Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 37
Filtrar
1.
Anal Biochem ; 630: 114313, 2021 10 01.
Artigo em Inglês | MEDLINE | ID: mdl-34302798

RESUMO

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.


Assuntos
Celulose/análogos & derivados , Papel , Proteínas Quinases/análise , Radiometria , Celulose/química , Celulose/metabolismo , Proteínas Quinases/metabolismo
2.
J Cell Sci ; 129(12): 2416-29, 2016 06 15.
Artigo em Inglês | MEDLINE | ID: mdl-27179075

RESUMO

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.


Assuntos
Caderinas/metabolismo , Cortactina/metabolismo , Canais de Cátion TRPP/metabolismo , Citoesqueleto de Actina/metabolismo , Junções Aderentes/metabolismo , Animais , Antígenos CD , Células CACO-2 , Adesão Celular , Colo/metabolismo , Epitélio/metabolismo , Células HEK293 , Humanos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Modelos Biológicos , Fosforilação , Fosfosserina/metabolismo , Vinculina/metabolismo
3.
Proc Natl Acad Sci U S A ; 111(18): E1872-9, 2014 May 06.
Artigo em Inglês | MEDLINE | ID: mdl-24753582

RESUMO

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.


Assuntos
Neoplasias da Mama/metabolismo , Isoenzimas/metabolismo , Metaloproteinase 14 da Matriz/metabolismo , Proteína Quinase C/metabolismo , Adenocarcinoma/genética , Adenocarcinoma/metabolismo , Adenocarcinoma/patologia , Adulto , Idoso , Transporte Biológico Ativo , Neoplasias da Mama/genética , Neoplasias da Mama/patologia , Carcinoma Ductal de Mama/genética , Carcinoma Ductal de Mama/metabolismo , Carcinoma Ductal de Mama/patologia , Linhagem Celular Tumoral , Cortactina/metabolismo , Grânulos Citoplasmáticos/metabolismo , Progressão da Doença , Dinamina II/metabolismo , Endossomos/metabolismo , Matriz Extracelular/metabolismo , Feminino , Humanos , Isoenzimas/antagonistas & inibidores , Isoenzimas/genética , Metaloproteinase 14 da Matriz/genética , Pessoa de Meia-Idade , Invasividade Neoplásica , Fosforilação , Proteína Quinase C/antagonistas & inibidores , Proteína Quinase C/genética , Interferência de RNA , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , RNA Neoplásico/genética , RNA Neoplásico/metabolismo , RNA Interferente Pequeno/genética , Regulação para Cima
4.
Cell Microbiol ; 17(12): 1797-810, 2015 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-26096820

RESUMO

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.


Assuntos
Actinas/metabolismo , Cortactina/metabolismo , Endocitose , Interações Hospedeiro-Patógeno , Proteína Quinase C/metabolismo , Transdução de Sinais , Trypanosoma cruzi/fisiologia , Células Epiteliais/parasitologia , Células Epiteliais/fisiologia , Células HeLa , Humanos , Microscopia Confocal , Análise de Sequência de DNA , Imagem com Lapso de Tempo
5.
J Biol Chem ; 287(12): 9473-83, 2012 Mar 16.
Artigo em Inglês | MEDLINE | ID: mdl-22228765

RESUMO

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.


Assuntos
Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Proteína Quinase C/metabolismo , Proteína rhoA de Ligação ao GTP/metabolismo , Motivos de Aminoácidos , Animais , Proteínas Reguladoras de Apoptose , Proteínas de Ligação ao GTP , Células HEK293 , Humanos , Peptídeos e Proteínas de Sinalização Intracelular/química , Peptídeos e Proteínas de Sinalização Intracelular/genética , Camundongos , Células NIH 3T3 , Fosforilação , Proteína Quinase C/genética , Proteína rhoA de Ligação ao GTP/genética
6.
Am J Physiol Heart Circ Physiol ; 303(3): H323-31, 2012 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-22636676

RESUMO

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.


