Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
Add more filters










Publication year range
1.
Toxicol Appl Pharmacol ; 355: 93-102, 2018 09 15.
Article in English | MEDLINE | ID: mdl-29960001

ABSTRACT

NOD-like receptor (NLR) family, pyrin domain-containing 3 (NLRP3) inflammasome is a component of innate immunity, and is responsible for producing mature IL-1ß and -18. Several flavonoids were found to affect inflammasome pathway, but the mechanism of action is still obscure. To elucidate the effects on NLRP3 inflammasome pathway and to determine the structure-activity relationships, NLRP3 inflammasome in differentiated THP-1 cells was activated via treatment with monosodium urate (MSU) crystals. Levels of mature IL-1ß, NLRP3 inflammasome components and apoptosis-associated speck-like protein containing a CARD (caspase recruitment domain) (ASC) oligomerization were investigated and the mechanisms of action were also elucidated. Among the 56 flavonoids initially tested, only flavone, 2',4'-dihhydroxyflavone, 3',4'-dichloroflavone, 4',5,7-trihydroxyflavone (apigenin), 3,4',5,7-tetrahydroxyflavone (kaempferol) and 3,3',4',5,7-pentahydroxyflavone (quercetin) significantly inhibited IL-1ß production at 10 µM. Apigenin, kaempferol and 3',4'-dichloroflavone inhibited ASC oligomerization without affecting the ASC level in cell lysates. Apigenin also inhibited absent in melanoma 2 (AIM2) inflammasome-related pathway, but not NLR family CARD domain-containing protein 4 (NLRC4) inflammasome activation. The action of apigenin on NLRP3 inflammasome activation is mediated partly via inhibition of phosphorylation of spleen tyrosine kinase/protein tyrosine kinase 2 (Syk/Pyk2) pathway. Furthermore, orally administered apigenin (100 mg/kg) strongly reduced the number of neutrophils and monocytes in MSU-induced peritonitis in mice. The present study, for the first time, demonstrated the structure-activity profiles of flavonoids in NLRP3 inflammasome activation and mechanisms of cellular action. Certain flavonoids including apigenin are expected to ameliorate the inflammatory symptoms in autoinflammatory diseases associated with NLRP3 inflammasome activation.


Subject(s)
Flavonoids/pharmacology , Inflammasomes/drug effects , NLR Family, Pyrin Domain-Containing 3 Protein/drug effects , Animals , Caspase 1/drug effects , Cell Line , Focal Adhesion Kinase 2/drug effects , Humans , Inflammation/chemically induced , Inflammation/prevention & control , Male , Mice , Mice, Inbred C57BL , Mice, Inbred NOD , Monocytes/drug effects , Peritonitis/chemically induced , Peritonitis/prevention & control , Structure-Activity Relationship , Syk Kinase/drug effects , Uric Acid
2.
J Cell Mol Med ; 19(11): 2633-46, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26282580

ABSTRACT

Lymphangioleiomyomatosis (LAM) is associated with dysfunction of the tuberous sclerosis complex (TSC) leading to enhanced cell proliferation and migration. This study aims to examine whether doxycycline, a tetracycline antibiotic, can inhibit the enhanced migration of TSC2-deficient cells, identify signalling pathways through which doxycycline works and to assess the effectiveness of combining doxycycline with rapamycin (mammalian target of rapamycin complex 1 inhibitor) in controlling cell migration, proliferation and wound closure. TSC2-positive and TSC2-negative mouse embryonic fibroblasts (MEF), 323-TSC2-positive and 323-TSC2-null MEF and Eker rat uterine leiomyoma (ELT3) cells were treated with doxycycline or rapamycin alone, or in combination. Migration, wound closure and proliferation were assessed using a transwell migration assay, time-lapse microscopy and manual cell counts respectively. RhoA-GTPase activity, phosphorylation of p70S6 kinase (p70S6K) and focal adhesion kinase (FAK) in TSC2-negative MEF treated with doxycycline were examined using ELISA and immunoblotting techniques. The enhanced migration of TSC2-null cells was reduced by doxycycline at concentrations as low as 20 pM, while the rate of wound closure was reduced at 2-59 µM. Doxycycline decreased RhoA-GTPase activity and phosphorylation of FAK in these cells but had no effect on the phosphorylation of p70S6K, ERK1/2 or AKT. Combining doxycycline with rapamycin significantly reduced the rate of wound closure at lower concentrations than achieved with either drug alone. This study shows that doxycycline inhibits TSC2-null cell migration. Thus doxycycline has potential as an anti-migratory agent in the treatment of diseases with TSC2 dysfunction.


Subject(s)
Doxycycline/therapeutic use , Focal Adhesion Kinase 2/drug effects , Lymphangioleiomyomatosis/etiology , Sirolimus/therapeutic use , Tuberous Sclerosis/drug therapy , rho-Associated Kinases/drug effects , Animals , Doxycycline/administration & dosage , Lymphangioleiomyomatosis/drug therapy , Mice , Rats
3.
Ann Neurol ; 77(6): 953-71, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25707991

ABSTRACT

OBJECTIVE: Currently no effective disease-modifying agents exist for the treatment of Alzheimer disease (AD). The Fyn tyrosine kinase is implicated in AD pathology triggered by amyloid-ß oligomers (Aßo) and propagated by Tau. Thus, Fyn inhibition may prevent or delay disease progression. Here, we sought to repurpose the Src family kinase inhibitor oncology compound, AZD0530, for AD. METHODS: The pharmacokinetics and distribution of AZD0530 were evaluated in mice. Inhibition of Aßo signaling to Fyn, Pyk2, and Glu receptors by AZD0530 was tested by brain slice assays. After AZD0530 or vehicle treatment of wild-type and AD transgenic mice, memory was assessed by Morris water maze and novel object recognition. For these cohorts, amyloid precursor protein (APP) metabolism, synaptic markers (SV2 and PSD-95), and targets of Fyn (Pyk2 and Tau) were studied by immunohistochemistry and by immunoblotting. RESULTS: AZD0530 potently inhibits Fyn and prevents both Aßo-induced Fyn signaling and downstream phosphorylation of the AD risk gene product Pyk2, and of NR2B Glu receptors in brain slices. After 4 weeks of treatment, AZD0530 dosing of APP/PS1 transgenic mice fully rescues spatial memory deficits and synaptic depletion, without altering APP or Aß metabolism. AZD0530 treatment also reduces microglial activation in APP/PS1 mice, and rescues Tau phosphorylation and deposition abnormalities in APP/PS1/Tau transgenic mice. There is no evidence of AZD0530 chronic toxicity. INTERPRETATION: Targeting Fyn can reverse memory deficits found in AD mouse models, and rescue synapse density loss characteristic of the disease. Thus, AZD0530 is a promising candidate to test as a potential therapy for AD.


Subject(s)
Alzheimer Disease/drug therapy , Behavior, Animal/drug effects , Benzodioxoles/pharmacology , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins c-fyn/antagonists & inhibitors , Quinazolines/pharmacology , Signal Transduction/drug effects , Amyloid beta-Peptides/drug effects , Animals , Benzodioxoles/pharmacokinetics , Disease Models, Animal , Focal Adhesion Kinase 2/drug effects , Mice , Mice, Inbred C57BL , Mice, Transgenic , Protein Kinase Inhibitors/pharmacokinetics , Quinazolines/pharmacokinetics
4.
Circ Res ; 111(9): 1137-46, 2012 Oct 12.
Article in English | MEDLINE | ID: mdl-22896586

