Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 31
Filter
Add more filters

Publication year range
1.
Biochem Biophys Res Commun ; 553: 126-133, 2021 05 14.
Article in English | MEDLINE | ID: mdl-33770577

ABSTRACT

Circular RNA (circRNA) homeodomain-interacting protein kinase 3 (circ_HIPK3) has recently reported as regulator in spinal cord injury (SCI). The regulatory mechanism of circ_HIPK3 in SCI was further researched in this study. Circ_HIPK3 expression was inhibited by CoCl2 in AGE1.HN cells. The CoCl2-induced cell cycle arrest, cell proliferation inhibition and apoptosis promotion were mitigated by overexpression of circ_HIPK3. Circ_HIPK3 could target miR-222-3p and circ_HIPK3 repressed the CoCl2-induced neuronal cell injury by sponging miR-222-3p. DUSP19 was a target gene of miR-222-3p and circ_HIPK3 affected the expression of DUSP19 via binding to miR-222-3p. The regulation of circ_HIPK3 in CoCl2-induced injury of AGE1.HN cells was associated with the upregulation of DUSP19. Circ_HIPK3 acted as a pathogenic inhibitor in the progression of SCI via the miR-222-3p-mediated DUSP19 upregulation.


Subject(s)
Apoptosis/drug effects , Cobalt/pharmacology , Dual-Specificity Phosphatases/genetics , MicroRNAs/genetics , Neurons/drug effects , Neurons/pathology , RNA, Circular/genetics , Base Sequence , Cell Line , Dual-Specificity Phosphatases/biosynthesis , Dual-Specificity Phosphatases/deficiency , Dual-Specificity Phosphatases/metabolism , Humans , RNA, Circular/deficiency
2.
Biochemistry (Mosc) ; 86(12): 1624-1634, 2021 Dec.
Article in English | MEDLINE | ID: mdl-34937541

ABSTRACT

Esophageal squamous cell carcinoma (ESCC) is a predominant subtype of esophageal cancer (EC) and has a poor prognosis due to its aggressive nature. Accordingly, it is necessary to find novel prognostic biomarkers and therapeutic targets for ESCC. Lysine-specific histone demethylase 1 (LSD1) plays a core role in the regulation of ESCC oncogenesis. However, the detailed mechanism of LSD1-regulated ESCC growth has not been elucidated. This study aims to explore molecular mechanism underlying the LSD1-regulated ESCC's oncogenesis. After LSD1 silencing, we detected differentially expressed genes (DEGs) in human ESCC cell line, TE-1, by transcriptome sequencing. Subsequently, we investigated expression pattern of the selected molecules in the ESCC tissues and cell lines by qRT-PCR and Western blotting. Furthermore, we explored the roles of selected molecules in ESCC using gene silencing and overexpression assays. Transcriptome sequencing showed that the expression of dual specificity phosphatase 4 (DUSP4) in TE-1 was significantly attenuated after the LSD1 silencing. In addition, the DUSP4 mRNA expression level was significantly higher in the ESCC tissues, especially in those derived from patients with invasion or metastasis. Moreover, the DUSP4 expression was positively associated with the LSD1 expression in the ESCC tissues. DUSP4 overexpression promoted proliferation, invasion, and migration of the ESCC cells, while DUSP4 silencing had an opposite effect. DUSP4 overexpression also enhanced tumorigenicity of the ESCC cells in vivo, while DUSP4 silencing inhibited tumor growth. Importantly, inhibition of cell proliferation, invasion, and migration by the LSD1 inhibitor (ZY0511) was reversed by DUSP4 overexpression. Conclusively, we found that LSD1 promotes ESCC's oncogenesis by upregulating DUSP4, the potential therapeutic and diagnostic target in ESCC.


Subject(s)
Carcinogenesis/metabolism , Dual-Specificity Phosphatases/biosynthesis , Esophageal Neoplasms/enzymology , Esophageal Squamous Cell Carcinoma/enzymology , Gene Expression Regulation, Enzymologic , Gene Expression Regulation, Neoplastic , Histone Demethylases/metabolism , Mitogen-Activated Protein Kinase Phosphatases/biosynthesis , Neoplasm Proteins/metabolism , Up-Regulation , Carcinogenesis/genetics , Cell Line, Tumor , Dual-Specificity Phosphatases/genetics , Esophageal Neoplasms/genetics , Esophageal Neoplasms/pathology , Esophageal Squamous Cell Carcinoma/genetics , Esophageal Squamous Cell Carcinoma/pathology , Female , Histone Demethylases/genetics , Humans , Male , Mitogen-Activated Protein Kinase Phosphatases/genetics , Neoplasm Proteins/genetics
3.
J Cell Biochem ; 120(4): 5666-5676, 2019 04.
Article in English | MEDLINE | ID: mdl-30335891

