Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 127
Filter
1.
Dev Cell ; 56(15): 2223-2236.e5, 2021 08 09.
Article in English | MEDLINE | ID: mdl-34324859

ABSTRACT

Cancer tissue often comprises multiple tumor clones with distinct oncogenic alterations such as Ras or Src activation, yet the mechanism by which tumor heterogeneity drives cancer progression remains elusive. Here, we show in Drosophila imaginal epithelium that clones of Ras- or Src-activated benign tumors interact with each other to mutually promote tumor malignancy. Mechanistically, Ras-activated cells upregulate the cell-surface ligand Delta while Src-activated cells upregulate its receptor Notch, leading to Notch activation in Src cells. Elevated Notch signaling induces the transcriptional repressor Zfh1/ZEB1, which downregulates E-cadherin and cell death gene hid, leading to Src-activated invasive tumors. Simultaneously, Notch activation in Src cells upregulates the cytokine Unpaired/IL-6, which activates JAK-STAT signaling in neighboring Ras cells. Elevated JAK-STAT signaling upregulates the BTB-zinc-finger protein Chinmo, which downregulates E-cadherin and thus generates Ras-activated invasive tumors. Our findings provide a mechanistic explanation for how tumor heterogeneity triggers tumor progression via cell-cell interactions.


Subject(s)
Neoplasms/metabolism , Oncogene Protein pp60(v-src)/metabolism , Proto-Oncogene Proteins p21(ras)/metabolism , Animals , Cadherins/metabolism , Carcinogenesis/metabolism , Cell Transformation, Neoplastic/genetics , Drosophila Proteins/metabolism , Drosophila melanogaster/metabolism , Epithelium/metabolism , Gene Expression Regulation, Neoplastic/genetics , Genes, ras/genetics , Genes, ras/physiology , Imaginal Discs/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Membrane Proteins/metabolism , Nerve Tissue Proteins/metabolism , Oncogene Protein pp60(v-src)/physiology , Proto-Oncogene Proteins p21(ras)/physiology , Receptors, Notch/genetics , Receptors, Notch/metabolism , Repressor Proteins/metabolism , Signal Transduction/physiology , Transcription Factors/metabolism , Zinc Fingers
2.
BMC Urol ; 20(1): 145, 2020 Sep 09.
Article in English | MEDLINE | ID: mdl-32907556

ABSTRACT

BACKGROUND: Bladder cancer (BC) is one of the most common malignancies of the urinary tract. The role of transient receptor potential melastatin 7 (TRPM7) in BC remains unclear. The aim of this study was to investigate the function and signal transduction pathway of TRPM7 in BC. METHODS: T24 and UMUC3 cells were used to evaluate the molecular mechanism of TRPM7 by immunoblot analysis. Small interfering RNA was used to knockdown TRPM7, and the effect of silencing TRPM7 was studied by wound healing, migration, and invasion assays in T24 and UMUC3 cells. Xenograft model study was obtained to analyze the effect of TRPM7 inhibition in vivo. RESULTS: Silencing of TRPM7 decreased the migration and invasion ability of T24 and UMUC3 cells. The phosphorylation of Src, Akt, and JNK (c-Jun N-terminal kinase) was also suppressed by TRPM7 silencing. Src, Akt, and JNK inhibitors effectively inhibited the migration and invasion of T24 and UMUC3 cells. In addition, the TRPM7 inhibitor, carvacrol, limited the tumor size in a xenograft model. CONCLUSION: Our data reveal that TRPM7 regulates the migration and invasion of T24 and UMUC3 cells via the Src, Akt, and JNK signaling pathway. Therefore, TRPM7 suppression could be a potential treatment for BC patients.


Subject(s)
MAP Kinase Signaling System/physiology , Oncogene Protein pp60(v-src)/physiology , Protein Serine-Threonine Kinases/genetics , Proto-Oncogene Proteins c-akt/physiology , TRPM Cation Channels/genetics , Urinary Bladder Neoplasms/genetics , Urinary Bladder Neoplasms/pathology , Animals , Cell Movement , Cell Proliferation , Female , Gene Knockdown Techniques , Humans , Mice , Mice, Inbred BALB C , Neoplasm Invasiveness , Signal Transduction , Tumor Cells, Cultured , Urinary Bladder Neoplasms/etiology
3.
Biochem J ; 458(1): 81-93, 2014 Feb 15.
Article in English | MEDLINE | ID: mdl-24266736

