Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 333
Filter
Add more filters










Publication year range
2.
Curr Mol Med ; 16(1): 83-90, 2016.
Article in English | MEDLINE | ID: mdl-26695692

ABSTRACT

Pancreatic ductal adenocarcinoma (PDAC) is one of the most potent and perilous diseases known, with a median survival rate of 3-5 months due to the combination of only advanced stage diagnosis and ineffective therapeutic options. Metformin (1,1-Dimethylbiguanide hydrochloride), the leading drug used for type 2 diabetes mellitus, emerges as a potential therapy for PDAC and other human cancers. Metformin exerts its anticancer action via a variety of adenosine monophosphate (AMP)-activated protein kinase (AMPK)- dependent and/or AMPK-independent mechanisms. We present data here showing that metformin downregulated pancreatic transcription factor pancreatic duodenal homeobox-1 (PDX-1), suggesting a potential novel mechanism by which metformin exerts its anticancer action. Metformin inhibited PDX-1 expression at both protein and mRNA levels and PDX-1 transactivity as well in PDAC cells. Extracellular signal-regulated kinase (ERK) was identified as a PDX-1-interacting protein by antibody array screening in GFP-PDX-1 stable HEK293 cells. Co-transfection of ERK1 with PDX-1 resulted in an enhanced PDX-1 expression in HEK293 cells in a dose-dependent manner. Immunoprecipitation/Western blotting analysis confirmed the ERK-PDX-1 interaction in PANC-1 cells stimulated by epidermal growth factor (EGF). EGF induced an enhanced PDX-1 expression in PANC-1 cells and this stimulation was inhibited by MEK inhibitor PD0325901. Metformin inhibited EGF-stimulated PDX-1 expression with an accompanied inhibition of ERK kinase activation in PANC- 1 cells. Taken together, our studies show that PDX-1 is a potential novel target for metformin in PDAC cells and that metformin may exert its anticancer action in PDAC by down-regulating PDX-1 via a mechanism involving inhibition of ERK signaling.


Subject(s)
Adenocarcinoma/drug therapy , Carcinoma, Pancreatic Ductal/drug therapy , Homeodomain Proteins/metabolism , MAP Kinase Signaling System/drug effects , Metformin/pharmacology , Pancreatic Neoplasms/drug therapy , Trans-Activators/metabolism , Adenocarcinoma/metabolism , Carcinoma, Pancreatic Ductal/metabolism , Cell Line , Cell Line, Tumor , Diabetes Mellitus, Type 2/metabolism , Down-Regulation/drug effects , Epidermal Growth Factor/metabolism , Extracellular Signal-Regulated MAP Kinases , Genes, Homeobox/drug effects , HEK293 Cells , Humans , Pancreas/drug effects , Pancreas/metabolism , Pancreatic Neoplasms/metabolism , Protein Kinase Inhibitors/pharmacology , Signal Transduction/drug effects , Pancreatic Neoplasms
3.
Am J Transplant ; 14(4): 806-19, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24580843

ABSTRACT

Antibody (Ab) crosslinking of HLA I molecules on the surface of endothelial cells triggers proliferative and pro-survival intracellular signaling, which is implicated in the process of chronic allograft rejection, also known as transplant vasculopathy (TV). The purpose of this study was to investigate the role of mammalian target of rapamycin (mTOR) in HLA I Ab-induced signaling cascades. Everolimus provides a tool to establish how the mTOR signal network regulates HLA I-mediated migration, proliferation and survival. We found that everolimus inhibits mTOR complex 1 (mTORC1) by disassociating Raptor from mTOR, thereby preventing class I-induced phosphorylation of mTOR, p70S6K, S6RP and 4E-BP1, and resultant class I-stimulated cell migration and proliferation. Furthermore, we found that everolimus inhibits class I-mediated mTORC2 activation (1) by disassociating Rictor and Sin1 from mTOR; (2) by preventing class I-stimulated Akt phosphorylation and (3) by preventing class I-mediated ERK phosphorylation. These results suggest that everolimus is more effective than sirolimus at antagonizing both mTORC1 and mTORC2, the latter of which is critical in endothelial cell functional changes leading to TV in solid organ transplantation after HLA I crosslinking. Our findings point to a potential therapeutic effect of everolimus in prevention of chronic Ab-mediated rejection.


Subject(s)
Antibodies/pharmacology , Aorta/immunology , Cell Movement/drug effects , Cell Proliferation/drug effects , Endothelium, Vascular/immunology , Histocompatibility Antigens Class I/immunology , Sirolimus/analogs & derivatives , Sirolimus/pharmacology , Antibodies/immunology , Aorta/cytology , Aorta/metabolism , Blotting, Western , Cells, Cultured , Endothelium, Vascular/cytology , Endothelium, Vascular/metabolism , Everolimus , Humans , Immunoprecipitation , Immunosuppressive Agents/pharmacology , Mechanistic Target of Rapamycin Complex 1 , Mechanistic Target of Rapamycin Complex 2 , Multiprotein Complexes/metabolism , Signal Transduction/drug effects , TOR Serine-Threonine Kinases/metabolism , Wound Healing
4.
Am J Transplant ; 13(2): 299-311, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23279566

