Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
Add more filters










Publication year range
1.
Vet Microbiol ; 294: 110124, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38795403

ABSTRACT

PEDV, a single-stranded RNA virus, causes significant economic losses in the pig industry. Sin3-associated protein 18 (SAP18) is known for its role in transcriptional inhibition and RNA splicing. However, research on SAP18's involvement in PEDV infection is limited. Here, we identified an interaction between SAP18 and PEDV nonstructural protein 10 (Nsp10) using immunoprecipitation-mass spectrometry (IP-MS) and confirmed it through immunoprecipitation and laser confocal microscopy. Additionally, PEDV Nsp10 reduced SAP18 protein levels and induced its cytoplasmic accumulation. Overexpressing SAP18 suppressed PEDV replication, meanwhile its knockdown via short interfering RNA (siRNA) enhanced replication. SAP18 overexpression boosted IRF3 and NF-κB P65 phosphorylation, nuclear translocation, and IFN-ß antiviral response. Furthermore, SAP18 upregulated RIG-I expression and facilitated its dephosphorylation, while SAP18 knockdown had the opposite effect. Finally, SAP18 interacted with phosphatase 1 (PP1) catalytic subunit alpha (PPP1CA), promoting PPP1CA-RIG-I interaction during PEDV infection. These findings highlight SAP18's role in activating the type I interferon pathway and inhibiting viral replication by promoting RIG-I dephosphorylation through its interaction with PPP1CA.


Subject(s)
Porcine epidemic diarrhea virus , Viral Nonstructural Proteins , Virus Replication , Animals , Viral Nonstructural Proteins/metabolism , Viral Nonstructural Proteins/genetics , Porcine epidemic diarrhea virus/physiology , Porcine epidemic diarrhea virus/genetics , Phosphorylation , Swine , Cell Line , DEAD Box Protein 58/metabolism , DEAD Box Protein 58/genetics , Chlorocebus aethiops
2.
J Biomol Struct Dyn ; : 1-17, 2024 Mar 12.
Article in English | MEDLINE | ID: mdl-38469816

ABSTRACT

NIMA-related kinase 7 (NEK7) and phosphoprotein phosphatase-1 catalytic subunit alpha (PPP1CA) are the most common proteins overexpressed in pancreatic ductal adenocarcinoma, which is the most common type of pancreatic cancer. The goal of the current study was to identify a possible NEK7 and PPP1CA therapeutic inhibitor. For this investigation, 5000 compounds were retrieved from the IMPPAT library of phytochemicals, which were docked with our respective target proteins. Also, a reference compound, gemcitabine, which is a Food and Drug Administration (FDA) approved drug, was docked with the target proteins. The binding energy of the reference compound for both the targeted proteins was -6.5 kcal/mol. The common ligand with the lowest binding energy for both targets is boeravinone B (PubChem ID: 14018348) with -9.2 kcal/mol of NEK7 and -7.6 kcal/mol for PPP1CA. The compound was further investigated through density function theory (DFT) and molecular dynamic simulation analysis. The root mean square deviation (RMSD), root mean square fluctuation (RMSF), radius of gyration (Rg), and hydrogen bonding analysis indicated the stability of the boeravinone B with the target proteins (NEK7 and PPP1CA).Communicated by Ramaswamy H. Sarma.