ABSTRACT

The proto-oncogenic tyrosine kinase c-Src is up-regulated in various human cancers, implicating its role in tumour progression. Upon activation, c-Src translocates to focal adhesions and initiates intracellular signalling cascades that promote malignant transformation, but the underlying mechanisms for c-Src translocation remain unclear. In the present study we show that c-Src up-regulation perturbs sphingolipid/cholesterol-enriched membrane microdomains by activating ceramide synthesis, resulting in promotion of c-Src translocation. Using an inducible c-Src expression system in Csk (C-terminal Src kinase)-deficient fibroblasts, we found that translocation of c-Src to focal adhesions/podosomes occurs in the later stages of cell transformation. Activated c-Src is liberated from microdomains and promotes the phosphorylation of FAK (focal adhesion kinase) and cortactin localized to focal adhesions/podosomes. In parallel with these events, anabolic metabolism of ceramides is activated by up-regulation of the de novo synthesis pathway. Inhibition of ceramide conversion into glucosylceramide promotes liberation of c-Src from microdomains, and inhibition of de novo ceramide synthesis restores the microdomain distribution of c-Src and suppresses malignant phenotypes such as increased cell motility and anchorage-independent cell growth. These results suggest that c-Src-induced activation of ceramide synthesis impairs the integrity of microdomains and contributes to malignant progression by promoting the translocation of c-Src to focal adhesions/podosomes.


Subject(s)
Cell Transformation, Neoplastic , Ceramides/metabolism , Focal Adhesions , Membrane Microdomains/physiology , Oncogene Protein pp60(v-src)/physiology , Animals , Mice , Reverse Transcriptase Polymerase Chain Reaction
4.
Neurochem Int ; 63(5): 413-22, 2013 Nov.
Article in English | MEDLINE | ID: mdl-23911694

ABSTRACT

The PC12 (rat pheochromocytoma) cell line is a popular model system to study neuronal differentiation. Upon prolonged nerve growth factor (NGF) exposure these tumor cells stop to divide, become polygonal, grow projections and start to look and behave like sympathetic neurons. Differentiation of PC12 cells can also be induced by peptidyl-aldehyde proteasome inhibitors, such as Z-Leu-Leu-Leu-al (also known as MG-132) or via infection of the cells with Rous sarcoma virus. The signal transduction pathways underlying process formation, however, are still not fully understood. The liganded NGF receptor initiates a protein kinase cascade a member of which is Extracellular Signal-Regulated Kinase (ERK). Active ERK1/2 enzymes phosphorylate various cytoplasmic proteins and can also be translocated into the nucleus, where they regulate gene expression by activating key transcription factors. Using immunological methods we detected phosphorylation of TrkA, prolongedactivation of Src, and ERK1/2 with nuclear translocation of the latter during MG-132-induced process formation of PC12 cells. Activated Src remained predominantly cytoplasmic. MG-132-induced sustained ERK1/2 activation, nuclear translocation and neuritogenesis required the intact function of Src since these phenomena were markedly reduced or failed upon chemical inhibition of Src tyrosine protein kinase activity.


Subject(s)
Leupeptins/pharmacology , Oncogene Protein pp60(v-src)/physiology , Proteasome Inhibitors/pharmacology , Animals , Blotting, Western , Enzyme Activation , Extracellular Signal-Regulated MAP Kinases/metabolism , Kinetics , Microscopy, Fluorescence , Oncogene Protein pp60(v-src)/metabolism , PC12 Cells , Phosphorylation , Rats
5.
PLoS One ; 5(10): e13572, 2010 Oct 22.
Article in English | MEDLINE | ID: mdl-21042581

ABSTRACT

BACKGROUND: We have been investigating how interruption of differentiation contributes to the oncogenic process and the possibility to reverse the transformed phenotype by restoring differentiation. In a previous report, we correlated the capacity of intracellular Notch (ICN) to suppress v-Src-mediated transformation of quail neuroretina (QNR/v-src(ts)) cells with the acquisition by these undifferentiated cells of glial differentiation markers. METHODOLOGY/PRINCIPAL FINDINGS: In this work, we have identified autocrine TGF-ß3 signaling activation as a major effector of Notch-induced phenotypic changes, sufficient to induce transition in differentiation markers expression, suppress morphological transformation and significantly inhibit anchorage-independent growth. We also show that this signaling is constitutive of and contributes to ex-vivo autonomous QNR cell differentiation and that its down-regulation is essential to achieve v-Src-induced transformation. CONCLUSIONS/SIGNIFICANCE: These results support the possibility that Notch signaling induces differentiation and suppresses transformation by a novel mechanism, involving secreted proteins. They also underline the importance of extracellular signals in controlling the balance between normal and transformed phenotypes.