ABSTRACT

RATIONALE: Angiotensin II (Ang II) has pleiotropic effects on vascular smooth muscle cells (VSMCs). It has been demonstrated to promote the proliferative phenotype of VSMCs in mouse ascending aorta, but the underlying mechanisms remain incompletely understood. OBJECTIVE: The present study was designed to explore whether the Ca(2+)-permeable transient receptor potential melastatin 7 (TRPM7) channel is involved in Ang II-induced phenotype switching of ascending aortic VSMCs and to dissect the molecular mechanisms by which TRPM7 modulates VSMC phenotype. METHODS AND RESULTS: As revealed by current recording, Ang II infusion increased TRPM7 whole-cell currents in ascending aortic VSMCs. The increase in TRPM7 currents was found to result from enhanced expression of TRPM7 protein rather than elevated single-channel activity (open probability and slope conductance) and/or reduced Mg(2+)-mediated channel block. Mechanistically, Ang II elevated TRPM7 expression via Ang II type 1 receptor-mediated ERK1/2 signaling. As indicated by the expression levels of VSMC differentiation marker genes, phenotypic switching of ascending aorta occurred during Ang II infusion. Meanwhile, ERK1/2-Elk-1 signaling pathway known to suppress VSMC differentiation was activated in Ang II-infused ascending aorta. Knockdown of TRPM7 with small interfering RNA established a causative role of TRPM7 in Ang II-induced phenotypic change and promotion of cell proliferation. Moreover, TRPM7 was shown to be required for Pyk2-ERK1/2-Elk-1 pathway activation by Ang II, which potentiated TRPM7 channel function and thus activated the Ca(2+)-sensitive kinase Pyk2. Finally, TRPM7 knockdown attenuated Ang II-induced displacement of myocardin from SM22 promoter, but the effects could be reversed by expression of constitutively active c-Src. CONCLUSIONS: Our data establish that upregulation of TRPM7 channels by Ang II contributes to the development of the proliferative phenotype of ascending aortic VSMCs, and TRPM7 channel suppresses VSMC gene expression via Ca(2+) influx-mediated activation of Pyk2-ERK1/2-Elk-1 pathway.


Subject(s)
Angiotensin II/pharmacology , Cell Differentiation/drug effects , Muscle, Smooth, Vascular/cytology , TRPM Cation Channels/metabolism , Up-Regulation/drug effects , Animals , Aorta/cytology , Aorta/drug effects , Cell Differentiation/physiology , Cell Proliferation/drug effects , Cells, Cultured , Focal Adhesion Kinase 2/drug effects , Focal Adhesion Kinase 2/physiology , MAP Kinase Signaling System/drug effects , MAP Kinase Signaling System/physiology , Male , Mice , Mice, Inbred C57BL , Models, Animal , Muscle, Smooth, Vascular/drug effects , Phenotype , Signal Transduction/drug effects , Signal Transduction/physiology , Up-Regulation/physiology , ets-Domain Protein Elk-1/drug effects , ets-Domain Protein Elk-1/physiology
5.
Exp Cell Res ; 315(18): 3210-9, 2009 Nov 01.
Article in English | MEDLINE | ID: mdl-19563802

ABSTRACT

We previously reported that the hypothalamic hormone oxytocin (OT), best known for its uterotonic activity, also stimulates migration and invasion in human umbilical vein endothelial cells (HUVECs), thus suggesting a possible role for the peptide in the regulation of angiogenesis. We identified the Gq coupling of OT receptors (OTRs) and phospholipase C (PLC) as the main effectors of OT's action in HUVECs. Moreover, the pro-migratory effect of OT required the OTR-induced activation of the phosphatidylinositol-3-kinase (PI-3-K)/AKT/endothelial nitric oxide synthase (eNOS) pathway. To better characterize the proposed pro-angiogenic effect of OT in HUVECs, we have now utilized a three-dimensional (3-D) in vitro angiogenesis assay, and demonstrated that OT stimulates the outgrowth of capillary-like structures from HUVEC spheroids to an extent comparable to that of vascular endothelial growth factor (VEGF). This OT effect was abolished by inhibitors of PLC, PI-3-K and Src kinase. It was also found that OT phosphorylates proline-rich tyrosine kinase-2 (Pyk-2) and Src kinase in a PLC- and calcium-dependent manner. Furthermore, knockdown of Pyk-2 expression by RNA interference markedly impaired Src phosphorylation, migration and endothelial cell sprouting induced by OT. In conclusion, by using a pharmacological and genetic approach, the OT pro-angiogenic action and the cascade of intracellular signals responsible for it were defined by showing for the first time that OT, by interacting with its Gq-coupled receptor, induces HUVEC capillary outgrowth via Pyk-2 phosphorylation, which activates Src which in turn activates the PI-3-K/AKT pathway.


