Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 549
Filter
Add more filters

Publication year range
1.
Mol Cell ; 67(6): 947-961.e5, 2017 Sep 21.
Article in English | MEDLINE | ID: mdl-28890336

ABSTRACT

The Hsp90 system in the eukaryotic cytosol is characterized by a cohort of co-chaperones that bind to Hsp90 and affect its function. Although progress has been made regarding the underlying biochemical mechanisms, how co-chaperones influence Hsp90 client proteins in vivo has remained elusive. By investigating the effect of 12 Hsp90 co-chaperones on the activity of different client proteins in yeast, we find that deletion of co-chaperones can have a neutral or negative effect on client activity but can also lead to more active clients. Only a few co-chaperones are active on all clients studied. Closely related clients and even point mutants can depend on different co-chaperones. These effects are direct because differences in client-co-chaperone interactions can be reconstituted in vitro. Interestingly, some co-chaperones affect client conformation in vivo. Thus, co-chaperones adapt the Hsp90 cycle to the requirements of the client proteins, ensuring optimal activation.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Cell Plasticity , HSP90 Heat-Shock Proteins/metabolism , Saccharomyces cerevisiae Proteins/metabolism , Saccharomyces cerevisiae/metabolism , Adaptor Proteins, Signal Transducing/genetics , Genotype , HSP90 Heat-Shock Proteins/genetics , Mutation , Oncogene Protein pp60(v-src)/genetics , Oncogene Protein pp60(v-src)/metabolism , Phenotype , Receptors, Steroid/genetics , Receptors, Steroid/metabolism , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae Proteins/genetics , Signal Transduction
2.
Genes Dev ; 30(7): 798-811, 2016 Apr 01.
Article in English | MEDLINE | ID: mdl-27013234

ABSTRACT

Cell-cell adhesion protein αE-catenin inhibits skin squamous cell carcinoma (SCC) development; however, the mechanisms responsible for this function are not completely understood. We report here that αE-catenin inhibits ß4 integrin-mediated activation of SRC tyrosine kinase.SRCis the first discovered oncogene, but the protein substrate critical for SRC-mediated transformation has not been identified. We found that YAP1, the pivotal effector of the Hippo signaling pathway, is a direct SRC phosphorylation target, and YAP1 phosphorylation at three sites in its transcription activation domain is necessary for SRC-YAP1-mediated transformation. We uncovered a marked increase in this YAP1 phosphorylation in human and mouse SCC tumors with low/negative expression of αE-catenin. We demonstrate that the tumor suppressor function of αE-catenin involves negative regulation of the ß4 integrin-SRC signaling pathway and that SRC-mediated phosphorylation and activation of YAP1 are an alternative to the canonical Hippo signaling pathway that directly connect oncogenic tyrosine kinase signaling with YAP1.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Carcinoma, Squamous Cell/physiopathology , Oncogene Protein pp60(v-src)/metabolism , Phosphoproteins/metabolism , Protein Serine-Threonine Kinases/metabolism , Protein-Tyrosine Kinases/metabolism , Signal Transduction , alpha Catenin/metabolism , Animals , Carcinoma, Squamous Cell/enzymology , Carcinoma, Squamous Cell/genetics , Cell Cycle Proteins , Cell Nucleus/metabolism , Cell Proliferation , Cell Transformation, Neoplastic/genetics , Cells, Cultured , Gene Expression Regulation, Neoplastic , Humans , Keratinocytes/cytology , Keratinocytes/pathology , Mice , Phosphorylation , Protein Transport , YAP-Signaling Proteins
3.
FASEB J ; 35(1): e21242, 2021 01.
Article in English | MEDLINE | ID: mdl-33368671

ABSTRACT

When cells with excess DNA, such as tetraploid cells, undergo cell division, it can contribute to cellular transformation via asymmetrical chromosome segregation-generated genetic diversity. Cell cycle progression of tetraploid cells is suppressed by large tumor suppressor 2 (LATS2) kinase-induced inhibitory phosphorylation of the transcriptional coactivator Yes-associated protein (YAP). We recently reported that the oncogene v-Src induces tetraploidy and promotes cell cycle progression of tetraploid cells by suppressing LATS2 activity. We explore here the mechanism by which v-Src suppresses LATS2 activity and the role of LATS2 in v-Src-expressing cells. LATS2 was directly phosphorylated by v-Src and the proto-oncogene c-Src, resulting in decreased LATS2 kinase activity. This kinase-deficient LATS2 accumulated in a YAP transcriptional activity-dependent manner, and knockdown of either LATS2 or the LATS2-binding partner moesin-ezrin-radixin-like protein (Merlin) accelerated v-Src-induced membrane bleb formation. Upon v-Src expression, the interaction of Merlin with LATS2 was increased possibly due to a decrease in Merlin phosphorylation at Ser518, the dephosphorylation of which is required for the open conformation of Merlin and interaction with LATS2. LATS2 was colocalized with Merlin at the plasma membrane in a manner that depends on the Merlin-binding region of LATS2. The bleb formation in v-Src-expressing and LATS2-knockdown cells was rescued by the reexpression of wild-type or kinase-dead LATS2 but not the LATS2 mutant lacking the Merlin-binding region. These results suggest that the kinase-deficient LATS2 plays a role with Merlin at the plasma membrane in the maintenance of cortical rigidity in v-Src-expressing cells, which may cause tumor suppression.


