Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 8 de 8
Filter
Add more filters










Database
Language
Publication year range
1.
J Exp Clin Cancer Res ; 38(1): 484, 2019 Dec 05.
Article in English | MEDLINE | ID: mdl-31805999

ABSTRACT

BACKGROUND: Protein tyrosine phosphatase receptor delta (PTPRD) is frequently inactivated in various types of cancers. Here, we explored the underlying mechanism of PTPRD-loss-induced cancer metastasis and investigated an efficient treatment option for PTPRD-inactivated gastric cancers (GCs). METHODS: PTPRD expression was evaluated by immunohistochemistry. Microarray analysis was used to identify differentially expressed genes in PTPRD-inactivated cancer cells. Quantitative reverse transcription (qRT-PCR), western blotting, and/or enzyme-linked immunosorbent assays were used to investigate the PTPRD-CXCL8 axis and the expression of other related genes. An in vitro tube formation assay was performed using HUVECs. The efficacy of metformin was assessed by MTS assay. RESULTS: PTPRD was frequently downregulated in GCs and the loss of PTPRD expression was associated with advanced stage, worse overall survival, and a higher risk of distant metastasis. Microarray analysis revealed a significant increase in CXCL8 expression upon loss of PTPRD. This was validated in various GC cell lines using transient and stable PTPRD knockdown. PTPRD-loss-induced angiogenesis was mediated by CXCL8, and the increase in CXCL8 expression was mediated by both ERK and STAT3 signaling. Thus, specific inhibitors targeting ERK or STAT3 abrogated the corresponding signaling nodes and inhibited PTPRD-loss-induced angiogenesis. Additionally, metformin was found to efficiently inhibit PTPRD-loss-induced angiogenesis, decrease cell viability in PTPRD-inactivated cancers, and reverse the decrease in PTPRD expression. CONCLUSIONS: Thus, the PTPRD-CXCL8 axis may serve as a potential therapeutic target, particularly for the suppression of metastasis in PTPRD-inactivated GCs. Hence, we propose that the therapeutic efficacy of metformin in PTPRD-inactivated cancers should be further investigated.


Subject(s)
Interleukin-8/genetics , Metformin/pharmacology , Receptor-Like Protein Tyrosine Phosphatases, Class 2/genetics , Stomach Neoplasms/blood supply , Cell Line, Tumor , Down-Regulation , Gene Silencing , Humans , Hypoglycemic Agents/pharmacology , Interleukin-8/metabolism , Neoplasm Metastasis , Neovascularization, Pathologic/genetics , Neovascularization, Pathologic/metabolism , Neovascularization, Pathologic/pathology , Receptor-Like Protein Tyrosine Phosphatases, Class 2/biosynthesis , Receptor-Like Protein Tyrosine Phosphatases, Class 2/deficiency , Signal Transduction/drug effects , Stomach Neoplasms/drug therapy , Stomach Neoplasms/genetics , Stomach Neoplasms/pathology , Transfection
2.
Dig Dis Sci ; 64(11): 3164-3172, 2019 11.
Article in English | MEDLINE | ID: mdl-31041642

ABSTRACT

BACKGROUND: The protein tyrosine phosphatase delta (PTPRD) is a tumor suppressor, and its role in gastric cancer (GC) remains poorly understood. METHODS: The expressions of PTPRD were determined based on public data. In addition, the mRNA expressions of PTPRD in the GC samples, and the expressions of PTPRD in the GC cell lines including HGC27, SGC790, and BGC823, and gastric epithelial cell line GES-1 were determined by western blotting and quantitative real-time PCR. Furthermore, PTPRD siRNA was transfected into the HGC27 cell line, and then, cell proliferation, migration, and invasion were evaluated. The activity of signal transducer and activator of transcription 3 (STAT3) pathways in HGC27 cells transfected with PTPRD siRNA was determined by western blotting. RESULTS: PTPRD deletion was found in the GC patients, and this deletion was found to be correlated with poor prognosis in the GC patients. Expression of PTPRD was significantly downregulated in gastric carcinoma specimens and tumor cell lines when compared with those in normal controls. PTPRD also plays a key role in the GC cells proliferation, invasion, and migration. Silencing PTPRD expression by siRNA dramatically promoted GC cells proliferation, invasion, and migration. Mechanism study demonstrated that phosphorylation of STAT3 was inhibited by silencing PTPRD expression and the according changes including inhibition of cell migration and invasion were observed. CONCLUSION: This study supports PTPRD as a tumor suppressor and could be served as a marker for prognostic of GC. Silencing PTPRD could be a potential therapeutic in GC.