3.
Mol Cancer ; 23(1): 34, 2024 02 15.
Article in English | MEDLINE | ID: mdl-38360682

ABSTRACT

BACKGROUND: Clear cell renal cell carcinoma (ccRCC) is the most prevalent kidney cancer with high aggressive phenotype and poor prognosis. Accumulating evidence suggests that circRNAs have been identified as pivotal mediators in cancers. However, the role of circRNAs in ccRCC progression remains elusive. METHODS: The differentially expressed circRNAs in 4 paired human ccRCC and adjacent noncancerous tissues ccRCC were screened using circRNA microarrays and the candidate target was selected based on circRNA expression level using weighted gene correlation network analysis (WGCNA) and the gene expression omnibus (GEO) database. CircPDHK1 expression in ccRCC and adjacent noncancerous tissues (n = 148) were evaluated along with clinically relevant information. RT-qPCR, RNase R digestion, and actinomycin D (ActD) stability test were conducted to identify the characteristics of circPDHK1. The subcellular distribution of circPDHK1 was analyzed by subcellular fractionation assay and fluorescence in situ hybridization (FISH). Immunoprecipitation-mass spectrometry (IP-MS) and immunofluorescence (IF) were employed to evaluate the protein-coding ability of circPDHK1. ccRCC cells were transfected with siRNAs, plasmids or lentivirus approach, and cell proliferation, migration and invasion, as well as tumorigenesis and metastasis in nude mice were assessed to clarify the functional roles of circPDHK1 and its encoded peptide PDHK1-241aa. RNA-sequencing, western blot analysis, immunoprecipitation (IP) and chromatin immunoprecipitation (ChIP) assays were further employed to identify the underlying mechanisms regulated by PDHK1-241aa. RESULTS: CircPDHK1 was upregulated in ccRCC tissues and closely related to WHO/ISUP stage, T stage, distant metastasis, VHL mutation and Ki-67 levels. CircPDHK1 had a functional internal ribosome entry site (IRES) and encoded a novel peptide PDHK1-241aa. Functionally, we confirmed that PDHK1-241aa and not the circPDHK1 promoted the proliferation, migration and invasion of ccRCC. Mechanistically, circPDHK1 was activated by HIF-2A at the transcriptional level. PDHK1-241aa was upregulated and interacted with PPP1CA, causing the relocation of PPP1CA to the nucleus. This thereby inhibited AKT dephosphorylation and activated the AKT-mTOR signaling pathway. CONCLUSIONS: Our data indicated that circPDHK1-encoded PDHK1-241aa promotes ccRCC progression by interacting with PPP1CA to inhibit AKT dephosphorylation. This study provides novel insights into the multiplicity of circRNAs and highlights the potential use of circPDHK1 or PDHK1-241aa as a therapeutic target for ccRCC.


Subject(s)
Carcinoma, Renal Cell , Kidney Neoplasms , Animals , Mice , Humans , Carcinoma, Renal Cell/metabolism , Proto-Oncogene Proteins c-akt/metabolism , RNA, Circular/genetics , Mice, Nude , In Situ Hybridization, Fluorescence , Cell Line, Tumor , Signal Transduction/genetics , Kidney Neoplasms/genetics , TOR Serine-Threonine Kinases/metabolism , Cell Proliferation/genetics , Peptides/genetics , Gene Expression Regulation, Neoplastic , Protein Phosphatase 1/genetics , Protein Phosphatase 1/metabolism
4.
J Exp Clin Cancer Res ; 42(1): 334, 2023 Dec 06.
Article in English | MEDLINE | ID: mdl-38057879

ABSTRACT

BACKGROUND: With the advancements in bioinformatic technology, an increasing number of circular RNAs (circRNAs) have been discovered and their crucial roles in the development and progression of various malignancies have been confirmed through multiple pathways. However, the specific mechanisms involving protein-binding circRNAs in colorectal cancer (CRC) remain largely unexplored. METHODS: Differential circRNA expression was assessed using a human circRNA microarray in five CRC tissue and paired normal samples. CircGPRC5A expression was then confirmed in the CRC tissues and paired normal samples using qRT-PCR. The biological function of circGPRC5A in CRC were studied in vitro and in vivo. Western blotting, fluorescence in situ hybridization, immunofluorescence, RNA pulldown, mass spectrometry, immunoprecipitation, quantitative phosphoproteomics, and RNA-binding protein immunoprecipitation assays were used to study circGPRC5A. RESULTS: Our analysis revealed that circGPRC5A expression was higher in CRC tissues compared to normal tissues and was associated with tumor size, tumor stage and lymph node status. CircGPRC5A promoted CRC cell proliferation, migration, and metastasis in vitro and in vivo. CircGPRC5A could stabilize PPP1CA protein by inhibiting the binding between UBA1 and PPP1CA, and increasing YAP dephosphorylation. CONCLUSIONS: Our study revealed that circGPRC5A plays an essential function in CRC progression by stabilizing PPP1CA protein and enhancing YAP dephosphorylation. CircGPRC5A could act as a novel and potential target for CRC.


