Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 5 de 5
1.
PLoS One ; 14(3): e0213463, 2019.
Article En | MEDLINE | ID: mdl-30893332

Distant metastasis leads oral cancer patients into a poor survival rate and a high recurrence stage. During tumor progression, dysregulated microRNAs (miRNAs) have been reported to involve tumor initiation and modulate oral cancer malignancy. MiR-450a was significantly upregulated in oral squamous cell carcinoma (OSCC) patients without functional reports. This study was attempted to uncover the molecular mechanism of novel miR-450a in OSCC. Mir-450a expression was examined by quantitative RT-PCR, both in OSCC cell lines and patients. Specific target of miR-450a was determined by software prediction, luciferase reporter assay, and correlation with target protein expression. The functions of miR-450a and TMEM182 were accessed by adhesion and transwell invasion analyses. Determination of the expression and cellular localization of TMEM182 was examined by RT-PCR and by immunofluorescence staining. The signaling pathways involved in regulation of miR-450a were investigated using the kinase inhibitors. Overexpression of miR-450a in OSCC cells impaired cell adhesion ability and induced invasiveness, which demonstrated the functional role of miR-450a as an onco-miRNA. Interestingly, tumor necrosis factor alpha (TNF-α)-mediated expression of TMEM182 was regulated by miR-450a induction. MiR-450a-reduced cellular adhesion was abolished by TMEM182 restoration. Furthermore, the oncogenic activity of TNF-α/miR-450a/TMEM182 axis was primarily through activating extracellular signal-regulated kinase 1/2 (ERK1/2) signaling pathway. ERK1/2 inhibitor prevented the TNF-α-induced miR-450a expression and enhanced adhesion ability. Our data suggested that TNF-α-induced ERK1/2-dependent miR-450a against TMEM182 expression exerted a great influence on increasing OSCC motility. Overall, our results provide novel molecular insights into how TNF-α contributes to oral carcinogenesis through miR-450a that targets TMEM182.


Carcinoma, Squamous Cell/metabolism , Membrane Proteins/metabolism , MicroRNAs/metabolism , Mouth Neoplasms/metabolism , Tumor Necrosis Factor-alpha/metabolism , Adult , Aged , Carcinoma, Squamous Cell/genetics , Carcinoma, Squamous Cell/secondary , Cell Adhesion/genetics , Cell Adhesion/physiology , Cell Line, Tumor , Cell Movement/genetics , Cell Movement/physiology , Female , Humans , MAP Kinase Signaling System , Male , Membrane Proteins/genetics , MicroRNAs/genetics , Middle Aged , Mouth Neoplasms/genetics , Mouth Neoplasms/pathology , Neoplasm Invasiveness/genetics , Neoplasm Invasiveness/pathology
2.
Cell Biosci ; 4: 25, 2014.
Article En | MEDLINE | ID: mdl-24904742

BACKGROUND: Protein phosphates 4 (PP4), encoded by the ppp4c gene, is a ubiquitously expressed phosphatase that has been implicated in the regulation of cytokine signaling and lymphocyte survival; recent reports suggest that PP4 may be involved in pre-TCR signaling and B cell development. However, whether PP4 also modulates the functions of peripheral T cells has not been investigated due to the lack of a suitable in vivo model. Treg cells are a specialized subset of CD4 helper T cells that can suppress the proliferation of activated effector T cells. In the absence of this negative regulation, autoimmune syndromes and inflammatory diseases, such as human Crohn's disease, will arise. RESULTS: In this report, we generated mice with T cell-specific ablation of the ppp4c gene (CD4cre:PP4(f/f)) and a Foxp3-GFP reporter gene to examine the roles of PP4 in Treg development and function. Characterizations of the CD4cre:PP4(f/f) mice showed that PP4 deficiency induced partial αß T lymphopenia and T cell hypo-proliferation. Further analyses revealed significant reductions in the numbers of thymic and peripheral Treg cells, as well as in the efficiency of in vitro Treg polarization. In addition, PP4-deficient Treg cells exhibited reduced suppressor functions that were associated with decreased IL-10, CTLA4, GITR and CD103 expression. More interestingly, the CD4cre:PP4(f/f) mice developed spontaneous rectal prolapse and colitis with symptoms similar to human Crohn's disease. The pathogenesis of colitis required the presence of commensal bacteria, and was correlated with reduced Treg cells in the gut. Nevertheless, PP4-deficient Treg cells were still capable of suppressing experimental colitis, suggesting that multiple factors contributed to the onset of the spontaneous colitis. CONCLUSIONS: While the molecular mechanisms remain to be investigated, our results clearly show that PP4 plays a non-redundant role for the differentiation, suppressor activity and gut homeostasis of Treg cells. The onset of spontaneous colitis in the CD4cre:PP4(f/f) mice further suggests that PP4 is essential for the maintenance of protective gut immunity. The CD4cre:PP4(f/f) mice thus may serve as a good model for studying the interactions between Treg cells and gut commensal bacteria for the regulation of mucosal immunity.