Subject(s)
Disease Progression , Receptor-Like Protein Tyrosine Phosphatases, Class 2/biosynthesis , STAT3 Transcription Factor/metabolism , Stomach Neoplasms/metabolism , Adult , Aged , Aged, 80 and over , Biomarkers, Tumor/metabolism , Cell Line, Tumor , Cell Proliferation/physiology , Female , Gene Expression Regulation, Neoplastic , Humans , Male , Middle Aged , Receptor-Like Protein Tyrosine Phosphatases, Class 2/genetics , STAT3 Transcription Factor/genetics , Stomach Neoplasms/mortality , Stomach Neoplasms/pathology , Survival Rate/trends
3.
Biomed Pharmacother ; 109: 991-999, 2019 Jan.
Article in English | MEDLINE | ID: mdl-30551553

ABSTRACT

BACKGROUND: Aberrant expression of miR-532-3p was involved in progression and development of multiple cancers, whereas miR-532-3p has not been reported in hepatocellular carcinoma (HCC). The aim of this study was to elucidate the functions of miR-532-3p in progression of HCC. METHODS: Real-time PCR in HCC tissues and cell lines and database analysis were conducted for detection of the expression of miR-532-3p in HCC. Then, the association of miR-532-3p with clinicopathological features and prognosis of HCC patients were statistically measured. Subsequently, we attempted to observe the effects of miR-532-3p on migration, invasion and proliferation of HCC cells by Wound healing assay, Transwell assays, MTT assay and EdU assay. Furthermore, bioinformatics tools, database analysis, luciferase reporter gene assay and rescue experiments were conducted to explore the target of miR-532-3p in HCC, and to explore whether the target mediated the effects of miR-532-3p on HCC cells. RESULTS: Our findings and data from databases consistently indicated that the miR-532-3p expression level was higher in HCC. In addition, high miR-532-3p expression was found to be closely related to larger tumor size (P = 0.0027), presence of vascular invasion (P = 0.015), and advanced TNM stage (P = 0.015). In addition, experiments in vitro revealed that miR-532-3p promotes migration, invasion and proliferation of HCC cells. Furthermore, receptor protein tyrosine phosphatase T (PTPRT) was identified as the target and mediator of miR-532-3p in HCC cells. CONCLUSION: Our results demonstrate that miR-532-3p, which is frequently up-regulated in HCC, contributes to HCC cells mobility and proliferation through targeting PTPRT.


Subject(s)
Carcinoma, Hepatocellular/metabolism , Disease Progression , Liver Neoplasms/metabolism , MicroRNAs/biosynthesis , Receptor-Like Protein Tyrosine Phosphatases, Class 2/biosynthesis , Adult , Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/pathology , Cell Proliferation/physiology , Female , Hep G2 Cells , Humans , Liver Neoplasms/genetics , Liver Neoplasms/pathology , Male , MicroRNAs/genetics , Middle Aged , Receptor-Like Protein Tyrosine Phosphatases, Class 2/genetics
4.
Histol Histopathol ; 33(11): 1181-1188, 2018 Nov.
Article in English | MEDLINE | ID: mdl-29745967

