Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 7 de 7
Filter
Add more filters










Database
Language
Publication year range
1.
PLoS One ; 7(9): e46485, 2012.
Article in English | MEDLINE | ID: mdl-23029530

ABSTRACT

Expression of the hominoid-specific TBC1D3 oncoprotein enhances growth factor receptor signaling and subsequently promotes cellular proliferation and survival. Here we report that TBC1D3 is degraded in response to growth factor signaling, suggesting that TBC1D3 expression is regulated by a growth factor-driven negative feedback loop. To gain a better understanding of how TBC1D3 is regulated, we studied the effects of growth factor receptor signaling on TBC1D3 post-translational processing and turnover. Using a yeast two-hybrid screen, we identified CUL7, the scaffolding subunit of the CUL7 E3 ligase complex, as a TBC1D3-interacting protein. We show that CUL7 E3 ligase ubiquitinates TBC1D3 in response to serum stimulation. Moreover, TBC1D3 recruits F-box 8 (Fbw8), the substrate recognition domain of CUL7 E3 ligase, in pull-down experiments and in an in vitro assay. Importantly, alkaline phosphatase treatment of TBC1D3 suppresses its ability to recruit Fbw8, indicating that TBC1D3 phosphorylation is critical for its ubiquitination and degradation. We conclude that serum- and growth factor-stimulated TBC1D3 ubiquitination and degradation are regulated by its interaction with CUL7-Fbw8.


Subject(s)
Cullin Proteins/metabolism , F-Box Proteins/metabolism , GTPase-Activating Proteins/metabolism , Protein Processing, Post-Translational , Proteolysis , Proto-Oncogene Proteins/metabolism , HeLa Cells , Humans , Intercellular Signaling Peptides and Proteins/physiology , Leupeptins/pharmacology , Phosphorylation , Proteasome Inhibitors/pharmacology , Protein Binding , Two-Hybrid System Techniques , Ubiquitin-Protein Ligases/metabolism , Ubiquitination
2.
PLoS One ; 7(2): e31225, 2012.
Article in English | MEDLINE | ID: mdl-22348058

ABSTRACT

Insulin/IGF-1 signaling plays a pivotal role in the regulation of cellular homeostasis through its control of glucose metabolism as well as due to its effects on cell proliferation. Aberrant regulation of insulin signaling has been repeatedly implicated in uncontrolled cell growth and malignant transformations. TBC1D3 is a hominoid specific gene previously identified as an oncogene in breast and prostate cancers. Our efforts to identify the molecular mechanisms of TBC1D3-induced oncogenesis revealed the role of TBC1D3 in insulin/IGF-1 signaling pathway. We document here that TBC1D3 intensifies insulin/IGF-1-induced signal transduction through intricate, yet elegant fine-tuning of signaling mechanisms. We show that TBC1D3 expression substantially delayed ubiquitination and degradation of insulin receptor substrate-1 (IRS-1). This effect is achieved through suppression of serine phosphorylation at S636/639, S307 and S312 of IRS-1, which are key phosphorylation sites required for IRS-1 degradation. Furthermore, we report that the effect of TBC1D3 on IRS-1:S636/639 phosphorylation is mediated through TBC1D3-induced activation of protein phosphatase 2A (PP2A), followed by suppression of T389 phosphorylation on p70 S6 kinase (S6K). TBC1D3 specifically interacts with PP2A regulatory subunit B56γ, indicating that TBC1D3 and PP2A B56γ operate jointly to promote S6K:T389 dephosphorylation. These findings suggest that TBC1D3 plays an unanticipated and potentially unique role in the fine-tuning of insulin/IGF-1 signaling, while providing novel insights into the regulation of tumorigenesis by a hominoid-specific protein.


Subject(s)
GTPase-Activating Proteins/genetics , Insulin Receptor Substrate Proteins/metabolism , Insulin/metabolism , Proteolysis , Proto-Oncogene Proteins/genetics , Ribosomal Protein S6 Kinases, 70-kDa/metabolism , Signal Transduction , GTPase-Activating Proteins/metabolism , Humans , Phosphorylation , Protein Phosphatase 2/metabolism , Proto-Oncogene Proteins/metabolism
3.
Sci Signal ; 2(89): pe59, 2009 Sep 22.
Article in English | MEDLINE | ID: mdl-19797272

ABSTRACT

The identification and characterization of human-specific genes and the cellular processes that the encoded proteins control have the potential to help us understand at the molecular level what makes humans different from other species. The sequencing of the human genome and the genomes of closely related primates has revealed the presence of a small number of human- or human-lineage-specific genes that have no orthologs in lower species. Human-specific and human-lineage-specific genes are likely to function as regulators of cell signaling events, and by fine-tuning pathways, the encoded proteins may contribute to human-specific characteristics and behaviors. In addition, human-specific genes may represent biomarkers for examining human-specific characteristics of various diseases. Investigation of the gene encoding TBC1D3 is one example of a search that may lead to understanding the evolution and the function of human-specific genes, because it is absent in lower species and present in high copy number in the human genome.


