Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
Add more filters










Publication year range
1.
Biomed Res Int ; 2018: 2461363, 2018.
Article in English | MEDLINE | ID: mdl-30186853

ABSTRACT

BACKGROUND: MiR-29a is known as a repressor of human cancer. However, its relevance in glioma proliferation and invasion remains largely unknown. In this study, we aimed to investigate the function and mechanism of miR-29a in glioma tumorigenesis. METHODS: The expression of miR-29a was determined by using qRT-PCR. CCK-8, wound healing, and transwell invasion assays were carried out to analyze the effects of miR-29a in glioblastoma cells. qRT-PCR, luciferase reporter, and western blot experiments were done to validate the targeting of TRAF4/Akt pathway by miR-29a. The expression correlation between levels of TRAF4 and miR-29a was analyzed. Regulation of miR-29a expression by enhanced/reduced TRAF4/Akt expression was finally confirmed by qRT-PCR. RESULTS: MiR-29a was decreased in the glioma tissues, especially in those at higher grades. Following its mimic transfection, we validated that miR-29a inhibited cell proliferation, migration, and invasion. Consistently, miR-29a inhibition induced the opposite effects on cell proliferation, migration, and invasion. We confirmed TRAF4 as a direct target of miR-29a, which might mediate the Akt pathway activation. We showed a significantly negative expression correlation between TRAF4 and miR-29a in normal and glioma tissues. Finally we observed an upregulation of miR-29a in TRAF4/Akt activated cells. CONCLUSION: MiR-29a is critical tumor suppressor for glioma tumorigenesis by forming a negative feedback loop of TRAF4/Akt signaling and represents a potent therapeutic candidate for treating gliomas.


Subject(s)
Brain Neoplasms/metabolism , Cell Proliferation , Glioma/metabolism , MicroRNAs/physiology , TNF Receptor-Associated Factor 4/physiology , Carcinogenesis , Cell Line, Tumor , Cell Movement , Gene Expression Regulation, Neoplastic , Humans , Proto-Oncogene Proteins c-akt
2.
Sci China Life Sci ; 57(12): 1172-6, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25249198

ABSTRACT

The tumor-promoting arm of transforming growth factor beta (TGF-ß) receptor signaling contributes to advanced cancer progression and is considered a master regulator of breast cancer metastasis. In mammals, there are six distinct members in the tumor-necrosis factor receptor (TNFR)-associated factor (TRAF) family (TRAF1-TRAF6), with the function of TRAF4 not being extensively studied in the past decade. Although numerous studies have suggested that there is elevated TRAF4 expression in human cancer, it is still unknown in which oncogenic pathway TRAF4 is mainly implicated. This review highlights TGF-ß-induced SMAD-dependent signaling and non-SMAD signaling as the major pathways regulated by TRAF4 involved in breast cancer metastasis.


Subject(s)
Biomarkers, Tumor/metabolism , Breast Neoplasms/metabolism , Cell Transformation, Neoplastic , Signal Transduction/physiology , TNF Receptor-Associated Factor 4/physiology , Transforming Growth Factor beta/metabolism , Breast Neoplasms/pathology , Female , Humans , Neoplasm Metastasis , Prognosis , Smad Proteins/metabolism , Ubiquitin-Protein Ligases/metabolism
3.
Biol Aujourdhui ; 208(4): 299-310, 2014.
Article in French | MEDLINE | ID: mdl-25840457

ABSTRACT

Eukaryotic epithelial cells form a sheet of contiguous cells, called epithelium, by means of the establishment of well-developed junctional complexes. These junctional complexes ensure the cell cohesion in the tissue and separate the plasma membrane into an apical and a basolateral compartment. This apicobasal polarity, which is crucial for both the architecture and the function of epithelia, is mainly maintained by tight junctions (TJS). Indeed, TJS weakening or loss disrupts the integrity of the epithelium, a process participating to the formation and progression of carcinomas. It has recently been shown that TRAF4, a protein dynamically localized in TJS and commonly overexpressed in carcinomas, plays a variety of functions in tumor progression. Here, we review recent data implicating TRAF4 in carcinogenesis. First, the conserved TRAF proteins family will be presented, and then the molecular mechanism addressing TRAF4 to TJS which involves lipid binding by the TRAF domain will be described. The various roles of TRAF4 in carcinogenesis will be discussed. Finally, we will highlight the ability of all TRAF proteins to bind lipids and discuss its potential functional relevance.