ABSTRACT

Breast cancer is one of the most prevalent cancers in women. Triple-negative breast cancer consists 15% to 20% of breast cancer cases and has a poor prognosis. Cancerous transformation has several causes one of which is dysregulation of microRNAs (miRNAs) expression. Exosomes can transfer miRNAs to neighboring and distant cells. Thus, exosomal miRNAs can transfer cancerous phenotype to distant cells. We used gene expression omnibus (GEO) datasets and miRNA target prediction tools to find overexpressed miRNA in breast cancer cells and their target genes, respectively. Exosomes were extracted from MDA-MB-231 and MCF-7 cells and characterized. Overexpression of the miRNAs of MDA-MB-231 cells and their exosomes were analyzed using quantitative Real-time PCR. The target genes expression was also evaluated in the cell lines. Luciferase assay was performed to confirm the miRNAs: mRNAs interactions. Finally, MCF-7 cells were treated with MDA-MB-231 cells' exosomes. The target genes expression was evaluated in the recipient cells. GSE60714 results indicated that miR-9 and miR-155 were among the overexpressed miRNAs in highly metastatic triple negative breast cancer cells and their exosomes. Bioinformatic studies showed that these two miRNAs target PTEN and DUSP14 tumor suppressor genes. Quantitative Real-time PCR confirmed the overexpression of the miRNAs and downregulation of their targets. Luciferase assay confirmed that the miRNAs target PTEN and DUSP14. Treatment of MCF-7 cells with MDA-MB-231 cells' exosomes resulted in target genes downregulation in MCF-7 cells. We found that miR-9 and miR-155 were enriched in metastatic breast cancer exosomes. Therefore, exosomal miRNAs can transfer from cancer cells to other cells and can suppress their target genes in the recipient cells.


Subject(s)
Breast Neoplasms/metabolism , Dual-Specificity Phosphatases/biosynthesis , Exosomes/metabolism , MicroRNAs/metabolism , Mitogen-Activated Protein Kinase Phosphatases/biosynthesis , PTEN Phosphohydrolase/biosynthesis , RNA, Neoplasm/metabolism , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Dual-Specificity Phosphatases/genetics , Exosomes/genetics , Exosomes/pathology , Female , Humans , MCF-7 Cells , MicroRNAs/genetics , Mitogen-Activated Protein Kinase Phosphatases/genetics , Neoplasm Metastasis , PTEN Phosphohydrolase/genetics , RNA, Neoplasm/genetics
4.
PLoS Genet ; 12(4): e1005970, 2016 Apr.
Article in English | MEDLINE | ID: mdl-27070551

ABSTRACT

Oocyte meiotic progression and maternal-to-zygote transition are accompanied by dynamic epigenetic changes. The functional significance of these changes and the key epigenetic regulators involved are largely unknown. Here we show that Setdb1, a lysine methyltransferase, controls the global level of histone H3 lysine 9 di-methyl (H3K9me2) mark in growing oocytes. Conditional deletion of Setdb1 in developing oocytes leads to meiotic arrest at the germinal vesicle and meiosis I stages, resulting in substantially fewer mature eggs. Embryos derived from these eggs exhibit severe defects in cell cycle progression, progressive delays in preimplantation development, and degeneration before reaching the blastocyst stage. Rescue experiments by expressing wild-type or inactive Setdb1 in Setdb1-deficient oocytes suggest that the catalytic activity of Setdb1 is essential for meiotic progression and early embryogenesis. Mechanistically, up-regulation of Cdc14b, a dual-specificity phosphatase that inhibits meiotic progression, greatly contributes to the meiotic arrest phenotype. Setdb1 deficiency also leads to derepression of transposons and increased DNA damage in oocytes, which likely also contribute to meiotic defects. Thus, Setdb1 is a maternal-effect gene that controls meiotic progression and is essential for early embryogenesis. Our results uncover an important link between the epigenetic machinery and the major signaling pathway governing meiotic progression.


Subject(s)
Blastocyst/cytology , Dual-Specificity Phosphatases/biosynthesis , Histone-Lysine N-Methyltransferase/genetics , Histones/metabolism , Meiosis/genetics , Animals , Cell Cycle/genetics , DNA Damage/genetics , Dual-Specificity Phosphatases/genetics , Embryonic Development/genetics , Female , Methylation , Mice , Mice, Inbred C57BL , Mice, Knockout , Oocytes/cytology , Oocytes/growth & development , Oocytes/metabolism , RNA Interference , RNA, Small Interfering , Retroelements/genetics
5.
Exp Mol Pathol ; 105(3): 272-278, 2018 12.
Article in English | MEDLINE | ID: mdl-30171833

ABSTRACT

Regulation of growth and differentiation of neuroblastoma (NB) cells is the rational of some maintenance therapies for high-risk NB. MAP kinase phosphatases (MKPs) are potential physiologic regulators of neuronal differentiation and survival, but their expression patterns in NB are scarcely known. Here, an expression analysis of the MKP family has been performed using human NB tumor samples and human NB cell lines (SH-SY5Y, SMS-KCNR, and IMR-32) undergoing retinoic acid (RA)-induced differentiation or subjected to stimuli that activate the MAPK ERK1/2 pathway. We have identified candidate MKPs that could modulate differentiation and growth of NB cells. pERK1/2 high expression correlated with high expression of the MKP DUSP5 in NB tumors, and was associated with poor prognosis. ERK1/2 activation on SH-SY5Y cells was accompanied by increased cell proliferation, and correlated with the expression levels of DUSP5. Accordingly, siRNA knock-down of DUSP5 augmented proliferation of SH-SY5Y cells. Our findings provide insights into the dynamic expression of MKPs in NB cells, disclose DUSP5 as a potential marker of NB poor prognosis, and suggest a role for DUSP5 in limiting ERK1/2-mediated NB proliferation.


