 15 mg or more twice daily reduced macular edema and improved vision in some patients. This is a preliminary demonstration of clinical safety and efficacy of a VE-PTP inhibitor and Tie2 activator.


Subject(s)
Aniline Compounds/administration & dosage , Diabetic Retinopathy/drug therapy , Enzyme Inhibitors/administration & dosage , Macular Edema/drug therapy , Receptor, TIE-2/metabolism , Receptor-Like Protein Tyrosine Phosphatases, Class 3/antagonists & inhibitors , Sulfonic Acids/administration & dosage , Adult , Aged , Aged, 80 and over , Aniline Compounds/adverse effects , Blood Pressure/drug effects , Diabetic Retinopathy/metabolism , Enzyme Inhibitors/adverse effects , Female , Fluorescein Angiography , Humans , Injections, Subcutaneous , Macular Edema/metabolism , Male , Middle Aged , Sulfonic Acids/adverse effects , Tomography, Optical Coherence , Visual Acuity/physiology
18.
J Natl Cancer Inst ; 105(16): 1188-201, 2013 Aug 21.
Article in English | MEDLINE | ID: mdl-23899555

ABSTRACT

BACKGROUND: The solid tumor microvasculature is characterized by structural and functional abnormality and mediates several deleterious aspects of tumor behavior. Here we determine the role of vascular endothelial protein tyrosine phosphatase (VE-PTP), which deactivates endothelial cell (EC) Tie-2 receptor tyrosine kinase, thereby impairing maturation of tumor vessels. METHODS: AKB-9778 is a first-in-class VE-PTP inhibitor. We examined its effects on ECs in vitro and on embryonic angiogenesis in vivo using zebrafish assays. We studied the impact of AKB-9778 therapy on the tumor vasculature, tumor growth, and metastatic progression using orthotopic models of murine mammary carcinoma as well as spontaneous and experimental metastasis models. Finally, we used endothelial nitric oxide synthase (eNOS)-deficient mice to establish the role of eNOS in mediating the effects of VE-PTP inhibition. All statistical tests were two-sided. RESULTS: AKB-9778 induced ligand-independent Tie-2 activation in ECs and impaired embryonic zebrafish angiogenesis. AKB-9778 delayed the early phase of mammary tumor growth by maintaining vascular maturity (P < .01, t test); slowed growth of micrometastases (P < .01, χ(2) test) by preventing extravasation of tumor cells (P < 0.01, Fisher exact test), resulting in a trend toward prolonged survival (27.0 vs 36.5 days; hazard ratio of death = 0.33, 95% confidence interval = 0.11 to 1.03; P = .05, Mantel-Cox test); and stabilized established primary tumor blood vessels, enhancing tumor perfusion (P = .03 for 4T1 tumor model and 0.05 for E0771 tumor model, by two-sided t tests) and, hence, radiation response (P < .01, analysis of variance; n = 7 mice per group). The effects of AKB-9778 on tumor vessels were mediated in part by endothelial nitric oxide synthase activation. CONCLUSIONS: Our results demonstrate that pharmacological VE-PTP inhibition can normalize the structure and function of tumor vessels through Tie-2 activation, which delays tumor growth, slows metastatic progression, and enhances response to concomitant cytotoxic treatments.


Subject(s)
Antineoplastic Agents/pharmacology , Breast Neoplasms/pathology , Enzyme Inhibitors/pharmacology , Lung Neoplasms/prevention & control , Neovascularization, Pathologic/drug therapy , Receptor-Like Protein Tyrosine Phosphatases, Class 3/antagonists & inhibitors , Zebrafish Proteins/metabolism , Angiogenesis Inhibitors/pharmacology , Animals , Breast Neoplasms/blood supply , Disease Progression , Drug Synergism , Enzyme Activation/drug effects , Female , Human Umbilical Vein Endothelial Cells , Lung Neoplasms/secondary , Mice , Mice, Inbred C57BL , Nitric Oxide Synthase Type III/metabolism , Receptor, TIE-2/metabolism , Xenograft Model Antitumor Assays , Zebrafish
19.
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
20.
ACS Chem Biol ; 7(10): 1666-76, 2012 Oct 19.
Article in English | MEDLINE | ID: mdl-22759068

ABSTRACT

PTPRJ is a receptor-type protein tyrosine phosphatase whose expression is strongly reduced in the majority of investigated cancer cell lines and tumor specimens. PTPRJ negatively interferes with mitogenic signals originating from several oncogenic receptor tyrosine kinases, including HGFR, PDGFR, RET, and VEGFR-2. Here we report the isolation and characterization of peptides from a random peptide phage display library that bind and activate PTPRJ. These agonist peptides, which are able to both circularize and form dimers in acqueous solution, were assayed for their biochemical and biological activity on both human cancer cells and primary endothelial cells (HeLa and HUVEC, respectively). Our results demonstrate that binding of PTPRJ-interacting peptides to cell cultures dramatically reduces the extent of both MAPK phosphorylation and total phosphotyrosine levels; conversely, they induce a significant increase of the cell cycle inhibitor p27(Kip1). Moreover, PTPRJ agonist peptides both reduce proliferation and trigger apoptosis of treated cells. Our data indicate that peptide agonists of PTPRJ positively modulate the PTPRJ activity and may lead to novel targeted anticancer therapies.


Subject(s)
Antineoplastic Agents/pharmacology , Peptides/pharmacology , Apoptosis/drug effects , Cyclin-Dependent Kinase Inhibitor p27/metabolism , HeLa Cells , Human Umbilical Vein Endothelial Cells/metabolism , Humans , Mitogen-Activated Protein Kinases/metabolism , Models, Molecular , Peptide Library , Phosphorylation , Phosphotyrosine/metabolism , Receptor-Like Protein Tyrosine Phosphatases, Class 3/antagonists & inhibitors
SELECTION OF CITATIONS
SEARCH DETAIL
...