ABSTRACT

Protein tyrosine phosphatase receptor type S is a tumor suppressor gene, located at chromosome 19p13.3, frequently inactivated through deletions or epigenetic mechanisms in many types of cancers. In this study, we investigate protein tyrosine phosphatase receptor S (PTPRS) expression level, clinicopathological and prognostic significance in 205 cases of esophageal squamous cell carcinoma (ESCC). Paraffin embedded tissue with immunohistochemistry methods was adopted to exam PTPRS expression in ESCC and paired normal esophageal mucosa tissues on Tissue Microarrays (TMAs). The protein tyrosine phosphatase receptor S was significantly down-regulated in ESCC (58.0%) relative to normal tissues (43.9%) (P=0.006). Statistical analysis revealed that reduced PTPRS expression was significantly associated with TNM stage (P=0.013), invasion depth (P<0.001), local lymph node metastasis (P=0.042) and tumor differentiation (P=0.001). Furthermore, Kaplan-Meier survival analysis revealed that low expression of PTPRS significantly correlated with poor survival of ESCC patients (P=0.002). Cox regression analysis confirmed PTPRS expression as an independent predictor of the overall survival of ESCC patients (HR=1.573, P=0.049). The 5-year overall survival rates in patients with high and low PTPRS expression were 50.6% and 37.2%, respectively. PTPRS deficiency is independently associated with shorter survival and increased recurrence in patients. Our data offer convincing evidence that loss of PTPRS expression may predict an aggressive clinical course in ESCC patients. PTPRS may function as a tumor suppressor and play an important role in ESCC growth and metastasis.


Subject(s)
Biomarkers, Tumor/biosynthesis , Esophageal Neoplasms/metabolism , Esophageal Squamous Cell Carcinoma/metabolism , Receptor-Like Protein Tyrosine Phosphatases, Class 2/biosynthesis , Adult , Aged , Esophageal Neoplasms/physiopathology , Esophageal Squamous Cell Carcinoma/physiopathology , Female , Gene Expression Regulation, Neoplastic , Humans , Male , Middle Aged , Prognosis , Receptor-Like Protein Tyrosine Phosphatases, Class 2/genetics
5.
FASEB J ; 31(11): 4935-4945, 2017 11.
Article in English | MEDLINE | ID: mdl-28751527

ABSTRACT

In utero exposure to diesel exhaust air pollution has been associated with increased adult susceptibility to heart failure in mice, but the mechanisms by which this exposure promotes susceptibility to heart failure are poorly understood. To identify the potential transcriptional effects that mediate this susceptibility, we have performed RNA sequencing analysis on adult hearts from mice that were exposed to diesel exhaust in utero and that have subsequently undergone transverse aortic constriction. We identified 3 target genes, Mir133a-2, Ptprf, and Pamr1, which demonstrate dysregulation after exposure and aortic constriction. Examination of expression patterns in human heart tissues indicates a correlation between expression and heart failure. We subsequently assessed DNA methylation modifications at these candidate loci in neonatal cultured cardiac myocytes after in utero exposure to diesel exhaust and found that the promoter for Mir133a-2 is differentially methylated. These target genes in the heart are the first genes to be identified that likely play an important role in mediating adult sensitivity to heart failure. We have also shown a change in DNA methylation within cardiomyocytes as a result of in utero exposure to diesel exhaust.-Goodson, J. M., Weldy, C. S., MacDonald, J. W., Liu, Y., Bammler, T. K., Chien, W.-M., Chin, M. T. In utero exposure to diesel exhaust particulates is associated with an altered cardiac transcriptional response to transverse aortic constriction and altered DNA methylation.


