Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 288
1.
Int J Mol Sci ; 22(22)2021 Nov 11.
Article En | MEDLINE | ID: mdl-34830088

For developmental processes, we know most of the gene networks controlling specific cell responses. We still have to determine how these networks cooperate and how signals become integrated. The JNK pathway is one of the key elements modulating cellular responses during development. Yet, we still know little about how the core components of the pathway interact with additional regulators or how this network modulates cellular responses in the whole organism in homeostasis or during tissue morphogenesis. We have performed a promoter analysis, searching for potential regulatory sequences of puckered (puc) and identified different specific enhancers directing gene expression in different tissues and at different developmental times. Remarkably, some of these domains respond to the JNK activity, but not all. Altogether, these analyses show that puc expression regulation is very complex and that JNK activities participate in non-previously known processes during the development of Drosophila.


Drosophila Proteins , Gene Expression Regulation, Enzymologic , Morphogenesis/genetics , Phosphoprotein Phosphatases , Response Elements , Signal Transduction/genetics , Animals , Drosophila Proteins/biosynthesis , Drosophila Proteins/genetics , Drosophila melanogaster , Phosphoprotein Phosphatases/biosynthesis , Phosphoprotein Phosphatases/genetics
2.
Mol Neurobiol ; 58(7): 3529-3544, 2021 Jul.
Article En | MEDLINE | ID: mdl-33745117

Anxiety is a serious psychiatric disorder, with a higher incidence rate in women than in men. Protein phosphatase Mg2+/Mn2+-dependent 1F (PPM1F), a serine/threonine phosphatase, has been shown to have multiple biological and cellular functions. However, the effects of PPM1F and its neuronal substrates on anxiety remain largely unclear. In this study, we showed that chronic restraint stress (CRS) induced anxiety-related behaviors only in female mice, while acute restraint stress (ARS) produced anxiety-related behaviors in both male and female mice in light-dark and elevated plus maze tests and induced upregulation of PPM1F and downregulation of brain-derived neurotrophic factor (BDNF) expression in the hippocampus. Adeno-associated virus-mediated overexpression of PPM1F or conditional knockout of BDNF in dentate gyrus (DG) led to a more pronounced anxiety-related behavior in female than in male mice as indicated by the behavioral evaluations. Meanwhile, overexpression of PPM1F in the DG decreased total Bdnf exon-specific messenger RNA expression in the hippocampus with the decreased binding activity of phosphorylated H3S10 to its individual promoters in female mice. Furthermore, we identified that overexpression of PPM1F decreased the phosphorylation levels of AKT and JNK in the hippocampus of female mice. These results may suggest that PPM1F regulates anxiety-related behaviors by modulating BDNF expression and H3S10 phosphorylation-mediated epigenetic modification, which may be served as potentially pathological genes associated with anxiety or other mental diseases.


Anxiety/metabolism , Brain-Derived Neurotrophic Factor/biosynthesis , Dentate Gyrus/metabolism , MAP Kinase Signaling System/physiology , Phosphoprotein Phosphatases/biosynthesis , Proto-Oncogene Proteins c-akt/metabolism , Animals , Anxiety/prevention & control , Anxiety/psychology , Brain-Derived Neurotrophic Factor/genetics , Female , Gene Expression , Histones/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Phosphoprotein Phosphatases/genetics , Proto-Oncogene Proteins c-akt/antagonists & inhibitors
3.
Exp Neurol ; 340: 113657, 2021 06.
Article En | MEDLINE | ID: mdl-33639208

Major depressive disorder (MDD) is a common, serious, debilitating mental illness. Protein phosphatase Mg2+/Mn2+-dependent 1F (PPM1F), a serine/threonine phosphatase, has been reported to have multiple biological and cellular functions. However, the effects of PPM1F and its neuronal substrates on depressive behaviors remain largely unknown. Here, we showed that PPM1F is widely distributed in the hippocampus, and chronic unpredictable stress (CUS) can induce increased expression of PPM1F in the hippocampus, which was correlated with depression-associated behaviors. Overexpression of PPM1F mediated by adeno-associated virus (AAV) in the dentate gyrus (DG) produced depression-related behaviors and enhanced susceptibility to subthreshold CUS (SCUS) in both male and female mice, while, knockout of PPM1F in DG produced antidepressant phonotypes under stress conditions. Whole-cell patch-clamp recordings demonstrated that overexpression of PPM1F increased the neuronal excitability of the granule cells in the DG. Consistent with neuronal hyperexcitability, overexpression of PPM1F regulated the expression of certain ion channel genes and induced decreased phosphorylation of Ca2+/calmodulin-dependent protein kinase II (CAMKII) and Adenosine 5'-monophosphate (AMP)-activated protein kinase (AMPK) in hippocampus. These results suggest that PPM1F in the DG regulates depression-related behaviors by modulating neuronal excitability, which might be an important pathological gene for depression or other mental diseases.


