Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
Add more filters










Publication year range
1.
Alcohol Clin Exp Res ; 44(5): 1037-1045, 2020 05.
Article in English | MEDLINE | ID: mdl-32154588

ABSTRACT

BACKGROUND: Pleiotrophin (PTN) and midkine (MK) are cytokines that are up-regulated in the prefrontal cortex (PFC) after alcohol administration and have been shown to reduce alcohol intake and reward. Both cytokines are endogenous inhibitors of receptor protein tyrosine phosphatase (RPTP) ß/ζ (a.k.a. PTPRZ1). Recently, a new compound named MY10 was designed with the aim of mimicking the activity of PTN and MK. MY10 has already shown promising results regulating alcohol-related behaviors in mice. METHODS: We have now tested the effects of MY10 on alcohol operant self-administration and Drinking In the Dark-Multiple Scheduled Access (DID-MSA) paradigms in rats. Gene expression of relevant genes in the PTN/MK signaling pathway in the PFC was analyzed by real-time PCR. RESULTS: MY10, at the highest dose tested (100 mg/kg), reduced alcohol consumption in the alcohol operant self-administration paradigm (p = 0.040). In the DID-MSA paradigm, rats drank significantly less alcohol (p = 0.019) and showed a significant decrease in alcohol preference (p = 0.002). We observed that the longer the exposure to alcohol, the greater the suppressing effects of MY10 on alcohol consumption. It was demonstrated that the effects of MY10 were specific to alcohol since saccharin intake was not affected by MY10 (p = 0.804). MY10 prevented the alcohol-induced down-regulation of Ptprz1 (p = 0.004) and anaplastic lymphoma kinase (Alk; p = 0.013) expression. CONCLUSIONS: Our results support and provide further evidence regarding the efficacy of MY10 on alcohol-related behaviors and suggest the consideration of the blockade of RPTPß/ζ as a target for reducing excessive alcohol consumption.


Subject(s)
Alcohol Drinking/drug therapy , Enzyme Inhibitors/administration & dosage , Receptor-Like Protein Tyrosine Phosphatases, Class 5/antagonists & inhibitors , Animals , Carrier Proteins/genetics , Carrier Proteins/pharmacology , Cytokines/genetics , Cytokines/pharmacology , Dose-Response Relationship, Drug , Gene Expression/drug effects , Male , Midkine/genetics , Midkine/pharmacology , Rats , Rats, Wistar , Receptor-Like Protein Tyrosine Phosphatases, Class 5/genetics , Signal Transduction/genetics
2.
J Neurotrauma ; 37(3): 534-554, 2020 02 01.
Article in English | MEDLINE | ID: mdl-31426714

ABSTRACT

L1 cell adhesion molecule (L1CAM) supports spinal cord cellular milieu after contusion and compression lesions, contributing to neuroprotection, promoting axonal outgrowth, and reducing outgrowth-inhibitory molecules in lesion proximity. We extended investigations into L1CAM molecular targets and explored long-distance effects of L1CAM rostral and caudal to complete spinal cord transection (SCT) in adult rats. L1CAM overexpression in neurons and glia after Th10/Th11 SCT was achieved using adeno-associated viral vector serotype 5 (AAV5) injected into an L1-lumbar segment immediately after transection. At 5 weeks, a L1CAM mRNA profound decrease detected rostral and caudal to the transection site was alleviated by AAV5-L1CAM treatment, with increased endogenous L1CAM rostral to the SCT. Transected corticospinal tract fibers showed attenuated retraction after treatment, accompanied by a multi-segmental increase of lesion-reduced expression of adenylate cyclase 1 (Adcy1), synaptophysin, growth-associated protein 43, and myelin basic protein genes caudal to transection, and Adcy1 rostral to transection. In parallel, chondroitin sulfate proteoglycan phosphacan elevated after SCT was downregulated after treatment. Low-molecular L1CAM isoforms generated after spinalization indicated the involvement of sheddases in L1CAM processing and long-distance effects. A disintegrin and metalloproteinase (ADAM)10 sheddase immunoreactivity, stronger in AAV5-L1CAM than AAV5- enhanced green fluorescent protein (EGFP)-transduced motoneurons indicated local ADAM10 upregulation by L1CAM. The results suggest that increased L1CAM availability and penetration of diffusible L1CAM fragments post-lesion induce both local and long-distance neuronal and glial responses toward better neuronal maintenance, neurite growth, and myelination. Despite the fact that intervention promoted beneficial molecular changes, kinematic analysis of hindlimb movements showed minor improvement, indicating that spinalized rats require longer L1CAM treatment to regain locomotor functions.