3.
J Biol Chem ; 287(38): 32216-21, 2012 Sep 14.
Article En | MEDLINE | ID: mdl-22833682

Retinoic acid-inducible gene I (RIG-I)-like receptors (RLRs) are key RNA viral sensors for triggering antiviral immunity. The underlying mechanisms for RLRs to trigger antiviral immunity have yet to be explored. Here we report the identification of TAPE (TBK1-associated protein in endolysosomes) as a novel regulator of the RLR pathways. TAPE functionally and physically interacts with RIG-I, MDA5, and IPS-1 to activate the IFN-ß promoter. TAPE knockdown impairs IFN-ß activation induced by RLRs but not IPS-1. TAPE-deficient cells are defective in cytokine production upon RLR ligand stimulation. During RNA virus infection, TAPE knockdown or deficiency diminishes cytokine production and antiviral responses. Our data demonstrate a critical role for TAPE in linking RLRs to antiviral immunity.


DEAD-box RNA Helicases/metabolism , DNA-Binding Proteins/chemistry , Repressor Proteins/chemistry , Animals , Antiviral Agents/chemistry , Antiviral Agents/pharmacology , Cell Line, Tumor , Chlorocebus aethiops , DEAD Box Protein 58 , DNA-Binding Proteins/metabolism , Gene Expression Regulation , HEK293 Cells , Humans , Immune System , Macrophages/metabolism , Mice , Mice, Knockout , Protein Binding , Protein Serine-Threonine Kinases/chemistry , RNA Interference , Receptors, Immunologic , Repressor Proteins/metabolism , Signal Transduction , Vero Cells
4.
PLoS One ; 7(6): e40193, 2012.
Article En | MEDLINE | ID: mdl-22768254

BACKGROUND: Pluripotent embryonic stem cells are considered to be an unlimited cell source for tissue regeneration and cell-based therapy. Investigating the molecular mechanism underlying the regulation of embryonic stem cell expansion is thus important. 14-3-3 proteins are implicated in controlling cell division, signaling transduction and survival by interacting with various regulatory proteins. However, the function of 14-3-3 in embryonic stem cell proliferation remains unclear. METHODOLOGY AND PRINCIPAL FINDINGS: In this study, we show that all seven 14-3-3 isoforms were detected in mouse embryonic stem cells. Retinoid acid suppressed selectively the expression of 14-3-3σ isoform. Knockdown of 14-3-3σ with siRNA reduced embryonic stem cell proliferation, while only 14-3-3σ transfection increased cell growth and partially rescued retinoid acid-induced growth arrest. Since the growth-enhancing action of 14-3-3σ was abrogated by ß-catenin knockdown, we investigated the influence of 14-3-3σ overexpression on ß-catenin/GSK-3ß. 14-3-3σ bound GSK-3ß and increased GSK-3ß phosphorylation in a PI-3K/Akt-dependent manner. It disrupted ß-catenin binding by the multiprotein destruction complex. 14-3-3σ overexpression attenuated ß-catenin phosphorylation and rescued the decline of ß-catenin induced by retinoid acid. Furthermore, 14-3-3σ enhanced Wnt3a-induced ß-catenin level and GSK-3ß phosphorylation. DKK, an inhibitor of Wnt signaling, abolished Wnt3a-induced effect but did not interfere GSK-3ß/14-3-3σ binding. SIGNIFICANCE: Our findings show for the first time that 14-3-3σ plays an important role in regulating mouse embryonic stem cell proliferation by binding and sequestering phosphorylated GSK-3ß and enhancing Wnt-signaled GSK-3ß inactivation. 14-3-3σ is a novel target for embryonic stem cell expansion.