Dentate Gyrus/metabolism , Depression/metabolism , Neurons/metabolism , Phosphoprotein Phosphatases/biosynthesis , Animals , Depression/genetics , Depression/psychology , Female , Locomotion/physiology , Male , Mice , Mice, Inbred C57BL , Phosphoprotein Phosphatases/genetics
4.
World J Urol ; 38(11): 2849-2856, 2020 Nov.
Article En | MEDLINE | ID: mdl-31965287

OBJECTIVE: Slingshot homolog-1 (SSH-1) shows an important role in the occurrence and development in various tumors. While, the expression and prognostic implications of SSH-1 in bladder urothelial carcinoma (UC) remain unclear and thus were addressed in this study. METHODS: Immunohistochemistry (IHC) was performed on tissue microarrays composed of 624 bladder UC specimens after transurethral resection of bladder tumor (TUR-BT) to detect SSH-1 expression. The clinic-pathological features were compared between SSH-1( +) and SSH-1(-) subgroups. The Kaplan-Meier curve with log-rank test and univariate/multivariate Cox regression model with stepwise backward elimination methods were performed for survival analyses. RESULTS: In this study, 359 (57.53%) specimens were detected with SSH-1 expression. SSH-1 positivity was significantly associated with higher pathological grade (p = 0.020), lymphovascular invasion (p = 0.006), tumor recurrence (p < 0.001) and progression (p < 0.001) in bladder UC. Besides, SSH-1 positivity predicted a shorter overall survival (OS, p = 0.024), recurrence-free survival (RFS, p < 0.001), progression-free survival (PFS, p = 0.002) and cancer-specific survival (CSS, p = 0.047). Multivariate Cox proportional hazard analysis showed that tumor size (p = 0.007), lymphovascular invasion (p = 0.003), recurrence (p < 0.001), progression (p < 0.001) and SSH-1 expression (p = 0.015) were predictors of poor prognosis in bladder UC patients. CONCLUSIONS: SSH-1 expression was associated with undesirable clinic-pathological characteristics and poor post-operative prognosis in bladder UC patients. SSH-1 might play an important role in bladder UC and serve as a promising predictor of oncological outcomes in patients with bladder UC.


Carcinoma, Transitional Cell/metabolism , Carcinoma, Transitional Cell/surgery , Cystectomy , Phosphoprotein Phosphatases/biosynthesis , Urinary Bladder Neoplasms/metabolism , Urinary Bladder Neoplasms/surgery , Aged , Carcinoma, Transitional Cell/mortality , Carcinoma, Transitional Cell/pathology , Cystectomy/methods , Female , Humans , Male , Middle Aged , Neoplasm Staging , Prognosis , Retrospective Studies , Survival Rate , Urethra , Urinary Bladder Neoplasms/mortality , Urinary Bladder Neoplasms/pathology
5.
Hypertension ; 73(5): e14-e24, 2019 05.
Article En | MEDLINE | ID: mdl-30929511

Regulatory T cells (Treg cells) play important roles in hypertension and organ damages. MicroRNA-31 (miR-31) is a critical regulator for Treg cell generation. However, the role of miR-31 in hypertension has not been elucidated. We aim to study the functionality of miR-31 and the detailed mechanism in Ang II (Angiotensin II)-induced hypertensive mouse model. We found: In vitro, miR-31 expression was higher in T helper 17 cells and lower in Treg cells than that of naïve T cells. The genetic deficiency of miR-31 promoted Treg cell differentiation, whereas no impact on T helper 17 cells differentiation. Ang II-induced hypertension resulted in increased expression of miR-31 in the aorta, splenic CD4+ T cells, and kidney leukocytes. MiR-31 deficiency strikingly decreased systolic blood pressure and diastolic blood pressure and attenuated renal and vascular damage. MiR-31 deletion altered the accumulation of Treg cells and macrophages and expression of inflammatory cytokines in kidneys in Ang II-induced hypertensive mice. Ang II treatment reduced the levels of anti-inflammatory cytokines and increased proinflammatory cytokines in plasma that were blunted by the miR-31 deletion. Ppp6C (protein phosphatase 6c; a direct target of miR-31) specific deletion in Treg cells led to marked impairment of Treg cell induction, increased Ang II-induced blood pressure elevation, and organ damage in mice. In conclusion, we provided novel evidence of miR-31 as an emerging key posttranscriptional regulator of hypertension-associated immunosuppression through targeting ppp6C which is a critical regulator in the differentiation of Treg cells. This study offers new perspectives on miRNA-based therapeutic approaches.


Gene Expression Regulation , Hypertension/genetics , Immunity, Cellular/genetics , MicroRNAs/genetics , Phosphoprotein Phosphatases/genetics , T-Lymphocytes, Regulatory/immunology , Angiotensin II/toxicity , Animals , Cells, Cultured , Disease Models, Animal , Hypertension/chemically induced , Hypertension/immunology , Immunosuppression Therapy , Mice , Mice, Inbred C57BL , Mice, Knockout , MicroRNAs/biosynthesis , Phosphoprotein Phosphatases/biosynthesis , RNA/genetics , T-Lymphocytes, Regulatory/metabolism , T-Lymphocytes, Regulatory/pathology
6.
J Biochem Mol Toxicol ; 33(6): e22314, 2019 Jun.
Article En | MEDLINE | ID: mdl-30811740

The main topic of this study was to investigate the effect of benzo[a]pyrene (BP) on microRNAs and their target genes expression levels in primary cell cultures from normal and malignant endometrial tissue. MicroRNA-126 (miR-126) and miR-190a were most sensitive to BP treatment. The treatment of both cultures with BP was accompanied by a decrease of miR-126 level and an increase of EGFL7 gene expression level. BP-induced upregulation of miR-190a was detected only in normal cells and it was accompanied with decrease of mRNA levels of TP53INP1 and PHLPP1 genes. Taking into account that BP promoted the proliferation of normal cells and amplified apoptosis of cancer cells, it is possible that miR-190a is involved in general cellular response to BP. The findings of this study indicate that miR-190a and its target genes may be involved in the regulation of cell fate under BP treatment.


