Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 33
Filter
Add more filters










Publication year range
1.
Nat Commun ; 15(1): 5119, 2024 Jun 15.
Article in English | MEDLINE | ID: mdl-38879572

ABSTRACT

One open question in the biology of growth factor receptors is how a quantitative input (i.e., ligand concentration) is decoded by the cell to produce specific response(s). Here, we show that an EGFR endocytic mechanism, non-clathrin endocytosis (NCE), which is activated only at high ligand concentrations and targets receptor to degradation, requires a tripartite organelle platform involving the plasma membrane (PM), endoplasmic reticulum (ER) and mitochondria. At these contact sites, EGFR-dependent, ER-generated Ca2+ oscillations are sensed by mitochondria, leading to increased metabolism and ATP production. Locally released ATP is required for cortical actin remodeling and EGFR-NCE vesicle fission. The same biochemical circuitry is also needed for an effector function of EGFR, i.e., collective motility. The multiorganelle signaling platform herein described mediates direct communication between EGFR signaling and mitochondrial metabolism, and is predicted to have a broad impact on cell physiology as it is activated by another growth factor receptor, HGFR/MET.


Subject(s)
Adenosine Triphosphate , Endocytosis , Endoplasmic Reticulum , ErbB Receptors , Mitochondria , Signal Transduction , Mitochondria/metabolism , ErbB Receptors/metabolism , Endoplasmic Reticulum/metabolism , Humans , Adenosine Triphosphate/metabolism , Animals , Cell Membrane/metabolism , Calcium Signaling/physiology , Calcium/metabolism
2.
Adv Sci (Weinh) ; : e2308255, 2024 May 17.
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.

3.
Cell Mol Life Sci ; 80(9): 244, 2023 Aug 10.
Article in English | MEDLINE | ID: mdl-37561190

ABSTRACT

Breast cancer (BC) is the most frequently diagnosed cancer and one of the major causes of cancer death. Despite enormous progress in its management, both from the therapeutic and early diagnosis viewpoints, still around 700,000 patients succumb to the disease each year, worldwide. Late recurrency is the major problem in BC, with many patients developing distant metastases several years after the successful eradication of the primary tumor. This is linked to the phenomenon of metastatic dormancy, a still mysterious trait of the natural history of BC, and of several other types of cancer, by which metastatic cells remain dormant for long periods of time before becoming reactivated to initiate the clinical metastatic disease. In recent years, it has become clear that cancers are best understood if studied as ecosystems in which the impact of non-cancer-cell-autonomous events-dependent on complex interaction between the cancer and its environment, both local and systemic-plays a paramount role, probably as significant as the cell-autonomous alterations occurring in the cancer cell. In adopting this perspective, a metabolic vision of the cancer ecosystem is bound to improve our understanding of the natural history of cancer, across space and time. In BC, many metabolic pathways are coopted into the cancer ecosystem, to serve the anabolic and energy demands of the cancer. Their study is shedding new light on the most critical aspect of BC management, of metastatic dissemination, and that of the related phenomenon of dormancy and fostering the application of the knowledge to the development of metabolic therapies.


Subject(s)
Breast Neoplasms , Humans , Female , Breast Neoplasms/pathology , Ecosystem
4.
Nat Rev Mol Cell Biol ; 22(9): 625-643, 2021 09.
Article in English | MEDLINE | ID: mdl-34075221

ABSTRACT

Endocytosis allows cells to transport particles and molecules across the plasma membrane. In addition, it is involved in the termination of signalling through receptor downmodulation and degradation. This traditional outlook has been substantially modified in recent years by discoveries that endocytosis and subsequent trafficking routes have a profound impact on the positive regulation and propagation of signals, being key for the spatiotemporal regulation of signal transmission in cells. Accordingly, endocytosis and membrane trafficking regulate virtually every aspect of cell physiology and are frequently subverted in pathological conditions. Two key aspects of endocytic control over signalling are coming into focus: context-dependency and long-range effects. First, endocytic-regulated outputs are not stereotyped but heavily dependent on the cell-specific regulation of endocytic networks. Second, endocytic regulation has an impact not only on individual cells but also on the behaviour of cellular collectives. Herein, we will discuss recent advancements in these areas, highlighting how endocytic trafficking impacts complex cell properties, including cell polarity and collective cell migration, and the relevance of these mechanisms to disease, in particular cancer.