Subject(s)
Carcinoma/pathology , Neoplasm Proteins/physiology , TNF Receptor-Associated Factor 4/physiology , Amino Acid Motifs , Amino Acid Sequence , Binding Sites , Carcinoma/metabolism , Cell Compartmentation , Cell Movement , Cell Polarity/physiology , Conserved Sequence , Disease Progression , Epithelial Cells/pathology , Epithelial-Mesenchymal Transition , Humans , Membrane Lipids/metabolism , Membrane Proteins/metabolism , Models, Molecular , Molecular Sequence Data , Multigene Family , Phosphatidylinositol Phosphates/metabolism , Protein Conformation , Sequence Alignment , Sequence Homology, Amino Acid , Signal Transduction/physiology , TNF Receptor-Associated Factor 4/genetics , Tight Junctions/physiology , Transforming Growth Factor beta/physiology , Tumor Necrosis Factor Receptor-Associated Peptides and Proteins/chemistry , Tumor Necrosis Factor Receptor-Associated Peptides and Proteins/genetics , Tumor Necrosis Factor Receptor-Associated Peptides and Proteins/physiology
4.
PLoS Biol ; 11(12): e1001726, 2013 Dec.
Article in English | MEDLINE | ID: mdl-24311986

ABSTRACT

Tumor necrosis factor (TNF) receptor-associated factor 4 (TRAF4) is frequently overexpressed in carcinomas, suggesting a specific role in cancer. Although TRAF4 protein is predominantly found at tight junctions (TJs) in normal mammary epithelial cells (MECs), it accumulates in the cytoplasm of malignant MECs. How TRAF4 is recruited and functions at TJs is unclear. Here we show that TRAF4 possesses a novel phosphoinositide (PIP)-binding domain crucial for its recruitment to TJs. Of interest, this property is shared by the other members of the TRAF protein family. Indeed, the TRAF domain of all TRAF proteins (TRAF1 to TRAF6) is a bona fide PIP-binding domain. Molecular and structural analyses revealed that the TRAF domain of TRAF4 exists as a trimer that binds up to three lipids using basic residues exposed at its surface. Cellular studies indicated that TRAF4 acts as a negative regulator of TJ and increases cell migration. These functions are dependent from its ability to interact with PIPs. Our results suggest that TRAF4 overexpression might contribute to breast cancer progression by destabilizing TJs and favoring cell migration.


Subject(s)
Cell Movement/physiology , TNF Receptor-Associated Factor 4/physiology , Tight Junctions/physiology , Animals , COS Cells , Cell Membrane/physiology , Chlorocebus aethiops , Humans , Phosphatidylinositols/physiology , Recombinant Proteins
5.
Cancer Res ; 73(23): 6938-50, 2013 Dec 01.
Article in English | MEDLINE | ID: mdl-24154876

ABSTRACT

TRAF4 is an adapter protein overexpressed in certain cancers, but its contributions to tumorigenesis are unclear. In lung cancer cells and primary lung tumors, we found that TRAF4 is overexpressed. RNA interference-mediated attenuation of TRAF4 expression blunted the malignant phenotype in this setting, exerting inhibitory effects on cell proliferation, anchorage-independent growth, and tumor development in a xenograft mouse model. Unexpectedly, we discovered that TRAF4, but not Skp2, was required for activation of the pivotal cell survival kinase Akt through ubiquitination. Furthermore, TRAF4 attenuation impaired glucose metabolism by inhibiting expression of Glut1 and HK2 mediated by the Akt pathway. Overall, our work suggests that TRAF4 offers a candidate molecular target for lung cancer prevention and therapy.