Subject(s)
Endothelial Cells/drug effects , Focal Adhesion Kinase 2/metabolism , Neovascularization, Physiologic , Oxytocin/pharmacology , Type C Phospholipases/metabolism , src-Family Kinases/metabolism , Cell Movement/drug effects , Cell Movement/physiology , Cells, Cultured , Chromones/pharmacology , Endothelial Cells/cytology , Endothelial Cells/metabolism , Enzyme Inhibitors/pharmacology , Estrenes/pharmacology , Focal Adhesion Kinase 2/drug effects , Focal Adhesion Kinase 2/genetics , GTP-Binding Protein alpha Subunits, Gq-G11/drug effects , GTP-Binding Protein alpha Subunits, Gq-G11/metabolism , Humans , Morpholines/pharmacology , Phosphatidylinositol 3-Kinases/metabolism , Phosphodiesterase Inhibitors/pharmacology , Phosphoinositide-3 Kinase Inhibitors , Phosphorylation/drug effects , Phosphorylation/physiology , Phosphotransferases (Alcohol Group Acceptor)/antagonists & inhibitors , Phosphotransferases (Alcohol Group Acceptor)/metabolism , Proto-Oncogene Proteins c-akt/drug effects , Proto-Oncogene Proteins c-akt/metabolism , Pyrimidines/pharmacology , Pyrrolidinones/pharmacology , RNA, Small Interfering/metabolism , Receptors, Oxytocin/drug effects , Receptors, Oxytocin/metabolism , Signal Transduction/drug effects , Signal Transduction/physiology , Sphingosine/analogs & derivatives , Sphingosine/pharmacology , Type C Phospholipases/antagonists & inhibitors , Umbilical Veins/cytology , Umbilical Veins/drug effects , Vascular Endothelial Growth Factor A/pharmacology , src-Family Kinases/antagonists & inhibitors
6.
Cell Tissue Res ; 337(2): 243-55, 2009 Aug.
Article in English | MEDLINE | ID: mdl-19484266

ABSTRACT

The related cytoplasmic non-receptor tyrosine kinases Pyk2 (proline-rich tyrosine kinase 2) and FAK (focal adhesion kinase) have been implicated in phenylephrine-induced G-protein-coupled receptor-mediated signaling mechanisms leading to cardiomyocyte hypertrophy. We report that, in phenylephrine-stimulated neonatal rat ventricular myocytes (NRVM), Pyk2 augments expression of the hypertrophic marker atrial natriuretic factor (ANF) but reduces cytoskeletal organization and cell spreading. In contrast, FAK attenuates ANF production but does not alter cytoskeletal organization and cell spreading. Pyk2 and FAK exhibit differential localization in both unstimulated and phenylephrine-stimulated myocytes. Pyk2 catalytic activity is required for Pyk2 to augment ANF secretion but is not necessary to reduce cell spreading. Pyk2 autophosphorylation is required but not sufficient for Pyk2 to augment ANF secretion. Expression of the Pyk2 FERM domain as an autonomous fragment inhibits phenylephrine-mediated ANF secretion and reduces cell spreading. In addition, expression of the Pyk2 FERM domain inhibits the ability of Pyk2 to augment ANF secretion; this is correlated with reduced Pyk2 autophosphorylation. These data indicate that Pyk2 and FAK have different roles and occupy different positions in signaling pathways leading to the development of cardiomyocyte hypertrophy.