Subject(s)
Cell Membrane Structures/enzymology , Oncogene Protein pp60(v-src)/metabolism , Protein Serine-Threonine Kinases/metabolism , Tumor Suppressor Proteins/metabolism , Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Signal Transducing/metabolism , Animals , Cell Membrane Structures/genetics , HCT116 Cells , HT29 Cells , HeLa Cells , Humans , Mice , NIH 3T3 Cells , Neurofibromin 2/genetics , Neurofibromin 2/metabolism , Oncogene Protein pp60(v-src)/genetics , Protein Serine-Threonine Kinases/genetics , Proto-Oncogene Mas , Transcription Factors/genetics , Transcription Factors/metabolism , Tumor Suppressor Proteins/genetics , YAP-Signaling Proteins
4.
Cell Mol Life Sci ; 78(23): 7257-7273, 2021 Dec.
Article in English | MEDLINE | ID: mdl-34677645

ABSTRACT

The Hsp70 and Hsp90 molecular chaperone systems are critical regulators of protein homeostasis (proteostasis) in eukaryotes under normal and stressed conditions. The Hsp70 and Hsp90 systems physically and functionally interact to ensure cellular proteostasis. Co-chaperones interact with Hsp70 and Hsp90 to regulate and to promote their molecular chaperone functions. Mammalian Hop, also called Stip1, and its budding yeast ortholog Sti1 are eukaryote-specific co-chaperones, which have been thought to be essential for substrate ("client") transfer from Hsp70 to Hsp90. Substrate transfer is facilitated by the ability of Hop to interact simultaneously with Hsp70 and Hsp90 as part of a ternary complex. Intriguingly, in prokaryotes, which lack a Hop ortholog, the Hsp70 and Hsp90 orthologs interact directly. Recent evidence shows that eukaryotic Hsp70 and Hsp90 can also form a prokaryote-like binary chaperone complex in the absence of Hop, and that this binary complex displays enhanced protein folding and anti-aggregation activities. The canonical Hsp70-Hop-Hsp90 ternary chaperone complex is essential for optimal maturation and stability of a small subset of clients, including the glucocorticoid receptor, the tyrosine kinase v-Src, and the 26S/30S proteasome. Whereas many cancers have increased levels of Hop, the levels of Hop decrease in the aging human brain. Since Hop is not essential in all eukaryotic cells and organisms, tuning Hop levels or activity might be beneficial for the treatment of cancer and neurodegeneration.


Subject(s)
HSP70 Heat-Shock Proteins/metabolism , HSP90 Heat-Shock Proteins/metabolism , Heat-Shock Proteins/metabolism , Neoplasms/pathology , Neurodegenerative Diseases/pathology , Aging/metabolism , Animals , Brain/metabolism , Heat-Shock Proteins/genetics , Humans , Oncogene Protein pp60(v-src)/metabolism , Proteasome Endopeptidase Complex/metabolism , Protein Binding , Protein Folding , Proteostasis/physiology , Receptors, Glucocorticoid/metabolism
5.
J Cell Mol Med ; 25(3): 1677-1687, 2021 02.
Article in English | MEDLINE | ID: mdl-33465289

ABSTRACT

v-Src oncogene causes cell transformation through its strong tyrosine kinase activity. We have revealed that v-Src-mediated cell transformation occurs at a low frequency and it is attributed to mitotic abnormalities-mediated chromosome instability. v-Src directly phosphorylates Tyr-15 of cyclin-dependent kinase 1 (CDK1), thereby causing mitotic slippage and reduction in Eg5 inhibitor cytotoxicity. However, it is not clear whether v-Src modifies cytotoxicities of the other anticancer drugs targeting cell division. In this study, we found that v-Src restores cancer cell viability reduced by various microtubule-targeting agents (MTAs), although v-Src does not alter cytotoxicity of DNA-damaging anticancer drugs. v-Src causes mitotic slippage of MTAs-treated cells, consequently generating proliferating tetraploid cells. We further demonstrate that v-Src also restores cell viability reduced by a polo-like kinase 1 (PLK1) inhibitor. Interestingly, treatment with Aurora kinase inhibitor strongly induces cell death when cells express v-Src. These results suggest that the v-Src modifies cytotoxicities of anticancer drugs targeting cell division. Highly activated Src-induced resistance to MTAs through mitotic slippage might have a risk to enhance the malignancy of cancer cells through the increase in chromosome instability upon chemotherapy using MTAs.


Subject(s)
Antineoplastic Agents/pharmacology , Cell Division/drug effects , Cell Survival/drug effects , Oncogene Protein pp60(v-src)/metabolism , Biomarkers , Cell Cycle Proteins/antagonists & inhibitors , Cell Line, Tumor , Flow Cytometry , Humans , Immunophenotyping , Microtubules/metabolism , Mitosis/drug effects , Mitosis/genetics , Phosphorylation , Protein Kinase Inhibitors/pharmacology , Protein Serine-Threonine Kinases/antagonists & inhibitors , Proto-Oncogene Proteins/antagonists & inhibitors , Tubulin Modulators/pharmacology , Polo-Like Kinase 1
6.
Molecules ; 26(12)2021 Jun 12.
Article in English | MEDLINE | ID: mdl-34204651

ABSTRACT

The driving forces and conformational pathways leading to amphitropic protein-membrane binding and in some cases also to protein misfolding and aggregation is the subject of intensive research. In this study, a chimeric polypeptide, A-Cage-C, derived from α-Lactalbumin is investigated with the aim of elucidating conformational changes promoting interaction with bilayers. From previous studies, it is known that A-Cage-C causes membrane leakages associated with the sporadic formation of amorphous aggregates on solid-supported bilayers. Here we express and purify double-labelled A-Cage-C and prepare partially deuterated bicelles as a membrane mimicking system. We investigate A-Cage-C in the presence and absence of these bicelles at non-binding (pH 7.0) and binding (pH 4.5) conditions. Using in silico analyses, NMR, conformational clustering, and Molecular Dynamics, we provide tentative insights into the conformations of bound and unbound A-Cage-C. The conformation of each state is dynamic and samples a large amount of overlapping conformational space. We identify one of the clusters as likely representing the binding conformation and conclude tentatively that the unfolding around the central W23 segment and its reorientation may be necessary for full intercalation at binding conditions (pH 4.5). We also see evidence for an overall elongation of A-Cage-C in the presence of model bilayers.


Subject(s)
Oncogene Protein pp60(v-src)/chemistry , Peptide Fragments/chemistry , Peptides/chemistry , Lactalbumin/chemistry , Magnetic Resonance Spectroscopy/methods , Membrane Proteins/chemistry , Membrane Proteins/metabolism , Membranes , Molecular Dynamics Simulation , Oncogene Protein pp60(v-src)/metabolism , Peptide Fragments/metabolism , Peptides/metabolism , Protein Binding , Protein Conformation
7.
J Neurosci ; 39(29): 5634-5646, 2019 07 17.
Article in English | MEDLINE | ID: mdl-31092585

ABSTRACT

Addictive behaviors, including relapse, are thought to depend in part on long-lasting drug-induced adaptations in dendritic spine signaling and morphology in the nucleus accumbens (NAc). While the influence of activity-dependent actin remodeling in these phenomena has been studied extensively, the role of microtubules and associated proteins remains poorly understood. We report that pharmacological inhibition of microtubule polymerization in the NAc inhibited locomotor sensitization to cocaine and contextual reward learning. We then investigated the roles of microtubule end-binding protein 3 (EB3) and SRC kinase in the neuronal and behavioral responses to volitionally administered cocaine. In synaptoneurosomal fractions from the NAc of self-administering male rats, the phosphorylation of SRC at an activating site was induced after 1 d of withdrawal, while EB3 levels were increased only after 30 d of withdrawal. Blocking SRC phosphorylation during early withdrawal by virally overexpressing SRCIN1, a negative regulator of SRC activity known to interact with EB3, abolished the incubation of cocaine craving in both male and female rats. Conversely, mimicking the EB3 increase observed after prolonged withdrawal increased the motivation to consume cocaine in male rats. In mice, the overexpression of either EB3 or SRCIN1 increased dendritic spine density and altered the spine morphology of NAc medium spiny neurons. Finally, a cocaine challenge after prolonged withdrawal recapitulated most of the synaptic protein expression profiles observed at early withdrawal. These findings suggest that microtubule-associated signaling proteins such as EB3 cooperate with actin remodeling pathways, notably SRC kinase activity, to establish and maintain long-lasting cellular and behavioral alterations following cocaine self-administration.SIGNIFICANCE STATEMENT Drug-induced morphological restructuring of dendritic spines of nucleus accumbens neurons is thought to be one of the cellular substrates of long-lasting drug-associated memories. The molecular basis of these persistent changes has remained incompletely understood. Here we implicate for the first time microtubule function in this process, together with key players such as microtubule-bound protein EB3 and synaptic SRC phosphorylation. We propose that microtubule and actin remodeling cooperate during withdrawal to maintain the plastic structural changes initially established by cocaine self-administration. This work opens new translational avenues for further characterization of microtubule-associated regulatory molecules as putative drug targets to tackle relapse to drug taking.


Subject(s)
Cocaine/administration & dosage , Locomotion/physiology , Microtubule-Associated Proteins/metabolism , Oncogene Protein pp60(v-src)/metabolism , Substance Withdrawal Syndrome/metabolism , Synapses/metabolism , Animals , Cocaine-Related Disorders/metabolism , Cocaine-Related Disorders/pathology , Female , Locomotion/drug effects , Male , Mice , Mice, Inbred C57BL , Microtubules/drug effects , Microtubules/metabolism , Microtubules/pathology , Phosphorylation/drug effects , Phosphorylation/physiology , Rats , Self Administration , Substance Withdrawal Syndrome/pathology , Synapses/drug effects , Synapses/pathology
8.
Nucleic Acids Res ; 46(17): 8689-8699, 2018 09 28.
Article in English | MEDLINE | ID: mdl-30102385

ABSTRACT

DEAD-box proteins are an essential class of enzymes involved in all stages of RNA metabolism. The study of DEAD-box proteins is challenging in a native setting since they are structurally similar, often essential and display dosage sensitivity. Pharmacological inhibition would be an ideal tool to probe the function of these enzymes. In this work, we describe a chemical genetic strategy for the specific inactivation of individual DEAD-box proteins with small molecule inhibitors using covalent complementarity. We identify a residue of low conservation within the P-loop of the nucleotide-binding site of DEAD-box proteins and show that it can be mutated to cysteine without a substantial loss of enzyme function to generate electrophile-sensitive mutants. We then present a series of small molecules that rapidly and specifically bind and inhibit electrophile-sensitive DEAD-box proteins with high selectivity over the wild-type enzyme. Thus, this approach can be used to systematically generate small molecule-sensitive alleles of DEAD-box proteins, allowing for pharmacological inhibition and functional characterization of members of this enzyme family.