Benzo(a)pyrene/toxicity , Calcium-Binding Proteins/biosynthesis , Carrier Proteins/biosynthesis , EGF Family of Proteins/biosynthesis , Endometrial Neoplasms/metabolism , Endometrium/metabolism , Gene Expression Regulation, Neoplastic/drug effects , Heat-Shock Proteins/biosynthesis , MicroRNAs/biosynthesis , Neoplasm Proteins/biosynthesis , Nuclear Proteins/biosynthesis , Phosphoprotein Phosphatases/biosynthesis , RNA, Neoplasm/biosynthesis , Endometrial Neoplasms/pathology , Endometrium/pathology , Female , Humans
7.
Proc Natl Acad Sci U S A ; 115(37): 9288-9293, 2018 09 11.
Article En | MEDLINE | ID: mdl-30150402

Th17 cells favor glycolytic metabolism, and pyruvate dehydrogenase (PDH) is the key bifurcation enzyme, which in its active dephosphorylated form advances the oxidative phosphorylation from glycolytic pathway. The transcriptional factor, inducible cAMP early repressor/cAMP response element modulator (ICER/CREM), has been shown to be induced in Th17 cells and to be overexpressed in CD4+ T cells from the patients with systemic lupus erythematosus (SLE). We found that glycolysis and lactate production in in vitro Th17-polarized T cells was reduced and that the expression of pyruvate dehydrogenase phosphatase catalytic subunit 2 (PDP2), an enzyme that converts the inactive PDH to its active form, and PDH enzyme activity were increased in Th17 cells from ICER/CREM-deficient animals. ICER was found to bind to the Pdp2 promoter and suppress its expression. Furthermore, forced expression of PDP2 in CD4+ cells reduced the in vitro Th17 differentiation, whereas shRNA-based suppression of PDP2 expression increased in vitro Th17 differentiation and augmented experimental autoimmune encephalomyelitis. At the translational level, PDP2 expression was decreased in memory Th17 cells from patients with SLE and forced expression of PDP2 in CD4+ T cells from lupus-prone MRL/lpr mice and patients with SLE suppressed Th17 differentiation. These data demonstrate the direct control of energy production during Th17 differentiation in health and disease by the transcription factor ICER/CREM at the PDH metabolism bifurcation level.


Cell Differentiation , Gene Expression Regulation, Enzymologic , Phosphoprotein Phosphatases/biosynthesis , Response Elements , Th17 Cells/enzymology , Animals , Catalytic Domain , Cyclic AMP Response Element Modulator/genetics , Cyclic AMP Response Element Modulator/immunology , Cyclic AMP Response Element Modulator/metabolism , Encephalomyelitis, Autoimmune, Experimental/enzymology , Encephalomyelitis, Autoimmune, Experimental/genetics , Encephalomyelitis, Autoimmune, Experimental/immunology , Encephalomyelitis, Autoimmune, Experimental/pathology , Female , Humans , Lupus Erythematosus, Systemic/enzymology , Lupus Erythematosus, Systemic/genetics , Lupus Erythematosus, Systemic/immunology , Lupus Erythematosus, Systemic/pathology , Male , Mice , Mice, Knockout , Phosphoprotein Phosphatases/genetics , Phosphoprotein Phosphatases/immunology , Th17 Cells/immunology , Th17 Cells/pathology
8.
Anal Biochem ; 549: 99-106, 2018 05 15.
Article En | MEDLINE | ID: mdl-29559334

Casein kinase 1 (CK1) is a widely expressed Ser/Thr kinase in eukaryotic organisms that is involved in various cellular processes (e.g., circadian rhythm and apoptosis). Therefore, preparing highly active CK1 and investigating its properties in vitro have important implications for understanding the biological roles of the kinase. However, recombinant CK1 undergoes autoinactivation via autophosphorylation in Escherichia coli cells and thus is undesirably prepared as a phosphorylated and inactivated kinase. To circumvent this problem, we established a protein expression system using E. coli strain BL21(DE3)pλPP in which λ protein phosphatase (λPPase) is constitutively expressed. Using this system, recombinant CK1 isoforms (α, δ and ε) were readily prepared as unphosphorylated forms. Furthermore, we found that CK1s prepared using BL21(DE3)pλPP showed markedly higher activity than those prepared by the conventional BL21(DE3). Finally, we demonstrated that the kinase activity of CK1δ from BL21(DE3)pλPP was higher than that prepared by a conventional method consisting of troublesome steps such as in vitro λPPase treatment. Thus, this simple method using BL21(DE3)pλPP is valuable for preparing highly active CK1s. It may also be applicable to other kinases that are difficult to prepare because of phosphorylation in E. coli cells.