Subject(s)
Cardiomegaly/enzymology , Cytoskeleton/metabolism , Focal Adhesion Kinase 1/metabolism , Focal Adhesion Kinase 2/metabolism , Myocytes, Cardiac/enzymology , Animals , Atrial Natriuretic Factor/agonists , Atrial Natriuretic Factor/biosynthesis , Cardiac Myosins/metabolism , Cardiomegaly/pathology , Cardiotonic Agents/pharmacology , Cell Movement/drug effects , Cell Movement/physiology , Cells, Cultured , Cytoskeleton/drug effects , Focal Adhesion Kinase 1/drug effects , Focal Adhesion Kinase 2/drug effects , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/pathology , Myosin Light Chains/metabolism , Phenylephrine/pharmacology , Phosphorylation/drug effects , Phosphorylation/physiology , Rats , Rats, Sprague-Dawley , Signal Transduction/drug effects , Signal Transduction/physiology , Transfection
7.
J Neuroimmunol ; 213(1-2): 47-59, 2009 Aug 18.
Article in English | MEDLINE | ID: mdl-19541372

ABSTRACT

The neuroregulatory activities of PMS-601, a platelet activating factor antagonist, were investigated in laboratory and animal models of HIV-1 encephalitis (HIVE). For the former, PMS-601 reduced monocyte-derived macrophage pro-inflammatory secretions, multinucleated giant cell (MGC) formation, and neuronal loss independent of antiretroviral responses. PMS-601 treatment of HIVE severe combined immunodeficient mice showed reduced microgliosis, MGCs and neurodegeneration. These observations support the further development of PMS-601 as an adjunctive therapy for HIV-1 associated neurocognitive disorders.


Subject(s)
AIDS Dementia Complex/drug therapy , Brain/drug effects , Cognition Disorders/drug therapy , Piperazines/pharmacology , Platelet Activating Factor/antagonists & inhibitors , AIDS Dementia Complex/immunology , AIDS Dementia Complex/metabolism , Animals , Anti-Inflammatory Agents/pharmacology , Anti-Inflammatory Agents/therapeutic use , Brain/metabolism , Brain/physiopathology , Cell Death/drug effects , Cell Death/immunology , Cells, Cultured , Cognition Disorders/immunology , Cognition Disorders/metabolism , Disease Models, Animal , Focal Adhesion Kinase 2/drug effects , Focal Adhesion Kinase 2/metabolism , Giant Cells/drug effects , Giant Cells/immunology , Giant Cells/metabolism , Gliosis/drug therapy , Gliosis/immunology , Gliosis/metabolism , Humans , Immunologic Factors/pharmacology , Immunologic Factors/therapeutic use , Inflammation Mediators/agonists , Inflammation Mediators/metabolism , Male , Mice , Mice, SCID , Microglia/drug effects , Microglia/immunology , Microglia/metabolism , Monocytes/drug effects , Monocytes/immunology , Monocytes/metabolism , NF-kappa B/drug effects , NF-kappa B/metabolism , Piperazines/therapeutic use , Platelet Activating Factor/metabolism
8.
BMC Cancer ; 8: 150, 2008 May 28.
Article in English | MEDLINE | ID: mdl-18507841

ABSTRACT

BACKGROUND: Suppressor of cytokine signaling 3 (SOCS3) is considered to inhibit cytokine responses and play a negative role in migration of various cells. Proline-rich tyrosine kinase 2 (PYK2) is a non-receptor kinase and has been found crucial to cell motility. However, little is known about whether SOCS3 could regulate PYK2 pro-migratory function in lung cancer. METHODS: The methylation status of SOCS3 was investigated in HBE and A549 cell lines by methylation-specific PCR. A549 cells were either treated with a demethylation agent 5-aza-2'-deoxycytidine or transfected with three SOCS3 mutants with various functional domains deleted. Besides, cells were pretreated with a proteasome inhibitor beta-lactacystin where indicated. The effects of SOCS3 up-regulation on PYK2 expression, PYK2 and ERK1/2 phosphorylations were assessed by western blot using indicated antibodies. RT-PCR was used to estimate PYK2 mRNA levels. Transwell experiments were performed to evaluate cell migration. RESULTS: SOCS3 expression was found impaired in A549 cells and higher PYK2 activity was correlated with enhanced cell migration. We identified that SOCS3 was aberrantly methylated in the exon 2, and 5-aza-2'-deoxycytidine restored SOCS3 expression. Reactivation of SOCS3 attenuated PYK2 expression and phosphorylation, cell migration was inhibited as well. Transfection studies indicated that exogenous SOCS3 interacted with PYK2, and both the Src homology 2 (SH2) and the kinase inhibitory region (KIR) domains of SOCS3 contributed to PYK2 binding. Furthermore, SOCS3 was found to inhibit PYK2-associated ERK1/2 activity in A549 cells. SOCS3 possibly promoted degradation of PYK2 in a SOCS-box-dependent manner and interfered with PYK2-related signaling events, such as cell migration. CONCLUSION: These data indicate that SOCS3 negatively regulates cell motility and decreased SOCS3 induced by methylation may confer a migration advantage to A549 cells. These results also suggest a negative role of SOCS3 in PYK2 signaling, and a previously unidentified regulatory mechanism for PYK2 function.


Subject(s)
Cell Movement/physiology , DNA Methylation , Focal Adhesion Kinase 2/metabolism , Signal Transduction/physiology , Suppressor of Cytokine Signaling Proteins/metabolism , Adenocarcinoma/drug therapy , Adenocarcinoma/metabolism , Blotting, Western , Cell Movement/drug effects , Deoxycytidine/pharmacology , Down-Regulation , Focal Adhesion Kinase 2/drug effects , Humans , In Vitro Techniques , Lung Neoplasms/drug therapy , Lung Neoplasms/metabolism , Phosphorylation , Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Sensitivity and Specificity , Signal Transduction/drug effects , Suppressor of Cytokine Signaling 3 Protein , Suppressor of Cytokine Signaling Proteins/drug effects , Tumor Cells, Cultured , Up-Regulation
9.
Eur J Neurosci ; 24(11): 3271-84, 2006 Dec.
Article in English | MEDLINE | ID: mdl-17156388

ABSTRACT

Here, we show that chronic nicotine exposure induces changes in Src signaling for the modulation of N-methyl-D-aspartate receptor (NMDAR) function and LTP induction in CA1 pyramidal cells. Activation of muscarinic receptors normally potentiates NMDAR responses in pyramidal cells via a Gq/protein kinase C (PKC)/proline-rich tyrosine kinase 2/Src signaling cascade. However, muscarinic, PKC and Src stimulation had no effect on NMDAR responses after chronic nicotine treatment. The lack of effect was apparently due to enhanced tyrosine phosphorylation, and therefore further stimulation of the signaling cascade caused no effect on NMDAR responses. Interestingly, another Src-family kinase potentiated NMDAR responses after, but not before, chronic nicotine treatment. In control pyramidal cells, Src inhibitor peptides prevented tetanus-induced long-term potentiation (LTP). Conversely, in chronic nicotine-exposed cells, the inhibitor was ineffective in blocking tetanus-induced LTP. Furthermore, in control pyramidal cells, applying exogenous Src and administration of an endogenous Src-family kinase activator increased alpha-amino-3-hydroxy-5-methylisoxazole-4-propionate receptor (AMPAR)-mediated responses. This increase was blocked by Src inhibitor peptides and occluded tetanus-induced LTP, as reported previously. In contrast, in chronic nicotine-treated pyramidal cells, applying exogenous Src had no effect on AMPAR-mediated responses and a tetanus-induced LTP. Interestingly, however, administration of an endogenous Src-family kinase activator enhanced AMPAR-mediated responses, which occluded tetanus-induced LTP. This enhancement was not prevented by co-application of Src inhibitor peptides. Thus, it appears that chronic nicotine exposure recruits another member of the Src-family for the regulation of NMDAR function and LTP induction. The nicotine-induced distinct signaling cascades may be involved in long-lasting memories of nicotine misuse.