Subject(s)
Aortic Diseases , DNA Methylation/drug effects , Maternal Exposure/adverse effects , Myocardium/metabolism , Prenatal Exposure Delayed Effects/metabolism , Transcription, Genetic/drug effects , Vehicle Emissions/toxicity , Animals , Aortic Diseases/chemically induced , Aortic Diseases/congenital , Aortic Diseases/metabolism , Aortic Diseases/pathology , Female , Gene Expression Regulation/drug effects , Male , Mice , MicroRNAs/biosynthesis , Myocardium/pathology , Pregnancy , Prenatal Exposure Delayed Effects/pathology , Receptor-Like Protein Tyrosine Phosphatases, Class 2/biosynthesis , Serine Endopeptidases/biosynthesis , Serine Proteases
6.
Biochemistry ; 52(22): 3899-912, 2013 Jun 04.
Article in English | MEDLINE | ID: mdl-23663107

ABSTRACT

Ubiquilin-1 is an Alzheimer's disease-associated protein, which is known to modulate amyloid precursor protein (APP) processing, amyloid-ß (Aß) secretion, and presenilin-1 (PS1) accumulation. Here, we aim to elucidate the molecular mechanisms by which full-length transcript variant 1 of ubiquilin-1 (TV1) affects APP processing and γ-secretase function in human neuroblastoma cells stably overexpressing APP (SH-SY5Y-APP751). We found that TV1 overexpression significantly increased the level of APP intracellular domain (AICD) generation. However, there was no increase in the levels of secreted Aß40, Aß42, or total Aß, suggesting that ubiquilin-1 in particular enhances γ-secretase-mediated ε-site cleavage. This is supported by the finding that TV1 also significantly increased the level of intracellular domain generation of another γ-secretase substrate, leukocyte common antigen-related (LAR) phosphatase. However, in these cells, the increase in AICD levels was abolished, suggesting a preference of the γ-secretase for LAR over APP. TV2, another ubiquilin-1 variant that lacks the protein fragment encoded by exon 8, did not increase the level of AICD generation like TV1 did. The subcellular and plasma membrane localization of APP or γ-secretase complex components PS1 and nicastrin was not altered in TV1-overexpressing cells. Moreover, the effects of TV1 were not mediated by altered expression or APP binding of FE65, an adaptor protein thought to regulate AICD generation and stability. These data suggest that ubiquilin-1 modulates γ-secretase-mediated ε-site cleavage and thus may play a role in regulating γ-secretase cleavage of various substrates.


Subject(s)
Amyloid Precursor Protein Secretases/metabolism , Amyloid beta-Protein Precursor/metabolism , Carrier Proteins/physiology , Cell Cycle Proteins/physiology , Adaptor Proteins, Signal Transducing , Amyloid Precursor Protein Secretases/drug effects , Autophagy-Related Proteins , Carrier Proteins/biosynthesis , Cell Cycle Proteins/biosynthesis , Cell Line, Tumor , Humans , Peptide Fragments/biosynthesis , Receptor-Like Protein Tyrosine Phosphatases, Class 2/biosynthesis , Receptor-Like Protein Tyrosine Phosphatases, Class 2/drug effects
7.
Exp Lung Res ; 37(6): 327-43, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21649524

ABSTRACT

In response to injury, airway epithelia utilize an epidermal growth factor (EGF) receptor (EGFR) signaling program to institute repair and restitution. Protein tyrosine phosphatases (PTPs) counterregulate EGFR autophosphorylation and downstream signaling. PTPµ is highly expressed in lung epithelia and can be localized to intercellular junctions where its ectodomain homophilically interacts with PTPµ ectodomain expressed on neighboring cells. We asked whether PTPµ expression might be altered in response to epithelial injury and whether altered PTPµ expression might influence EGFR signaling. In A549 cells, diverse injurious stimuli dramatically reduced PTPµ protein expression. Under basal conditions, small interfering RNA (siRNA)-induced silencing of PTPµ increased EGFR Y992 and Y1068 phosphorylation. In the presence of EGF, PTPµ knockdown increased EGFR Y845, Y992, Y1045, Y1068, Y1086, and Y1173 but not Y1148 phosphorylation. Reduced PTPµ expression increased EGF-stimulated phosphorylation of Y992, a docking site for phospholipase C (PLC)γ(1), activation of PLCγ(1) itself, and increased cell migration in both wounding and chemotaxis assays. In contrast, overexpression of PTPµ decreased EGF-stimulated EGFR Y992 and Y1068 phosphorylation. Therefore, airway epithelial injury profoundly reduces PTPµ expression, and PTPµ depletion selectively increases phosphorylation of specific EGFR tyrosine residues, PLCγ(1) activation, and cell migration, providing a novel mechanism through which epithelial integrity may be restored.