Bacteriophage lambda/enzymology , Casein Kinase I , Escherichia coli , Gene Expression , Phosphoprotein Phosphatases/biosynthesis , Viral Proteins/biosynthesis , Bacteriophage lambda/genetics , Casein Kinase I/biosynthesis , Casein Kinase I/chemistry , Casein Kinase I/genetics , Casein Kinase I/isolation & purification , Escherichia coli/chemistry , Escherichia coli/enzymology , Escherichia coli/genetics , Humans , Phosphoprotein Phosphatases/genetics , Recombinant Proteins/biosynthesis , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/isolation & purification , Viral Proteins/genetics
9.
Virchows Arch ; 472(5): 727-737, 2018 May.
Article En | MEDLINE | ID: mdl-29352327

Cofilin phospho-regulation is important for actin filament turnover and is implicated in cancer. Phosphorylation of cofilin is mediated by LIM kinases (LIMKs) and dephosphorylation by Slingshot phosphatases (SSH). LIMKs and SSH promote cancer cell invasion and metastasis and represent novel anti-cancer targets. However, little is known regarding LIMK/cofilin and SSH in human colorectal cancer (CRC). In this study, we aimed to address their expression and significance in human CRC. We evaluated expression of non-phosphorylated (active) and phosphorylated cofilin, LIMK1, LIMK2, and SSH1 by immunohistochemistry in 143 human CRC samples in relation to clinicopathologic parameters, response of metastatic disease to chemotherapy, and epithelial-mesenchymal transition (EMT) markers ß-catenin, E-cadherin, and ZEB. We show that active cofilin, LIMK1, LIMK2, and SSH1 are overexpressed in human CRC and are associated with tumor progression parameters. SSH1 is an independent predictor of lymph node metastasis by multivariate analysis. LIMK1 and SSH1 expression is also higher in non-responders to chemotherapy, and SSH1 is shown by multivariate analysis to independently predict response of metastatic disease to chemotherapy. Active cofilin, LIMK1, LIMK2, and SSH1 also correlated with the EMT markers examined. In addition, immunofluorescence analysis showed increased expression of active cofilin, LIMK1, LIMK2, and SSH1 in HT29 colon cancer cells resistant to 5-fluorouracil compared to parental HT29 cells. Our results suggest that F-actin regulators LIMK/cofilin pathway and SSH1 are associated with CRC progression and chemoresistance representing promising tumor biomarkers and therapeutic targets in CRC.


Biomarkers, Tumor/analysis , Colorectal Neoplasms/pathology , Drug Resistance, Neoplasm/physiology , Actin Depolymerizing Factors/analysis , Actin Depolymerizing Factors/biosynthesis , Adult , Aged , Aged, 80 and over , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Colorectal Neoplasms/drug therapy , Disease Progression , Female , Humans , Lim Kinases/analysis , Lim Kinases/biosynthesis , Male , Middle Aged , Phosphoprotein Phosphatases/analysis , Phosphoprotein Phosphatases/biosynthesis , Signal Transduction/physiology
10.
Eur Rev Med Pharmacol Sci ; 21(11): 2617-2625, 2017 06.
Article En | MEDLINE | ID: mdl-28678323

OBJECTIVE: Gastric cancer is a leading cause of cancer deaths and has a poor prognosis after diagnosis. Previous studies showed that Magnesium-Dependent Phosphatase-1 (MDP-1) might be a key component for glycosylation in human protein repair, and an alteration of its function has been involved in some aspects of cellular metabolic networks linked to either normal or pathological processe. In this study, we investigate the MDP-1 status in patients with gastric carcinoma, and determine the potential relationship between MDP-1 and clinical outcome. PATIENTS AND METHODS: One hundred and seventy-one consecutive patients with stage I-III gastric carcinoma who had received a D2 gastrectomy were recruited. The MDP-1 expression was determined by immunohistochemistry (IHC). Disease-free survival (DFS) and overall survival (OS) were evaluated. RESULTS: We generate an IHC score on a continuous scale of 0-7. The IHC cutoff point generated by ROC analysis and the threshold IHC score was 2. Low MDP-1 expression was scored for 61 (35.7%) and high MDP-1 expression for 110 (64.3%) patients. We saw a significant down-regulation of MDP-1 expression in G3-4 and stage III tumor tissue compared with G1-2 and stage I-II tumors, p=0.023 and p=0.047. In univariate survival analysis, high expression of MDP-1 predicted a significantly better DFS (56.0 months vs. 25.0 months, p=0.029) and OS (59.0 months vs. 41.0 months, p=0.043) compared with low expression. In a multivariate analysis, the tumor stage was a significant predictor for DFS and OS even after adjustment for all other covariates. The MDP-1 status was a joint predictor for DFS and OS with a multivariate HR 0.728, 95% CI 0.530-0.999, p=0.049 and a multivariate HR 0.745, 95% CI 0.543-1.022, p=0.068, respectively. CONCLUSIONS: We showed that down-regulation of MDP-1 expression was correlated with poorly differentiated carcinoma and later tumor stage, and it predicted a significantly poorer DFS and OS. Down-regulation of MDP-1 expression was a predictor of a poor prognosis for gastric cancer patients, and it may refer to tumor cells that have lost a protective enzymatic system.


Phosphoprotein Phosphatases/biosynthesis , Stomach Neoplasms/enzymology , Adult , Aged , Disease-Free Survival , Down-Regulation , Female , Gastrectomy , Humans , Immunohistochemistry , Male , Middle Aged , Neoplasm Staging , Prognosis , ROC Curve , Stomach Neoplasms/mortality , Stomach Neoplasms/surgery
11.
Acta Biochim Pol ; 64(1): 11-16, 2017.
Article En | MEDLINE | ID: mdl-28090608

Osteosarcoma is the most common primary malignant bone tumor in adolescents and young adults. However, the involvement of serine/threonine phosphatase type 5 (PP5) in osteosarcoma remains unclear. The aim of this study was to evaluate the functional role of PP5 in osteosarcoma cells. Firstly, we found that PP5 is widely expressed in several human osteosarcoma cell lines. Then we used lentivirus-delivered siRNA to silence PP5 expression in Saos-2 and U2OS cell lines. Knockdown of endogenous PP5 expression by shRNA-expressing lentivirus significantly decreased the viability and proliferation of the osteosarcoma cells. Moreover, FACS analysis showed that knockdown of PP5 expression induced a significant arrest in the G0/G1 phase of the cell cycle, which was associated with the inhibition of cell proliferation. Therefore, knockdown of PP5 is likely to provide a novel alternative to targeted therapy of osteosarcoma and deserves further investigation.


Cell Proliferation/drug effects , Nuclear Proteins/genetics , Osteosarcoma/pathology , Phosphoprotein Phosphatases/genetics , Cell Cycle Checkpoints/drug effects , Cell Line, Tumor , Cell Proliferation/genetics , Gene Silencing/drug effects , Humans , Nuclear Proteins/biosynthesis , Nuclear Proteins/physiology , Osteosarcoma/drug therapy , Osteosarcoma/enzymology , Phosphoprotein Phosphatases/biosynthesis , Phosphoprotein Phosphatases/physiology , RNA, Small Interfering/pharmacology , Resting Phase, Cell Cycle , Up-Regulation
12.
In Vitro Cell Dev Biol Anim ; 53(3): 248-257, 2017 Mar.
Article En | MEDLINE | ID: mdl-27815660

Phosphoglycerate mutase 5 (PGAM5) is a mitochondrial membrane protein that plays crucial roles in necroptosis and apoptosis. Though PGAM5 is known to be required for inducing intrinsic apoptosis through interacting with BCL2 associated X protein (Bax) and dynamin-related protein 1 (Drp1), the expression and role of PGAM5 in cardiomyocyte apoptosis driven by myocardial ischemia/reperfusion injury(MIRI) has not been studied. The present study shows that PGAM5 expression decreased after MIRI in vivo, positively correlated with Bcl-xL expression, negatively correlated with Kelch-ECH associating protein 1 (Keap1) expression. Furthermore, PGAM5 expression also decreased in cardiomyocytes after hypoxia/reoxygenation (H/R) treatment in vitro. PGAM5 silence promoted cardiomyocyte apoptosis and inhibited Bcl-xL expression, but with no effect on Keap1 expression. Accordingly, Keap1 overexpression further inhibited Bcl-xL and PGAM5 expression. Additionally, PGAM5-Bcl-xL-Keap1 interaction was identified, suggesting that PGAM5 might participate in the degradation of Bcl-xL mediated by Keap1. In summary, PGAM5 controls cardiomyocyte apoptosis induced by MIRI through regulating Keap1-mediated Bcl-xL degradation, which may supply a novel molecular target for acute myocardial infarction (AMI) therapy. Graphical abstract ᅟ.


Kelch-Like ECH-Associated Protein 1/biosynthesis , Myocardial Infarction/genetics , Myocardial Reperfusion Injury/genetics , Phosphoprotein Phosphatases/genetics , bcl-X Protein/biosynthesis , Animals , Apoptosis/genetics , Disease Models, Animal , Dynamins/biosynthesis , Humans , Kelch-Like ECH-Associated Protein 1/genetics , Mitochondria/genetics , Mitochondria/metabolism , Myocardial Infarction/pathology , Myocardial Reperfusion Injury/pathology , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/pathology , Necrosis/genetics , Necrosis/pathology , Phosphoprotein Phosphatases/biosynthesis , Rats , bcl-X Protein/genetics
13.
J Proteome Res ; 16(2): 698-711, 2017 02 03.
Article En | MEDLINE | ID: mdl-27976581

Cancer metastasis is a complex mechanism involving multiple processes. Previously, our integrative proteome, transcriptome, and phosphoproteome study reported that the levels of serine/threonine phosphatase POPX2 were positively correlated with cancer cell motility through modulating MAPK signaling. Surprisingly, here we found that POPX2 knockdown cells induced more numerous and larger tumor nodules in lungs in longer term animal studies. Interestingly, our analysis of DNA microarray data from cancer patient samples that are available in public databases shows that low POPX2 expression is linked to distant metastasis and poor survival rate. These observations suggest that lower levels of POPX2 may favor tumor progression in later stages of metastasis. We hypothesize that POPX2 may do so by modulation of angiogenesis. Secretome analysis of POPX2-knockdown MDA-MB-231 cells using LC-MS/MS-based SILAC quantitative proteomics and cytokine array show that silencing of POPX2 leads to increased secretion of exosomes, which may, in turn, induce multiple pro-angiogenic cytokines. This study, combined with our previous findings, suggests that a single ubiquitously expressed phosphatase POPX2 influences cancer metastasis via modulating multiple biological processes including MAPK signaling and exosome cytokine secretion.