Subject(s)
Cell Differentiation/physiology , Neurons/cytology , Oncogene Protein pp60(v-src)/antagonists & inhibitors , Receptors, Notch/metabolism , Signal Transduction , Transforming Growth Factor beta/physiology , Animals , Base Sequence , Blotting, Western , Coturnix , DNA Primers , Fluorescent Antibody Technique , Myosin Light Chains/metabolism , Oncogene Protein pp60(v-src)/physiology , Phosphorylation , RNA, Messenger/genetics , Reverse Transcriptase Polymerase Chain Reaction , Transforming Growth Factor beta/genetics
6.
J Invest Surg ; 23(6): 303-8, 2010 Dec.
Article in English | MEDLINE | ID: mdl-21208094

ABSTRACT

Sarcomas are rare tumors, and malignant fibrous histiocytoma (MFH) is the most common soft tissue sarcoma found in adults. Increasing evidence suggests a possible role for AKT activation in soft tissue sarcoma. In the present study, we established a primary human MFH cell line (named MSUMFH cells) from a fresh surgically resected MFH tumor. These cells morphologically resembled human normal fibroblasts that are the presumptive cells of origin of MFH tumor cells. As there is, unfortunately, no standard marker other than morphology to identify MFH at the cellular level, we compared MSUMFH cells to primary nonmalignant fibroblasts and the primary tumor specimen to characterize its signaling. AKT was hyperactivated in both the MSUMFH cell line and original primary MFH tumor cells compared to normal fibroblasts. The AKT hyperactivity in the MSUMFH cell line was not accompanied by activation of focal adhesion kinase (FAK) or downregulated expression of PTEN, each of which is a putative upstream regulator of AKT. In contrast, this AKT hyperactivation required PI-3K and Src in MSUMFH cells. This PI-3K and Src-dependent AKT-activated MSUMFH cell line that we established in this study may be beneficial for the future cell-based study of MFH biology.


Subject(s)
Histiocytoma, Malignant Fibrous/physiopathology , Proto-Oncogene Proteins c-akt/physiology , Signal Transduction , Histiocytoma, Malignant Fibrous/pathology , Humans , Oncogene Protein pp60(v-src)/physiology , Phosphatidylinositol 3-Kinases/metabolism
7.
Clin Cancer Res ; 15(19): 6035-41, 2009 Oct 01.
Article in English | MEDLINE | ID: mdl-19755386

ABSTRACT

PURPOSE: Waldenstrom macroglobulinemia is a lymphoplasmacytic lymphoma characterized by widespread involvement of the bone marrow. Despite different options of therapy, Waldenstrom macroglobulinemia is still incurable. Src tyrosine kinase has been shown to play a central role in the regulation of a variety of biological processes, such as cell proliferation, migration, adhesion, and survival in solid tumors. We sought to determine whether the protein tyrosine kinase Src regulates adhesion, migration, and survival in Waldenstrom macroglobulinemia. EXPERIMENTAL DESIGN: We tested the expression of Src tyrosine kinase in Waldenstrom macroglobulinemia and normal cells, and the effect of the specific Src inhibitor AZD0530 on the adhesion, migration, cell cycle, and survival of a Waldenstrom macroglobulinemia cell line and patient samples. Moreover, we tested the effect of AZD0530 on cytoskeletal and cell cycle signaling in Waldenstrom macroglobulinemia. RESULTS: We show that Src is overexpressed in Waldenstrom macroglobulinemia cells compared with control B cells, and that the use of the Src inhibitor AZD0530 led to significant inhibition of adhesion, migration, and cytoskeletal signaling induced by SDF1. Moreover, inhibition of Src activity induced G(1) cell cycle arrest; however, it had minimal effect on survival of Waldenstrom macroglobulinemia cells, and no significant effect on survival of normal cells. CONCLUSIONS: Taken together, these results delineate the role of Src kinase activity in Waldenstrom macroglobulinemia and provide the framework for future clinical trials using Src inhibitors in combination with other drugs to improve the outcome of patients with Waldenstrom macroglobulinemia.


Subject(s)
Chemotaxis , Oncogene Protein pp60(v-src)/physiology , Waldenstrom Macroglobulinemia/pathology , Benzodioxoles/pharmacology , Cell Adhesion/drug effects , Cell Cycle/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Chemokine CXCL12/pharmacology , Chemotaxis/drug effects , Cytotoxins/pharmacology , Drug Evaluation, Preclinical , Humans , Oncogene Protein pp60(v-src)/antagonists & inhibitors , Oncogene Protein pp60(v-src)/metabolism , Phosphorylation/drug effects , Protein Kinase Inhibitors/pharmacology , Quinazolines/pharmacology , Waldenstrom Macroglobulinemia/metabolism
8.
J Neurosci ; 29(17): 5690-700, 2009 Apr 29.
Article in English | MEDLINE | ID: mdl-19403835

ABSTRACT

Signal relay by guidance receptors at the axonal growth cone is a process essential for the assembly of a functional nervous system. We investigated the in vivo function of Src family kinases (SFKs) as growth cone guidance signaling intermediates in the context of spinal lateral motor column (LMC) motor axon projection toward the ventral or dorsal limb mesenchyme. Using in situ mRNA detection we determined that Src and Fyn are expressed in LMC motor neurons of chick and mouse embryos at the time of limb trajectory selection. Inhibition of SFK activity by C-terminal Src kinase (Csk) overexpression in chick LMC axons using in ovo electroporation resulted in LMC axons selecting the inappropriate dorsoventral trajectory within the limb mesenchyme, with medial LMC axon projecting into the dorsal and ventral limb nerve with apparently random incidence. We also detected LMC axon trajectory choice errors in Src mutant mice demonstrating a nonredundant role for Src in motor axon guidance in agreement with gain and loss of Src function in chick LMC neurons which led to the redirection of LMC axons. Finally, Csk-mediated SFK inhibition attenuated the retargeting of LMC axons caused by EphA or EphB over-expression, implying the participation of SFKs in Eph-mediated LMC motor axon guidance. In summary, our findings demonstrate that SFKs are essential for motor axon guidance and suggest that they play an important role in relaying ephrin:Eph signals that mediate the selection of motor axon trajectory in the limb.