Subject(s)
ErbB Receptors/metabolism , Receptor-Like Protein Tyrosine Phosphatases, Class 2/biosynthesis , Respiratory Mucosa/metabolism , Cell Line , Cell Migration Assays/methods , Epidermal Growth Factor/metabolism , Epithelial Cells/metabolism , Epithelial Cells/pathology , Gene Knockdown Techniques/methods , Humans , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Phospholipase C gamma/metabolism , Phosphorylation , RNA, Small Interfering/genetics , Receptor-Like Protein Tyrosine Phosphatases, Class 2/genetics , Receptor-Like Protein Tyrosine Phosphatases, Class 2/metabolism , Respiratory Mucosa/enzymology , Respiratory Mucosa/pathology , Signal Transduction , Tyrosine/metabolism
8.
Mol Biol Cell ; 21(1): 29-35, 2010 Jan 01.
Article in English | MEDLINE | ID: mdl-19864455

ABSTRACT

Epidermal growth factor receptor (EGFR) signaling pathways promote human keratinocyte survival and proliferation. In contrast, transforming growth factor-beta (TGF-beta) signaling pathways are strongly anti-proliferative. Receptor type protein tyrosine phosphatase-kappa (RPTP-kappa) specifically dephosphorylates EGFR, thereby blocking EGFR-dependent signaling, and inhibiting proliferation. We report here that RPTP-kappa mediates functional integration of EGFR and TGF-beta signaling pathways in human keratinocytes. TGF-beta up-regulates RPTP-kappa mRNA and protein, in a dose and time dependent manner. Induction of RPTP-kappa by TGF-beta significantly decreases basal and EGF-stimulated EGFR tyrosine phosphorylation. shRNA-mediated reduction of TGF-beta-induced RPTP-kappa significantly attenuates the ability of TGF-beta to inhibit proliferation. RPTP-kappa induction is dependent on activation of transcription factors Smad3 and Smad4. Inhibition of TGF-beta receptor kinase completely prevents induction of RPTP-kappa. Chromatin immunoprecipitation assays reveal that TGF-beta stimulates Smad3 and Smad4 binding to RPTP-kappa gene promoter. Smad3/4 binding is localized to an 186-base pair region, which contains a consensus Smad3-binding element. These data describe a novel mechanism of cross-talk between EGFR and TGF-beta pathways, in which RPTP-kappa functions to integrate growth-promoting and growth-inhibiting signaling pathways.


Subject(s)
ErbB Receptors/metabolism , Keratinocytes/drug effects , Keratinocytes/enzymology , Receptor-Like Protein Tyrosine Phosphatases, Class 2/metabolism , Signal Transduction , Transforming Growth Factor beta/metabolism , Adult , Cell Proliferation/drug effects , Enzyme Activation/drug effects , Enzyme Induction/drug effects , ErbB Receptors/antagonists & inhibitors , Gene Knockdown Techniques , Humans , Keratinocytes/cytology , Phosphotyrosine/metabolism , Promoter Regions, Genetic/genetics , Protein Binding/drug effects , RNA, Messenger/genetics , RNA, Messenger/metabolism , Receptor Cross-Talk/drug effects , Receptor-Like Protein Tyrosine Phosphatases, Class 2/biosynthesis , Receptor-Like Protein Tyrosine Phosphatases, Class 2/genetics , Receptors, Transforming Growth Factor beta/metabolism , Signal Transduction/drug effects , Smad Proteins/metabolism , Time Factors , Transforming Growth Factor beta/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...