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.
PLoS One ; 14(5): e0216967, 2019.
Article in English | MEDLINE | ID: mdl-31120930

ABSTRACT

Many receptor tyrosine kinases (RTKs, such as EGFR, MET) are negatively regulated by ubiquitination and degradation mediated by Cbl proteins, a family of RING finger (RF) ubiquitin ligases (E3s). Loss of Cbl protein function is associated with malignant transformation driven by increased RTK activity. RF E3s, such as the Cbl proteins, interact with a ubiquitin-conjugating enzyme (E2) to confer specificity to the ubiquitination process and direct the transfer of ubiquitin from the E2 to one or more lysines on the target proteins. Using in vitro E3 assays and yeast two-hybrid screens, we found that Ube2d, Ube2e families, Ube2n/2v1, and Ube2w catalyze autoubiquitination of the Cbl protein and Ube2d2, Ube2e1, and Ube 2n/2v1 catalyze Cbl-mediated substrate ubiquitination of the EGFR and SYK. Phosphorylation of the Cbl protein by by Src resulted in increased E3 activity compared to unphosphorylated cbl or Cbl containing a phosphomimetic Y371E mutation. Ubiquitin chain formation depended on the E2 tested with Cbl with Ube2d2 forming both K48 and K63 linked chains, Ube2n/2v1 forming only K63 linked chains, and Ube2w inducing monoubiquitination. In cells, the Ube2d family, Ube2e family, and Ube2n/2v1 contributed to EGFR ubiquitination. Our data suggest that multiple E2s can interact with Cbl and modulate its E3 activity in vitro and in cells.


Subject(s)
Proto-Oncogene Proteins c-cbl/metabolism , Ubiquitin-Conjugating Enzymes/metabolism , ErbB Receptors/metabolism , Gene Expression Regulation , Gene Silencing , HEK293 Cells , HeLa Cells , Humans , Mutation , Phosphorylation , Protein Binding , Proto-Oncogene Proteins c-cbl/genetics , Two-Hybrid System Techniques , Ubiquitin/metabolism , Ubiquitination
3.
Biochem Mol Biol Educ ; 45(1): 13-24, 2017 Jan 02.
Article in English | MEDLINE | ID: mdl-27231204

ABSTRACT

Translational science is an emerging field that holds great promise to accelerate the development of novel medical interventions. As the field grows, so does the demand for highly trained biomedical scientists to fill the positions that are being created. Many graduate and postdoctorate training programs do not provide their trainees with sufficient education to take advantage of this growing employment sector. To help better prepare the trainees at the National Institutes of Health for possible careers in translation, we have created the Translational Science Training Program (TSTP). The TSTP is an intensive 2- to 3-day training program that introduces NIH postdoctoral trainees and graduate students to the science and operation of turning basic research discoveries into a medical therapeutic, device or diagnostic, and also exposes them to the variety of career options in translational science. Through a combination of classroom teaching from practicing experts in the various disciplines of translation and small group interactions with pre-clinical development teams, participants in the TSTP gain knowledge that will aid them in obtaining a career in translational science and building a network to make the transition to the field. © 2016 by The International Union of Biochemistry and Molecular Biology, 45(1):13-24, 2017.


Subject(s)
Biomedical Research/education , Career Choice , Education, Graduate/organization & administration , Research Personnel/education , Translational Research, Biomedical/education , Translational Research, Biomedical/organization & administration , Humans , National Institutes of Health (U.S.) , United States
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
6.
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
7.
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
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.
Prostate ; 65(4): 375-89, 2005 Dec 01.
Article in English | MEDLINE | ID: mdl-16037992

ABSTRACT

BACKGROUND: Tumor suppressor p53 mutations are associated with the transition of prostate cancer to metastatic, hormone-refractory disease and stable expression of p53 gain-of-function (p53GOF) alleles support growth of LNCaP in androgen-depleted medium. In this study, we performed gene expression profiling of four LNCaP-p53GOF sublines to test the hypothesis that different p53GOF mutants mediated androgen independence via modulation of a common set of genes. METHODS: Expression profiling was performed using Affymetrix HG-U95Av2 arrays followed by hierarchical clustering to identify expression patterns associated with particular molecular alterations. p53GOF-mediated regulation of Id-1 expression was validated by RT-PCR and dual-luciferase reporter assays. RNA interference was used to investigate the effects of Id-1 and Id-3 suppression. RESULTS: LNCaP-p53GOF sublines possessed a molecular signature consisting of 95 differentially regulated genes that could be segregated into two clusters of transcripts induced (n=50) and repressed (n=45) by p53GOF expression. To begin validating these genes as effectors of the p53 mutants, we evaluated one of the overexpressed genes, Id-1. RT-PCR confirmed the microarray results and revealed elevated Id-1 levels in LNCaP-p53-P151S (loss-of-function only mutant), thereby implicating p53 mutational inactivation, but not gain-of-function, as a basis for Id-1 deregulation. Reporter assays demonstrated enhanced Id-1 promoter activity in an LNCaP-p53GOF subline. The contribution of Id-1 to p53GOF-mediated biology was demonstrated by the ability of RNAi-mediated gene silencing to decrease both basal and androgen-independent proliferation. CONCLUSIONS: While different p53GOF mutants result in overall distinct expression profiles, they share a common set of differentially-expressed genes that can be used to signify their presence and provide insight into mechanisms underlying androgen independence.