ABSTRACT

Donor-specific HLA antibodies significantly lower allograft survival, but as yet there are no satisfactory therapies for prevention of antibody-mediated rejection. Intracapillary macrophage infiltration is a hallmark of antibody-mediated rejection, and macrophages are important in both acute and chronic rejection. The purpose of this study was to investigate the Fc-independent effect of HLA I antibodies on endothelial cell activation, leading to monocyte recruitment. We used an in vitro model to assess monocyte binding to endothelial cells in response to HLA I antibodies. We confirmed our results in a mouse model of antibody-mediated rejection, in which B6.RAG1(-/-) recipients of BALB/c cardiac allografts were passively transferred with donor-specific MHC I antibodies. Our findings demonstrate that HLA I antibodies rapidly increase intracellular calcium and endothelial presentation of P-selectin, which supports monocyte binding. In the experimental model, donor-specific MHC I antibodies significantly increased macrophage accumulation in the allograft. Concurrent administration of rPSGL-1-Ig abolished antibody-induced monocyte infiltration in the allograft, but had little effect on antibody-induced endothelial injury. Our data suggest that antagonism of P-selectin may ameliorate accumulation of macrophages in the allograft during antibody-mediated rejection.


Subject(s)
Antibodies/immunology , Graft Rejection/immunology , Histocompatibility Antigens Class I/immunology , Monocytes/cytology , P-Selectin/antagonists & inhibitors , Animals , Antibodies, Monoclonal/chemistry , Aorta/cytology , Calcium/metabolism , Cells, Cultured , Endothelial Cells/cytology , Exocytosis , Heart Transplantation/methods , Humans , Immunization, Passive , Leukocyte Common Antigens/metabolism , Macrophages/cytology , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Transplantation, Homologous/immunology
5.
Am J Physiol Gastrointest Liver Physiol ; 294(5): G1245-56, 2008 May.
Article in English | MEDLINE | ID: mdl-18308856

ABSTRACT

Tachykinins, acting through NK(3) receptors (NK(3)R), contribute to excitatory transmission to intrinsic primary afferent neurons (IPANs) of the small intestine. Although this transmission is dependent on protein kinase C (PKC), its maintenance could depend on protein kinase D (PKD), a downstream target of PKC. Here we show that PKD1/2-immunoreactivity occurred exclusively in IPANs of the guinea pig ileum, demonstrated by double staining with the IPAN marker NeuN. PKCepsilon was also colocalized with PKD1/2 in IPANs. PKCepsilon and PKD1/2 trafficking was studied in enteric neurons within whole mounts of the ileal wall. In untreated preparations, PKCepsilon and PKD1/2 were cytosolic and no signal for activated (phosphorylated) PKD was detected. The NK(3)R agonist senktide evoked a transient translocation of PKCepsilon and PKD1/2 from the cytosol to the plasma membrane and induced PKD1/2 phosphorylation at the plasma membrane. PKCepsilon translocation was maximal at 10 s and returned to the cytosol within 2 min. Phosphorylated-PKD1/2 was detected at the plasma membrane within 15 s and translocated to the cytosol by 2 min, where it remained active up to 30 min after NK(3)R stimulation. PKD1/2 activation was reduced by a PKCepsilon inhibitor and prevented by NK(3)R inhibition. NK(3)R-mediated PKCepsilon and PKD activation was confirmed in HEK293 cells transiently expressing NK(3)R and green fluorescent protein-tagged PKCepsilon, PKD1, PKD2, or PKD3. Senktide caused membrane translocation and activation of kinases within 30 s. After 15 min, phosphorylated PKD had returned to the cytosol. PKD activation was confirmed through Western blotting. Thus stimulation of NK(3)R activates PKCepsilon and PKD in sequence, and sequential activation of these kinases may account for rapid and prolonged modulation of IPAN function.


Subject(s)
Myenteric Plexus/physiology , Protein Kinase C-epsilon/metabolism , Protein Kinase C/metabolism , Receptors, Neurokinin-3/physiology , Acetates/pharmacology , Animals , Cell Line , Diterpenes/pharmacology , Female , Guinea Pigs , Humans , Ileum/innervation , Kinetics , Male , Myenteric Plexus/drug effects , Neurons, Afferent/drug effects , Neurons, Afferent/physiology , Peptide Fragments/pharmacology , Phorbol 12,13-Dibutyrate/pharmacology , Phosphorylation/drug effects , Protein Kinase C/genetics , Protein Kinase C-epsilon/antagonists & inhibitors , Protein Kinase C-epsilon/genetics , Protein Kinase D2 , Protein Kinases/genetics , Protein Kinases/metabolism , Protein Transport/drug effects , Quinolines/pharmacology , Receptors, G-Protein-Coupled/agonists , Receptors, Neurokinin-3/agonists , Receptors, Neurokinin-3/antagonists & inhibitors , Substance P/analogs & derivatives , Substance P/pharmacology , Transfection
6.
J Cell Physiol ; 189(3): 291-305, 2001 Dec.
Article in English | MEDLINE | ID: mdl-11748587