Assuntos
Proteínas de Transporte/metabolismo , Acoplamento Excitação-Contração , Contração Miocárdica , Miócitos Cardíacos/enzimologia , Proteína Quinase C/metabolismo , Antagonistas Adrenérgicos beta/farmacologia , Animais , Proteínas de Transporte/genética , Estimulação Elétrica , Acoplamento Excitação-Contração/efeitos dos fármacos , Imunoprecipitação , Masculino , Camundongos , Camundongos Knockout , Contração Miocárdica/efeitos dos fármacos , Miócitos Cardíacos/efeitos dos fármacos , Miofibrilas/enzimologia , Fosforilação , Ligação Proteica , Ratos , Ratos Endogâmicos Lew , Serina
7.
J Biol Chem ; 285(24): 18672-83, 2010 Jun 11.
Artigo em Inglês | MEDLINE | ID: mdl-20363754

RESUMO

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.


Assuntos
Actinas/química , Cortactina/química , Proteína Quinase C/química , Motivos de Aminoácidos , Linhagem Celular , Linhagem Celular Tumoral , Movimento Celular , Transferência Ressonante de Energia de Fluorescência , Humanos , Imuno-Histoquímica/métodos , Neuregulina-1/química , Fosforilação , Ligação Proteica , Família de Proteínas da Síndrome de Wiskott-Aldrich/química
8.
J Exp Med ; 201(5): 793-804, 2005 Mar 07.
Artigo em Inglês | MEDLINE | ID: mdl-15738054

RESUMO

The molecular basis of thymocyte negative selection, a crucial mechanism in establishing central tolerance, is not yet resolved. Histone deacetylases (HDACs) have emerged as key transcriptional regulators in several major developmental programs. Recently, we showed that the class IIa member, HDAC7, regulates negative selection by repressing expression of Nur77, an orphan nuclear receptor involved in antigen-induced apoptosis of thymocytes. Engagement of the T cell receptor (TCR) alleviates this repression through phosphorylation-dependent nuclear exclusion of HDAC7. However, the identity of the TCR-activated kinase that phosphorylates and inactivates HDAC7 was still unknown. Here, we demonstrate that TCR-induced nuclear export of HDAC7 and Nur77 expression is mediated by activation of protein kinase D (PKD). Indeed, active PKD stimulates HDAC7 nuclear export and Nur77 expression. In contrast, inhibition of PKD prevents TCR-mediated nuclear exclusion of HDAC7 and associated Nur77 activation. Furthermore, we show that HDAC7 is an interaction partner and a substrate for PKD. We identify four serine residues in the NH(2) terminus of HDAC7 as targets for PKD. More importantly, a mutant of HDAC7 specifically deficient in phosphorylation by PKD, inhibits TCR-mediated apoptosis of T cell hybridomas. These findings indicate that PKD is likely to play a key role in the signaling pathways controlling negative selection.


Assuntos
Apoptose , Proteínas de Ligação a DNA/metabolismo , Regulação da Expressão Gênica , Histona Desacetilases/metabolismo , Proteína Quinase C/metabolismo , Receptores de Antígenos de Linfócitos T/metabolismo , Receptores Citoplasmáticos e Nucleares/metabolismo , Receptores de Esteroides/metabolismo , Fatores de Transcrição/metabolismo , Transporte Ativo do Núcleo Celular/efeitos dos fármacos , Sequência de Aminoácidos , Linhagem Celular , Ativação Enzimática , Histona Desacetilases/química , Histona Desacetilases/genética , Humanos , Dados de Sequência Molecular , Mutação/genética , Membro 1 do Grupo A da Subfamília 4 de Receptores Nucleares , Ésteres de Forbol/farmacologia , Fosforilação , Regiões Promotoras Genéticas/genética , Inibidores de Proteínas Quinases/farmacologia , Alinhamento de Sequência , Transdução de Sinais , Timo/metabolismo
9.
EMBO J ; 26(22): 4619-33, 2007 Nov 14.
Artigo em Inglês | MEDLINE | ID: mdl-17962809