Subject(s)
Signal Transduction/genetics , Animals , Biological Evolution , GTPase-Activating Proteins/genetics , GTPase-Activating Proteins/physiology , Hominidae/genetics , Hominidae/physiology , Humans , Models, Genetic , Oncogene Proteins/genetics , Oncogene Proteins/physiology , Proto-Oncogene Proteins , Receptors, Growth Factor/physiology , Signal Transduction/physiology , Species Specificity
4.
J Biol Chem ; 283(19): 13233-42, 2008 May 09.
Article in English | MEDLINE | ID: mdl-18319245

ABSTRACT

Hominoid- and human-specific genes may have evolved to modulate signaling pathways of a higher order of complexity. TBC1D3 is a hominoid-specific oncogene encoded by a cluster of eight paralogs on chromosome 17. Initial work indicates that TBC1D3 is widely expressed in human tissues ( Hodzic, D., Kong, C., Wainszelbaum, M. J., Charron, A. J., Su, X., and Stahl, P. D. (2006) Genomics 88, 731-736 ). In this study, we show that TBC1D3 expression has a powerful effect on cell proliferation that is further enhanced by epidermal growth factor (EGF) in both human and mouse cell lines. EGF activation of the Erk and protein kinase B/Akt pathways is enhanced, both in amplitude and duration, by TBC1D3 expression, whereas RNA interference silencing of TBC1D3 suppresses the activation. Light microscopy and Western blot experiments demonstrate that increased signaling in response to EGF is coupled with a significant delay in EGF receptor (EGFR) trafficking and degradation, which significantly extends the life span of EGFR. Moreover, TBC1D3 suppresses polyubiquitination of the EGFR and the recruitment of c-Cbl. Using the Ras binding domain of Raf1 to monitor GTP-Ras we show that TBC1D3 expression enhances Ras activation in quiescent cells, which is further increased by EGF treatment. We speculate that TBC1D3 may alter Ras GTP loading. We conclude that the expression of TBC1D3 generates a delay in EGFR degradation, a decrease in ubiquitination, and a failure to recruit adapter proteins that ultimately dysregulate EGFR signal transduction and enhance cell proliferation. Altered growth factor receptor trafficking and GTP-Ras turnover may be sites where recently evolved genes such as TBC1D3 selectively modulate signaling in hominoids and humans.


Subject(s)
ErbB Receptors/metabolism , GTPase-Activating Proteins/metabolism , MAP Kinase Signaling System , Oncogene Proteins/metabolism , ras Proteins/metabolism , Cell Line , Cell Membrane/metabolism , Cell Proliferation/drug effects , Enzyme Activation , Epidermal Growth Factor/pharmacology , GTPase-Activating Proteins/genetics , Humans , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Oncogene Proteins/genetics , Protein Binding , Proto-Oncogene Proteins , Proto-Oncogene Proteins c-akt/metabolism , Proto-Oncogene Proteins c-cbl/metabolism , Substrate Specificity , Ubiquitination
5.
Genomics ; 88(6): 731-736, 2006 Dec.
Article in English | MEDLINE | ID: mdl-16863688

ABSTRACT

TBC1D3 is a member of the TBC1 domain family of proteins that stimulates the intrinsic GTPase activity of RAB5A, an essential actor in early endosome trafficking. Oncogenic properties of TBC1D3 have been demonstrated previously both in vitro and in mouse models. Although the oncogenic mechanism of TBC1D3 has yet to be elucidated, the TBC1D3 locus (chromosome 17q12) is amplified in 15% of primary prostate tumors. Here, we describe eight highly related TBC1D3 paralogues located within that genomic region, potentially encoding six variant TBC1D3 proteins. We found that human tissues display specific transcription patterns of these paralogues. Furthermore, that pattern was altered in several primary prostate tumors in comparison to healthy prostate tissues. Potential TBC1D3 oncogenic mechanisms are discussed in light of these results.