Breast Neoplasms/genetics , Phosphoprotein Phosphatases/genetics , Proteome/genetics , Proteomics , Cell Line, Tumor , Female , Gene Expression Regulation, Neoplastic , Gene Knockdown Techniques , Humans , Mitogen-Activated Protein Kinase Kinases/genetics , Phosphoprotein Phosphatases/biosynthesis , Signal Transduction
14.
Oncol Rep ; 36(5): 2926-2934, 2016 Nov.
Article En | MEDLINE | ID: mdl-27599670

The objective of this study was to investigate the effect of silencing gene protein phosphatase 1H (PPM1H) on malignant phenotype of human pancreatic cancer cell line BxPC-3. In order to explore the function of PPM1H in pancreatic cancer cells, real-time PCR and western blotting were used to detect the expression of PPM1H in different pancreatic cancer cell lines. Human pancreatic cancer cell line BxPC-3 was treated with 10 ng/ml TGF-ß1 and 200 ng/ml BMP2 for 72 h, respectively, and the mRNA and protein expression levels of PPM1H and EMT-related markers (E-cadherin, vimentin) were detected by real-time PCR and western blotting, respectively. Using exogenous RNA interference technology to silence the PPM1H gene, the expression of PPM1H and EMT-related markers at mRNA and protein levels were detected by real-time PCR and western blotting. The cell migration and invasion were measured using Transwell assays. Finally, cell counting kit-8 (CCK-8) and flow cytometry were used to determine the effect of PPM1H on cell proliferation and apoptosis of BxPC-3 cells. The expression levels of PPM1H in all of the examined pancreatic cancer cell lines (BxPC-3, MIA-PACA2, PANC-1, SW1990, PANC-03.27) were lower than that of normal pancreatic ductal epithelial cells (HPDE6-C7) at both mRNA and protein levels. Both TGF-ß1 and BMP2 treatment induced EMT and downregulation of PPM1H in BxPC-3 cells. By using RNA interference to transiently knock down PPM1H expression in BxPC-3 cells, we found that the expression of E-cadherin was downregulated while vimentin was up-regulated. The data suggested that silencing PPM1H gene can induce EMT in BxPC-3 cells. In addition, Transwell migration assays showed that silencing PPM1H gene can promote the invasion and metastasis of BxPC-3 cells. Cell proliferation and apotosis detection demonstrated that silencing PPM1H gene can promote the proliferation and inhibit apoptosis of BxPC-3 cells. In conclusion, PPM1H is aberrantly expressed in human pancreatic cancer cell lines and can be downregulated when EMT is induced by cytokine stimulation. Silencing PPM1H gene can induce EMT in BxPC-3 cells, and promote the invasion and metastasis of BxPC-3 cells. Moreover, silencing PPM1H gene can promote the proliferation and inhibit apoptosis of BxPC-3 cells. PPM1H may be a new tumor-suppressor factor for pancreatic cancer and provides new insight into molecular targets for gene therapy of pancreatic cancer.


Biomarkers, Tumor/genetics , Epithelial-Mesenchymal Transition/genetics , Pancreatic Neoplasms/genetics , Phosphoprotein Phosphatases/genetics , Antigens, CD , Apoptosis/genetics , Biomarkers, Tumor/antagonists & inhibitors , Biomarkers, Tumor/biosynthesis , Bone Morphogenetic Protein 2/administration & dosage , Cadherins/biosynthesis , Cell Line, Tumor , Cell Movement/genetics , Cell Proliferation/genetics , Gene Expression Regulation, Neoplastic , Gene Silencing , Humans , Pancreatic Neoplasms/pathology , Phosphoprotein Phosphatases/antagonists & inhibitors , Phosphoprotein Phosphatases/biosynthesis , RNA Interference , Transforming Growth Factor beta/administration & dosage , Vimentin/biosynthesis
15.
Clin Cancer Res ; 21(16): 3783-93, 2015 Aug 15.
Article En | MEDLINE | ID: mdl-25977341

PURPOSE: The carcinogenic capacity of B[a]P/B[a]PDE is supported by epidemiologic studies. However, the molecular mechanisms responsible for B[a]P/B[a]PDE-caused lung cancer have not been well investigated. We evaluated here the role of novel target PHLPP2 in lung inflammation and carcinogenesis upon B[a]P/B[a]PDE exposure. EXPERIMENTAL DESIGN: We used the Western blotting, RT-PCR, [(35)S]methionine pulse and immunohistochemistry staining to determine PHLPP2 downregulation following B[a]P/B[a]PDE exposure. Both B[a]PDE-induced Beas-2B cell transformation model and B[a]P-caused mouse lung cancer model were used to elucidate the mechanisms leading to PHLPP2 downregulation and lung carcinogenesis. The important findings were also extended to in vivo human studies. RESULTS: We found that B[a]P/B[a]PDE exposure downregulated PHLPP2 expression in human lung epithelial cells in vitro and in mouse lung tissues in vivo. The ectopic expression of PHLPP2 dramatically inhibited cell transformation upon B[a]PDE exposure. Mechanistic studies showed that miR-205 induction was crucial for inhibition of PHLPP2 protein translation by targeting PHLPP2-3'-UTR. Interestingly, PHLPP2 expression was inversely associated with tumor necrosis factor alpha (TNFα) expression, with low PHLPP2 and high TNFα expression in lung cancer tissues compared with the paired adjacent normal lung tissues. Additional studies revealed that PHLPP2 exhibited its antitumorigenic effect of B[a]P/B[a]PDE through the repression of inflammatory TNFα transcription. CONCLUSIONS: Our studies not only first time identify PHLPP2 downregulation by lung carcinogen B[a]P/B[a]PDE, but also elucidate a novel molecular mechanisms underlying lung inflammation and carcinogenesis upon B[a]P/B[a]PDE exposure.