Subject(s)
Down-Regulation/physiology , Neural Cell Adhesion Molecule L1/biosynthesis , Neuronal Plasticity/physiology , Receptor-Like Protein Tyrosine Phosphatases, Class 5/biosynthesis , Spinal Cord Injuries/metabolism , Up-Regulation/physiology , Animals , Gene Expression , Male , Neural Cell Adhesion Molecule L1/genetics , Rats , Rats, Wistar , Receptor-Like Protein Tyrosine Phosphatases, Class 5/antagonists & inhibitors , Receptor-Like Protein Tyrosine Phosphatases, Class 5/genetics , Spinal Cord Injuries/genetics , Thoracic Vertebrae/injuries
3.
Behav Brain Res ; 369: 111933, 2019 09 02.
Article in English | MEDLINE | ID: mdl-31054277

ABSTRACT

Pleiotrophin (PTN) and Midkine (MK) are neurotrophic factors that are upregulated in the prefrontal cortex after alcohol administration and have been shown to reduce ethanol drinking and reward. PTN and MK are endogenous inhibitors of Receptor Protein Tyrosine Phosphatase (RPTP) ß/ζ. Interestingly, pharmacological inhibition of RPTPß/ζ reduces ethanol consumption and blocks ethanol-induced conditioned place preference (CPP) in wild type mice. Since PTN-knockout (Ptn-/-) mice are more sensitive to the conditioning effects of alcohol, we aimed to test the effects of MY10, a small-molecule inhibitor of RPTPß/ζ, on ethanol-induced CPP in Ptn-/- mice. The data presented here demonstrate for the first time that a regular dose of MY10, known to block ethanol consumption and reward in wild type mice, also blocks the rewarding effects of ethanol in the more vulnerable individuals lacking PTN, the endogenous inhibitor of RPTPß/ζ. In addition, since MY10 readily penetrates the blood brain barrier (BBB), we tested its effects in a series of behavioural tests in Ptn+/+ and Ptn-/- mice. The data indicate that MY10 does not cause gross behavioural effects in wild type mice. However, MY10 tended to induce anxiolytic effects in Ptn-/- mice in the elevated plus maze paradigm. Overall, the data indicate that MY10 rescues Ptn-/- mice from their increased susceptibility to the conditioning effects of ethanol and may induce anxiolytic effects in individuals with reduced or absent PTN functions. Further studies are needed to confirm the potential of pharmacological inhibition of RPTPß/ζ as a new therapeutic strategy in the treatment of anxiety-related disorders.


Subject(s)
Alcohol Drinking/metabolism , Conditioning, Classical/drug effects , Receptor-Like Protein Tyrosine Phosphatases, Class 5/antagonists & inhibitors , Animals , Carrier Proteins/genetics , Carrier Proteins/metabolism , Cytokines/genetics , Cytokines/metabolism , Ethanol/metabolism , Ethanol/pharmacology , Inhibition, Psychological , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Midkine/genetics , Midkine/metabolism , Nerve Growth Factors/metabolism , Prefrontal Cortex/drug effects , Prefrontal Cortex/metabolism , Receptor-Like Protein Tyrosine Phosphatases, Class 5/metabolism , Reward , Signal Transduction/drug effects
4.
Mar Drugs ; 16(9)2018 Sep 07.
Article in English | MEDLINE | ID: mdl-30205507

ABSTRACT

The mangrove ecosystem is considered as an attractive biodiversity hotspot that is intensively studied in the hope of discovering new useful chemical scaffolds, including those with potential medicinal application. In the past two decades, mangrove-derived microorganisms, along with mangrove plants, proved to be rich sources of bioactive secondary metabolites as exemplified by the constant rise in the number of publications, which suggests the great potential of this important ecological niche. The present review summarizes selected examples of bioactive compounds either from mangrove endophytes or from soil-derived mangrove fungi and bacteria, covering the literature from 2014 to March 2018. Accordingly, 163 natural products are described in this review, possessing a wide range of potent bioactivities, such as cytotoxic, antibacterial, antifungal, α-glucosidase inhibitory, protein tyrosine phosphatase B inhibitory, and antiviral activities, among others.


