Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 37
Filter
Add more filters










Publication year range
1.
Anal Biochem ; 630: 114313, 2021 10 01.
Article in English | MEDLINE | ID: mdl-34302798

ABSTRACT

Radiometric assays have widely been used for measuring protein kinase activity for decades. In addition, several non-radiometric kinase assay formats have been developed over the years, including luciferase-based and fluorescence-based assays. However, radiometric assays are still considered as the "gold standard" for protein kinase assays, because of their direct readout, high sensitivity, reproducibility, reliability, and very low background signals. These radiometric assays rely on P81 phosphocellulose paper to capture the phosphorylated substrate and wash out unreacted [γ-32P] ATP. However, recently the production of P81 was discontinued by the manufacturer, causing major concern within the protein kinase research community. The advantages of radiometric assays over other kinase assay methods call for an urgent alternative to the discontinued P81 paper. In this report, we demonstrate that the LSA-50 paper is a worthy alternative for radiometric protein kinase assays originally using P81 phosphocellulose paper.


Subject(s)
Cellulose/analogs & derivatives , Paper , Protein Kinases/analysis , Radiometry , Cellulose/chemistry , Cellulose/metabolism , Protein Kinases/metabolism
2.
ChemMedChem ; 16(14): 2158-2171, 2021 07 20.
Article in English | MEDLINE | ID: mdl-33829655

ABSTRACT

Protein kinase D (PKD) is a serine/threonine kinase family belonging to the Ca2+/calmodulin-dependent protein kinase group. Since its discovery two decades ago, many efforts have been put in elucidating PKD's structure, cellular role and functioning. The PKD family consists of three highly homologous isoforms: PKD1, PKD2 and PKD3. Accumulating cell-signaling research has evidenced that dysregulated PKD plays a crucial role in the pathogenesis of cardiac hypertrophy and several cancer types. These findings led to a broad interest in the design of small-molecule protein kinase D inhibitors. In this review, we present an extensive overview on the past and recent advances in the discovery and development of PKD inhibitors. The focus extends from broad-spectrum kinase inhibitors used in PKD signaling experiments to intentionally developed, bioactive PKD inhibitors. Finally, attention is paid to PKD inhibitors that have been identified as an off-target through large kinome screening panels.


Subject(s)
Drug Development , Protein Kinase C/antagonists & inhibitors , Protein Kinase Inhibitors/pharmacology , Animals , Dose-Response Relationship, Drug , Humans , Molecular Structure , Protein Kinase C/metabolism , Protein Kinase Inhibitors/chemical synthesis , Structure-Activity Relationship
3.
Eur J Med Chem ; 205: 112638, 2020 Nov 01.
Article in English | MEDLINE | ID: mdl-32835918

ABSTRACT

The multiple roles of protein kinase D (PKD) in various cancer hallmarks have been repeatedly reported. Therefore, the search for novel PKD inhibitors and their evaluation as antitumor agents has gained considerable attention. In this work, novel pyrazolo[3,4-d]pyrimidine based pan-PKD inhibitors with structural variety at position 1 were synthesized and evaluated for biological activity. Starting from 3-IN-PP1, a known PKD inhibitor with IC50 values in the range of 94-108 nM, compound 17m was identified with an improved biochemical inhibitory activity against PKD (IC50 = 17-35 nM). Subsequent cellular assays demonstrated that 3-IN-PP1 and 17m inhibited PKD-dependent cortactin phosphorylation. Furthermore, 3-IN-PP1 displayed potent anti-proliferative activity against PANC-1 cells. Finally, a screening against different cancer cell lines demonstrated that 3-IN-PP1 is a potent and versatile antitumoral agent.


Subject(s)
Drug Design , Protein Kinase C/antagonists & inhibitors , Protein Kinase Inhibitors/chemical synthesis , Protein Kinase Inhibitors/pharmacology , Pyrazoles/chemistry , Pyrimidines/chemical synthesis , Pyrimidines/pharmacology , Cell Line, Tumor , Cell Proliferation/drug effects , Chemistry Techniques, Synthetic , Humans , Inhibitory Concentration 50 , Phosphorylation/drug effects , Protein Kinase Inhibitors/chemistry , Pyrimidines/chemistry
4.
FEBS Lett ; 592(14): 2432-2443, 2018 07.
Article in English | MEDLINE | ID: mdl-29933512

ABSTRACT

The protein kinase D (PKD) family is regulated through multi-site phosphorylation, including autophosphorylation. For example, PKD displays in vivo autophosphorylation on Ser-742 (and Ser-738 in vitro) in the activation loop and Ser-910 in the C-tail (hPKD1 numbering). In this paper, we describe the surprising observation that PKD also displays in vitro autocatalytic activity towards a Tyr residue in the P + 1 loop of the activation segment. We define the molecular determinants for this unusual activity and identify a Cys residue (C705 in PKD1) in the catalytic loop as of utmost importance. In cells, PKD Tyr autophosphorylation is suppressed through the association of an inhibitory factor. Our findings provide important novel insights into PKD (auto)regulation.


