Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 34
Filter
1.
J Biol Chem ; 291(32): 16787-801, 2016 08 05.
Article in English | MEDLINE | ID: mdl-27281823

ABSTRACT

The α-subunits of hypoxia-inducible factors (HIF1α and HIF2α) promote transcription of genes that regulate glycolysis and cell survival and growth. Sprouty2 (Spry2) is a modulator of receptor tyrosine kinase signaling and inhibits cell proliferation by a number of different mechanisms. Because of the seemingly opposite actions of HIFα subunits and Spry2 on cellular processes, we investigated whether Spry2 regulates the levels of HIF1α and HIF2α proteins. In cell lines from different types of tumors in which the decreased protein levels of Spry2 have been associated with poor prognosis, silencing of Spry2 elevated HIF1α protein levels. Increases in HIF1α and HIF2α protein levels due to silencing of Spry2 also up-regulated HIFα target genes. Using HIF1α as a prototype, we show that Spry2 decreases HIF1α stability and enhances the ubiquitylation of HIF1α by a von Hippel-Lindau protein (pVHL)-dependent mechanism. Spry2 also exists in a complex with HIF1α. Because Spry2 can also associate with pVHL, using a mutant form of Spry2 (3P/3A-Spry2) that binds HIF1α, but not pVHL, we show that WT-Spry2, but not the 3P/3A-Spry2 decreases HIF1α protein levels. In accordance, expression of WT-Spry2, but not 3P/3A-Spry2 results in a decrease in HIF1α-sensitive glucose uptake. Together our data suggest that Spry2 acts as a scaffold to bring more pVHL/associated E3 ligase in proximity of HIF1α and increase its ubiquitylation and degradation. This represents a novel action for Spry2 in modulating biological processes regulated by HIFα subunits.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/metabolism , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Membrane Proteins/metabolism , Transcription, Genetic , Ubiquitination , Basic Helix-Loop-Helix Transcription Factors/genetics , Cell Line, Tumor , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Intracellular Signaling Peptides and Proteins/genetics , Membrane Proteins/genetics , Protein Stability , Ubiquitin-Protein Ligases/genetics , Ubiquitin-Protein Ligases/metabolism , Von Hippel-Lindau Tumor Suppressor Protein/metabolism
2.
Exp Cell Res ; 349(1): 139-151, 2016 Nov 15.
Article in English | MEDLINE | ID: mdl-27737732

ABSTRACT

Earlier studies from our laboratory have identified Anacardic acid (AA) as a potent inhibitor of gelatinases (MMP-2 and 9), which are over-expressed in a wide variety of cancers (Omanakuttan et al., 2012). Disruption of the finely tuned matrix metalloproteinase (MMP) activator/inhibitor balance plays a decisive role in determining the fate of the cell. The present study demonstrates for the first time, that in addition to regulating the expression as well as activity of gelatinases, AA also inhibits the expression of its endogenous activators like MMP-14 and Extracellular Matrix MetalloProteinase Inducer (EMMPRIN) and induces the expression of its endogenous inhibitor, REversion-inducing Cysteine-rich protein with Kazal motifs (RECK). In addition to modulating gelatinases, AA also inhibits the expression of various components of the Epidermal Growth Factor (EGF) pathway like EGF, Protein Kinase B (Akt) and Mitogen-activated protein kinases (MAPK). Furthermore, AA also activates the expression of Sprouty 2 (Spry2), a negative regulator of EGF pathway, and silencing Spry2 results in up-regulation of expression of gelatinases as well as MMP-14. The present study thus elucidates a novel mechanism of action of AA and provides a strong basis for utilizing this molecule as a template for cancer therapeutics.


Subject(s)
Anacardic Acids/pharmacology , Basigin/metabolism , GPI-Linked Proteins/metabolism , Gelatinases/antagonists & inhibitors , Intracellular Signaling Peptides and Proteins/metabolism , Matrix Metalloproteinase 14/metabolism , Membrane Proteins/metabolism , Cell Adhesion/drug effects , Cell Line, Tumor , Cell Movement/drug effects , Epidermal Growth Factor/metabolism , Gelatinases/metabolism , Gene Expression Regulation, Enzymologic/drug effects , Humans , Matrix Metalloproteinase 2/genetics , Matrix Metalloproteinase 2/metabolism , Matrix Metalloproteinase 9/genetics , Matrix Metalloproteinase 9/metabolism , Matrix Metalloproteinase Inhibitors/pharmacology , Models, Biological , Neoplasm Invasiveness , Signal Transduction/drug effects , Tissue Inhibitor of Metalloproteinase-2/metabolism
3.
Mol Pharmacol ; 85(2): 357-67, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24307699

