Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
Add more filters










Publication year range
1.
PLoS One ; 14(7): e0219143, 2019.
Article in English | MEDLINE | ID: mdl-31260484

ABSTRACT

Receptor Tyrosine Kinase (RTK) signaling is essential for normal biological processes and disruption of this regulation can lead to tumor initiation and progression. Cbl proteins (Cbl, Cbl-b and Cbl-c) are a family of RING finger (RF) ubiquitin ligases that negatively regulate a variety of RTKs, including EGFR, MET, and RET. Recent studies have identified Cbl mutations associated with human myeloid neoplasias in approximately 5% of the cases. Cbl-c is the most recently identified human Cbl protein and is expressed exclusively in epithelial cells. We identified a novel cDNA that was isolated from a mouse mammary cancer from the C3(1) Large T Antigen transgenic model. This mutant cDNA encodes a protein that has a deletion in the RF domain of Cbl-c, thereby resembling known Cbl family mutations associated with myeoloid neoplasias. Genomic analysis of both parental and transgenic lines shows no evidence of germline mutation indicating that this mutation is likely a somatic mutation. The mutant protein enhances transformation of NIH 3T3 cells when expressed in combination with SV40 Large T antigen. Together these data are consistent with a second hit mutation. In overexpression studies, this mutant Cbl-c protein fails to mediate ubiquitination of activated EGFR and acts in a dominant negative fashion to prevent ubiquitination and downregulation of the activated EGFR by wild type Cbl proteins. Mechanistically, the mutant Cbl-c binds to the EGFR and prevents recruitment of the wild type Cbl protein. Furthermore, data mining reveals Cbl-c mutations associated with solid tumors in humans. Subsequent cell-based analysis demonstrates a similar loss of E3 function and dominant negative effects for one of these human mutations. These data suggest that like Cbl mutations in myeloid neoplasms, loss of Cbl-c function may contribute to the pathogenesis of solid tumors in murine models and in humans.


Subject(s)
Loss of Function Mutation , Neoplasms/genetics , Proto-Oncogene Proteins c-cbl/genetics , Amino Acid Sequence , Animals , Antigens, Viral, Tumor/genetics , Base Sequence , Cell Transformation, Neoplastic/genetics , Female , HEK293 Cells , Humans , Male , Mammary Neoplasms, Experimental/genetics , Mice , Mice, Transgenic , Mutant Proteins/chemistry , Mutant Proteins/genetics , Mutant Proteins/metabolism , NIH 3T3 Cells , Neoplasms/metabolism , Proto-Oncogene Proteins c-cbl/chemistry , Proto-Oncogene Proteins c-cbl/metabolism , RING Finger Domains/genetics , Receptor Protein-Tyrosine Kinases/metabolism , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Sequence Deletion , Signal Transduction
2.
Breast Cancer Res ; 16(2): R41, 2014 Apr 17.
Article in English | MEDLINE | ID: mdl-24745479

