Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
Add more filters










Publication year range
1.
Proc Natl Acad Sci U S A ; 120(7): e2212909120, 2023 02 14.
Article in English | MEDLINE | ID: mdl-36745811

ABSTRACT

Phosphorylation is a ubiquitous mechanism by which signals are transduced in cells. Protein kinases, enzymes that catalyze the phosphotransfer reaction are, themselves, often regulated by phosphorylation. Paradoxically, however, a substantial fraction of more than 500 human protein kinases are capable of catalyzing their own activation loop phosphorylation. Commonly, these kinases perform this autophosphorylation reaction in trans, whereby transient dimerization leads to the mutual phosphorylation of the activation loop of the opposing protomer. In this study, we demonstrate that protein kinase D (PKD) is regulated by the inverse mechanism of dimerization-mediated trans-autoinhibition, followed by activation loop autophosphorylation in cis. We show that PKD forms a stable face-to-face homodimer that is incapable of either autophosphorylation or substrate phosphorylation. Dissociation of this trans-autoinhibited dimer results in activation loop autophosphorylation, which occurs exclusively in cis. Phosphorylation serves to increase PKD activity and prevent trans-autoinhibition, thereby switching PKD on. Our findings not only reveal the mechanism of PKD regulation but also have profound implications for the regulation of many other eukaryotic kinases.


Subject(s)
Protein Kinase C , Humans , Phosphorylation/physiology , Protein Kinase C/metabolism
2.
Elife ; 72018 07 20.
Article in English | MEDLINE | ID: mdl-30028295

ABSTRACT

Protein kinase D (PKD) is a family of serine/threonine kinases that is required for the structural integrity and function of the Golgi complex. Despite its importance in the regulation of Golgi function, the molecular mechanisms regulating PKD activity are still incompletely understood. Using the genetically encoded PKD activity reporter G-PKDrep we now uncover a Rho signaling network comprising GEF-H1, the RhoGAP DLC3, and the Rho effector PLCε that regulate the activation of PKD at trans-Golgi membranes. We further show that this molecular network coordinates the formation of TGN-derived Rab6-positive transport carriers delivering cargo for localized exocytosis at focal adhesions.


Subject(s)
Focal Adhesions/physiology , Microtubules/metabolism , Protein Kinase C/metabolism , Signal Transduction , trans-Golgi Network/metabolism , Cytoskeleton , HEK293 Cells , HeLa Cells , Humans , Phosphoinositide Phospholipase C/genetics , Phosphoinositide Phospholipase C/metabolism , Protein Transport , Rho Guanine Nucleotide Exchange Factors/genetics , Rho Guanine Nucleotide Exchange Factors/metabolism , TRPP Cation Channels/genetics , TRPP Cation Channels/metabolism , rab GTP-Binding Proteins/genetics , rab GTP-Binding Proteins/metabolism , rhoA GTP-Binding Protein/genetics , rhoA GTP-Binding Protein/metabolism
3.
Mol Biol Cell ; 28(2): 285-295, 2017 01 15.
Article in English | MEDLINE | ID: mdl-27852895

ABSTRACT

Ras and Rab interactor 1 (RIN1) is predominantly expressed in the nervous system. RIN1-knockout animals have deficits in latent inhibition and fear extinction in the amygdala, suggesting a critical role for RIN1 in preventing the persistence of unpleasant memories. At the molecular level, RIN1 signals through Rab5 GTPases that control endocytosis of cell-surface receptors and Abl nonreceptor tyrosine kinases that participate in actin cytoskeleton remodeling. Here we report that RIN1 controls the plasticity of cultured mouse hippocampal neurons. Our results show that RIN1 affects the morphology of dendritic protrusions and accelerates dendritic filopodial motility through an Abl kinase-dependent pathway. Lack of RIN1 results in enhanced mEPSC amplitudes, indicating an increase in surface AMPA receptor levels compared with wild-type neurons. We further provide evidence that the Rab5 GEF activity of RIN1 regulates surface GluA1 subunit endocytosis. Consequently loss of RIN1 blocks surface AMPA receptor down-regulation evoked by chemically induced long-term depression. Our findings indicate that RIN1 destabilizes synaptic connections and is a key player in postsynaptic AMPA receptor endocytosis, providing multiple ways of negatively regulating memory stabilization during neuronal plasticity.


