Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 60
1.
Elife ; 112022 02 28.
Article En | MEDLINE | ID: mdl-35225231

The Fbw7 ubiquitin ligase targets many proteins for proteasomal degradation, which include oncogenic transcription factors (TFs) (e.g., c-Myc, c-Jun, and Notch). Fbw7 is a tumor suppressor and tumors often contain mutations in FBXW7, the gene that encodes Fbw7. The complexity of its substrate network has obscured the mechanisms of Fbw7-associated tumorigenesis, yet this understanding is needed for developing therapies. We used an integrated approach employing RNA-Seq and high-resolution mapping (cleavage under target and release using nuclease) of histone modifications and TF occupancy (c-Jun and c-Myc) to examine the combinatorial effects of misregulated Fbw7 substrates in colorectal cancer (CRC) cells with engineered tumor-associated FBXW7 null or missense mutations. Both Fbw7 mutations caused widespread transcriptional changes associated with active chromatin and altered TF occupancy: some were common to both Fbw7 mutant cell lines, whereas others were mutation specific. We identified loci where both Jun and Myc were coregulated by Fbw7, suggesting that substrates may have synergistic effects. One coregulated gene was CIITA, the master regulator of MHC Class II gene expression. Fbw7 loss increased MHC Class II expression and Fbw7 mutations were correlated with increased CIITA expression in TCGA colorectal tumors and cell lines, which may have immunotherapeutic implications for Fbw7-associated cancers. Analogous studies in neural stem cells in which FBXW7 had been acutely deleted closely mirrored the results in CRC cells. Gene set enrichment analyses revealed Fbw7-associated pathways that were conserved across both cell types that may reflect fundamental Fbw7 functions. These analyses provide a framework for understanding normal and neoplastic context-specific Fbw7 functions.


Colorectal Neoplasms , F-Box Proteins , F-Box-WD Repeat-Containing Protein 7/genetics , Cell Cycle Proteins/metabolism , Colorectal Neoplasms/pathology , F-Box Proteins/genetics , F-Box Proteins/metabolism , Humans , Mutation , Ubiquitin-Protein Ligases/genetics , Ubiquitin-Protein Ligases/metabolism
2.
Sci Adv ; 8(4): eabl7872, 2022 Jan 28.
Article En | MEDLINE | ID: mdl-35089787

c-Myc (hereafter, Myc) is a cancer driver whose abundance is regulated by the SCFFbw7 ubiquitin ligase and proteasomal degradation. Fbw7 binds to a phosphorylated Myc degron centered at threonine 58 (T58), and mutations of Fbw7 or T58 impair Myc degradation in cancers. Here, we identify a second Fbw7 phosphodegron at Myc T244 that is required for Myc ubiquitylation and acts in concert with T58 to engage Fbw7. While Ras-dependent Myc serine 62 phosphorylation (pS62) is thought to stabilize Myc by preventing Fbw7 binding, we find instead that pS62 greatly enhances Fbw7 binding and is an integral part of a high-affinity degron. Crystallographic studies revealed that both degrons bind Fbw7 in their diphosphorylated forms and that the T244 degron is recognized via a unique mode involving Fbw7 arginine 689 (R689), a mutational hotspot in cancers. These insights have important implications for Myc-associated tumorigenesis and therapeutic strategies targeting Myc stability.

3.
Nat Commun ; 12(1): 2043, 2021 04 06.
Article En | MEDLINE | ID: mdl-33824312

The tumour suppressor FBW7 is a substrate adaptor for the E3 ubiquitin ligase complex SKP1-CUL1-F-box (SCF), that targets several oncoproteins for proteasomal degradation. FBW7 is widely mutated and FBW7 protein levels are commonly downregulated in cancer. Here, using an shRNA library screen, we identify the HECT-domain E3 ubiquitin ligase TRIP12 as a negative regulator of FBW7 stability. We find that SCFFBW7-mediated ubiquitylation of FBW7 occurs preferentially on K404 and K412, but is not sufficient for its proteasomal degradation, and in addition requires TRIP12-mediated branched K11-linked ubiquitylation. TRIP12 inactivation causes FBW7 protein accumulation and increased proteasomal degradation of the SCFFBW7 substrate Myeloid Leukemia 1 (MCL1), and sensitizes cancer cells to anti-tubulin chemotherapy. Concomitant FBW7 inactivation rescues the effects of TRIP12 deficiency, confirming FBW7 as an essential mediator of TRIP12 function. This work reveals an unexpected complexity of FBW7 ubiquitylation, and highlights branched ubiquitylation as an important signalling mechanism regulating protein stability.