ABSTRACT

INTRODUCTION: Tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) binds to its receptors, TRAIL-receptor 1 (TRAIL-R1) and TRAIL-receptor 2 (TRAIL-R2), leading to apoptosis by activation of caspase-8 and the downstream executioner caspases, caspase-3 and caspase-7 (caspase-3/7). Triple-negative breast cancer (TNBC) cell lines with a mesenchymal phenotype are sensitive to TRAIL, whereas other breast cancer cell lines are resistant. The underlying mechanisms that control TRAIL sensitivity in breast cancer cells are not well understood. Here, we performed small interfering RNA (siRNA) screens to identify molecular regulators of the TRAIL pathway in breast cancer cells. METHODS: We conducted siRNA screens of the human kinome (691 genes), phosphatome (320 genes), and about 300 additional genes in the mesenchymal TNBC cell line MB231. Forty-eight hours after transfection of siRNA, parallel screens measuring caspase-8 activity, caspase-3/7 activity, or cell viability were conducted in the absence or presence of TRAIL for each siRNA, relative to a negative control siRNA (siNeg). A subset of genes was screened in cell lines representing epithelial TNBC (MB468), HER2-amplified breast cancer (SKBR3), and estrogen receptor-positive breast cancer (T47D). Selected putative negative regulators of the TRAIL pathway were studied by using small-molecule inhibitors. RESULTS: The primary screens in MB231 identified 150 genes, including 83 kinases, 4 phosphatases, and 63 nonkinases, as potential negative regulators of TRAIL. The identified genes are involved in many critical cell processes, including apoptosis, growth factor-receptor signaling, cell-cycle regulation, transcriptional regulation, and DNA repair. Gene-network analysis identified four genes (PDPK1, IKBKB, SRC, and BCL2L1) that formed key nodes within the interaction network of negative regulators. A secondary screen of a subset of the genes identified in additional cell lines representing different breast cancer subtypes and sensitivities to TRAIL validated and extended these findings. Further, we confirmed that small-molecule inhibition of SRC or BCL2L1, in combination with TRAIL, sensitizes breast cancer cells to TRAIL-induced apoptosis, including cell lines resistant to TRAIL-induced cytotoxicity. CONCLUSIONS: These data identify novel molecular regulators of TRAIL-induced apoptosis in breast cancer cells and suggest strategies for the enhanced application of TRAIL as a therapy for breast cancer.


Subject(s)
Apoptosis/drug effects , Apoptosis/genetics , Gene Expression Regulation, Neoplastic/drug effects , RNA Interference , TNF-Related Apoptosis-Inducing Ligand/pharmacology , Biphenyl Compounds/pharmacology , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Caspase 3/genetics , Caspase 3/metabolism , Caspase 7/genetics , Caspase 7/metabolism , Caspase 8/genetics , Caspase 8/metabolism , Cell Line, Tumor , Cell Survival/drug effects , Cell Survival/genetics , Cysteine Proteinase Inhibitors/pharmacology , Drug Resistance, Neoplasm/genetics , Humans , Immunoblotting , Nitrophenols/pharmacology , Oligopeptides/pharmacology , Piperazines/pharmacology , Pyrimidines/pharmacology , Sulfonamides/pharmacology , bcl-X Protein/antagonists & inhibitors , bcl-X Protein/genetics , bcl-X Protein/metabolism , src-Family Kinases/antagonists & inhibitors , src-Family Kinases/genetics , src-Family Kinases/metabolism
3.
PLoS One ; 9(1): e87116, 2014.
Article in English | MEDLINE | ID: mdl-24466333

ABSTRACT

The Cbl proteins (Cbl, Cbl-b, and Cbl-c) are a highly conserved family of RING finger ubiquitin ligases (E3s) that function as negative regulators of tyrosine kinases in a wide variety of signal transduction pathways. In this study, we identify a new Cbl-c interacting protein, Enigma (PDLIM7). This interaction is specific to Cbl-c as Enigma fails to bind either of its closely related homologues, Cbl and Cbl-b. The binding between Enigma and Cbl-c is mediated through the LIM domains of Enigma as removal of all three LIM domains abrogates this interaction, while only LIM1 is sufficient for binding. Here we show that Cbl-c binds wild-type and MEN2A isoforms of the receptor tyrosine kinase, RET, and that Cbl-c enhances ubiquitination and degradation of activated RET. Enigma blocks Cbl-c-mediated RETMEN2A ubiquitination and degradation. Cbl-c decreased downstream ERK activation by RETMEN2A and co-expression of Enigma blocked the Cbl-c-mediated decrease in ERK activation. Enigma showed no detectable effect on Cbl-c-mediated ubiquitination of activated EGFR suggesting that this effect is specific to RET. Through mapping studies, we show that Cbl-c and Enigma bind RETMEN2A at different residues. However, binding of Enigma to RETMENA prevents Cbl-c recruitment to RETMEN2A. Consistent with these biochemical data, exploratory analyses of breast cancer patients with high expression of RET suggest that high expression of Cbl-c correlates with a good outcome, and high expression of Enigma correlates with a poor outcome. Together, these data demonstrate that Cbl-c can ubiquitinate and downregulate RETMEN2A and implicate Enigma as a positive regulator of RETMEN2A through blocking of Cbl-mediated ubiquitination and degradation.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Breast Neoplasms/metabolism , Cytoskeletal Proteins/metabolism , LIM Domain Proteins/metabolism , Multiple Endocrine Neoplasia Type 2a/metabolism , Pancreatic Neoplasms/metabolism , Proto-Oncogene Proteins c-cbl/metabolism , Proto-Oncogene Proteins c-ret/metabolism , Blotting, Western , Breast Neoplasms/mortality , Breast Neoplasms/pathology , Cells, Cultured , Female , HEK293 Cells , Humans , Immunoprecipitation , Microarray Analysis , Pancreatic Neoplasms/pathology , Proteolysis , Two-Hybrid System Techniques , Ubiquitination
4.
PLoS One ; 7(11): e49428, 2012.
Article in English | MEDLINE | ID: mdl-23145173