Subject(s)
Chromosomes, Human, Pair 17/genetics , GTPase-Activating Proteins/genetics , Gene Dosage , Genetic Variation , Oncogene Proteins/genetics , Oncogenes/genetics , Chromosome Mapping , Female , GTPase-Activating Proteins/metabolism , Humans , Male , Oncogene Proteins/metabolism , Organ Specificity , Placenta/metabolism , Prostate/metabolism , Prostatic Neoplasms/genetics , Proto-Oncogene Proteins , RNA, Messenger/genetics , RNA, Messenger/metabolism
6.
Exp Cell Res ; 312(12): 2238-51, 2006 Jul 15.
Article in English | MEDLINE | ID: mdl-16650848

ABSTRACT

The endosomal compartment and the plasma membrane form a complex partnership that controls signal transduction and trafficking of different molecules. The specificity and functionality of the early endocytic pathway are regulated by a growing number of Rab GTPases, particularly Rab5. In this study, we demonstrate that IL4 (a Th-2 cytokine) and prostaglandin E2 (PGE2) synergistically induce Rab5 and several Rab effector proteins, including Rin1 and EEA1, and promote the formation of an enlarged early endocytic (EEE) compartment. Endosome enlargement is linked to a substantial induction of the mannose receptor (MR), a well-characterized macrophage endocytic receptor. Both MR levels and MR-mediated endocytosis are enhanced approximately 7-fold. Fluid-phase endocytosis is also elevated in treated cells. Light microscopy and fractionation studies reveal that MR colocalizes predominantly with Rab5a and partially with Rab11, an endosomal recycling pathway marker. Using retroviral expression of Rab5a:S34N, a dominant negative mutant, and siRNA Rab5a silencing, we demonstrate that Rab5a is essential for the large endosome phenotype and for localization of MR in these structures. We speculate that the EEE is maintained by activated Rab5, and that the EEE phenotype is part of some macrophage developmental program such as cell fusion, a characteristic of IL4-stimulated cells.


Subject(s)
Dinoprostone/pharmacology , Endosomes/drug effects , Interleukin-4/pharmacology , Macrophages/drug effects , rab5 GTP-Binding Proteins/metabolism , Animals , Autoantigens/metabolism , Carrier Proteins/metabolism , Drug Synergism , Endocytosis/drug effects , Endosomes/physiology , Endosomes/ultrastructure , Intracellular Signaling Peptides and Proteins , Lectins, C-Type/analysis , Lectins, C-Type/metabolism , Macrophages/metabolism , Macrophages/physiology , Male , Mannans/metabolism , Mannose Receptor , Mannose-Binding Lectins/analysis , Mannose-Binding Lectins/metabolism , Membrane Proteins/metabolism , Mice , Microscopy, Electron , Pinocytosis/drug effects , RNA, Small Interfering/genetics , Receptors, Cell Surface/analysis , Receptors, Cell Surface/metabolism , Vesicular Transport Proteins , rab GTP-Binding Proteins/analysis , rab GTP-Binding Proteins/metabolism , rab5 GTP-Binding Proteins/analysis , rab5 GTP-Binding Proteins/genetics , rab7 GTP-Binding Proteins , ras GTPase-Activating Proteins/metabolism
7.
Biochem J ; 375(Pt 3): 705-12, 2003 Nov 01.
Article in English | MEDLINE | ID: mdl-12887332

ABSTRACT

Intestinal extracts of Triatoma infestans induce cell differentiation of Trypanosoma cruzi epimastigotes into the infective metacyclic form. Part of this effect can be explained by the presence of haemoglobin fragments, which stimulate trypanosomal adenylate cyclase. In this work we examined the metacyclogenic activity of lipids present in this intestinal extract. We found that lipid extracts of the intestinal extract have significant stimulatory effects that reside with the free-fatty-acid fraction, especially oleic acid. These compounds stimulate de novo diacylglycerol formation and protein kinase C activity in the parasite. Moreover, metacyclogenesis is stimulated by phorbol esters and cell-permeant diacylglycerol, while protein kinase C down-regulation or incubation with inhibitors of this kinase abrogates this effect. These results indicate that free fatty acids are a novel signal, inducing metacyclogenesis, acting through a pathway involving diacylglycerol biosynthesis and protein kinase C activation.


Subject(s)
Cell Differentiation/drug effects , Fatty Acids, Nonesterified/pharmacology , Sulfonamides , Trypanosoma cruzi/drug effects , 1-(5-Isoquinolinesulfonyl)-2-Methylpiperazine/pharmacology , Animals , Bucladesine/pharmacology , Cell Division/drug effects , Cyclic AMP-Dependent Protein Kinases/metabolism , Diglycerides/biosynthesis , Diglycerides/pharmacology , Enzyme Activation/drug effects , Enzyme Inhibitors/pharmacology , Fatty Acids, Nonesterified/pharmacokinetics , Intestines/chemistry , Isoquinolines/pharmacology , Oleic Acid/pharmacokinetics , Oleic Acid/pharmacology , Phosphorylation/drug effects , Protein Kinase C/antagonists & inhibitors , Protein Kinase C/metabolism , Tetradecanoylphorbol Acetate/pharmacology , Triatoma/chemistry , Trypanosoma cruzi/cytology , Trypanosoma cruzi/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL