Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
Add more filters










Publication year range
1.
Physiol Res ; 68(1): 89-98, 2019 03 06.
Article in English | MEDLINE | ID: mdl-30433799

ABSTRACT

The contraction of gastrointestinal (GI) smooth muscles is regulated by both Ca(2+)-dependent and Ca(2+) sensitization mechanisms. Proline-rich tyrosine kinase 2 (Pyk2) is involved in the depolarization-induced contraction of vascular smooth muscle via a Ca(2+) sensitization pathway. However, the role of Pyk2 in GI smooth muscle contraction is unclear. The spontaneous contraction of colonic smooth muscle was measured by using isometric force transducers. Protein and phosphorylation levels were determined by using western blotting. Pyk2 protein was expressed in colonic tissue, and spontaneous colonic contractions were inhibited by PF-431396, a Pyk2 inhibitor, in the presence of tetrodotoxin (TTX). In cultured colonic smooth muscle cells (CSMCs), PF-431396 decreased the levels of myosin light chain (MLC20) phosphorylated at Ser19 and ROCK2 protein expression, but myosin light chain kinase (MLCK) expression was not altered. However, Y-27632, a Rho kinase inhibitor, increased phosphorylation of Pyk2 at Tyr402 and concomitantly decreased ROCK2 levels; the expression of MLCK in CSMCs did not change. The expression of P(Tyr402)-Pyk2 and ROCK2 was increased when CSMCs were treated with Ach. Pyk2 is involved in the process of colonic smooth muscle contraction through the RhoA/ROCK pathway. These pathways may provide very important targets for investigating GI motility disorders.


Subject(s)
Colon/metabolism , Focal Adhesion Kinase 2/biosynthesis , Muscle Contraction/physiology , Muscle, Smooth/metabolism , rho GTP-Binding Proteins/biosynthesis , rho-Associated Kinases/biosynthesis , Animals , Cell Culture Techniques , Colon/drug effects , Enzyme Inhibitors/pharmacology , Focal Adhesion Kinase 2/antagonists & inhibitors , Male , Mice , Mice, Inbred ICR , Muscle Contraction/drug effects , Muscle, Smooth/drug effects , Organ Culture Techniques , Signal Transduction/drug effects , Signal Transduction/physiology , rhoA GTP-Binding Protein
2.
Exp Neurol ; 307: 62-73, 2018 09.
Article in English | MEDLINE | ID: mdl-29803828

ABSTRACT

Pyk2 is a Ca2+-activated non-receptor tyrosine kinase enriched in forebrain neurons and involved in synaptic regulation. Human genetic studies associated PTK2B, the gene coding Pyk2, with risk for Alzheimer's disease (AD). We previously showed that Pyk2 is important for hippocampal function, plasticity, and spine structure. However, its potential role in AD is unknown. To address this question we used human brain samples and 5XFAD mice, an amyloid mouse model of AD expressing mutated human amyloid precursor protein and presenilin1. In the hippocampus of 5XFAD mice and in human AD patients' cortex and hippocampus, Pyk2 total levels were normal. However, Pyk2 Tyr-402 phosphorylation levels, reflecting its autophosphorylation-dependent activity, were reduced in 5XFAD mice at 8 months of age but not 3 months. We crossed these mice with Pyk2-/- mice to generate 5XFAD animals devoid of Pyk2. At 8 months the phenotype of 5XFAD x Pyk2-/- double mutant mice was not different from that of 5XFAD. In contrast, overexpression of Pyk2 in the hippocampus of 5XFAD mice, using adeno-associated virus, rescued autophosphorylated Pyk2 levels and improved synaptic markers and performance in several behavioral tasks. Both Pyk2-/- and 5XFAD mice showed an increase of potentially neurotoxic Src cleavage product, which was rescued by Pyk2 overexpression. Manipulating Pyk2 levels had only minor effects on Aß plaques, which were slightly decreased in hippocampus CA3 region of double mutant mice and increased following overexpression. Our results show that Pyk2 is not essential for the pathogenic effects of human amyloidogenic mutations in the 5XFAD mouse model. However, the slight decrease in plaque number observed in these mice in the absence of Pyk2 and their increase following Pyk2 overexpression suggest a contribution of this kinase in plaque formation. Importantly, a decreased function of Pyk2 was observed in 5XFAD mice, indicated by its decreased autophosphorylation and associated Src alterations. Overcoming this deficit by Pyk2 overexpression improved the behavioral and molecular phenotype of 5XFAD mice. Thus, our results in a mouse model of AD suggest that Pyk2 impairment may play a role in the symptoms of the disease.


Subject(s)
Alzheimer Disease/enzymology , Alzheimer Disease/pathology , Disease Models, Animal , Focal Adhesion Kinase 2/biosynthesis , Gene Expression Regulation, Enzymologic , Aged, 80 and over , Alzheimer Disease/genetics , Animals , Cerebral Cortex/enzymology , Cerebral Cortex/pathology , Female , Focal Adhesion Kinase 2/genetics , Gene Expression Regulation, Enzymologic/physiology , Hippocampus/enzymology , Hippocampus/pathology , Humans , Locomotion/physiology , Male , Maze Learning/physiology , Mice , Mice, Transgenic , Plaque, Amyloid/enzymology , Plaque, Amyloid/genetics , Plaque, Amyloid/pathology
3.
J Am Soc Nephrol ; 29(4): 1198-1209, 2018 04.
Article in English | MEDLINE | ID: mdl-29436516

ABSTRACT

Nonobstructive hydronephrosis, defined as dilatation of the renal pelvis with or without dilatation of the ureter, is the most common antenatal abnormality detected by fetal ultrasound. Yet, the etiology of nonobstructive hydronephrosis is poorly defined. We previously demonstrated that defective development of urinary tract pacemaker cells (utPMCs) expressing hyperpolarization-activated cyclic nucleotide-gated channel 3 (HCN3) and the stem cell marker cKIT causes abnormal ureteric peristalsis and nonobstructive hydronephrosis. However, further investigation of utPMC development and function is limited by lack of knowledge regarding the embryonic derivation, development, and molecular apparatus of these cells. Here, we used lineage tracing in mice to identify cells that give rise to utPMCs. Neural crest cells (NCCs) indelibly labeled with tdTomato expressed HCN3 and cKIT. Furthermore, purified HCN3+ and cKIT+ utPMCs were enriched in Sox10 and Tfap-2α, markers of NCCs. Sequencing of purified RNA from HCN3+ cells revealed enrichment of a small subset of RNAs, including RNA encoding protein kinase 2ß (PTK2ß), a Ca2+-dependent tyrosine kinase that regulates ion channel activity in neurons. Immunofluorescence analysis in situ revealed PTK2ß expression in NCCs as early as embryonic day 12.5 and in HCN3+ and cKIT+ utPMCs as early as embryonic day 15.5, with sustained expression in HCN3+ utPMCs until postnatal week 8. Pharmacologic inhibition of PTK2ß in murine pyeloureteral tissue explants inhibited contraction frequency. Together, these results demonstrate that utPMCs are derived from NCCs, identify new markers of utPMCs, and demonstrate a functional contribution of PTK2ß to utPMC function.


Subject(s)
Focal Adhesion Kinase 2/physiology , Gene Expression Regulation, Developmental , Interstitial Cells of Cajal/enzymology , Kidney Pelvis/physiology , Neural Crest/enzymology , Peristalsis/physiology , Ureter/physiology , Animals , Antigens, Differentiation/analysis , Focal Adhesion Kinase 2/biosynthesis , Focal Adhesion Kinase 2/genetics , Genes, Reporter , Gestational Age , Hydronephrosis/enzymology , Hydronephrosis/physiopathology , Hyperpolarization-Activated Cyclic Nucleotide-Gated Channels/analysis , Interstitial Cells of Cajal/physiology , Kidney Pelvis/cytology , Kidney Pelvis/embryology , Kidney Pelvis/growth & development , Mice , Mice, Inbred C57BL , Mice, Transgenic , Neural Crest/physiology , Potassium Channels/analysis , Proto-Oncogene Proteins c-kit/analysis , RNA, Messenger/biosynthesis , SOXE Transcription Factors/analysis , Signal Transduction , Transcription Factor AP-2/analysis , Ureter/cytology , Ureter/embryology , Ureter/growth & development
4.
Br J Pharmacol ; 175(3): 456-468, 2018 02.
Article in English | MEDLINE | ID: mdl-29139546

ABSTRACT

BACKGROUND AND PURPOSE: Haem oxygenase-1 (HO-1) could provide cytoprotection against various inflammatory diseases. However, the mechanisms underlying the protective effect of CO-releasing molecule-2 (CORM-2)-induced HO-1 expression against TNF-α-induced inflammatory responses in human pulmonary alveolar epithelial cells (HPAEpiCs) remain unknown. EXPERIMENTAL APPROACH: CORM-2-induced HO-1 protein and mRNA expression, and signalling pathways were determined by Western blot and real-time PCR, coupled with respective pharmacological inhibitors or transfection with siRNAs. The effect of CORM-2 on TNF-α-induced increase in leukocyte counts in BAL fluid and VCAM-1 expression in lung was determined by cell counting and Western blot analysis. KEY RESULTS: CORM-2 attenuated the TNF-α-induced pulmonary haematoma, VCAM-1 expression and increase in leukocytes through an up-regulation of HO-1 in mice; this effect of CORM-2 was reversed by the HO-1 inhibitor zinc protoporphyrin IX. Furthermore, CORM-2 increased HO-1 protein and mRNA expression as well as the phosphorylation of PYK2, PKCα and ERK1/2 (p44/p42 MAPK) in HPAEpiCs; these effects were attenuated by their respective pharmacological inhibitors or transfection with siRNAs. Inhibition of PKCα by Gö6976 or Gö6983 attenuated CORM-2-induced stimulation of PKCα and ERK1/2 phosphorylation but had no effect on PYK2 phosphorylation. Moreover, inhibition of PYK2 by PF431396 reduced the phosphorylation of all three protein kinases. Finally, PYK2/PKCα/ERK1/2-mediated stimulation of activator protein 1 was shown to play a key role in CORM-2-induced HO-1 expression via an up-regulation of c-Fos mRNA. CONCLUSIONS AND IMPLICATIONS: CORM-2 activates a PYK2/PKCα/ERK1/2/AP-1 pathway leading to HO-1 expression in HPAEpiCs. This HO-1/CO system might have potential as a therapeutic target in pulmonary inflammation.


Subject(s)
Focal Adhesion Kinase 2/biosynthesis , Heme Oxygenase-1/biosynthesis , Organometallic Compounds/pharmacology , Pneumonia/metabolism , Protein Kinase C-alpha/biosynthesis , Tumor Necrosis Factor-alpha/toxicity , Animals , Cell Line , Humans , Male , Mice , Mice, Inbred ICR , Organometallic Compounds/therapeutic use , Pneumonia/chemically induced , Pneumonia/prevention & control , Tumor Necrosis Factor-alpha/antagonists & inhibitors , Up-Regulation/physiology
5.
Hum Mol Genet ; 26(22): 4519-4529, 2017 11 15.
Article in English | MEDLINE | ID: mdl-28973302

ABSTRACT

Genetic variants at PTK2B-CLU locus pose as high-risk factors for many age-related disorders. However, the role of these variants in disease progression is less characterized. In this study, we aimed to investigate the functional significance of a clusterin intronic SNP, rs2279590, that has been associated with pseudoexfoliation, Alzheimer's disease (AD) and diabetes. We have previously shown that the alleles at rs2279590 differentially regulate clusterin (CLU) gene expression in lens capsule tissues. This polymorphism resides in an active regulatory region marked by H3K27Ac and DNase I hypersensitive site and is an eQTL for CLU expression. Here, we report the presence of an enhancer element in surrounding region of rs2279590. Deletion of a 115 bp intronic region flanking the rs2279590 variant through CRISPR-Cas9 genome editing in HEK293 cells demonstrated a decreased clusterin gene expression. Electrophoretic mobility shift and chromatin immunoprecipitation assays show that rs2279590 with allele 'A' constitutes a transcription factor binding site for heat shock factor-1 (HSF1) but not with allele 'G'. By binding to allele 'A', HSF1 abrogates the enhancer effect of the locus as validated by reporter assays. Interestingly, rs2279590 locus has a widespread enhancer effect on two nearby genes, protein tyrosine kinase 2 beta (PTK2B) and epoxide hydrolase-2 (EPHX2); both of which have been previously associated with AD as risk factors. To summarize, our study unveils a mechanistic role of the common variant rs2279590 that can affect a variety of aging disorders by regulating the expression of a specific set of genes.


Subject(s)
Alzheimer Disease/genetics , Clusterin/genetics , Epoxide Hydrolases/genetics , Exfoliation Syndrome/genetics , Focal Adhesion Kinase 2/genetics , Alleles , Alzheimer Disease/metabolism , Clusterin/biosynthesis , Enhancer Elements, Genetic , Epoxide Hydrolases/biosynthesis , Exfoliation Syndrome/metabolism , Focal Adhesion Kinase 2/biosynthesis , Gene Frequency , Genetic Predisposition to Disease , Genetic Variation , Genotype , HEK293 Cells , Humans , Polymorphism, Single Nucleotide , Quantitative Trait Loci , Risk Factors
6.
Oncol Rep ; 34(6): 3280-7, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26352169

ABSTRACT

In the present study, we aimed to demonstrate whether praline-rich tyrosine kinase-2 (Pyk2) participates in the chemokine receptor 7 (CCR7) downstream signaling network, and to determine the role of this molecule and the related mechanism in the CCR7-mediated regulation of viability and metastasis in vivo and in vitro of squamous cell carcinoma of the head and neck (SCCHN). We constructed the stable Pyk2 related non-kinase (PRNK)-expressing SCCHN cell line, and examined the viability, apoptosis, migration, invasion and adhesion ability in the transfected and untransfected SCCHN cells. An SCCHN tumor model in nude mice was designed and the tumor growth rate was assayed. E-cadherin and vimentin expression was assessed when Pyk2 was inactivated. We found that the stable PRNK-expressing SCCHN cells exhibited low viability, a high rate of apoptosis, low migratory ability, low invasive ability and low adhesion capacity. In the nude mouse body, the tumors formed by these cells grew slowly when compared to the tumor growth in the control group. When Pyk2 was inactivated, CCR7-induced E-cadherin and vimentin expression levels were altered. Thus, Pyk2 is a key downstream signaling molecules of CCR7 in SCCHN, which promotes SCCHN tumorigenesis and progression.


Subject(s)
Carcinoma, Squamous Cell/genetics , Focal Adhesion Kinase 2/genetics , Head and Neck Neoplasms/genetics , Neoplasm Invasiveness/genetics , Receptors, CCR7/genetics , Animals , Apoptosis/genetics , Carcinogenesis , Carcinoma, Squamous Cell/pathology , Cell Line, Tumor , Cell Movement/genetics , Cell Survival/genetics , Focal Adhesion Kinase 2/antagonists & inhibitors , Focal Adhesion Kinase 2/biosynthesis , Focal Adhesions/genetics , Gene Expression Regulation, Neoplastic , Head and Neck Neoplasms/pathology , Humans , Mice , Neoplasm Metastasis , Signal Transduction , Squamous Cell Carcinoma of Head and Neck , Xenograft Model Antitumor Assays
7.
PLoS One ; 9(8): e104159, 2014.
Article in English | MEDLINE | ID: mdl-25090023

ABSTRACT

Microvascular lesions resulting from endothelial cell dysfunction are produced in the brain, lung, kidney, and retina of patients of complex chronic diseases. The environmental and molecular risk factors which may contribute in the development of microvascular damage are unclear. The mechanism(s) responsible for initiating microvascular damage remain poorly defined, although several inciting factors have been proposed, including environmental toxicants-induced oxidative stress. Enhanced neovascularization has been implicated in either the development or progression of proliferative vascular lesions. Here, we present evidence for how PCB-induced ROS may contribute to the development of a neovascular phenotype with the aim of elucidating the role of environmental toxicants in endothelial dysfunction with a specific focus on the inhibitor of differentiation protein ID3. We used a combination of phenotype and immunohistochemical analysis followed by validating with protein expression and post-translational modifications with Western Blot and MALDI-TOF/TOF analysis. We also looked for a correlation between ID3 expression in vascular tissue. Our results showed that PCB-induced ROS mediated a highly tube branched neovascular phenotype that also depended on ID3 and Pyk2; and PCB153 treatment increased the size of endothelial spheroids under conditions typically used for clonal selection of stem cell spheroids. High ID3 protein expression correlated with a greater degree of malignancy and oxidative DNA damage marker 8-OHdG in blood vessels from human subjects. PCB153 treatment increased both serine and tyrosine phosphorylation of endothelial ID3. Stable ID3 overexpression increased cell survival of human microvascular endothelial cell line hCMEC/D3. In summary, our data provide evidence that ID3 may play a critical role in regulating vascular endothelial cell survival and development of microvascular lesions induced by persistent environmental pollutants such as PCB153. Findings of this study are important because they provide a new paradigm by which PCBs may contribute to the growth of microvascular lesions.


Subject(s)
Cell Differentiation/drug effects , Endothelial Cells/pathology , Inhibitor of Differentiation Proteins/biosynthesis , Microvessels/pathology , Neoplasm Proteins/biosynthesis , Polychlorinated Biphenyls/toxicity , Air Pollutants/toxicity , Cell Differentiation/genetics , DNA Damage/drug effects , Endothelial Cells/drug effects , Focal Adhesion Kinase 2/biosynthesis , Focal Adhesion Kinase 2/genetics , Gene Expression Regulation/drug effects , Human Umbilical Vein Endothelial Cells , Humans , Inhibitor of Differentiation Proteins/genetics , Microvessels/drug effects , Microvessels/metabolism , Neoplasm Proteins/genetics , Oxidation-Reduction/drug effects
8.
Oncogene ; 32(16): 2005-15, 2013 Apr 18.
Article in English | MEDLINE | ID: mdl-22710711

ABSTRACT

Epithelial-mesenchymal transition (EMT) programs are essential in promoting breast cancer invasion, systemic dissemination and in arousing proliferative programs in breast cancer micrometastases, a reaction that is partially dependent on focal adhesion kinase (FAK). Many functions of FAK are shared by its homolog, protein tyrosine kinase 2 (Pyk2), raising the question as to whether Pyk2 also participates in driving the metastatic outgrowth of disseminated breast cancer cells. In addressing this question, we observed Pyk2 expression to be (i) significantly upregulated in recurrent human breast cancers; (ii) differentially expressed across clonal isolates of human MDA-MB-231 breast cancer cells in a manner predictive for metastatic outgrowth, but not for invasiveness; and (iii) dramatically elevated in ex vivo cultures of breast cancer cells isolated from metastatic lesions as compared with cells that produced the primary tumor. We further show that metastatic human and murine breast cancer cells robustly upregulate their expression of Pyk2 during EMT programs stimulated by transforming growth factor-ß (TGF-ß). Genetic and pharmacological inhibition of Pyk2 demonstrated that the activity of this protein tyrosine kinase was dispensable for the ability of breast cancer cells to undergo invasion in response to TGF-ß, and to form orthotopic mammary tumors in mice. In stark contrast, Pyk2-deficiency prevented TGF-ß from stimulating the growth of breast cancer cells in 3D-organotypic cultures that recapitulated pulmonary microenvironments, as well as inhibited the metastatic outgrowth of disseminated breast cancer cells in the lungs of mice. Mechanistically, Pyk2 expression was inversely related to that of E-cadherin, such that elevated Pyk2 levels stabilized ß1 integrin expression necessary to initiate the metastatic outgrowth of breast cancer cells. Thus, we have delineated novel functions for Pyk2 in mediating distinct elements of the EMT program and metastatic cascade regulated by TGF-ß, particularly the initiation of secondary tumor outgrowth by disseminated cells.


Subject(s)
Breast Neoplasms/enzymology , Epithelial-Mesenchymal Transition/physiology , Focal Adhesion Kinase 2/biosynthesis , Mammary Neoplasms, Experimental/enzymology , Transforming Growth Factor beta/pharmacology , Animals , Biomarkers, Tumor/biosynthesis , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Cell Line, Tumor , Epithelial-Mesenchymal Transition/drug effects , Female , Humans , Lung Neoplasms/enzymology , Lung Neoplasms/secondary , Mammary Neoplasms, Experimental/genetics , Mammary Neoplasms, Experimental/pathology , Mice , Mice, Inbred BALB C , Signal Transduction/drug effects , Smad4 Protein/metabolism , Transforming Growth Factor beta/metabolism , src-Family Kinases/metabolism
9.
J Biol Chem ; 287(21): 17257-17268, 2012 May 18.
Article in English | MEDLINE | ID: mdl-22447931

ABSTRACT

The proliferation and differentiation of osteoblast (OB) precursors are essential for elaborating the bone-forming activity of mature OBs. However, the mechanisms regulating OB proliferation and function are largely unknown. We reported that OB proliferation is enhanced by megakaryocytes (MKs) via a process that is regulated in part by integrin signaling. The tyrosine kinase Pyk2 has been shown to regulate cell proliferation and survival in a variety of cells. Pyk2 is also activated by integrin signaling and regulates actin remodeling in bone-resorbing osteoclasts. In this study, we examined the role of Pyk2 and actin in the MK-mediated increase in OB proliferation. Calvarial OBs were cultured in the presence of MKs for various times, and Pyk2 signaling cascades in OBs were examined by Western blotting, subcellular fractionation, and microscopy. We found that MKs regulate the temporal expression of Pyk2 and its subcellular localization. We also found that MKs regulate the expression of two alternatively spliced isoforms of Pyk2 in OBs, which may regulate OB differentiation and proliferation. MKs also induced cytoskeletal reorganization in OBs, which was associated with the caspase-mediated cleavage of actin, an increase in focal adhesions, and the formation of apical membrane ruffles. Moreover, BrdU incorporation in MK-stimulated OBs was blocked by the actin-polymerizing agent, jasplakinolide. Collectively, our studies reveal that Pyk2 and actin play an important role in MK-regulated signaling cascades that control OB proliferation and may be important for therapeutic interventions aimed at increasing bone formation in metabolic diseases of the skeleton.