Subject(s)
Biological Factors/pharmacology , Drug Discovery , Soil Microbiology , Wetlands , Anti-Bacterial Agents/isolation & purification , Anti-Bacterial Agents/metabolism , Anti-Bacterial Agents/pharmacology , Antifungal Agents/isolation & purification , Antifungal Agents/metabolism , Antifungal Agents/pharmacology , Antineoplastic Agents/isolation & purification , Antineoplastic Agents/metabolism , Antineoplastic Agents/pharmacology , Antiviral Agents/isolation & purification , Antiviral Agents/metabolism , Antiviral Agents/pharmacology , Bacteria/metabolism , Biodiversity , Biological Factors/isolation & purification , Biological Factors/metabolism , Endophytes/metabolism , Fungi/metabolism , Glycoside Hydrolase Inhibitors/isolation & purification , Glycoside Hydrolase Inhibitors/metabolism , Glycoside Hydrolase Inhibitors/pharmacology , Plants/microbiology , Receptor-Like Protein Tyrosine Phosphatases, Class 5/antagonists & inhibitors
5.
Neuropharmacology ; 137: 86-95, 2018 07 15.
Article in English | MEDLINE | ID: mdl-29753117

ABSTRACT

Pleiotrophin (PTN) and Midkine (MK) are neurotrophic factors that are upregulated in the prefrontal cortex after alcohol administration and have been shown to reduce ethanol drinking and reward. PTN and MK are the endogenous inhibitors of Receptor Protein Tyrosine Phosphatase (RPTP) ß/ζ (a.k.a. PTPRZ1, RPTPß, PTPζ), suggesting a potential role for this phosphatase in the regulation of alcohol effects. To determine if RPTPß/ζ regulates ethanol consumption, we treated mice with recently developed small-molecule inhibitors of RPTPß/ζ (MY10, MY33-3) before testing them for binge-like drinking using the drinking in the dark protocol. Mice treated with RPTPß/ζ inhibitors, particularly with MY10, drank less ethanol than controls. MY10 treatment blocked ethanol conditioned place preference, showed limited effects on ethanol-induced ataxia, and potentiated the sedative effects of ethanol. We also tested whether RPTPß/ζ is involved in ethanol signaling pathways. We found that ethanol treatment of neuroblastoma cells increased phosphorylation of anaplastic lymphoma kinase (ALK) and TrkA, known substrates of RPTPß/ζ. Treatment of neuroblastoma cells with MY10 or MY33-3 also increased levels of phosphorylated ALK and TrkA. However, concomitant treatment of neuroblastoma cells with ethanol and MY10 or MY33-3 prevented the increase in pTrkA and pALK. These results demonstrate for the first time that ethanol engages TrkA signaling and that RPTPß/ζ modulates signaling pathways activated by alcohol and behavioral responses to this drug. The data support the hypothesis that RPTPß/ζ might be a novel target of pharmacotherapy for reducing excessive alcohol consumption.


Subject(s)
Binge Drinking/enzymology , Central Nervous System Depressants/pharmacology , Ethanol/pharmacology , Receptor-Like Protein Tyrosine Phosphatases, Class 5/antagonists & inhibitors , Alcohol Deterrents/chemical synthesis , Alcohol Deterrents/chemistry , Alcohol Deterrents/pharmacology , Anaplastic Lymphoma Kinase/metabolism , Animals , Binge Drinking/drug therapy , Cell Line, Tumor , Enzyme Inhibitors/chemical synthesis , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/pharmacology , Humans , Male , Mice, Inbred C57BL , Receptor, trkA/metabolism , Receptor-Like Protein Tyrosine Phosphatases, Class 5/metabolism
6.
Eur J Med Chem ; 144: 318-329, 2018 Jan 20.
Article in English | MEDLINE | ID: mdl-29275231