Subject(s)
Biomarkers, Tumor/analysis , Dual-Specificity Phosphatases/biosynthesis , Neuroblastoma/pathology , Child , Child, Preschool , Female , Humans , Infant , Male , Neuroblastoma/metabolism , Neuroblastoma/mortality , Prognosis
6.
J Neurochem ; 137(5): 770-81, 2016 06.
Article in English | MEDLINE | ID: mdl-26924229

ABSTRACT

Amyloid beta peptide (Aß) is a pathological hallmark of Alzheimer's disease (AD) and is generated through the sequential cleavage of amyloid precursor protein (APP) by ß- and γ-secretases. Hypoxia is a known risk factor for AD and stimulates Aß generation by γ-secretase; however, the underlying mechanisms remain unclear. In this study, we showed that dual-specificity phosphatase 26 (DUSP26) regulates Aß generation through changes in subcellular localization of the γ-secretase complex and its substrate C99 under hypoxic conditions. DUSP26 was identified as a novel γ-secretase regulator from a genome-wide functional screen using a cDNA expression library. The phosphatase activity of DUSP26 was required for the increase in Aß42 generation through γ-secretase, but this regulation did not affect the amount of the γ-secretase complex. Interestingly, DUSP26 induced the accumulation of C99 in the axons by stimulating anterograde transport of C99-positive vesicles. Additionally, DUSP26 induced c-Jun N-terminal kinase (JNK) activation for APP processing and axonal transport of C99. Under hypoxic conditions, DUSP26 expression levels were elevated together with JNK activation, and treatment with JNK inhibitor SP600125, or the DUSP26 inhibitor NSC-87877, reduced hypoxia-induced Aß generation by diminishing vesicle trafficking of C99 to the axons. Finally, we observed enhanced DUSP26 expression and JNK activation in the hippocampus of AD patients. Our results suggest that DUSP26 mediates hypoxia-induced Aß generation through JNK activation, revealing a new regulator of γ-secretase-mediated APP processing under hypoxic conditions. We propose the role of phosphatase dual-specificity phosphatase 26 (DUSP26) in the selective regulation of Aß42 production in neuronal cells under hypoxic stress. Induction of DUSP26 causes JNK-dependent shift in the subcellular localization of γ-secretase and C99 from the cell body to axons for Aß42 generation. These findings provide a new strategy for developing new therapeutic targets to arrest AD progression.


Subject(s)
Amyloid beta-Peptides/biosynthesis , Amyloid beta-Protein Precursor/metabolism , Axonal Transport/physiology , Dual-Specificity Phosphatases/biosynthesis , Dual-Specificity Phosphatases/pharmacology , Mitogen-Activated Protein Kinase Phosphatases/biosynthesis , Mitogen-Activated Protein Kinase Phosphatases/pharmacology , Peptide Fragments/biosynthesis , Alzheimer Disease/metabolism , Axonal Transport/drug effects , Brain/drug effects , Brain/metabolism , Cell Hypoxia/drug effects , Cell Hypoxia/physiology , Cell Line, Tumor , HEK293 Cells , Humans , Organ Culture Techniques
7.
Tumour Biol ; 36(11): 8531-5, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26032091

ABSTRACT

Dual specificity phosphatase 22 (DUSP22) is a novel dual specificity phosphatase that has been demonstrated to be a cancer suppressor gene associated with numerous biological and pathological processes. However, little is known of DUSP22 expression profiling in colorectal cancer and its prognostic value. Our study aims to investigate the role of DUSP22 expression in the prognosis of colorectal cancer. We detected the mRNA expression in 92 paired primary colorectal cancer tissues and the corresponding adjacent normal tissues by using QuantiGenePlex assay. The Friedman test was used to determine the statistical difference of gene expression. Kaplan-Meier survival analysis was performed. Mann-Whitney test and Kruskal-Wallis test were used to conduct data analyses to determine the prognostic value. Statistical significance was set at P < 0.05. In 74 of 92 cases, DUSP22 mRNA was reduced in primary colorectal cancer tissues, compared to the adjacent normal tissues. The mRNA levels of DUSP22 were significantly lower in colorectal cancer tissues than in adjacent normal tissues (0.0290 vs. 0.0658; P < 0.001). Low expression of DUSP22 correlated significantly with large tumor size (P = 0.013). No association was observed between DUSP22 mRNA expression and differentiation, histopathological type, tumor invasion, lymph node metastases, metastases, TNM stage, and Duke's phase (all P > 0.05). Kaplan-Meier analysis indicated that DUSP22 expression had no significant relationship with overall survival in all patients (P > 0.05). Interestingly, low expression level of DUSP22 in stage IV patients had a poor survival measures with a marginal P value (P = 0.07). Reduced DUSP22 expression was found in colorectal cancer specimens. Low expression level of DUSP22 in stage IV patients had a poor survival outcome. Further study is required for the investigation of the role of DUSP22 in colorectal cancer.


Subject(s)
Biomarkers, Tumor/biosynthesis , Colorectal Neoplasms/genetics , Dual-Specificity Phosphatases/biosynthesis , Mitogen-Activated Protein Kinase Phosphatases/biosynthesis , Prognosis , Adult , Aged , Biomarkers, Tumor/genetics , Colorectal Neoplasms/pathology , Dual-Specificity Phosphatases/genetics , Female , Gene Expression Regulation, Neoplastic , Humans , Kaplan-Meier Estimate , Lymphatic Metastasis , Male , Middle Aged , Mitogen-Activated Protein Kinase Phosphatases/genetics , Neoplasm Staging , RNA, Messenger/biosynthesis , RNA, Messenger/genetics
8.
Breast Cancer Res Treat ; 148(1): 211-20, 2014 Nov.
Article in English | MEDLINE | ID: mdl-25281216