Subject(s)
Avian Proteins/physiology , Axons/enzymology , Extremities/embryology , Extremities/innervation , Motor Neurons/enzymology , Oncogene Protein pp60(v-src)/physiology , Proto-Oncogene Proteins c-fyn/physiology , Animals , Avian Proteins/antagonists & inhibitors , Avian Proteins/biosynthesis , Avian Proteins/genetics , Chick Embryo , Mice , Oncogene Protein pp60(v-src)/antagonists & inhibitors , Oncogene Protein pp60(v-src)/biosynthesis , Oncogene Protein pp60(v-src)/genetics , Proto-Oncogene Proteins c-fyn/biosynthesis , Proto-Oncogene Proteins c-fyn/genetics
9.
J Neurochem ; 110(3): 779-90, 2009 Aug.
Article in English | MEDLINE | ID: mdl-19457108

ABSTRACT

Fetal alcohol syndrome is a leading cause of mental retardation. The neuropathology found in patients with fetal alcohol syndrome overlaps with those with mutations in the gene for cell adhesion molecule (L1). We have previously shown that L1-mediated neurite outgrowth and L1 activation of extracellular receptor kinases 1/2 are inhibited at low concentrations of ethanol. One possible mechanism for this effect is through disruption of a tyrosine-based sorting signal, Y(1176)RSLE, on the cytoplasmic domain of L1. Our goal was to determine if ethanol inhibited the sorting signal or its phosphorylation state. Using cerebellar granule neurons and dorsal root ganglion neurons, we found that ethanol had no effect on L1 distribution to the growth cone or its ability to be expressed on the cell surface as determined by confocal microscopy. In cerebellar granule neurons, clustering of L1 resulted in increased dephosphorylation of Y(1176), increased L1 tyrosine phosphorylation, and an increase in the activation of pp60(src) as measured by immunoblot. All changes were inhibited by 25 mM ethanol. Using PP2 to inhibit pp60(src) activation resulted in inhibition of increases in L1 tyrosine and extracellular receptor kinases 1/2 phosphorylation, and Y(1176) dephosphorylation. We conclude that ethanol disrupts L1 trafficking/signaling following its expression on the surface of the growth cone, and prior to its activation of pp60(src).


Subject(s)
Ethanol/toxicity , Neural Cell Adhesion Molecule L1/antagonists & inhibitors , Neural Cell Adhesion Molecule L1/metabolism , Oncogene Protein pp60(v-src)/antagonists & inhibitors , Oncogene Protein pp60(v-src)/metabolism , Tyrosine/antagonists & inhibitors , Tyrosine/metabolism , Animals , Cells, Cultured , Chick Embryo , Mice , Neural Cell Adhesion Molecule L1/physiology , Oncogene Protein pp60(v-src)/physiology , Phosphorylation/drug effects , Phosphorylation/physiology , Protein Interaction Mapping , Rats , Signal Transduction/drug effects , Signal Transduction/physiology
10.
Cell Commun Adhes ; 15(1): 75-84, 2008 May.
Article in English | MEDLINE | ID: mdl-18649180

ABSTRACT

Modulation of gap junction structures and gap junctional communication is important in maintaining tissue homeostasis and can be controlled via phosphorylation of connexin 43 (Cx43) through several different signaling pathways. Transformation of cells by v-src has been shown to down-regulate gap junction communication coincident with an increase in tyrosine phosphorylation on Cx43. Activation of mitogen-activated protein kinase (MAPK) and protein kinase C (PKC) also lead to down-regulation via phosphorylation on specific serine residues. Using phosphospecific anti-Cx43 antibodies generated by the authors' laboratory to specific tyrosines (src substrates) and serine residues (MAPK and PKC substrates) to probe LA-25 cells (which express temperature-sensitive v-src), the authors show that distinct tyrosine and serines residues are phosphorylated in response to v-src activity. They show that tyrosine phosphorylation appears to occur predominantly in gap junction plaques when src is active. In addition, src activation led to increased phosphorylation of apparent MAPK and PKC sites in Cx43. These results indicate all three signaling pathways could contribute to gap junction down-regulation during src transformation in LA-25 cells.