Subject(s)
Colorectal Neoplasms , MicroRNAs , Humans , Cell Proliferation/genetics , Colorectal Neoplasms/pathology , Gene Expression Regulation, Neoplastic , In Situ Hybridization, Fluorescence , MicroRNAs/genetics , Protein Phosphatase 1/metabolism , RNA/genetics , RNA, Circular/genetics , RNA, Circular/metabolism
5.
Int J Mol Sci ; 24(24)2023 Dec 10.
Article in English | MEDLINE | ID: mdl-38139150

ABSTRACT

The vertebrate PPP1R15 family consists of the proteins GADD34 (growth arrest and DNA damage-inducible protein 34, the product of the PPP1R15A gene) and CReP (constitutive repressor of eIF2α phosphorylation, the product of the PPP1R15B gene), both of which function as targeting/regulatory subunits for protein phosphatase 1 (PP1) by regulating subcellular localization, modulating substrate specificity and assembling complexes with target proteins. The primary cellular function of these proteins is to facilitate the dephosphorylation of eukaryotic initiation factor 2-alpha (eIF2α) by PP1 during cell stress. In this review, we will provide a comprehensive overview of the cellular function, biochemistry and pharmacology of GADD34 and CReP, starting with a brief introduction of eIF2α phosphorylation via the integrated protein response (ISR). We discuss the roles GADD34 and CReP play as feedback inhibitors of the unfolded protein response (UPR) and highlight the critical function they serve as inhibitors of the PERK-dependent branch, which is particularly important since it can mediate cell survival or cell death, depending on how long the stressful stimuli lasts, and GADD34 and CReP play key roles in fine-tuning this cellular decision. We briefly discuss the roles of GADD34 and CReP homologs in model systems and then focus on what we have learned about their function from knockout mice and human patients, followed by a brief review of several diseases in which GADD34 and CReP have been implicated, including cancer, diabetes and especially neurodegenerative disease. Because of the potential importance of GADD34 and CReP in aspects of human health and disease, we will discuss several pharmacological inhibitors of GADD34 and/or CReP that show promise as treatments and the controversies as to their mechanism of action. This review will finish with a discussion of the biochemical properties of GADD34 and CReP, their regulation and the additional interacting partners that may provide insight into the roles these proteins may play in other cellular pathways. We will conclude with a brief outline of critical areas for future study.


Subject(s)
Neurodegenerative Diseases , Protein Phosphatase 1 , Animals , Humans , Mice , Eukaryotic Initiation Factor-2/metabolism , Mice, Knockout , Phosphorylation , Protein Biosynthesis , Protein Phosphatase 1/genetics , Protein Phosphatase 1/metabolism , Proteins/metabolism
6.
Cell Chem Biol ; 30(2): 188-202.e6, 2023 02 16.
Article in English | MEDLINE | ID: mdl-36720221

ABSTRACT

Reversible protein phosphorylation, catalyzed by protein kinases and phosphatases, is a fundamental process that controls protein function and intracellular signaling. Failure of phospho-control accounts for many human diseases. While a kinase phosphorylates multiple substrates, a substrate is often phosphorylated by multiple kinases. This renders phospho-control at the substrate level challenging, as it requires inhibition of multiple kinases, which would thus affect other kinase substrates. Here, we describe the development and application of the affinity-directed phosphatase (AdPhosphatase) system for targeted dephosphorylation of specific phospho-substrates. By deploying the Protein Phosphatase 1 or 2A catalytic subunits conjugated to an antigen-stabilized anti-GFP nanobody, we can promote the dephosphorylation of two independent phospho-proteins, FAM83D or ULK1, knocked in with GFP-tags using CRISPR-Cas9, with exquisite specificity. By redirecting protein phosphatases to neo-substrates through nanobody-mediated proximity, AdPhosphatase can alter the phospho-status and function of target proteins and thus, offers a new modality for potential drug discovery approaches.