7,8-Dihydro-7,8-dihydroxybenzo(a)pyrene 9,10-oxide/toxicity , Benzo(a)pyrene/toxicity , Cell Transformation, Neoplastic/genetics , Inflammation/genetics , Lung Neoplasms/genetics , MicroRNAs/genetics , Phosphoprotein Phosphatases/biosynthesis , Animals , Carcinogenesis/genetics , Carcinogens/toxicity , Cell Proliferation/drug effects , Cell Transformation, Neoplastic/drug effects , Gene Expression Regulation, Neoplastic , Humans , Inflammation/chemically induced , Inflammation/pathology , Lung Neoplasms/chemically induced , Lung Neoplasms/pathology , Mice , Phosphoprotein Phosphatases/genetics
16.
Biomed Pharmacother ; 70: 164-9, 2015 Mar.
Article En | MEDLINE | ID: mdl-25776496

Previous studies have shown that microRNAs play essential roles in cancer growth and progression. Although a number of microRNAs were differentially expressed in glioblastoma (GBM). In this study, we evaluated the miR-522s role in cell proliferation in GBM. Expression of miR-522 is markedly upregulated in GBM tissues and GBM cells compared with the matched non-tumor adjacent brain tissues (TAT) and normal human astrocytes (NHAs). In functional assays, miR-522 promoted GBM cell proliferation, which could be reversed by inhibitor of miR-522. We further identified PH domain leucine-rich repeats protein phosphatase-1 (PHLPP1) as a putative target of miR-522, which is likely a main contributor to the promotion of tumor cell growth observed in our assays. Our results demonstrated that miR-522 promoted tumor cell proliferation and hence may represent a novel therapeutically relevant cellular target to treatment of GBM patients.


Brain Neoplasms/metabolism , Cell Proliferation/physiology , Gene Expression Regulation, Neoplastic , Glioblastoma/metabolism , MicroRNAs/physiology , Nuclear Proteins/biosynthesis , Phosphoprotein Phosphatases/biosynthesis , Brain Neoplasms/pathology , Cell Line, Tumor , Glioblastoma/pathology , Humans
17.
Cancer Lett ; 360(2): 171-6, 2015 May 01.
Article En | MEDLINE | ID: mdl-25684665

Slingshot-1L (SSH1L), a cofilin-phosphatase, plays a role in actin dynamics and cell migration by reactivating cofilin-1. However, the expression of SSH1L in malignant diseases is poorly understood. The overexpression of SSH1L in cancerous tissue compared to the matched surrounding non-cancerous tissues from patients with late stages (III-IV) of PC was detected in 90% (9/10) of cases by western blotting. The expression of SSH1L was shown to be upregulated in tumor cells from 10.7% (11/102) of patients with pancreatic cancer (PC) by immunohistochemistry (IHC). The positive rate of SSH1L in patients with PC at stage VI (TNM) categorized as grade 3 was of 50% (2/4) and 15% (6/40), respectively. Moreover, SSH1L expression was shown to be up-regulated in the PC cell lines (KLM1, PANC-1 and MIAPaCa-2) with high metastatic potential. Loss of SSH1L expression was associated with an increase in the phosphorylation of cofilin-1 at serine-3 and further inhibited cell migration (but not proliferation) in KLM1, PANC-1 and MIAPaCa-2. Actin polymerization inhibitor cytochalasin-D was sufficient to abrogate cell migration of PC without changing SSH1L expression. These results reveal that SSH1L is upregulated in a subset of PCs and that the SSH1L/cofilin-1 signal pathway is associated positively in PC with cell migration. Our study may thus provide potential targets to prevent and/or treat PC invasion and metastasis in patients with SSH1L-positive PC.


Pancreatic Neoplasms/enzymology , Pancreatic Neoplasms/pathology , Phosphoprotein Phosphatases/biosynthesis , Actins/antagonists & inhibitors , Actins/metabolism , Aged , Cell Movement/physiology , Cofilin 1/metabolism , Cytochalasin D/pharmacology , Female , Humans , Male , Middle Aged , Neoplasm Grading , Neoplasm Metastasis , Neoplasm Staging , Phosphorylation
18.
Appl Biochem Biotechnol ; 175(1): 526-34, 2015 Jan.
Article En | MEDLINE | ID: mdl-25326185

Ser/Thr protein phosphatase 5 (PPP5C) has been reported to participate in tumor progression. However, its functional role in hepatocellular carcinoma (HCC) remains unknown yet. In this study, we firstly evaluated the expression levels of PPP5C in six HCC cell lines by real-time PCR and found that PPP5C was widely expressed in HCC cells. To explore the role of PPP5C in HCC cell growth, lentivirus-mediated short hairpin RNA (shRNA) was employed to silence PPP5C expression in HepG2 and Bel-7404 cells. The expression of PPP5C was significantly downregulated in PPP5C knockdown cells. Knockdown of PPP5C markedly suppressed the proliferation and colony formation ability of HCC cells. Moreover, cell cycle analysis showed that PPP5C depletion in HepG2 cells led to G0/G1 phase and G2/M phase arrest. We demonstrate for the first time that PPP5C is essential for growth of HCC cells, which suggests that inhibition of PPP5C by RNAi may be a potential therapeutic strategy for the treatment of HCC.