ABSTRACT

In order to develop a model system for identifying signaling pathways and cell cycle events involved in gastrin-mediated mitogenesis, we have used high efficiency retroviral-mediated transfection of cholecystokinin (CCK)(B)/gastrin receptor into Swiss 3T3 cells. The retrovirally-transfected CCK(B)/gastrin receptor binds 125I-CCK-8 with high affinity (Kd = 1.1 nM) and is functionally coupled to intracellular signaling pathways including rapid and transient increase in Ca2+ fluxes, protein kinase C-dependent protein kinase D activation, and MEK-dependent ERK1/2 activation. In the presence of insulin, CCK-8 or gastrin induced a 66.5 +/- 8.8-fold (mean +/- SEM, n = 24 in eight independent experiments) increase in cellular DNA synthesis, reaching a level similar to that achieved by stimulation with a saturating concentration of fresh serum, and much greater than the response to each agonist added alone. CCK-8 also induced a striking increase in the expression of cyclins D1, D3, and E and hyperphosphorylation of Rb acting synergistically with insulin. Similar effects were observed when CCK(B)/gastrin receptor was activated in the presence of EGF or bombesin. Our results demonstrate that activation of CCK(B)/gastrin receptor retrovirally-transfected into Swiss 3T3 induces a potent synergistic effect on DNA synthesis, accumulation of cyclins D1, D3, and E and hyperphosphorylation of Rb in combination with insulin, EGF, or bombesin. Thus, the CCK(B)/gastrin receptor transfected into Swiss 3T3 cells provides a novel model system to elucidate mitogenic signal transduction pathways and cell cycle events activated via this receptor.


Subject(s)
Cyclin D1/biosynthesis , Cyclin E/biosynthesis , Cyclins/biosynthesis , Receptors, Cholecystokinin/physiology , 3T3 Cells , Animals , Calcium/metabolism , Cell Cycle , Cyclin D3 , DNA/biosynthesis , Drug Synergism , Genetic Vectors , Insulin/pharmacology , Mice , Mitogen-Activated Protein Kinases/metabolism , Protein Kinase C/metabolism , Receptor, Cholecystokinin B , Receptors, Cholecystokinin/genetics , Retinoblastoma Protein/metabolism , Retroviridae/genetics , Sincalide/metabolism , Sincalide/pharmacology , Transfection
7.
J Biol Chem ; 276(52): 49228-35, 2001 Dec 28.
Article in English | MEDLINE | ID: mdl-11641411

ABSTRACT

Protein kinase D (PKD)/protein kinase C mu is a serine/threonine protein kinase activated by growth factors, antigen-receptor engagement, and G protein-coupled receptor (GPCR) agonists via a phosphorylation-dependent mechanism that requires protein kinase C (PKC) activity. In order to investigate the dynamic mechanisms associated with GPCR signaling, the intracellular distribution of PKD was analyzed in live cells by imaging fluorescent protein-tagged PKD and in fixed cells by immunocytochemistry. We found that PKD shuttled between the cytoplasm and the nucleus in both fibroblasts and epithelial cells. Cell stimulation with mitogenic GPCR agonists that activate PKD induced a transient nuclear accumulation of PKD that was prevented by inhibiting PKC activity. The nuclear import of PKD requires its cys2 domain in conjunction with a nuclear import receptor, while its nuclear export requires its pleckstrin homology domain and a competent Crm1-dependent nuclear export pathway. This study thus characterizes the regulated nuclear transport of a signaling molecule in response to mitogenic GPCR agonists and positions PKD as a serine kinase whose kinase activity and intracellular localization is coordinated by PKC.


Subject(s)
Active Transport, Cell Nucleus , Cell Nucleus/metabolism , GTP-Binding Proteins/metabolism , Protein Kinase C/metabolism , Receptors, Cell Surface/metabolism , Recombinant Fusion Proteins/metabolism , 3T3 Cells , Actins/genetics , Actins/metabolism , Animals , Bombesin/pharmacology , Cytoplasm/metabolism , Green Fluorescent Proteins , Immunohistochemistry , Luminescent Proteins/metabolism , Mice , Protein Structure, Tertiary , Vasopressins/pharmacology , Red Fluorescent Protein
8.
Infect Immun ; 69(11): 6839-45, 2001 Nov.
Article in English | MEDLINE | ID: mdl-11598057

ABSTRACT

Cytotoxic necrotizing factor 1 (CNF) is a toxin produced by some isolates of Escherichia coli that cause extraintestinal infections. CNF can initiate signaling pathways that are mediated by the Rho family of small GTPases through a covalent modification that results in constitutive activation. In addition to regulating the assembly of actin stress fibers and focal adhesion complexes, RhoA can also regulate gene expression at the level of transcription. Here we demonstrate for the first time, by using a luciferase-based reporter system, that the transcription of cyclooxygenase-2 (COX-2) is strongly upregulated in NIH 3T3 fibroblasts treated with CNF and that this effect is dependent upon the activation of RhoA by the toxin. Subsequent protein tyrosine phosphorylation events modulate the induction, but the transcription signal is not mediated by Rho-associated kinase (p160/ROCK) and so must rely upon another effector that is activated by RhoA. CNF therefore induces COX-2 expression via a RhoA-dependent signaling pathway that diverges from the pathway that regulates cytoskeletal rearrangements in response to RhoA activation.


Subject(s)
Bacterial Toxins/pharmacology , Cytotoxins/pharmacology , Escherichia coli Proteins , Escherichia coli , Gene Expression Regulation, Enzymologic/drug effects , Isoenzymes/genetics , Prostaglandin-Endoperoxide Synthases/genetics , rhoA GTP-Binding Protein/metabolism , 3T3 Cells , Animals , Cyclooxygenase 2 , Cytochalasin D/pharmacology , Escherichia coli/chemistry , Intracellular Signaling Peptides and Proteins , Mice , Phosphorylation , Protein Serine-Threonine Kinases/metabolism , Transcription, Genetic , Transcriptional Activation , Tyrosine/metabolism , rho-Associated Kinases
9.
Biochem Biophys Res Commun ; 287(1): 21-6, 2001 Sep 14.
Article in English | MEDLINE | ID: mdl-11549247

ABSTRACT

Protein kinase D (PKD)/protein kinase Cmu is a serine/threonine protein kinase that has been localized in the cytosol and in several intracellular compartments including Golgi, mitochondria and plasma membrane. Using real time imaging of fluorescent protein (GFP)-tagged PKD, we have found that the accumulation of PKD in the Golgi compartment, following a temperature shift from 37 to 20 degrees C, was mediated by the cysteine-rich domain (CRD) of PKD. The CRD of PKD also mediates its interaction with the plasma membrane, further supporting the conclusion that the CRD of PKD may act as a subcellular localization signal.


Subject(s)
Cysteine/metabolism , Golgi Apparatus/metabolism , Protein Kinase C/metabolism , 3T3 Cells , Animals , Catalysis , Mice , Protein Kinase C/chemistry , Protein Structure, Tertiary , Subcellular Fractions
10.
Am J Physiol Cell Physiol ; 281(3): C886-98, 2001 Sep.
Article in English | MEDLINE | ID: mdl-11502566

ABSTRACT

We examined the role of epidermal growth factor (EGF) receptor (EGFR) tyrosine kinase activation in G protein-coupled receptor (GPCR) agonist-induced mitogenesis in Swiss 3T3 and Rat-1 cells. Addition of EGFR tyrosine kinase inhibitors (e.g., tyrphostin AG-1478) abrogated bombesin-induced extracellular signal-regulated kinase (ERK) activation in Rat-1 cells but not in Swiss 3T3 cells, indicating the importance of cell context in determining the role of EGFR in ERK activation. In striking contrast, treatment with tyrphostin AG-1478 markedly (~70%) inhibited DNA synthesis induced by bombesin in both Swiss 3T3 and Rat-1 cells. Similar inhibition of bombesin-induced DNA synthesis in Swiss 3T3 cells was obtained using four structurally different inhibitors of EGFR tyrosine kinase. Furthermore, kinetic analysis indicates that EGFR function is necessary for bombesin-induced mitogenesis in mid-late G(1) in both Swiss 3T3 and Rat-1 cells. Our results indicate that EGFR kinase activity is necessary in mid-late G(1) for promoting the accumulation of cyclins D1 and E and implicate EGFR function in the coupling of GPCR signaling to the activation of the cell cycle.


Subject(s)
Bombesin/pharmacology , Bradykinin/pharmacology , Cell Cycle/physiology , ErbB Receptors/physiology , Mitogen-Activated Protein Kinases/metabolism , 3T3 Cells , Animals , Cell Cycle/drug effects , Cell Division/drug effects , Cell Line , Cyclin D1/metabolism , Cyclin E/metabolism , DNA/biosynthesis , Enzyme Activation , Enzyme Inhibitors/pharmacology , G1 Phase , Kinetics , Mice , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3 , Quinazolines , Rats , Receptors, Bombesin/genetics , Receptors, Bombesin/physiology , Recombinant Proteins/metabolism , Transfection , Tyrphostins/pharmacology
11.
J Biol Chem ; 276(42): 38619-27, 2001 Oct 19.
Article in English | MEDLINE | ID: mdl-11507098

ABSTRACT

Protein kinase D (PKD/PKCmu) immunoprecipitated from COS-7 cells transiently transfected with either a constitutively active mutant of Rho (RhoQ63L) or the Rho-specific guanine nucleotide exchange factor pOnco-Lbc (Lbc) exhibited a marked increase in basal activity. Addition of aluminum fluoride to cells co-transfected with PKD and wild type Galpha(13) also induced PKD activation. Co-transfection of Clostridium botulinum C3 toxin blocked activation of PKD by RhoQ63L, Lbc, or aluminum fluoride-stimulated Galpha(13). Treatment with the protein kinase C inhibitors GF I or Ro 31-8220 prevented the increase in PKD activity induced by RhoQ63L, Lbc, or aluminum fluoride-stimulated Galpha(13). PKD activation in response to Galpha(13) signaling was also completely prevented by mutation of Ser-744 and Ser-748 to Ala in the kinase activation loop of PKD. Co-expression of C. botulinum C3 toxin and a COOH-terminal fragment of Galpha(q) that acts in a dominant-negative fashion blocked PKD activation in response to agonist stimulation of bombesin receptor. Expression of the COOH-terminal region of Galpha(13) also attenuated PKD activation in response to bombesin receptor stimulation. Our results show that Galpha(13) contributes to PKD activation through a Rho- and protein kinase C-dependent signaling pathway and indicate that PKD activation is mediated by both Galpha(q) and Galpha(13) in response to bombesin receptor stimulation.


Subject(s)
Heterotrimeric GTP-Binding Proteins/chemistry , Heterotrimeric GTP-Binding Proteins/metabolism , Protein Kinase C/metabolism , Signal Transduction , rho GTP-Binding Proteins/metabolism , A Kinase Anchor Proteins , Adaptor Proteins, Signal Transducing , Alanine/chemistry , Aluminum Compounds/pharmacology , Animals , COS Cells , DNA, Complementary/metabolism , Enzyme Activation , Fluorides/pharmacology , GTP-Binding Protein alpha Subunits, G12-G13 , Genes, Dominant , Green Fluorescent Proteins , Indoles/pharmacology , Luminescent Proteins/metabolism , Minor Histocompatibility Antigens , Models, Biological , Mutation , Precipitin Tests , Protein Binding , Protein Kinase C/chemistry , Protein Structure, Tertiary , Proto-Oncogene Proteins/metabolism , Receptors, Bombesin/metabolism , Recombinant Fusion Proteins/metabolism , Serine/chemistry , Transfection
12.
J Biol Chem ; 276(43): 40298-305, 2001 Oct 26.
Article in English | MEDLINE | ID: mdl-11514571

ABSTRACT

We examined whether protein kinase D (PKD) overexpression in Swiss 3T3 cells potentiates the proliferative response to either the G protein-coupled receptor agonists bombesin and vasopressin or the biologically active phorbol ester phorbol 12,13-dibutyrate (PDBu). In order to generate Swiss 3T3 cells stably overexpressing PKD, cultures of these cells were infected with retrovirus encoding murine PKD and green fluorescent protein (GFP) expressed as two separate proteins translated from the same mRNA. GFP was used as a marker for selection of PKD-positive cells. PKD overexpressed in Swiss 3T3 cells was dramatically activated by cell treatment with bombesin or PDBu as judged by in vitro kinase autophosphorylation assays and exogenous substrate phosphorylation. Concomitantly, these stimuli induced PKD phosphorylation at Ser(744), Ser(748), and Ser(916). PKD activation and phosphorylation were prevented by exposure of the cells to protein kinase C-specific inhibitors. Addition of bombesin, vasopressin, or PDBu to cultures of Swiss 3T3 cells overexpressing PKD induced a striking increase in DNA synthesis and cell number compared with cultures of Swiss 3T3-GFP cells. In contrast, stimulation of DNA synthesis in response to epidermal growth factor, which acts via protein kinase C/PKD-independent pathways, was not enhanced. Our results demonstrate that overexpression of PKD selectively potentiates mitogenesis induced by bombesin, vasopressin, or PDBu in Swiss 3T3 cells.


Subject(s)
Bombesin/pharmacology , Cell Cycle Proteins/metabolism , Phorbol 12,13-Dibutyrate/pharmacology , Protein Kinase C/metabolism , Vasopressins/pharmacology , 3T3 Cells , Animals , Cell Cycle Proteins/genetics , Cell Division , DNA/biosynthesis , Mice , Neuropeptides/pharmacology , Protein Kinase C/genetics , Receptors, Cell Surface/agonists , Recombinant Proteins/biosynthesis
13.
Infect Immun ; 69(9): 5931-5, 2001 Sep.
Article in English | MEDLINE | ID: mdl-11500476

ABSTRACT

Cytotoxic necrotizing factor 1 and Pasteurella multocida toxin induced dose- and time-dependent increases in focal adhesion kinase (FAK) Tyr397 phosphorylation in Swiss 3T3 cells. FAK autophosphorylation was sensitive to inhibitors of p160/ROCK and coincided with the formation of stable complexes between FAK and Src family members.