RESUMO

Protein kinase D2 (PKD2), a member of the PKD family of serine/threonine kinases, is localized in various subcellular compartments including the nucleus where the kinase accumulates upon activation of G-protein-coupled receptors. We define three critical post-translational modifications required for nuclear accumulation of PKD2 in response to activation of the CCK2 receptor (CCK2R): phosphorylation at Ser706 and Ser710 within the activation loop by PKC eta leading to catalytic activity and phosphorylation at Ser244 within the zinc-finger domain, which is crucial for blocking nuclear export of active PKD2 by preventing its interaction with the Crm-1 export machinery. We identify CK1delta and epsilon as upstream activated kinases by CCK2R that phosphorylate PKD2 at Ser244. Moreover, nuclear accumulation of active PKD2 is a prerequisite for efficient phosphorylation of its nuclear substrate, HDAC7. Only nuclear, active PKD2 mediates CCK2R-induced HDAC7 phosphorylation and Nur77 expression. Thus, we define a novel, compartment-specific signal transduction pathway downstream of CCK2R that phosphorylates PKD2 at three specific sites, results in nuclear accumulation of the active kinase and culminates in efficient phosphorylation of nuclear PKD2 substrates in human gastric cancer cells.


Assuntos
Caseína Quinase 1 épsilon/metabolismo , Caseína Quinase Idelta/metabolismo , Proteínas Quinases/metabolismo , Receptor de Colecistocinina B/metabolismo , Animais , Células COS , Linhagem Celular , Linhagem Celular Tumoral , Núcleo Celular/metabolismo , Chlorocebus aethiops , Proteínas de Ligação a DNA/metabolismo , Gastrinas/farmacologia , Células HeLa , Histona Desacetilases/metabolismo , Humanos , Membro 1 do Grupo A da Subfamília 4 de Receptores Nucleares , Fosforilação , Proteína Quinase C/metabolismo , Proteína Quinase D2 , Receptores Citoplasmáticos e Nucleares/metabolismo , Receptores de Esteroides/metabolismo , Serina/metabolismo , Fatores de Transcrição/metabolismo
10.
Gut ; 59(10): 1316-30, 2010 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-20732914

RESUMO

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.


Assuntos
Neoplasias Gastrointestinais/patologia , Proteínas Quinases/fisiologia , Animais , Comunicação Celular/fisiologia , Embrião de Galinha , Membrana Corioalantoide/irrigação sanguínea , Membrana Corioalantoide/enzimologia , Técnicas de Cocultura , Células Endoteliais/patologia , Endotélio Vascular/enzimologia , Neoplasias Gastrointestinais/irrigação sanguínea , Neoplasias Gastrointestinais/enzimologia , Técnicas de Silenciamento de Genes , Humanos , Camundongos , Camundongos Nus , Transplante de Neoplasias , Neovascularização Patológica/enzimologia , Neovascularização Patológica/patologia , Neoplasias Pancreáticas/irrigação sanguínea , Neoplasias Pancreáticas/enzimologia , Neoplasias Pancreáticas/patologia , Proteína Quinase D2 , Proteínas Quinases/genética , Proteínas Quinases/metabolismo , Transplante Heterólogo , Células Tumorais Cultivadas , Fator A de Crescimento do Endotélio Vascular/fisiologia
11.
ChemMedChem ; 16(14): 2158-2171, 2021 07 20.
Artigo em Inglês | MEDLINE | ID: mdl-33829655

RESUMO

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.


Assuntos
Desenvolvimento de Medicamentos , Proteína Quinase C/antagonistas & inibidores , Inibidores de Proteínas Quinases/farmacologia , Animais , Relação Dose-Resposta a Droga , Humanos , Estrutura Molecular , Proteína Quinase C/metabolismo , Inibidores de Proteínas Quinases/síntese química , Relação Estrutura-Atividade
12.
Eur J Med Chem ; 205: 112638, 2020 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-32835918

RESUMO

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.


