Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 57
Filter
Add more filters










Publication year range
1.
Adv Sci (Weinh) ; 11(28): e2308255, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38757578

ABSTRACT

Metabolic alterations in cancers can be exploited for diagnostic, prognostic, and therapeutic purposes. This is exemplified by 18F-fluorodeoxyglucose (FDG)-positron emission tomography (FDG-PET), an imaging tool that relies on enhanced glucose uptake by tumors for diagnosis and staging. By performing transcriptomic analysis of breast cancer (BC) samples from patients stratified by FDG-PET, a 54-gene signature (PETsign) is identified that recapitulates FDG uptake. PETsign is independently prognostic of clinical outcome in luminal BCs, the most common and heterogeneous BC molecular subtype, which requires improved stratification criteria to guide therapeutic decision-making. The prognostic power of PETsign is stable across independent BC cohorts and disease stages including the earliest BC stage, arguing that PETsign is an ab initio metabolic signature. Transcriptomic and metabolomic analysis of BC cells reveals that PETsign predicts enhanced glycolytic dependence and reduced reliance on fatty acid oxidation. Moreover, coamplification of PETsign genes occurs frequently in BC arguing for their causal role in pathogenesis. CXCL8 and EGFR signaling pathways feature strongly in PETsign, and their activation in BC cells causes a shift toward a glycolytic phenotype. Thus, PETsign serves as a molecular surrogate for FDG-PET that could inform clinical management strategies for BC patients.


Subject(s)
Breast Neoplasms , Fluorodeoxyglucose F18 , Positron-Emission Tomography , Humans , Breast Neoplasms/metabolism , Breast Neoplasms/genetics , Female , Positron-Emission Tomography/methods , Fluorodeoxyglucose F18/metabolism , Prognosis , Gene Expression Profiling/methods , Transcriptome/genetics
2.
J Neurosci Res ; 101(8): 1345-1359, 2023 08.
Article in English | MEDLINE | ID: mdl-37031448

ABSTRACT

Classical dynamins (DNMs) are GTPase proteins engaged in endocytosis, a fundamental process for cargo internalization from the plasma membrane. In mammals, three DNM genes are present with different expression patterns. DNM1 is expressed at high levels in neurons, where it takes place in the recycling of synaptic vesicles; DNM2 is ubiquitously expressed, while DNM3 is found in the brain and in the testis. Due to the conservation of genes in comparison to mammals, we took advantage of a zebrafish model for functional characterization of dnm1a, ortholog of mammalian DNM1. Our data strongly demonstrated that dnm1a has a nervous tissue-specific expression pattern and plays a role in the formation of both axon and synapse. This is the first in vivo study that collects evidence about the effects of dnm1a loss of function in zebrafish, thus providing a new excellent model to be used in different scientific fields.


Subject(s)
Nerve Tissue , Zebrafish , Animals , Male , Axons , Neurons/metabolism , Synapses/metabolism , Mammals
3.
Cell Rep ; 40(12): 111396, 2022 09 20.
Article in English | MEDLINE | ID: mdl-36130505

ABSTRACT

Deubiquitinating enzymes are key regulators of the ubiquitin-proteasome system and cell cycle, and their dysfunction leads to tumorigenesis. Our in vivo drop-out screens in patient-derived xenograft models identify USP7 as a regulator of melanoma. We show that USP7 downregulation induces cellular senescence, arresting melanoma growth in vivo and proliferation in vitro in BRAF- and NRAS-mutant melanoma. We provide a comprehensive understanding of targets and networks affected by USP7 depletion by performing a global transcriptomic and proteomics analysis. We show that RRM2 is a USP7 target and is regulated by USP7 during S phase of the cell cycle. Ectopic expression of RRM2 in USP7-depleted cells rescues the senescent phenotype. Pharmacological inhibition of USP7 by P5091 phenocopies the shUSP7-induced senescent phenotype. We show that the bifunctional histone deacetylase (HDAC)/LSD1 inhibitor domatinostat has an additive antitumor effect, eliminating P5091-induced senescent cells, paving the way to a therapeutic combination for individuals with melanoma.