Subject(s)
Adenosine Monophosphate/analogs & derivatives , DEAD Box Protein 58/chemistry , DEAD-box RNA Helicases/chemistry , Oncogene Protein pp60(v-src)/chemistry , Saccharomyces cerevisiae Proteins/chemistry , Acrylamides/chemical synthesis , Acrylamides/metabolism , Acrylates/chemical synthesis , Acrylates/metabolism , Adenosine Monophosphate/metabolism , Amino Acid Motifs , Amino Acid Sequence , Amino Acid Substitution , Binding Sites , Cloning, Molecular , Crotonates/chemical synthesis , Crotonates/metabolism , Crystallography, X-Ray , DEAD Box Protein 58/antagonists & inhibitors , DEAD Box Protein 58/genetics , DEAD Box Protein 58/metabolism , DEAD-box RNA Helicases/antagonists & inhibitors , DEAD-box RNA Helicases/genetics , DEAD-box RNA Helicases/metabolism , Escherichia coli/genetics , Escherichia coli/metabolism , Gene Expression , Humans , Kinetics , Models, Molecular , Oncogene Protein pp60(v-src)/antagonists & inhibitors , Oncogene Protein pp60(v-src)/genetics , Oncogene Protein pp60(v-src)/metabolism , Protein Binding , Protein Conformation, alpha-Helical , Protein Conformation, beta-Strand , Protein Interaction Domains and Motifs , Receptors, Immunologic , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/metabolism , Saccharomyces cerevisiae Proteins/antagonists & inhibitors , Saccharomyces cerevisiae Proteins/genetics , Saccharomyces cerevisiae Proteins/metabolism
9.
Proc Natl Acad Sci U S A ; 114(47): 12495-12500, 2017 11 21.
Article in English | MEDLINE | ID: mdl-29109268

ABSTRACT

The cytoplasmic tyrosine kinase SRC controls cell growth, proliferation, adhesion, and motility. The current view is that SRC acts primarily downstream of cell-surface receptors to control intracellular signaling cascades. Here we reveal that SRC functions in cell-to-cell communication by controlling the biogenesis and the activity of exosomes. Exosomes are viral-like particles from endosomal origin that can reprogram recipient cells. By gain- and loss-of-function studies, we establish that SRC stimulates the secretion of exosomes having promigratory activity on endothelial cells and that syntenin is mandatory for SRC exosomal function. Mechanistically, SRC impacts on syndecan endocytosis and on syntenin-syndecan endosomal budding, upstream of ARF6 small GTPase and its effector phospholipase D2, directly phosphorylating the conserved juxtamembrane DEGSY motif of the syndecan cytosolic domain and syntenin tyrosine 46. Our study uncovers a function of SRC in cell-cell communication, supported by syntenin exosomes, which is likely to contribute to tumor-host interactions.


Subject(s)
Cell Communication/genetics , Exosomes/metabolism , Human Umbilical Vein Endothelial Cells/drug effects , Oncogene Protein pp60(v-src)/genetics , Syntenins/genetics , ADP-Ribosylation Factor 6 , ADP-Ribosylation Factors/genetics , ADP-Ribosylation Factors/metabolism , Amino Acid Motifs , Cell Movement , Cell Proliferation , Culture Media, Conditioned/pharmacology , Endocytosis , Endosomes/metabolism , Gene Expression Regulation , Human Umbilical Vein Endothelial Cells/cytology , Human Umbilical Vein Endothelial Cells/metabolism , Humans , MCF-7 Cells , Oncogene Protein pp60(v-src)/metabolism , Phospholipase D/genetics , Phospholipase D/metabolism , Phosphorylation , Signal Transduction , Syndecans/genetics , Syndecans/metabolism , Syntenins/metabolism
10.
J Biol Chem ; 293(40): 15524-15537, 2018 10 05.
Article in English | MEDLINE | ID: mdl-30135207

ABSTRACT

The nonreceptor tyrosine kinase v-Src is an oncogene first identified in Rous sarcoma virus. The oncogenic effects of v-Src have been intensively studied; however, its effects on chromosomal integrity are not fully understood. Here, using HeLa S3/v-Src cells having inducible v-Src expression, we found that v-Src causes mitotic slippage in addition to cytokinesis failure, even when the spindle assembly checkpoint is not satisfied because of the presence of microtubule-targeting agents. v-Src's effect on mitotic slippage was also observed in cells after a knockdown of C-terminal Src kinase (Csk), a protein-tyrosine kinase that inhibits Src-family kinases and was partially inhibited by PP2, an Src-family kinase inhibitor. Proteomic analysis and in vitro kinase assay revealed that v-Src phosphorylates cyclin-dependent kinase 1 (Cdk1) at Tyr-15. This phosphorylation attenuated Cdk1 kinase activity, resulting in a decrease in the phosphorylation of Cdk1 substrates. Furthermore, v-Src-induced mitotic slippage reduced the sensitivity of the cells to microtubule-targeting agents, and cells that survived the microtubule-targeting agents exhibited polyploidy. These results suggest that v-Src causes mitotic slippage by attenuating Cdk1 kinase activity via direct phosphorylation of Cdk1 at Tyr-15. On the basis of these findings, we propose a model for v-Src-induced oncogenesis, in which v-Src-promoted mitotic slippage due to Cdk1 phosphorylation generates genetic diversity via abnormal cell division of polyploid cells and also increases the tolerance of cancer cells to microtubule-targeting agents.


Subject(s)
Antineoplastic Agents, Phytogenic/pharmacology , CDC2 Protein Kinase/genetics , Microtubules/drug effects , Mitosis/drug effects , Oncogene Protein pp60(v-src)/genetics , Paclitaxel/pharmacology , CDC2 Protein Kinase/antagonists & inhibitors , CDC2 Protein Kinase/metabolism , Cell Proliferation/drug effects , HeLa Cells , Humans , Microtubules/metabolism , Microtubules/ultrastructure , Oncogene Protein pp60(v-src)/antagonists & inhibitors , Oncogene Protein pp60(v-src)/metabolism , Phosphoproteins/genetics , Phosphoproteins/metabolism , Phosphorylation , Polyploidy , Protein Kinase Inhibitors/pharmacology , Pyrimidines/pharmacology , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Time-Lapse Imaging
11.
Genes Cells ; 23(11): 974-981, 2018 Nov.
Article in English | MEDLINE | ID: mdl-30175422

ABSTRACT

At the initial stage of carcinogenesis, transformation occurs in single cells within the epithelium. Recent studies have revealed that the newly emerging transformed cells are often apically eliminated from epithelial tissues. However, the underlying molecular mechanisms of this cancer preventive phenomenon still remain elusive. In this study, we first demonstrate that myosin-II accumulates in Src-transformed cells when they are surrounded by normal epithelial cells. Knock-down of the heavy chains of myosin-II substantially diminishes apical extrusion of Src cells, suggesting that accumulated myosin-II positively regulates the apical elimination of transformed cells. Furthermore, we have identified ß-spectrin as a myosin-II-binding protein under the coculture of normal and Src-transformed epithelial cells. ß-spectrin is also accumulated in Src cells that are surrounded by normal cells, and the ß-spectrin accumulation is regulated by myosin-II. Moreover, knock-down of ß-spectrin significantly suppresses apical extrusion of Src cells. Collectively, these results indicate that accumulation of the myosin-II-spectrin complex plays a positive role in apical extrusion of Src-transformed epithelial cells. Further elucidation of the molecular mechanisms of apical extrusion would lead to the establishment of a novel type of cancer preventive medicine.


Subject(s)
Actin Cytoskeleton/metabolism , Cell Transformation, Neoplastic/pathology , Epithelial Cells/pathology , Myosin Type II/metabolism , Oncogene Protein pp60(v-src)/metabolism , Spectrin/metabolism , Animals , Cell Communication , Cell Transformation, Neoplastic/metabolism , Cells, Cultured , Dogs , Epithelial Cells/metabolism , Signal Transduction
12.
J Biol Chem ; 292(1): 172-184, 2017 Jan 06.
Article in English | MEDLINE | ID: mdl-27872192

ABSTRACT

Tumor invasion into surrounding stromal tissue is a hallmark of high grade, metastatic cancers. Oncogenic transformation of human epithelial cells in culture can be triggered by activation of v-Src kinase, resulting in increased cell motility, invasiveness, and tumorigenicity and provides a valuable model for studying how changes in gene expression cause cancer phenotypes. Here, we show that epithelial cells transformed by activated Src show increased levels of DNA methylation and that the methylation inhibitor 5-azacytidine (5-AzaC) potently blocks the increased cell motility and invasiveness induced by Src activation. A proteomic screen for chromatin regulators acting downstream of activated Src identified the replication-dependent histone chaperone CAF1 as an important factor for Src-mediated increased cell motility and invasion. We show that Src causes a 5-AzaC-sensitive decrease in both mRNA and protein levels of the p150 (CHAF1A) and p60 (CHAF1B), subunits of CAF1. Depletion of CAF1 in untransformed epithelial cells using siRNA was sufficient to recapitulate the increased motility and invasive phenotypes characteristic of transformed cells without activation of Src. Maintaining high levels of CAF1 by exogenous expression suppressed the increased cell motility and invasiveness phenotypes when Src was activated. These data identify a critical role of CAF1 in the dysregulation of cell invasion and motility phenotypes seen in transformed cells and also highlight an important role for epigenetic remodeling through DNA methylation for Src-mediated induction of cancer phenotypes.


Subject(s)
Azacitidine/pharmacology , Breast/pathology , Cell Movement , Cell Transformation, Neoplastic/pathology , Epithelial Cells/pathology , Oncogene Protein pp60(v-src)/metabolism , Transcription Factors/metabolism , Antimetabolites, Antineoplastic/pharmacology , Breast/drug effects , Breast/metabolism , Cell Transformation, Neoplastic/drug effects , Cell Transformation, Neoplastic/metabolism , Cells, Cultured , Chromatin Assembly and Disassembly , Epithelial Cells/drug effects , Epithelial Cells/metabolism , Female , Humans , Mass Spectrometry , Neoplasm Invasiveness , Oncogene Protein pp60(v-src)/genetics , Protein Subunits , Proteomics , Signal Transduction , Transcription Factors/genetics
13.
J Virol ; 91(1)2017 Jan 01.
Article in English | MEDLINE | ID: mdl-27795443