ABSTRACT

Triple-negative breast cancer (TNBC) is an aggressive cancer with limited treatment options. Dual specificity phosphatase 4 (DUSP4) has recently been suggested as a potential marker of chemotherapy resistance for TNBC. DUSP4 gene expression levels were measured in breast cancer tissue from 469 TNBC patients aged 20-75 years who participated in the Shanghai Breast Cancer Survival Study, and their association with recurrence/breast cancer mortality and total mortality was evaluated. Information on breast cancer diagnosis, treatment, and disease progression was collected via medical chart review and multiple in-person follow-up surveys. A Cox regression model was applied in the data analyses. Over a median follow-up of 5.3 years (range: 0.7-8.9 years), 100 deaths and 92 recurrences/breast cancer deaths were documented. Expression levels of transcript variant 1 (NM_001394) and transcript variant 2 (NM_057158) of the DUSP4 gene were studied and were highly correlated (r = 0.76). Low DUSP4 expression levels, particularly of variant 1, were associated with both increased recurrence/breast cancer mortality and increased overall mortality. Hazard ratios with adjustment for age at diagnosis and TNM stage associated with below versus above the median expression level were 1.97 (95 % confidence interval (CI): 1.27-3.05) for recurrence/breast cancer mortality and 2.09 (95 % CI: 1.38-3.17) for overall mortality. Additional adjustment for expression levels of MKI67 and TP53, common treatment types, breast cancer subtype, and grade did not materially alter the observed associations. Low DUSP4 expression levels predict recurrence and mortality in TNBC patients independently from known clinical and molecular predictors.


Subject(s)
Biomarkers, Tumor/genetics , Dual-Specificity Phosphatases/biosynthesis , Mitogen-Activated Protein Kinase Phosphatases/biosynthesis , Neoplasm Recurrence, Local/genetics , Triple Negative Breast Neoplasms/genetics , Adult , Aged , Dual-Specificity Phosphatases/analysis , Dual-Specificity Phosphatases/genetics , Female , Gene Expression Regulation, Neoplastic , Humans , Middle Aged , Mitogen-Activated Protein Kinase Phosphatases/analysis , Mitogen-Activated Protein Kinase Phosphatases/genetics , Neoplasm Recurrence, Local/pathology , Proportional Hazards Models , Transcriptome , Triple Negative Breast Neoplasms/pathology , Young Adult
9.
BMC Biochem ; 15: 27, 2014 Dec 18.
Article in English | MEDLINE | ID: mdl-25519881

ABSTRACT

BACKGROUND: The mitogen-activated protein kinases (MAPKs) pathway is critical for cellular signaling, and proteins such as phosphatases that regulate this pathway are important for normal tissue development. Based on our previous work on dual specificity phosphatase-5 (DUSP5), and its role in embryonic vascular development and disease, we hypothesized that mutations in DUSP5 will affect its function. RESULTS: In this study, we tested this hypothesis by generating full-length glutathione-S-transferase-tagged DUSP5 and serine 147 proline mutant (S147P) proteins from bacteria. Light scattering analysis, circular dichroism, enzymatic assays and molecular modeling approaches have been performed to extensively characterize the protein form and function. We demonstrate that both proteins are active and, interestingly, the S147P protein is hypoactive as compared to the DUSP5 WT protein in two distinct biochemical substrate assays. Furthermore, due to the novel positioning of the S147P mutation, we utilize computational modeling to reconstruct full-length DUSP5 and S147P to predict a possible mechanism for the reduced activity of S147P. CONCLUSION: Taken together, this is the first evidence of the generation and characterization of an active, full-length, mutant DUSP5 protein which will facilitate future structure-function and drug development-based studies.


Subject(s)
Dual-Specificity Phosphatases/biosynthesis , Amino Acid Sequence , Amino Acid Substitution , Catalytic Domain , Dual-Specificity Phosphatases/chemistry , Dual-Specificity Phosphatases/genetics , Extracellular Signal-Regulated MAP Kinases/chemistry , Humans , Molecular Dynamics Simulation , Molecular Sequence Data , Protein Binding , Protein Biosynthesis
10.
Int J Neurosci ; 123(11): 802-9, 2013 Nov.
Article in English | MEDLINE | ID: mdl-23679081

ABSTRACT

The c-Jun N-terminal kinase (JNK) undergoes complete inactivation following the intense activation induced by cerebral ischemia and reperfusion in rat hippocampi. This study examines the molecular mechanism underlying JNK dephosphorylation and inactivation evoked by dual-specificity phosphates following cerebral ischemia. The results revealed upregulation of dual-specificity phosphatase M3/6 (DUSP8) activity at 4 h of reperfusion in rat hippocampi. This was accompanied by the dephosphorylation of JNK. The M3/6 inhibitor, anisomycin, was found to enhance JNK activity following postischemic reperfusion, suggesting that M3/6 is closely associated with JNK inactivation following cerebral ischemia. Cerebral ischemia also induced an increase in heat shock protein (HSP70) levels, which is involved in the upregulation of soluble cytoplasmic M3/6 levels. The inhibition of HSP70 using quercetin resulted in an elevation of JNK activity by decreasing the cytoplasmic solubility of M3/6. The findings of the current study suggest that M3/6 is implicated in the inactivation of JNK in response to cerebral ischemia, which requires the molecular chaperone HSP70 to facilitate the correction of folding defects.