Subject(s)
Protein Kinase C/chemistry , Protein Kinase C/metabolism , Tyrosine/metabolism , Amino Acid Sequence , Animals , COS Cells , Cells, Cultured , Chlorocebus aethiops , Drosophila , Enzyme Activation/genetics , HEK293 Cells , Homeostasis/genetics , Humans , Mutagenesis, Site-Directed , Phosphorylation/genetics , Protein Kinase C/genetics , Tyrosine/genetics
5.
Oxid Med Cell Longev ; 2018: 2138502, 2018.
Article in English | MEDLINE | ID: mdl-29854077

ABSTRACT

Oxidative stress is a condition that arises when cells are faced with levels of reactive oxygen species (ROS) that destabilize the homeostatic redox balance. High levels of ROS can cause damage to macromolecules including DNA, lipids, and proteins, eventually resulting in cell death. Moderate levels of ROS however serve as signaling molecules that can drive and potentiate several cellular phenotypes. Increased levels of ROS are associated with a number of diseases including neurological disorders and cancer. In cancer, increased ROS levels can contribute to cancer cell survival and proliferation via the activation of several signaling pathways. One of the downstream effectors of increased ROS is the protein kinase D (PKD) family of kinases. In this review, we will discuss the regulation and function of this family of ROS-activated kinases and describe their unique isoform-specific features, in terms of both kinase regulation and signaling output.


Subject(s)
Oxidative Stress/drug effects , Protein Isoforms/metabolism , Protein Kinase C/metabolism , Humans
6.
Oncogene ; 37(10): 1263-1278, 2018 03.
Article in English | MEDLINE | ID: mdl-29259300

ABSTRACT

Protein kinase D2 (PKD2) is a serine/threonine kinase that belongs to the PKD family of calcium-calmodulin kinases, which comprises three isoforms: PKD1, PKD2, and PKD3. PKD2 is activated by many stimuli including growth factors, phorbol esters, and G-protein-coupled receptor agonists. PKD2 participation to uncontrolled growth, survival, neovascularization, metastasis, and invasion has been documented in various tumor types including pancreatic, colorectal, gastric, hepatic, lung, prostate, and breast cancer, as well as glioma multiforme and leukemia. This review discusses the versatile functions of PKD2 from the perspective of cancer hallmarks as described by Hanahan and Weinberg. The PKD2 status, signaling pathways affected in different tumor types and the molecular mechanisms that lead to tumorigenesis and tumor progression are presented. The latest developments of small-molecule inhibitors selective for PKD/PKD2, as well as the need for further chemotherapies that prevent, slow down, or eliminate tumors are also discussed in this review.


Subject(s)
Neoplasms/genetics , Neoplasms/pathology , Protein Kinases/physiology , Animals , Cell Proliferation/genetics , Humans , Neoplasm Metastasis/genetics , Neovascularization, Pathologic/genetics , Neovascularization, Pathologic/pathology , Protein Kinase D2 , Protein Kinases/genetics , Signal Transduction/genetics
7.
Medchemcomm ; 8(3): 640-646, 2017 Mar 01.
Article in English | MEDLINE | ID: mdl-28890776

ABSTRACT

In this study, we set out to rationally optimize PKD inhibitors based on the pyrazolo[3,4-d]pyrimidine scaffold. The lead compound for this study was 1-NM-PP1, which was previously found by us and others to inhibit PKD. In our screening we identified one compound (3-IN-PP1) displaying a 10-fold increase in potency over 1-NM-PP1, opening new possibilities for specific protein kinase inhibitors for kinases that show sensitivity towards pyrazolo[3,4-d]pyrimidine derived compounds. Interestingly the observed SAR was not in complete agreement with the commonly observed binding mode where the pyrazolo[3,4-d]pyrimidine compounds are bound in a similar fashion as PKD's natural ligand ATP. Therefore we suggest an alternate binding mode where the compounds are flipped 180 degrees. This possible alternate binding mode for pyrazolo[3,4-d]pyrimidine based compounds could pave the way for a new class of specific protein kinase inhibitors for kinases sensitive towards pyrazolo[3,4-d]pyrmidines.

8.
Mol Metab ; 6(7): 715-724, 2017 07.
Article in English | MEDLINE | ID: mdl-28702327

ABSTRACT

OBJECTIVE: A potential strategy to treat obesity - and the associated metabolic consequences - is to increase energy expenditure. This could be achieved by stimulating thermogenesis through activation of brown adipose tissue (BAT) and/or the induction of browning of white adipose tissue (WAT). Over the last years, it has become clear that several metalloproteinases play an important role in adipocyte biology. Here, we investigated the potential role of ADAMTS5. METHODS: Mice deficient in ADAMTS5 (Adamts5-/-) and wild-type (Adamts5+/+) littermates were kept on a standard of Western-type diet for 15 weeks. Energy expenditure and heat production was followed by indirect calorimetry. To activate thermogenesis, mice were treated with the ß3-adrenergic receptor (ß3-AR) agonist CL-316,243 or alternatively, exposed to cold for 2 weeks. RESULTS: Compared to Adamts5+/+ mice, Adamts5-/- mice have significantly more interscapular BAT and marked browning of their subcutaneous (SC) WAT. Thermogenic pathway analysis indicated, in the absence of ADAMTS5, enhanced ß3-AR signaling via activation of the cAMP response element-binding protein (CREB). Additional ß3-AR stimulation with CL-316,243 promoted browning of WAT in Adamts5+/+ mice but had no additive effect in Adamts5-/- mice. However, cold exposure induced more pronounced browning of WAT in Adamts5-/- mice. CONCLUSIONS: These data indicate that ADAMTS5 plays a functional role in development of BAT and browning of WAT. Hence, selective targeting of ADAMTS5 could provide a novel therapeutic strategy for treatment/prevention of obesity and metabolic diseases.


Subject(s)
ADAMTS5 Protein/genetics , Adipose Tissue, Brown/metabolism , Adipose Tissue, White/metabolism , Cyclic AMP Response Element-Binding Protein/metabolism , ADAMTS5 Protein/deficiency , ADAMTS5 Protein/metabolism , Adipose Tissue, White/drug effects , Adrenergic beta-3 Receptor Agonists/pharmacology , Animals , Cells, Cultured , Dioxoles/pharmacology , Energy Metabolism , Male , Mice , Mice, Inbred C57BL , Thermogenesis
9.
Sci Rep ; 7(1): 887, 2017 04 20.
Article in English | MEDLINE | ID: mdl-28428613

ABSTRACT

Protein kinases are essential molecules in life and their crucial function requires tight regulation. Many kinases are regulated via phosphorylation within their activation loop. This loop is embedded in the activation segment, which additionally contains the Mg2+ binding loop and a P + 1 loop that is important in substrate binding. In this report, we identify Abl-mediated phosphorylation of a highly conserved Tyr residue in the P + 1 loop of protein kinase D2 (PKD2) during oxidative stress. Remarkably, we observed that the three human PKD isoforms display very different degrees of P + 1 loop Tyr phosphorylation and we identify one of the molecular determinants for this divergence. This is paralleled by a different activation mechanism of PKD1 and PKD2 during oxidative stress. Tyr phosphorylation in the P + 1 loop of PKD2 increases turnover for Syntide-2, while substrate specificity and the role of PKD2 in NF-κB signaling remain unaffected. Importantly, Tyr to Phe substitution renders the kinase inactive, jeopardizing its use as a non-phosphorylatable mutant. Since large-scale proteomics studies identified P + 1 loop Tyr phosphorylation in more than 70 Ser/Thr kinases in multiple conditions, our results do not only demonstrate differential regulation/function of PKD isoforms under oxidative stress, but also have implications for kinase regulation in general.


Subject(s)
Oxidative Stress , Protein Kinase C/metabolism , Protein Processing, Post-Translational , Conserved Sequence , HEK293 Cells , HeLa Cells , Humans , Intercellular Signaling Peptides and Proteins , Isoenzymes/chemistry , Isoenzymes/genetics , Isoenzymes/metabolism , NF-kappa B/metabolism , Peptides/metabolism , Phosphorylation , Protein Domains , Protein Kinase C/chemistry , Protein Kinase C/genetics , Tyrosine/genetics , Tyrosine/metabolism
10.
J Cell Sci ; 129(12): 2416-29, 2016 06 15.
Article in English | MEDLINE | ID: mdl-27179075

ABSTRACT

Dynamic regulation of cell-cell adhesion by the coordinated formation and dissolution of E-cadherin-based adherens junctions is crucial for tissue homeostasis. The actin-binding protein cortactin interacts with E-cadherin and enables F-actin accumulation at adherens junctions. Here, we were interested to study the broader functional interactions of cortactin in adhesion complexes. In line with literature, we demonstrate that cortactin binds to E-cadherin, and that a posttranslational modification of cortactin, RhoA-induced phosphorylation by protein kinase D1 (PKD1; also known as PRKD1) at S298, impairs adherens junction assembly and supports their dissolution. Two new S298-phosphorylation-dependent interactions were also identified, namely, that phosphorylation of cortactin decreases its interaction with ß-catenin and the actin-binding protein vinculin. In addition, binding of vinculin to ß-catenin, as well as linkage of vinculin to F-actin, are also significantly compromised upon phosphorylation of cortactin. Accordingly, we found that regulation of cell-cell adhesion by phosphorylation of cortactin downstream of RhoA and PKD1 is vitally dependent on vinculin-mediated protein interactions. Thus, cortactin, unexpectedly, is an important integration node for the dynamic regulation of protein complexes during breakdown and formation of adherens junctions.


Subject(s)
Cadherins/metabolism , Cortactin/metabolism , TRPP Cation Channels/metabolism , Actin Cytoskeleton/metabolism , Adherens Junctions/metabolism , Animals , Antigens, CD , Caco-2 Cells , Cell Adhesion , Colon/metabolism , Epithelium/metabolism , HEK293 Cells , Humans , Mice, Inbred C57BL , Mice, Transgenic , Models, Biological , Phosphorylation , Phosphoserine/metabolism , Vinculin/metabolism
11.
Cell Microbiol ; 17(12): 1797-810, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26096820

ABSTRACT

Trypanosoma cruzi extracellular amastigotes (EAs) display unique mechanisms for cell invasion that are highly dependent on host actin filaments. Protein kinase D1 (PKD1) phosphorylates and modulates the activity of cortactin, a key regulator of actin dynamics. We evaluated the role of host cortactin and PKD1 in actin filament dynamics during HeLa cell invasion by EAs. Host cortactin, PKD1 and actin are recruited by EAs based on experiments in fixed and live cells by time lapse confocal microscopy. EAs trigger PKD1 and extracellular signal-regulated kinase 1/2 activation, but not Src family kinases, and selectively phosphorylate cortactin. Heat-killed EAs and non-infective epimastigotes both triggered distinct host responses and did not recruit the molecules studied herein. EA invasion was influenced by depletion or overexpression of host cortactin and PKD1, respectively, suggesting the involvement of both proteins in this event. Collectively, these results show new host cell mechanisms subverted during EA internalization into non-phagocytic cells.


Subject(s)
Actins/metabolism , Cortactin/metabolism , Endocytosis , Host-Pathogen Interactions , Protein Kinase C/metabolism , Signal Transduction , Trypanosoma cruzi/physiology , Epithelial Cells/parasitology , Epithelial Cells/physiology , HeLa Cells , Humans , Microscopy, Confocal , Sequence Analysis, DNA , Time-Lapse Imaging
12.
Cancer Res ; 74(23): 7125-36, 2014 Dec 01.
Article in English | MEDLINE | ID: mdl-25297628

ABSTRACT

The kinase PRKD2 (protein kinase D) is a crucial regulator of tumor cell-endothelial cell communication in gastrointestinal tumors and glioblastomas, but its mechanistic contributions to malignant development are not understood. Here, we report that the oncogenic chaperone HSP90 binds to and stabilizes PRKD2 in human cancer cells. Pharmacologic inhibition of HSP90 with structurally divergent small molecules currently in clinical development triggered proteasome-dependent degradation of PRKD2, augmenting apoptosis in human cancer cells of various tissue origins. Conversely, ectopic expression of PRKD2 protected cancer cells from the apoptotic effects of HSP90 abrogation, restoring blood vessel formation in two preclinical models of solid tumors. Mechanistic studies revealed that PRKD2 is essential for hypoxia-induced accumulation of hypoxia-inducible factor-1α (HIF1α) and activation of NF-κB in tumor cells. Notably, ectopic expression of PRKD2 was able to partially restore HIF1α and secreted VEGF-A levels in hypoxic cancer cells treated with HSP90 inhibitors. Taken together, our findings indicate that signals from hypoxia and HSP90 pathways are interconnected and funneled by PRKD2 into the NF-κB/VEGF-A signaling axis to promote tumor angiogenesis and tumor growth.


Subject(s)
HSP90 Heat-Shock Proteins/metabolism , Neovascularization, Pathologic/metabolism , Protein Kinase C/metabolism , Animals , Apoptosis/physiology , Cell Hypoxia/physiology , Cell Line, Tumor , Female , HCT116 Cells , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Mice , Mice, Nude , NF-kappa B/metabolism , Proteasome Endopeptidase Complex/metabolism , Signal Transduction/physiology , Vascular Endothelial Growth Factor A/metabolism
13.
Proc Natl Acad Sci U S A ; 111(18): E1872-9, 2014 May 06.
Article in English | MEDLINE | ID: mdl-24753582

ABSTRACT

Dissemination of carcinoma cells requires the pericellular degradation of the extracellular matrix, which is mediated by membrane type 1-matrix metalloproteinase (MT1-MMP). In this article, we report a co-up-regulation and colocalization of MT1-MMP and atypical protein kinase C iota (aPKCι) in hormone receptor-negative breast tumors in association with a higher risk of metastasis. Silencing of aPKC in invasive breast-tumor cell lines impaired the delivery of MT1-MMP from late endocytic storage compartments to the surface and inhibited matrix degradation and invasion. We provide evidence that aPKCι, in association with MT1-MMP-containing endosomes, phosphorylates cortactin, which is present in F-actin-rich puncta on MT1-MMP-positive endosomes and regulates cortactin association with the membrane scission protein dynamin-2. Thus, cell line-based observations and clinical data reveal the concerted activity of aPKC, cortactin, and dynamin-2, which control the trafficking of MT1-MMP from late endosome to the plasma membrane and play an important role in the invasive potential of breast-cancer cells.


Subject(s)
Breast Neoplasms/metabolism , Isoenzymes/metabolism , Matrix Metalloproteinase 14/metabolism , Protein Kinase C/metabolism , Adenocarcinoma/genetics , Adenocarcinoma/metabolism , Adenocarcinoma/pathology , Adult , Aged , Biological Transport, Active , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Carcinoma, Ductal, Breast/genetics , Carcinoma, Ductal, Breast/metabolism , Carcinoma, Ductal, Breast/pathology , Cell Line, Tumor , Cortactin/metabolism , Cytoplasmic Granules/metabolism , Disease Progression , Dynamin II/metabolism , Endosomes/metabolism , Extracellular Matrix/metabolism , Female , Humans , Isoenzymes/antagonists & inhibitors , Isoenzymes/genetics , Matrix Metalloproteinase 14/genetics , Middle Aged , Neoplasm Invasiveness , Phosphorylation , Protein Kinase C/antagonists & inhibitors , Protein Kinase C/genetics , RNA Interference , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA, Neoplasm/genetics , RNA, Neoplasm/metabolism , RNA, Small Interfering/genetics , Up-Regulation
14.
Am J Physiol Heart Circ Physiol ; 303(3): H323-31, 2012 Aug 01.
Article in English | MEDLINE | ID: mdl-22636676

ABSTRACT

Cardiac myosin-binding protein C (cMyBP-C) is involved in the regulation of cardiac myofilament contraction. Recent evidence showed that protein kinase D (PKD) is one of the kinases that phosphorylate cMyBP-C. However, the mechanism by which PKD-induced cMyBP-C phosphorylation affects cardiac contractile responses is not known. Using immunoprecipitation, we showed that, in contracting cardiomyocytes, PKD binds to cMyBP-C and phosphorylates it at Ser(315). The effect of PKD-mediated phosphorylation of cMyBP-C on cardiac myofilament function was investigated in permeabilized ventricular myocytes, isolated from wild-type (WT) and from cMyBP-C knockout (KO) mice, incubated in the presence of full-length active PKD. In WT myocytes, PKD increased both myofilament Ca(2+) sensitivity (pCa(50)) and maximal Ca(2+)-activated tension of contraction (T(max)). In cMyBP-C KO skinned myocytes, PKD increased pCa(50) but did not alter T(max). This suggests that cMyBP-C is not involved in PKD-mediated sensitization of myofilaments to Ca(2+) but is essential for PKD-induced increase in T(max). Furthermore, the phosphorylation of both PKD-Ser(916) and cMyBP-C-Ser(315) was contraction frequency-dependent, suggesting that PKD-mediated cMyBP-C phosphorylation is operational primarily during periods of increased contractile activity. Thus, during high contraction frequency, PKD facilitates contraction of cardiomyocytes by increasing Ca(2+) sensitivity and by an increased T(max) through phosphorylation of cMyBP-C.