Carrier Proteins/metabolism , F-Box-WD Repeat-Containing Protein 7/metabolism , Proteasome Endopeptidase Complex/metabolism , Proteolysis , Ubiquitin-Protein Ligases/metabolism , Ubiquitination , Biocatalysis , Drug Resistance, Neoplasm , HCT116 Cells , HEK293 Cells , Humans , Lysine/metabolism , Myeloid Cell Leukemia Sequence 1 Protein/metabolism , Protein Binding , Protein Processing, Post-Translational , Protein Stability , RNA, Small Interfering/metabolism , Substrate Specificity , Ubiquitin-Conjugating Enzymes/metabolism
4.
Proc Natl Acad Sci U S A ; 117(45): 28287-28296, 2020 11 10.
Article En | MEDLINE | ID: mdl-33093209

Head and neck squamous cell carcinoma (HNSCC) associated with high-risk human papilloma virus (HPV) infection is a growing clinical problem. The WEE1 kinase inhibitor AZD1775 (WEE1i) overrides cell cycle checkpoints and is being studied in HNSCC regimens. We show that the HPV16 E6/E7 oncoproteins sensitize HNSCC cells to single-agent WEE1i treatment through activation of a FOXM1-CDK1 circuit that drives mitotic gene expression and DNA damage. An isogenic cell system indicated that E6 largely accounts for these phenotypes in ways that extend beyond p53 inactivation. A targeted genomic analysis implicated FOXM1 signaling downstream of E6/E7 expression and analyses of primary tumors and The Cancer Genome Atlas (TCGA) data revealed an activated FOXM1-directed promitotic transcriptional signature in HPV+ versus HPV- HNSCCs. Finally, we demonstrate the causality of FOXM1 in driving WEE1i sensitivity. These data suggest that elevated basal FOXM1 activity predisposes HPV+ HNSCC to WEE1i-induced toxicity and provide mechanistic insights into WEE1i and HPV+ HNSCC therapies.


Cell Cycle Proteins/drug effects , Forkhead Box Protein M1/metabolism , Papillomavirus Infections/drug therapy , Protein-Tyrosine Kinases/drug effects , Pyrazoles/antagonists & inhibitors , Pyrimidinones/antagonists & inhibitors , Squamous Cell Carcinoma of Head and Neck/drug therapy , CDC2 Protein Kinase/metabolism , Cell Cycle Checkpoints , Cell Cycle Proteins/metabolism , Cell Line, Tumor , DNA Damage/drug effects , Head and Neck Neoplasms , Humans , Oncogene Proteins, Viral/metabolism , Papillomavirus E7 Proteins/metabolism , Protein-Tyrosine Kinases/metabolism , Repressor Proteins/metabolism , Up-Regulation
5.
Sci Adv ; 6(16): eaaz9899, 2020 04.
Article En | MEDLINE | ID: mdl-32494624

Cyclin-dependent kinase 2 (CDK2) controls cell division and is central to oncogenic signaling. We used an "in situ" approach to identify CDK2 substrates within nuclei isolated from cells expressing CDK2 engineered to use adenosine 5'-triphosphate analogs. We identified 117 candidate substrates, ~40% of which are known CDK substrates. Previously unknown candidates were validated to be CDK2 substrates, including LSD1, DOT1L, and Rad54. The identification of many chromatin-associated proteins may have been facilitated by labeling conditions that preserved nuclear architecture and physiologic CDK2 regulation by endogenous cyclins. Candidate substrates include proteins that regulate histone modifications, chromatin, transcription, and RNA/DNA metabolism. Many of these proteins also coexist in multi-protein complexes, including epigenetic regulators, that may provide new links between cell division and other cellular processes mediated by CDK2. In situ phosphorylation thus revealed candidate substrates with a high validation rate and should be readily applicable to other nuclear kinases.