Subject(s)
Brain Ischemia/enzymology , Dual-Specificity Phosphatases/metabolism , Hippocampus/enzymology , JNK Mitogen-Activated Protein Kinases/metabolism , Animals , Anisomycin/pharmacology , Anisomycin/therapeutic use , Brain Ischemia/drug therapy , Dual-Specificity Phosphatases/antagonists & inhibitors , Dual-Specificity Phosphatases/biosynthesis , Hippocampus/drug effects , JNK Mitogen-Activated Protein Kinases/antagonists & inhibitors , Male , Rats , Rats, Sprague-Dawley , Up-Regulation/physiology
11.
J Biol Chem ; 286(41): 35407-35417, 2011 Oct 14.
Article in English | MEDLINE | ID: mdl-21852236

ABSTRACT

Glutathione peroxidase-1 (GPx-1) is a crucial antioxidant enzyme, the deficiency of which promotes atherogenesis. Accordingly, we examined the mechanisms by which GPx-1 deficiency enhances endothelial cell activation and inflammation. In human microvascular endothelial cells, we found that GPx-1 deficiency augments intercellular adhesion molecule-1 (ICAM-1) and vascular cell adhesion molecule-1 (VCAM-1) expression by redox-dependent mechanisms that involve NFκB. Suppression of GPx-1 enhanced TNF-α-induced ROS production and ICAM-1 expression, whereas overexpression of GPx-1 attenuated these TNF-α-mediated responses. GPx-1 deficiency prolonged TNF-α-induced IκBα degradation and activation of ERK1/2 and JNK. JNK or NFκB inhibition attenuated TNF-α induction of ICAM-1 and VCAM-1 expression in GPx-1-deficient and control cells, whereas ERK1/2 inhibition attenuated only VCAM-1 expression. To analyze further signaling pathways involved in GPx-1-mediated protection from TNF-α-induced ROS, we performed microarray analysis of human microvascular endothelial cells treated with TNF-α in the presence and absence of GPx-1. Among the genes whose expression changed significantly, dual specificity phosphatase 4 (DUSP4), encoding an antagonist of MAPK signaling, was down-regulated by GPx-1 suppression. Targeted DUSP4 knockdown enhanced TNF-α-mediated ERK1/2 pathway activation and resulted in increased adhesion molecule expression, indicating that GPx-1 deficiency may augment TNF-α-mediated events, in part, by regulating DUSP4.


Subject(s)
Endothelial Cells/enzymology , Glutathione Peroxidase/metabolism , MAP Kinase Signaling System , Tumor Necrosis Factor-alpha/metabolism , Cells, Cultured , Dual-Specificity Phosphatases/biosynthesis , Dual-Specificity Phosphatases/genetics , Enzyme Activation/genetics , Gene Expression Regulation, Enzymologic/genetics , Gene Knockdown Techniques , Glutathione Peroxidase/genetics , Humans , I-kappa B Kinase/genetics , I-kappa B Kinase/metabolism , Intercellular Adhesion Molecule-1/biosynthesis , Intercellular Adhesion Molecule-1/genetics , Mitogen-Activated Protein Kinase 1/genetics , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/genetics , Mitogen-Activated Protein Kinase 3/metabolism , Mitogen-Activated Protein Kinase Phosphatases/biosynthesis , Mitogen-Activated Protein Kinase Phosphatases/genetics , Oxidation-Reduction , Reactive Oxygen Species/metabolism , Tumor Necrosis Factor-alpha/genetics , Vascular Cell Adhesion Molecule-1/biosynthesis , Vascular Cell Adhesion Molecule-1/genetics , Glutathione Peroxidase GPX1
12.
Breast Cancer Res ; 14(6): 324, 2012 Nov 05.
Article in English | MEDLINE | ID: mdl-23127286

ABSTRACT

Lack of eradication of disseminated breast cancer by chemotherapy is a central clinical problem. Even tumors that show substantial shrinkage after drug treatment frequently relapse and eventually become refractory to all drugs available. The mechanisms underlying this lack of eradication are largely undefined and it is therefore difficult to develop curative strategies using systemic anti-cancer therapy. In a recent article low DUSP4 expression was reported to activate RAS-ERK signaling in residual breast cancer after neoadjuvant chemotherapy. This may be a druggable characteristic because MEK inhibition increases docetaxel sensitivity in a xenograft model.


Subject(s)
Breast Neoplasms/drug therapy , Dual-Specificity Phosphatases/biosynthesis , Mitogen-Activated Protein Kinase Phosphatases/biosynthesis , Mitogen-Activated Protein Kinases/antagonists & inhibitors , Neoplasm Recurrence, Local/prevention & control , Neoplasm, Residual/drug therapy , Antineoplastic Agents/pharmacology , Benzimidazoles/pharmacology , Breast Neoplasms/pathology , Cell Proliferation , Docetaxel , Drug Resistance, Neoplasm , Drug Synergism , Dual-Specificity Phosphatases/genetics , Extracellular Signal-Regulated MAP Kinases/biosynthesis , Extracellular Signal-Regulated MAP Kinases/genetics , Female , Humans , MAP Kinase Signaling System/drug effects , Mitogen-Activated Protein Kinase Phosphatases/genetics , Neoplasm Recurrence, Local/drug therapy , Protein Kinase Inhibitors/therapeutic use , Proto-Oncogene Proteins p21(ras)/biosynthesis , Proto-Oncogene Proteins p21(ras)/genetics , RNA Interference , Taxoids/pharmacology
13.
Biochem Biophys Res Commun ; 420(1): 29-35, 2012 Mar 30.
Article in English | MEDLINE | ID: mdl-22390936

ABSTRACT

Cyclin-dependent kinase inhibitor 3 (CDKN3) belongs to the protein phosphatases family and has a dual function in cell cycling. The function of this gene has been studied in several kinds of cancers, but its role in human hepatocellular carcinoma (HCC) remains to be elucidated. In this study, we found that CDKN3 was frequently overexpressed in both HCC cell lines and clinical samples, and this overexpression was correlated with poor tumor differentiation and advanced tumor stage. Functional studies showed that overexpression of CDKN3 could promote cell proliferation by stimulating G1-S transition but has no impact on cell apoptosis and invasion. Microarray-based co-expression analysis identified a total of 61 genes co-expressed with CDKN3, with most of them involved in cell proliferation, and BIRC5 was located at the center of CDKN3 co-expression network. These results suggest that CDKN3 acts as an oncogene in human hepatocellular carcinoma and antagonism of CDKN3 may be of interest for the treatment of HCC.