ABSTRACT

The increase in AP-1 activity is a hallmark of cell transformation by tyrosine kinases. Previously, we reported that blocking AP-1 using the c-Jun dominant negative mutant TAM67 induced senescence, adipogenesis, or apoptosis in v-Src-transformed chicken embryo fibroblasts (CEFs) whereas inhibition of JunD by short hairpin RNA (shRNA) specifically induced apoptosis. To investigate the role of AP-1 in Src-mediated transformation, we undertook a gene profiling study to characterize the transcriptomes of v-Src-transformed CEFs expressing either TAM67 or the JunD shRNA. Our study revealed a cluster of 18 probe sets upregulated exclusively in response to AP-1/JunD impairment and v-Src transformation. Four of these probe sets correspond to genes involved in the interferon pathway. One gene in particular, death-associated protein kinase 1 (DAPK1), is a C/EBPß-regulated mediator of apoptosis in gamma interferon (IFN-γ)-induced cell death. Here, we show that inhibition of DAPK1 abrogates cell death in v-Src-transformed cells expressing the JunD shRNA. Chromatin immunoprecipitation data indicated that C/EBPß was recruited to the DAPK1 promoter while the expression of a dominant negative mutant of C/EBPß abrogated the induction of DAPK1 in response to the inhibition of AP-1. In contrast, as determined by chromatin immunoprecipitation (ChIP) assays, JunD was not detected on the DAPK1 promoter under any conditions, suggesting that JunD promotes survival by indirectly antagonizing the expression of DAPK1 in v-Src transformed cells. IMPORTANCE: Transformation by the v-Src oncoprotein causes extensive changes in gene expression in primary cells such as chicken embryo fibroblasts. These changes, determining the properties of transformed cells, are controlled in part at the transcriptional level. Much attention has been devoted to transcription factors such as AP-1 and NF-κB and the control of genes associated with a more aggressive phenotype. In this report, we describe a novel mechanism of action determined by the JunD component of AP-1, a factor enhancing cell survival in v-Src-transformed cells. We show that the loss of JunD results in the aberrant activation of a genetic program leading to cell death. This program requires the activation of the tumor suppressor death-associated protein kinase 1 (DAPK1). Since DAPK1 is phosphorylated and inhibited by v-Src, these results highlight the importance of this kinase and the multiple mechanisms controlled by v-Src to antagonize the tumor suppressor function of DAPK1.


Subject(s)
Death-Associated Protein Kinases/genetics , Fibroblasts/metabolism , Oncogene Protein pp60(v-src)/genetics , Proto-Oncogene Proteins c-jun/genetics , Transcription Factor AP-1/genetics , Animals , Apoptosis/genetics , Base Sequence , CCAAT-Enhancer-Binding Protein-beta/genetics , CCAAT-Enhancer-Binding Protein-beta/metabolism , Cell Survival , Cells, Cultured , Chick Embryo , Chickens , Chromatin Immunoprecipitation , Death-Associated Protein Kinases/metabolism , Fibroblasts/cytology , Gene Expression Profiling , Gene Expression Regulation , Oncogene Protein pp60(v-src)/metabolism , Plasmids/chemistry , Plasmids/metabolism , Protein Binding , Proto-Oncogene Proteins c-jun/antagonists & inhibitors , Proto-Oncogene Proteins c-jun/metabolism , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Signal Transduction , Transcription Factor AP-1/antagonists & inhibitors , Transcription Factor AP-1/metabolism , Transfection
14.
Proc Natl Acad Sci U S A ; 112(25): E3189-98, 2015 Jun 23.
Article in English | MEDLINE | ID: mdl-26056257

ABSTRACT

Hsp90 is a molecular chaperone involved in the activation of numerous client proteins, including many kinases. The most stringent kinase client is the oncogenic kinase v-Src. To elucidate how Hsp90 chaperones kinases, we reconstituted v-Src kinase chaperoning in vitro and show that its activation is ATP-dependent, with the cochaperone Cdc37 increasing the efficiency. Consistent with in vivo results, we find that Hsp90 does not influence the almost identical c-Src kinase. To explain these findings, we designed Src kinase chimeras that gradually transform c-Src into v-Src and show that their Hsp90 dependence correlates with compactness and folding cooperativity. Molecular dynamics simulations and hydrogen/deuterium exchange of Hsp90-dependent Src kinase variants further reveal increased transitions between inactive and active states and exposure of specific kinase regions. Thus, Hsp90 shifts an ensemble of conformations of v-Src toward high activity states that would otherwise be metastable and poorly populated.


Subject(s)
HSP90 Heat-Shock Proteins/metabolism , Oncogene Protein pp60(v-src)/metabolism , Animals , Chickens , Molecular Dynamics Simulation , Oncogene Protein pp60(v-src)/chemistry , Protein Conformation , Recombinant Fusion Proteins/metabolism
15.
J Biol Chem ; 291(30): 15867-80, 2016 07 22.
Article in English | MEDLINE | ID: mdl-27235399

ABSTRACT

Connexin43 (Cx43) assembly and degradation, the regulation of electrical and metabolic coupling, as well as modulating the interaction with other proteins, involve phosphorylation. Here, we identified and characterized the biological significance of a novel tyrosine kinase that phosphorylates Cx43, tyrosine kinase 2 (Tyk2). Activation of Tyk2 led to a decrease in Cx43 gap junction communication by increasing the turnover rate of Cx43 from the plasma membrane. Tyk2 directly phosphorylated Cx43 residues Tyr-247 and Tyr-265, leading to indirect phosphorylation on residues Ser-279/Ser-282 (MAPK) and Ser-368 (PKC). Although this phosphorylation pattern is similar to what has been observed following Src activation, the response caused by Tyk2 occurred when Src was inactive in NRK cells. Knockdown of Tyk2 at the permissive temperature (active v-Src) in LA-25 cells decreased Cx43 phosphorylation, indicating that although activation of Tyk2 and v-Src leads to phosphorylation of the same Cx43CT residues, they are not identical in level at each site. Additionally, angiotensin II activation of Tyk2 increased the intracellular protein level of Cx43 via STAT3. These findings indicate that, like Src, Tyk2 can also inhibit gap junction communication by phosphorylating Cx43.