Subject(s)
rab GTP-Binding Proteins/metabolism , rab GTP-Binding Proteins/physiology , Animals , Cell Movement/physiology , Dendrites/metabolism , Dendrites/physiology , Endocytosis/physiology , Hippocampus/physiology , Intracellular Signaling Peptides and Proteins/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Neuronal Plasticity/physiology , Neurons/metabolism , Proto-Oncogene Proteins c-abl/metabolism , Pseudopodia/metabolism , Pseudopodia/physiology , Receptors, AMPA/metabolism , Receptors, AMPA/physiology , Signal Transduction/physiology , Synaptic Membranes/physiology , rab5 GTP-Binding Proteins/metabolism
4.
EMBO Rep ; 12(6): 527-33, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21525957

ABSTRACT

By using mass spectrometry, we have identified Ser 402 as a new phosphorylation site within the catalytic domain of human slingshot 1 (SSH1). Phosphorylation at this site inhibits substrate binding and, thus, phosphatase activity in vitro, resulting in enrichment of phosphorylated cofilin in monolayer cell culture. We further demonstrate that protein kinase D (PKD) is upstream from Ser 402 phosphorylation. Accordingly, expression of active PKD in Drosophila phenotypically mimics the loss of SSH activity by inducing accumulation of phosphorylated cofilin and filamentous actin. We thus identify a universal mechanism by which PKD controls SSH1 phosphatase activity.


Subject(s)
Phosphoprotein Phosphatases/metabolism , Serine/metabolism , Actin Depolymerizing Factors/metabolism , Actins/metabolism , Amino Acid Sequence , Amino Acid Substitution/genetics , Enzyme Activation/genetics , HEK293 Cells , HeLa Cells , Humans , Molecular Sequence Data , Phosphoprotein Phosphatases/genetics , Phosphorylation , Protein Binding , Protein Kinase C/chemistry , Protein Kinase C/metabolism , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Sequence Alignment
5.
Mol Biol Cell ; 22(5): 570-80, 2011 Mar 01.
Article in English | MEDLINE | ID: mdl-21209314

ABSTRACT

The multifunctional signal adapter protein Ras and Rab interactor 1 (RIN1) is a Ras effector protein involved in the regulation of epithelial cell processes such as cell migration and endocytosis. RIN1 signals via two downstream pathways, namely the activation of Rab5 and Abl family kinases. Protein kinase D (PKD) phosphorylates RIN1 at serine 351 in vitro, thereby regulating interaction with 14-3-3 proteins. Here, we report the identification of serine 292 in RIN1 as an in vivo PKD phosphorylation site. PKD-mediated phosphorylation at this site was confirmed with a phospho-specific antibody and by mass spectrometry. We demonstrate that phosphorylation at serine 292 controls RIN1-mediated inhibition of cell migration by modulating the activation of Abl kinases. We further provide evidence that RIN1 in vivo phosphorylation at serine 351 occurs independently of PKD. Collectively, our data identify a novel PKD signaling pathway through RIN1 and Abl kinases that is involved in the regulation of actin remodeling and cell migration.


Subject(s)
Cell Movement , Intracellular Signaling Peptides and Proteins/chemistry , Intracellular Signaling Peptides and Proteins/metabolism , Protein Kinase C/metabolism , Actins/metabolism , Amino Acid Motifs , Amino Acid Sequence , Antibodies/immunology , HEK293 Cells , Humans , Molecular Sequence Data , Phosphorylation , Phosphoserine/metabolism , Protein Transport , Protein-Tyrosine Kinases/metabolism , Subcellular Fractions/metabolism , Tumor Suppressor Proteins/metabolism
6.
Cancer Res ; 69(14): 5634-8, 2009 Jul 15.
Article in English | MEDLINE | ID: mdl-19567672

ABSTRACT

Protein kinase D (PKD) has been identified as a negative regulator of epithelial cell migration; however, its molecular substrates and downstream signaling pathways that mediate this activity have remained elusive. In this study, we provide evidence that the cofilin phosphatase slingshot 1 like (SSH1L), an important regulator of the complex actin remodeling machinery, is a novel in vivo PKD substrate. PKD-mediated phosphorylation of serines 937 and 978 regulates SSH1L subcellular localization by binding of 14-3-3 proteins and thus impacts the control of local cofilin activation and actin remodeling during cell migration. In line with this, we show that the loss of PKD decreases cofilin phosphorylation, induces a more spread cell morphology, and stimulates chemotactic migration of breast cancer cells in an SSHL1-dependent fashion. Our data thus identify PKD as a central regulator of the cofilin signaling network via direct phosphorylation and regulation of SSH1L.