Subject(s)
Cell Membrane/metabolism , Endocytosis/physiology , Endosomes/metabolism , Animals , Biological Transport , Cell Movement , Cell Polarity , Humans , Morphogenesis , Signal Transduction
5.
Nature ; 592(7856): 799-803, 2021 04.
Article in English | MEDLINE | ID: mdl-33854232

ABSTRACT

Mammalian development, adult tissue homeostasis and the avoidance of severe diseases including cancer require a properly orchestrated cell cycle, as well as error-free genome maintenance. The key cell-fate decision to replicate the genome is controlled by two major signalling pathways that act in parallel-the MYC pathway and the cyclin D-cyclin-dependent kinase (CDK)-retinoblastoma protein (RB) pathway1,2. Both MYC and the cyclin D-CDK-RB axis are commonly deregulated in cancer, and this is associated with increased genomic instability. The autophagic tumour-suppressor protein AMBRA1 has been linked to the control of cell proliferation, but the underlying molecular mechanisms remain poorly understood. Here we show that AMBRA1 is an upstream master regulator of the transition from G1 to S phase and thereby prevents replication stress. Using a combination of cell and molecular approaches and in vivo models, we reveal that AMBRA1 regulates the abundance of D-type cyclins by mediating their degradation. Furthermore, by controlling the transition from G1 to S phase, AMBRA1 helps to maintain genomic integrity during DNA replication, which counteracts developmental abnormalities and tumour growth. Finally, we identify the CHK1 kinase as a potential therapeutic target in AMBRA1-deficient tumours. These results advance our understanding of the control of replication-phase entry and genomic integrity, and identify the AMBRA1-cyclin D pathway as a crucial cell-cycle-regulatory mechanism that is deeply interconnected with genomic stability in embryonic development and tumorigenesis.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Cyclin D/metabolism , Genomic Instability , S Phase , Animals , Cell Line , Cell Proliferation , Checkpoint Kinase 1/antagonists & inhibitors , Cyclin-Dependent Kinases/metabolism , DNA Replication , Gene Expression Regulation, Developmental , Genes, Tumor Suppressor , Humans , Mice , Mice, Knockout , Synthetic Lethal Mutations
6.
Front Oncol ; 9: 848, 2019.
Article in English | MEDLINE | ID: mdl-31544066

ABSTRACT

Tumors driven by mutant KRAS are among the most aggressive and refractory to treatment. Unfortunately, despite the efforts, targeting alterations of this GTPase, either directly or by acting on the downstream signaling cascades, has been, so far, largely unsuccessful. However, recently, novel therapeutic opportunities are emerging based on the effect that this oncogenic lesion exerts in rewiring the cancer cell metabolism. Cancer cells that become dependent on KRAS-driven metabolic adaptations are sensitive to the inhibition of these metabolic routes, revealing novel therapeutic windows of intervention. In general, mutant KRAS fosters tumor growth by shifting cancer cell metabolism toward anabolic pathways. Depending on the tumor, KRAS-driven metabolic rewiring occurs by up-regulating rate-limiting enzymes involved in amino acid, fatty acid, or nucleotide biosynthesis, and by stimulating scavenging pathways such as macropinocytosis and autophagy, which, in turn, provide building blocks to the anabolic routes, also maintaining the energy levels and the cell redox potential (1). This review will discuss the most recent findings on mutant KRAS metabolic reliance in tumor models of pancreatic and non-small-cell lung cancer, also highlighting the role that these metabolic adaptations play in resistance to target therapy. The effects of constitutive KRAS activation in glycolysis elevation, amino acids metabolism reprogramming, fatty acid turnover, and nucleotide biosynthesis will be discussed also in the context of different genetic landscapes.

7.
Nat Commun ; 10(1): 4176, 2019 09 13.
Article in English | MEDLINE | ID: mdl-31519908

ABSTRACT

The centrosome is the master orchestrator of mitotic spindle formation and chromosome segregation in animal cells. Centrosome abnormalities are frequently observed in cancer, but little is known of their origin and about pathways affecting centrosome homeostasis. Here we show that autophagy preserves centrosome organization and stability through selective turnover of centriolar satellite components, a process we termed doryphagy. Autophagy targets the satellite organizer PCM1 by interacting with GABARAPs via a C-terminal LIR motif. Accordingly, autophagy deficiency results in accumulation of large abnormal centriolar satellites and a resultant dysregulation of centrosome composition. These alterations have critical impact on centrosome stability and lead to mitotic centrosome fragmentation and unbalanced chromosome segregation. Our findings identify doryphagy as an important centrosome-regulating pathway and bring mechanistic insights to the link between autophagy dysfunction and chromosomal instability. In addition, we highlight the vital role of centriolar satellites in maintaining centrosome integrity.