Subject(s)
Protein Kinases , Protein Phosphatase 2 , Humans , Cell Cycle Proteins/metabolism , Microtubule-Associated Proteins/metabolism , Phosphorylation , Protein Kinases/metabolism , Protein Phosphatase 2/metabolism , Substrate Specificity , Phosphoric Monoester Hydrolases/metabolism
7.
J Neurochem ; 161(1): 20-39, 2022 Apr.
Article in English | MEDLINE | ID: mdl-35050500

ABSTRACT

Vitamins B1 (thiamine) and B6 (pyridox (al/ine/amine)) are crucial for central nervous system (CNS) function and neurogenesis due to the coenzyme action of their phosphorylated derivatives in the brain metabolism of glucose and neurotransmitters. Here, the non-coenzyme action of thiamine on the major mammalian producers of pyridoxal-5'-phosphate (PLP), such as pyridoxal kinase (PdxK) and pyridoxine 5'-phosphate oxidase (PNPO), is characterized. Among the natural thiamine compounds, thiamine triphosphate (ThTP) is the best effector of recombinant human PdxK (hPdxK) in vitro, inhibiting hPdxK in the presence of Mg2+ but activating the Zn2+ -dependent reaction. Inhibition of hPdxK by thiamine antagonists decreases from amprolium to pyrithiamine to oxythiamine, highlighting possible dysregulation of both the B1 - and B6 -dependent metabolism in the chemical models of thiamine deficiency. Compared with the canonical hPdxK, the D87H and V128I variants show a twofold increase in Kapp of thiamine inhibition, and the V128I and H246Q variants show a fourfold and a twofold decreased Kapp of thiamine diphosphate (ThDP), respectively. Thiamine administration changes diurnal regulation of PdxK activity and phosphorylation at Ser213 and Ser285, expression of the PdxK-related circadian kinases/phosphatases in the rat brain, and electrocardiography (ECG). In contrast to PdxK, PNPO is not affected by thiamine or its derivatives, either in vitro or in vivo. Dephosphorylation of the PdxK Ser285, potentially affecting mobility of the ATP-binding loop, inversely correlates with the enzyme activity. Dephosphorylation of the PdxK Ser213, which is far away from the active site, does not correlate with the activity. The correlations analysis suggests the PdxK Ser213 to be a target of kinase MAP2K1 and phosphatase Ppp1ca. Diurnal effects of thiamine administration on the metabolically linked ThDP- and PLP-dependent enzymes may support the brain homeostatic mechanisms and physiological fitness.


Subject(s)
Pyridoxal Kinase , Thiamine , Animals , Brain/metabolism , Mammals/metabolism , Phosphates , Pyridoxal Kinase/chemistry , Pyridoxal Kinase/metabolism , Pyridoxal Phosphate/metabolism , Pyridoxal Phosphate/pharmacology , Rats , Thiamine/pharmacology
8.
Gastroenterol Hepatol Bed Bench ; 15(4): 395-405, 2022.
Article in English | MEDLINE | ID: mdl-36762213

ABSTRACT

Aim: This study investigated the association between methylation status and expression levels of BTG2, PPP1CA, and PEG3 genes in colon cancer. Background: Aberrant DNA methylation is one of the most important epigenetic modifications in the development of cancer. Evidence indicates that hypermethylation of various tumor suppressor genes could be a potential mechanism of colon tumorigenesis. Methods: The expression levels of BTG2, PPP1CA, and PEG3 genes were evaluated in HT-29/219, HCT116, SW48, SW742, SW480, and LS180 cell lines using quantitative Real-Time PCR. The methylation status of BTG2 and PPP1CA was determined by methylation-specific PCR (MSP) method, and the methylation pattern of PEG3 was evaluated by bisulfite sequencing PCR (BSP). To investigate the effect of methylation on the expression of these genes, all colon cancer cell lines were treated by 5-Azacitidine (5-Aza) and/or Trichostatin A (TSA). Results: The expression levels of BTG2, PPP1CA, and PEG3 were highly heterogeneous and quantitatively correlated to their promoter methylation status in the studied colon cancer cell lines. Treatment by 5-Aza and/or TSA increased the expression of the above-named genes in colon cancer cell lines. Conclusion: Overall, it seems that BTG2, PPP1CA, and PEG3 act as tumor suppressor genes in colon cancer, and methylation is a potential mechanism for their loss of expression. Therefore, these genes may be considered as suitable targets for demethylation approaches and, eventually, colon cancer treatment. Combined treatment by 5-Aza and TSA may be a promising therapeutic strategy for colon cancer treatment. Further studies may contribute to confirm these results.