Subject(s)
Carrier Proteins/metabolism , Excitation Contraction Coupling , Myocardial Contraction , Myocytes, Cardiac/enzymology , Protein Kinase C/metabolism , Adrenergic beta-Antagonists/pharmacology , Animals , Carrier Proteins/genetics , Electric Stimulation , Excitation Contraction Coupling/drug effects , Immunoprecipitation , Male , Mice , Mice, Knockout , Myocardial Contraction/drug effects , Myocytes, Cardiac/drug effects , Myofibrils/enzymology , Phosphorylation , Protein Binding , Rats , Rats, Inbred Lew , Serine
15.
J Biol Chem ; 287(12): 9473-83, 2012 Mar 16.
Article in English | MEDLINE | ID: mdl-22228765

ABSTRACT

The members of the protein kinase D (PKD) family of serine/threonine kinases are major targets for tumor-promoting phorbol esters, G protein-coupled receptors, and activated protein kinase C isoforms (PKCs). The expanding list of cellular processes in which PKDs exert their function via phosphorylation of various substrates include proliferation, apoptosis, migration, angiogenesis, and vesicle trafficking. Therefore, identification of novel PKD substrates is necessary to understand the profound role of this kinase family in signal transduction. Here, we show that rhotekin, an effector of RhoA GTPase, is a novel substrate of PKD. We identified Ser-435 in rhotekin as the potential site targeted by PKD in vivo. Expression of a phosphomimetic S435E rhotekin mutant resulted in an increase of endogenous active RhoA GTPase levels. Phosphorylation of rhotekin by PKD2 modulates the anchoring of the RhoA in the plasma membrane. Consequently, the S435E rhotekin mutant displayed enhanced stress fiber formation when expressed in serum-starved fibroblasts. Our data thus identify a novel role of PKD as a regulator of RhoA activity and actin stress fiber formation through phosphorylation of rhotekin.


Subject(s)
Intracellular Signaling Peptides and Proteins/metabolism , Protein Kinase C/metabolism , rhoA GTP-Binding Protein/metabolism , Amino Acid Motifs , Animals , Apoptosis Regulatory Proteins , GTP-Binding Proteins , HEK293 Cells , Humans , Intracellular Signaling Peptides and Proteins/chemistry , Intracellular Signaling Peptides and Proteins/genetics , Mice , NIH 3T3 Cells , Phosphorylation , Protein Kinase C/genetics , rhoA GTP-Binding Protein/genetics
16.
Neuro Oncol ; 13(7): 710-24, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21727210

ABSTRACT

Glioblastoma multiforme, a highly aggressive tumor of the central nervous system, has a dismal prognosis that is due in part to its resistance to radio- and chemotherapy. The protein kinase C (PKC) family of serine threonine kinases has been implicated in the formation and proliferation of glioblastoma multiforme. Members of the protein kinase D (PKD) family, which consists of PKD1, -2 and, -3, are prominent downstream targets of PKCs and could play a major role in glioblastoma growth. PKD2 was highly expressed in both low-grade and high-grade human gliomas. The number of PKD2-positive tumor cells increased with glioma grading (P < .001). PKD2 was also expressed in CD133-positive glioblastoma stem cells and various glioblastoma cell lines in which the kinase was found to be constitutively active. Inhibition of PKDs by pharmacological inhibitors resulted in substantial inhibition of glioblastoma proliferation. Furthermore, specific depletion of PKD2 by siRNA resulted in a marked inhibition of anchorage-dependent and -independent proliferation and an accumulation of glioblastoma cells in G0/G1, accompanied by a down-regulation of cyclin D1 expression. In addition, PKD2-depleted glioblastoma cells exhibited substantially reduced tumor formation in vivo on chicken chorioallantoic membranes. These findings identify PKD2 as a novel mediator of glioblastoma cell growth in vitro and in vivo and thereby as a potential therapeutic target for this devastating disease.