6.
Sci Rep ; 9(1): 5488, 2019 04 02.
Article En | MEDLINE | ID: mdl-30940846

Insertional mutagenesis is a powerful means of identifying cancer drivers in animal models. We used the Sleeping Beauty (SB) transposon/transposase system to identify activated oncogenes in hematologic cancers in wild-type mice and mice that express a stabilized cyclin E protein (termed cyclin ET74AT393A). Cyclin E governs cell division and is misregulated in human cancers. Cyclin ET74AT393A mice develop ineffective erythropoiesis that resembles early-stage human myelodysplastic syndrome, and we sought to identify oncogenes that might cooperate with cyclin E hyperactivity in leukemogenesis. SB activation in hematopoietic precursors caused T-cell leukemia/lymphomas (T-ALL) and pure red blood cell erythroleukemias (EL). Analysis of >12,000 SB integration sites revealed markedly different oncogene activations in EL and T-ALL: Notch1 and Ikaros were most common in T-ALL, whereas ETS transcription factors (Erg and Ets1) were targeted in most ELs. Cyclin E status did not impact leukemogenesis or oncogene activations. Whereas most SB insertions were lost during culture of EL cell lines, Erg insertions were retained, indicating Erg's key role in these neoplasms. Surprisingly, cyclin ET74AT393A conferred growth factor independence and altered Erg-dependent differentiation in EL cell lines. These studies provide new molecular insights into erythroid leukemia and suggest potential therapeutic targets for human leukemia.


Cyclin E/genetics , Leukemia, Erythroblastic, Acute/genetics , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/genetics , Transposases/genetics , Animals , Cell Culture Techniques , DNA Transposable Elements , Disease Models, Animal , Genetic Predisposition to Disease , Mice , Mutagenesis, Insertional , Oncogene Proteins/genetics , Transcriptional Regulator ERG/genetics
7.
PLoS Pathog ; 15(1): e1007543, 2019 01.
Article En | MEDLINE | ID: mdl-30689667

Merkel cell polyomavirus (MCPyV) accounts for 80% of all Merkel cell carcinoma (MCC) cases through expression of two viral oncoproteins: the truncated large T antigen (LT-t) and small T antigen (ST). MCPyV ST is thought to be the main driver of cellular transformation and has also been shown to increase LT protein levels through the activity of its Large-T Stabilization Domain (LSD). The ST LSD was reported to bind and sequester several ubiquitin ligases, including Fbw7 and ß-TrCP, and thereby stabilize LT-t and several other Fbw7 targets including c-Myc and cyclin E. Therefore, the ST LSD is thought to contribute to transformation by promoting the accumulation of these oncoproteins. Targets of Fbw7 and ß-TrCP contain well-defined, conserved, phospho-degrons. However, as neither MCPyV LT, LT-t nor ST contain the canonical Fbw7 phospho-degron, we sought to further investigate the proposed model of ST stabilization of LT-t and transformation. In this study, we provide several lines of evidence that fail to support a specific interaction between MCPyV T antigens and Fbw7 or ß-TrCP by co-immunoprecipitation or functional consequence. Although MCPyV ST does indeed increase LT protein levels through its Large-T Stabilization domain (LSD), this is accomplished independently of Fbw7. Therefore, our study indicates a need for further investigation into the role and mechanism(s) of MCPyV T antigens in viral replication, latency, transformation, and tumorigenesis.


Antigens, Polyomavirus Transforming/metabolism , F-Box-WD Repeat-Containing Protein 7/metabolism , Merkel cell polyomavirus/metabolism , Antigens, Neoplasm/metabolism , Antigens, Viral, Tumor/metabolism , Carcinoma, Merkel Cell/metabolism , HEK293 Cells , Humans , Ligases/metabolism , Merkel Cells , Merkel cell polyomavirus/immunology , Merkel cell polyomavirus/pathogenicity , Oncogene Proteins/metabolism , Polyomavirus Infections/metabolism , Protein Domains , Tumor Virus Infections/virology , Ubiquitin/metabolism , Virus Replication , beta-Transducin Repeat-Containing Proteins/metabolism
8.
Proc Natl Acad Sci U S A ; 115(21): 5462-5467, 2018 05 22.
Article En | MEDLINE | ID: mdl-29735700