Subject(s)
Bacterial Proteins , Bacterial Toxins/pharmacology , Cytotoxins/pharmacology , Escherichia coli Proteins , Escherichia coli/metabolism , Pasteurella multocida/metabolism , Protein-Tyrosine Kinases/metabolism , 3T3 Cells , Animals , Focal Adhesion Kinase 1 , Focal Adhesion Protein-Tyrosine Kinases , Mice , Phosphorylation , Protein-Tyrosine Kinases/antagonists & inhibitors , Tyrosine , src-Family Kinases/metabolism
14.
Exp Cell Res ; 266(2): 292-302, 2001 Jun 10.
Article in English | MEDLINE | ID: mdl-11399057

ABSTRACT

A rapid increase in tyrosine phosphorylation of focal adhesion kinase (FAK), paxillin, and Crk-associated substrate (CAS) are prominent early events triggered by many G protein-coupled receptors (GPCRs), but the mechanisms involved remain unclear. Here, we examined whether the Rho-associated protein serine/threonine kinase family (ROCK) is a critical Rho effector in the pathway that links GPCR activation to the tyrosine phosphorylation of FAK, CAS, and paxillin. Treatment of Swiss 3T3 cells with Y-27632, a preferential inhibitor of ROCK, dramatically inhibited the formation of actin stress fibers, the assembly of focal contacts, and the increase in tyrosine phosphorylation of FAK and paxillin induced by bombesin in these cells. Surprisingly, we found that treatment with Y-27632 did not produce any detectable effect on bombesin-elicited CAS tyrosine phosphorylation even at the highest concentrations of Y-27632 tested. HA-1077, a preferential inhibitor of ROCK activity structurally unrelated to Y-27632, also attenuated the increase in the tyrosine phosphorylation of FAK and paxillin but did not affect the tyrosine phosphorylation of CAS induced by bombesin in Swiss 3T3 cells. The results demonstrate that ROCK-dependent tyrosine phosphorylation of FAK and paxillin can be dissociated from a ROCK-independent pathway leading to tyrosine phosphorylation of CAS.


Subject(s)
1-(5-Isoquinolinesulfonyl)-2-Methylpiperazine/analogs & derivatives , Amides/pharmacology , Bombesin/pharmacology , Cytoskeletal Proteins/metabolism , Phosphoproteins/metabolism , Protein Serine-Threonine Kinases/antagonists & inhibitors , Protein-Tyrosine Kinases/metabolism , Proteins , Pyridines/pharmacology , 1-(5-Isoquinolinesulfonyl)-2-Methylpiperazine/pharmacology , 3T3 Cells , Animals , Crk-Associated Substrate Protein , Enzyme Inhibitors/pharmacology , Focal Adhesion Kinase 1 , Focal Adhesion Protein-Tyrosine Kinases , Focal Adhesions/drug effects , Focal Adhesions/ultrastructure , Intracellular Signaling Peptides and Proteins , Lysophospholipids/pharmacology , Mice , Paxillin , Phosphorylation/drug effects , Phosphotyrosine/metabolism , Platelet-Derived Growth Factor/pharmacology , Protein Serine-Threonine Kinases/metabolism , Retinoblastoma-Like Protein p130 , Stress Fibers/drug effects , Stress Fibers/ultrastructure , rho-Associated Kinases
15.
Biochem Biophys Res Commun ; 282(2): 404-8, 2001 Mar 30.
Article in English | MEDLINE | ID: mdl-11401472

ABSTRACT

Phosphorylation of c-Jun at Ser 63/73 by the c-Jun N-terminal kinase (JNK) potentiates the transactivation function of c-Jun. Protein kinase D (PKD), a downstream effector of protein kinase C (PKC), has been implicated in the attenuation of epidermal growth factor (EGF)-induced activation of JNK. In order to determine whether activated PKD is sufficient to modulate the EGF-JNK-c-Jun pathway, we have developed a cellular model system, utilizing human embryonic kidney cells (HEK 293), in which stably transfected, constitutively active or kinase dead mutants of PKD can be inducibly expressed by the insect hormone, ecdysone. Induced expression of constitutively active, but not kinase dead PKD, suppressed EGF stimulated c-Jun phosphorylation at Ser 63, demonstrating that activated PKD is sufficient to suppress c-Jun phosphorylation. This is the first demonstration that PKD modulates phosphorylation of the proto-oncogene c-Jun at a site critical for its ability to mediate cell proliferation and differentiation.


Subject(s)
Epidermal Growth Factor/pharmacology , Protein Kinase C/metabolism , Proto-Oncogene Proteins c-jun/metabolism , Animals , COS Cells , Cell Line , Ecdysone/pharmacology , Humans , JNK Mitogen-Activated Protein Kinases , Mitogen-Activated Protein Kinases/metabolism , Mutation , Phosphorylation , Protein Kinase C/genetics , Proto-Oncogene Mas , Proto-Oncogene Proteins c-jun/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Serine/metabolism , Transfection
16.
J Biol Chem ; 276(35): 32606-15, 2001 Aug 31.
Article in English | MEDLINE | ID: mdl-11410586

ABSTRACT

The importance of activation loop phosphorylation in the regulation of protein kinase D (PKD/protein kinase C (PKC) mu) activity has become controversial. In order to clarify the mechanism(s) of PKD activation, we developed a novel phosphospecific antibody recognizing phosphorylated Ser(748) in PKD (pS748). Western blot analysis with the pS748 antibody, carried out with a variety of PKD forms and in a variety of cell types including full-length PKD transfected in COS-7 and HEK 293 cells, a green fluorescent protein-PKD fusion protein transfected in either Swiss 3T3 fibroblasts or Madin-Darby canine kidney epithelial cells, and endogenous PKD expressed in A20 lymphocytes and Rat-1 fibroblasts, indicated that Ser(748) phosphorylation was absent from unstimulated cells. In contrast, dramatic increases in Ser(748) phosphorylation were induced by phorbol esters, bombesin, or cross-linking of B lymphocyte antigen receptors or by cotransfection with active PKCepsilon or PKCeta. Western analysis using a second phosphospecific antibody, which primarily recognizes PKD phosphorylated at Ser(744), revealed that Ser(744) phosphorylation accompanies Ser(748) phosphorylation during PKD activation in vivo. Ser(744)/Ser(748) phosphorylation requires PKC but not PKD activity, indicative of transphosphorylation. Our results provide new experimental evidence indicating that activation loop phosphorylation at Ser(744) and Ser(748) occurs during PKD activation in vivo and support the notion of a PKC-PKD phosphorylation cascade.


Subject(s)
Protein Kinase C/chemistry , Protein Kinase C/metabolism , Serine , 3T3 Cells , Amino Acid Sequence , Amino Acid Substitution , Animals , Antibodies , COS Cells , Cell Line , Chlorocebus aethiops , Enzyme Activation , Green Fluorescent Proteins , Humans , Kinetics , Luminescent Proteins/analysis , Mice , Molecular Sequence Data , Mutagenesis, Site-Directed , Peptide Fragments/chemistry , Peptide Fragments/immunology , Phosphorylation , Protein Kinase C/genetics , Rats , Recombinant Fusion Proteins/analysis , Recombinant Fusion Proteins/metabolism , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism , Transfection
17.
J Biol Chem ; 276(35): 32616-26, 2001 Aug 31.
Article in English | MEDLINE | ID: mdl-11410587

ABSTRACT

Protein kinase D (PKD)/protein kinase C (PKC) mu is a serine/threonine protein kinase that can be activated by physiological stimuli like growth factors, antigen-receptor engagement and G protein-coupled receptor (GPCR) agonists via a phosphorylation-dependent mechanism that requires PKC activity. In order to investigate the dynamic mechanisms associated with GPCR signaling, the intracellular translocation of a green fluorescent protein-tagged PKD was analyzed by real-time visualization in fibroblasts and epithelial cells stimulated with bombesin, a GPCR agonist. We found that bombesin induced a rapidly reversible plasma membrane translocation of green fluorescent protein-tagged PKD, an event that can be divided into two distinct mechanistic steps. The first step, which is exclusively mediated by the cysteine-rich domain in the N terminus of PKD, involved its translocation from the cytosol to the plasma membrane. The second step, i.e. the rapid reverse translocation of PKD from the plasma membrane to the cytosol, required its catalytic domain and surprisingly PKC activity. These findings provide evidence for a novel mechanism by which PKC coordinates the translocation and activation of PKD in response to bombesin-induced GPCR activation.


Subject(s)
Bombesin/pharmacology , Protein Kinase C/metabolism , Receptors, Bombesin/physiology , 3T3 Cells , Amino Acid Substitution , Animals , Cell Line , Cell Membrane/metabolism , Cysteine , Cytosol/metabolism , Dogs , Enzyme Inhibitors/pharmacology , Epithelial Cells/metabolism , Fibroblasts/metabolism , Green Fluorescent Proteins , Kinetics , Luminescent Proteins/metabolism , Mice , Mutagenesis, Site-Directed , Peptide Fragments/chemistry , Peptide Fragments/metabolism , Point Mutation , Protein Kinase C/chemistry , Protein Kinase C/genetics , Protein Transport , Receptors, Bombesin/drug effects , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/metabolism , Transfection , Red Fluorescent Protein
18.
Exp Cell Res ; 267(1): 37-44, 2001 Jul 01.
Article in English | MEDLINE | ID: mdl-11412036

ABSTRACT

G-protein-coupled receptors (GPCRs) are membrane proteins that exhibit a decreased mobile fraction compared to a freely mobile plasma membrane protein. Recently, interest has focused on proteins other than heterotrimeric G-proteins that interact with GPCRs as scaffolding structures that affect receptor signal transduction. In order to investigate the physical state of receptors before and after agonist, we used fluorescence recovery after photobleaching of the bombesin/gastrin-releasing peptide (GRP) receptor fused to the intrinsically fluorescent green fluorescent protein (GFP-GRP receptor) expressed in KNRK cells to measure both the fraction of mobile receptors and their diffusion rate before and after agonist stimulation. In live cells at 37 degrees C, addition of GRP (100 nM) caused a rapid decrease in GFP-GRP receptor mobile fraction from 0.8 +/- 0.1 to 0.49 +/- 0.05, which was independent of endocytosis. Concurrently, the remaining mobile GFP-GRPreceptors showed an increase in the diffusion rate with the half-time of fluorescent recovery, tau(1/2) = 46 +/- 7 s for untreated cells, decreasing to tau(1/2) = 30 +/- 6 s for cells treated with GRP. Prior treatment with the Src-specific inhibitor PP-2 (10 microM) blocked GFP-GRP receptor immobilization while treatment with the inactive analog PP-3 (10 microM) did not affect receptor immobilization. These data suggest that agonist-bound GPCR have increased plasma membrane diffusion rates but an increased affinity for immobilization into a multiprotein complex that is mediated by Src activity.


