Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
Add more filters










Publication year range
1.
Cancers (Basel) ; 15(3)2023 Jan 30.
Article in English | MEDLINE | ID: mdl-36765819

ABSTRACT

Over the last 30 years, the study of the cellular response to ionizing radiation (IR) has increased exponentially. Among the various signaling pathways affected by IR, p38 MAPK has been shown to be activated both in vitro and in vivo, with involvement in key processes triggered by IR-mediated genotoxic insult, such as the cell cycle, apoptosis or senescence. However, we do not yet have a definitive clue about the role of p38 MAPK in terms of radioresistance/sensitivity and its potential use to improve current radiotherapy. In this review, we summarize the current knowledge on this family of MAPKs in response to IR as well as in different aspects related to radiotherapy, such as their role in the control of REDOX, fibrosis, and in the radiosensitizing effect of several compounds.

2.
Cancers (Basel) ; 14(14)2022 Jul 19.
Article in English | MEDLINE | ID: mdl-35884568

ABSTRACT

Sarcomas are a heterogeneous group of tumors in which the role of ERK5 is poorly studied. To clarify the role of this MAPK in sarcomatous pathology, we used a murine 3-methyl-cholanthrene (3MC)-induced sarcoma model. Our data show that 3MC induces pleomorphic sarcomas with muscle differentiation, showing an increased expression of ERK5. Indeed, this upregulation was also observed in human sarcomas of muscular origin, such as leiomyosarcoma or rhabdomyosarcoma. Moreover, in cell lines derived from these 3MC-induced tumors, abrogation of Mapk7 expression by using specific shRNAs decreased in vitro growth and colony-forming capacity and led to a marked loss of tumor growth in vivo. In fact, transcriptomic profiling in ERK5 abrogated cell lines by RNAseq showed a deregulated gene expression pattern for key biological processes such as angiogenesis, migration, motility, etc., correlating with a better prognostic in human pathology. Finally, among the various differentially expressed genes, Klf2 is a key mediator of the biological effects of ERK5 as indicated by its specific interference, demonstrating that the ERK5-KLF2 axis is an important determinant of sarcoma biology that should be further studied in human pathology.

3.
Int J Mol Sci ; 21(20)2020 Oct 12.
Article in English | MEDLINE | ID: mdl-33053909

ABSTRACT

The p38 mitogen-activated protein kinase (MAPK) signaling pathway is implicated in cancer biology and has been widely studied over the past two decades as a potential therapeutic target. Most of the biological and pathological implications of p38MAPK signaling are often associated with p38α (MAPK14). Recently, several members of the p38 family, including p38γ and p38δ, have been shown to play a crucial role in several pathologies including cancer. However, the specific role of p38ß (MAPK11) in cancer is still elusive, and further investigation is needed. Here, we summarize what is currently known about the role of p38ß in different types of tumors and its putative implication in cancer therapy. All evidence suggests that p38ß might be a key player in cancer development, and could be an important therapeutic target in several pathologies, including cancer.


Subject(s)
Disease Susceptibility , Mitogen-Activated Protein Kinase 11/metabolism , Neoplasms/etiology , Neoplasms/metabolism , Animals , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/metabolism , Gene Expression Regulation, Neoplastic , Humans , Mitogen-Activated Protein Kinase 11/genetics , Multigene Family , Neoplasms/pathology , Signal Transduction
4.
J Immunol ; 197(8): 3371-3381, 2016 10 15.
Article in English | MEDLINE | ID: mdl-27574297

ABSTRACT

The involvement of NOTCH signaling in macrophage activation by Toll receptors has been clearly established, but the factors and pathways controlling NOTCH signaling during this process have not been completely delineated yet. We have characterized the role of TSPAN33, a tetraspanin implicated in a disintegrin and metalloproteinase (ADAM) 10 maturation, during macrophage proinflammatory activation. Tspan33 expression increases in response to TLR signaling, including responses triggered by TLR4, TLR3, and TLR2 activation, and it is enhanced by IFN-γ. In this study, we report that induction of Tspan33 expression by TLR and IFN-γ is largely dependent on NOTCH signaling, as its expression is clearly diminished in macrophages lacking Notch1 and Notch2 expression, but it is enhanced after overexpression of a constitutively active intracellular domain of NOTCH1. TSPAN33 is the member of the TspanC8 tetraspanin subgroup more intensely induced during macrophage activation, and its overexpression increases ADAM10, but not ADAM17, maturation. TSPAN33 favors NOTCH processing at the membrane by modulating ADAM10 and/or Presenilin1 activity, thus increasing NOTCH signaling in activated macrophages. Moreover, TSPAN33 modulates TLR-induced proinflammatory gene expression, at least in part, by increasing NF-κB-dependent transcriptional activity. Our results suggest that TSPAN33 represents a new control element in the development of inflammation by macrophages that could constitute a potential therapeutic target.


Subject(s)
Macrophage Activation , Macrophages/metabolism , Receptors, Notch/metabolism , Signal Transduction , Tetraspanins/metabolism , Toll-Like Receptors/metabolism , Animals , Humans , Inflammation/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , RAW 264.7 Cells , Tetraspanins/genetics , U937 Cells
5.
Oncotarget ; 6(17): 15551-65, 2015 Jun 20.
Article in English | MEDLINE | ID: mdl-26036632

ABSTRACT

Resistance to cisplatin is a major challenge in the current cancer therapy. In order to explore new therapeutic strategies to cisplatin resistance, we evaluated, in a model of lung cancer (H1299 and H460 cell lines), the nature of the pathways leading to cell death. We observed that H1299 displayed a natural resistance to cisplatin due to an inability to trigger an apoptotic response that correlates with the induction of autophagy. However, pharmacological and genetic approaches showed how autophagy was a mechanism associated to cell death rather than to resistance. Indeed, pro-autophagic stimuli such as mTOR or Akt inhibition mediate cell death in both cell lines to a similar extent. We next evaluated the response to a novel platinum compound, monoplatin, able to promote cell death in an exclusive autophagy-dependent manner. In this case, no differences were observed between both cell lines. Furthermore, in response to monoplatin, two molecular hallmarks of cisplatin response (p53 and MAPKs) were not implicated, indicating the ability of this pro-autophagic compound to overcome cisplatin resistance. In summary, our data highlight how induction of autophagy could be used in cisplatin resistant tumours and an alternative treatment for p53 mutated patient in a synthetic lethally approach.


Subject(s)
Autophagy/drug effects , Cisplatin/pharmacology , Lung Neoplasms/drug therapy , Platinum Compounds/pharmacology , Proto-Oncogene Proteins c-akt/metabolism , Tumor Suppressor Protein p53/metabolism , Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Autophagy-Related Protein 5 , Cell Line, Tumor , Drug Resistance, Neoplasm , HEK293 Cells , Humans , Microtubule-Associated Proteins/genetics , RNA Interference , RNA, Small Interfering , TOR Serine-Threonine Kinases/metabolism
6.
Cardiovasc Res ; 93(2): 232-41, 2012 Feb 01.
Article in English | MEDLINE | ID: mdl-22068159

ABSTRACT

AIMS: The epidermal growth factor-like protein Delta-like 1 (DLK1) regulates multiple differentiation processes. It resembles NOTCH ligands structurally and is considered a non-canonical ligand. Given the crucial role of the NOTCH pathway in angiogenesis, we hypothesized that DLK1 could regulate angiogenesis by interfering with NOTCH. We therefore investigated the expression and function of DLK1 in the vascular endothelium and its role in the regulation of angiogenesis. METHODS AND RESULTS: We report DLK1 expression in the endothelium of different species, including human, cow, pig, and mouse. Angiogenesis was studied by using in vitro and in vivo models of angiotube formation in endothelial cells, retinal phenotypes in Dlk1-null mice, and vessel development in zebrafish. DLK1 overexpression strongly inhibited angiotube formation, whereas lung endothelial cells from Dlk1-null mice were highly angiogenic. In vivo studies demonstrated DLK1-mediated inhibition of neovessel formation and revealed an altered pattern of angiogenesis in the retinas of Dlk1-null mice. The expression of human DLK1 in zebrafish embryos severely altered the formation of intersegmental vessels, while knockdown of the orthologous gene was associated with ectopic and increased tumour-induced angiogenesis. NOTCH-dependent signalling as determined by gene expression reporters was inhibited by the presence of DLK1 in vascular endothelial cells. In contrast, Dlk1-null mice showed increased levels of NOTCH downstream targets, such as Snail and Slug. CONCLUSION: Our results unveil a novel inhibitory role for DLK1 in the regulation of angiogenesis, mediated by antagonism of the NOTCH pathway, and establish the basis for investigating its action in pathological settings.


Subject(s)
Intercellular Signaling Peptides and Proteins/physiology , Neovascularization, Physiologic , Animals , Calcium-Binding Proteins , Cattle , Cells, Cultured , Endothelial Cells/metabolism , Mice , Neovascularization, Pathologic/etiology , Receptors, Notch/antagonists & inhibitors , Retinal Neovascularization/etiology , Signal Transduction , Wound Healing , Zebrafish
7.
Biochim Biophys Acta ; 1813(6): 1153-64, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21419176

ABSTRACT

The protein DLK2, highly homologous to DLK1, belongs to the EGF-like family of membrane proteins, which includes NOTCH receptors and their DSL-ligands. The molecular mechanisms by which DLK proteins regulate cell differentiation and proliferation processes are not fully established yet. In previous reports, we demonstrated that DLK1 interacts with itself and with specific EGF-like repeats of the NOTCH1 extracellular region involved in the binding to NOTCH1 canonical ligands. Moreover, the interaction of DLK1 with NOTCH1 caused an inhibition of basal NOTCH signaling in preadipocytes and mesenchymal multipotent cells. In this work, we demonstrate, for the first time, that DLK2 interacts with itself, with DLK1, and with the same NOTCH1 receptor region as DLK1 does. We demonstrate also that the interaction of DLK2 with NOTCH1 similarly results in an inhibition of NOTCH signaling in preadipocytes and Mouse Embryo fibloblasts. In addition, we demonstrate that a membrane DLK1 variant, lacking the sequence recognized by the protease TACE, also inhibits NOTCH signaling. Furthermore, both DLK1 and DLK2 are able to decrease NOTCH activity also when triggered by specific NOTCH ligands. However, the decrease in NOTCH signaling induced by overexpression of Dlk2 is reversed by the overexpression of Dlk1, and viceversa. We conclude that DLK1 and DLK2 act as inhibitory non-canonical protein ligands for the NOTCH1 receptor that modulate NOTCH signaling.


Subject(s)
Fibroblasts/metabolism , Intercellular Signaling Peptides and Proteins/metabolism , Receptor, Notch1/metabolism , Signal Transduction , 3T3 Cells , 3T3-L1 Cells , Adaptor Proteins, Signal Transducing , Adipocytes/cytology , Adipocytes/metabolism , Animals , Binding, Competitive , Blotting, Western , Calcium-Binding Proteins/genetics , Calcium-Binding Proteins/metabolism , Cells, Cultured , Embryo, Mammalian/cytology , Fibroblasts/cytology , HEK293 Cells , Humans , Immunoprecipitation , Intercellular Signaling Peptides and Proteins/genetics , Intracellular Signaling Peptides and Proteins/genetics , Intracellular Signaling Peptides and Proteins/metabolism , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mice , Mice, Inbred BALB C , Mice, Knockout , Protein Binding , Receptor, Notch1/genetics , Serrate-Jagged Proteins , Two-Hybrid System Techniques
8.
J Bone Miner Res ; 26(7): 1457-71, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21308776

ABSTRACT

Delta-like 1/fetal antigen 1 (DLK1/FA-1) is a transmembrane protein belonging to the Notch/Delta family that acts as a membrane-associated or a soluble protein to regulate regeneration of a number of adult tissues. Here we examined the role of DLK1/FA-1 in bone biology using osteoblast-specific Dlk1-overexpressing mice (Col1-Dlk1). Col1-Dlk1 mice displayed growth retardation and significantly reduced total body weight and bone mineral density (BMD). Micro-computed tomographis (µCT) scanning revealed a reduced trabecular and cortical bone volume fraction. Tissue-level histomorphometric analysis demonstrated decreased bone-formation rate and enhanced bone resorption in Col1-Dlk1 mice compared with wild-type mice. At a cellular level, Dlk1 markedly reduced the total number of bone marrow (BM)-derived colony-forming units fibroblasts (CFU-Fs), as well as their osteogenic capacity. In a number of in vitro culture systems, Dlk1 stimulated osteoclastogenesis indirectly through osteoblast-dependent increased production of proinflammatory bone-resorbing cytokines (eg, Il7, Tnfa, and Ccl3). We found that ovariectomy (ovx)-induced bone loss was associated with increased production of Dlk1 in the bone marrow by activated T cells. Interestingly, Dlk1(-/-) mice were significantly protected from ovx-induced bone loss compared with wild-type mice. Thus we identified Dlk1 as a novel regulator of bone mass that functions to inhibit bone formation and to stimulate bone resorption. Increasing DLK1 production by T cells under estrogen deficiency suggests its possible use as a therapeutic target for preventing postmenopausal bone loss.


Subject(s)
Bone Resorption/metabolism , Bone Resorption/pathology , Bone and Bones/pathology , Estrogens/deficiency , Intercellular Signaling Peptides and Proteins/metabolism , Animals , Body Patterning , Body Weight , Bone Resorption/blood , Bone and Bones/abnormalities , Bone and Bones/metabolism , Calcium-Binding Proteins , Cell Differentiation , Collagen Type I/metabolism , Estrogens/metabolism , Female , Gene Expression Regulation , Immunologic Factors/genetics , Immunologic Factors/metabolism , Intercellular Signaling Peptides and Proteins/blood , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , NF-kappa B/metabolism , Organ Size , Osteoblasts/metabolism , Osteoblasts/pathology , Ovariectomy , Phenotype , Signal Transduction , T-Lymphocytes/metabolism
9.
Eur J Immunol ; 39(9): 2556-70, 2009 Sep.
Article in English | MEDLINE | ID: mdl-19662631

ABSTRACT

Macrophages present different Notch receptors and ligands on their surface. Following macrophage activation by LPS or other TLR ligands, Notch1 expression is upregulated. We report here that Notch signaling increases both basal and LPS-induced NF-kappaB activation, favoring the expression of genes implicated in the inflammatory response, such as the cytokines TNF-alpha and IL-6, or enzymes, such as iNOS. Delta4 seems to be the most effective ligand to induce Notch activation and increasing NF-kappaB transcriptional activity in macrophages. We show that Notch1 signaling promotes NF-kappaB translocation to the nucleus and DNA binding by increasing both phosphorylation of the IkappaB kinase alpha/beta complex and the expression of some NF-kappaB family members. Treatment of macrophages with the gamma-secretase inhibitor DAPT, which prevents the cleavage and activation of Notch receptors, inhibits all these processes, diminishing NF-kappaB activity following LPS stimulation. Additionally, we show that the active intracellular Notch fragment can directly interact with TNF-alpha and iNOS promoters. Our results suggest that Notch signaling results in an amplification of the macrophage-dependent inflammatory response by enhancing NF-kappaB signaling.


Subject(s)
Macrophage Activation/immunology , Macrophages, Peritoneal/immunology , NF-kappa B/immunology , Receptor, Notch1/immunology , Amyloid Precursor Protein Secretases/antagonists & inhibitors , Amyloid Precursor Protein Secretases/immunology , Amyloid Precursor Protein Secretases/metabolism , Animals , Cell Line , Enzyme Inhibitors/pharmacology , Humans , I-kappa B Kinase/immunology , I-kappa B Kinase/metabolism , Intracellular Signaling Peptides and Proteins , Lipopolysaccharides/pharmacology , Macrophage Activation/drug effects , Macrophages, Peritoneal/drug effects , Macrophages, Peritoneal/metabolism , Male , Membrane Proteins/immunology , Membrane Proteins/metabolism , Mice , NF-kappa B/metabolism , Receptor, Notch1/metabolism , Signal Transduction/drug effects , Signal Transduction/immunology , Up-Regulation/drug effects , Up-Regulation/immunology
10.
J Mol Biol ; 367(5): 1270-80, 2007 Apr 13.
Article in English | MEDLINE | ID: mdl-17320102

ABSTRACT

The Dlk1 gene appears to function as a regulator of adipogenesis. Adult Dlk1-deficient mice are obese, but adipose tissue still develops in transgenic mice overexpressing an Fc-dlk1 fusion protein, and neither type of genetically modified mice displays serious abnormalities. It was therefore possible that one yet unidentified gene might either compensate or antagonize for the absence or for overexpression, respectively, of Dlk1 in those animals. In database searches, we found a novel gene, EGFL9, encoding for a protein whose structural features are virtually identical to those of dlk1, suggesting it may function in a similar way. As dlk1 does, the protein encoded by EGFL9/Dlk2 affects adipogenesis of 3T3-L1 preadipocytes and mesenchymal C3H10T1/2 cells; however, it does so in an opposite way to that of dlk1. In addition, expression levels of both genes appear to be inversely correlated in both cell lines. Moreover, enforced changes in the expression of one gene affect the expression levels of the other. Our data suggest that adipogenesis may be modulated by the coordinated expression of Dlk1 and EGFL9/Dlk2.


Subject(s)
Adipogenesis/genetics , Intercellular Signaling Peptides and Proteins/genetics , Intercellular Signaling Peptides and Proteins/physiology , 3T3-L1 Cells , Adipocytes/cytology , Adipocytes/metabolism , Amino Acid Sequence , Animals , Calcium-Binding Proteins , Cells, Cultured , Gene Expression Regulation , Intercellular Signaling Peptides and Proteins/metabolism , Mice , Mice, Inbred Strains , Molecular Sequence Data , Phylogeny , Sequence Homology, Amino Acid , Tissue Distribution , Transfection
11.
J Immunol ; 176(9): 5362-73, 2006 May 01.
Article in English | MEDLINE | ID: mdl-16622004

ABSTRACT

Notch signaling has been extensively implicated in cell-fate determination along the development of the immune system. However, a role for Notch signaling in fully differentiated immune cells has not been clearly defined. We have analyzed the expression of Notch protein family members during macrophage activation. Resting macrophages express Notch-1, -2, and -4, as well as the Notch ligands Jagged-1 and -2. After treatment with LPS and/or IFN-gamma, we observed a p38 MAPK-dependent increase in Notch-1 and Jagged-1 mRNA and protein levels. To study the role of Notch signaling in macrophage activation, we forced the transient expression of truncated, active intracellular Notch-1 (Notch-IC) proteins in Raw 264.7 cells and analyzed their effects on the activity of transcription factors involved in macrophage activation. Notch-IC increased STAT-1-dependent transcription. Furthermore, Raw 264.7 Notch-IC stable transfectants increased STAT1-dependent transcription in response to IFN-gamma, leading to higher expression of IFN regulatory factor-1, suppressor of cytokine signaling-1, ICAM-1, and MHC class II proteins. This effect was independent from an increase of STAT1 Tyr or Ser phosphorylation. However, inducible NO synthase expression and NO production decreased under the same conditions. Our results show that Notch up-regulation and subsequent signaling following macrophage activation modulate gene expression patterns known to affect the function of mature macrophages.


Subject(s)
Antigen Presentation/immunology , Macrophage Activation/immunology , Macrophages/immunology , Macrophages/metabolism , Receptor, Notch1/metabolism , Signal Transduction , Active Transport, Cell Nucleus , Animals , Calcium-Binding Proteins/genetics , Calcium-Binding Proteins/metabolism , Cell Line , DNA/metabolism , Gene Expression Regulation/drug effects , Genes, Reporter/genetics , Inflammation/immunology , Inflammation/metabolism , Intercellular Signaling Peptides and Proteins , Interferon-gamma/biosynthesis , Jagged-1 Protein , Lipopolysaccharides/pharmacology , Macrophages/drug effects , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mice , Nitric Oxide Synthase Type II/metabolism , Phosphorylation , Protein Kinase Inhibitors/pharmacology , RNA, Messenger/genetics , Receptor, Notch1/genetics , STAT1 Transcription Factor/genetics , STAT1 Transcription Factor/metabolism , STAT3 Transcription Factor/genetics , STAT3 Transcription Factor/metabolism , Serrate-Jagged Proteins , Transcription Factor AP-1/metabolism , Up-Regulation/drug effects , p38 Mitogen-Activated Protein Kinases/metabolism
12.
Exp Cell Res ; 303(2): 343-59, 2005 Feb 15.
Article in English | MEDLINE | ID: mdl-15652348

ABSTRACT

The protein dlk, encoded by the Dlk1 gene, belongs to the Notch epidermal growth factor (EGF)-like family of receptors and ligands, which participate in cell fate decisions during development. The molecular mechanisms by which dlk regulates cell differentiation remain unknown. By using the yeast two-hybrid system, we found that dlk interacts with Notch1 in a specific manner. Moreover, by using luciferase as a reporter gene under the control of a CSL/RBP-Jk/CBF-1-dependent promoter in the dlk-negative, Notch1-positive Balb/c 14 cell line, we found that addition of synthetic dlk EGF-like peptides to the culture medium or forced expression of dlk decreases endogenous Notch activity. Furthermore, the expression of the gene Hes-1, a target for Notch1 activation, diminishes in confluent Balb/c14 cells transfected with an expression construct encoding for the extracellular EGF-like region of dlk. The expression of Dlk1 and Notch1 increases in 3T3-L1 cells maintained in a confluent state for several days, which is associated with a concomitant decrease in Hes-1 expression. On the other hand, the decrease of Dlk1 expression in 3T3-L1 cells by antisense cDNA transfection is associated with an increase in Hes-1 expression. These results suggest that dlk functionally interacts in vivo with Notch1, which may lead to the regulation of differentiation processes modulated by Notch1 activation and signaling, including adipogenesis.


Subject(s)
Membrane Proteins/metabolism , Receptors, Cell Surface/metabolism , Transcription Factors/metabolism , 3T3-L1 Cells , Animals , Base Sequence , Basic Helix-Loop-Helix Transcription Factors , Cell Line , DNA, Antisense/genetics , Epidermal Growth Factor/chemistry , Epidermal Growth Factor/genetics , Epidermal Growth Factor/metabolism , Gene Expression Regulation , Genes, Reporter , Homeodomain Proteins/chemistry , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Humans , In Vitro Techniques , Intracellular Signaling Peptides and Proteins , Luciferases/genetics , Membrane Proteins/chemistry , Membrane Proteins/genetics , Mice , Receptor, Notch1 , Receptors, Cell Surface/chemistry , Receptors, Cell Surface/genetics , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Repetitive Sequences, Amino Acid , Transcription Factor HES-1 , Transcription Factors/chemistry , Transcription Factors/genetics , Transfection , Two-Hybrid System Techniques
13.
Exp Cell Res ; 274(2): 178-88, 2002 Apr 01.
Article in English | MEDLINE | ID: mdl-11900478