ABSTRACT

Cbl proteins (Cbl, Cbl-b and Cbl-c) are ubiquitin ligases that are critical regulators of tyrosine kinase signaling. In this study we identify a new Cbl-c interacting protein, Hydrogen peroxide Induced Construct 5 (Hic-5). The two proteins interact through a novel interaction mediated by the RING finger of Cbl-c and the LIM2 domain of Hic-5. Further, this interaction is mediated and dependent on specific zinc coordinating complexes within the RING finger and LIM domain. Binding of Hic-5 to Cbl-c leads to an increase in the ubiquitin ligase activity of Cbl-c once Cbl-c has been activated by Src phosphorylation or through an activating phosphomimetic mutation. In addition, co-transfection of Hic-5 with Cbl-c leads to an increase in Cbl-c mediated ubiquitination of the EGFR. These data suggest that Hic-5 enhances Cbl-c ubiquitin ligase activity once Cbl-c has been phosphorylated and activated. Interactions between heterologous RING fingers have been shown to activate E3s. This is the first demonstration of enhancement of ubiquitin ligase activity of a RING finger ubiquitin ligase by the direct interaction of a LIM zinc coordinating domain.


Subject(s)
Intracellular Signaling Peptides and Proteins/metabolism , LIM Domain Proteins/metabolism , Proto-Oncogene Proteins c-cbl/metabolism , RING Finger Domains , Cell Line , ErbB Receptors/metabolism , HEK293 Cells , HeLa Cells , Humans , Protein Interaction Domains and Motifs , Protein Interaction Mapping , Protein Structure, Tertiary , Proto-Oncogene Proteins c-cbl/physiology , Two-Hybrid System Techniques , Ubiquitination
5.
J Biol Chem ; 285(31): 23687-98, 2010 Jul 30.
Article in English | MEDLINE | ID: mdl-20525694

ABSTRACT

Cbl proteins are ubiquitin ligases (E3s) that play a significant role in regulating tyrosine kinase signaling. There are three mammalian family members: Cbl, Cbl-b, and Cbl-c. All have a highly conserved N-terminal tyrosine kinase binding domain, a catalytic RING finger domain, and a C-terminal proline-rich domain that mediates interactions with Src homology 3 (SH3) containing proteins. Although both Cbl and Cbl-b have been studied widely, little is known about Cbl-c. Published reports have demonstrated that the N terminus of Cbl and Cbl-b have an inhibitory effect on their respective E3 activity. However, the mechanism for this inhibition is still unknown. In this study we demonstrate that the N terminus of Cbl-c, like that of Cbl and Cbl-b, inhibits the E3 activity of Cbl-c. Furthermore, we map the region responsible for the inhibition to the EF-hand and SH2 domains. Phosphorylation of a critical tyrosine (Tyr-341) in the linker region of Cbl-c by Src or a phosphomimetic mutation of this tyrosine (Y341E) is sufficient to increase the E3 activity of Cbl-c. We also demonstrate for the first time that phosphorylation of Tyr-341 or the Y341E mutation leads to a decrease in affinity for the ubiquitin-conjugating enzyme (E2), UbcH5b. The decreased affinity of the Y341E mutant Cbl-c for UbcH5b results in a more rapid turnover of bound UbcH5b coincident with the increased E3 activity. These data suggest that the N terminus of Cbl-c contributes to the binding to the E2 and that phosphorylation of Tyr-341 leads to a decrease in affinity and an increase in the E3 activity of Cbl-c.


Subject(s)
Proto-Oncogene Proteins c-cbl/chemistry , Ubiquitin-Protein Ligases/chemistry , Ubiquitin/chemistry , Glutathione Transferase/metabolism , Humans , Kinetics , Mutagenesis, Site-Directed , Mutation , Phosphorylation , Protein Binding , Proto-Oncogene Proteins c-cbl/metabolism , Recombinant Proteins/chemistry , Signal Transduction , src Homology Domains
6.
Cancer Res ; 70(12): 4789-94, 2010 Jun 15.
Article in English | MEDLINE | ID: mdl-20501843

ABSTRACT

Cbl was originally discovered in 1989 as the cellular homolog of the v-Cbl oncogene, the transforming gene of the Cas NS-1 murine retrovirus that causes myeloid leukemia and lymphomas in mice. Cbl is a member of a family of RING finger ubiquitin ligases that negatively regulate signaling by tyrosine kinases and that function as adaptor proteins to regulate signaling positively. Until the past 2 years, there was little evidence that Cbl proteins were involved in human malignancies. Recent publications have shown homozygous mutations in Cbl in human myeloid neoplasms. Although in vitro and animal transformation models suggested that mutant forms of Cbl acted as an oncogene, the cellular role suggested that the protein could serve as a tumor suppressor gene. The recent data begin to reconcile this paradox as the loss of ubiquitin ligase function (the tumor suppressor function) is coupled to the maintenance of the positive signaling function (the oncogene function). These data also provide insight into potential therapeutic approaches to myeloid disorders harboring Cbl mutations.


Subject(s)
Leukemia, Myeloid/metabolism , Oncogenes/physiology , Proto-Oncogene Proteins c-cbl/physiology , Humans
7.
Breast Cancer Res Treat ; 113(2): 217-30, 2009 Jan.
Article in English | MEDLINE | ID: mdl-18266105

ABSTRACT

Tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) induces apoptosis in some but not all breast cancer cell lines. Breast cancers can be divided into those which express the estrogen (ER) and progesterone (PR) receptors, those with HER-2 amplification, and those without expression of ER, PR, or HER-2 amplification (referred to as basal or triple-negative breast cancer). We tested a panel of 20 breast cancer cell lines representing the different types of breast cancer to evaluate if the molecular phenotype of the breast cancer cells determined their response to TRAIL. The most striking finding was that eight of eleven triple-negative cell lines are sensitive to TRAIL-mediated apoptosis. The eight TRAIL-sensitive triple-negative cell lines have a mesenchymal phenotype while the three TRAIL-resistant triple-negative cell lines have an epithelial phenotype. Two of five cell lines with HER-2 amplification were sensitive to TRAIL and none of the five ER positive cell lines were sensitive. RNAi-mediated knockdown of TRAIL receptor expression demonstrated that TRAIL Receptor 2 (TRAIL-R2) mediates the effects of TRAIL, even when both TRAIL-R1 and TRAIL-R2 are expressed. Finally, inhibition of EGFR, expressed in both TRAIL-sensitive and TRAIL-resistant triple-negative breast cancer cell lines, using a small molecule tyrosine kinase inhibitor (AG1478), enhanced TRAIL-induced apoptosis in TRAIL-sensitive cell lines but did not convert resistant cells into TRAIL-sensitive cells. Together, these findings suggest that a subset of triple-negative breast cancer, those with mesenchymal features, may be the most likely to benefit from TRAIL targeted therapy. These findings could form the basis to select breast cancer patients for clinical trials of TRAIL-R2 ligands.