Subject(s)
Histone Deacetylase Inhibitors , Melanoma , Cell Line, Tumor , Histone Deacetylase Inhibitors/pharmacology , Histone Deacetylases , Histone Demethylases/genetics , Humans , Melanoma/pathology , Proteasome Endopeptidase Complex , Proto-Oncogene Proteins B-raf/genetics , Thiophenes , Ubiquitin-Specific Peptidase 7/metabolism , Ubiquitins
4.
Cancers (Basel) ; 12(11)2020 Nov 13.
Article in English | MEDLINE | ID: mdl-33202906

ABSTRACT

Metastases are the primary cause of cancer-related deaths. The underlying molecular and biological mechanisms remain, however, elusive, thus preventing the design of specific therapies. In melanomas, the metastatic process is influenced by the acquisition of metastasis-associated mutational and epigenetic traits and the activation of metastatic-specific signaling pathways in the primary melanoma. In the current study, we investigated the role of an adaptor protein of the Shc family (ShcD) in the acquisition of metastatic properties by melanoma cells, exploiting our cohort of patient-derived xenografts (PDXs). We provide evidence that the depletion of ShcD expression increases a spread cell shape and the capability of melanoma cells to attach to the extracellular matrix while its overexpression switches their morphology from elongated to rounded on 3D matrices, enhances cells' invasive phenotype, as observed on collagen gel, and favors metastasis formation in vivo. ShcD overexpression sustains amoeboid movement in melanoma cells, by suppressing the Rac1 signaling pathway through the confinement of DOCK4 in the cytoplasm. Inactivation of the ShcD signaling pathway makes melanoma cells more sensitive to therapeutic treatments. Consistently, ShcD expression predicts poor outcome in a cohort of 183 primary melanoma patients.

5.
Nat Commun ; 11(1): 3020, 2020 06 15.
Article in English | MEDLINE | ID: mdl-32541686

ABSTRACT

The subversion of endocytic routes leads to malignant transformation and has been implicated in human cancers. However, there is scarce evidence for genetic alterations of endocytic proteins as causative in high incidence human cancers. Here, we report that Epsin 3 (EPN3) is an oncogene with prognostic and therapeutic relevance in breast cancer. Mechanistically, EPN3 drives breast tumorigenesis by increasing E-cadherin endocytosis, followed by the activation of a ß-catenin/TCF4-dependent partial epithelial-to-mesenchymal transition (EMT), followed by the establishment of a TGFß-dependent autocrine loop that sustains EMT. EPN3-induced partial EMT is instrumental for the transition from in situ to invasive breast carcinoma, and, accordingly, high EPN3 levels are detected at the invasive front of human breast cancers and independently predict metastatic rather than loco-regional recurrence. Thus, we uncover an endocytic-based mechanism able to generate TGFß-dependent regulatory loops conferring cellular plasticity and invasive behavior.


Subject(s)
Adaptor Proteins, Vesicular Transport/metabolism , Breast Neoplasms/physiopathology , Endocytosis , Adaptor Proteins, Vesicular Transport/genetics , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cadherins/genetics , Cadherins/metabolism , Epithelial-Mesenchymal Transition , Female , Gene Expression Regulation, Neoplastic , Humans , Neoplasm Invasiveness , Neoplasm Metastasis , Signal Transduction , Transcription Factor 4/genetics , Transcription Factor 4/metabolism , Transforming Growth Factor beta/metabolism , beta Catenin/genetics , beta Catenin/metabolism
6.
G3 (Bethesda) ; 9(10): 3359-3367, 2019 10 07.
Article in English | MEDLINE | ID: mdl-31451549

ABSTRACT

MDM2 regulates a variety of cellular processes through its dual protein:protein interaction and ubiquitin ligase activities. One major function of MDM2 is to bind and ubiquitinate P53, thereby regulating its proteasomal degradation. This function is in turn controlled by the cell fate determinant NUMB, which binds to and inhibits MDM2 via a short stretch of 11 amino acids, contained in its phosphotyrosine-binding (PTB) domain, encoded by exon 3 of the NUMB gene. The NUMB-MDM2-P53 circuitry is relevant to the specification of the stem cell fate and its subversion has been shown to be causal in breast cancer leading to the emergence of cancer stem cells. While extensive work on the evolutionary aspects of the MDM2/P53 circuitry has provided hints as to how these two proteins have evolved together to maintain conserved and linked functions, little is known about the evolution of the NUMB gene and, in particular, how it developed the ability to regulate MDM2 function. Here, we show that NUMB is a metazoan gene, which acquired exon 3 in the common ancestor of the Chordate lineage, first being present in the Cephalochordate and Tunicate subphyla, but absent in invertebrates. We provide experimental evidence showing that since its emergence, exon 3 conferred to the PTB domain of NUMB the ability to bind and to regulate MDM2 functions.


Subject(s)
Chordata/classification , Chordata/genetics , Exons , Gene Expression Regulation , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Proto-Oncogene Proteins c-mdm2/genetics , Proto-Oncogene Proteins c-mdm2/metabolism , Animals , Evolution, Molecular , Models, Molecular , Nerve Tissue Proteins/chemistry , Phylogeny , Protein Conformation , Protein Interaction Domains and Motifs , Protein Isoforms , Proto-Oncogene Proteins c-mdm2/chemistry , Structure-Activity Relationship
7.
Cell Rep ; 27(10): 3049-3061.e6, 2019 06 04.
Article in English | MEDLINE | ID: mdl-31167147

ABSTRACT

Adaptor protein 2 (AP2) is a major constituent of clathrin-coated pits (CCPs). Whether it is essential for all forms of clathrin-mediated endocytosis (CME) in mammalian cells is an open issue. Here, we demonstrate, by live TIRF microscopy, the existence of a subclass of relatively short-lived CCPs lacking AP2 under physiological, unperturbed conditions. This subclass is retained in AP2-knockout cells and is able to support the internalization of epidermal growth factor receptor (EGFR) but not of transferrin receptor (TfR). The AP2-independent internalization mechanism relies on the endocytic adaptors eps15, eps15L1, and epsin1. The absence of AP2 impairs the recycling of the EGFR to the cell surface, thereby augmenting its degradation. Accordingly, under conditions of AP2 ablation, we detected dampening of EGFR-dependent AKT signaling and cell migration, arguing that distinct classes of CCPs could provide specialized functions in regulating EGFR recycling and signaling.


Subject(s)
Clathrin-Coated Vesicles/physiology , Signal Transduction , Adaptor Protein Complex 2/antagonists & inhibitors , Adaptor Protein Complex 2/genetics , Adaptor Protein Complex 2/metabolism , Adaptor Proteins, Signal Transducing/antagonists & inhibitors , Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Signal Transducing/metabolism , Adaptor Proteins, Vesicular Transport/antagonists & inhibitors , Adaptor Proteins, Vesicular Transport/genetics , Adaptor Proteins, Vesicular Transport/metabolism , Animals , Cell Line , Cell Movement , Endocytosis , Epidermal Growth Factor/metabolism , ErbB Receptors/metabolism , Gene Editing , HeLa Cells , Humans , Mice , Microscopy, Fluorescence , Proto-Oncogene Proteins c-akt/metabolism , RNA Interference , RNA, Small Interfering/metabolism , Transcriptional Activation
8.
EBioMedicine ; 42: 352-362, 2019 Apr.
Article in English | MEDLINE | ID: mdl-30846393

ABSTRACT

BACKGROUND: Breast cancers show variations in the number and biological aggressiveness of cancer stem cells that correlate with their clinico-prognostic and molecular heterogeneity. Thus, prognostic stratification of breast cancers based on cancer stem cells might help guide patient management. METHODS: We derived a 20-gene stem cell signature from the transcriptional profile of normal mammary stem cells, capable of identifying breast cancers with a homogeneous profile and poor prognosis in in silico analyses. The clinical value of this signature was assessed in a prospective-retrospective cohort of 2, 453 breast cancer patients. Models for predicting individual risk of metastasis were developed from expression data of the 20 genes in patients randomly assigned to a training set, using the ridge-penalized Cox regression, and tested in an independent validation set. FINDINGS: Analyses revealed that the 20-gene stem cell signature provided prognostic information in Triple-Negative and Luminal breast cancer patients, independently of standard clinicopathological parameters. Through functional studies in individual tumours, we correlated the risk score assigned by the signature with the proliferative and self-renewal potential of the cancer stem cell population. By retraining the 20-gene signature in Luminal patients, we derived the risk model, StemPrintER, which predicted early and late recurrence independently of standard prognostic factors. INTERPRETATION: Our findings indicate that the 20-gene stem cell signature, by its unique ability to interrogate the biology of cancer stem cells of the primary tumour, provides a reliable estimate of metastatic risk in Triple-Negative and Luminal breast cancer patients independently of standard clinicopathological parameters.


Subject(s)
Biomarkers, Tumor , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Multigene Family , Neoplastic Stem Cells/metabolism , Adult , Aged , Breast Neoplasms/mortality , Female , Gene Expression Profiling , Genomics/methods , Humans , Middle Aged , Neoplasm Grading , Neoplasm Metastasis , Neoplasm Staging , Prognosis , Proportional Hazards Models , Retrospective Studies , Transcriptome
9.
Life Sci Alliance ; 2(1)2019 02.
Article in English | MEDLINE | ID: mdl-30692166

ABSTRACT

EPS15 and its homologous EPS15L1 are endocytic accessory proteins. Studies in mammalian cell lines suggested that EPS15 and EPS15L1 regulate endocytosis in a redundant manner. However, at the organismal level, it is not known to which extent the functions of the two proteins overlap. Here, by exploiting various constitutive and conditional null mice, we report redundant and nonredundant functions of the two proteins. EPS15L1 displays a unique nonredundant role in the nervous system, whereas both proteins are fundamental during embryo development as shown by the embryonic lethality of -Eps15/Eps15L1-double KO mice. At the cellular level, the major process redundantly regulated by EPS15 and EPS15L1 is the endocytosis of the transferrin receptor, a pathway that sustains the development of red blood cells and controls iron homeostasis. Consequently, hematopoietic-specific conditional Eps15/Eps15L1-double KO mice display traits of microcytic hypochromic anemia, due to a cell-autonomous defect in iron internalization.


Subject(s)
Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Signal Transducing/metabolism , Endocytosis/physiology , Anemia, Hypochromic/genetics , Animals , Behavior Rating Scale , Embryonic Development/physiology , Erythrocytes/metabolism , Fibroblasts/metabolism , Gene Knockout Techniques , Genes, Lethal/physiology , Hippocampus/cytology , Iron/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Neurons/metabolism , Receptors, Transferrin/metabolism , Sequence Homology, Amino Acid , Structural Homology, Protein , Synapses/metabolism
10.
J Cell Biol ; 217(9): 3161-3182, 2018 09 03.
Article in English | MEDLINE | ID: mdl-30061108

ABSTRACT

The endocytic protein NUMB has been implicated in the control of various polarized cellular processes, including the acquisition of mesenchymal migratory traits through molecular mechanisms that have only been partially defined. Here, we report that NUMB is a negative regulator of a specialized set of understudied, apically restricted, actin-based protrusions, the circular dorsal ruffles (CDRs), induced by either PDGF or HGF stimulation. Through its PTB domain, NUMB binds directly to an N-terminal NPLF motif of the ARF6 guanine nucleotide exchange factor, EFA6B, and promotes its exchange activity in vitro. In cells, a NUMB-EFA6B-ARF6 axis regulates the recycling of the actin regulatory cargo RAC1 and is critical for the formation of CDRs that mark the acquisition of a mesenchymal mode of motility. Consistently, loss of NUMB promotes HGF-induced cell migration and invasion. Thus, NUMB negatively controls membrane protrusions and the acquisition of mesenchymal migratory traits by modulating EFA6B-ARF6 activity.


Subject(s)
ADP-Ribosylation Factors/metabolism , Cell Movement/physiology , Guanine Nucleotide Exchange Factors/metabolism , Membrane Proteins/metabolism , Mesoderm/metabolism , Nerve Tissue Proteins/metabolism , ADP-Ribosylation Factor 6 , Cell Line, Tumor , Cell Polarity , HeLa Cells , Hepatocyte Growth Factor/metabolism , Humans , Membrane Proteins/genetics , Mesoderm/cytology , Nerve Tissue Proteins/genetics , Platelet-Derived Growth Factor/metabolism , Protein Binding , Protein Domains , RNA Interference , RNA, Small Interfering/genetics , rac1 GTP-Binding Protein/metabolism
12.
Stem Cell Reports ; 10(4): 1412-1425, 2018 04 10.
Article in English | MEDLINE | ID: mdl-29551673

ABSTRACT

Cancer-initiating cells (CICs) have been implicated in tumor development and aggressiveness. In ovarian carcinoma (OC), CICs drive tumor formation, dissemination, and recurrence, as well as drug resistance, thus accounting for the high death-to-incidence ratio of this neoplasm. However, the molecular mechanisms that underlie such a pathogenic role of ovarian CICs (OCICs) remain elusive. Here, we have capitalized on primary cells either from OC or from its tissues of origin to obtain the transcriptomic profile associated with OCICs. Among the genes differentially expressed in OCICs, we focused on CD73, which encodes the membrane-associated 5'-ectonucleotidase. The genetic inactivation of CD73 in OC cells revealed that this molecule is causally involved in sphere formation and tumor initiation, thus emerging as a driver of OCIC function. Furthermore, functional inhibition of CD73 via either a chemical compound or a neutralizing antibody reduced sphere formation and tumorigenesis, highlighting the druggability of CD73 in the context of OCIC-directed therapies. The biological function of CD73 in OCICs required its enzymatic activity and involved adenosine signaling. Mechanistically, CD73 promotes the expression of stemness and epithelial-mesenchymal transition-associated genes, implying a regulation of OCIC function at the transcriptional level. CD73, therefore, is involved in OCIC biology and may represent a therapeutic target for innovative treatments aimed at OC eradication.


Subject(s)
5'-Nucleotidase/metabolism , Epithelial-Mesenchymal Transition , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/pathology , Ovarian Neoplasms/metabolism , Ovarian Neoplasms/pathology , Adenosine/metabolism , Cell Line, Tumor , Cell Proliferation , Epithelial-Mesenchymal Transition/genetics , Epithelium/pathology , Fallopian Tubes/pathology , Female , GPI-Linked Proteins/metabolism , Gene Expression Regulation, Neoplastic , Humans , Ovarian Neoplasms/genetics , Spheroids, Cellular
14.
J Cell Biol ; 217(2): 745-762, 2018 02 05.
Article in English | MEDLINE | ID: mdl-29269425

ABSTRACT

Numb functions as an oncosuppressor by inhibiting Notch signaling and stabilizing p53. This latter effect depends on the interaction of Numb with Mdm2, the E3 ligase that ubiquitinates p53 and commits it to degradation. In breast cancer (BC), loss of Numb results in a reduction of p53-mediated responses including sensitivity to genotoxic drugs and maintenance of homeostasis in the stem cell compartment. In this study, we show that the Numb-Mdm2 interaction represents a fuzzy complex mediated by a short Numb sequence encompassing its alternatively spliced exon 3 (Ex3), which is necessary and sufficient to inhibit Mdm2 and prevent p53 degradation. Alterations in the Numb splicing pattern are critical in BC as shown by increased chemoresistance of tumors displaying reduced levels of Ex3-containing isoforms, an effect that could be mechanistically linked to diminished p53 levels. A reduced level of Ex3-less Numb isoforms independently predicts poor outcome in BCs harboring wild-type p53. Thus, we have uncovered an important mechanism of chemoresistance and progression in p53-competent BCs.


Subject(s)
Breast Neoplasms/metabolism , Membrane Proteins/metabolism , Nerve Tissue Proteins/metabolism , Proto-Oncogene Proteins c-mdm2/metabolism , Alternative Splicing , Breast Neoplasms/pathology , Cell Line, Tumor , Female , Humans , Membrane Proteins/genetics , Nerve Tissue Proteins/genetics , Protein Isoforms/genetics , Protein Isoforms/metabolism , Proto-Oncogene Proteins c-mdm2/genetics , Tumor Suppressor Protein p53/metabolism
15.
Science ; 356(6338): 617-624, 2017 05 12.
Article in English | MEDLINE | ID: mdl-28495747

ABSTRACT

The integration of endocytic routes is critical to regulate receptor signaling. A nonclathrin endocytic (NCE) pathway of the epidermal growth factor receptor (EGFR) is activated at high ligand concentrations and targets receptors to degradation, attenuating signaling. Here we performed an unbiased molecular characterization of EGFR-NCE. We identified NCE-specific regulators, including the endoplasmic reticulum (ER)-resident protein reticulon 3 (RTN3) and a specific cargo, CD147. RTN3 was critical for EGFR/CD147-NCE, promoting the creation of plasma membrane (PM)-ER contact sites that were required for the formation and/or maturation of NCE invaginations. Ca2+ release at these sites, triggered by inositol 1,4,5-trisphosphate (IP3)-dependent activation of ER Ca2+ channels, was needed for the completion of EGFR internalization. Thus, we identified a mechanism of EGFR endocytosis that relies on ER-PM contact sites and local Ca2+ signaling.