ABSTRACT

Cardiomyocyte apoptosis contributes toward the loss of muscle mass in myocardial pathologies. Previous reports have implicated type I cAMP-dependent protein kinase (PKA) and p90 ribosomal S6 kinase (RSK) in cardiomyocyte apoptosis. However, the precise mechanisms and the isoform of RSK involved in this process remain undefined. Using adult rat ventricular myocytes and mouse-derived cardiac HL-1 cardiomyocytes, we demonstrate that hypoxia/reoxygenation (H/R)-induced apoptosis is accompanied by a decrease in the type I PKA regulatory subunit (PKARIα) and activation of RSK1. As previously described by us for other cell types, in cardiomyocytes, inactive RSK1 also interacts with PKARIα, whereas the active RSK1 interacts with the catalytic subunit of PKA. Additionally, small interfering (siRNA)-mediated silencing of PKARIα or disrupting the RSK1/PKARIα interactions with a small, cell-permeable peptide activates RSK1 and recapitulates the H/R-induced apoptosis. Inhibition of RSK1 or siRNA-mediated silencing of RSK1 attenuates H/R-induced apoptosis, demonstrating the role of RSK1 in cardiomyocyte apoptosis. Furthermore, silencing of RSK1 decreases the H/R-induced phosphorylation of sodium-hydrogen exchanger 1 (NHE1), and inhibition of NHE1 with 5'-N-ethyl-N-isopropyl-amiloride blocks H/R induced apoptosis, indicating the involvement of NHE1 in apoptosis. Overall, our findings demonstrate that H/R-mediated decrease in PKARIα protein levels leads to activation of RSK1, which via phosphorylation of NHE1 induces cardiomyocyte apoptosis.


Subject(s)
Apoptosis , Cyclic AMP-Dependent Protein Kinase RIalpha Subunit/physiology , Myocytes, Cardiac/physiology , Ribosomal Protein S6 Kinases, 90-kDa/physiology , Animals , Cation Transport Proteins/metabolism , Cells, Cultured , Mice , Myocytes, Cardiac/enzymology , Phosphorylation , Rats , Sodium-Hydrogen Exchanger 1 , Sodium-Hydrogen Exchangers/metabolism
5.
J Biol Chem ; 286(49): 42027-42036, 2011 Dec 09.
Article in English | MEDLINE | ID: mdl-22006925

ABSTRACT

Sprouty (Spry) proteins modulate the actions of receptor tyrosine kinases during development and tumorigenesis. Decreases in cellular levels of Spry, especially Sprouty2 (Spry2), have been implicated in the growth and progression of tumors of the breast, prostate, lung, and liver. During development and tumor growth, cells experience hypoxia. Therefore, we investigated how hypoxia modulates the levels of Spry proteins. Hypoxia elevated the levels of all four expressed Spry isoforms in HeLa cells. Amounts of endogenous Spry2 in LS147T and HEP3B cells were also elevated by hypoxia. Using Spry2 as a prototype, we demonstrate that silencing and expression of prolyl hydroxylase domain proteins (PHD1-3) increase and decrease, respectively, the cellular content of Spry2. Spry2 also preferentially interacted with PHD1-3 and von Hippel-Lindau protein (pVHL) during normoxia but not in hypoxia. Additionally, Spry2 is hydroxylated on Pro residues 18, 144, and 160, and substitution of these residues with Ala enhanced stability of Spry2 and abrogated its interactions with pVHL. Silencing of pVHL increased levels of Spry2 by decreasing its ubiquitylation and degradation and thereby augmented the ability of Spry2 to inhibit FGF-elicited activation of ERK1/2. Thus, prolyl hydroxylase mediated hydroxylation and subsequent pVHL-elicited ubiquitylation of Spry2 target it for degradation and, consequently, provide a novel mechanism of regulating growth factor signaling.


Subject(s)
Intracellular Signaling Peptides and Proteins/metabolism , Von Hippel-Lindau Tumor Suppressor Protein/metabolism , Gene Silencing , HEK293 Cells , HeLa Cells , Humans , Hypoxia , Membrane Proteins , Phosphorylation , Procollagen-Proline Dioxygenase/metabolism , Protein Structure, Tertiary , RNA, Small Interfering/metabolism , Receptor Protein-Tyrosine Kinases/metabolism , Signal Transduction , Ubiquitin/chemistry , Ubiquitin-Protein Ligases/metabolism
6.
J Biol Chem ; 285(10): 6970-9, 2010 Mar 05.
Article in English | MEDLINE | ID: mdl-20048145

ABSTRACT

Previously we showed that the inactive form of p90 ribosomal S6 kinase 1 (RSK1) interacts with the regulatory subunit, PKARIalpha, of protein kinase A (PKA), whereas the active RSK1 interacts with the catalytic subunit (PKAc) of PKA. Herein, we demonstrate that the N-terminal kinase domain (NTK) of RSK1 is necessary for interactions with PKARIalpha. Substitution of the activation loop phosphorylation site (Ser-221) in the NTK with the negatively charged Asp residue abrogated the association between RSK1 and PKARIalpha. This explains the lack of an interaction between active RSK1 and PKARIalpha. Full-length RSK1 bound to PKARIalpha with an affinity of 0.8 nm. The NTK domain of RSK1 competed with PKAc for binding to the pseudosubstrate region (amino acids 93-99) of PKARIalpha. Overexpressed RSK1 dissociated PKAc from PKARIalpha, increasing PKAc activity, whereas silencing of RSK1 increased PKAc/PKARIalpha interactions and decreased PKAc activity. Unlike PKAc, which requires Arg-95 and -96 in the pseudosubstrate region of PKARIalpha for their interactions, RSK1/PKARIalpha association requires all four Arg residues (Arg-93-96) in the pseudosubstrate site of PKARIalpha. A peptide (Wt-PS) corresponding to residues 91-99 of PKARIalpha competed for binding of RSK1 with PKARIalpha both in vitro and in intact cells. Furthermore, peptide Wt-PS (but not control peptide Mut-PS), by dissociating RSK1 from PKARIalpha, activated RSK1 in the absence of any growth factors and protected cells from apoptosis. Thus, by competing for binding to the pseudosubstrate region of PKARIalpha, RSK1 regulates PKAc activity in a cAMP-independent manner, and PKARIalpha by associating with RSK1 regulates its activation and its biological functions.


Subject(s)
Catalytic Domain , Cyclic AMP-Dependent Protein Kinase RIalpha Subunit/metabolism , Cyclic AMP-Dependent Protein Kinases/metabolism , Ribosomal Protein S6 Kinases, 90-kDa/metabolism , Animals , Binding Sites , Cell Line , Cyclic AMP/metabolism , Cyclic AMP-Dependent Protein Kinase RIalpha Subunit/genetics , Cyclic AMP-Dependent Protein Kinases/genetics , Humans , Isoenzymes/genetics , Isoenzymes/metabolism , Mice , Protein Binding , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Rats , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Ribosomal Protein S6 Kinases, 90-kDa/genetics
7.
J Biol Chem ; 285(1): 255-64, 2010 Jan 01.
Article in English | MEDLINE | ID: mdl-19864419

ABSTRACT

Sprouty (Spry) proteins are important regulators of receptor tyrosine kinase signaling in development and disease. Alterations in cellular Spry content have been associated with certain forms of cancers and also in cardiovascular diseases. Thus, understanding the mechanisms that regulate cellular Spry levels are important. Herein, we demonstrate that Spry1 and Spry2, but not Spry3 or Spry4, associate with the HECT domain family E3 ubiquitin ligase, Nedd4. The Spry2/Nedd4 association involves the WW domains of Nedd4 and requires phosphorylation of the Mnk2 kinase sites, Ser(112) and Ser(121), on Spry2. The phospho-Ser(112/121) region on Spry2 that binds WW domains of Nedd4 is a novel non-canonical WW domain binding region that does not contain Pro residues after phospho-Ser. Endogenous and overexpressed Nedd4 polyubiquitinate Spry2 via Lys(48) on ubiquitin and decrease its stability. Silencing of endogenous Nedd4 increased the cellular Spry2 content and attenuated fibroblast growth factor-elicited ERK1/2 activation that was reversed when elevations in Spry2 levels were prevented by Spry2-specific small interfering RNA. Mnk2 silencing decreased Spry2-Nedd4 interactions and also augmented the ability of Spry2 to inhibit fibroblast growth factor signaling. This is the first report demonstrating the regulation of cellular Spry content and its ability to modulate receptor tyrosine kinase signaling by a HECT domain-containing E3 ubiquitin ligase.


Subject(s)
Endosomal Sorting Complexes Required for Transport/chemistry , Endosomal Sorting Complexes Required for Transport/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Ubiquitin-Protein Ligases/chemistry , Ubiquitin-Protein Ligases/metabolism , Ubiquitination , Animals , Enzyme Activation , Extracellular Signal-Regulated MAP Kinases/metabolism , Fibroblast Growth Factors/metabolism , Gene Silencing , Humans , Membrane Proteins , Nedd4 Ubiquitin Protein Ligases , Phosphorylation , Phosphoserine/metabolism , Protein Binding , Protein Serine-Threonine Kinases/metabolism , Protein Stability , Protein Structure, Tertiary , Protein Transport , Rats , Signal Transduction
8.
Am J Physiol Heart Circ Physiol ; 300(2): H459-67, 2011 Feb.
Article in English | MEDLINE | ID: mdl-21076020

ABSTRACT

Angiogenesis is regulated by signals received by receptor tyrosine kinases such as vascular endothelial growth factor receptors. Mammalian Sprouty (Spry) proteins are known to function by specifically antagonizing the activation of the mitogen-activated protein kinase signaling pathway by receptor tyrosine kinases, a pathway known to promote angiogenesis. To examine the role of Spry2 in the regulation of angiogenesis during wound repair, we used a model of murine dermal wound healing. Full-thickness excisional wounds (3 mm) were made on the dorsum of anesthetized adult female FVB mice. Samples were harvested at multiple time points postwounding and analyzed using real-time RT-PCR, Western blot analysis, and immunofluorescent histochemistry. Spry2 mRNA and protein levels in the wound bed increased significantly during the resolving phases of healing, coincident with the onset of vascular regression in this wound model. In another experiment, intracellular levels of Spry2 or its dominant-negative mutant (Y55F) were elevated by a topical application to the wounds of controlled-release gel containing cell permeable, transactivator of transcription-tagged Spry2, Spry2Y55F, or green fluorescent protein (as control). Wound samples were analyzed for vascularity using CD31 immunofluorescent histochemistry as well as for total and phospho-Erk1/2 protein content. The treatment of wounds with Spry2 resulted in a significant decrease in vascularity and a reduced abundance of phospho-Erk1/2 compared with wounds treated with the green fluorescent protein control. In contrast, the wounds treated with the dominant-negative Spry2Y55F exhibited a moderate increase in vascularity and elevated phospho-Erk1/2 content. These results indicate that endogenous Spry2 functions to downregulate angiogenesis in the healing murine skin wound, potentially by inhibiting the mitogen-activated protein kinase signaling pathway.