Carcinoma, Hepatocellular/genetics , Cell Proliferation/genetics , Liver Neoplasms/genetics , Nuclear Proteins/genetics , Phosphoprotein Phosphatases/genetics , Apoptosis/genetics , Carcinoma, Hepatocellular/pathology , Gene Expression Regulation, Neoplastic , Gene Knockdown Techniques , Hep G2 Cells , Humans , In Vitro Techniques , Liver Neoplasms/pathology , Nuclear Proteins/antagonists & inhibitors , Nuclear Proteins/biosynthesis , Phosphoprotein Phosphatases/antagonists & inhibitors , Phosphoprotein Phosphatases/biosynthesis , RNA Interference
19.
Int J Clin Exp Pathol ; 8(11): 14741-8, 2015.
Article En | MEDLINE | ID: mdl-26823799

Sacral chordoma is a rare spine tumor with a high recurrence rate even after optimal therapy. Previous studies have demonstrated that the PI3K/AKT pathway plays a pivotal role in chordoma, and high expression of pAKT is associated with poor prognosis. Recently, PHLPP was recognized to be a tumor suppressor that targets AKT. We analyzed the expression of PHLPP1 and AKT2 in 37 chordoma samples and 11 fetal nucleus pulposus samples by immunohistochemical staining. Of the chordoma cases, 40.5% (15/37) showed strong cytoplasmic staining (score ≥3) for PHLPP1, which was significantly lower than the 90.9% (10/11) of fetal nucleus pulposus samples (P = 0.004). Conversely, strong immunohistochemical staining for AKT2 was observed in 75.7% (28/37) of chordoma samples, which was significantly higher than 36.4% (4/11) of fetal nucleus pulposus (P = 0.021). Kaplan-Meier survival curves and log-rank test showed that patients with high expression of PHLPP1 experienced longer progression free survival time than those with low PHLPP1 expression (P = 0.011). Further multivariate Cox regression analysis indicated that PHLPP1 expression level and surgical approaches were independent risk factors for chordoma recurrence (P = 0.023 and P = 0.022). However, PHLPP1 expression was not statistically related to patients' total survival time. Conclusively, our results suggest that PHLPP1 plays a crucial role in sacral chordoma, and may be a promising biomarker for prognosis. Meanwhile, manipulation of PHLPP1 expression is also a potential therapeutic approach for the treatment of sacral chordoma.


Biomarkers, Tumor/analysis , Chordoma/pathology , Nuclear Proteins/biosynthesis , Phosphoprotein Phosphatases/biosynthesis , Spinal Neoplasms/pathology , Adolescent , Adult , Aged , Aged, 80 and over , Chordoma/mortality , Disease-Free Survival , Female , Humans , Immunohistochemistry , Kaplan-Meier Estimate , Male , Middle Aged , Nuclear Proteins/analysis , Phosphoprotein Phosphatases/analysis , Polymerase Chain Reaction , Prognosis , Proportional Hazards Models , Sacrum/pathology
20.
Biomed Res Int ; 2014: 521794, 2014.
Article En | MEDLINE | ID: mdl-25530962

The phytohormone abscisic acid (ABA) plays an important role in plant development and environmental stress response. Additionally, ABA also regulates secondary metabolism such as artemisinin in the medicinal plant Artemisia annua L. Although an earlier study showed that ABA receptor, AaPYL9, plays a positive role in ABA-induced artemisinin content improvement, many components in the ABA signaling pathway remain to be elucidated in Artemisia annua L. To get insight of the function of AaPYL9, we isolated and characterized an AaPYL9-interacting partner, AaPP2C1. The coding sequence of AaPP2C1 encodes a deduced protein of 464 amino acids, with all the features of plant type clade A PP2C. Transcriptional analysis showed that the expression level of AaPP2C1 is increased after ABA, salt, and drought treatments. Yeast two-hybrid and bimolecular fluorescence complementation assays (BiFC) showed that AaPYL9 interacted with AaPP2C1. The P89S, H116A substitution in AaPYL9 as well as G199D substitution or deletion of the third phosphorylation site-like motif in AaPP2C1 abolished this interaction. Furthermore, constitutive expression of AaPP2C1 conferred ABA insensitivity compared with the wild type. In summary, our data reveals that AaPP2C1 is an AaPYL9-interacting partner and involved in the negative modulation of the ABA signaling pathway in A. annua L.


Abscisic Acid/genetics , Arabidopsis Proteins/genetics , Artemisinins/metabolism , Phosphoprotein Phosphatases/genetics , Abscisic Acid/metabolism , Arabidopsis/genetics , Arabidopsis Proteins/biosynthesis , Artemisia annua/genetics , Artemisia annua/growth & development , Gene Expression Regulation, Plant , Germination/genetics , Mutation , Phosphoprotein Phosphatases/biosynthesis , Phosphoprotein Phosphatases/metabolism , Plant Leaves/genetics , Plant Leaves/growth & development , Signal Transduction/genetics , Stress, Physiological/genetics
...