Subject(s)
Connexin 43/metabolism , Oncogene Protein pp60(v-src)/physiology , Serine/metabolism , Signal Transduction/physiology , Tyrosine/metabolism , Amino Acid Sequence , Animals , Cell Line , Connexin 43/genetics , Dogs , Gap Junctions/metabolism , Humans , Molecular Sequence Data , Phosphorylation , Rats , Serine/genetics , Tyrosine/genetics
11.
Oncogene ; 27(46): 6023-33, 2008 Oct 09.
Article in English | MEDLINE | ID: mdl-18574469

ABSTRACT

Sphingosine kinase 1 (SPHK1) is overexpressed in solid tumors and leukemia. However, the mechanism of SPHK1 overexpression by oncogenes has not been defined. We found that v-Src-transformed NIH3T3 cells showed a high SPHK1 mRNA, SPHK1 protein and SPHK enzyme activity. siRNA of SPHK1 inhibited the growth of v-Src-NIH3T3, suggesting the involvement of SPHK1 in v-Src-induced oncogenesis. v-Src-NIH3T3 showed activations of protein kinase C-alpha, signal transducers and activators of transcription 3 and c-Jun NH(2)-terminal kinase. Their inhibition suppressed SPHK1 expression in v-Src-NIH3T3, whereas their overexpression increased SPHK1 mRNA in NIH3T3. Unexpectedly, the nuclear run-on assay and the promoter analysis using 5'-promoter region of mouse SPHK1 did not show any significant difference between mock- and v-Src-NIH3T3. Furthermore, the half-life of SPHK1 mRNA in mock-NIH3T3 was nearly 15 min, whereas that of v-Src-NIH3T3 was much longer. Examination of two AU-rich region-binding proteins, AUF1 and HuR, that regulate mRNA decay reciprocally, showed decreased total AUF1 protein associated with increased tyrosine-phosphorylated form and increased serine-phosphorylated HuR protein in v-Src-NIH3T3. Modulation of AUF1 and HuR by their overexpression or siRNA revealed that SPHK1 mRNA in v-Src- and mock-NIH3T3 was regulated reciprocally by these factors. Our results showed, for the first time, a novel mechanism of v-Src-induced SPHK1 overexpression.


Subject(s)
Oncogene Protein pp60(v-src)/physiology , Phosphotransferases (Alcohol Group Acceptor)/genetics , RNA Stability/physiology , RNA-Binding Proteins/physiology , Animals , Antigens, Surface/genetics , Antigens, Surface/metabolism , Antigens, Surface/physiology , Cell Line, Transformed , Cell Proliferation/drug effects , ELAV Proteins , ELAV-Like Protein 1 , Gene Expression Regulation, Enzymologic , Half-Life , Heterogeneous Nuclear Ribonucleoprotein D0 , Heterogeneous-Nuclear Ribonucleoprotein D/genetics , Heterogeneous-Nuclear Ribonucleoprotein D/metabolism , Heterogeneous-Nuclear Ribonucleoprotein D/physiology , Mice , Models, Biological , NIH 3T3 Cells , Oncogene Protein pp60(v-src)/antagonists & inhibitors , Oncogene Protein pp60(v-src)/genetics , Phosphotransferases (Alcohol Group Acceptor)/metabolism , RNA Stability/genetics , RNA, Messenger/metabolism , RNA, Small Interfering/pharmacology , RNA-Binding Proteins/genetics , RNA-Binding Proteins/metabolism , Regulatory Sequences, Ribonucleic Acid/physiology , Signal Transduction/genetics , Signal Transduction/physiology , Transfection
12.
Apoptosis ; 13(4): 531-42, 2008 Apr.
Article in English | MEDLINE | ID: mdl-18322799

ABSTRACT

The molecular determinants which dictate survival and apoptosis/anoikis in human intestinal crypt cells remain to be fully understood. To this effect, the roles of beta1 integrin/Fak/Src signaling to the PI3-K/Akt-1, MEK/Erk, and p38 pathways, were investigated. The regulation of six Bcl-2 homologs (Bcl-2, Mcl-1, Bcl-X(L), Bax, Bak, Bad) was likewise analyzed. We report that: (1) Anoikis causes a down-activation of Fak, Src, Akt-1 and Erk1/2, a loss of Fak-Src association, and a sustained/enhanced activation of p38beta, which is required as apoptosis/anoikis driver; (2) PI3-K/Akt-1 up-regulates the expression of Bcl-X(L) and Mcl-1, down-regulates Bax and Bak, drives Bad phosphorylation (both serine112/136 residues) and antagonizes p38beta activation; (3) MEK/Erk up-regulates Bcl-2, drives Bad phosphorylation (serine112 residue), but does not antagonize p38bactivation; (4) PI3-K/Akt-1 is required for survival, whereas MEK/Erk is not; (5) Src acts as a cornerstone in the engagement of both pathways by beta1 integrins/Fak, and is crucial for survival; and (6) beta1 integrins/Fak and/or Src regulate Bcl-2 homologs as both PI3-K/Atk-1 and MEK/Erk combined. Hence, beta1 integrin/Fak/Src signaling translates into integrated mediating functions of p38beta activation and regulation of Bcl-2 homologs by PI3-K/Akt-1 and MEK/Erk, consequently determining their requirement (or not) for survival.