9.
Exp Eye Res ; 210: 108703, 2021 09.
Article in English | MEDLINE | ID: mdl-34280391

ABSTRACT

Diabetic retinopathy (DR) is a vision-loss complication caused by diabetes with high prevalence. During DR, the retinal microvascular injury and neurodegeneration derived from chronic hyperglycemia have attracted global attention to retinal Müller cells (RMCs), the major macroglia in the retina contributes to neuroprotection. Protein Phosphatase 1 Catalytic Subunit Alpha (PPP1CA) dephosphorylates the transcriptional coactivator Yes-associated protein (YAP) to promote the transcription of glutamine synthetase (GS). GS catalyzes the transformation of neurotoxic glutamate (Glu) into nontoxic glutamine (Gln) to activate the mammalian target of rapamycin complex 1 (mTORC1), which promotes the activation of RMCs. In this study, in vitro MIO-M1 cell and in vivo mouse high-fat diet and streptozotocin (STZ)-induced diabetic model to explore the role of the PPP1CA/YAP/GS/Gln/mTORC1 pathway on the activation of MRCs during DR. Results showed that PPP1CA promoted the dephosphorylation and nuclear translocation of YAP in high glucose (HG)-exposed MIO-M1 cells. YAP transcribed GS in HG-exposed MIO-M1 cells in a TEAD1-dependent and PPP1CA-dependent way. GS promoted the biosynthesis of Gln in HG-exposed MIO-M1 cells. Gln activated mTORC1 instead of mTORC2 in HG-exposed MIO-M1 cells. The proliferation and activation of HG-exposed MIO-M1 cells were PPP1CA/YAP/GS/Gln/mTORC1-dependent. Finally, RMC proliferation and activation during DR were inhibited by the PPP1CA/YAP/GS/Gln/mTORC1 blockade. The findings supplied a potential idea to protect RMCs and alleviate the development of DR.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Diabetic Retinopathy/metabolism , Ependymoglial Cells/metabolism , Glutamate-Ammonia Ligase/metabolism , Glutamine/metabolism , Mechanistic Target of Rapamycin Complex 1/metabolism , Protein Phosphatase 1/metabolism , Animals , Blotting, Western , Cell Proliferation , Cells, Cultured , Diabetes Mellitus, Experimental/metabolism , Enzyme-Linked Immunosorbent Assay , Glucose/pharmacology , Male , Mice , Mice, Inbred C57BL , Microscopy, Fluorescence , Real-Time Polymerase Chain Reaction , Signal Transduction/physiology , Streptozocin , YAP-Signaling Proteins
10.
EBioMedicine ; 48: 236-247, 2019 Oct.
Article in English | MEDLINE | ID: mdl-31521612