Subject(s)
Adenocarcinoma/metabolism , Carcinoma, Squamous Cell/metabolism , Lung Neoplasms/metabolism , Oncogene Protein v-akt/metabolism , TNF Receptor-Associated Factor 4/physiology , Adenocarcinoma/genetics , Animals , Carcinoma, Squamous Cell/genetics , Cells, Cultured , Enzyme Activation , Gene Expression Regulation, Neoplastic/drug effects , Humans , Lung Neoplasms/genetics , Mice , Mice, Nude , NIH 3T3 Cells , RNA, Small Interfering/pharmacology , TNF Receptor-Associated Factor 4/antagonists & inhibitors , Xenograft Model Antitumor Assays
6.
J Immunol ; 189(1): 33-7, 2012 Jul 01.
Article in English | MEDLINE | ID: mdl-22649194

ABSTRACT

The effector T cell subset, Th17, plays a significant role in the pathogenesis of multiple sclerosis and of other autoimmune diseases. The signature cytokine, IL-17, engages the IL-17R and recruits the E3-ligase NF-κB activator 1 (Act1) upon stimulation. In this study, we examined the role of TNFR-associated factor (TRAF)4 in IL-17 signaling and Th17-mediated autoimmune encephalomyelitis. Primary cells from TRAF4-deficient mice displayed markedly enhanced IL-17-activated signaling pathways and induction of chemokine mRNA. Adoptive transfer of MOG35-55 specific wild-type Th17 cells into TRAF4-deficient recipient mice induced an earlier onset of disease. Mechanistically, we found that TRAF4 and TRAF6 used the same TRAF binding sites on Act1, allowing the competition of TRAF4 with TRAF6 for the interaction with Act1. Taken together, the results of this study reveal the necessity of a unique role of TRAF4 in restricting the effects of IL-17 signaling and Th17-mediated disease.


Subject(s)
Encephalomyelitis, Autoimmune, Experimental/immunology , Encephalomyelitis, Autoimmune, Experimental/pathology , Interleukin-17/antagonists & inhibitors , Interleukin-17/physiology , Signal Transduction/immunology , TNF Receptor-Associated Factor 4/physiology , Animals , Encephalomyelitis, Autoimmune, Experimental/genetics , HEK293 Cells , HeLa Cells , Humans , Interleukin-17/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , RNA, Messenger/biosynthesis , Signal Transduction/genetics , TNF Receptor-Associated Factor 4/deficiency , TNF Receptor-Associated Factor 4/genetics , Th17 Cells/immunology , Th17 Cells/pathology
7.
J Immunol ; 183(7): 4560-8, 2009 Oct 01.
Article in English | MEDLINE | ID: mdl-19752230

ABSTRACT

The TNF-associated factor (TRAF) family, the crucial adaptor group in innate immune signaling, increased to 24 in amphioxus, the oldest lineage of the Chordata. To address how these expanded molecules evolved to adapt to the changing TRAF mediated signaling pathways, here we conducted genomic and functional comparisons of four distinct amphioxus TRAF groups with their human counterparts. We showed that lineage-specific duplication and rearrangement were responsible for the expansion of amphioxus TRAF1/2 and 3 lineages, whereas TRAF4 and 6 maintained a relatively stable genome and protein structure. Amphioxus TRAF1/2 and 3 molecules displayed various expression patterns in response to microbial infection, and some of them can attenuate the NF-kappaB activation mediated by human TRAF2 and 6. Amphioxus TRAF4 presented two unique functions: activation of the NF-kappaB pathway and involvement in somite formation. Although amphioxus TRAF6 was conserved in activating NF-kappaB pathway for antibacterial defense, the mechanism was not the same as that observed in humans. In summary, our findings reveal the evolutionary uniqueness of the TRAF family in this basal chordate, and suggest that genomic duplication and functional divergence of the TRAF family are important for the current form of the TRAF-mediated signaling pathways in humans.