14-3-3 Proteins/metabolism , Embryonic Stem Cells/cytology , Embryonic Stem Cells/enzymology , Glycogen Synthase Kinase 3/metabolism , beta Catenin/metabolism , Animals , Axin Protein/metabolism , Cell Nucleus/metabolism , Cell Proliferation , Glycogen Synthase Kinase 3 beta , Mice , Models, Biological , Multiprotein Complexes/metabolism , Protein Binding , Protein Stability , Protein Transport , RNA, Small Interfering/metabolism , Wnt Proteins
5.
PLoS One ; 5(2): e9187, 2010 Feb 12.
Article En | MEDLINE | ID: mdl-20169147

BACKGROUND: Rho kinases (ROCKs) mediate cell contraction, local adhesion, and cell motility, which are considered to be important in cell differentiation. We postulated that ROCKs are involved in controlling embryonic stem (ES) cell renewal and differentiation. METHODOLOGY/PRINCIPAL FINDINGS: CCE, a murine ES cell, was treated with Y-27632 for 48 to 96 hours and colony formation was evaluated. Y-27632 blocked CCE colony formation and induced CCE to grow as individual cells, regardless of the initial seeding cell density either at 10(4)/cm(2) ("high" seeding density) or 2x10(3)/cm(2) ("low" density). However, at high seeding density, Y-27632-treated cells exhibited reduction of alkaline phosphatase (AP) staining and Oct3/4 expression. They expressed SOX-1, nestin, and MAP2c, but not betaIII-tubulin or NG-2. They did not express endoderm or mesoderm lineage markers. After removal of Y-27632, the cells failed to form colonies or regain undifferentiated state. Silencing of ROCK-1 or ROCK-2 with selective small interference RNA induced CCE morphological changes similar to Y-27632. Silencing of ROCK-1 or ROCK-2 individually was sufficient to cause reduction of AP and Oct3/4, and expression of SOX-1, nestin, and MAP2c; and combined silencing of both ROCKs did not augment the effects exerted by individual ROCK siRNA. Y-27632-treated CCE cells seeded at 2x10(3) or 6.6x10(3) cells/cm(2) did not lose renewal factors or express differentiation markers. Furthermore, they were able to form AP-positive colonies after removal of Y-27632 and reseeding. Similar to ROCK inhibition by Y-27632, silencing of ROCK-1 or ROCK-2 in cells seeded at 2x10(3)/cm(2) did not change renewal factors. CONCLUSIONS/SIGNIFICANCE: We conclude that ROCKs promote ES cell colony formation, maintain them at undifferentiated state, and prevent them from neural differentiation at high seeding density. ROCK inhibition represents a new strategy for preparing large numbers of neural progenitor cells.


Cell Differentiation/physiology , Cell Proliferation , Embryonic Stem Cells/metabolism , rho-Associated Kinases/metabolism , Alkaline Phosphatase/metabolism , Amides/pharmacology , Animals , Blotting, Western , Cell Differentiation/drug effects , Cell Differentiation/genetics , Cell Line , Embryonic Stem Cells/cytology , Embryonic Stem Cells/drug effects , Enzyme Inhibitors/pharmacology , Gene Expression/drug effects , Intermediate Filament Proteins/genetics , Intermediate Filament Proteins/metabolism , Isoquinolines/pharmacology , Mice , Mice, Inbred Strains , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Nestin , Neurons/cytology , Neurons/drug effects , Neurons/metabolism , Octamer Transcription Factor-3/metabolism , Pyridines/pharmacology , RNA Interference , Reverse Transcriptase Polymerase Chain Reaction , Stem Cells/cytology , Stem Cells/drug effects , Stem Cells/metabolism , Sulfonamides/pharmacology , rho-Associated Kinases/antagonists & inhibitors , rho-Associated Kinases/genetics
...