Subject(s)
Brain Neoplasms/pathology , Brain/enzymology , Glioblastoma/pathology , TRPP Cation Channels/metabolism , Animals , Apoptosis , Blotting, Western , Brain Neoplasms/enzymology , Cell Cycle , Cell Proliferation , Chickens , Chorioallantoic Membrane/metabolism , Cyclin D1/metabolism , Glioblastoma/enzymology , Humans , Immunoenzyme Techniques , RNA, Small Interfering/genetics , TRPP Cation Channels/antagonists & inhibitors , TRPP Cation Channels/genetics
17.
Gut ; 59(10): 1316-30, 2010 Oct.
Article in English | MEDLINE | ID: mdl-20732914

ABSTRACT

BACKGROUND: Tumour angiogenesis is crucially dependent on the communication between the tumour and the associated endothelium. Protein kinase D (PKD) isoenzymes mediate vascular endothelial growth factor-A (VEGF-A) induced endothelial cell proliferation and migration and are also highly expressed in various tumours. AIM: To examine the role of PKDs for tumour proliferation and angiogenesis selectively in pancreatic and gastric tumours and in tumour-associated endothelium in vitro and in vivo. METHODS: PKD2 expression in human tumours was determined by immunohistochemistry. The effect of PKD2 depletion in endothelial cells by siRNAs was examined in sprouting assays, the chorioallantois model (CAM) and tumour xenografts. In murine endothelium in vivo PKD2 was knocked-down by splice switching oligonucleotides. Human PKD2 was depleted in xenografts by siRNAs and PKD2-miRs. PKD2 activation by hypoxia and its role for hypoxia-induced NR4/TR3- and VEGF-A promoter activity, expression and secretion was investigated in cell lines. RESULTS: PKD2 is expressed in gastrointestinal tumours and in the tumour-associated endothelium. Tumour growth and angiogenesis in the CAM and in tumour xenografts require PKD expression in endothelial cells. Conversely, hypoxia activates PKD2 in pancreatic cancer cells and PKD2 was identified as the major mediator of hypoxia-stimulated VEGF-A promoter activity, expression and secretion in tumour cells. PKD2 depletion in pancreatic tumours inhibited tumour-driven blood vessel formation and tumour growth in the CAM and in orthotopic pancreatic cancer xenografts. CONCLUSION: PKD2 regulates hypoxia-induced VEGF-A expression/secretion by tumour cells and VEGF-A stimulated blood vessel formation. PKD2 is a novel, essential mediator of tumour cell-endothelial cell communication and a promising therapeutic target to inhibit angiogenesis in gastrointestinal cancers.


Subject(s)
Gastrointestinal Neoplasms/pathology , Protein Kinases/physiology , Animals , Cell Communication/physiology , Chick Embryo , Chorioallantoic Membrane/blood supply , Chorioallantoic Membrane/enzymology , Coculture Techniques , Endothelial Cells/pathology , Endothelium, Vascular/enzymology , Gastrointestinal Neoplasms/blood supply , Gastrointestinal Neoplasms/enzymology , Gene Knockdown Techniques , Humans , Mice , Mice, Nude , Neoplasm Transplantation , Neovascularization, Pathologic/enzymology , Neovascularization, Pathologic/pathology , Pancreatic Neoplasms/blood supply , Pancreatic Neoplasms/enzymology , Pancreatic Neoplasms/pathology , Protein Kinase D2 , Protein Kinases/genetics , Protein Kinases/metabolism , Transplantation, Heterologous , Tumor Cells, Cultured , Vascular Endothelial Growth Factor A/physiology
18.
J Biol Chem ; 285(24): 18672-83, 2010 Jun 11.
Article in English | MEDLINE | ID: mdl-20363754

ABSTRACT

We here identify protein kinase D (PKD) as an upstream regulator of the F-actin-binding protein cortactin and the Arp actin polymerization machinery. PKD phosphorylates cortactin in vitro and in vivo at serine 298 thereby generating a 14-3-3 binding motif. In vitro, a phosphorylation-deficient cortactin-S298A protein accelerated VCA-Arp-cortactin-mediated synergistic actin polymerization and showed reduced F-actin binding, indicative of enhanced turnover of nucleation complexes. In vivo, cortactin co-localized with the nucleation promoting factor WAVE2, essential for lamellipodia extension, in the actin polymerization zone in Heregulin-treated MCF-7 cells. Using a 3-dye FRET-based approach we further demonstrate that WAVE2-Arp and cortactin prominently interact at these structures. Accordingly, cortactin-S298A significantly enhanced lamellipodia extension and directed cell migration. Our data thus unravel a previously unrecognized mechanism by which PKD controls cancer cell motility.