Subject(s)
Chordata/genetics , Chordata/physiology , Multigene Family/immunology , Proteomics , Tumor Necrosis Factor Receptor-Associated Peptides and Proteins/genetics , Tumor Necrosis Factor Receptor-Associated Peptides and Proteins/physiology , Animals , Cell Line , Drosophila melanogaster/chemistry , Drosophila melanogaster/genetics , Drosophila melanogaster/immunology , HeLa Cells , Humans , Molecular Sequence Data , Multigene Family/genetics , NF-kappa B/metabolism , Proteomics/methods , Signal Transduction/genetics , Signal Transduction/immunology , TNF Receptor-Associated Factor 2/chemistry , TNF Receptor-Associated Factor 2/genetics , TNF Receptor-Associated Factor 2/physiology , TNF Receptor-Associated Factor 3/chemistry , TNF Receptor-Associated Factor 3/genetics , TNF Receptor-Associated Factor 3/physiology , TNF Receptor-Associated Factor 4/chemistry , TNF Receptor-Associated Factor 4/genetics , TNF Receptor-Associated Factor 4/physiology , TNF Receptor-Associated Factor 6/chemistry , TNF Receptor-Associated Factor 6/genetics , TNF Receptor-Associated Factor 6/physiology
8.
PLoS One ; 3(10): e3518, 2008.
Article in English | MEDLINE | ID: mdl-18953416

ABSTRACT

BACKGROUND: Despite numerous in vivo evidences that Tumor Necrosis Factor Receptor-Associated Factor 4 (TRAF4) plays a key biological function, how it works at the cellular and molecular level remains elusive. METHODOLOGY/PRINCIPAL FINDINGS: In the present study, we show using immunofluorescence and immuohistochemistry that TRAF4 is a novel player at the tight junctions (TJs). TRAF4 is connected to assembled TJs in confluent epithelial cells, but accumulates in the cytoplasm and/or nucleus when TJs are open in isolated cells or EGTA-treated confluent cells. In vivo, TRAF4 is consistently found at TJs in normal human mammary epithelia as well as in well-differentiated in situ carcinomas. In contrast, TRAF4 is never localized at the plasma membrane of poorly-differentiated invasive carcinomas devoid of correct TJs, but is observed in the cytoplasm and/or nucleus of the cancer cells. Moreover, TRAF4 TJ subcellular localization is remarkably dynamic. Fluorescence recovery after photobleaching (FRAP) experiments show that TRAF4 is highly mobile and shuttles between TJs and the cytoplasm. Finally, we show that intracellular TRAF4 potentiates ERK1/2 phosphorylation in proliferating HeLa cells, an epithelial cell line known to be devoid of TJs. CONCLUSIONS/SIGNIFICANCE: Collectively, our data strongly support the new concept of TJs as a dynamic structure. Moreover, our results implicate TRAF4 in one of the emerging TJ-dependent signaling pathways that responds to cell polarity by regulating the cell proliferation/differentiation balance, and subsequently epithelium homeostasis. Drastic phenotypes or lethality in TRAF4-deficient mice and drosophila strongly argue in favor of such a function.