Subject(s)
Alleles , Genes, p53/genetics , Mutation , Neoplasms, Hormone-Dependent/genetics , Prostatic Neoplasms/genetics , Cell Line, Tumor , Gene Expression Profiling , Humans , Inhibitor of Differentiation Protein 1/biosynthesis , Inhibitor of Differentiation Protein 1/genetics , Male , Neoplasms, Hormone-Dependent/metabolism , Oligonucleotide Array Sequence Analysis , Promoter Regions, Genetic , Prostatic Neoplasms/metabolism , RNA Interference , RNA, Neoplasm/chemistry , RNA, Neoplasm/genetics , RNA, Small Interfering/genetics , Reverse Transcriptase Polymerase Chain Reaction
10.
J Immunol ; 174(7): 4010-8, 2005 Apr 01.
Article in English | MEDLINE | ID: mdl-15778358

ABSTRACT

The commitment of naive T cells to proliferate is a function of the strength and duration of stimuli mediated by the TCR and coreceptors. Ranges of 2-20 h of stimulation have been reported as necessary in vitro. Whether T cells actually experience uninterrupted stimulation for such long periods under physiological conditions is controversial. Here we ask whether commitment to proliferate requires continuous stimulation, or can T cells integrate intermittent periods of stimulation. T cells were stimulated for two short-term (subthreshold) periods (5-7 h) either sequentially or separated by an interval of rest. Naive lymph node T cells were able to integrate interrupted stimulation, even when the duration of rest was as long as 2 days. Furthermore, when short-term-stimulated T cells were separated by density, three populations were observed: low density blasts, intermediate density G(1) cells, and high density G(0) cells. Low density cells progressed to division without further stimulation, whereas G(0) and G(1) cells remained undivided. However, after a period of rest, a second subthreshold stimulation caused the G(1) but not the G(0) fraction to quickly proceed through the cell cycle. We conclude that noncycling T cells in the G(1) phase of the cell cycle remain in a state of readiness for prolonged periods of time, and may represent a population of memory-like effectors capable of responding rapidly to antigenic challenge.


Subject(s)
G1 Phase/immunology , Immunologic Memory , T-Lymphocytes/immunology , Animals , Biomarkers/analysis , Cell Cycle/immunology , Cell Proliferation , Interleukin-2/biosynthesis , Lymphocyte Activation/immunology , Mice , Mice, Inbred C57BL , Resting Phase, Cell Cycle/immunology , T-Lymphocytes/cytology
11.
Cancer Res ; 62(22): 6606-14, 2002 Nov 15.
Article in English | MEDLINE | ID: mdl-12438256

ABSTRACT

CWR22 has been a valuable xenograft model for the study of prostate cancer progression from an androgen-dependent tumor to one that grows in castrated animals. Herein, we report the identification and characterization of a novel androgen receptor (AR) mutation occurring in a relapsed tumor (CWR22R-2152) and in the CWR22Rv1 cell line established from it. The mutation was not detected in the original, hormone-dependent CWR22 xenograft, indicating that this change occurred during the progression to androgen independence. It is characterized by an in-frame tandem duplication of exon 3 that encodes the second zinc finger of the AR DNA-binding domain. Accordingly, immunoblot analyses demonstrated the expression of an AR species having an approximately 5-kDa increase in size relative to the LNCaP AR. This was accompanied by a COOH-terminally truncated AR species migrating with a relative mass of 75-80 kDa, referred to as ARDeltaLBD because it lacks the ligand-binding domain. By recreating the exon 3 duplication mutation in a wild-type AR expression construct, the generation of ARDeltaLBD could be recapitulated. Whereas ARDeltaLBD exhibited constitutive nuclear localization and DNA binding, these functions in the full-length AR remained androgen dependent. The CWR22Rv1 AR repertoire displayed dose-dependent, androgen-responsive transcriptional transactivation in reporter assays, albeit to a lesser extent in comparison with LNCaP. This cell line also expressed low levels of prostate-specific antigen mRNA and did not express or secrete detectable levels of prostate-specific antigen protein in androgen-depleted medium or in response to physiological androgenic stimulation. In summary, the CWR22Rv1 cell line displays both androgen-responsive and androgen-insensitive features due, at least in part, to a novel insertional mutation of the AR.


Subject(s)
Gene Duplication , Prostatic Neoplasms/genetics , Receptors, Androgen/genetics , Androgens/deficiency , Androgens/physiology , Animals , Base Sequence , Exons , Humans , Male , Mice , Molecular Sequence Data , Neoplasm Transplantation , Neoplasms, Hormone-Dependent/genetics , Prostatic Neoplasms/metabolism , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Receptors, Androgen/biosynthesis , Testosterone/pharmacology , Transfection , Transplantation, Heterologous , Tumor Cells, Cultured
SELECTION OF CITATIONS
SEARCH DETAIL