Subject(s)
Carrier Proteins/metabolism , Cell Membrane/metabolism , Endocytosis , ErbB Receptors/metabolism , Membrane Proteins/metabolism , Nerve Tissue Proteins/metabolism , Basigin/metabolism , Calcium Signaling , Cell Line , Endoplasmic Reticulum/metabolism , Humans
16.
Nat Commun ; 8: 14797, 2017 03 16.
Article in English | MEDLINE | ID: mdl-28300085

ABSTRACT

The docking protein p140Cap negatively regulates tumour cell features. Its relevance on breast cancer patient survival, as well as its ability to counteract relevant cancer signalling pathways, are not fully understood. Here we report that in patients with ERBB2-amplified breast cancer, a p140Cap-positive status associates with a significantly lower probability of developing a distant event, and a clear difference in survival. p140Cap dampens ERBB2-positive tumour cell progression, impairing tumour onset and growth in the NeuT mouse model, and counteracting epithelial mesenchymal transition, resulting in decreased metastasis formation. One major mechanism is the ability of p140Cap to interfere with ERBB2-dependent activation of Rac GTPase-controlled circuitries. Our findings point to a specific role of p140Cap in curbing the aggressiveness of ERBB2-amplified breast cancers and suggest that, due to its ability to impinge on specific molecular pathways, p140Cap may represent a predictive biomarker of response to targeted anti-ERBB2 therapies.


Subject(s)
Adaptor Proteins, Vesicular Transport/metabolism , Breast Neoplasms/metabolism , Receptor, ErbB-2/metabolism , rac GTP-Binding Proteins/metabolism , Adaptor Proteins, Vesicular Transport/genetics , Animals , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Cell Line, Tumor , Disease Progression , Female , Gene Expression Regulation, Neoplastic , Humans , Mice, Inbred BALB C , Mice, Transgenic , Neoplasm Metastasis , Neoplasms, Experimental/genetics , Neoplasms, Experimental/metabolism , Neoplasms, Experimental/pathology , Receptor, ErbB-2/genetics , rac GTP-Binding Proteins/genetics
17.
Cell Rep ; 18(13): 3242-3256, 2017 03 28.
Article in English | MEDLINE | ID: mdl-28355574

ABSTRACT

Our understanding of the molecular determinants of cancer is still inadequate because of cancer heterogeneity. Here, using epithelial ovarian cancer (EOC) as a model system, we analyzed a minute amount of patient-derived epithelial cells from either healthy or cancerous tissues by single-shot mass-spectrometry-based phosphoproteomics. Using a multi-disciplinary approach, we demonstrated that primary cells recapitulate tissue complexity and represent a valuable source of differentially expressed proteins and phosphorylation sites that discriminate cancer from healthy cells. Furthermore, we uncovered kinase signatures associated with EOC. In particular, CDK7 targets were characterized in both EOC primary cells and ovarian cancer cell lines. We showed that CDK7 controls cell proliferation and that pharmacological inhibition of CDK7 selectively represses EOC cell proliferation. Our approach defines the molecular landscape of EOC, paving the way for efficient therapeutic approaches for patients. Finally, we highlight the potential of phosphoproteomics to identify clinically relevant and druggable pathways in cancer.


Subject(s)
Ovarian Neoplasms/metabolism , Phosphoproteins/metabolism , Protein Kinases/metabolism , Proteomics/methods , Carcinoma, Ovarian Epithelial , Epithelial Cells/metabolism , Female , Humans , Neoplasm Proteins/metabolism , Neoplasms, Glandular and Epithelial/metabolism , Spliceosomes/metabolism , Tumor Cells, Cultured
18.
EMBO J ; 36(3): 260-273, 2017 02 01.
Article in English | MEDLINE | ID: mdl-28007894