Assuntos
Desenho de Fármacos , Proteína Quinase C/antagonistas & inibidores , Inibidores de Proteínas Quinases/síntese química , Inibidores de Proteínas Quinases/farmacologia , Pirazóis/química , Pirimidinas/síntese química , Pirimidinas/farmacologia , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Técnicas de Química Sintética , Humanos , Concentração Inibidora 50 , Fosforilação/efeitos dos fármacos , Inibidores de Proteínas Quinases/química , Pirimidinas/química
13.
Trends Cell Biol ; 12(4): 193-200, 2002 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-11978539

RESUMO

Recent research has identified protein kinase D (PKD, also called PKCmu) as a serine/threonine kinase with potentially important roles in growth factor signaling as well as in stress-induced signaling. Moreover, PKD has emerged as an important regulator of plasma membrane enzymes and receptors, in some cases mediating cross-talk between different signaling systems. The recent discovery of two additional kinases belonging to the PKD family and the plethora of proteins that interact with PKD point to a multifaceted regulation and a multifunctional role for these enzymes, with functions in processes as diverse as cell proliferation, apoptosis, immune cell regulation, tumor cell invasion and regulation of Golgi vesicle fission.


Assuntos
Proteína Quinase C/fisiologia , Sistemas do Segundo Mensageiro/fisiologia , Animais , Humanos , Proteína Quinase C/química , Proteína Quinase C/metabolismo , Proteínas Serina-Treonina Quinases/química , Proteínas Serina-Treonina Quinases/metabolismo , Proteínas Serina-Treonina Quinases/fisiologia , Receptor Cross-Talk
14.
Mol Cell Biol ; 25(6): 2364-83, 2005 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-15743830

RESUMO

Adaptive immune signaling can be coupled to stress-activated protein kinase (SAPK)/c-Jun N-terminal kinase (JNK) and NF-kappaB activation by the hematopoietic progenitor kinase 1 (HPK1), a mammalian hematopoiesis-specific Ste20 kinase. To gain insight into the regulation of leukocyte signal transduction, we investigated the molecular details of HPK1 activation. Here we demonstrate the capacity of the Src family kinase Lck and the SLP-76 family adaptor protein Clnk (cytokine-dependent hematopoietic cell linker) to induce HPK1 tyrosine phosphorylation and relocation to the plasma membrane, which in lymphocytes results in recruitment of HPK1 to the contact site of antigen-presenting cell (APC)-T-cell conjugates. Relocation and clustering of HPK1 cause its enzymatic activation, which is accompanied by phosphorylation of regulatory sites in the HPK1 kinase activation loop. We show that full activation of HPK1 is dependent on autophosphorylation of threonine 165 and phosphorylation of serine 171, which is a target site for protein kinase D (PKD) in vitro. Upon T-cell receptor stimulation, PKD robustly augments HPK1 kinase activity in Jurkat T cells and enhances HPK1-driven SAPK/JNK and NF-kappaB activation; conversely, antisense down-regulation of PKD results in reduced HPK1 activity. Thus, activation of major lymphocyte signaling pathways via HPK1 involves (i) relocation, (ii) autophosphorylation, and (iii) transphosphorylation of HPK1 by PKD.


Assuntos
Proteína Quinase C/metabolismo , Proteínas Serina-Treonina Quinases/metabolismo , Transdução de Sinais/imunologia , Linfócitos T/enzimologia , Proteínas Adaptadoras de Transdução de Sinal/fisiologia , Sequência de Aminoácidos , Aminoácidos/metabolismo , Animais , Células Apresentadoras de Antígenos/imunologia , Linhagem Celular , Membrana Celular/química , Membrana Celular/metabolismo , Humanos , Ativação Linfocitária/imunologia , Proteína Tirosina Quinase p56(lck) Linfócito-Específica/fisiologia , Proteína Quinase 8 Ativada por Mitógeno/metabolismo , Dados de Sequência Molecular , Mutação/genética , NF-kappa B/metabolismo , Mapeamento de Peptídeos , Fosforilação , Proteína Quinase C/genética , Proteínas Serina-Treonina Quinases/análise , Proteínas Serina-Treonina Quinases/genética , Estrutura Terciária de Proteína/genética , Estrutura Terciária de Proteína/fisiologia , Transporte Proteico/fisiologia , RNA Antissenso/genética , Receptores de Antígenos de Linfócitos T/agonistas , Receptores de Antígenos de Linfócitos T/imunologia , Linfócitos T/imunologia
15.
Biochem J ; 407(2): 303-11, 2007 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-17635105