Subject(s)
Anoikis/physiology , Cell Survival/physiology , Focal Adhesion Kinase 1/physiology , Integrin beta1/physiology , Intestinal Mucosa/physiology , Oncogene Protein pp60(v-src)/physiology , Signal Transduction/physiology , Cells, Cultured , Down-Regulation , Humans , Intestinal Mucosa/cytology , MAP Kinase Kinase Kinases/physiology , Mitogen-Activated Protein Kinase 1/physiology , Mitogen-Activated Protein Kinase 11/physiology , Mitogen-Activated Protein Kinase 3/physiology , Phosphatidylinositol 3-Kinases/physiology , Proto-Oncogene Proteins c-akt/physiology , Proto-Oncogene Proteins c-bcl-2/physiology , Up-Regulation
13.
Genes Cells ; 13(1): 1-12, 2008 Jan.
Article in English | MEDLINE | ID: mdl-18173743

ABSTRACT

The c-src proto-oncogene product, c-Src, is frequently over-expressed and activated in various human malignant cancers, implicating a role for c-Src in cancer progression. To verify the role of c-Src, we analyzed the transforming ability of c-Src in mouse embryonic fibroblasts that lack Csk, a negative regulator of Src family kinases. Although Csk deficiency is not sufficient for cell transformation, c-Src over-expression induced characteristic transformed phenotypes including anchorage-independent growth and tumorigenecity. These phenotypes were dose-dependently inhibited by the re-expression of Csk, indicating that there is a certain threshold for c-Src transformation, which is determined by the c-Src : Csk ratio. In contrast to v-Src, c-Src induced the phosphorylation of a limited number of cellular proteins and elicited a restricted change in gene expression profiles. The activation of some critical targets for v-Src transformation, such as STAT3, was not significantly induced by c-Src transformation. Several genes that are involved in cancer progression, that is, cyclin D1 and HIF-1alpha, were induced by v-Src, but not by c-Src. Furthermore, v-Src tumors exhibited aggressive growth and extensive angiogenesis, while c-Src tumors grew more slowly accompanied by the induction of hematomas. These findings demonstrate that c-Src has the potential to induce cell transformation, but it requires coordination with an additional pathway(s) to promote tumor progression in vivo.


Subject(s)
Cell Transformation, Neoplastic/metabolism , Fibroblasts/cytology , Oncogene Protein pp60(v-src)/physiology , Protein-Tyrosine Kinases/physiology , Animals , CSK Tyrosine-Protein Kinase , Cell Transformation, Neoplastic/pathology , Cyclin D1/metabolism , Fibroblasts/metabolism , Gene Expression Regulation , Hematoma/metabolism , Hematoma/pathology , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Mice , Mice, Nude , Neovascularization, Pathologic , Oligonucleotide Array Sequence Analysis , Phosphorylation , Protein-Tyrosine Kinases/genetics , Proto-Oncogene Mas , STAT3 Transcription Factor/metabolism , Signal Transduction , Transplantation, Heterologous , src-Family Kinases
14.
Oncogene ; 27(7): 942-56, 2008 Feb 07.
Article in English | MEDLINE | ID: mdl-17700527

ABSTRACT

Diacylglycerol (DAG) kinases (Dgk), which phosphorylate DAG to generate phosphatidic acid, act as either positive or negative key regulators of cell signaling. We previously showed that Src mediates growth factors-induced activation of Dgk-alpha, whose activity is required for cell motility, proliferation and angiogenesis. Here, we demonstrate that both hepatocytes growth factor (HGF) stimulation and v-Src transformation induce tyrosine phosphorylation of Dgk-alpha on Y335, through a mechanism requiring its proline-rich C-terminal sequence. Moreover, we show that both proline-rich sequence and phosphorylation of Y335 of Dgk-alpha mediate: (i) its enzymatic activation, (ii) its ability to interact respectively with SH3 and SH2 domains of Src, (iii) its recruitment to the membrane. In addition, we show that phosphorylation of Dgk-alpha on Y335 is required for HGF-induced motility, while its constitutive recruitment at the membrane by myristylation is sufficient to trigger spontaneous motility in absence of HGF. Providing the first evidence that tyrosine phosphorylation of Dgk-alpha is required for growth-factors-induced activation and membrane recruitment, these findings underscore its relevance as a rheostat, whose activation is a threshold to elicit growth factors-induced migratory signaling.