Subject(s)
Carcinoma, Hepatocellular/enzymology , Carcinoma, Hepatocellular/pathology , Cyclin-Dependent Kinase Inhibitor Proteins/biosynthesis , Dual-Specificity Phosphatases/biosynthesis , Liver Neoplasms/enzymology , Liver Neoplasms/pathology , Adult , Cell Proliferation , Cyclin-Dependent Kinase Inhibitor Proteins/genetics , Dual-Specificity Phosphatases/genetics , Female , Humans , Male , Middle Aged , Neoplasm Invasiveness , Young Adult
14.
Exp Mol Pathol ; 93(1): 40-9, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22525806

ABSTRACT

Oval cell activation occurs under conditions of severe liver injury when normal hepatocyte proliferation is blocked. Recent studies have shown that a subset of hepatocellular carcinomas expresses oval cell markers, suggesting that these cells are targets of hepatocarcinogens. However, the signaling pathways that control oval cell activation and proliferation are not well characterized. Based on the role of the nutrient signaling kinase complex, mTORC1, in liver development, we investigated the role of this pathway in oval cell activation. Oval cell proliferation was induced in male Fisher rats by a modification of the traditional choline deficient plus ethionine model (CDE) or by 2-acetylaminoflourene treatment followed by 2/3 partial hepatectomy with or without initiation by diethylnitrosamine. To assess the role of mTOR in the oval cell response and development of preneoplastic foci, the effect of the mTORC1 inhibitor, rapamycin, was studied in all models. Rapamycin induced a significant suppression of the oval cell response in both models, an effect that coincided with a decrease in oval cell proliferation. Rapamycin administration did not affect the abundance of neutrophils or natural killer cells in CDE-treated liver or the expression of key cytokines. Gene expression studies revealed the fetal hepatocyte marker MKP-4 to be expressed in oval cells. In an experimental model of hepatic carcinogenesis, rapamycin decreased the size of preneoplastic foci and the rate of cell proliferation within the foci. mTORC1 signaling plays a key role in the oval cell response and in the development of preneoplastic foci. This pathway may be a target for the chemoprevention of hepatocellular carcinoma.


Subject(s)
Antibiotics, Antineoplastic/pharmacology , Hepatocytes/drug effects , Precancerous Conditions/drug therapy , Sirolimus/pharmacology , Animals , Carcinoma, Hepatocellular/prevention & control , Cell Proliferation/drug effects , Cell Shape , Cell Transformation, Neoplastic/drug effects , Cell Transformation, Neoplastic/pathology , Choline Deficiency/metabolism , Diethylnitrosamine/toxicity , Dual-Specificity Phosphatases/biosynthesis , Ethionine/toxicity , Fluorenes/toxicity , Gene Expression Profiling , Hepatectomy/methods , Liver Neoplasms, Experimental/prevention & control , Male , Mechanistic Target of Rapamycin Complex 1 , Mitogen-Activated Protein Kinase Phosphatases/biosynthesis , Multiprotein Complexes , Precancerous Conditions/chemically induced , Precancerous Conditions/pathology , Proteins/metabolism , Rats , TOR Serine-Threonine Kinases
15.
J Cell Physiol ; 226(11): 2841-8, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21302282

ABSTRACT

Most acute coronary events occur in the upstream region of stenotic atherosclerotic plaques that experience laminar shear stress (LSS) elevated above normal physiological levels. Many studies have described the atheroprotective effect on endothelial behavior of normal physiological LSS (approximately 15 dynes/cm(2)) compared to static or oscillatory shear stress (OSS), but it is unknown whether the levels of elevated shear stress imposed by a stenotic plaque would preserve, enhance or reverse this effect. Therefore we used transcriptomics and related functional analyses to compare human endothelial cells exposed to laminar shear stress of 15 (LSS15-normal) or 75 dynes/cm(2) (LSS75-elevated). LSS75 upregulated expression of 145 and downregulated expression of 158 genes more than twofold relative to LSS15. Modulation of the metallothioneins (MT1-G, -M, -X) and NADPH oxidase subunits (NOX2, NOX4, NOX5, and p67phox) accompanied suppression of reactive oxygen species production at LSS75. Shear induced changes in dual specificity phosphatases (DUSPs 1, 5, 8, and 16 increasing and DUSPs 6 and 23 decreasing) were observed as well as reduced ERK1/2 but increased p38 MAP kinase phosphorylation. Amongst vasoactive substances, endothelin-1 expression decreased whereas vasoactive intestinal peptide (VIP) and prostacyclin expression increased, despite which intracellular cAMP levels were reduced. Promoter analysis by rVISTA identified a significant over representation of ATF and Nrf2 transcription factor binding sites in genes upregulated by LSS75 compared to LSS15. In summary, LSS75 induced a specific change in behavior, modifying gene expression, reducing ROS levels, altering MAP kinase signaling and reducing cAMP levels, opening multiple avenues for future study.