Subject(s)
Connexin 43/biosynthesis , Gap Junctions/enzymology , Gene Expression Regulation , TYK2 Kinase/metabolism , Animals , Cell Line , Connexin 43/genetics , Gap Junctions/genetics , Oncogene Protein pp60(v-src)/genetics , Oncogene Protein pp60(v-src)/metabolism , Phosphorylation/genetics , Rats , STAT3 Transcription Factor/genetics , STAT3 Transcription Factor/metabolism , TYK2 Kinase/genetics
16.
Neurochem Res ; 42(10): 2850-2860, 2017 Oct.
Article in English | MEDLINE | ID: mdl-28512713

ABSTRACT

Chronic neuroinflammation is a pathological feature of neurodegenerative diseases. Inhibition of microglia-mediated neuroinflammation might be a potential strategy for neurodegeneration. Matairesinol, a dibenzylbutyrolactone plant lignan, presents in a wide variety of foodstuffs. It has been found to possess anti-angiogenic, anti-oxidative, anti-cancer and anti-fungal activities. In the present study, we investigated the anti-neuroinflammation effects of matairesinol on lipopolysaccharide (LPS)-induced BV2 microglia cells and the related molecular mechanisms. The results showed that matairesinol inhibited microglia activation by reducing the production of nitric oxide, the expression of inducible nitric oxide synthase and cyclooxygenase-2 in a concentration-dependent manner (6.25, 12.5, 25 µM). In the molecular signaling pathway, LPS-induced nuclear factor-kappa B (NF-κB) transcriptional activity and translocation into the nucleus were remarkably suppressed by matairesinol through the inhibition of the extracellular signal-regulated kinase (ERK)1/2 signal transduction pathways, but not p38 MAPK or c-jun N-terminal kinase (JNK). Meanwhile, matairesinol also blocked LPS-mediated microglia migration and this was associated with inhibition of LPS-induced Src phosphorylation as well as Src expression in a concentration-dependent manner. Taken together, these results suggest that matairesinol inhibited inflammatory response and migration in LPS-induced BV2 microglia, and the mechanisms may be associated with the NF-κB activation and modulation of Src pathway.


Subject(s)
Furans/pharmacology , Lignans/pharmacology , MAP Kinase Signaling System/drug effects , Microglia/drug effects , NF-kappa B/metabolism , Signal Transduction/drug effects , Animals , Anti-Inflammatory Agents/pharmacology , Lipopolysaccharides/pharmacology , Mice , Mitogen-Activated Protein Kinases/metabolism , NF-kappa B/drug effects , Nitric Oxide/metabolism , Nitric Oxide Synthase Type II/metabolism , Oncogene Protein pp60(v-src)/metabolism , Phosphorylation , Proto-Oncogene Proteins c-akt/metabolism
17.
Mol Cell ; 33(5): 639-53, 2009 Mar 13.
Article in English | MEDLINE | ID: mdl-19285946

ABSTRACT

The Rac activator Tiam1 is required for adherens junction (AJ) maintenance, and its depletion results in AJ disassembly. Conversely, the oncoprotein Src potently induces AJ disassembly and epithelial-mesenchymal transition (EMT). Here, we show that Tiam1 is phosphorylated on Y384 by Src. This occurs predominantly at AJs, is required for Src-induced AJ disassembly and cell migration, and creates a docking site on Tiam1 for Grb2. We find that Tiam1 is associated with ERK. Following recruitment of the Grb2-Sos1 complex, ERK becomes activated and triggers the localized degradation of Tiam1 at AJs, likely involving calpain proteases. Furthermore, we demonstrate that, in human tumors, Y384 phosphorylation positively correlates with Src activity, and total Tiam1 levels are inversely correlated. Thus, our data implicate Tiam1 phosphorylation and consequent degradation in Src-mediated EMT and resultant cell motility and establish a paradigm for regulating local concentrations of Rho-GEFs.


Subject(s)
Adherens Junctions/enzymology , Guanine Nucleotide Exchange Factors/metabolism , Protein Processing, Post-Translational , src-Family Kinases/metabolism , Animals , Calpain/deficiency , Calpain/genetics , Calpain/metabolism , Cell Line , Cell Movement , Cloning, Molecular , Dogs , Extracellular Signal-Regulated MAP Kinases/metabolism , GRB2 Adaptor Protein/metabolism , Guanine Nucleotide Exchange Factors/deficiency , Guanine Nucleotide Exchange Factors/genetics , Humans , MAP Kinase Kinase Kinases/metabolism , Mice , Mice, Knockout , Mutagenesis, Site-Directed , Mutation , Neoplasm Invasiveness , Neoplasms/enzymology , Neoplasms/pathology , Oncogene Protein pp60(v-src)/metabolism , Phosphorylation , Proto-Oncogene Proteins c-myc/metabolism , Proto-Oncogene Proteins c-yes/metabolism , Proto-Oncogene Proteins pp60(c-src)/metabolism , Recombinant Fusion Proteins/metabolism , SOS1 Protein/metabolism , T-Lymphoma Invasion and Metastasis-inducing Protein 1 , Time Factors , Transfection , Tyrosine , src Homology Domains , src-Family Kinases/genetics
18.
EMBO J ; 31(6): 1506-17, 2012 Mar 21.
Article in English | MEDLINE | ID: mdl-22227520

ABSTRACT

Sti1/Hop is a modular protein required for the transfer of client proteins from the Hsp70 to the Hsp90 chaperone system in eukaryotes. It binds Hsp70 and Hsp90 simultaneously via TPR (tetratricopeptide repeat) domains. Sti1/Hop contains three TPR domains (TPR1, TPR2A and TPR2B) and two domains of unknown structure (DP1 and DP2). We show that TPR2A is the high affinity Hsp90-binding site and TPR1 and TPR2B bind Hsp70 with moderate affinity. The DP domains exhibit highly homologous α-helical folds as determined by NMR. These, and especially DP2, are important for client activation in vivo. The core module of Sti1 for Hsp90 inhibition is the TPR2A-TPR2B segment. In the crystal structure, the two TPR domains are connected via a rigid linker orienting their peptide-binding sites in opposite directions and allowing the simultaneous binding of TPR2A to the Hsp90 C-terminal domain and of TPR2B to Hsp70. Both domains also interact with the Hsp90 middle domain. The accessory TPR1-DP1 module may serve as an Hsp70-client delivery system for the TPR2A-TPR2B-DP2 segment, which is required for client activation in vivo.