Subject(s)
Actins/metabolism , Caspases/metabolism , Focal Adhesion Kinase 2/biosynthesis , Gene Expression Regulation, Enzymologic/physiology , Megakaryocytes/metabolism , Osteoblasts/metabolism , Alternative Splicing , Animals , Cell Differentiation/physiology , Cell Proliferation , Cells, Cultured , Focal Adhesions/metabolism , Isoenzymes/biosynthesis , Megakaryocytes/cytology , Mice , Osteoblasts/cytology
10.
Am J Physiol Cell Physiol ; 301(1): C242-51, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21451101

ABSTRACT

Aberrant vascular smooth muscle cell (VSMC) growth is associated with many vascular diseases including atherosclerosis, hypertension, and restenosis. Platelet-derived growth factor-BB (PDGF) induces VSMC proliferation through control of cell cycle progression and protein and DNA synthesis. Multiple signaling cascades control VSMC growth, including members of the mitogen-activated protein kinase (MAPK) family as well as phosphatidylinositol 3-kinase (PI3K) and its downstream effector AKT/protein kinase B (PKB). Little is known about how these signals are integrated by mitogens and whether there are common receptor-proximal signaling control points that synchronize the execution of physiological growth functions. The nonreceptor proline-rich tyrosine kinase 2 (PYK2) is activated by a variety of growth factors and G protein receptor agonists in VSMC and lies upstream of both PI3K and MAPK cascades. The present study investigated the role of PYK2 in PDGF signaling in cultured rat aortic VSMC. PYK2 downregulation attenuated PDGF-dependent protein and DNA synthesis, which correlated with inhibition of AKT and extracellular signal-regulated kinases 1 and 2 (ERK1/2) but not p38 MAPK activation. Inhibition of PDGF-dependent protein kinase B (AKT) and ERK1/2 signaling by inhibitors of upstream kinases PI3K and MEK, respectively, as well as downregulation of PYK2 resulted in modulation of the G(1)/S phase of the cell cycle through inhibition of retinoblastoma protein (Rb) phosphorylation and cyclin D(1) expression, as well as p27(Kip) upregulation. Cell division kinase 2 (cdc2) phosphorylation at G(2)/M was also contingent on PDGF-dependent PI3K-AKT and ERK1/2 signaling. These data suggest that PYK2 is an important upstream mediator in PDGF-dependent signaling cascades that regulate VSMC proliferation.


Subject(s)
Focal Adhesion Kinase 2/metabolism , Muscle, Smooth, Vascular/cytology , Myocytes, Smooth Muscle/cytology , Myocytes, Smooth Muscle/metabolism , Platelet-Derived Growth Factor/metabolism , Signal Transduction , Animals , Cell Cycle , Cell Proliferation , Cyclin D1/genetics , Cyclin D1/metabolism , Cyclin-Dependent Kinase Inhibitor p27/genetics , Cyclin-Dependent Kinases/metabolism , DNA/biosynthesis , Extracellular Signal-Regulated MAP Kinases/metabolism , Focal Adhesion Kinase 2/biosynthesis , Focal Adhesion Kinase 2/genetics , Mitogen-Activated Protein Kinase Kinases/metabolism , Muscle, Smooth, Vascular/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Phosphorylation , Protein Biosynthesis , Proto-Oncogene Proteins c-akt/metabolism , RNA Interference , RNA, Small Interfering , Rats , Retinoblastoma Protein/metabolism , p38 Mitogen-Activated Protein Kinases/metabolism
11.
Circ Res ; 102(12): 1520-8, 2008 Jun 20.
Article in English | MEDLINE | ID: mdl-18483407

ABSTRACT

In native and primary cultures of endothelial cells, fluid shear stress elicits the tyrosine phosphorylation of the endothelial NO synthase (eNOS), however, the consequences of this modification on enzyme activity are unclear. We found that fluid shear stress induces the association of eNOS with the proline-rich tyrosine kinase 2 (PYK2) in endothelial cells and that the eNOS immunoprecipitated from eNOS- and PYK2-overexpressing HEK293 cells was tyrosine-phosphorylated on Tyr657. In mouse carotid arteries, the overexpression of wild-type PYK2, but not a dominant-negative PYK2, decreased eNOS activity (approximately 50%), whereas in murine lung endothelial cells, the downregulation of PYK2 (small interfering RNA) increased ionomycin-induced NO production. Mutation of Tyr657 to the phosphomimetic residues aspartate (D) or glutamate (E) abolished enzyme activity, whereas a nonphosphorylatable mutant (phenylalanine [F]) showed activity comparable to the wild-type enzyme. Moreover, normal flow-induced vasodilatation was apparent in carotid arteries from eNOS(-/-) mice overexpressing either the wild-type eNOS or the Y657F mutant, whereas no flow-induced vasodilatation was apparent in arteries expressing the Y657E eNOS mutant. Insulin also activated PYK2 and stimulated eNOS in endothelial cells expressing the Y657F mutant but not wild-type eNOS. These data indicate that PYK2 mediates the tyrosine phosphorylation of eNOS on Tyr657 in response to fluid shear stress and insulin stimulation and that this modification attenuates the activity of the enzyme. The PYK2-dependent inhibition of NO production may serve to keep eNOS activity low and limit the detrimental consequences of maintained high NO output, ie, the generation of peroxynitrite.


Subject(s)
Endothelial Cells/enzymology , Focal Adhesion Kinase 2/physiology , Insulin/pharmacology , Nitric Oxide Synthase Type III/antagonists & inhibitors , Nitric Oxide Synthase Type II/antagonists & inhibitors , Animals , Aorta/cytology , Cells, Cultured/drug effects , Cells, Cultured/enzymology , Endothelial Cells/drug effects , Enzyme Activation/drug effects , Epithelial Cells/drug effects , Epithelial Cells/enzymology , Focal Adhesion Kinase 2/biosynthesis , Focal Adhesion Kinase 2/genetics , Ionomycin/pharmacology , Lung/cytology , Mice , Mutation, Missense , Nitric Oxide/biosynthesis , Nitric Oxide Synthase Type II/deficiency , Nitric Oxide Synthase Type II/genetics , Nitric Oxide Synthase Type III/chemistry , Nitric Oxide Synthase Type III/genetics , Nitric Oxide Synthase Type III/physiology , Point Mutation , RNA Interference , RNA, Small Interfering/pharmacology , Recombinant Fusion Proteins/physiology , Rheology , Stress, Mechanical , Sus scrofa
12.
J Mol Cell Cardiol ; 45(1): 93-105, 2008 Jul.
Article in English | MEDLINE | ID: mdl-18495152

ABSTRACT

PYK2 is a Ca(2+)-dependent, nonreceptor protein tyrosine kinase that is involved in the induction of left ventricular hypertrophy (LVH) and its transition to heart failure. We and others have previously investigated PYK2's function in vitro using cultured neonatal and adult rat ventricular myocytes as model systems. However, the function of PYK2 in the in vivo adult heart remains unclear. Here we evaluate the effect of PYK2 inhibition following myocardial infarction (MI) using adenoviral (Adv) overexpression of the C-terminal domain of PYK2, known as CRNK. First we demonstrate that CRNK functions as a dominant-negative inhibitor of PYK2-dependent signaling, presumably by displacing PYK2 from focal adhesions and costameres. Then, male Sprague-Dawley rats (~300 g) underwent permanent left anterior descending coronary artery ligation. One wk post-MI, either Adv-GFP (n=34) or Adv-CRNK (n=28) was administered (10(10) pfu, 0.1 ml) via catheter-based, Optison-mediated gene transfer. LV structure and function were evaluated by echocardiography 1 and 3 wk after gene transfer, and LV tissue was analyzed by real-time RT-PCR and Western blotting. CRNK overexpression was readily detected by Western blotting 1 wk following gene transfer. Adv-CRNK improved overall survival (P=0.03; Logrank Test) and LV fractional shortening (23+/-2% vs. 31+/-2% for Adv-GFP vs. Adv-CRNK infected animals, respectively; P<0.05). Whereas MI hearts exhibited increased beta-, and decreased alpha-myosin heavy chain (MHC) mRNA expression characteristic of LVH, Adv-CRNK reversed the MHC isoenzyme switch (3.3+/-1.4 fold increase in alpha MHC; 0.4+/-0.1 fold decrease in beta MHC; P<0.05 for both). In summary, CRNK gene transfer improves survival, increases LV function, and alters MHC gene expression suggesting an attenuation of LV remodeling post-MI.


Subject(s)
Adenoviridae , Focal Adhesion Kinase 2/biosynthesis , Myocardial Infarction/enzymology , Myosin Heavy Chains/metabolism , Transduction, Genetic , Ventricular Myosins/metabolism , Ventricular Remodeling , Animals , Animals, Newborn , Cells, Cultured , Focal Adhesion Kinase 2/genetics , Genetic Therapy , Heart Failure/enzymology , Heart Failure/genetics , Hypertrophy, Left Ventricular/enzymology , Hypertrophy, Left Ventricular/genetics , Isoenzymes/genetics , Isoenzymes/metabolism , Male , Myocardial Infarction/genetics , Myocardial Infarction/therapy , Myosin Heavy Chains/genetics , Protein Structure, Tertiary/genetics , Rats , Rats, Sprague-Dawley , Ventricular Function, Left/genetics , Ventricular Myosins/genetics , Ventricular Remodeling/genetics
13.
Biol Chem ; 389(1): 47-55, 2008 Jan.
Article in English | MEDLINE | ID: mdl-18095869

ABSTRACT

We previously observed that collagen IV regulates Caco-2 intestinal epithelial cell spreading and migration via Src-dependent p130(Cas) phosphorylation and stimulates focal adhesion kinase (FAK). However, the role of FAK and the related kinase, Pyk2, in Caco-2 spreading and migration is unclear. FAK- or Pyk2-specific siRNAs reduced protein levels by 90%. However, when detached cells were replated on collagen IV neither individual nor combined FAK and Pyk2 siRNAs affected the cell spreading rate. As combined FAK and Pyk2 siRNAs increased p130(Cas) protein levels, we cotransfected cells with 1 nm p130(Cas) siRNA to partially reduce p130(Cas) protein to control levels. Although p130(Cas) Tyr(P)(249) phosphorylation was reduced by 60%, cell spreading was unaffected. Combined siRNA reduction of FAK, Pyk2 and p130(Cas) increased cell spreading by 20% compared to p130(Cas) siRNA alone, suggesting that FAK and Pyk2 negatively regulate spreading in addition to stimulating spreading via p130(Cas). FAK-binding mutant SH3 domain-deleted rat p130(Cas) was not phosphorylated after adhesion and, unlike full-length p130(Cas), did not restore spreading after human-specific p130(Cas) siRNA knockdown of endogenous p130(Cas). Together, these data suggest that FAK positively regulates Caco-2 spreading on collagen IV via p130(Cas) phosphorylation, but also suggests that FAK may negatively regulate spreading through other mechanisms and the presence of additional FAK-independent pathways regulating p130(Cas).