Subject(s)
Autophagy/physiology , Centrioles/metabolism , Centrosome/metabolism , Mitosis/physiology , Autophagy/genetics , Cell Cycle/genetics , Cell Cycle/physiology , Cell Line, Tumor , Chromatography, Liquid , Humans , Immunoblotting , Magnetic Resonance Spectroscopy , Mass Spectrometry , Microscopy, Fluorescence , Microtubules/metabolism , Mitosis/genetics , Molecular Dynamics Simulation
8.
Int J Mol Sci ; 19(9)2018 Aug 30.
Article in English | MEDLINE | ID: mdl-30200238

ABSTRACT

RAB5 is a small GTPase that belongs to the wide family of Rab proteins and localizes on early endosomes. In its active GTP-bound form, RAB5 recruits downstream effectors that, in turn, are responsible for distinct aspects of early endosome function, including their movement along microtubules. We previously reported that, at the onset of mitosis, RAB5positive vesicles cluster around the spindle poles and, during metaphase, move along spindle microtubules. RNAi-mediated depletion of the three RAB5 isoforms delays nuclear envelope breakdown at prophase and severely affects chromosome alignment and segregation. Here we show that depletion of the Kinesin-2 motor complex impairs long-range movement of RAB5 endosomes in interphase cells and prevents localization of these vesicles at the spindle during metaphase. Similarly to the effect caused by RAB5 depletion, functional ablation of Kinesin-2 delays nuclear envelope breakdown resulting in prolonged prophase. Altogether these findings suggest that endosomal transport at the onset of mitosis is required to control timing of nuclear envelope breakdown.


Subject(s)
Endosomes/physiology , Kinesins/genetics , Kinesins/metabolism , Spindle Apparatus/metabolism , rab5 GTP-Binding Proteins/metabolism , Cell Line , Humans , Interphase , Metaphase , Microtubules/metabolism , RNA Interference
9.
Cancer Res ; 78(13): 3432-3444, 2018 07 01.
Article in English | MEDLINE | ID: mdl-29691252

ABSTRACT

USP6NL, also named RN-tre, is a GTPase-activating protein involved in control of endocytosis and signal transduction. Here we report that USP6NL is overexpressed in breast cancer, mainly of the basal-like/integrative cluster 10 subtype. Increased USP6NL levels were accompanied by gene amplification and were associated with worse prognosis in the METABRIC dataset, retaining prognostic value in multivariable analysis. High levels of USP6NL in breast cancer cells delayed endocytosis and degradation of the EGFR, causing chronic AKT (protein kinase B) activation. In turn, AKT stabilized the glucose transporter GLUT1 at the plasma membrane, increasing aerobic glycolysis. In agreement, elevated USP6NL sensitized breast cancer cells to glucose deprivation, indicating that their glycolytic capacity relies on this protein. Depletion of USP6NL accelerated EGFR/AKT downregulation and GLUT1 degradation, impairing cell proliferation exclusively in breast cancer cells that harbored increased levels of USP6NL. Overall, these findings argue that USP6NL overexpression generates a metabolic rewiring that is essential to foster the glycolytic demand of breast cancer cells and promote their proliferation.Significance: USP6NL overexpression leads to glycolysis addiction of breast cancer cells and presents a point of metabolic vulnerability for therapeutic targeting in a subset of aggressive basal-like breast tumors.Graphical Abstract: http://cancerres.aacrjournals.org/content/canres/78/13/3432/F1.large.jpg Cancer Res; 78(13); 3432-44. ©2018 AACR.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Breast Neoplasms/pathology , GTPase-Activating Proteins/metabolism , Glucose Transporter Type 1/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Adaptor Proteins, Signal Transducing/genetics , Breast Neoplasms/metabolism , Breast Neoplasms/mortality , Cell Line, Tumor , Cell Proliferation , Datasets as Topic , ErbB Receptors/metabolism , Female , GTPase-Activating Proteins/genetics , Gene Amplification , Glycolysis , Humans , Phosphorylation , Prognosis , Protein Stability , Proteolysis , Survival Analysis
10.
Cell Death Differ ; 25(7): 1259-1275, 2018 07.
Article in English | MEDLINE | ID: mdl-29555978

ABSTRACT

Semaphorin 4C (Sema4C) expression in human breast cancers correlates with poor disease outcome. Surprisingly, upon knock-down of Sema4C or its receptor PlexinB2 in diverse mammary carcinoma cells (but not their normal counterparts), we observed dramatic growth inhibition associated with impairment of G2/M phase transition, cytokinesis defects and the onset of cell senescence. Mechanistically, we demonstrated a Sema4C/PlexinB2/LARG-dependent signaling cascade that is required to maintain critical RhoA-GTP levels in cancer cells. Interestingly, we also found that Sema4C upregulation in luminal-type breast cancer cells drives a dramatic phenotypic change, with disassembly of polarity complexes, mitotic spindle misorientation, cell-cell dissociation and increased migration and invasiveness. We found that this signaling cascade is dependent on the PlexinB2 effectors ErbB2 and RhoA-dependent kinases. Moreover, Sema4C-overexpressing luminal breast cancer cells upregulated the transcription factors Snail, Slug and SOX-2, and formed estrogen-independent metastatic tumors in mice. In sum, our data indicate that Sema4C/PlexinB2 signaling is essential for the growth of breast carcinoma cells, featuring a novel potential therapeutic target. In addition, elevated Sema4C expression enables indolent luminal-type tumors to become resistant to estrogen deprivation, invasive and metastatic in vivo, which could account for its association with a subset of human breast cancers with poor prognosis.


Subject(s)
Breast Neoplasms/metabolism , Estrogens/pharmacology , Nerve Tissue Proteins/metabolism , Receptor, ErbB-2/metabolism , Semaphorins/metabolism , Signal Transduction/drug effects , Animals , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Estrogens/metabolism , Female , Humans , MCF-7 Cells , Mice , Mice, Nude , Mice, SCID , Nerve Tissue Proteins/genetics , Receptor, ErbB-2/genetics , Semaphorins/genetics , Signal Transduction/genetics
11.
Mol Oncol ; 12(1): 3-20, 2018 01.
Article in English | MEDLINE | ID: mdl-29124875

ABSTRACT

The physiological function of the epidermal growth factor receptor (EGFR) is to regulate epithelial tissue development and homeostasis. In pathological settings, mostly in lung and breast cancer and in glioblastoma, the EGFR is a driver of tumorigenesis. Inappropriate activation of the EGFR in cancer mainly results from amplification and point mutations at the genomic locus, but transcriptional upregulation or ligand overproduction due to autocrine/paracrine mechanisms has also been described. Moreover, the EGFR is increasingly recognized as a biomarker of resistance in tumors, as its amplification or secondary mutations have been found to arise under drug pressure. This evidence, in addition to the prominent function that this receptor plays in normal epithelia, has prompted intense investigations into the role of the EGFR both at physiological and at pathological level. Despite the large body of knowledge obtained over the last two decades, previously unrecognized (herein defined as 'noncanonical') functions of the EGFR are currently emerging. Here, we will initially review the canonical ligand-induced EGFR signaling pathway, with particular emphasis to its regulation by endocytosis and subversion in human tumors. We will then focus on the most recent advances in uncovering noncanonical EGFR functions in stress-induced trafficking, autophagy, and energy metabolism, with a perspective on future therapeutic applications.


Subject(s)
Neoplasms/pathology , Animals , Autophagy/physiology , Cell Membrane/metabolism , Endocytosis/physiology , Endosomes/metabolism , ErbB Receptors/genetics , ErbB Receptors/physiology , Humans , Ligands , Mice , Mice, Knockout , Stress, Physiological
12.
Cancer Res ; 77(8): 1813-1817, 2017 04 15.
Article in English | MEDLINE | ID: mdl-28373181

ABSTRACT

Alterations of endo/exocytic proteins have long been associated with malignant transformation, and genes encoding membrane trafficking proteins have been identified as bona fide drivers of tumorigenesis. Focusing on the mechanisms underlying the impact of endo/exocytic proteins in cancer, a scenario emerges in which altered trafficking routes/networks appear to be preferentially involved in the acquisition of prometastatic traits. This involvement in metastasis frequently occurs through the integration of programs leading to migratory/invasive phenotypes, survival and resistance to environmental stresses, epithelial-to-mesenchymal transition, and the emergence of cancer stem cells. These findings might have important implications in the clinical setting for the development of metastasis-specific drugs and for patient stratification to optimize the use of available therapies. Cancer Res; 77(8); 1813-7. ©2017 AACR.


Subject(s)
Neoplasms/metabolism , Neoplasms/pathology , Animals , Carcinogenesis/metabolism , Carcinogenesis/pathology , Cell Membrane/metabolism , Endocytosis/physiology , Exocytosis/physiology , Humans , Neoplasm Metastasis , rab GTP-Binding Proteins/metabolism
13.
Cancer Res ; 76(20): 6095-6106, 2016 10 15.
Article in English | MEDLINE | ID: mdl-27569217

ABSTRACT

Current therapeutic options for the pediatric cancer rhabdomyosarcoma have not improved significantly, especially for metastatic rhabdomyosarcoma. In the current work, we performed a deep miRNA profiling of the three major human rhabdomyosarcoma subtypes, along with cell lines and normal muscle, to identify novel molecular circuits with therapeutic potential. The signature we determined could discriminate rhabdomyosarcoma from muscle, revealing a subset of muscle-enriched miRNA (myomiR), including miR-22, which was strongly underexpressed in tumors. miR-22 was physiologically induced during normal myogenic differentiation and was transcriptionally regulated by MyoD, confirming its identity as a myomiR. Once introduced into rhabdomyosarcoma cells, miR-22 decreased cell proliferation, anchorage-independent growth, invasiveness, and promoted apoptosis. Moreover, restoring miR-22 expression blocked tumor growth and prevented tumor dissemination in vivo Gene expression profiling analysis of miR-22-expressing cells suggested TACC1 and RAB5B as possible direct miR-22 targets. Accordingly, loss- and gain-of-function experiments defined the biological relevance of these genes in rhabdomyosarcoma pathogenesis. Finally, we demonstrated the ability of miR-22 to intercept and overcome the intrinsic resistance to MEK inhibition based on ERBB3 upregulation. Overall, our results identified a novel miR-22 regulatory network with critical therapeutic implications in rhabdomyosarcoma. Cancer Res; 76(20); 6095-106. ©2016 AACR.


Subject(s)
Gene Regulatory Networks , High-Throughput Nucleotide Sequencing , MicroRNAs/physiology , Rhabdomyosarcoma/therapy , Animals , Cell Differentiation , Cell Line, Tumor , Female , Fetal Proteins/genetics , Fetal Proteins/physiology , Gene Expression Regulation, Neoplastic , Humans , Mice , Microtubule-Associated Proteins/genetics , Microtubule-Associated Proteins/physiology , Mitogen-Activated Protein Kinase Kinases/antagonists & inhibitors , MyoD Protein/metabolism , Nuclear Proteins/genetics , Nuclear Proteins/physiology , Promoter Regions, Genetic , Receptor, ErbB-3/genetics , Receptor, ErbB-3/physiology , Rhabdomyosarcoma/etiology , Rhabdomyosarcoma/genetics , Rhabdomyosarcoma/pathology , rab5 GTP-Binding Proteins/genetics , rab5 GTP-Binding Proteins/physiology
14.
Cancer Res ; 76(17): 5019-29, 2016 09 01.
Article in English | MEDLINE | ID: mdl-27364553

ABSTRACT

MET oncogene amplification is emerging as a major mechanism of acquired resistance to EGFR-directed therapy in lung and colorectal cancers. Furthermore, MET amplification predicts responsiveness to MET inhibitors currently in clinical trials. Among the anti-MET drugs available, ATP-competitive small-molecule kinase inhibitors abrogate receptor autophosphorylation and downstream activation of ERK1/2 and AKT, resulting in cell-cycle arrest. However, this antiproliferative effect allows persistence of a pool of cancer cells that are quiescent but alive. Once the inhibition is removed, rebound activation of MET-driven cell proliferative pathways and tumor growth may occur, an adverse event observed frequently in clinical settings after drug discontinuation. Here we show that inhibitor withdrawal prompts receptor phosphorylation to levels higher than those displayed at steady-state and generates a rebound effect pushing quiescent cancer cells back into the cell cycle, both in vitro and in experimental tumor models in vivo Mechanistically, we found that inhibitor treatment blocks MET endocytosis, causing a local increase in the number of receptors at the plasma membrane. Upon inhibitor washout, the receptor is readily rephosphorylated. The initial phosphorylation is not only increased but also prolonged in duration due to downmodulation of a phosphatase-mediated MET-negative feedback loop, which accompanies receptor internalization. Notably, treatment with a MET therapeutic antibody that induces proteolytic cleavage of the receptor at the cell surface substantially prevents this rebound effect, providing a rationale to combine or alternate these mechanistically different types of MET-targeted therapy. Cancer Res; 76(17); 5019-29. ©2016 AACR.


Subject(s)
Antibodies/pharmacology , Antineoplastic Agents/pharmacology , Neoplasm Recurrence, Local/pathology , Neoplasms, Experimental/pathology , Proto-Oncogene Proteins c-met/metabolism , Animals , Blotting, Western , Cell Line, Tumor , Drug Resistance, Neoplasm/drug effects , Female , Fluorescent Antibody Technique , Humans , Mice , Mice, Inbred NOD , Mice, SCID , Microscopy, Confocal , Neoplasm Recurrence, Local/metabolism , Neoplasms, Experimental/metabolism , Protein Kinase Inhibitors/pharmacology , Xenograft Model Antitumor Assays
15.
BMC Biol ; 14: 5, 2016 Jan 19.
Article in English | MEDLINE | ID: mdl-26787475

ABSTRACT

BACKGROUND: Tankyrases are poly(adenosine diphosphate)-ribose polymerases that contribute to biological processes as diverse as modulation of Wnt signaling, telomere maintenance, vesicle trafficking, and microtubule-dependent spindle pole assembly during mitosis. At interphase, polarized reshaping of the microtubule network fosters oriented cell migration. This is attained by association of adenomatous polyposis coli with the plus end of microtubules at the cortex of cell membrane protrusions and microtubule-based centrosome reorientation towards the migrating front. RESULTS: Here we report a new function for tankyrases, namely, regulation of directional cell locomotion. Using a panel of lung cancer cell lines as a model system, we found that abrogation of tankyrase activity by two different, structurally unrelated small-molecule inhibitors (one introduced and characterized here for the first time) or by RNA interference-based genetic silencing weakened cell migration, invasion, and directional movement induced by the motogenic cytokine hepatocyte growth factor. Mechanistically, the anti-invasive outcome of tankyrase inhibition could be ascribed to sequential deterioration of the distinct events that govern cell directional sensing. In particular, tankyrase blockade negatively impacted (1) microtubule dynamic instability; (2) adenomatous polyposis coli plasma membrane targeting; and (3) centrosome reorientation. CONCLUSIONS: Collectively, these findings uncover an unanticipated role for tankyrases in influencing at multiple levels the interphase dynamics of the microtubule network and the subcellular distribution of related polarity signals. These results encourage the further exploration of tankyrase inhibitors as therapeutic tools to oppose dissemination and metastasis of cancer cells.


Subject(s)
Cell Movement/drug effects , Enzyme Inhibitors/pharmacology , Lung Neoplasms/drug therapy , Lung Neoplasms/pathology , Lung/drug effects , Tankyrases/antagonists & inhibitors , Cell Line, Tumor , Enzyme Inhibitors/chemistry , Humans , Lung/metabolism , Lung/pathology , Lung Neoplasms/genetics , Lung Neoplasms/metabolism , Microtubules/metabolism , Microtubules/pathology , Neoplasm Invasiveness/genetics , Neoplasm Invasiveness/pathology , Neoplasm Invasiveness/prevention & control , RNA Interference , Tankyrases/genetics , Tankyrases/metabolism
16.
BMC Cell Biol ; 16: 15, 2015 May 07.
Article in English | MEDLINE | ID: mdl-25947155

ABSTRACT

BACKGROUND: Polycystin-1 (PC-1) is a large plasma membrane receptor, encoded by the PKD1 gene, which is mutated in most cases of Autosomal Dominant Polycystic Kidney Disease (ADPKD). The disease is characterized by renal cysts. The precise function of PC-1 remains elusive, although several studies suggest that it can regulate the cellular shape in response to external stimuli. We and others reported that PC-1 regulates the actin cytoskeleton and cell migration. RESULTS: Here we show that cells over-expressing PC-1 display enhanced adhesion rates to the substrate, while cells lacking PC-1 have a reduced capability to adhere. In search for the mechanism responsible for this new property of PC-1 we found that this receptor is able to regulate the stability of the microtubules, in addition to its capability to regulate the actin cytoskeleton. The two cytoskeletal components are acting in a coordinated fashion. Notably, we uncovered that PC-1 regulation of the microtubule cytoskeleton impacts on the turnover rates of focal adhesions in migrating cells and we link all these properties to the capability of PC-1 to regulate the activation state of Focal Adhesion Kinase (FAK). CONCLUSIONS: In this study we show several new features of the PC-1 receptor in modulating microtubules and adhesion dynamics, which are essential for its capability to regulate migration.


Subject(s)
Actin Cytoskeleton/metabolism , Focal Adhesions/metabolism , Microtubules/metabolism , TRPP Cation Channels/metabolism , Animals , Cell Adhesion , Cell Line , Cell Movement , Dogs , Fluorescence Recovery After Photobleaching , Focal Adhesion Protein-Tyrosine Kinases/metabolism , Madin Darby Canine Kidney Cells , Mice , RNA Interference , RNA, Small Interfering/metabolism , TRPP Cation Channels/antagonists & inhibitors , TRPP Cation Channels/genetics
17.
Mol Oncol ; 9(2): 377-88, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25306394

ABSTRACT

The inflammatory cytokine Tumor Necrosis Factor Alpha (TNF-α) is known to trigger invasive growth, a physiological property for tissue healing, turning into a hallmark of progression in cancer. However, the invasive response to TNF-α relies on poorly understood molecular mechanisms. We thus investigated whether it involves the MET oncogene, which regulates the invasive growth program by encoding the tyrosine kinase receptor for Hepatocyte Growth Factor (HGF). Here we show that the TNF-α pro-invasive activity requires MET function, as it is fully inhibited by MET-specific inhibitors (small-molecules, antibodies, and siRNAs). Mechanistically, we show that TNF-α induces MET transcription via NF-κB, and exploits MET to sustain MEK/ERK activation and Snail accumulation, leading to E-cadherin downregulation. We then show that TNF-α not only induces MET expression in cancer cells, but also HGF secretion by fibroblasts. Consistently, we found that, in human colorectal cancer tissues, high levels of TNF-α correlates with increased expression of both MET and HGF. These findings suggest that TNF-α fosters a HGF/MET pro-invasive paracrine loop in tumors. Targeting this ligand/receptor pair would contribute to prevent cancer progression associated with inflammation.


Subject(s)
Colorectal Neoplasms/metabolism , Gene Expression Regulation, Neoplastic , MAP Kinase Signaling System , Proto-Oncogene Proteins c-met/biosynthesis , Tumor Necrosis Factor-alpha/metabolism , Cell Line, Tumor , Colorectal Neoplasms/genetics , Hepatocyte Growth Factor/genetics , Hepatocyte Growth Factor/metabolism , Humans , Neoplasm Invasiveness/genetics , Paracrine Communication/genetics , Proto-Oncogene Proteins c-met/genetics , Snail Family Transcription Factors , Transcription Factors/genetics , Transcription Factors/metabolism , Tumor Necrosis Factor-alpha/genetics
18.
Dev Cell ; 30(5): 553-68, 2014 Sep 08.
Article in English | MEDLINE | ID: mdl-25203208

ABSTRACT

The role of endocytic proteins and the molecular mechanisms underlying epithelial cell cohesion and tumor dissemination are not well understood. Here, we report that the endocytic F-BAR-containing CDC42-interacting protein 4 (CIP4) is required for ERBB2- and TGF-ß1-induced cell scattering, breast cancer (BC) cell motility and invasion into 3D matrices, and conversion from ductal breast carcinoma in situ to invasive carcinoma in mouse xenograft models. CIP4 promotes the formation of an E-cadherin-CIP4-SRC complex that controls SRC activation, E-cadherin endocytosis, and localized phosphorylation of the myosin light chain kinase, thereby impinging on the actomyosin contractility required to generate tangential forces to break cell-cell junctions. CIP4 is upregulated in ERBB2-positive human BC, correlates with increased distant metastasis, and is an independent predictor of poor disease outcome in subsets of BC patients. Thus, it critically controls cell-cell cohesion and is required for the acquisition of an invasive phenotype in breast tumors.


Subject(s)
Epithelial Cells/cytology , Gene Expression Regulation, Neoplastic , Microtubule-Associated Proteins/metabolism , Triple Negative Breast Neoplasms/metabolism , Actomyosin/metabolism , Animals , Cadherins/metabolism , Carcinoma, Ductal, Breast/pathology , Carcinoma, Intraductal, Noninfiltrating/pathology , Cell Line, Tumor , Endocytosis , Female , Humans , Mammary Neoplasms, Experimental/metabolism , Mice , Minor Histocompatibility Antigens , Neoplasm Transplantation , Receptor, ErbB-2/metabolism , Transforming Growth Factor beta1/metabolism
19.
Curr Biol ; 23(23): 2355-64, 2013 Dec 02.
Article in English | MEDLINE | ID: mdl-24239119

ABSTRACT

BACKGROUND: Integrin-mediated adhesion of cells to the extracellular matrix (ECM) relies on the dynamic formation of focal adhesions (FAs), which are biochemical and mechanosensitive platforms composed of a large variety of cytosolic and transmembrane proteins. During migration, there is a constant turnover of ECM contacts that initially form as nascent adhesions at the leading edge, mature into FAs as actomyosin tension builds up, and are then disassembled at the cell rear, thus allowing for cell detachment. Although the mechanisms of FA assembly have largely been defined, the molecular circuitry that regulates their disassembly still remains elusive. RESULTS: Here, we show that RN-tre, a GTPase-activating protein (GAP) for Rabs including Rab5 and Rab43, is a novel regulator of FA dynamics and cell migration. RN-tre localizes to FAs and to a pool of Rab5-positive vesicles mainly associated with FAs undergoing rapid remodeling. We found that RN-tre inhibits endocytosis of ß1, but not ß3, integrins and delays the turnover of FAs, ultimately impairing ß1-dependent, but not ß3-dependent, chemotactic cell migration. All of these effects are mediated by its GAP activity and rely on Rab5. CONCLUSIONS: Our findings identify RN-tre as the Rab5-GAP that spatiotemporally controls FA remodeling during chemotactic cell migration.


Subject(s)
Cell Movement/genetics , Endocytosis/genetics , GTPase-Activating Proteins/metabolism , rab5 GTP-Binding Proteins/genetics , Adaptor Proteins, Signal Transducing/metabolism , Animals , Cell Adhesion/physiology , Cells, Cultured , Doxycycline/pharmacology , Endocytosis/physiology , Focal Adhesions/metabolism , GTPase-Activating Proteins/genetics , Integrin beta1/metabolism , Integrin beta3/metabolism , Mice , Mice, Knockout , RNA Interference , RNA, Small Interfering , Signal Transduction/physiology , rab GTP-Binding Proteins/biosynthesis , rab GTP-Binding Proteins/genetics , rab5 GTP-Binding Proteins/biosynthesis
20.
Cancer Res ; 72(22): 5801-11, 2012 Nov 15.
Article in English | MEDLINE | ID: mdl-22986738

ABSTRACT

Neuropilin-1 (NRP1) is a coreceptor for multiple extracellular ligands. NRP1 is widely expressed in cancer cells and in advanced human tumors; however, its functional relevance and signaling mechanisms are unclear. Here, we show that NRP1 expression controls viability and proliferation of different cancer cells, independent of its short intracellular tail. We found that the extracellular domain of NRP1 interacts with the EGF receptor (EGFR) and promotes its signaling cascade elicited upon EGF or TGF-α stimulation. Upon NRP1 silencing, the ability of ligand-bound EGFR to cluster on the cell surface, internalize, and activate the downstream AKT pathway is severely impaired. EGFR is frequently activated in human tumors due to overexpression, mutation, or sustained autocrine/paracrine stimulation. Here we show that NRP1-blocking antibodies and NRP1 silencing can counteract ligand-induced EGFR activation in cancer cells. Thus our findings unveil a novel molecular mechanism by which NRP1 can control EGFR signaling and tumor growth.


Subject(s)
ErbB Receptors/metabolism , Neuropilin-1/metabolism , Animals , Cell Growth Processes/physiology , Cell Line, Tumor , Cell Survival/physiology , Endocytosis , Gene Knockdown Techniques , Humans , Mice , Neuropilin-1/deficiency , Neuropilin-1/genetics , RNA, Small Interfering/administration & dosage , RNA, Small Interfering/genetics , Signal Transduction , Transfection , Transplantation, Heterologous
SELECTION OF CITATIONS
SEARCH DETAIL
...