Subject(s)
Hippocampus/enzymology , Long-Term Potentiation/physiology , Nicotine/pharmacology , Pyramidal Cells/enzymology , Signal Transduction/physiology , src-Family Kinases/metabolism , Animals , Enzyme Activation/drug effects , Enzyme Activation/physiology , Enzyme Inhibitors/pharmacology , Focal Adhesion Kinase 2/drug effects , Focal Adhesion Kinase 2/metabolism , Glutamic Acid/metabolism , Hippocampus/drug effects , Long-Term Potentiation/drug effects , Memory/drug effects , Memory/physiology , Nicotinic Agonists/pharmacology , Phosphorylation/drug effects , Protein Kinase C/drug effects , Protein Kinase C/metabolism , Pyramidal Cells/drug effects , Rats , Rats, Sprague-Dawley , Receptors, AMPA/drug effects , Receptors, AMPA/metabolism , Receptors, N-Methyl-D-Aspartate/drug effects , Receptors, N-Methyl-D-Aspartate/metabolism , Signal Transduction/drug effects , Synaptic Transmission/drug effects , Synaptic Transmission/physiology , Tobacco Use Disorder/enzymology , Tobacco Use Disorder/physiopathology , src-Family Kinases/drug effects
10.
Cell Signal ; 18(11): 1932-40, 2006 Nov.
Article in English | MEDLINE | ID: mdl-16574377

ABSTRACT

The calcium-dependent proline-rich tyrosine kinase Pyk2 is activated by tyrosine phosphorylation, associates with focal adhesion proteins, and has been linked to proliferative and migratory responses in a variety of mesenchymal and epithelial cell types. Full Pyk2 activation requires phosphorylation at functionally distinct sites, including autophosphorylation site Tyr-402 and catalytic domain site Tyr-580, though the mechanisms involved are unclear. The pathways mediating Pyk2 phosphorylation at Tyr-402 and Tyr-580 were therefore investigated. Both sites were rapidly and transiently phosphorylated following cell stimulation by Ang II or LPA. However, only Tyr-580 phosphorylation was rapidly enhanced by intracellular Ca(2+) release, or inhibited by Ca(2+) depletion. Conversely, Tyr-402 phosphorylation was highly sensitive to inhibition of actin stress fibers, or of Rho kinase (ROK), an upstream regulator of stress fiber assembly. Ang II also induced a delayed (30-60 min) secondary phosphorylation peak occurring at Tyr-402 alone. Unlike the homologous focal adhesion kinase (FAK), Pyk2 phosphorylation was sensitive neither to the Src inhibitor PP2, nor to truncation of its N-terminal region, which contains a putative autoinhibitory FERM domain. These results better define the mechanisms involved in Pyk2 activation, demonstrating that autophosphorylation is ROK- and stress fiber-dependent, while transphosphorylation within the kinase domain is Ca(2+)-dependent and Src-independent in intestinal epithelial cells. This contrasts with the tight sequential coupling of phosphorylation seen in FAK activation, and further underlines the differences between these closely related kinases.


Subject(s)
Calcium/physiology , Cytoskeleton/metabolism , Focal Adhesion Kinase 2/metabolism , Intestinal Mucosa/metabolism , Intracellular Signaling Peptides and Proteins/physiology , Protein Serine-Threonine Kinases/physiology , src-Family Kinases/physiology , Angiotensin II/pharmacology , Animals , Cells, Cultured , Focal Adhesion Kinase 2/chemistry , Focal Adhesion Kinase 2/drug effects , Humans , Intestinal Mucosa/chemistry , Intestinal Mucosa/cytology , Phosphorylation , Signal Transduction , Tyrosine , rho-Associated Kinases
SELECTION OF CITATIONS
SEARCH DETAIL
...