ABSTRACT

A new series of blood-brain barrier permeable molecules designed to mimic the activity of Pleiotrophin in the CNS has been designed and synthesized. These compounds exert their action by interacting with the intracellular domain PD1 of the Protein Tyrosine-Phosphatase Receptor Z1 (PTPRZ1), and inhibiting its tyrosine phosphatase activity. The most potent compounds 10a and 12b (IC50 = 0,1 µM) significantly increase the phosphorylation of key tyrosine residues of PTPRZ1 substrates involved in neuronal survival and differentiation, and display protective effects against amphetamine-induced toxicity. Docking and molecular dynamics experiments have been used to analyze the binding mode and to explain the observed selectivity against PTP1B. An In vivo experiment has demonstrated that 10a can cross the BBB, thus promoting the possibility of moving forward these candidates for the development of drugs for the treatment of CNS disorders, such as drug addiction and neurodegenerative diseases.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Carrier Proteins/pharmacology , Central Nervous System Diseases/drug therapy , Cytokines/pharmacology , Enzyme Inhibitors/pharmacology , Receptor-Like Protein Tyrosine Phosphatases, Class 5/antagonists & inhibitors , Animals , Anti-Inflammatory Agents, Non-Steroidal/chemical synthesis , Anti-Inflammatory Agents, Non-Steroidal/chemistry , Blood-Brain Barrier/drug effects , Blood-Brain Barrier/metabolism , Carrier Proteins/chemical synthesis , Carrier Proteins/chemistry , Cell Line , Cell Survival/drug effects , Central Nervous System Diseases/metabolism , Cytokines/chemical synthesis , Cytokines/chemistry , Dose-Response Relationship, Drug , Enzyme Inhibitors/chemical synthesis , Enzyme Inhibitors/chemistry , Humans , Mice , Models, Molecular , Molecular Structure , Rats , Receptor-Like Protein Tyrosine Phosphatases, Class 5/metabolism , Structure-Activity Relationship
7.
Sci Rep ; 7(1): 5609, 2017 07 17.
Article in English | MEDLINE | ID: mdl-28717188

ABSTRACT

The R5 subfamily of receptor-type protein tyrosine phosphatases (RPTPs) comprises PTPRZ and PTPRG. A recent study on primary human glioblastomas suggested a close association between PTPRZ1 (human PTPRZ) expression and cancer stemness. However, the functional roles of PTPRZ activity in glioma stem cells have remained unclear. In the present study, we found that sphere-forming cells from the rat C6 and human U251 glioblastoma cell lines showed high expression levels of PTPRZ-B, the short receptor isoform of PTPRZ. Stable PTPRZ knockdown altered the expression levels of stem cell transcription factors such as SOX2, OLIG2, and POU3F2 and decreased the sphere-forming abilities of these cells. Suppressive effects on the cancer stem-like properties of the cells were also observed following the knockdown of PTPRG. Here, we identified NAZ2329, a cell-permeable small molecule that allosterically inhibits both PTPRZ and PTPRG. NAZ2329 reduced the expression of SOX2 in C6 and U251 cells and abrogated the sphere-forming abilities of these cells. Tumor growth in the C6 xenograft mouse model was significantly slower with the co-treatment of NAZ2329 with temozolomide, an alkylating agent, than with the individual treatments. These results indicate that pharmacological inhibition of R5 RPTPs is a promising strategy for the treatment of malignant gliomas.


Subject(s)
Carcinogenesis/drug effects , Enzyme Inhibitors/pharmacology , Glioblastoma/prevention & control , Neoplastic Stem Cells/drug effects , Receptor-Like Protein Tyrosine Phosphatases, Class 5/antagonists & inhibitors , Small Molecule Libraries/pharmacology , Temozolomide/pharmacology , Animals , Antineoplastic Agents, Alkylating/pharmacology , Carcinogenesis/metabolism , Carcinogenesis/pathology , Female , Glioblastoma/enzymology , Glioblastoma/pathology , Humans , Mice , Mice, Inbred BALB C , Mice, Nude , Neoplastic Stem Cells/enzymology , Neoplastic Stem Cells/pathology , Rats , Receptor-Like Protein Tyrosine Phosphatases, Class 5/metabolism , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
8.
Sci Rep ; 6: 20473, 2016 Feb 09.
Article in English | MEDLINE | ID: mdl-26857455

ABSTRACT

Protein tyrosine phosphatase receptor-type Z (PTPRZ) is aberrantly over-expressed in glioblastoma and a causative factor for its malignancy. However, small molecules that selectively inhibit the catalytic activity of PTPRZ have not been discovered. We herein performed an in vitro screening of a chemical library, and identified SCB4380 as the first potent inhibitor for PTPRZ. The stoichiometric binding of SCB4380 to the catalytic pocket was demonstrated by biochemical and mass spectrometric analyses. We determined the crystal structure of the catalytic domain of PTPRZ, and the structural basis of the binding of SCB4380 elucidated by a molecular docking method was validated by site-directed mutagenesis studies. The intracellular delivery of SCB4380 by liposome carriers inhibited PTPRZ activity in C6 glioblastoma cells, and thereby suppressed their migration and proliferation in vitro and tumor growth in a rat allograft model. Therefore, selective inhibition of PTPRZ represents a promising approach for glioma therapy.