Subject(s)
Endothelial Cells/physiology , Shear Strength , Stress, Mechanical , Activating Transcription Factors/metabolism , Binding Sites , Cells, Cultured , Cyclic AMP/biosynthesis , Down-Regulation , Dual-Specificity Phosphatases/biosynthesis , Endothelin-1/biosynthesis , Epoprostenol/biosynthesis , Extracellular Signal-Regulated MAP Kinases/metabolism , Gene Expression Profiling , Humans , Metallothionein/biosynthesis , NADPH Oxidases/biosynthesis , NF-E2-Related Factor 2/metabolism , Reactive Oxygen Species/metabolism , Up-Regulation , Vasoactive Intestinal Peptide/biosynthesis
16.
Blood ; 113(5): 1184-91, 2009 Jan 29.
Article in English | MEDLINE | ID: mdl-18927432

ABSTRACT

Mitogen-activated protein kinases play an integral role in several cellular processes. To regulate mitogen-activated protein kinases, cells express members of a counteracting group of proteins called phosphatases. In this study, we have identified a specific role that one member of this family of phosphatases, dual-specific phosphatase-5 (Dusp-5) plays in vascular development in vivo. We have determined that dusp-5 is expressed in angioblasts and in established vasculature and that it counteracts the function of a serine threonine kinase, Snrk-1, which also plays a functional role in angioblast development. Together, Dusp-5 and Snrk-1 control angioblast populations in the lateral plate mesoderm with Dusp-5 functioning downstream of Snrk-1. Importantly, mutations in dusp-5 and snrk-1 have been identified in affected tissues of patients with vascular anomalies, implicating the Snrk-1-Dusp-5 signaling pathway in human disease.


Subject(s)
Blood Vessels/embryology , Dual-Specificity Phosphatases/biosynthesis , Gene Expression Regulation , Hemangioma/enzymology , Mesoderm/blood supply , Neoplasm Proteins/biosynthesis , Protein Serine-Threonine Kinases/biosynthesis , Zebrafish Proteins/biosynthesis , Zebrafish/embryology , Animals , Blood Vessels/pathology , Dual-Specificity Phosphatases/genetics , Gene Expression Regulation/genetics , Hemangioma/genetics , Hemangioma/pathology , Humans , Mesoderm/embryology , Mesoderm/pathology , Mutation , Neoplasm Proteins/genetics , Protein Serine-Threonine Kinases/genetics , Signal Transduction/genetics , Zebrafish Proteins/genetics
17.
Cell Prolif ; 54(12): e13140, 2021 Dec.
Article in English | MEDLINE | ID: mdl-34647385

ABSTRACT

OBJECTIVE: In this study, we aim to explore the role of bone marrow macrophage-derived exosomes in hepatic insulin resistance, investigate the substance in exosomes that regulates hepatic insulin signalling pathways, reveal the specific molecular mechanisms involved in hepatic insulin resistance and further explore the role of exosomes in type 2 diabetes. MATERIALS AND METHODS: High-fat diet (HFD)-fed mice were used as obesity-induced hepatic insulin resistance model, exosomes were isolated from BMMs which were extracted from HFD-fed mice by ultracentrifugation. Exosomes were analysed the spectral changes of microRNA expression using a microRNA array. The activation of the insulin signalling pathway and the level of glycogenesis were examined in hepatocytes after transfected with miR-143-5p mimics. Luciferase assay and western blot were used to assess the target of miR-143-5p. RESULTS: BMMs from HFD-fed mice were polarized towards M1, and miR-143-5p was significantly upregulated in exosomes of BMMs from HFD-fed mice. Overexpression of miR-143-5p in Hep1-6 cells led to decreased phosphorylation of AKT and GSK and glycogen synthesis. Dual-luciferase reporter assay and western blot demonstrated that mitogen-activated protein kinase phosphatase-5 (Mkp5, also known as Dusp10) was the target gene of miR-143-5p. Moreover, the overexpression of MKP5 could rescue the insulin resistance induced by transfection miR-143-5p mimics in Hep1-6. CONCLUSION: Bone marrow macrophage-derived exosomal miR-143-5p induces insulin resistance in hepatocytes through repressing MKP5.


Subject(s)
Bone Marrow Cells/metabolism , Dual-Specificity Phosphatases/biosynthesis , Gene Expression Regulation, Enzymologic , Hepatocytes/metabolism , Insulin Resistance , Macrophages/metabolism , MicroRNAs/metabolism , Animals , Diet, High-Fat , Exosomes , Mice
18.
Sci Rep ; 10(1): 21918, 2020 12 14.
Article in English | MEDLINE | ID: mdl-33318551

ABSTRACT

Cardiomyocytes of newborn mice proliferate after injury or exposure to growth factors. However, these responses are diminished after postnatal day-6 (P6), representing a barrier to building new cardiac muscle in adults. We have previously shown that exogenous thyroid hormone (T3) stimulates cardiomyocyte proliferation in P2 cardiomyocytes, by activating insulin-like growth factor-1 receptor (IGF-1R)-mediated ERK1/2 signaling. But whether exogenous T3 functions as a mitogen in post-P6 murine hearts is not known. Here, we show that exogenous T3 increases the cardiomyocyte endowment of P8 hearts, but the proliferative response is confined to cardiomyocytes of the left ventricular (LV) apex. Exogenous T3 stimulates proliferative ERK1/2 signaling in apical cardiomyocytes, but not in those of the LV base, which is inhibited by expression of the nuclear phospho-ERK1/2-specific dual-specificity phosphatase, DUSP5. Developmentally, between P7 and P14, DUSP5 expression increases in the myocardium from the LV base to its apex; after this period, it is uniformly expressed throughout the LV. In young adult hearts, exogenous T3 increases cardiomyocyte numbers after DUSP5 depletion, which might be useful for eliciting cardiac regeneration.