Subject(s)
Cell Membrane/metabolism , Cell Movement , Diacylglycerol Kinase/metabolism , Hepatocyte Growth Factor/pharmacology , Myristic Acids/chemistry , Oncogene Protein pp60(v-src)/physiology , Tyrosine/metabolism , Animals , COS Cells/metabolism , Cell Communication , Cells, Cultured , Chlorocebus aethiops , Dogs , Enzyme Activation , Humans , Kidney/cytology , Kidney/metabolism , Phosphorylation , Proline/metabolism , Signal Transduction
15.
Oncogene ; 27(4): 528-39, 2008 Jan 17.
Article in English | MEDLINE | ID: mdl-17637741

ABSTRACT

In this study, we delineate the intracellular signalling pathways modulated by a conditional v-Src tyrosine kinase that lead to unrestrained proliferation and block of differentiation of primary avian myoblasts. By inhibiting Ras-MAPK kinase and phosphatidylinositol 3-kinase with different means, we find that both pathways play crucial roles in controlling v-Src-sustained growth factor and anchorage independence for proliferation. The Ras-MAPK kinase pathway also contributes to block of differentiation independently of cell proliferation since inhibition of this pathway both in proliferating and growth-arrested v-Src-transformed myoblasts induces expression of muscle-specific genes, fusion into multinucleated myotubes and assembly of specialized contractile structures. Importantly, we find that the p38 MAPK pathway is inhibited by v-Src in myoblasts and its forced activation results in growth inhibition and expression of differentiation, indicating p38 MAPK as a critical target of v-Src in growth transformation and myogenic differentiation. Furthermore, we show that downregulation of p38 MAPK activation may occur via Ras-MAPK kinase, thus highlighting a cross-regulation between the two pathways. Finally, we report that the simultaneous inhibition of MAPK kinase and calpain, combined to activation of p38 MAPK, are sufficient to reconstitute largely the differentiation potential of v-Src-transformed myoblasts.


Subject(s)
Cell Transformation, Neoplastic/genetics , Myoblasts, Skeletal/pathology , Oncogene Protein pp60(v-src)/physiology , Signal Transduction/genetics , Animals , Cell Adhesion/physiology , Cell Differentiation/physiology , Cell Line, Transformed , Cell Proliferation , Cell Transformation, Neoplastic/metabolism , Cells, Cultured , Chick Embryo , Mitogen-Activated Protein Kinase Kinases/physiology , Models, Biological , Myoblasts, Skeletal/metabolism , Phosphatidylinositol 3-Kinases/physiology , Proto-Oncogene Proteins p21(ras)/physiology , Quail , Transfection
16.
Exp Cell Res ; 313(6): 1090-105, 2007 Apr 01.
Article in English | MEDLINE | ID: mdl-17335807

ABSTRACT

Cancer cells depend on chemotaxis for invasion and frequently overexpress and/or activate Src. We previously reported that v-Src accelerates motility by promoting phosphoinositide 3-kinase (PI3-K) signalling but abrogates chemotaxis. We here addressed the mechanism of the loss of chemotactic response to platelet-derived growth factor (PDGF) gradients in fibroblasts harbouring a thermosensitive v-Src kinase. At non-permissive temperature, PDGF receptor (PDGFR) signalling, assessed by phosphoY(751)-specific antibodies (a docking site for PI3-K), was not detected without PDGF and showed a concentration-dependent PDGF response. Both immunolabeling of PI3-K (p110) and live cell imaging of its product (phosphatidylinositol 3,4,5 tris-phosphate) showed PI3-K recruitment and activation at lamellipodia polarized towards a PDGF gradient. Centrosomes and PDGFR- and Src-bearing endosomes were also oriented towards this gradient. Upon v-Src thermoactivation, (i) Y(751) phosphorylation was moderately induced without PDGF and synergistically increased with PDGF; (ii) PI3-K was recruited and activated all along the plasma membrane without PDGF and did not polarize in response to a PDGF gradient; and (iii) polarization of centrosomes and of PDGFR-bearing endosomes were also abrogated. Thus, PDGF can further increase PDGFR auto-phosphorylation despite strong Src kinase activity, but diffuse downstream activation of PI3-K by Src abrogates cell polarization and chemotaxis: "signalling requires silence".


Subject(s)
Cell Membrane/enzymology , Chemotaxis , Oncogene Protein pp60(v-src)/physiology , Phosphatidylinositol 3-Kinases/metabolism , Platelet-Derived Growth Factor/pharmacology , Receptors, Platelet-Derived Growth Factor/metabolism , Animals , Cell Line , Cell Membrane/metabolism , Centrosome/physiology , Cytoplasm/enzymology , Cytoplasm/metabolism , Cytoskeleton/physiology , Enzyme Activation , Fibroblasts/metabolism , Focal Adhesions/metabolism , Microtubule-Organizing Center/physiology , Proto-Oncogene Proteins c-akt/genetics , Rats , Signal Transduction
17.
Biochem Biophys Res Commun ; 356(2): 512-6, 2007 May 04.
Article in English | MEDLINE | ID: mdl-17367752