Subject(s)
Collagen Type IV/pharmacology , Crk-Associated Substrate Protein/metabolism , Focal Adhesion Kinase 2/physiology , Signal Transduction/physiology , Caco-2 Cells , Cell Adhesion/drug effects , Cell Proliferation/drug effects , Culture Media , Focal Adhesion Kinase 2/antagonists & inhibitors , Focal Adhesion Kinase 2/biosynthesis , Humans , Phosphorylation , RNA, Small Interfering/pharmacology , Transfection
14.
Oncogene ; 27(12): 1737-48, 2008 Mar 13.
Article in English | MEDLINE | ID: mdl-17906699

ABSTRACT

Neuropeptide hormones like bombesin/gastrin-releasing peptide, galanin or bradykinin, acting via auto and paracrine growth loops, represent the principal mitogens of small cell lung cancer (SCLC). These mitogenic neuropeptides activate G(q/11)-coupled receptors which stimulate phospholipase Cbeta activity, followed by rises of the intracellular calcium concentration ([Ca2+](i)) and activation of protein kinase C (PKC). We report here that proline-rich tyrosine kinase Pyk2 is highly expressed in SCLC cells and provides a functional link between neuropeptide-induced increases in [Ca2+](i) and tumor cell proliferation. Activation of Pyk2 and its association with Src kinases critically depends on the elevation of [Ca2+](i), but is independent of PKC. Src kinase activities are crucial for neuropeptide-mediated GTP-loading of Ras and activation of extracellular signal-regulated kinases in SCLC cells. Pyk2 and Src kinases essentially contribute to anchorage-independent proliferation of SCLC cells. Inhibition of either Pyk2 or Src kinases by lentiviral RNAi or pharmacological inhibition with PP2, respectively, attenuated basal and neuropeptide-elicited survival and proliferation of SCLC cells in liquid culture and in soft agar. Thus, neuropeptides stimulate anchorage-independent survival and proliferation of SCLC cells via pathways involving Pyk2 and Src kinases. Therefore, Ca2+-induced Pyk2/Src complex formation may be a rewarding molecular target for novel therapeutic strategies in SCLC cells.


Subject(s)
Carcinoma, Small Cell/enzymology , Cell Proliferation , Focal Adhesion Kinase 2/physiology , Galanin/physiology , Lung Neoplasms/enzymology , src-Family Kinases/physiology , Calcium/physiology , Carcinoma, Small Cell/pathology , Cell Line , Cell Line, Tumor , Enzyme Activation/genetics , Focal Adhesion Kinase 2/biosynthesis , Focal Adhesion Kinase 2/genetics , Focal Adhesion Kinase 2/metabolism , Humans , Lung Neoplasms/pathology , Proline/metabolism , src-Family Kinases/metabolism
15.
Biochimie ; 88(1): 77-84, 2006 Jan.
Article in English | MEDLINE | ID: mdl-16040187

ABSTRACT

The expression and localization of prolin-rich tyrosine kinase 2 (Pyk2) were studied in chick embryo epiphyseal chondrocytes. Two immunoreactive bands were detected in chondrocytes, a major band with an apparent Mr of 123 kDa and a minor band with an apparent Mr of 68 kDa. The major band appears to migrate as a doublet with apparent Mr of 116/123 kDa. Increased levels of the three forms of Pyk2 were observed in v-src transformed chondrocytes as compared to control uninfected chondrocytes. Immunofluorescent staining shows that Pyk2 is clearly visible in the cytosol and in the perinuclear region of control and v-src-chondrocytes and displays a pattern very similar to the distribution of the mitochondrial marker Mito Tracker. More, immunofluorescent staining shows that Pyk2 is nuclear in most chondrocytes. By subcellular fractionation, the p116/123 Pyk2 doublet, was found to be accumulated mainly in the cytoplasm while the p68 Pyk2 form, was found to be accumulated exclusively in the nucleus. The differential nuclear/cytoplasmic distribution of the Pyk2 forms remains unchanged after v-Src-induced transformation. The p68 Pyk2 form could no longer be detected by using a N-terminus domain-specific anti-Pyk2 antibody. Consistently, Pyk2 immunoreactivity was restricted to the cytoplasm of control and v-src transformed chondrocytes. Thus it appears that the p68 Pyk2 form that accumulates in the nucleus has a deletion in the N-terminus region.


Subject(s)
Chondrocytes/ultrastructure , Focal Adhesion Kinase 2/biosynthesis , Oncogene Protein pp60(v-src)/genetics , Animals , Cell Nucleus/enzymology , Cells, Cultured , Chick Embryo , Chondrocytes/metabolism , Cytoplasm/enzymology , Epiphyses , Mitochondria/enzymology , Transformation, Genetic
16.
Anticancer Res ; 25(6B): 4135-9, 2005.
Article in English | MEDLINE | ID: mdl-16309208

ABSTRACT

BACKGROUND: The important metastatic potential of lung cancers is directly correlated with cell adhesion. Cell-extracellular matrix interactions occur in specialized structures termed focal adhesion (FA) complexes. Our aims were to investigate: (i) the expression of the major FA components in three lung cancer cell lines (non metastatic: A549, or metastatic: Calu-1 and H460), (ii) the modifications of the FA complex occurring when apoptosis was induced by Vinorelbine in the A549 cells. MATERIALS AND METHODS: The FA complex was characterized by flow cytometry, immunocytochemical staining and Western blot. RESULTS: The expressions of alpha3, betsaP, paxillin, p-paxillin and Grb2 varied depending on the histological type of the tumor. In apoptotic cells, the expressions of the PYK2, p-p38, PI3K and Grb2 adhesion proteins were increased. CONCLUSION: Our data suggest that these adhesion proteins may be implicated in the transduction of death signals.