Subject(s)
Cell Movement , Cofilin 1/metabolism , Phosphoprotein Phosphatases/metabolism , Protein Kinase C/metabolism , Animals , COS Cells , Cell Line , Cell Line, Tumor , Chlorocebus aethiops , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Humans , Immunoblotting , Microscopy, Confocal , Microscopy, Fluorescence , Mutation , Phosphoprotein Phosphatases/genetics , Phosphorylation , Protein Kinase C/genetics , RNA, Small Interfering/genetics , Serine/metabolism , Transfection
7.
Traffic ; 10(7): 858-67, 2009 Jul.
Article in English | MEDLINE | ID: mdl-19416469

ABSTRACT

The protein kinase D (PKD) family comprises multifunctional serine/threonine-specific protein kinases with three mammalian isoforms: PKD1, PKD2 and PKD3. A prominent PKD function is the regulation of basolateral-targeted transport carrier fission from the trans-Golgi network (TGN). To visualize site-specific PKD activation at this organelle, we designed a molecular reporter consisting of a PKD-specific substrate sequence fused to enhanced green fluorescent protein (EGFP), specifically targeted to the TGN via the p230 GRIP domain. Quantitative analyses using a phosphospecific antibody and ratiometric fluorescence imaging revealed that Golgi-specific phosphorylation of the reporter was strictly dependent on stimulation of endogenous PKD or transient expression of active PKD constructs. Conversely, PKD-specific pharmacological inhibitors and siRNA-mediated PKD knockdown suppressed reporter phosphorylation. Using this reporter we investigated a potential role for PKD in the regulation of Golgi complex morphology. Interestingly, nocodazole-induced Golgi complex break-up and dispersal was associated with local PKD activation as measured by reporter phosphorylation and this was efficiently blocked by expression of a dominant-negative PKD mutant or PKD depletion. Our data thus identify a novel link between PKD activity and the microtubule cytoskeleton, whereby Golgi complex integrity is regulated.


Subject(s)
Genes, Reporter , Golgi Apparatus , Isoenzymes/metabolism , Nocodazole/pharmacology , Protein Kinase C/metabolism , Tubulin Modulators/pharmacology , Amino Acid Sequence , Animals , Cell Line , Enzyme Activation , Golgi Apparatus/drug effects , Golgi Apparatus/metabolism , Humans , Molecular Sequence Data , Protein Kinase C/antagonists & inhibitors , Protein Kinase C/genetics , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism
8.
J Cell Sci ; 119(Pt 17): 3613-21, 2006 Sep 01.
Article in English | MEDLINE | ID: mdl-16912074

ABSTRACT

Phosphatidylinositol-4-kinase-IIIbeta (PI4KIIIbeta) is activated at the Golgi compartment by PKD-mediated phosphorylation. Subsequent mechanisms responsible for continuous PtdIns(4)P production at Golgi membranes and potential interaction partners of activated PI4KIIIbeta are unknown. Here we identify phosphoserine/-threonine binding 14-3-3 proteins as novel regulators of PI4KIIIbeta activity downstream of this phosphorylation. The PI4KIIIbeta-14-3-3 interaction, evident from GST pulldowns, co-immunoprecipitations and bimolecular fluorescence complementation, was augmented by phosphatase inhibition with okadaic acid. Binding of 14-3-3 proteins to PI4KIIIbeta involved the PKD phosphorylation site Ser294, evident from reduced 14-3-3 binding to a S294A PI4KIIIbeta mutant. Expression of dominant negative 14-3-3 proteins resulted in decreased PI4KIIIbeta Ser294 phosphorylation, whereas wildtype 14-3-3 proteins increased phospho-PI4KIIIbeta levels. This was because of protection of PI4KIIIbeta Ser294 phosphorylation from phosphatase-mediated dephosphorylation. The functional significance of the PI4KIIIbeta-14-3-3 interaction was evident from a reduction of PI4KIIIbeta activity upon dominant negative 14-3-3 protein expression. We propose that 14-3-3 proteins function as positive regulators of PI4KIIIbeta activity by protecting the lipid kinase from active site dephosphorylation, thereby ensuring a continuous supply of PtdIns(4)P at the Golgi compartment.