Subject(s)
Apoptosis/drug effects , Breast Neoplasms/pathology , Recombinant Fusion Proteins/pharmacology , Breast Neoplasms/chemistry , Breast Neoplasms/classification , Cell Line, Tumor/chemistry , Cell Line, Tumor/drug effects , Cell Line, Tumor/pathology , Drug Delivery Systems , Epithelium , ErbB Receptors/antagonists & inhibitors , ErbB Receptors/physiology , Female , Genes, erbB-2 , Humans , Mesoderm , Neoplasm Proteins/analysis , Phenotype , Protein Kinase Inhibitors/pharmacology , Quinazolines , RNA Interference , Receptors, Estrogen/analysis , Receptors, Progesterone/analysis , Receptors, TNF-Related Apoptosis-Inducing Ligand/drug effects , Tyrphostins/pharmacology
8.
Trends Biochem Sci ; 31(2): 79-88, 2006 Feb.
Article in English | MEDLINE | ID: mdl-16406635

ABSTRACT

Cbl proteins are regulators of signal transduction through many pathways and, consequently, regulate cell function and development. They are ubiquitin ligases that ubiquitinate and target many signaling molecules for degradation. The Cbl proteins themselves are regulated by an increasingly complex network of interactions that fine-tune the effects that Cbl proteins have on signaling. The negative regulation of Cbl protein function can occur via cis-acting structural elements that prevent inappropriate ubiquitin ligase activity, degradation of the Cbl proteins, inhibition without degradation owing to interaction with other signaling proteins, deubiquitination of Cbl substrates, and regulation of assembly of the endosomal ESCRT-I complex. Defects in the regulatory mechanisms that control Cbl function are implicated in the development of immunological and malignant diseases.


Subject(s)
Gene Expression Regulation, Enzymologic , Proto-Oncogene Proteins c-cbl/genetics , Signal Transduction/drug effects , Animals , Autoimmune Diseases/physiopathology , CD28 Antigens/metabolism , Calcium-Binding Proteins/physiology , Carrier Proteins/metabolism , Carrier Proteins/physiology , Cell Cycle Proteins/physiology , Cortactin/physiology , Endosomal Sorting Complexes Required for Transport , Humans , Membrane Proteins , Neoplasms/physiopathology , Nerve Tissue Proteins/physiology , Oncogene Proteins, Viral/physiology , Protein Tyrosine Phosphatases , Proto-Oncogene Proteins c-cbl/metabolism , Proto-Oncogene Proteins c-kit/metabolism , Signal Transduction/physiology , Ubiquitin-Protein Ligases/metabolism , cdc42 GTP-Binding Protein/physiology , src-Family Kinases/metabolism
9.
Oncogene ; 23(42): 7104-15, 2004 Sep 16.
Article in English | MEDLINE | ID: mdl-15273720

ABSTRACT

Cbl proteins are ubiquitin protein ligases, which ubiquitinate activated tyrosine kinases and target them for degradation. Both c-Cbl and Cbl-b have an ubiquitin associated (UBA) domain at their C-terminal end. We observed that high molecular weight ubiquitinated proteins constitutively coimmunoprecipitated with transfected and endogenous Cbl-b, but not c-Cbl. The binding site for these ubiquitinated proteins was mapped to the UBA domain of Cbl-b (UBAb). GST-fusion proteins containing the UBAb interacted with ubiquitinated proteins and polyubiquitin chains in vitro, whereas those containing the UBA domain of c-Cbl (UBAc) did not. The UBAb had a much greater affinity for polyubiquitin chains than for monoubiquitin. Analysis of the UBAb and UBAc demonstrate that the affinity for ubiquitin is determined by multiple amino-acid differences between the two domains. Overexpression of the UBAb, but not overexpression of the UBAc, inhibited a variety of ubiquitin-mediated processes such as degradation of ubiquitinated proteins (i.e. EGFR, Mdm-2, and Siah-1). This in vivo result is consistent with the differences in ubiquitin binding observed in vitro between the UBAb and UBAc. This difference in ubiquitin-binding may reflect distinct regulatory functions of c-Cbl and Cbl-b.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Proto-Oncogene Proteins/metabolism , Ubiquitin-Protein Ligases/metabolism , Ubiquitin/metabolism , Adaptor Proteins, Signal Transducing/genetics , Amino Acid Sequence , Base Sequence , Binding Sites , Cell Line , HeLa Cells , Humans , Kidney , Molecular Sequence Data , Mutagenesis, Site-Directed , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins c-cbl , Recombinant Proteins/metabolism , Transfection , Ubiquitin-Protein Ligases/genetics
10.
Mol Cell Biol ; 23(21): 7708-18, 2003 Nov.
Article in English | MEDLINE | ID: mdl-14560016

ABSTRACT

Cbl family proteins are evolutionarily conserved ubiquitin ligases that negatively regulate signaling from tyrosine kinase-coupled receptors. The mammalian cbl family consists of c-Cbl, Cbl-b, and the recently cloned Cbl-3 (also known as Cbl-c). In this study, we describe the detailed expression pattern of murine Cbl-3 and report the generation and characterization of Cbl-3-deficient mice. Cbl-3 exhibits an expression pattern distinct from those of c-Cbl and Cbl-b, with high levels of Cbl-3 expression in epithelial cells of the gastrointestinal tract and epidermis, as well as the respiratory, urinary, and reproductive systems. Cbl-3 expression was not detected in nonepithelial cells, but within epithelial tissues, the levels of Cbl-3 expression varied from undetectable in the alveoli of the lungs to very strong in the cecum and colon. Despite this restricted expression pattern, Cbl-3-deficient mice were viable, healthy, and fertile and displayed no histological abnormalities up to 18 months of age. Proliferation of epithelial cells in the epidermises and gastrointestinal tracts was unaffected by the loss of Cbl-3. Moreover, Cbl-3 was not required for attenuation of epidermal growth factor-stimulated Erk activation in primary keratinocytes. Thus, Cbl-3 is dispensable for normal epithelial development and function.


Subject(s)
Epithelial Cells/metabolism , Epithelium/growth & development , Retroviridae Proteins, Oncogenic/metabolism , Animals , Cells, Cultured , Enzyme Activation , Epithelial Cells/cytology , Epithelium/anatomy & histology , Gene Expression Regulation , Gene Targeting , Humans , In Situ Hybridization , Keratinocytes/cytology , Keratinocytes/metabolism , Mice , Mice, Knockout , Mitogen-Activated Protein Kinases/metabolism , Proto-Oncogene Proteins c-cbl , Retroviridae Proteins, Oncogenic/genetics , Tissue Distribution
11.
Gene ; 308: 103-13, 2003 Apr 10.
Article in English | MEDLINE | ID: mdl-12711395

ABSTRACT

The genomic organization of cbl genes from a variety of mammalian and non-mammalian species was determined by a combination of cloning and database searches. Humans and mice have three cbl genes (c-cbl,(1) cblb, and cblc) which show remarkable conservation of the intron/exon structure over the region of the genes which encode the highly conserved N-terminal region of the proteins including the RING finger. Searches of genomic, cDNA, and EST databases revealed that one or more cbl genes exist in chordates, insects, and worms. Comparison of the complexity and genomic organization of the cbl gene family and the predicted Cbl proteins from various species suggests that the three mammalian cbl genes arose by two duplications of an ancestral gene. The genomic organization of the cbl genes from various species provides insight into the evolution of the cbl gene family.


Subject(s)
Genes/genetics , Proto-Oncogene Proteins/genetics , Ubiquitin-Protein Ligases , Amino Acid Sequence , Animals , Base Sequence , Databases, Nucleic Acid , Evolution, Molecular , Exons , Humans , Introns , Mice , Multigene Family/genetics , Phylogeny , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins c-cbl , Sequence Homology, Amino Acid
SELECTION OF CITATIONS
SEARCH DETAIL