The Fbw7 (F-box/WD repeat-containing protein 7) ubiquitin ligase targets multiple oncoproteins for degradation and is commonly mutated in cancers. Like other pleiotropic tumor suppressors, Fbw7's complex biology has impeded our understanding of how Fbw7 mutations promote tumorigenesis and hindered the development of targeted therapies. To address these needs, we employed a transfer learning approach to derive gene-expression signatures from The Cancer Gene Atlas datasets that predict Fbw7 mutational status across tumor types and identified the pathways enriched within these signatures. Genes involved in mitochondrial function were highly enriched in pan-cancer signatures that predict Fbw7 mutations. Studies in isogenic colorectal cancer cell lines that differed in Fbw7 mutational status confirmed that Fbw7 mutations increase mitochondrial gene expression. Surprisingly, Fbw7 mutations shifted cellular metabolism toward oxidative phosphorylation and caused context-specific metabolic vulnerabilities. Our approach revealed unexpected metabolic reprogramming and possible therapeutic targets in Fbw7-mutant cancers and provides a framework to study other complex, oncogenic mutations.


Colorectal Neoplasms/metabolism , Colorectal Neoplasms/pathology , F-Box-WD Repeat-Containing Protein 7/genetics , F-Box-WD Repeat-Containing Protein 7/metabolism , Metabolome , Mitochondria/metabolism , Mutation , Cell Respiration , Colorectal Neoplasms/genetics , Gene Expression Profiling , Humans , Mitochondria/pathology , Oxidative Phosphorylation , Oxidative Stress , Phosphorylation , Ubiquitin , Ubiquitination
9.
Nat Commun ; 9(1): 34, 2018 01 02.
Article En | MEDLINE | ID: mdl-29295984

In eukaryotes, RAD54 catalyzes branch migration (BM) of Holliday junctions, a basic process during DNA repair, replication, and recombination. RAD54 also stimulates RAD51 recombinase and has other activities. Here, we investigate the structural determinants for different RAD54 activities. We find that the RAD54 N-terminal domain (NTD) is responsible for initiation of BM through two coupled, but distinct steps; specific binding to Holliday junctions and RAD54 oligomerization. Furthermore, we find that the RAD54 oligomeric state can be controlled by NTD phosphorylation at S49, a CDK2 consensus site, which inhibits RAD54 oligomerization and, consequently, BM. Importantly, the effect of phosphorylation on RAD54 oligomerization is specific for BM, as it does not affect stimulation of RAD51 recombinase by RAD54. Thus, the transition of the oligomeric states provides an important control of the biological functions of RAD54 and, likely, other multifunctional proteins.


Adenosine Triphosphatases/metabolism , DNA Helicases/metabolism , DNA, Cruciform/metabolism , Nuclear Proteins/metabolism , Amino Acid Sequence , Animals , Binding Sites/genetics , Cell Line , DNA Helicases/chemistry , DNA Helicases/genetics , DNA Repair , DNA, Cruciform/chemistry , DNA, Cruciform/genetics , DNA-Binding Proteins , Humans , Hydrolysis , Nuclear Proteins/chemistry , Nuclear Proteins/genetics , Nucleic Acid Conformation , Phosphorylation , Protein Multimerization , Recombination, Genetic , Sequence Homology, Amino Acid , Sf9 Cells , Spodoptera
10.
Oncotarget ; 8(31): 50680-50691, 2017 Aug 01.
Article En | MEDLINE | ID: mdl-28881594

The miR-106a~363 cluster encodes 6 miRNAs on the X-chromosome which are abundant in blood cells and overexpressed in a variety of malignancies. The constituent miRNA of miR-106a~363 have functional activities in vitro that are predicted to be both oncogenic and tumor suppressive, yet little is known about their physiological functions in vivo. Mature miR-106a~363 (Mirc2) miRNAs are processed from an intragenic, non-protein encoding gene referred to as Xpcl1 (or Kis2), situated at an X-chromosomal locus frequently targeted by retroviruses in murine lymphomas. The oncogenic potential of miR-106a~363 Xpcl1 has not been proven, nor its potential role in T cell development. We show that miR106a~363 levels normally drop at the CD4+/CD8+ double positive (DP) stage of thymocyte development. Forced expression of Xpcl1 at this stage impairs thymocyte maturation and induces T-cell lymphomas. Surprisingly, miR-106a~363 Xpcl1 also induces p27 transcription via Foxo3/4 transcription factors. As a haploinsufficient tumor suppressor, elevated p27 is expected to inhibit lymphomagenesis. Consistent with this, concurrent p27 Kip1 deletion dramatically accelerated lymphomagenesis, indicating that p27 is rate limiting for tumor development by Xpcl1. Whereas down-regulation of miR-106a~363 is important for normal T cell differentiation and for the prevention of lymphomas, eliminating p27 reveals Xpcl1's full oncogenic potential.

11.
Cell Rep ; 19(1): 162-174, 2017 04 04.
Article En | MEDLINE | ID: mdl-28380355

Ultraviolet (UV) radiation is a carcinogen that generates DNA lesions. Here, we demonstrate an unexpected role for DGCR8, an RNA binding protein that canonically functions with Drosha to mediate microRNA processing, in the repair of UV-induced DNA lesions. Treatment with UV induced phosphorylation on serine 153 (S153) of DGCR8 in both human and murine cells. S153 phosphorylation was critical for cellular resistance to UV, the removal of UV-induced DNA lesions, and the recovery of RNA synthesis after UV exposure but not for microRNA expression. The RNA-binding and Drosha-binding activities of DGCR8 were not critical for UV resistance. DGCR8 depletion was epistatic to defects in XPA, CSA, and CSB for UV sensitivity. DGCR8 physically interacted with CSB and RNA polymerase II. JNKs were involved in the UV-induced S153 phosphorylation. These findings suggest that UV-induced S153 phosphorylation mediates transcription-coupled nucleotide excision repair of UV-induced DNA lesions in a manner independent of microRNA processing.


DNA Damage , MicroRNAs/metabolism , RNA-Binding Proteins/metabolism , Ribonuclease III/metabolism , Animals , Anisomycin/metabolism , Anthracenes/metabolism , DNA/metabolism , DNA/radiation effects , DNA Repair , HCT116 Cells , HeLa Cells , Humans , MAP Kinase Kinase 4/antagonists & inhibitors , MAP Kinase Kinase 4/metabolism , Mice , Phosphorylation , RNA Polymerase II/metabolism , RNA-Binding Proteins/genetics , Ribonuclease III/genetics , Ultraviolet Rays
12.
Mol Cell Biol ; 37(8)2017 04 15.
Article En | MEDLINE | ID: mdl-28137908

Cyclin E, in conjunction with its catalytic partner cyclin-dependent kinase 2 (CDK2), regulates cell cycle progression as cells exit quiescence and enter S-phase. Multiple mechanisms control cyclin E periodicity during the cell cycle, including phosphorylation-dependent cyclin E ubiquitylation by the SCFFbw7 ubiquitin ligase. Serine 384 (S384) is the critical cyclin E phosphorylation site that stimulates Fbw7 binding and cyclin E ubiquitylation and degradation. Because S384 is autophosphorylated by bound CDK2, this presents a paradox as to how cyclin E can evade autocatalytically induced degradation in order to phosphorylate its other substrates. We found that S384 phosphorylation is dynamically regulated in cells and that cyclin E is specifically dephosphorylated at S384 by the PP2A-B56 phosphatase, thereby uncoupling cyclin E degradation from cyclin E-CDK2 activity. Furthermore, the rate of S384 dephosphorylation is high in interphase but low in mitosis. This provides a mechanism whereby interphase cells can oppose autocatalytic cyclin E degradation and maintain cyclin E-CDK2 activity while also enabling cyclin E destruction in mitosis, when inappropriate cyclin E expression is genotoxic.


Biocatalysis , Cyclin E/metabolism , Protein Phosphatase 2/metabolism , Proteolysis , Cell Cycle , Cell Cycle Proteins/deficiency , Cell Cycle Proteins/metabolism , F-Box Proteins/metabolism , F-Box-WD Repeat-Containing Protein 7 , HCT116 Cells , HeLa Cells , Humans , Phosphorylation , Phosphoserine/metabolism , Protein Stability , Protein Subunits/metabolism , Ubiquitin-Protein Ligases/deficiency , Ubiquitin-Protein Ligases/metabolism
13.
Reproduction ; 151(1): 51-8, 2016 Jan.
Article En | MEDLINE | ID: mdl-26515778

Nucleoporin 50 kDa (NUP50), a component of the nuclear pore complex, is highly expressed in male germ cells, but its role in germ cells is largely unknown. In this study, we analyzed the expression and function of NUP50 during the embryonic development of germ cells using NUP50-deficient mice. NUP50 was expressed in germ cells of both sexes at embryonic day 15.5 (E15.5), E13.5, and E12.5. In addition, NUP50 expression was also detected in primordial germ cells (PGCs) migrating into the genital ridges at E9.5. The gonads of Nup50-/- embryos of both sexes contained few PGCs at both E11.5 and E12.5 and no developing germ cells at E15.5. The migratory PGCs in Nup50-/- embryos at E9.5 showed increased apoptosis but a normal rate of proliferation, resulting in the progressive loss of germ cells at later stages. Taken together, these results suggest that NUP50 plays an essential role in the survival of PGCs during embryonic development.