Subject(s)
Membrane Proteins/genetics , Membrane Proteins/physiology , Neovascularization, Physiologic/genetics , Neovascularization, Physiologic/physiology , Wound Healing/genetics , Wound Healing/physiology , Adaptor Proteins, Signal Transducing , Animals , Blotting, Western , Cell Membrane Permeability , Cells, Cultured , Endothelial Cells/physiology , Female , Fluorescent Antibody Technique , Intracellular Signaling Peptides and Proteins , Mice , Mitogen-Activated Protein Kinases/metabolism , Protein Serine-Threonine Kinases , RNA, Messenger/biosynthesis , RNA, Messenger/isolation & purification , Recombinant Proteins/pharmacology , Regional Blood Flow/physiology , Reverse Transcriptase Polymerase Chain Reaction , Trans-Activators/genetics , Wounds and Injuries/physiopathology
9.
J Biol Chem ; 284(48): 33070-8, 2009 Nov 27.
Article in English | MEDLINE | ID: mdl-19808666

ABSTRACT

Previously, we reported that the catalytic subunit of cAMP-dependent protein kinase (PKAc) binds to the active p90 ribosomal S6 kinase 1 (RSK1) (Chaturvedi, D., Poppleton, H. M., Stringfield, T., Barbier, A., and Patel, T. B. (2006) Mol. Cell. Biol. 26, 4586-4600). Herein, by overexpressing hemagglutinin-tagged RSK1 fragments in HeLa cells we have identified the region of RSK1 that is responsible for the interaction with PKAc. PKAc bound to the last 13 amino acids of RSK1, which overlaps the Erk1/2 docking site. This interaction between PKAc and RSK1 required the phosphorylation of Ser-732 in the C terminus of RSK1. Depending upon its phosphorylation status, RSK1 switched interactions between Erk1/2 and PKAc. In addition, a peptide corresponding to the last 13 amino acids of RSK1 with substitution of Ser-732 with Glu (peptide E), but not Ala (peptide A), decreased interactions between endogenous active RSK1 and PKAc. RSK1 attenuated the ability of cAMP to activate PKA in vitro and this modulation was abrogated by peptide E, but not by peptide A. Similarly, in intact cells, cAMP-mediated phosphorylation of Bcl-xL/Bcl-2-associated death promoter on Ser-115, the PKA site, was reduced when RSK1 was activated by epidermal growth factor, and this effect was blocked by peptide E, but not by peptide A. These findings demonstrate that interactions between endogenous RSK1 and PKAc in intact cells regulate the ability of cAMP to activate PKA and identify a novel mechanism by which PKA activity is regulated by the Erk1/2 pathway.


Subject(s)
Amino Acids/metabolism , Cyclic AMP-Dependent Protein Kinases/metabolism , Ribosomal Protein S6 Kinases, 90-kDa/metabolism , Amino Acids/genetics , Animals , Binding Sites , Blotting, Western , Catalytic Domain , Cell Line , Cyclic AMP/pharmacology , Cyclic AMP-Dependent Protein Kinases/genetics , Enzyme Activation/drug effects , HeLa Cells , Humans , Immunoprecipitation , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Mutation , Phosphorylation , Protein Binding , RNA Interference , Ribosomal Protein S6 Kinases, 90-kDa/genetics , Serine/genetics , Serine/metabolism , Threonine/genetics , Threonine/metabolism , Transfection
10.
Mol Pharmacol ; 76(4): 679-91, 2009 Oct.
Article in English | MEDLINE | ID: mdl-19570949

ABSTRACT

Receptor tyrosine kinase (RTK) signaling is spatially and temporally regulated by a number of positive and negative regulatory mechanisms. These regulatory mechanisms control the amplitude and duration of the signals initiated at the cell surface to have a normal or aberrant biological outcome in development and disease, respectively. In the past decade, the Sprouty (Spry) family of proteins has been identified as modulators of RTK signaling in normal development and disease. This review summarizes recent advances concerning the biological activities modulated by Spry family proteins, their interactions with signaling proteins, and their involvement in cardiovascular diseases and cancer. The diversity of mechanisms in the regulation of Spry expression and activity in cell systems emphasizes the crucial role of Spry proteins in development and growth across the animal kingdom.