ABSTRACT

SWAP-70 is a phosphatidylinositol trisphosphate (PtdIns(3,4,5)P(3)) binding protein, which acts in F-actin rearrangement. The role of SWAP-70 in oncogenic transformation of mouse embryo fibroblasts (MEFs) by v-Src was examined by use of MEFs defective in SWAP-70. v-Src morphologically transformed MEFs lacking SWAP-70, but growth of the transformed cells in culture was slower than that of cells supplemented with exogenous SWAP-70. The v-Src-transformed MEFs deficient in SWAP-70 were unable to grow in soft agar while those expressing SWAP70 readily formed colonies, suggesting that SWAP-70 is required for anchorage independent growth of v-Src transformed MEFs. When transplanted in nude mice, tumors formed by the v-Src transformed SWAP-70(-/-) MEFs were smaller than those formed by cells expressing exogenous SWAP-70. These results suggest that SWAP-70 may be required for oncogenic transformation and contributes to cell growth in MEFs transformed by v-Src.


Subject(s)
Cell Line, Transformed/physiology , DNA-Binding Proteins/physiology , Embryo, Mammalian/cytology , Fibroblasts/physiology , Guanine Nucleotide Exchange Factors/physiology , Nuclear Proteins/physiology , Oncogene Protein pp60(v-src)/physiology , Animals , Cell Transformation, Neoplastic/pathology , Mice , Minor Histocompatibility Antigens , Signal Transduction/physiology
18.
J Cell Physiol ; 210(3): 750-6, 2007 Mar.
Article in English | MEDLINE | ID: mdl-17187346

ABSTRACT

Fibronectin (FN) matrix assembly is an integrin-mediated process that is regulated by both the extracellular environment and intracellular signaling pathways. The activity of Src-family kinases is important for initiation of FN assembly by normal fibroblasts. Here we report that in HT1080 fibrosarcoma cells, Src kinase activity is required not only for the assembly of FN matrix but also for the maintenance of FN matrix fibrils at the cell surface. Dexamethasone-induced FN fibril formation by these cells was completely blocked for at least 24 h when Src-family kinase activity was inhibited by either PP1 or SU6656. Inhibition of Src after significant matrix had already been assembled, resulted in an increased rate of loss of detergent-insoluble FN. Binding of activation-dependent integrin antibodies reveals a role for Src in maintaining integrin activity. The requirement for Src kinase activity appears to depend, in part, on phosphorylation of paxillin at tyrosine 118 (Y118). Phospho-paxillin co-localized with FN fibrils, and overexpression of GFP-paxillin but not of GFP-paxillinY118F enhanced cell-mediated assembly of FN. Our results indicate that Src maintains FN matrix at the cell surface through its effect on integrin activity and paxillin phosphorylation.


Subject(s)
Fibronectins/metabolism , Oncogene Protein pp60(v-src)/physiology , Paxillin/metabolism , Antineoplastic Agents, Hormonal/pharmacology , Cell Line, Transformed , Cell Line, Tumor , Dexamethasone/pharmacology , Fibrosarcoma/pathology , Humans , Integrins/drug effects , Integrins/physiology , Phosphorylation
19.
Comb Chem High Throughput Screen ; 9(9): 711-8, 2006 Nov.
Article in English | MEDLINE | ID: mdl-17100576

ABSTRACT

The conversion of the genomic information produced by the recent sequencing projects into a comprehensive understanding of the human proteome has yet to occur. A new technology that represents a potential bridge between genomics and proteomics is reverse transfection. Reverse transfection cell microarrays are produced by overlaying cDNA arrays with mammalian cells, generating localized clusters of transfected cells with each cluster overexpressing a unique protein. This miniaturized cell-based microarray format affords parallel functional analysis of thousands of cDNA constructs in a high throughput format. In this report we document the development of a co-transfection methodology for reverse transfection applications. The demonstrated high co-transfection efficiency with a "marker" plasmid encoding for GFP enables the identification of transfected cells and eliminates the need for epitope-tagged constructs in cell-based high throughput screening applications using reverse transfection. This co-transfection method was used to study in parallel the structure/function of multiple versions of the v-Src protein using automated fluorescence microscopy. The wild-type v-Src protein and four mutants having insertions or deletions in the SH2 or SH3 domains displayed high levels of tyrosine kinase activity in HEK293T cells. Three other mutated v-Src proteins, including a kinase-dead version, were shown to be defective for tyrosine kinase activity. This reverse co-transfection approach is applicable for high throughput screening of both cDNA libraries and positional scanning recombinant protein libraries.


Subject(s)
Mutation , Oncogene Protein pp60(v-src)/physiology , Transfection , Cell Line , Fluorescent Antibody Technique , Humans , Microscopy, Confocal , Microscopy, Fluorescence , Oncogene Protein pp60(v-src)/genetics , Plasmids
SELECTION OF CITATIONS
SEARCH DETAIL
...