ABSTRACT

BACKGROUND: USP11 is an ubiquitin-specific protease that plays an important role in tumor progression via different mechanisms. However, the expression and prognostic significance of USP11 in colorectal cancer (CRC) remain unknown. METHODS: Bioinformatics analyses, qRT-PCR, western blotting, and immunohistochemistry were applied for investigating USP11 expression in CRC tissues. Kaplan-Meier analysis with log-rank test was used for survival analyses. LC-MS/MS was performed for identifying potential protein interactions with USP11. In vitro and in vivo assays were used for exploring the function of USP11 during the progression of CRC. FINDINGS: USP11 was overexpressed in CRC tissues and functioned as an oncogene. Overexpression or knockdown of USP11 promoted or inhibited, respectively, the growth and metastasis of CRC cells in vitro and in vivo. Mechanically, USP11 stabilized PPP1CA by deubiquitinating and protecting it from proteasome-mediated degradation. Moreover, the USP11/PPP1CA complex promoted CRC progression by activating the ERK/MAPK signaling pathway. INTERPRETATION: USP11 promoted tumor growth and metastasis in CRC via the ERK/MAPK pathway by stabilizing PPP1CA, suggesting USP11 is a potential prognostic marker. FUND: This work was supported by National Natural Science Foundation of China (NSFC81530044, NSFC81220108021, NSFC81802343), Technology Major Project of China Grants 2017ZX10203206, Shanghai Sailing Program (19YF1409600) and The project of Shanghai Jiaotong University (YG2017QN30).


Subject(s)
Colorectal Neoplasms/etiology , Colorectal Neoplasms/metabolism , MAP Kinase Signaling System , Protein Phosphatase 1/metabolism , Thiolester Hydrolases/genetics , Aged , Aged, 80 and over , Animals , Cell Line, Tumor , Colorectal Neoplasms/mortality , Colorectal Neoplasms/pathology , Disease Models, Animal , Female , Gene Expression Profiling , Gene Expression Regulation, Neoplastic , Humans , Immunohistochemistry , Kaplan-Meier Estimate , Male , Mice , Middle Aged , Neoplasm Staging , Prognosis , Protein Binding , Protein Phosphatase 1/genetics , Proteolysis , Thiolester Hydrolases/metabolism , Transcriptome
11.
Anticancer Res ; 37(12): 6705-6714, 2017 12.
Article in English | MEDLINE | ID: mdl-29187447

ABSTRACT

BACKGROUND/AIM: Bladder cancer (BC) has a high recurrence rate and may progress to being a muscle-invasive lesion, that is potentially associated with a poor prognosis. We identified tumor-associated proteins that were recognized by autoantibodies in sera from patients with high-grade non-muscle-invasive bladder cancer (HG-NMIBC) by proteomic analysis. MATERIALS AND METHODS: The serum levels of these autoantibodies against identified proteins were validated by dot blot analysis with sera from 95 patients with BC and 35 healthy controls. The expression of identified proteins was immunohistochemically analyzed in 115 BC tissues. RESULTS: Autoantibody against protein phosphatase 1, catalytic subunit, alpha isoform (PPP1CA) protein was detected in pretreated sera from patients with HG-NMIBC who showed progression. The serum IgG level of anti-PPP1CA autoantibody was significantly correlated with pathological stage, grade, lymphovascular invasion, and prognosis. The immunoreactions for PPP1CA protein in BC was significantly correlated with pathological stage, grade, and lymphovascular invasion. CONCLUSION: PPP1CA is a candidate sero-diagnostic and prognostic marker for patients with BC.


Subject(s)
Autoantibodies/immunology , Proteome/immunology , Proteomics/methods , Urinary Bladder Neoplasms/immunology , Adult , Aged , Aged, 80 and over , Autoantibodies/blood , Disease Progression , Female , Humans , Immunoglobulin G/blood , Immunoglobulin G/immunology , Immunohistochemistry , Male , Middle Aged , Neoplasm Grading , Prognosis , Protein Phosphatase 1/immunology , Protein Phosphatase 1/metabolism , Urinary Bladder Neoplasms/blood , Urinary Bladder Neoplasms/metabolism
12.
Pathol Oncol Res ; 22(2): 287-92, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26253838

ABSTRACT

BACKGROUND: Protein phosphatase 1 α (PP1A) is an enzyme intimately associated with cell cycle, the over expression of which has been demonstrated in glioblastoma (GBM). Further, the nuclear expression of PP1A has been shown to be highly specific to GBM. In addition, PP1A has been shown to be a connecting molecule in the p53 containing GBM sub network. In view of these, we evaluated the prognostic relevance of PP1A. METHODS: GBM tissues were examined for protein expression of PP1A by immunohistochemistry (IHC). Nuclear expression of PP1A was scored in all tumor tissue samples. Survival analyses were performed by Cox-Regression and Kaplan-Meier survival analysis with Log Rank tests. IDH1, ATRX and p53 IHC and stratification of all GBM cases were performed and subgroup specific evaluation of nuclear PP1A correlation with overall and progression free survival was performed. RESULTS: PP1A protein expression showed no correlation with prognosis in all cases of GBM or on stratification based on IDH1 or ATRX expression. However on p53 stratification nuclear PP1A expression emerged as strong independent predictor of poor overall survival only in p53 positive GBMs both in univariate and multivariate analysis. CONCLUSIONS: While PP1A expression uniquely associates with poor prognosis only in p53 expressing GBMs, there is a notable absence of such correlation in p53 negative GBMs; thus skewing the overall relation of this molecule with prognosis in GBM. PP1A emerging as a strong prognostic marker in p53 expressing GBMs, enables us to foresee this molecule as a potential therapeutic target.


Subject(s)
Biomarkers, Tumor/metabolism , Brain Neoplasms/pathology , Glioblastoma/pathology , Protein Phosphatase 1/metabolism , Tumor Suppressor Protein p53/metabolism , Adult , Brain Neoplasms/enzymology , Cohort Studies , Female , Follow-Up Studies , Gene Expression Regulation, Neoplastic , Glioblastoma/enzymology , Humans , Immunoenzyme Techniques , Male , Neoplasm Staging , Prognosis , Survival Rate
13.
Gastric Cancer ; 18(4): 729-39, 2015 Oct.
Article in English | MEDLINE | ID: mdl-25240408

ABSTRACT

BACKGROUND: MiR-125b functions as an oncogene in many cancers; however, its clinical significance and molecular mechanism in gastric cancers have never been sufficiently investigated. Here, we elucidated the functions and molecular regulated pathways of MiR-125b in gastric cancer. METHODS: We investigated MiR-125b expression in fresh tissues from 50 gastric cancer patients and 6 gastric cancer cell lines using RT-PCR, and explored its prognostic value by hybridizing MiR-125b in situ for 300 clinical gastric tumor tissues with pathological diagnosis and clinical parameters. The effects of MiR-125b on gastric cancer cells and downstream target genes and proteins were analyzed by MTT, transwell assay, RT-PCR, and western blot on the basis of silencing MiR-125b in vitro. Luciferase reporter plasmid was constructed to demonstrate MiR-125b's direct target. RESULTS: MiR-125b was upregulated in gastric cancer tissues and cell lines, and significantly promoted cellular proliferation, migration, and invasion by downregulating the expression of PPP1CA and upregulating Rb phosphorylation. MiR-125b expression was significantly correlated with tumor size and depth of invasion, lymph nodes, distant metastasis, and TNM stage. The high-MiR-125b-expression group had a significantly poorer prognosis than the low-expression group (P < 0.05) in stages I, II, and III, and the 5-year survival rate in of the high-expression group was significantly lower than that of the low-expression group. CONCLUSIONS: MiR-125b functions as an oncogene by targeting downregulated PPP1CA and upregulated Rb phosphorylation in gastric cancer. MiR-125b not only promotes cellular proliferation, migration, and invasion in vitro, but also acts as an independent prognostic factor in gastric cancer.


Subject(s)
Cell Movement , Gene Expression Regulation, Neoplastic/genetics , MicroRNAs/genetics , Protein Phosphatase 1/metabolism , Retinoblastoma Protein/metabolism , Signal Transduction , Stomach Neoplasms/pathology , Adult , Aged , Blotting, Western , Cell Line , Cell Proliferation/genetics , Female , Humans , In Situ Hybridization , Male , Middle Aged , Neoplasm Invasiveness , Prognosis , RNA, Small Interfering , Reverse Transcriptase Polymerase Chain Reaction , Stomach Neoplasms/genetics , Stomach Neoplasms/mortality , Tissue Array Analysis , Transfection
SELECTION OF CITATIONS
SEARCH DETAIL