Subject(s)
Actins/chemistry , Cortactin/chemistry , Protein Kinase C/chemistry , Amino Acid Motifs , Cell Line , Cell Line, Tumor , Cell Movement , Fluorescence Resonance Energy Transfer , Humans , Immunohistochemistry/methods , Neuregulin-1/chemistry , Phosphorylation , Protein Binding , Wiskott-Aldrich Syndrome Protein Family/chemistry
19.
Int Immunopharmacol ; 10(3): 277-83, 2010 Mar.
Article in English | MEDLINE | ID: mdl-19932769

ABSTRACT

Protein kinase D (PKD) is a serine-threonine kinase involved in the activation of a variety of cells. In mast cells, activation of PKD by cross-linking of high affinity receptor for IgE (FcepsilonRI) has been reported, but little is known for its effects on cytokine production. We investigated the roles of PKD on FcepsilonRI-induced activator protein-1 (AP-1) activation and proinflammatory cytokine productions in mast cells. Pharmacological inhibition of PKD strongly inhibited production of interleukin (IL)-13 and tumor necrosis factor (TNF)-alpha induced by FcepsilonRI stimulation, and the overexpression of PKD significantly increased the IL-13 and TNF-alpha production. Reporter assay revealed that the overexpression of PKD enhanced FcepsilonRI-induced IL-13 promoter activation, and that the 5'-flanking region of IL-13 gene from positions -110 to -52 was under the regulation of PKD. The overexpression of PKD enhanced the induction of AP-1 luciferase activity by FcepsilonRI stimulation, while it had no effect on luciferase activities dependent upon NF-kappaB and NF-AT activated by FcepsilonRI stimulation. In EMSA, c-Jun and c-Fos appear to be the major components of AP-1 complexes activated by FcepsilonRI stimulation. Moreover the overexpression of PKD strongly enhanced the phosphorylation of both c-Jun and c-Fos following FcepsilonRI stimulation. Although stress-activated protein kinase/c-Jun N-terminal kinase (JNK) is known to be an important regulator for c-Jun phosphorylation and AP-1 activation, overexpression and inhibition of PKD had no effects on JNK phosphorylation. These results suggest that PKD may play a pivotal role in FcepsilonRI-induced cytokine production in mast cells through the activation of c-Jun, c-Fos, and AP-1.


Subject(s)
Cytokines/biosynthesis , Mast Cells/metabolism , Protein Kinase C/metabolism , Receptors, IgE/agonists , Transcription Factor AP-1/metabolism , Animals , Blotting, Western , DNA/genetics , Electrophoretic Mobility Shift Assay , Enzyme Activation/drug effects , Enzyme Inhibitors/pharmacology , Enzyme-Linked Immunosorbent Assay , Genetic Vectors , Indicators and Reagents , Interleukin-13/biosynthesis , Interleukin-13/genetics , Mast Cells/drug effects , Phosphorylation , Plasmids/genetics , Protein Kinase C/biosynthesis , Rats , Retroviridae/genetics , Signal Transduction/drug effects , Transfection , Tumor Necrosis Factor-alpha/biosynthesis
20.
Cell Signal ; 21(2): 282-92, 2009 Feb.
Article in English | MEDLINE | ID: mdl-19000756

ABSTRACT

EVL-I is a splice variant of EVL (Ena/VASP like protein), whose in vivo function and regulation are still poorly understood. We found that Protein Kinase D (PKD) interacts in vitro and in vivo with EVL-I and phosphorylates EVL-I in a 21 amino acid alternately-included insert in the EVH2 domain. Following knockdown of the capping protein CPbeta and spreading on laminin, phosphorylated EVL-I can support filopodia formation and the phosphorylated EVL-I is localized at filopodial tips. Furthermore, we found that the lamellipodial localization of EVL-I is unaffected by phosphorylation, but that impairment of EVL-I phosphorylation is associated with ruffling of lamellipodia upon PDBu stimulation. Besides the lamellipodial and filopodial localization of phosphorylated EVL-I in fibroblasts, we determined that EVL-I is hyperphosphorylated and localized in the cell-cell contacts of certain breast cancer cells and mouse embryo keratinocytes. Taken together, our results show that phosphorylated EVL-I is present in lamellipodia, filopodia and cell-cell contacts and suggest the existence of signaling pathways that may affect EVL-I via phosphorylation of its EVH2 domain.


Subject(s)
Cell Adhesion Molecules/metabolism , Protein Kinase C/metabolism , Alternative Splicing , Amino Acid Sequence , Animals , Cell Line , Cells, Cultured , Cytoskeleton , Fibroblasts/metabolism , Humans , Immunoprecipitation , Mice , Phorbol Esters/pharmacology , Phosphorylation , Pseudopodia/metabolism , Pseudopodia/ultrastructure
SELECTION OF CITATIONS
SEARCH DETAIL
...