Subject(s)
Proteins/physiology , Animals , Disease , Humans , Protein Binding , Proteins/metabolism , Receptor Protein-Tyrosine Kinases/metabolism , Signal Transduction/physiology
11.
Mol Cell Biol ; 26(12): 4586-600, 2006 Jun.
Article in English | MEDLINE | ID: mdl-16738324

ABSTRACT

Cyclic AMP (cAMP)-dependent protein kinase (PKA) and ribosomal S6 kinase 1 (RSK1) share several cellular proteins as substrates. However, to date no other similarities between the two kinases or interactions between them have been reported. Here, we describe novel interactions between subunits of PKA and RSK1 that are dependent upon the activation state of RSK1 and determine its subcellular distribution and biological actions. Inactive RSK1 interacts with the type I regulatory subunit (RI) of PKA. Conversely, active RSK1 interacts with the catalytic subunit of PKA (PKAc). Binding of RSK1 to RI decreases the interactions between RI and PKAc, while the binding of active RSK1 to PKAc increases interactions between PKAc and RI and decreases the ability of cAMP to stimulate PKA. The RSK1/PKA subunit interactions ensure the colocalization of RSK1 with A-kinase PKA anchoring proteins (AKAPs). Disruption of the interactions between PKA and AKAPs decreases the nuclear accumulation of active RSK1 and, thus, increases its cytosolic content. This subcellular redistribution of active RSK1 is manifested by increased phosphorylation of its cytosolic substrates tuberous sclerosis complex 2 and BAD by epidermal growth factor along with decreased cellular apoptosis.


Subject(s)
Cyclic AMP-Dependent Protein Kinases/metabolism , Ribosomal Protein S6 Kinases, 90-kDa/metabolism , A Kinase Anchor Proteins , Adaptor Proteins, Signal Transducing/metabolism , Animals , Cell Line , Cell Nucleus/enzymology , Cyclic AMP/pharmacology , Cyclic AMP-Dependent Protein Kinases/chemistry , Enzyme Activation/drug effects , Epidermal Growth Factor/pharmacology , HeLa Cells , Humans , Mice , Multiprotein Complexes , Phosphorylation , Protein Binding , Protein Subunits , Ribosomal Protein S6 Kinases, 90-kDa/chemistry , Signal Transduction/drug effects , Subcellular Fractions/enzymology
12.
Biol Psychiatry ; 62(5): 505-12, 2007 Sep 01.
Article in English | MEDLINE | ID: mdl-17336937

ABSTRACT

BACKGROUND: Reductions in cell number are found within the medial prefrontal cortex (PFC) in major depression and bipolar disorder, conditions for which electroconvulsive therapy (ECT) is a highly effective treatment. We investigated whether electroconvulsive seizure (ECS) in rats stimulates cellular proliferation in the PFC immediately and four weeks after the treatments. In parallel, we examined if ECS also alters the expression of Sprouty2 (SPRY2), an inhibitor of cell proliferation. METHODS: Sprague-Dawley rats received 10 days of ECS treatments and bromodeoxyuridine (BrdU) injections. After a four week survival period, we estimated the density and number of BrdU-, proliferating cell nuclear antigen (PCNA)-, and SPRY2-immunoreactive cells in the medial (infralimbic) PFC (ILPFC). We also determined the percentage of BrdU-labeled cells that were immunoreactive for markers specific to oligodendrocytes, astrocytes, endothelial cells and neurons. RESULTS: ECS dramatically enhanced the proliferation of new cells in the infralimbic PFC, and this effect persisted four weeks following the treatments. The percentage of new cells expressing oligodendrocyte precursor cell markers increased slightly following ECS. In contrast, ECS dramatically reduced the number of cells expressing SPRY2. CONCLUSIONS: ECS stimulates long-lasting increases in glial proliferation within the ILPFC. ECS also decreases SPRY2 expression in the same region, an effect that might contribute to increased glial proliferation.


Subject(s)
Cell Proliferation/radiation effects , Electroshock , Nerve Tissue Proteins/metabolism , Neuroglia/pathology , Prefrontal Cortex/metabolism , Seizures/etiology , Analysis of Variance , Animals , Antigens/metabolism , Bromodeoxyuridine/metabolism , Cell Count/methods , Gene Expression Regulation/physiology , Gene Expression Regulation/radiation effects , Male , Nerve Tissue Proteins/genetics , Neurons/metabolism , Neurons/pathology , Prefrontal Cortex/pathology , Prefrontal Cortex/physiopathology , Prefrontal Cortex/radiation effects , Proteoglycans/metabolism , Rats , Rats, Sprague-Dawley , Seizures/metabolism , Seizures/pathology
13.
PLoS One ; 12(2): e0171616, 2017.
Article in English | MEDLINE | ID: mdl-28196140

ABSTRACT

Receptor Tyrosine Kinase (RTK) signaling plays a major role in tumorigenesis and normal development. Sprouty2 (Spry2) attenuates RTK signaling and inhibits processes such as angiogenesis, cell proliferation, migration and survival, which are all upregulated in tumors. Indeed in cancers of the liver, lung, prostate and breast, Spry2 protein levels are markedly decreased correlating with poor patient prognosis and shorter survival. Thus, it is important to understand how expression of Spry2 is regulated. While prior studies have focused on the post-translation regulation of Spry2, very few studies have focused on the transcriptional regulation of SPRY2 gene. Here, we demonstrate that in the human hepatoma cell line, Hep3B, the transcription of SPRY2 is inhibited by the transcription regulating hypoxia inducible factors (HIFs). HIFs are composed of an oxygen regulated alpha subunit (HIF1α or HIF2α) and a beta subunit (HIF1ß). Intriguingly, silencing of HIF1α and HIF2α elevates SPRY2 mRNA and protein levels suggesting HIFs reduce the transcription of the SPRY2 promoter. In silico analysis identified ten hypoxia response elements (HREs) in the proximal promoter and first intron of SPRY2. Using chromatin immunoprecipitation (ChIP), we show that HIF1α/2α bind near the putative HREs in the proximal promoter and intron of SPRY2. Our studies demonstrated that not only is the SPRY2 promoter methylated, but silencing HIF1α/2α reduced the methylation. ChIP assays also showed DNA methyltransferase1 (DNMT1) binding to the proximal promoter and first intron of SPRY2 and silencing HIF1α/2α decreased this association. Additionally, silencing of DNMT1 mimicked the HIF1α/2α silencing-mediated increase in SPRY2 mRNA and protein. While simultaneous silencing of HIF1α/2α and DNMT1 increased SPRY2 mRNA a little more, the increase was not additive suggesting a common mechanism by which DNMT1 and HIF1α/2α regulate SPRY2 transcription. Together these data suggest that the transcription of SPRY2 is inhibited by HIFs, in part, via DNMT1- mediated methylation.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/genetics , Gene Expression Regulation, Neoplastic , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Intracellular Signaling Peptides and Proteins/genetics , Membrane Proteins/genetics , Transcriptional Activation , Basic Helix-Loop-Helix Transcription Factors/metabolism , Blotting, Western , Cell Hypoxia , Cell Line, Tumor , Chromatin Immunoprecipitation , DNA (Cytosine-5-)-Methyltransferase 1 , DNA (Cytosine-5-)-Methyltransferases/genetics , DNA (Cytosine-5-)-Methyltransferases/metabolism , DNA Methylation , HEK293 Cells , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Membrane Proteins/metabolism , Promoter Regions, Genetic/genetics , Protein Binding , RNA Interference , Response Elements/genetics , Reverse Transcriptase Polymerase Chain Reaction
14.
Oncotarget ; 8(49): 86799-86815, 2017 Oct 17.
Article in English | MEDLINE | ID: mdl-29156836

ABSTRACT

BRCA1 is an important player in the DNA damage response signaling, and its deficiency results in genomic instability. A complete loss or significantly reduced BRCA1 protein expression is often found in sporadic breast cancer cases despite the absence of genetic or epigenetic aberrations, suggesting the existence of other regulatory mechanisms controlling BRCA1 protein expression. Herein, we demonstrate that Fyn-related kinase (Frk)/Rak plays an important role in maintaining genomic stability, possibly in part through positively regulating BRCA1 protein stability and function via tyrosine phosphorylation on BRCA1 Tyr1552. In addition, Rak deficiency confers cellular sensitivity to DNA damaging agents and poly(ADP-ribose) polymerase (PARP) inhibitors. Overall, our findings highlight a critical role of Rak in the maintenance of genomic stability, at least in part, through protecting BRCA1 and provide novel treatment strategies for patients with breast tumors lacking Rak.

15.
Arterioscler Thromb Vasc Biol ; 25(3): 533-8, 2005 Mar.
Article in English | MEDLINE | ID: mdl-15653567

ABSTRACT

OBJECTIVE: To determine whether the human sprouty 2 (hSPRY2) protein, an inhibitor of receptor tyrosine kinase actions, regulates vascular smooth muscle cell (VSMC) proliferation, migration, and neointima formation in injured carotid artery. METHODS AND RESULTS: The hSPRY2 protein or green fluorescent protein (GFP; control) was transduced into VSMCs by placing an N-terminal TAT epitope on the proteins. The transduction of TAT-tagged hSPRY2 (TAT-hSPRY2) but not TAT-GFP inhibited the ability of serum and different growth factors to stimulate migration of VSMCs. Likewise, TAT-hSPRY2 also inhibited VSMC proliferation in response to serum. The hSPRY2 microtubule association (amino acids 123-177) and membrane translocation (amino acids 178-194) domains were necessary for the biological actions of hSPRY2. In the rat carotid artery injury model, exposure of the injured vessel for 1 hour to TAT-hSPRY2, but not TAT-GFP, markedly inhibited growth of the neointima over the 28-day postangioplasty period as well as VSMC proliferation. The exogenously applied TAT-hSPRY2 was retained in the carotid arteries for at least 3 days after injury, and endogenous SPRY2 expression was maximized around day 14 after injury. The latter is perhaps a compensatory mechanism to regulate neointima formation. CONCLUSIONS: We conclude that TAT-tagged proteins are efficiently transduced into VSMCs in vitro and in vivo, that hSPRY2 inhibits growth and migration of VSMCs, and that this protein can decrease neointimal growth after blood vessel injury.