Subject(s)
HSP90 Heat-Shock Proteins/chemistry , HSP90 Heat-Shock Proteins/metabolism , Heat-Shock Proteins/chemistry , Heat-Shock Proteins/metabolism , Molecular Chaperones/chemistry , Molecular Chaperones/metabolism , Saccharomyces cerevisiae Proteins/chemistry , Saccharomyces cerevisiae Proteins/metabolism , Adenosine Triphosphatases/metabolism , Binding Sites , HSP70 Heat-Shock Proteins/chemistry , HSP70 Heat-Shock Proteins/metabolism , Models, Molecular , Oncogene Protein pp60(v-src)/metabolism , Protein Binding , Protein Structure, Secondary , Protein Structure, Tertiary , Receptors, Glucocorticoid/metabolism , Saccharomyces cerevisiae/metabolism
19.
Clin Exp Pharmacol Physiol ; 43(10): 1004-12, 2016 10.
Article in English | MEDLINE | ID: mdl-27314562

ABSTRACT

Protein tyrosine-phosphatases (PTPs) play important roles in various biological processes. Deregulation in PTP function has been implicated in carcinogenesis and tumour progression in many cancer types. However, the role of protein tyrosine phosphatase receptor type B (PTPRB) in non-small-cell lung cancer (NSCLC) tumorigenesis has not been investigated. Lentiviral vector expressing PTPRB cDNA or shRNA was infected into A549 and H1299 cell lines, followed by cell proliferation, colony formation, soft agar and invasion assays. A549 xenograft mouse model was used to evaluate in vivo function of PTPRB. Quantitative polymerase chain reaction (PCR) was used to measure PTPRB expression in NSCLC patient samples. Kaplan Meier analysis was performed to assess association between PTPRB expression and patient overall survival (OS). Multivariate analysis was performed to evaluate prognostic significance of PTPRB. Overexpression of PTPRB reduced cell proliferation rate, colony formation efficiency, soft agar growth and cell invasion in A549 and H1299 cells, as well as tumour growth rate in A549 xenograft. Knockdown of PTPRB increased Src phosphorylation and cell invasion, which was reversed by Src inhibitor PP2. Additionally, PTPRB was down-regulated in NSCLC patient and was associated with patient OS. PTPRB regulates Src phosphorylation and tumorigenesis in NSCLC. PTPRB may serve as an independent prognostic biomarker for NSCLC patients.


Subject(s)
Carcinoma, Non-Small-Cell Lung/metabolism , Disease Progression , Lung Neoplasms/metabolism , Oncogene Protein pp60(v-src)/metabolism , Receptor-Like Protein Tyrosine Phosphatases, Class 3/physiology , A549 Cells , Adult , Animals , Carcinoma, Non-Small-Cell Lung/mortality , Carcinoma, Non-Small-Cell Lung/pathology , Cell Line, Tumor , Female , Humans , Lung Neoplasms/mortality , Lung Neoplasms/pathology , Male , Mice , Mice, Nude , Middle Aged , Neoplasm Invasiveness/pathology , Phosphorylation/physiology , Survival Rate/trends , Xenograft Model Antitumor Assays/methods
20.
Int J Mol Sci ; 17(6)2016 Jun 02.
Article in English | MEDLINE | ID: mdl-27271602

ABSTRACT

An increase in Src activity is commonly observed in epithelial cancers. Aberrant activation of the kinase activity is associated with malignant progression. However, the mechanisms that underlie the Src-induced malignant progression of cancer are not completely understood. We show here that v-Src, an oncogene that was first identified from a Rous sarcoma virus and a mutant variant of c-Src, leads to an increase in the number of anaphase and telophase cells having chromosome bridges. v-Src increases the number of γH2AX foci, and this increase is inhibited by treatment with PP2, a Src kinase inhibitor. v-Src induces the phosphorylation of KAP1 at Ser824, Chk2 at Thr68, and Chk1 at Ser345, suggesting the activation of the ATM/ATR pathway. Caffeine decreases the number of cells having chromosome bridges at a concentration incapable of inhibiting Chk1 phosphorylation at Ser345. These results suggest that v-Src induces chromosome bridges via generation of DNA damage and the subsequent DNA damage response, possibly by homologous recombination. A chromosome bridge gives rise to the accumulation of DNA damage directly through chromosome breakage and indirectly through cytokinesis failure-induced multinucleation. We propose that v-Src-induced chromosome bridge formation is one of the causes of the v-Src-induced malignant progression of cancer cells.


Subject(s)
Caffeine/pharmacology , Chromosomes/genetics , Chromosomes/metabolism , DNA Damage/drug effects , Oncogene Protein pp60(v-src)/metabolism , Cell Line, Tumor , DNA-Binding Proteins/metabolism , Histones/metabolism , Humans , Phosphorylation
SELECTION OF CITATIONS
SEARCH DETAIL