Subject(s)
Carcinoma, Non-Small-Cell Lung/metabolism , Carcinoma, Non-Small-Cell Lung/pathology , Focal Adhesions/physiology , Lung Neoplasms/metabolism , Lung Neoplasms/pathology , Apoptosis/physiology , Cell Line, Tumor , Flow Cytometry , Focal Adhesion Kinase 2/biosynthesis , Focal Adhesion Protein-Tyrosine Kinases/biosynthesis , GRB2 Adaptor Protein/biosynthesis , Humans , Immunohistochemistry , Integrin alpha3/biosynthesis , Integrin beta1/biosynthesis , Paxillin/biosynthesis , Phosphatidylinositol 3-Kinases/biosynthesis
17.
Protein Expr Purif ; 44(2): 121-9, 2005 Dec.
Article in English | MEDLINE | ID: mdl-15946859

ABSTRACT

High-level recombinant expression of protein kinases in eukaryotic cells or Escherichia coli commonly gives products that are phosphorylated by autocatalysis or by the action of endogenous kinases. Here, we report that phosphorylation occurred on serine residues adjacent to hexahistidine affinity tags (His-tags) derived from several commercial expression vectors and fused to overexpressed kinases. The result was observed with a variety of recombinant kinases expressed in either insect cells or E. coli. Multiple phosphorylations of His-tagged full-length Aurora A, a protein serine/threonine kinase, were detected by mass spectrometry when it was expressed in insect cells in the presence of okadaic acid, a protein phosphatase inhibitor. Peptide mapping by liquid chromatography-mass spectrometry detected phosphorylations on all three serine residues in an N-terminal tag, alpha-N-acetyl-MHHHHHHSSGLPRGS. The same sequence was also phosphorylated, but only at a low level, when a His-tagged protein tyrosine kinase, Pyk2 was expressed in insect cells and activated in vitro. When catalytic domains of Aurora A and several other protein serine/threonine kinases were expressed in E. coli, serines in the affinity tag sequence GSSHHHHHHSSGLVPRGS were also variably phosphorylated. His-Aurora A with hyperphosphorylation of the serine residues in the tag aggregated and resisted thrombin-catalyzed removal of the tag. Treatment with alkaline phosphatase partly restored sensitivity to thrombin. The same His-tag sequence was also detected bearing alpha-N-d-gluconoylation in addition to multiple phosphorylations. The results show that histidine-tag sequences can receive complicated posttranslational modification, and that the hyperphosphorylation and resulting heterogeneity of the recombinant fusion proteins can interfere with downstream applications.


Subject(s)
Histidine/metabolism , Oligopeptides/metabolism , Protein Serine-Threonine Kinases/metabolism , Recombinant Proteins/biosynthesis , Serine/metabolism , Acetylation , Alkaline Phosphatase/chemistry , Animals , Aurora Kinases , Baculoviridae/genetics , Catalytic Domain/genetics , Cell Line , Escherichia coli/genetics , Escherichia coli/metabolism , Focal Adhesion Kinase 2/biosynthesis , Focal Adhesion Kinase 2/metabolism , Gas Chromatography-Mass Spectrometry , Gluconates/metabolism , Humans , Light , Molecular Weight , Okadaic Acid/pharmacology , Phosphorylation , Protein Processing, Post-Translational/drug effects , Protein Serine-Threonine Kinases/biosynthesis , Protein Serine-Threonine Kinases/chemistry , Protein Serine-Threonine Kinases/genetics , Recombinant Proteins/chemistry , Scattering, Radiation , Spodoptera , Thrombin/chemistry
18.
Beijing Da Xue Xue Bao Yi Xue Ban ; 37(3): 261-4, 2005 Jun 18.
Article in Chinese | MEDLINE | ID: mdl-15968315

ABSTRACT

OBJECTIVE: To investigate whether proline-rich tyrosine kinase-2 (Pyk2) is expressed differently in normal gastric mucosas and gastric carcinoma tissues and further to evaluate its significance. METHODS: Expressions of Pyk2 in 59 cases of normal gastric mucosas and 52 cases of gastric carcinoma tissues were analysed by immunohistochemical methods. RESULTS: Immunohistochemical studies showed that the positive rates of Pyk2 protein expression in normal gastric mucosas and gastric carcinoma tissues were 86.44% (51/59) and 19.23% (10/52) respectively. The difference between normal gastric mucosas and gastric carcinoma tissues was statistically significant (P<0.05). The positive rates of Pyk2 expression in highly differentiated gastric carcinoma and moderately/lowly differentiated gastric carcinoma were 47.37% (9/19) and 3.03% (1/33) respectively. Statistically significant differences (P<0.05) were observed in the levels of Pyk2 expression between highly differentiated gastric carcinoma and moderately/lowly differentiated gastric carcinoma. The positive rates of Pyk2 expression at different TNM stages gastric carcinoma were respectively: stage I 66.67% (6/9), stage II 30% (3/10), stage III 3.45% (1/29), stage IV 0% (0/4). The differences were statistically significant [(II+III+IV) v I, chi2=15.767, P<0.05]. CONCLUSION: In this study, we demonstrate that Pyk2 is expressed in normal gastric mucosas, whereas its expression declines significantly or almost disappears in gastric carcinoma tissues. The expression of Pyk2 progressively decreases with increasing grade of malignancy and TNM stages of gastric carcinoma. This phenomenon indicates that Pyk2 expression may be involved in the generation and development of gastric carcinoma.


Subject(s)
Adenocarcinoma/metabolism , Focal Adhesion Kinase 2/biosynthesis , Stomach Neoplasms/metabolism , Adenocarcinoma/pathology , Focal Adhesion Kinase 2/genetics , Gastric Mucosa/metabolism , Humans , Neoplasm Staging , Stomach Neoplasms/pathology
SELECTION OF CITATIONS
SEARCH DETAIL
...