RESUMO

IRBIT is an IP3R [IP3 (inositol 1,4,5-trisphosphate) receptor]-binding protein that competes with IP3 for binding to the IP3R. Phosphorylation of IRBIT is essential for the interaction with the IP3R. The unique N-terminal region of IRBIT, residues 1-104 for mouse IRBIT, contains a PEST (Pro-Glu-Ser-Thr) domain with many putative phosphorylation sites. In the present study, we have identified a well-conserved PP1 (protein phosphatase-1)-binding site preceeding this PEST domain which enabled the binding of PP1 to IRBIT both in vitro and in vivo. IRBIT emerged as a mediator of its own dephosphorylation by associated PP1 and, hence, as a novel substrate specifier for PP1. Moreover, IRBIT-associated PP1 specifically dephosphorylated Ser68 of IRBIT. Phosphorylation of Ser68 was required for subsequent phosphorylation of Ser71 and Ser74, but the latter two sites were not targeted by PP1. We found that phosphorylation of Ser71 and Ser74 were sufficient to enable inhibition of IP3 binding to the IP3R by IRBIT. Finally, we have shown that mutational inactivation of the docking site for PP1 on IRBIT increased the affinity of IRBIT for the IP3R. This pinpoints PP1 as a key player in the regulation of IP3R-controlled Ca2+ signals.


Assuntos
Adenosil-Homocisteinase/metabolismo , Canais de Cálcio/metabolismo , Glicoproteínas de Membrana/metabolismo , Proteína Fosfatase 1/fisiologia , Receptores Citoplasmáticos e Nucleares/metabolismo , Animais , Sítios de Ligação , Receptores de Inositol 1,4,5-Trifosfato , Camundongos , Fosforilação , Ligação Proteica
16.
Mol Biol Cell ; 16(9): 4375-85, 2005 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-15975900

RESUMO

Protein kinase D2 (PKD2) belongs to the PKD family of serine/threonine kinases that is activated by phorbol esters and G protein-coupled receptors (GPCRs). Its C-terminal regulatory domain comprises two cysteine-rich domains (C1a/C1b) followed by a pleckstrin homology (PH) domain. Here, we examined the role of the regulatory domain in PKD2 phorbol ester binding, catalytic activity, and subcellular localization: The PH domain is a negative regulator of kinase activity. C1a/C1b, in particular C1b, is required for phorbol ester binding and gastrin-stimulated PKD2 activation, but it has no inhibitory effect on the catalytic activity. Gastrin triggers nuclear accumulation of PKD2 in living AGS-B cancer cells. C1a/C1b, not the PH domain, plays a complex role in the regulation of nucleocytoplasmic shuttling: We identified a nuclear localization sequence in the linker region between C1a and C1b and a nuclear export signal in the C1a domain. In conclusion, our results define the critical components of the PKD2 regulatory domain controlling phorbol ester binding, catalytic activity, and nucleocytoplasmic shuttling and reveal marked differences to the regulatory properties of this domain in PKD1. These findings could explain functional differences between PKD isoforms and point to a functional role of PKD2 in the nucleus upon activation by GPCRs.