ABSTRACT

The EGF-like membrane protein dlk plays a crucial role in the control of cell differentiation. Overexpression of the protein prevents, whereas inhibition of its expression increases, adipocyte differentiation of 3T3-L1 cells in response to Insulin-like Growth Factor I (IGF-1) or insulin. We have investigated whether dlk modulates the signaling pathways known to control this process. We found that the levels of dlk expression modulated signaling through the IGF-1 receptor, causing changes in the activation levels and kinetics of Extracellular-Regulated Kinase/Mitogen-Activated Protein Kinase (ERK/MAPK) that correlated with differentiation outcome. These changes occurred in response to IGF-1 or insulin but not in response to Epidermal Growth Factor. However, the levels of expression of IGF-1 receptor, or the activation of Insulin Receptor Substrate-1 in response to IGF-1, were not affected by the levels of dlk expression. Therefore, dlk appears to modulate ERK/MAPK signaling in response to specific differentiation signals.


Subject(s)
Cell Differentiation/physiology , Cell Membrane/metabolism , Insulin-Like Growth Factor I/metabolism , MAP Kinase Signaling System/physiology , Membrane Proteins/deficiency , Mitogen-Activated Protein Kinases/metabolism , Receptor, IGF Type 1/metabolism , 3T3 Cells , Adipose Tissue/drug effects , Adipose Tissue/metabolism , Animals , Cell Differentiation/drug effects , Cell Membrane/drug effects , Enzyme Inhibitors/pharmacology , Gene Expression Regulation/drug effects , Gene Expression Regulation/physiology , Insulin/pharmacology , Insulin-Like Growth Factor I/pharmacology , Intracellular Signaling Peptides and Proteins , MAP Kinase Signaling System/drug effects , Membrane Proteins/genetics , Mice , Mitogen-Activated Protein Kinases/drug effects , Phosphatidylinositol 3-Kinases/metabolism , Phosphoinositide-3 Kinase Inhibitors , Receptor, IGF Type 1/drug effects
14.
Biochem Biophys Res Commun ; 291(2): 193-204, 2002 Feb 22.
Article in English | MEDLINE | ID: mdl-11846389

ABSTRACT

Levels of dlk, an EGF-like homeotic protein, are critical for several differentiation processes. Because growth and differentiation are, in general, exclusive of each other, and increasing evidence indicates that Dlk1 expression changes in tumorigenic processes, we studied whether dlk could also affect cell growth. We found that, in response to glucocorticoids, Balb/c 3T3 cells with diminished levels of dlk expression develop foci-like cells that have lost contact inhibition, display altered morphology, and grow faster than control cell lines. Balb/c 3T3 cells spontaneously growing more rapidly are also dlk-negative cells. Moreover, screening by the yeast two-hybrid system, using Dlk1 constructs as baits, resulted in the isolation of GAS1 and acrogranin cDNAs. Interestingly, these proteins are cysteine-rich molecules involved in the control of cell growth. Taken together, these observations suggest that dlk may participate in a network of interactions controlling how the cells respond to growth or differentiation signals.


Subject(s)
Cell Division , Growth Substances , Homeodomain Proteins/metabolism , Homeodomain Proteins/physiology , Intercellular Signaling Peptides and Proteins , Membrane Proteins/metabolism , Membrane Proteins/physiology , 3T3 Cells , Animals , Cell Cycle Proteins , Epidermal Growth Factor/genetics , GPI-Linked Proteins , Glycoproteins/chemistry , Glycoproteins/genetics , Glycoproteins/metabolism , Granulins , Homeodomain Proteins/chemistry , Homeodomain Proteins/genetics , Humans , Intracellular Signaling Peptides and Proteins , Membrane Proteins/chemistry , Membrane Proteins/genetics , Mice , Mice, Inbred BALB C , Progranulins , Protein Structure, Tertiary , RNA, Messenger/biosynthesis , Repetitive Sequences, Amino Acid , Two-Hybrid System Techniques , Yeasts/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...