Embryonic Germ Cells/physiology , Nuclear Pore Complex Proteins/deficiency , Nuclear Pore Complex Proteins/physiology , Animals , Apoptosis , Cell Proliferation , Cell Survival/physiology , Embryo, Mammalian/cytology , Embryonic Development , Female , Fluorescent Antibody Technique , Gestational Age , Gonads/chemistry , Gonads/embryology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Nuclear Pore Complex Proteins/analysis
14.
Cell Rep ; 13(11): 2425-2439, 2015 Dec 22.
Article En | MEDLINE | ID: mdl-26673326

To identify therapeutic targets for glioblastoma (GBM), we performed genome-wide CRISPR-Cas9 knockout (KO) screens in patient-derived GBM stem-like cells (GSCs) and human neural stem/progenitors (NSCs), non-neoplastic stem cell controls, for genes required for their in vitro growth. Surprisingly, the vast majority GSC-lethal hits were found outside of molecular networks commonly altered in GBM and GSCs (e.g., oncogenic drivers). In vitro and in vivo validation of GSC-specific targets revealed several strong hits, including the wee1-like kinase, PKMYT1/Myt1. Mechanistic studies demonstrated that PKMYT1 acts redundantly with WEE1 to inhibit cyclin B-CDK1 activity via CDK1-Y15 phosphorylation and to promote timely completion of mitosis in NSCs. However, in GSCs, this redundancy is lost, most likely as a result of oncogenic signaling, causing GBM-specific lethality.


CRISPR-Cas Systems/genetics , Cell Cycle Proteins/genetics , Genome, Human , Membrane Proteins/genetics , Neoplastic Stem Cells/metabolism , Nuclear Proteins/genetics , Protein Serine-Threonine Kinases/genetics , Protein-Tyrosine Kinases/genetics , CDC2 Protein Kinase/antagonists & inhibitors , CDC2 Protein Kinase/metabolism , Cell Cycle Proteins/antagonists & inhibitors , Cell Cycle Proteins/metabolism , Cell Survival/drug effects , Cyclin B/metabolism , ErbB Receptors/metabolism , Gene Library , Glioblastoma/metabolism , Glioblastoma/pathology , Humans , Membrane Proteins/antagonists & inhibitors , Membrane Proteins/metabolism , Microscopy, Video , Mitosis , Neoplastic Stem Cells/cytology , Nuclear Proteins/antagonists & inhibitors , Nuclear Proteins/metabolism , Phosphorylation , Protein Serine-Threonine Kinases/antagonists & inhibitors , Protein Serine-Threonine Kinases/metabolism , Protein-Tyrosine Kinases/antagonists & inhibitors , Protein-Tyrosine Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Pyrazoles/pharmacology , Pyrimidines/pharmacology , Pyrimidinones , RNA Interference , Time-Lapse Imaging , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism
15.
Cancer Cell ; 26(4): 455-64, 2014 Oct 13.
Article En | MEDLINE | ID: mdl-25314076

Tumor suppressors with widespread impact on carcinogenesis control broad spectra of oncogenic pathways. Protein degradation is an emerging mechanism by which tumor suppressors regulate a diversity of pathways and is exemplified by the SCF(Fbw7) ubiquitin ligase. Rapidly accumulating data indicate that SCF(Fbw7) regulates a network of crucial oncoproteins. Importantly, the FBXW7 gene, which encodes Fbw7, is one of the most frequently mutated genes in human cancers. These studies are yielding important new insights into tumorigenesis and may soon enable therapies targeting the Fbw7 pathway. Here, we focus on the mechanisms and consequences of Fbw7 deregulation in cancers and discuss possible therapeutic approaches.