Subject(s)
Homeostasis , Mammary Glands, Human/metabolism , TNF Receptor-Associated Factor 4/physiology , Tight Junctions/metabolism , Animals , Cell Communication/physiology , Cell Differentiation/physiology , Cell Line, Transformed , Cell Membrane/metabolism , Cell Nucleus/metabolism , Cell Polarity/physiology , Cell Proliferation , Cells, Cultured , Dogs , HeLa Cells , Homeostasis/physiology , Humans , Mammary Glands, Human/physiology , Mitogen-Activated Protein Kinase 3/metabolism , Models, Biological , Protein Transport/physiology , Signal Transduction/physiology , TNF Receptor-Associated Factor 4/metabolism
9.
Cancer Biol Ther ; 6(12): 1986-90, 2007 Dec.
Article in English | MEDLINE | ID: mdl-18087216

ABSTRACT

p63, a member of the p53 family, is overexpressed in squamous cell carcinoma of the head and neck (SCCHN) and some other tumors of epithelial origin. As a transcription factor, p63 can bind to p53-type response elements and there is some overlap between p53 family transcriptional targets. Tumor necrosis factor receptor associated factor 4 (TRAF4) is a p53 regulated gene which is overexpressed in many human carcinomas. We investigated the involvement of p63 in regulation of TRAF4 and the expression of the TRAF4 protein in SCCHN. Disrupting endogenous p63 expression resulted in downregulation of TRAF4 mRNA and protein in an SCCHN cell line. Endogenous p63 bound to the TRAF4 promoter in vivo and reporter assays showed that p63, p73 and p53 can all transactivate TRAF4, with TAp63 isoforms being the most potent activators. The level of TRAF4 activation by TAp63 was two-fold higher than by p53, and TRAF4 was ten-fold more responsive to TAp63 than another p63-target, IGFBP3. Nuclear expression of TRAF4 was seen in normal oral epithelium and highly/moderately differentiated SCCHN, whereas cytoplasmic expression of TRAF4 was seen in poorly differentiated SCCHN. These results indicate that TRAF4 is a common target of p53 family members and that localization of TRAF4 is associated with differentiation of SCCHN cells.


Subject(s)
Carcinoma, Squamous Cell/pathology , Gene Expression Regulation, Neoplastic/physiology , Head and Neck Neoplasms/pathology , Neoplasm Proteins/physiology , TNF Receptor-Associated Factor 4/physiology , Trans-Activators/physiology , Tumor Suppressor Proteins/physiology , Carcinoma, Squamous Cell/metabolism , Cell Differentiation , Cell Nucleus/metabolism , Cytoplasm/metabolism , Epithelial Cells/metabolism , Gene Expression Regulation, Neoplastic/drug effects , Genes, Tumor Suppressor , Genes, p53 , Head and Neck Neoplasms/metabolism , Humans , Mouth Mucosa/cytology , Multigene Family , Neoplasm Proteins/biosynthesis , Neoplasm Proteins/genetics , Protein Isoforms/physiology , RNA, Small Interfering/pharmacology , Recombinant Fusion Proteins/physiology , TNF Receptor-Associated Factor 4/biosynthesis , TNF Receptor-Associated Factor 4/genetics , Transcription Factors , Transcription, Genetic , Tumor Suppressor Protein p53/physiology
10.
Adv Exp Med Biol ; 597: 60-71, 2007.
Article in English | MEDLINE | ID: mdl-17633017

ABSTRACT

The fourth member of the TRAF protein family (TRAF4) presents several characteristics that distinguish it from the other members of the family. These characteristics concern the primary sequence of the protein, a strong evolutionary conservation, and a tightly regulated physiological expression during development. The subcellular localization of TRAF4 is controversial as it has been detected at the cell membrane, in the cytoplasm and in the nucleus. Using mouse and fly models, it has been established that TRAF4 is a key molecule in diverse ontogenic processes, particularly in the nervous system. However, the molecular mechanisms of action of TRAF4 remain evasive as it was found to interact with diverse types of proteins, leading either to pro-apoptotic or anti-apoptotic functions. Finally, few studies implicated TRAF4 in human diseases.


Subject(s)
TNF Receptor-Associated Factor 4/physiology , Animals , Humans , TNF Receptor-Associated Factor 4/chemistry , TNF Receptor-Associated Factor 4/genetics , TNF Receptor-Associated Factor 4/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...