Subject(s)
1-Phosphatidylinositol 4-Kinase/metabolism , 14-3-3 Proteins/metabolism , Isoenzymes/metabolism , 1-Phosphatidylinositol 4-Kinase/genetics , 14-3-3 Proteins/chemistry , 14-3-3 Proteins/genetics , Amino Acid Sequence , Animals , Binding Sites , Cell Line , Dimerization , Humans , Isoenzymes/genetics , Phosphorylation , Protein Binding , Protein Kinase C/metabolism , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Second Messenger Systems/physiology , Serine/metabolism
9.
Gene Expr Patterns ; 6(8): 849-56, 2006 Oct.
Article in English | MEDLINE | ID: mdl-16750940

ABSTRACT

Protein kinase D belongs to the subfamily of CaMK. In mammals, three isoforms are known. They have been linked to diverse cellular functions including regulation of cell proliferation, differentiation, apoptosis and motility as well as secretory transport from the trans-Golgi compartment to the plasma membrane. Accordingly, the mammalian PKDs show different intracellular locations, with reported dynamic redistribution, between cytosol, Golgi, plasma membranes and the nucleus, depending on the cell type and exogenous stimuli. The genome of Drosophila melanogaster harbours just one, highly conserved PKD homologue, which is expressed throughout development. PKD mRNA expression during late embryogenesis is restricted to ectodermal derivatives including those involved in cuticle secretion. In imaginal tissues, transcription appears more uniform. PKD protein is detected predominantly in the cytosol with an enrichment in lateral apodemes of late embryos as well as in larval fascicles. In secretory tissues like salivary glands, the protein is concentrated in dotted structures. A PKD-GFP transgene reveals a similar punctuate protein accumulation juxtaposed to a resident Golgi-marker. In cultured cells, transfected Drosophila PKD-GFP colocalizes with a marker of the trans-Golgi compartment like human PKD1-GFP. Similar to the mammalian homologues, Drosophila PKD may be multifunctional including a role in secretory transport in accordance with its subcellular distribution.


Subject(s)
Drosophila melanogaster/embryology , Drosophila melanogaster/metabolism , Golgi Apparatus/metabolism , Protein Kinase C/metabolism , Amino Acid Sequence , Animals , Biological Transport/physiology , COS Cells , Cells, Cultured , Chlorocebus aethiops , Drosophila melanogaster/genetics , Embryo, Nonmammalian , Embryonic Development/physiology , Gene Dosage , Green Fluorescent Proteins/metabolism , Humans , Models, Biological , Molecular Sequence Data , Protein Kinase C/genetics , Protein Kinase C/physiology , Recombinant Fusion Proteins/metabolism , Sequence Homology, Amino Acid , Tissue Distribution , Transcription, Genetic , Transfection
10.
Nat Cell Biol ; 7(9): 880-6, 2005 Sep.
Article in English | MEDLINE | ID: mdl-16100512

ABSTRACT

Protein kinase D (PKD) regulates the fission of vesicles originating from the trans-Golgi network. We show that phosphatidylinositol 4-kinase IIIbeta (PI4KIIIbeta) - a key player in the structure and function of the Golgi complex - is a physiological substrate of PKD. Of the three PKD isoforms, only PKD1 and PKD2 phosphorylated PI4KIIIbeta at a motif that is highly conserved from yeast to humans. PKD-mediated phosphorylation stimulated lipid kinase activity of PI4KIIIbeta and enhanced vesicular stomatitis virus G-protein transport to the plasma membrane. The identification of PI4KIIIbeta as one of the PKD substrates should help to reveal the molecular events that enable transport-carrier formation.


Subject(s)
1-Phosphatidylinositol 4-Kinase/metabolism , Chlorocebus aethiops , Golgi Apparatus/enzymology , Protein Kinase C/metabolism , Transport Vesicles/enzymology , Amino Acid Motifs/physiology , Animals , Biological Transport, Active/physiology , COS Cells , Cell Membrane/enzymology , Conserved Sequence/physiology , Enzyme Activation/physiology , Evolution, Molecular , Golgi Apparatus/metabolism , Humans , Lipid Metabolism , Membrane Glycoproteins/metabolism , Phosphorylation , Protein Isoforms/metabolism , Viral Envelope Proteins/metabolism
11.
J Cell Biol ; 156(1): 65-74, 2002 Jan 07.
Article in English | MEDLINE | ID: mdl-11777941

ABSTRACT

We here describe the structural requirements for Golgi localization and a sequential, localization-dependent activation process of protein kinase C (PKC) mu involving auto- and transphosphorylation. The structural basis for Golgi compartment localization was analyzed by confocal microscopy of HeLa cells expressing various PKC mu-green fluorescent protein fusion proteins costained with the Golgi compartment-specific markers p24 and p230. Deletions of either the NH(2)-terminal hydrophobic or the cysteine region, but not of the pleckstrin homology or the acidic domain, of PKC mu completely abrogated Golgi localization of PKC mu. As an NH(2)-terminal PKC mu fragment was colocalized with p24, this region of PKC mu is essential and sufficient to mediate association with Golgi membranes. Fluorescence recovery after photobleaching studies confirmed the constitutive, rapid recruitment of cytosolic PKC mu to, and stable association with, the Golgi compartment independent of activation loop phosphorylation. Kinase activity is not required for Golgi complex targeting, as evident from microscopical and cell fractionation studies with kinase-dead PKC mu found to be exclusively located at intracellular membranes. We propose a sequential activation process of PKC mu, in which Golgi compartment recruitment precedes and is essential for activation loop phosphorylation (serines 738/742) by a transacting kinase, followed by auto- and transphosphorylation of NH(2)-terminal serine(s) in the regulatory domain. PKC mu activation loop phosphorylation is indispensable for substrate phosphorylation and thus PKC mu function at the Golgi compartment.


Subject(s)
Golgi Apparatus/metabolism , Protein Kinase C/chemistry , Protein Kinase C/metabolism , Blotting, Western , Cell Line , Enzyme Activation , Green Fluorescent Proteins , HeLa Cells , Humans , Intracellular Membranes/metabolism , Luminescent Proteins/metabolism , Microscopy, Confocal , Microscopy, Fluorescence , Models, Biological , Mutation , Phosphorylation , Phosphoserine/metabolism , Protein Binding , Protein Kinase C/genetics , Protein Structure, Tertiary
12.
J Biol Chem ; 277(8): 6490-6, 2002 Feb 22.
Article in English | MEDLINE | ID: mdl-11741879

ABSTRACT

Protein kinase C (PKC), a family of lipid-activated serine kinases, is involved in multiple functions in the regulation of growth control. The PKC-related isoform PKC mu/PKD has been implicated in mitogenic signal cascades because of the activation of p42/p44 MAPK leading to Elk1-mediated gene transcription, and PKC mu/PKD has been shown to be activated via a PKC-dependent pathway. By using confocal analyses, we demonstrate here that PKC mu partially colocalizes with PKC eta in different cell types. Colocalization depends on the presence of the PKC mu pleckstrin homology domain. Coexpression of constitutively active PKC eta with PKC mu leads to a significant enhancement of the PKC mu substrate phosphorylation capacity as a result of an increased phosphorylation of the activation loop Ser(738/742) of PKC mu, whereas Ser(910) autophosphorylation remains unaffected. In vitro phosphorylation experiments show that PKC eta directly phosphorylates PKC mu on activation loop serines. Consequently, the p42 MAPK cascade is triggered leading to an increase in reporter gene activity driven by a serum-responsive element in HEK293 cells. At the same time, PKC eta-mediated JNK activation is reduced, providing evidence for a mutual regulation of PKC mu/PKC eta affecting different arms of the p38/ERK/JNK pathways. Our data provide evidence for the sequential involvement of selective PKC isoforms in kinase cascades and identify the relevant domains in PKC mu for interaction with and activation by PKC eta as pleckstrin homology domain and activation loop.


Subject(s)
Mitogen-Activated Protein Kinases/metabolism , Protein Kinase C/metabolism , Breast Neoplasms , Cell Line , DNA Primers , Enzyme Activation , Female , Genes, Reporter , Glutathione Transferase/genetics , Green Fluorescent Proteins , Humans , JNK Mitogen-Activated Protein Kinases , Jurkat Cells , Luminescent Proteins/genetics , Protein Kinase C/genetics , Recombinant Fusion Proteins/metabolism , Transfection
SELECTION OF CITATIONS
SEARCH DETAIL
...