Cell Cycle Proteins/physiology , F-Box Proteins/physiology , Genes, Tumor Suppressor , Neoplasms/pathology , Ubiquitin-Protein Ligases/physiology , Cell Cycle Proteins/genetics , F-Box Proteins/genetics , F-Box-WD Repeat-Containing Protein 7 , Humans , Mutation , Neoplasms/genetics , Ubiquitin-Protein Ligases/genetics
17.
Oncotarget ; 5(24): 12704-14, 2014 Dec 30.
Article En | MEDLINE | ID: mdl-25587029

As a cell cycle inhibitor and tumor suppressor, p27 is frequently misregulated in human cancers. Increased degradation is the most common mechanism of misregulation, however in some cancers, p27 is mislocalized from its cell cycle inhibitory location in the nucleus, to the cytoplasm. In normal cells cytoplasmic p27 has functions that are distinct from its cell cycle-regulatory nuclear functions. Therefore, an important question is whether localization of p27 to the cytoplasm in tumor cells is primarily a mechanism for cancelling its inhibitory effect on cell proliferation, or whether cytoplasmic p27 has more direct oncogenic actions. To study p27 mislocalization in human cancers we screened a panel of common breast cancer cell lines. We observed that p27 accumulated in the cytoplasm exclusively in cell lines that are Her2+. To address the significance of p27 mislocalization in Her2+ breast cancer cells we interrogated the cellular response to the dual-Her2/EGFR kinase inhibitor, lapatinib. Knockdown of p27 using shRNA sensitized Her2+ cells to lapatinib-induced apoptosis. Moreover, expression of a constitutively cytoplasmic form of p27 (p27ΔNLS) reversed the lapatinib-induced apoptosis, suggesting that cytoplasmic p27 contributed to lapatinib resistance in Her2+ breast cancer cells by suppressing apoptosis. Our results suggest that p27 localization may be useful as a predictive biomarker of therapeutic response in patients with Her2+ breast cancers.


Breast Neoplasms/drug therapy , Breast Neoplasms/metabolism , Cyclin-Dependent Kinase Inhibitor p27/metabolism , Quinazolines/pharmacology , Receptor, ErbB-2/antagonists & inhibitors , Receptor, ErbB-2/metabolism , Animals , Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Breast Neoplasms/enzymology , Breast Neoplasms/pathology , Cell Cycle Checkpoints/drug effects , Cell Cycle Checkpoints/physiology , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Proliferation/physiology , Cyclin-Dependent Kinase Inhibitor p27/antagonists & inhibitors , Cyclin-Dependent Kinase Inhibitor p27/genetics , Cytoplasm/metabolism , Drug Resistance, Neoplasm , Female , Gene Knockdown Techniques , Humans , Lapatinib , Mice , Molecular Targeted Therapy , Phosphorylation , Protein Kinase Inhibitors/pharmacology , RNA, Small Interfering/administration & dosage , RNA, Small Interfering/genetics , Signal Transduction
18.
Genes Dev ; 27(23): 2531-6, 2013 Dec 01.
Article En | MEDLINE | ID: mdl-24298052

The Fbw7 tumor suppressor targets a broad network of proteins for ubiquitylation. Here we show critical functions for Fbw7 dimerization in regulating the specificity and robustness of degradation. Dimerization enables Fbw7 to target substrates through concerted binding to two suboptimal and independent recognition sites. Accordingly, an endogenous dimerization-deficient Fbw7 mutation stabilizes suboptimal substrates. Dimerization increases Fbw7's robustness by preserving its function in the setting of mutations that disable Fbw7 monomers, thereby buffering against pathogenic mutations. Finally, dimerization regulates Fbw7 stability, and this likely involves Fbw7 trans-autoubiquitylation. Our study reveals novel functions of Fbw7 dimerization and an unanticipated complexity in substrate degradation.