Assuntos
Proteínas de Transporte Nucleocitoplasmático/química , Proteínas de Transporte Nucleocitoplasmático/fisiologia , Ésteres de Forbol/metabolismo , Proteínas Quinases/química , Proteínas Quinases/fisiologia , Catálise , Linhagem Celular , Linhagem Celular Tumoral , Humanos , Carioferinas/fisiologia , Proteínas de Transporte Nucleocitoplasmático/genética , Proteínas de Transporte Nucleocitoplasmático/metabolismo , Dibutirato de 12,13-Forbol/metabolismo , Proteína Quinase C/química , Proteína Quinase C/genética , Proteína Quinase C/metabolismo , Proteína Quinase C/fisiologia , Proteína Quinase D2 , Proteínas Quinases/genética , Proteínas Quinases/metabolismo , Sinais Direcionadores de Proteínas/fisiologia , Estrutura Terciária de Proteína , Transporte Proteico/fisiologia , Receptores Citoplasmáticos e Nucleares/fisiologia , Neoplasias Gástricas/enzimologia , Trítio , Proteína Exportina 1
17.
Oxid Med Cell Longev ; 2018: 2138502, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29854077

RESUMO

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.


Assuntos
Estresse Oxidativo/efeitos dos fármacos , Isoformas de Proteínas/metabolismo , Proteína Quinase C/metabolismo , Humanos
18.
Oncogene ; 37(10): 1263-1278, 2018 03.
Artigo em Inglês | MEDLINE | ID: mdl-29259300

RESUMO

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.


Assuntos
Neoplasias/genética , Neoplasias/patologia , Proteínas Quinases/fisiologia , Animais , Proliferação de Células/genética , Humanos , Metástase Neoplásica/genética , Neovascularização Patológica/genética , Neovascularização Patológica/patologia , Proteína Quinase D2 , Proteínas Quinases/genética , Transdução de Sinais/genética
19.
FEBS Lett ; 592(14): 2432-2443, 2018 07.
Artigo em Inglês | MEDLINE | ID: mdl-29933512

RESUMO

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.


Assuntos
Proteína Quinase C/química , Proteína Quinase C/metabolismo , Tirosina/metabolismo , Sequência de Aminoácidos , Animais , Células COS , Células Cultivadas , Chlorocebus aethiops , Drosophila , Ativação Enzimática/genética , Células HEK293 , Homeostase/genética , Humanos , Mutagênese Sítio-Dirigida , Fosforilação/genética , Proteína Quinase C/genética , Tirosina/genética
20.
Assay Drug Dev Technol ; 5(5): 637-43, 2007 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-17939756

RESUMO

The protein kinase D (PKD) family is a novel group of kinases that are involved in the regulation of cell proliferation and apoptosis, and several other physiological processes. Hence, these enzymes are attractive targets for pharmacological intervention, but no specific PKD inhibitors are known. With this in mind, we have developed a high-throughput, non-radioactive enzyme-linked immunosorbent assay (ELISA) method to monitor the PKD activity with myelin basic protein (MBP) as substrate. We determined that MBP is phosphorylated by PKD on Ser-160 and that this phosphorylation can be quantified in ELISAs, by the use of phosphorylation site-specific antibodies. Antibodies were developed that are highly specific for the MBP peptide sequence surrounding the phosphorylated Ser-160. We show that our high-throughput kinase assay is useful not only for determining the cellular PKD activity but also to screen for PKD-inhibitory compounds. Our ELISA has advantages over the current radioisotope kinase assay in terms of simplicity and environmental safety.


Assuntos
Anticorpos/química , Inibidores Enzimáticos/farmacologia , Proteína Quinase C/antagonistas & inibidores , Trifosfato de Adenosina/metabolismo , Anticorpos/análise , Especificidade de Anticorpos , Proliferação de Células/efeitos dos fármacos , Colódio , DNA/biossíntese , Ensaio de Imunoadsorção Enzimática , Espectrometria de Massas , Membranas Artificiais , Proteína Básica da Mielina/química , Proteína Básica da Mielina/imunologia , Fosforilação , Proteína Quinase C/química , Proteína Quinase C/isolamento & purificação , Especificidade por Substrato
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA