Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
Add more filters











Publication year range
1.
Mol Cancer ; 23(1): 105, 2024 May 16.
Article in English | MEDLINE | ID: mdl-38755661

ABSTRACT

BACKGROUND: The main drawback of BRAF/MEK inhibitors (BRAF/MEKi)-based targeted therapy in the management of BRAF-mutated cutaneous metastatic melanoma (MM) is the development of therapeutic resistance. We aimed to assess in this context the role of mTORC2, a signaling complex defined by the presence of the essential RICTOR subunit, regarded as an oncogenic driver in several tumor types, including MM. METHODS: After analyzing The Cancer Genome Atlas MM patients' database to explore both overall survival and molecular signatures as a function of intra-tumor RICTOR levels, we investigated the effects of RICTOR downregulation in BRAFV600E MM cell lines on their response to BRAF/MEKi. We performed proteomic screening to identify proteins modulated by changes in RICTOR expression, and Seahorse analysis to evaluate the effects of RICTOR depletion on mitochondrial respiration. The combination of BRAFi with drugs targeting proteins and processes emerged in the proteomic screening was carried out on RICTOR-deficient cells in vitro and in a xenograft setting in vivo. RESULTS: Low RICTOR levels in BRAF-mutated MM correlate with a worse clinical outcome. Gene Set Enrichment Analysis of low-RICTOR tumors display gene signatures suggestive of activation of the mitochondrial Electron Transport Chain (ETC) energy production. RICTOR-deficient BRAFV600E cells are intrinsically tolerant to BRAF/MEKi and anticipate the onset of resistance to BRAFi upon prolonged drug exposure. Moreover, in drug-naïve cells we observed a decline in RICTOR expression shortly after BRAFi exposure. In RICTOR-depleted cells, both mitochondrial respiration and expression of nicotinamide phosphoribosyltransferase (NAMPT) are enhanced, and their pharmacological inhibition restores sensitivity to BRAFi. CONCLUSIONS: Our work unveils an unforeseen tumor-suppressing role for mTORC2 in the early adaptation phase of BRAFV600E melanoma cells to targeted therapy and identifies the NAMPT-ETC axis as a potential therapeutic vulnerability of low RICTOR tumors. Importantly, our findings indicate that the evaluation of intra-tumor RICTOR levels has a prognostic value in metastatic melanoma and may help to guide therapeutic strategies in a personalized manner.


Subject(s)
Drug Resistance, Neoplasm , Mechanistic Target of Rapamycin Complex 2 , Melanoma , Protein Kinase Inhibitors , Proto-Oncogene Proteins B-raf , Rapamycin-Insensitive Companion of mTOR Protein , Animals , Humans , Mice , Cell Line, Tumor , Down-Regulation , Drug Resistance, Neoplasm/genetics , Gene Expression Regulation, Neoplastic , Mechanistic Target of Rapamycin Complex 2/metabolism , Mechanistic Target of Rapamycin Complex 2/genetics , Melanoma/genetics , Melanoma/drug therapy , Melanoma/metabolism , Melanoma/pathology , Mutation , Protein Kinase Inhibitors/pharmacology , Protein Kinase Inhibitors/therapeutic use , Proteomics/methods , Proto-Oncogene Proteins B-raf/antagonists & inhibitors , Proto-Oncogene Proteins B-raf/genetics , Rapamycin-Insensitive Companion of mTOR Protein/metabolism , Rapamycin-Insensitive Companion of mTOR Protein/genetics , Xenograft Model Antitumor Assays , MAP Kinase Kinase Kinases/antagonists & inhibitors
2.
Cell Commun Signal ; 16(1): 73, 2018 11 03.
Article in English | MEDLINE | ID: mdl-30390666

ABSTRACT

BACKGROUND: p130 Crk-associated substrate (p130CAS; also known as BCAR1) is a scaffold protein that modulates many essential cellular processes such as cell adhesion, proliferation, survival, cell migration, and intracellular signaling. p130Cas has been shown to be highly expressed in a variety of human cancers of epithelial origin. However, few data are available regarding the role of p130Cas during normal epithelial development and homeostasis. METHODS: To this end, we have generated a genetically modified mouse in which p130Cas protein was specifically ablated in the epidermal tissue. RESULTS: By using this murine model, we show that p130Cas loss results in increased cell proliferation and reduction of cell adhesion to extracellular matrix. In addition, epidermal deletion of p130Cas protein leads to premature expression of "late" epidermal differentiation markers, altered membrane E-cadherin/catenin proteins localization and aberrant tyrosine phosphorylation of E-cadherin/catenin complexes. Interestingly, these alterations in adhesive properties in absence of p130Cas correlate with abnormalities in progenitor cells balance resulting in the amplification of a more committed cell population. CONCLUSION: Altogether, these results provide evidence that p130Cas is an important regulator of epidermal cell fate and homeostasis.


Subject(s)
Cell Adhesion , Cell Differentiation , Crk-Associated Substrate Protein/deficiency , Crk-Associated Substrate Protein/genetics , Epidermis/metabolism , Gene Deletion , Homeostasis/genetics , Animals , Cell Proliferation , Extracellular Matrix/metabolism , Keratinocytes/cytology , Mice , Mice, Inbred C57BL , Phenotype
3.
Cell Commun Signal ; 16(1): 90, 2018 11 26.
Article in English | MEDLINE | ID: mdl-30477510

ABSTRACT

Following publication of the original article [1], the authors reported an error in the name of the 11th author. The author's name was incorrectly published as "Vincenzo Calautti", instead of "Enzo Calautti".

4.
Cell Death Differ ; 24(4): 731-746, 2017 04.
Article in English | MEDLINE | ID: mdl-28211872

ABSTRACT

How metabolic pathways required for epidermal tissue growth and remodeling influence the ability of keratinocytes to survive stressful conditions is still largely unknown. The mechanistic target of rapamycin complex 2 (mTORC2) regulates growth and metabolism of several tissues, but its functions in epidermal cells are poorly defined. Rictor is an adaptor protein essential for mTORC2 activity. To explore the roles of mTORC2 in the epidermis, we have conditionally deleted rictor in mice via K14-Cre-mediated homologous recombination and found that its deficiency causes moderate tissue hypoplasia, reduced keratinocyte proliferation and attenuated hyperplastic response to TPA. Noteworthy, rictor-deficient keratinocytes displayed increased lifespan, protection from senescence, and enhanced tolerance to cellular stressors such as growth factors deprivation, epirubicin and X-ray in vitro and radioresistance in vivo. Rictor-deficient keratinocytes exhibited changes in global gene expression profiles consistent with metabolic alterations and enhanced stress tolerance, a shift in cell catabolic processes from glycids and lipids to glutamine consumption and increased production of mitochondrial reactive oxygen species (ROS). Mechanistically, the resiliency of rictor-deficient epidermal cells relies on these ROS increases, indicating stress resistance via mitohormesis. Thus, our findings reveal a new link between metabolic changes and stress adaptation of keratinocytes centered on mTORC2 activity, with potential implications in skin aging and therapeutic resistance of epithelial tumors.


Subject(s)
Mitochondria/metabolism , Rapamycin-Insensitive Companion of mTOR Protein/genetics , Acetylcysteine/pharmacology , Animals , Apoptosis/drug effects , Cell Proliferation/drug effects , Cells, Cultured , Cellular Senescence/drug effects , Cellular Senescence/radiation effects , Epirubicin/toxicity , Glutamic Acid/metabolism , Hyperplasia , Keratin-14/genetics , Keratinocytes/cytology , Keratinocytes/drug effects , Keratinocytes/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Mitochondria/drug effects , Radiation Tolerance/radiation effects , Rapamycin-Insensitive Companion of mTOR Protein/antagonists & inhibitors , Rapamycin-Insensitive Companion of mTOR Protein/deficiency , Reactive Oxygen Species/metabolism , Skin/drug effects , Skin/metabolism , Skin/pathology , Tetradecanoylphorbol Acetate/analogs & derivatives , Tetradecanoylphorbol Acetate/pharmacology , Transcriptome/drug effects , Transcriptome/radiation effects , X-Rays
5.
Exp Dermatol ; 24(1): 29-34, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25382846

ABSTRACT

STAT3, a pleiotropic transcription factor acting downstream of cytokines and growth factors, is known to enhance proliferation, migration, invasion and aerobic glycolysis in tumors upon aberrant activation. In the murine epidermis, STAT3 is necessary for experimentally induced carcinogenesis. Skin tumorigenesis is conversely enhanced by overexpression in keratinocytes of the constitutively active STAT3C mutant, which also induces robust, psoriasis-like epidermal hyperplasia. We show here that STAT3C expression at physiological levels in knock-in mice leads to mild epidermal hyperplasia and attenuated expression of terminal differentiation markers. Altered differentiation is confirmed in isolated primary epidermal keratinocytes in vitro, correlating with enhanced proliferative and clonogenic potential, attenuated senescence and, strikingly, high-frequency spontaneous immortalization. These results suggest that moderate levels of continuous STAT3 activation, which closely resemble those triggered by chronic inflammation or persistent growth factor stimulation, may establish a preneoplastic state in part by promoting the escape of epidermal progenitor cells from differentiation and senescence checkpoints.


Subject(s)
Cell Differentiation , Cellular Senescence , Epidermal Cells , Keratinocytes/metabolism , STAT3 Transcription Factor/metabolism , Animals , Animals, Newborn , Cell Movement , Cell Proliferation , Glycolysis , Hyperplasia/metabolism , Keratinocytes/cytology , Mice , Mice, Transgenic , Real-Time Polymerase Chain Reaction , Skin/metabolism , Skin Aging , Stem Cells/cytology , beta-Galactosidase/metabolism
6.
Stem Cells ; 32(3): 754-69, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24123662

ABSTRACT

Signals downstream of Akt can either favor or oppose stem cell (SC) maintenance, but how this dual role can be achieved is still undefined. Using human limbal keratinocyte stem cells (LKSCs), a SC type used in transplantation therapies for corneal regeneration, we show that Akt signaling is prominent in SC populations both in vivo and in vitro, and that Akt1 promotes while Akt2 opposes SC self-renewal. Noteworthy, loss of Akt2 signaling enhances LKSC maintenance ex vivo, whereas Akt1 depletion anticipates SC exhaustion. Mechanistically, the antagonistic functions of Akt1 and Akt2 in SC control are mainly dictated by their differential subcellular distribution, being nuclear Akt2 selectively implicated in FOXO inhibition. Akt2 downregulation favors LKSC maintenance as a result of a gain of FOXO functions, which attenuates the mechanistic target of rapamycin complex one signaling via tuberous sclerosis one gene induction, and promotes growth factor signaling through Akt1. Consistently, Akt2 deficiency also enhances limbal SCs in vivo. Thus, our findings reveal distinct roles for nuclear versus cytosolic Akt signaling in normal epithelial SC control and suggest that the selective Akt2 inhibition may provide novel pharmacological strategies for human LKSC expansion in therapeutic settings and mechanistic research.


Subject(s)
Cell Nucleus/enzymology , Forkhead Transcription Factors/metabolism , Keratinocytes/cytology , Multiprotein Complexes/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Stem Cells/cytology , TOR Serine-Threonine Kinases/metabolism , 3T3 Cells , Adult , Animals , Cell Proliferation , Clone Cells , Enzyme Activation , Forkhead Box Protein O1 , Forkhead Box Protein O3 , Humans , Isoenzymes/metabolism , Limbus Corneae/cytology , Mechanistic Target of Rapamycin Complex 1 , Mice , Phenotype , Phosphorylation , Proto-Oncogene Proteins c-akt/deficiency , Repressor Proteins/metabolism , Signal Transduction , Stem Cells/enzymology , Transcription, Genetic
7.
J Biol Chem ; 280(38): 32856-65, 2005 Sep 23.
Article in English | MEDLINE | ID: mdl-16036919

ABSTRACT

Signaling pathways regulating the differentiation program of epidermal cells overlap widely with those activated during apoptosis. How differentiating cells remain protected from premature death, however, is still poorly defined. We show here that the phosphoinositide 3-kinase (PI3K)/Akt pathway is activated at early stages of mouse keratinocyte differentiation both in culture and in the intact epidermis in vivo. Expression of active Akt in keratinocytes promotes growth arrest and differentiation, whereas pharmacological blockade of PI3K inhibits the expression of "late" differentiation markers and leads to death of cells that would otherwise differentiate. Mechanistically, the activation of the PI3K/Akt pathway in keratinocyte differentiation depends on the activity of the epidermal growth factor receptor and Src families of tyrosine kinases and the engagement of E-cadherin-mediated adhesion. During this process, PI3K associates increasingly with cadherin-catenin protein complexes bearing tyrosine phosphorylated YXXM motifs. Thus, the PI3K signaling pathway regulates the choice between epidermal cell differentiation and death at the cross-talk between tyrosine kinases and cadherin-associated catenins.


Subject(s)
Keratinocytes/cytology , Phosphatidylinositol 3-Kinases/metabolism , Amino Acid Motifs , Animals , Apoptosis , Bromodeoxyuridine/pharmacology , Cadherins/chemistry , Cadherins/metabolism , Calcium/metabolism , Cell Adhesion , Cell Differentiation , Cell Proliferation , Cells, Cultured , Enzyme Activation , Epidermal Cells , ErbB Receptors/metabolism , Fluorescent Dyes/pharmacology , Green Fluorescent Proteins/metabolism , Immunoprecipitation , Keratinocytes/metabolism , Mice , Mice, Inbred C57BL , Microscopy, Fluorescence , Mitogen-Activated Protein Kinase 3/metabolism , Models, Biological , Phosphorylation , Protein-Tyrosine Kinases/chemistry , Protein-Tyrosine Kinases/metabolism , Signal Transduction , Time Factors , Tyrosine/chemistry , src-Family Kinases/metabolism
8.
J Biol Chem ; 279(45): 47172-6, 2004 Nov 05.
Article in English | MEDLINE | ID: mdl-15371457

ABSTRACT

Syndecan-4 is a transmembrane heparan sulfate proteoglycan that co-operates with integrins during cell-matrix interactions for the assembly of focal adhesions and actin stress fibers and in the phosphorylation of focal adhesion kinase (FAK) on Tyr397. These cellular events are regulated by the small GTPase Rho, and in the absence of syndecan-4 ligation, cellular levels of GTP-bound Rho are decreased implicating syndecan-4 in the regulation of the small GTPases. In the present study we report that, compared with wild type cells, fibronectin-adherent syndecan-4-null fibroblasts showed enhanced lamellipodia and increased Rac1 activity that could be down-regulated by re-expression of syndecan-4 in the mutant cells. Consistent with the role for Rac1 in activating p38 and JNK signaling, syndecan-4-null cells display higher levels of active p38 MAPK and JNK that were abolished by the expression of a dominant-negative RacN17 mutant. Since p38 and JNK regulate gene expression by phosphorylating and activating transcription factors, we compared both the phosphorylation state and the transcriptional activity of the ATF-2 transcription factor, as a direct p38 and JNK target in syndecan-4-null and wild type cells. In the absence of syndecan-4, both ATF-2 phosphorylation and transcriptional activity were significantly more elevated compared with wild type cells, and both activities were decreased either by the re-expression of syndecan-4 or by the expression of RacN17. Our results reveal a novel function for syndecan-4 in modulating nuclear transcriptional activity and indicate an underlying mechanism that acts at the level of Rac1-p38/JNK signaling.


Subject(s)
Cyclic AMP Response Element-Binding Protein/metabolism , Gene Expression Regulation , Membrane Glycoproteins/physiology , Proteoglycans/physiology , Transcription Factors/metabolism , rac1 GTP-Binding Protein/metabolism , Activating Transcription Factor 2 , Adenoviridae/genetics , Animals , Cell Nucleus/metabolism , DNA/metabolism , Fibroblasts/metabolism , Fibronectins/metabolism , Focal Adhesions , Green Fluorescent Proteins/metabolism , Humans , JNK Mitogen-Activated Protein Kinases/metabolism , Luciferases/metabolism , MAP Kinase Kinase 4 , Membrane Glycoproteins/metabolism , Mice , Mitogen-Activated Protein Kinase Kinases/metabolism , Phosphorylation , Proteoglycans/metabolism , Signal Transduction , Syndecan-4 , Transcription, Genetic , Transcriptional Activation , Tyrosine/chemistry , p38 Mitogen-Activated Protein Kinases/metabolism
9.
J Biol Chem ; 277(14): 12270-4, 2002 Apr 05.
Article in English | MEDLINE | ID: mdl-11805099

ABSTRACT

Syndecan-4 and integrins are the primary transmembrane receptors of focal adhesions in cells adherent to extracellular matrix molecules. Syndesmos is a cytoplasmic protein that interacts specifically with the cytoplasmic domain of syndecan-4, and it co-localizes with syndecan-4 in focal contacts. In the present study we sought possible interactors with syndesmos. We find that syndesmos interacts with the focal adhesion adaptor protein paxillin. The binding of syndesmos to paxillin is direct, and these interactions are triggered by the activation of protein kinase C. Syndesmos also binds the paxillin homolog, Hic-5. The connection of syndecan-4 with paxillin through syndesmos parallels the connection between paxillin and integrins and may thus reflect the cooperative signaling of these two receptors in the assembly of focal adhesions and actin stress fibers.


Subject(s)
Carrier Proteins/chemistry , Cytoskeletal Proteins/chemistry , DNA-Binding Proteins/chemistry , Phosphoproteins/chemistry , Actins/metabolism , Amino Acid Sequence , Animals , Cell Adhesion , Cells, Cultured , Chick Embryo , Cytoskeletal Proteins/metabolism , DNA-Binding Proteins/metabolism , Glutathione Transferase/metabolism , Intracellular Signaling Peptides and Proteins , LIM Domain Proteins , Models, Biological , Molecular Sequence Data , Paxillin , Phosphoproteins/metabolism , Precipitin Tests , Protein Binding , Protein Biosynthesis , Protein Kinase C/metabolism , Protein Structure, Tertiary , Recombinant Fusion Proteins/metabolism , Sequence Homology, Amino Acid , Signal Transduction , Transcription, Genetic
10.
Glycoconj J ; 19(4-5): 305-13, 2002.
Article in English | MEDLINE | ID: mdl-12975609

ABSTRACT

Studies examining the role of the cell-surface heparan sulfate proteoglycan syndecan-4 have yielded a plethora of information regarding its role in both cell-matrix and growth-factor mediated signaling events. Many of the initial conclusions drawn from such research placed syndecan-4 in a keystone position within various signaling pathways though the generation of syndecan-4 null mice have surprised many in the field by indicating otherwise. These contradictory results place researchers in the frustrating and yet exhilarating position of still asking the question, "What role does syndecan-4 play in life?"


Subject(s)
Membrane Glycoproteins/physiology , Proteoglycans/physiology , Animals , Cell Movement , Focal Adhesions , Gene Expression , Growth Substances/physiology , Heparan Sulfate Proteoglycans/physiology , Membrane Glycoproteins/deficiency , Membrane Glycoproteins/genetics , Mice , Mice, Knockout , Protein Kinase C/physiology , Proteoglycans/deficiency , Proteoglycans/genetics , Signal Transduction , Syndecan-4
SELECTION OF CITATIONS
SEARCH DETAIL