Subject(s)
Carotid Artery Injuries/pathology , Carotid Artery Injuries/therapy , Genetic Therapy , Muscle, Smooth, Vascular/physiology , Proteins/genetics , Animals , Carotid Artery Injuries/physiopathology , Cell Division/physiology , Cell Movement/physiology , Cells, Cultured , Gene Expression , Humans , Intracellular Signaling Peptides and Proteins , Membrane Proteins , Muscle, Smooth, Vascular/pathology , Proteins/physiology , Rats , Receptor Protein-Tyrosine Kinases/metabolism , Transduction, Genetic , Tunica Intima/pathology , Tunica Intima/physiology
16.
Methods Mol Biol ; 327: 49-59, 2006.
Article in English | MEDLINE | ID: mdl-16780212

ABSTRACT

Besides stimulating the mitogen-activated protein kinase, phospholipase Cgamma, and phosphatidylinositol 3-kinase cascades, in certain tissues and cells such as the heart, partotid gland, and luteal cells, activation of the epidermal growth factor (EGF) receptor also stimulates second-messenger systems that involve the heterotrimeric G proteins. For instance, in the heart EGF increases contractility and heart rate by elevating cellular cyclic adenosine monophosphate (cAMP) levels. This is the result of EGF-elicited activation of adenylyl cyclase via the stimulatory guanosine 5'-triphosphate (GTP)-binding protein Gs. In this context, the single transmembrane EGF receptor acts like a heptahelical G protein-coupled receptor. Here we have described the methods used to study interactions between the EGF receptor and heterotrimeric G proteins. Moreover, we have also described how the stoichiometry of EGF receptor association with the alpha subunit of Gs can be monitored in vitro. Several other single transmembrane receptors and proteins can also activate heterotrimeric G proteins, and, therefore, the methodologies described in this chapter can be adapted to other systems.


Subject(s)
Adenylyl Cyclases/chemistry , Adenylyl Cyclases/metabolism , ErbB Receptors/metabolism , GTP-Binding Protein alpha Subunits, Gs/metabolism , Animals , Cattle , GTP Phosphohydrolases/metabolism , GTP-Binding Protein alpha Subunits, Gs/isolation & purification , GTP-Binding Protein beta Subunits/isolation & purification , GTP-Binding Protein gamma Subunits/isolation & purification , Guanosine 5'-O-(3-Thiotriphosphate)/metabolism , Mice , Protein Binding
17.
Methods Mol Biol ; 327: 1-24, 2006.
Article in English | MEDLINE | ID: mdl-16780209

ABSTRACT

Since the isolation of epidermal growth factor (EGF) from mouse submaxillary glands in the early 1950s by Cohen and coworkers, this growth factor has been shown to have various effects on numerous cellular systems. The biological and physiological role that EGF plays during development and in adult animals led to the identification of its receptor (EGFR) as well as the other members of the EGF family of growth factors and their receptors. In this chapter we provide a historical overview of the discovery of EGF, identification of the other members of EGF family, early studies on the actions of EGF, as well as the discovery and structural characterization of its receptor. Further, we have reviewed the transactivation of the EGFR by agonists for G protein-coupled receptors (GPCRs) and other extracellular stimuli unrelated to EGF-like ligands. Finally, an overview of the role of the EGFR family members in various diseases, including different forms of cancer, is provided.


Subject(s)
ErbB Receptors/metabolism , Animals , Cytokines/metabolism , ErbB Receptors/chemistry , ErbB Receptors/genetics , Growth Hormone/metabolism , Integrins/metabolism , Oncogene Protein pp60(v-src)/metabolism , Receptors, G-Protein-Coupled/metabolism , Transcriptional Activation/genetics
18.
PLoS One ; 10(3): e0120693, 2015.
Article in English | MEDLINE | ID: mdl-25822989

ABSTRACT

Both the development and relief of stress-related psychiatric conditions such as major depression (MD) and post-traumatic stress disorder (PTSD) have been linked to neuroplastic changes in the brain. One such change involves the birth of new neurons (neurogenesis), which occurs throughout adulthood within discrete areas of the mammalian brain, including the dorsal hippocampus (HIP). Stress can trigger MD and PTSD in humans, and there is considerable evidence that it can decrease HIP neurogenesis in laboratory animals. In contrast, antidepressant treatments increase HIP neurogenesis, and their efficacy is eliminated by ablation of this process. These findings have led to the working hypothesis that HIP neurogenesis serves as a biomarker of neuroplasticity and stress resistance. Here we report that local alterations in the expression of Sprouty2 (SPRY2), an intracellular inhibitor of growth factor function, produces profound effects on both HIP neurogenesis and behaviors that reflect sensitivity to stressors. Viral vector-mediated disruption of endogenous Sprouty2 function (via a dominant negative construct) within the dorsal HIP of adult rats stimulates neurogenesis and produces signs of stress resilience including enhanced extinction of conditioned fear. Conversely, viral vector-mediated elevation of SPRY2 expression intensifies the behavioral consequences of stress. Studies of these manipulations in HIP primary cultures indicate that SPRY2 negatively regulates fibroblast growth factor-2 (FGF2), which has been previously shown to produce antidepressant- and anxiolytic-like effects via actions in the HIP. Our findings strengthen the relationship between HIP plasticity and stress responsiveness, and identify a specific intracellular pathway that could be targeted to study and treat stress-related disorders.