Subject(s)
Cell Membrane/metabolism , Receptors, Bombesin/metabolism , Diffusion , Endocytosis , Green Fluorescent Proteins , Luminescent Proteins/genetics , Luminescent Proteins/metabolism , Membrane Fluidity , Oncogene Protein pp60(v-src)/antagonists & inhibitors , Protein Transport , Receptors, Bombesin/agonists , Receptors, Bombesin/genetics , Recombinant Fusion Proteins/agonists , Recombinant Fusion Proteins/metabolism , src-Family Kinases/antagonists & inhibitors
19.
J Cell Physiol ; 188(1): 106-19, 2001 Jul.
Article in English | MEDLINE | ID: mdl-11382927

ABSTRACT

Treatment of intact Swiss 3T3 cells with calyculin-A, an inhibitor of myosin light chain (MLC) phosphatase, induces tyrosine phosphorylation of p125(Fak) in a sharply concentration- and time-dependent manner. Maximal stimulation was 4.2 +/- 2.1-fold (n = 14). The stimulatory effect of calyculin-A was observed at low nanomolar concentrations (<10 nM); at higher concentrations (>10 nM) tyrosine phosphorylation of p125(Fak) was strikingly decreased. Calyculin-A induced tyrosine phosphorylation of p125(Fak) through a protein kinase C- and Ca(2+)-independent pathway. Exposure to either cytochalasin-D or latrunculin-A, which disrupt actin organization by different mechanisms, abolished tyrosine phosphorylation of p125(Fak) in response to calyculin-A. Treatment with high concentrations of platelet-derived growth factor (20 ng/ml) which also disrupt actin stress fibers, completely inhibited tyrosine phosphorylation of p125(Fak) in response to calyculin-A. This agent also induced tyrosine phosphorylation of the focal adhesion-associated proteins p130(Cas) and paxillin. These tyrosine phosphorylation events were associated with a striking increase in the assembly of focal adhesions. The Rho kinase (ROK) inhibitor HA1077 that blocked focal adhesion formation by bombesin, had no effect on the focal adhesion assembly induced by calyculin-A. Thus, calyculin-A induces transient focal adhesion assembly and tyrosine phosphorylation of p125(Fak), p130(Cas), and paxillin, acting downstream of ROK.


Subject(s)
1-(5-Isoquinolinesulfonyl)-2-Methylpiperazine/analogs & derivatives , Cytoskeletal Proteins/metabolism , Enzyme Inhibitors/pharmacology , Focal Adhesions/metabolism , Oxazoles/pharmacology , Phosphoproteins/metabolism , Protein-Tyrosine Kinases/metabolism , Proteins , 1-(5-Isoquinolinesulfonyl)-2-Methylpiperazine/pharmacology , 3T3 Cells , Animals , Bombesin/pharmacology , Bridged Bicyclo Compounds, Heterocyclic/pharmacology , Calcium/metabolism , Crk-Associated Substrate Protein , Cytochalasin D/pharmacology , Cytoskeleton/drug effects , Dose-Response Relationship, Drug , Focal Adhesion Kinase 1 , Focal Adhesion Protein-Tyrosine Kinases , Immunoblotting , Indoles/pharmacology , Maleimides/pharmacology , Marine Toxins , Mice , Microscopy, Fluorescence , Myosin-Light-Chain Phosphatase , Nucleic Acid Synthesis Inhibitors/pharmacology , Paxillin , Phosphoprotein Phosphatases/antagonists & inhibitors , Phosphorylation , Phosphotyrosine/metabolism , Protein Kinase C/antagonists & inhibitors , Protein Kinase C/metabolism , Retinoblastoma-Like Protein p130 , Thiazoles/pharmacology , Thiazolidines , Time Factors
20.
FEBS Lett ; 489(1): 101-6, 2001 Jan 26.
Article in English | MEDLINE | ID: mdl-11231022

ABSTRACT

Addition of gastrin or cholecystokinin octapeptide (CCK-8) to cultures of Rat-1 cells stably transfected with the CCK2 (CCK(B)/gastrin) receptor induced protein kinase D (PKD) activation that was detectable within 1 min and reached a maximum ( approximately 10-fold) after 2.5 min of hormonal stimulation. Half-maximal PKD activation for both CCK-8 and gastrin was achieved at 10 nM. Treatment with various concentrations of the selective PKC inhibitors Ro 31-8220 or GF-I potently blocked PKD activation induced by subsequent addition of CCK-8 in a concentration-dependent fashion. Our results indicate that PKC-dependent PKD activation is a novel early event in the action of gastrin and CCK-8 via CCK2 receptors.


Subject(s)
Protein Kinase C/metabolism , Receptors, Cholecystokinin/metabolism , Animals , Cells, Cultured , Enzyme Activation , Gastrins/metabolism , Humans , Rats , Receptor, Cholecystokinin B , Sincalide/metabolism , Transfection
SELECTION OF CITATIONS
SEARCH DETAIL
...