Subject(s)
Enzyme Inhibitors , Glioblastoma , Molecular Docking Simulation , Neoplasm Proteins , Neoplasms, Experimental , Receptor-Like Protein Tyrosine Phosphatases, Class 5 , Animals , Cell Line, Tumor , Crystallography, X-Ray , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/pharmacology , Glioblastoma/drug therapy , Glioblastoma/enzymology , Glioblastoma/genetics , Male , Mutagenesis, Site-Directed , Neoplasm Proteins/antagonists & inhibitors , Neoplasm Proteins/chemistry , Neoplasm Proteins/genetics , Neoplasm Proteins/metabolism , Neoplasms, Experimental/drug therapy , Neoplasms, Experimental/enzymology , Neoplasms, Experimental/genetics , Rats , Rats, Wistar , Receptor-Like Protein Tyrosine Phosphatases, Class 5/antagonists & inhibitors , Receptor-Like Protein Tyrosine Phosphatases, Class 5/chemistry , Receptor-Like Protein Tyrosine Phosphatases, Class 5/genetics , Receptor-Like Protein Tyrosine Phosphatases, Class 5/metabolism
9.
Int J Cancer ; 135(5): 1101-9, 2014 Sep 01.
Article in English | MEDLINE | ID: mdl-24496747

ABSTRACT

While the cytogenetic and genetic characteristics of childhood acute lymphoblastic leukemias (ALL) are well studied, less clearly understood are the contributing epigenetic mechanisms that influence the leukemia phenotype. Our previous studies and others identified gene mutation (RAS) and DNA methylation (FHIT) to be associated with the most common cytogenetic subgroup of childhood ALL, high hyperdiploidy (having five more chromosomes). We screened DNA methylation profiles, using a genome-wide high-dimension platform of 166 childhood ALLs and 6 normal pre-B cell samples and observed a strong association of DNA methylation status at the PTPRG locus in human samples with levels of PTPRG gene expression as well as with RAS gene mutation status. In the 293 cell line, we found that PTPRG expression induces dephosphorylation of ERK, a downstream RAS target that may be critical for mutant RAS-induced cell growth. In addition, PTPRG expression is upregulated by RAS activation under DNA hypomethylating conditions. An element within the PTPRG promoter is bound by the RAS-responsive transcription factor RREB1, also under hypomethylating conditions. In conclusion, we provide evidence that DNA methylation of the PTPRG gene is a complementary event in oncogenesis induced by RAS mutations. Evidence for additional roles for PTPR family member genes is also suggested. This provides a potential therapeutic target for RAS-related leukemias as well as insight into childhood ALL etiology and pathophysiology.


Subject(s)
DNA Methylation/genetics , Gene Expression Regulation, Leukemic , Precursor Cell Lymphoblastic Leukemia-Lymphoma/genetics , Receptor-Like Protein Tyrosine Phosphatases, Class 5/genetics , Transcriptional Activation/genetics , ras Proteins/genetics , Cell Transformation, Neoplastic/genetics , Child , Child, Preschool , DNA-Binding Proteins/genetics , Enzyme Activation , Epigenesis, Genetic , Extracellular Signal-Regulated MAP Kinases/metabolism , HEK293 Cells , Humans , Mutation , Phosphorylation/genetics , Promoter Regions, Genetic , Receptor-Like Protein Tyrosine Phosphatases, Class 5/antagonists & inhibitors , Receptor-Like Protein Tyrosine Phosphatases, Class 5/biosynthesis , Transcription Factors/genetics , ras Proteins/metabolism
10.
J Med Chem ; 54(19): 6548-62, 2011 Oct 13.
Article in English | MEDLINE | ID: mdl-21882820

ABSTRACT

Protein tyrosine phosphatases (PTPs) catalyze the dephosphorylation of tyrosine residues, a process that involves a conserved tryptophan-proline-aspartate (WPD) loop in catalysis. In previously determined structures of PTPs, the WPD-loop has been observed in either an "open" conformation or a "closed" conformation. In the current work, X-ray structures of the catalytic domain of receptor-like protein tyrosine phosphatase γ (RPTPγ) revealed a ligand-induced "superopen" conformation not previously reported for PTPs. In the superopen conformation, the ligand acts as an apparent competitive inhibitor and binds in a small hydrophobic pocket adjacent to, but distinct from, the active site. In the open and closed WPD-loop conformations of RPTPγ, the side chain of Trp1026 partially occupies this pocket. In the superopen conformation, Trp1026 is displaced allowing a 3,4-dichlorobenzyl substituent to occupy this site. The bound ligand prevents closure of the WPD-loop over the active site and disrupts the catalytic cycle of the enzyme.