Subject(s)
Dual-Specificity Phosphatases/biosynthesis , Gene Expression Regulation, Enzymologic/drug effects , Heart Ventricles/enzymology , Myocardium/enzymology , Myocytes, Cardiac/enzymology , Triiodothyronine/pharmacology , Animals , MAP Kinase Signaling System , Mice , Mice, Transgenic , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism
19.
Neurol Res ; 42(1): 31-38, 2020 Jan.
Article in English | MEDLINE | ID: mdl-31813339

ABSTRACT

Objective: Spinal cord injury (SCI) is a common injury that seriously threatens human health. NF-κB may be involved in the secondary injury of SCI that is mediated by inflammation and aggravates damage. Our study was aimed to investigate the role of NF-κB signaling in DUSP19-mediated cleaved Caspase-3 expression and the release of inflammatory factors in vivo and in vitro.Materials and Methods: DUSP19 mRNA expression and the content of IL-6 and IL-8 in patients with traumatic SCI (TSCI) were measured by real-time PCR and ELISA, respectively. The levels of p-NF-κBp65, NF-κBp65 and cleaved Caspase-3 expression and the concentrations of IL-6 and IL-8 were measured by western blotting and ELISA, respectively.Results: Patients with TSCI showed lower DUSP19 expression and higher concentration of IL-6 and IL-8 compared with healthy controls. DUSP19 overexpression inhibited p-NF-κBp65 level, cleaved Caspase-3 expression, and production of IL-8 and IL-6 in the mice induced by TSCI. DUSP19 silencing increased p-NF-κBp65 level, cleaved Caspase-3 expression, and concentration of IL-6 and IL-8 in mouse primary microglia cells. DUSP19 overexpression had an inverse effect. Importantly, DUSP19 silencing and overexpression mediated p-NF-κBp65 level, cleaved Caspase-3 expression, and concentration of IL-6 and IL-8 in mouse primary microglia cells were reversed by NF-κB inhibitor pyrrolidine dithiocarbamate (PDTC) and NF-κB activator 12-myristate 13-acetate (PMA), respectively.Conclusion: These results suggested that DUSP19-mediated SCI-induced apoptosis and inflammation via NF-κB signaling and might therefore serve as a potential therapeutic target for SCI.


Subject(s)
Apoptosis/physiology , Dual-Specificity Phosphatases/biosynthesis , Microglia/metabolism , NF-kappa B/metabolism , Spinal Cord Injuries/metabolism , Animals , Cells, Cultured , Dual-Specificity Phosphatases/antagonists & inhibitors , Dual-Specificity Phosphatases/genetics , Humans , Inflammation/genetics , Inflammation/metabolism , Inflammation/pathology , Male , Mice , Mice, Inbred C57BL , Microglia/pathology , Prospective Studies , Random Allocation , Signal Transduction/physiology , Spinal Cord Injuries/genetics , Spinal Cord Injuries/pathology
20.
Cancer Biol Ther ; 19(5): 427-435, 2018 05 04.
Article in English | MEDLINE | ID: mdl-29509059

ABSTRACT

OBJECTIVE: To explore the relationship between miR-122-5p and DUSP4 and their effects on gastric cancer (GC) cell mobility and invasiveness. METHODS: Abnormally expressed miRNAs and mRNAs were analyzed using microarrays. The miR-122-5p and DUSP4 mRNA expression levels in GC tissues and cells were determined by RT-qPCR. The target relationship between miR-122-5p and DUSP4 was validated by dual luciferase reporter assay. GC cell mobility and invasiveness were respectively observed by wound healing assay and transwell invasion assay. Western blot and immunohistochemistry were used for detection of the expressions of DUSP4 protein and MMP2 and MMP9 proteins related to cell invasion and migration. The migration and invasion abilities of gastric cancer cells in vivo were evaluated according to the number of lung metastatic nodules in mice. RESULTS: The expression of miR-122-5p in GC tissues and cells was significantly down-regulated, whereas DUSP4 expression was up-regulated. Bioinformatics prediction strategies and dual luciferase reporter assay verified the binding sites of miR-122-5p on 3'UTR of DUSP4 and the target relationship between miR-122-5p and DUSP4. Overexpression of miR-122-5p and knockdown of DUSP4 in BGC-823 cells observantly suppressed GC cell mobility and invasiveness, whereas downregulation of miR-122-5p expression promoted cell metastasis. MiR-122-5p inhibited GC cell mobility and invasiveness and pulmonary tumor metastasis via downregulation of DUSP4. CONCLUSION: MiR-122-5p restrained migration and invasion abilities of GC cells by repressing DUSP4.


Subject(s)
Dual-Specificity Phosphatases/genetics , MicroRNAs/genetics , Mitogen-Activated Protein Kinase Phosphatases/genetics , Stomach Neoplasms/genetics , Cell Movement/genetics , Down-Regulation , Dual-Specificity Phosphatases/biosynthesis , Dual-Specificity Phosphatases/metabolism , HEK293 Cells , Humans , MicroRNAs/biosynthesis , MicroRNAs/metabolism , Mitogen-Activated Protein Kinase Phosphatases/biosynthesis , Mitogen-Activated Protein Kinase Phosphatases/metabolism , Neoplasm Invasiveness , Stomach Neoplasms/metabolism , Stomach Neoplasms/pathology , Transfection
SELECTION OF CITATIONS
SEARCH DETAIL