Subject(s)
Hippocampus/metabolism , Nerve Tissue Proteins/metabolism , Neurogenesis/physiology , Stress, Psychological/metabolism , Animals , Depression/metabolism , Depression/physiopathology , Depressive Disorder, Major/metabolism , Depressive Disorder, Major/physiopathology , Fibroblast Growth Factor 2/metabolism , Hippocampus/physiopathology , Male , Neuronal Plasticity/physiology , Neurons/metabolism , Neurons/physiology , Rats , Rats, Sprague-Dawley , Stress Disorders, Post-Traumatic/metabolism , Stress Disorders, Post-Traumatic/physiopathology , Stress, Psychological/physiopathology
19.
Cancer Res ; 75(20): 4372-83, 2015 Oct 15.
Article in English | MEDLINE | ID: mdl-26282169

ABSTRACT

Non-small cell lung cancers (NSCLC) that have developed resistance to EGF receptor (EGFR) tyrosine kinase inhibitor (TKI), including gefitinib and erlotinib, are clinically linked to an epithelial-to-mesenchymal transition (EMT) phenotype. Here, we examined whether modulating EMT maintains the responsiveness of EGFR-mutated NSCLCs to EGFR TKI therapy. Using human NSCLC cell lines harboring mutated EGFR and a transgenic mouse model of lung cancer driven by mutant EGFR (EGFR-Del19-T790M), we demonstrate that EGFR inhibition induces TGFß secretion followed by SMAD pathway activation, an event that promotes EMT. Chronic exposure of EGFR-mutated NSCLC cells to TGFß was sufficient to induce EMT and resistance to EGFR TKI treatment. Furthermore, NSCLC HCC4006 cells with acquired resistance to gefitinib were characterized by a mesenchymal phenotype and displayed a higher prevalence of the EGFR T790M mutated allele. Notably, combined inhibition of EGFR and the TGFß receptor in HCC4006 cells prevented EMT but was not sufficient to prevent acquired gefitinib resistance because of an increased emergence of the EGFR T790M allele compared with cells treated with gefitinib alone. Conversely, another independent NSCLC cell line, PC9, reproducibly developed EGFR T790M mutations as the primary mechanism underlying EGFR TKI resistance, even though the prevalence of the mutant allele was lower than that in HCC4006 cells. Thus, our findings underscore heterogeneity within NSCLC cells lines harboring EGFR kinase domain mutations that give rise to divergent resistance mechanisms in response to treatment and anticipate the complexity of EMT suppression as a therapeutic strategy.


Subject(s)
Antineoplastic Agents/pharmacology , Carcinoma, Non-Small-Cell Lung/genetics , Drug Resistance, Neoplasm/genetics , Lung Neoplasms/genetics , Mutation , Protein Kinase Inhibitors/pharmacology , Animals , Carcinoma, Non-Small-Cell Lung/metabolism , Carcinoma, Non-Small-Cell Lung/pathology , Cell Line, Tumor , Cluster Analysis , Epithelial-Mesenchymal Transition/drug effects , Epithelial-Mesenchymal Transition/genetics , Gefitinib , Gene Expression Profiling , Humans , Lung Neoplasms/metabolism , Lung Neoplasms/pathology , Mice , Phenotype , Quinazolines/pharmacology , Receptors, Transforming Growth Factor beta/metabolism , Signal Transduction/drug effects , Smad Proteins/metabolism , Transforming Growth Factor beta1/metabolism , Transforming Growth Factor beta1/pharmacology
20.
Brain Res Dev Brain Res ; 136(1): 35-42, 2002 May 30.
Article in English | MEDLINE | ID: mdl-12036515

ABSTRACT

Recently, human protein associated with MYC, PAM, has been cloned and characterized as a large protein that interacts with the transcriptional-activating domain of Myc. The regional expression pattern of PAM in brains has not been yet been defined. Expression patterns of PAM in both rat and mouse brains were examined by using in situ hybridization. Here, we demonstrate that PAM mRNA is highly expressed in specific anatomical regions including hippocampus, dentate gyrus and cerebellum. In these areas, PAM mRNA is restricted to pyramidal cells of hippocampus and granule cells of dentate gyrus and cerebellum. During development, PAM mRNA expression is differentially regulated. It is turned on after birth and up-regulated during the first postnatal 2 weeks. Thereafter, PAM mRNA expression remains elevated into adulthood. The regional distribution of PAM in brain is similar to that observed for several adenylyl cyclase isoforms such as type I isoform. However, no obvious alterations of PAM mRNA expression are detected in brains of mice deficient in type I or type 8 or type 1 and type 8 isoforms of adenylyl cyclase. Thus, adenylyl cyclase does not appear to alter the expression of PAM.


Subject(s)
Adaptor Proteins, Signal Transducing , Carrier Proteins/genetics , Cerebellum/growth & development , Dentate Gyrus/growth & development , Gene Expression Regulation, Developmental , Mixed Function Oxygenases , Adenylyl Cyclases/metabolism , Animals , Cerebellum/cytology , Cerebellum/physiology , Dentate Gyrus/cytology , Dentate Gyrus/physiology , In Situ Hybridization , Mice , Mice, Inbred C57BL , Mice, Inbred ICR , Mice, Knockout , Mitosis/physiology , Neurons/cytology , RNA, Messenger/analysis , Rats , Rats, Long-Evans , Ubiquitin-Protein Ligases
SELECTION OF CITATIONS
SEARCH DETAIL