Subject(s)
Models, Molecular , Receptor-Like Protein Tyrosine Phosphatases, Class 5/antagonists & inhibitors , Thiophenes/chemistry , Amino Acid Sequence , Catalytic Domain , Crystallography, X-Ray , Humans , Hydrophobic and Hydrophilic Interactions , Ligands , Molecular Sequence Data , Protein Binding , Protein Conformation , Receptor-Like Protein Tyrosine Phosphatases, Class 5/chemistry , Structure-Activity Relationship , Thiophenes/chemical synthesis
11.
J Biomol Screen ; 16(5): 476-85, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21406618

ABSTRACT

Protein tyrosine phosphatase-γ (PTP-γ) is a receptor-like PTP whose biological function is poorly understood. A recent mouse PTP-γ genetic deletion model associated the loss of PTP-γ gene expression with a potential antidepressant phenotype. This led the authors to screen a subset of the Bristol-Myers Squibb (BMS) compound collection to identify selective small-molecule inhibitors of receptor-like PTP-γ (RPTP-γ) for use in evaluating enzyme function in vivo. Here, they report the design of a high-throughput fluorescence resonance energy transfer (FRET) assay based on the Z'-LYTE technology to screen for inhibitors of RPTP-γ. A subset of the BMS diverse compound collection was screened and several compounds identified as RPTP-γ inhibitors in the assay. After chemical triage and clustering, compounds were assessed for potency and selectivity by IC(50) determination with RPTP-γ and two other phosphatases, PTP-1B and CD45. One hundred twenty-nine RPTP-γ selective (defined as IC(50) value greater than 5- to 10-fold over PTP-1B and CD45) inhibitors were identified and prioritized for evaluation. One of these hits, 3-(3, 4-dichlorobenzylthio) thiophene-2-carboxylic acid, was the primary chemotype for the initiation of a medicinal chemistry program.


Subject(s)
Drug Discovery/methods , Enzyme Inhibitors/metabolism , High-Throughput Screening Assays , Receptor-Like Protein Tyrosine Phosphatases, Class 5/antagonists & inhibitors , Dimethyl Sulfoxide/pharmacology , Enzyme Inhibitors/chemistry , Enzyme Stability/drug effects , Reproducibility of Results , Research Design , Sensitivity and Specificity , Solvents/pharmacology
12.
Curr Pharm Des ; 17(5): 449-61, 2011.
Article in English | MEDLINE | ID: mdl-21375485

ABSTRACT

Parkinson´s disease (PD) is generally a sporadic disease, and only a small proportion of cases have a clear genetic component. During the last few years, a possible specific cause triggering death of dopaminergic neurons in the substantia nigra, drug of abuse-induced neurotoxicity, is being considered as a potential mechanism to develop PD, especially in the case of abuse of amphetamine and its derivatives. Recent evidences have shown pleiotrophin, a growth factor with important functions in remodeling and repair of injured neural tissue, as an important factor involved in the pathogenesis of both diseases by preventing neurodegeneration in Parkinson's disease, neurotoxicity induced by drug abuse and by its ability to modulate drugs addictive effects. This review discusses targeting growth factors such as glial-derived neurotrophic factor (GDNF) and brain-derived neurotrophic factor (BDNF) to treat Parkinson's disease and/or drug addiction and compiles recent evidences to propose the pleiotrophin/receptor protein tyrosine phosphatase ß/ζ signaling pathway as a new therapeutic target to treat Parkinson's disease and to prevent drug of abuse-induced neurotoxicity and addictive effects.


Subject(s)
Drug Discovery/methods , Nerve Degeneration/drug therapy , Nerve Growth Factors/therapeutic use , Parkinson Disease/drug therapy , Receptor-Like Protein Tyrosine Phosphatases, Class 5/antagonists & inhibitors , Substance-Related Disorders/drug therapy , Animals , Carrier Proteins/biosynthesis , Carrier Proteins/physiology , Cytokines/biosynthesis , Cytokines/physiology , Disease Models, Animal , Humans , Nerve Degeneration/chemically induced , Nerve Degeneration/complications , Nerve Degeneration/physiopathology , Nerve Growth Factors/pharmacology , Parkinson Disease/complications , Parkinson Disease/physiopathology , Signal Transduction/drug effects , Signal Transduction/physiology , Substance-Related Disorders/complications , Substance-Related Disorders/physiopathology
13.
J Cell Mol Med ; 15(11): 2353-63, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21129151

ABSTRACT

Substantial genetic evidence suggests that chromosome 11q is involved in regulating initiation and progression of malignant melanomas. Mutations of the MEN1 gene, located in chromosome 11q13, predispose individuals to the multiple endocrine neoplasia type 1 (MEN1) familial syndrome. MEN1 patients develop primary malignant melanoma, suggesting a potential link between MEN1 syndrome and development of melanomas, but the precise molecular mechanism is poorly understood. Here we show that the MEN1 gene suppresses malignant phenotypes of melanoma cells through multiple signalling pathways. Ectopic expression of menin, the product of MEN1 gene, significantly inhibited melanoma cell proliferation and migration in vitro and in vivo. The inhibition was partly achieved through suppressing expression of growth factor pleiotrophin (PTN) and receptor protein tyrosine phosphatase (RPTP) ß/ζ, accompanied with the reduced expression of phosphatidylinositol 3-kinase (pI3K) and decreased phosphorylation of focal adhesion kinase (FAK) and extracellular signal regulated kinase (ERK1/2). Interestingly, reduced expression of menin was associated with hypermethylation of the CpG islands of the MEN1 promoter in melanoma cells. Taken together, these findings suggest a previously unappreciated function for menin in suppressing malignant phenotypes of melanomas and unravel a novel mechanism involving in regulating PTN signalling by menin in development and progression of melanomas.


Subject(s)
Carrier Proteins/metabolism , Cytokines/metabolism , Melanoma, Experimental/metabolism , Melanoma/metabolism , Phosphatidylinositol 3-Kinase/metabolism , Proto-Oncogene Proteins/metabolism , Receptor-Like Protein Tyrosine Phosphatases, Class 5/metabolism , Animals , Carrier Proteins/antagonists & inhibitors , Carrier Proteins/biosynthesis , Cell Line, Tumor , Cell Movement , Cell Proliferation , Chromatin Immunoprecipitation , CpG Islands/genetics , Cytokines/antagonists & inhibitors , Cytokines/biosynthesis , Extracellular Signal-Regulated MAP Kinases/metabolism , Female , Focal Adhesion Protein-Tyrosine Kinases/metabolism , Humans , Intercellular Signaling Peptides and Proteins/genetics , Intercellular Signaling Peptides and Proteins/metabolism , Melanoma/pathology , Melanoma, Experimental/pathology , Mice , Mice, Inbred C57BL , Phenotype , Phosphatidylinositol 3-Kinase/biosynthesis , Phosphoinositide-3 Kinase Inhibitors , Phosphorylation , Promoter Regions, Genetic , Receptor-Like Protein Tyrosine Phosphatases, Class 5/antagonists & inhibitors , Receptor-Like Protein Tyrosine Phosphatases, Class 5/biosynthesis , Signal Transduction , Transplantation, Heterologous
14.
Am J Physiol Cell Physiol ; 297(4): C990-1000, 2009 Oct.
Article in English | MEDLINE | ID: mdl-19657061

ABSTRACT

The maxi-anion channel with a large single-channel conductance of >300 pS, and unknown molecular identity, is functionally expressed in a large variety of cell types. The channel is activated by a number of experimental maneuvers such as exposing cells to hypotonic or ischemic stress. The most effective and consistent method of activating it is patch membrane excision. However, the activation mechanism of the maxi-anion channel remains poorly understood at present. In the present study, involvement of phosphorylation/dephosphorylation in excision-induced activation was examined. In mouse mammary fibroblastic C127 cells, activity of the channel was suppressed by intracellular application of Mg-ATP, but not Mg-5'-adenylylimidodiphosphate (AMP-PNP), in a concentration-dependent manner. When a cocktail of broad-spectrum tyrosine phosphatase inhibitors was applied, channel activation was completely abolished, whereas inhibitors of serine/threonine protein phosphatases had no effect. On the other hand, protein tyrosine kinase inhibitors brought the channel out of an inactivated state. In mouse adult skin fibroblasts (MAFs) in primary culture, similar maxi-anion channels were found to be activated on membrane excision, in a manner sensitive to tyrosine phosphatase inhibitors. In MAFs isolated from animals deficient in receptor protein tyrosine phosphatase (RPTP)zeta, activation of the maxi-anion channel was significantly slower and less prominent compared with that observed in wild-type MAFs; however, channel activation was restored by transfection of the RPTPzeta gene. Thus it is concluded that activation of the maxi-anion channel involves protein dephosphorylation mediated by protein tyrosine phosphatases that include RPTPzeta in mouse fibroblasts, but not in C127 cells.