ABSTRACT

The F-box protein FBXW7 is the substrate-recruiting subunit of an SCF ubiquitin ligase and a major tumor-suppressor protein that is altered in several human malignancies. Loss of function of FBXW7 results in the stabilization of numerous proteins that orchestrate cell proliferation and survival. Little is known about proteins that directly regulate the function of this protein. In the current work, we have mapped the interactome of the enigmatic pseudophosphatase STYX We reasoned that a catalytically inactive phosphatase might have adopted novel mechanisms of action. The STYX interactome contained several F-box proteins, including FBXW7. We show that STYX binds to the F-box domain of FBXW7 and disables its recruitment into the SCF complex. Therefore, STYX acts as a direct inhibitor of FBXW7, affecting the cellular levels of its substrates. Furthermore, we find that levels of STYX and FBXW7 are anti-correlated in breast cancer patients, which affects disease prognosis. We propose the STYX-FBXW7 interaction as a promising drug target for future investigations.


Subject(s)
Cell Cycle Proteins/antagonists & inhibitors , F-Box Proteins/antagonists & inhibitors , Intracellular Signaling Peptides and Proteins/metabolism , Nuclear Proteins/metabolism , SKP Cullin F-Box Protein Ligases/antagonists & inhibitors , Ubiquitin-Protein Ligases/antagonists & inhibitors , Breast Neoplasms/diagnosis , Breast Neoplasms/pathology , F-Box-WD Repeat-Containing Protein 7 , Humans
19.
Elife ; 52016 Sep 26.
Article in English | MEDLINE | ID: mdl-27668431

ABSTRACT

TFE-fusion renal cell carcinomas (TFE-fusion RCCs) are caused by chromosomal translocations that lead to overexpression of the TFEB and TFE3 genes (Kauffman et al., 2014). The mechanisms leading to kidney tumor development remain uncharacterized and effective therapies are yet to be identified. Hence, the need to model these diseases in an experimental animal system (Kauffman et al., 2014). Here, we show that kidney-specific TFEB overexpression in transgenic mice, resulted in renal clear cells, multi-layered basement membranes, severe cystic pathology, and ultimately papillary carcinomas with hepatic metastases. These features closely recapitulate those observed in both TFEB- and TFE3-mediated human kidney tumors. Analysis of kidney samples revealed transcriptional induction and enhanced signaling of the WNT ß-catenin pathway. WNT signaling inhibitors normalized the proliferation rate of primary kidney cells and significantly rescued the disease phenotype in vivo. These data shed new light on the mechanisms underlying TFE-fusion RCCs and suggest a possible therapeutic strategy based on the inhibition of the WNT pathway.

20.
EMBO Rep ; 17(7): 1061-80, 2016 07.
Article in English | MEDLINE | ID: mdl-27255086

ABSTRACT

The mechanisms of tumor cell dissemination and the contribution of membrane trafficking in this process are poorly understood. Through a functional siRNA screening of human RAB GTPases, we found that RAB2A, a protein essential for ER-to-Golgi transport, is critical in promoting proteolytic activity and 3D invasiveness of breast cancer (BC) cell lines. Remarkably, RAB2A is amplified and elevated in human BC and is a powerful and independent predictor of disease recurrence in BC patients. Mechanistically, RAB2A acts at two independent trafficking steps. Firstly, by interacting with VPS39, a key component of the late endosomal HOPS complex, it controls post-endocytic trafficking of membrane-bound MT1-MMP, an essential metalloprotease for matrix remodeling and invasion. Secondly, it further regulates Golgi transport of E-cadherin, ultimately controlling junctional stability, cell compaction, and tumor invasiveness. Thus, RAB2A is a novel trafficking determinant essential for regulation of a mesenchymal invasive program of BC dissemination.


Subject(s)
Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cadherins/metabolism , Golgi Apparatus/metabolism , Matrix Metalloproteinase 14/metabolism , rab GTP-Binding Proteins/metabolism , Biomarkers, Tumor , Breast Neoplasms/genetics , Breast Neoplasms/mortality , Cell Line, Tumor , Endosomes/metabolism , Exocytosis , Extracellular Matrix/metabolism , Female , Gene Expression , Gene Expression Profiling , Gene Silencing , Homeodomain Proteins/metabolism , Humans , Neoplasm Invasiveness , Prognosis , Protein Transport , Proteolysis , Recurrence , Tumor Suppressor Proteins/metabolism , rab GTP-Binding Proteins/genetics
SELECTION OF CITATIONS
SEARCH DETAIL