Cell Cycle Proteins/metabolism , F-Box Proteins/metabolism , Ubiquitin-Protein Ligases/metabolism , Amino Acid Motifs , Cell Cycle Proteins/chemistry , Dimerization , F-Box Proteins/chemistry , F-Box-WD Repeat-Containing Protein 7 , HCT116 Cells , Humans , Ligases/chemistry , Ligases/metabolism , Mutation , Protein Binding , Protein Stability , Substrate Specificity , Ubiquitin-Protein Ligases/chemistry , Ubiquitination
19.
PLoS Biol ; 11(6): e1001586, 2013.
Article En | MEDLINE | ID: mdl-23776410

FBW7 is a crucial component of an SCF-type E3 ubiquitin ligase, which mediates degradation of an array of different target proteins. The Fbw7 locus comprises three different isoforms, each with its own promoter and each suspected to have a distinct set of substrates. Most FBW7 targets have important functions in developmental processes and oncogenesis, including Notch proteins, which are functionally important substrates of SCF(Fbw7). Notch signalling controls a plethora of cell differentiation decisions in a wide range of species. A prominent role of this signalling pathway is that of mediating lateral inhibition, a process where exchange of signals that repress Notch ligand production amplifies initial differences in Notch activation levels between neighbouring cells, resulting in unequal cell differentiation decisions. Here we show that the downstream Notch signalling effector HES5 directly represses transcription of the E3 ligase Fbw7ß, thereby directly bearing on the process of lateral inhibition. Fbw7(Δ/+) heterozygous mice showed haploinsufficiency for Notch degradation causing impaired intestinal progenitor cell and neural stem cell differentiation. Notably, concomitant inactivation of Hes5 rescued both phenotypes and restored normal stem cell differentiation potential. In silico modelling suggests that the NICD/HES5/FBW7ß positive feedback loop underlies Fbw7 haploinsufficiency. Thus repression of Fbw7ß transcription by Notch signalling is an essential mechanism that is coupled to and required for the correct specification of cell fates induced by lateral inhibition.


Cell Lineage , F-Box Proteins/metabolism , Feedback, Physiological , Intestines/cytology , Neural Stem Cells/cytology , Receptors, Notch/metabolism , Repressor Proteins/metabolism , Animals , Basic Helix-Loop-Helix Transcription Factors/metabolism , Cell Count , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Cell Differentiation , F-Box Proteins/genetics , F-Box-WD Repeat-Containing Protein 7 , Gene Expression Regulation , Genetic Loci , Goblet Cells/cytology , Goblet Cells/metabolism , HCT116 Cells , Haploinsufficiency , Humans , Mice, Knockout , Models, Biological , Neural Stem Cells/metabolism , Transcription, Genetic , Ubiquitin-Protein Ligases/genetics , Ubiquitin-Protein Ligases/metabolism
20.
Proc Natl Acad Sci U S A ; 110(22): 8954-9, 2013 May 28.
Article En | MEDLINE | ID: mdl-23671119

Cyclin-dependent kinases (Cdks) coordinate cell division, and their activities are tightly controlled. Phosphorylation of threonine 14 (T14) and tyrosine 15 (Y15) inhibits Cdks and regulates their activities in numerous physiologic contexts. Although the roles of Cdk1 inhibitory phosphorylation during mitosis are well described, studies of Cdk2 inhibitory phosphorylation during S phrase have largely been indirect. To specifically study the functions of Cdk2 inhibitory phosphorylation, we used gene targeting to make an endogenous Cdk2 knockin allele in human cells, termed Cdk2AF, which prevents Cdk2 T14 and Y15 phosphorylation. Cdk2AF caused premature S-phase entry, rapid cyclin E degradation, abnormal DNA replication, and genome instability. Cdk2AF cells also exhibited strikingly abnormal responses to replication stress, accumulated irreparable DNA damage, and permanently exited the cell cycle after transient exposure to S-phase inhibitors. Our results reveal the specific and essential roles of Cdk2 inhibitory phosphorylation in the successful execution of the replication stress checkpoint response and in maintaining genome integrity.


Cyclin-Dependent Kinase 2/metabolism , DNA Replication/physiology , S Phase/physiology , Signal Transduction/physiology , Cell Cycle Proteins/metabolism , Cells, Cultured , Cyclin E/metabolism , Cyclin-Dependent Kinase 2/genetics , DNA Damage/physiology , DNA-Binding Proteins/metabolism , Electrophoresis, Gel, Pulsed-Field , Flow Cytometry , Gene Knock-In Techniques , Genomic Instability/physiology , Humans , Microfluidics , Nuclear Proteins/metabolism , Phosphorylation , Protein-Tyrosine Kinases/metabolism , RNA Interference , RNA, Small Interfering/genetics , S Phase Cell Cycle Checkpoints/physiology , Transcription Factors/metabolism
...