Subject(s)
Ion Channels/metabolism , Tyrosine/metabolism , Adenosine Triphosphate/pharmacology , Adenosine Triphosphate/physiology , Adenylyl Imidodiphosphate/pharmacology , Adenylyl Imidodiphosphate/physiology , Animals , Anions/metabolism , Cell Line , Enzyme Inhibitors/pharmacology , Fibroblasts/metabolism , Ion Channel Gating , Magnesium , Mice , Phosphorylation , Receptor-Like Protein Tyrosine Phosphatases, Class 5/antagonists & inhibitors , Receptor-Like Protein Tyrosine Phosphatases, Class 5/genetics , Receptor-Like Protein Tyrosine Phosphatases, Class 5/physiology , Signal Transduction
15.
Mini Rev Med Chem ; 9(4): 440-7, 2009 Apr.
Article in English | MEDLINE | ID: mdl-19356122

ABSTRACT

This review compiles the scientific basis to propose the pleiotrophin/receptor protein tyrosine phosphatase beta/zeta signaling pathway as a new therapeutic target to prevent drug of abuse-induced toxicity. In addition, potential guidelines are provided for the development of new therapeutic compounds derived from that knowledge. This approach may be relevant since efficient therapeutic strategies are currently lacking in this field, even when drug-induced neurotoxicity seems to underlie the neurodegenerative disorders diagnosed in drug addicts.


Subject(s)
Carrier Proteins/physiology , Carrier Proteins/therapeutic use , Cytokines/physiology , Cytokines/therapeutic use , Neurotoxicity Syndromes/complications , Neurotoxicity Syndromes/drug therapy , Receptor-Like Protein Tyrosine Phosphatases, Class 5/antagonists & inhibitors , Substance-Related Disorders/complications , Animals , Drug Delivery Systems , Drug Discovery , Humans , Illicit Drugs/adverse effects , Signal Transduction/drug effects , Signal Transduction/physiology
16.
Angiogenesis ; 12(1): 25-33, 2009.
Article in English | MEDLINE | ID: mdl-19116766

ABSTRACT

OBJECTIVES: The endothelial cell (EC)-selective receptor tyrosine kinase, Tie2, and its ligands angiopoietin Ang-1 and Ang-2, are essential for blood vessel maintenance and repair. Ang-1 is an agonist of Tie2 receptor activation, whereas Ang-2 is a context-dependent antagonist/agonist. Therefore, we investigated the role of the EC-selective phosphatase, human protein tyrosine phosphatase beta (HPTPbeta), in regulating Tie2 activity. METHODS AND RESULTS: siRNA silencing of HPTPbeta enhanced Ang-1 and Ang-2-induced Tie2 phosphorylation at 10 min (2.5-fold, P < 0.001; and 1.8-fold, P < 0.05, respectively). The cell survival response to Ang-1, but not Ang-2, was enhanced by HPTPbeta silencing as measured by flow cytometry (0.85-fold to 0.66-fold, P < 0.05) and ELISA (0.88-fold to 0.53-fold, P < 0.01). Hypoxia, which upregulated HPTPbeta expression in endothelial cells, impaired Ang-1-induced Tie2 phosphorylation. CONCLUSIONS: These results reveal a novel role for HPTPbeta in modulating Ang-1-Tie2 signaling and endothelial cell survival.


Subject(s)
Angiopoietins/metabolism , Endothelial Cells/enzymology , Receptor, TIE-2/metabolism , Receptor-Like Protein Tyrosine Phosphatases, Class 5/metabolism , Signal Transduction , Angiopoietin-1/pharmacology , Angiopoietin-2/pharmacology , Angiopoietins/pharmacology , Cell Hypoxia/drug effects , Cell Movement/drug effects , Cell Survival/drug effects , Cells, Cultured , Endothelial Cells/cytology , Endothelial Cells/drug effects , Gene Expression Regulation, Enzymologic/drug effects , Humans , Phosphorylation/drug effects , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA, Small Interfering/metabolism , Receptor-Like Protein Tyrosine Phosphatases, Class 5/antagonists & inhibitors , Receptor-Like Protein Tyrosine Phosphatases, Class 5/genetics , Signal Transduction/drug effects , Vanadates/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...